1
|
Jia Z, Yu X, Wang X, Li J. Therapeutic Effects of Coenzyme Q10 in the Treatment of Ischemic Stroke. Curr Nutr Rep 2024; 13:679-690. [PMID: 39227555 DOI: 10.1007/s13668-024-00568-2] [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] [Accepted: 08/20/2024] [Indexed: 09/05/2024]
Abstract
PURPOSE OF REVIEW Ischemic stroke is the second deadly disease worldwide, but current treatment is very limited. The brain, rich in lipids and high in oxygen consumption, is susceptible to damage from oxidative stress after ischemic stroke. Thus, antioxidants are promising neuroprotective agents for treatment and prevention of ischemic stroke. Coenzyme Q10 is the only lipophilic antioxidant that can be synthesized de novo by cells and plays a key role as an electron carrier in the oxidative phosphorylation of the mitochondrial electron transport chain. However, the reduced form of coenzyme Q10 (Ubiquinol) levels are significantly deficient in the brain. The aim of this article is to review the therapeutic effects and mechanisms of coenzyme Q10 in ischemic stroke. RECENT FINDINGS Current studies have found that coenzyme Q10 protects and treats ischemic stroke through multiple mechanisms based on the evidence from in vitro experiments, in vivo experiments, and clinical observations. For the first time, we reviewed the neuroprotective effects of coenzyme Q10 in ischemic stroke. Coenzyme Q10 exerts neuroprotective effects after ischemic stroke through anti-oxidative stress, anti-nitrosative stress, anti-inflammation, and anti-cell death. Here, we provided the evidence on the therapeutic and preventive effects of coenzyme Q10 in ischemic stroke and suggested the potential value of coenzyme Q10 as a medication candidate.
Collapse
Affiliation(s)
- Zhilei Jia
- Science and Technology Innovation Platform Management Center of Jilin Province, Changchun, Jilin, 130000, China
| | - Xiaoya Yu
- Science and Technology Innovation Platform Management Center of Jilin Province, Changchun, Jilin, 130000, China
| | - Xu Wang
- School of Public Health, Jilin University, Changchun, Jilin, 130021, China.
- College of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, 130117, China.
| | - Jinhua Li
- School of Public Health, Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
2
|
Sharma C, Kim S, Nam Y, Jung UJ, Kim SR. Mitochondrial Dysfunction as a Driver of Cognitive Impairment in Alzheimer's Disease. Int J Mol Sci 2021; 22:ijms22094850. [PMID: 34063708 PMCID: PMC8125007 DOI: 10.3390/ijms22094850] [Citation(s) in RCA: 119] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 04/30/2021] [Accepted: 05/01/2021] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is the most frequent cause of age-related neurodegeneration and cognitive impairment, and there are currently no broadly effective therapies. The underlying pathogenesis is complex, but a growing body of evidence implicates mitochondrial dysfunction as a common pathomechanism involved in many of the hallmark features of the AD brain, such as formation of amyloid-beta (Aβ) aggregates (amyloid plaques), neurofibrillary tangles, cholinergic system dysfunction, impaired synaptic transmission and plasticity, oxidative stress, and neuroinflammation, that lead to neurodegeneration and cognitive dysfunction. Indeed, mitochondrial dysfunction concomitant with progressive accumulation of mitochondrial Aβ is an early event in AD pathogenesis. Healthy mitochondria are critical for providing sufficient energy to maintain endogenous neuroprotective and reparative mechanisms, while disturbances in mitochondrial function, motility, fission, and fusion lead to neuronal malfunction and degeneration associated with excess free radical production and reduced intracellular calcium buffering. In addition, mitochondrial dysfunction can contribute to amyloid-β precursor protein (APP) expression and misprocessing to produce pathogenic fragments (e.g., Aβ1-40). Given this background, we present an overview of the importance of mitochondria for maintenance of neuronal function and how mitochondrial dysfunction acts as a driver of cognitive impairment in AD. Additionally, we provide a brief summary of possible treatments targeting mitochondrial dysfunction as therapeutic approaches for AD.
Collapse
Affiliation(s)
- Chanchal Sharma
- School of Life Sciences, Kyungpook National University, Daegu 41566, Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Sehwan Kim
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Korea; (S.K.); (Y.N.)
| | - Youngpyo Nam
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Korea; (S.K.); (Y.N.)
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Korea;
| | - Sang Ryong Kim
- School of Life Sciences, Kyungpook National University, Daegu 41566, Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Korea; (S.K.); (Y.N.)
- Correspondence: ; Tel.: +82-53-950-7362; Fax: +82-53-943-2762
| |
Collapse
|
3
|
Wang Y, Wu Q, Anand BG, Karthivashan G, Phukan G, Yang J, Thinakaran G, Westaway D, Kar S. Significance of cytosolic cathepsin D in Alzheimer's disease pathology: Protective cellular effects of PLGA nanoparticles against β-amyloid-toxicity. Neuropathol Appl Neurobiol 2020; 46:686-706. [PMID: 32716575 DOI: 10.1111/nan.12647] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/25/2020] [Accepted: 07/12/2020] [Indexed: 12/16/2022]
Abstract
BACKGROUND Evidence suggests that amyloid β (Aβ) peptides play an important role in the degeneration of neurons during the development of Alzheimer's disease (AD), the prevalent cause of dementia affecting the elderly. The endosomal-lysosomal system, which acts as a major site for Aβ metabolism, has been shown to exhibit abnormalities in vulnerable neurons of the AD brain, reflected by enhanced levels/expression of lysosomal enzymes including cathepsin D (CatD). At present, the implication of CatD in selective neuronal vulnerability in AD pathology remains unclear. METHODS We evaluated the role of CatD in the degeneration of neurons in Aβ-treated cultures, transgenic AD mouse model (that is 5xFAD) and post mortem AD brain samples. RESULTS Our results showed that Aβ1-42 -induced toxicity in cortical cultured neurons is associated with impaired lysosomal integrity, enhanced levels of carbonylated proteins and tau phosphorylation. The cellular and cytosolic levels/activity of CatD are also elevated in cultured neurons following exposure to Aβ peptide. Additionally, we observed that CatD cellular and subcellular levels/activity are increased in the affected cortex, but not in the unaffected cerebellum, of 5xFAD mice and post mortem AD brains. Interestingly, treatment of cultured neurons with nanoparticles PLGA, which targets lysosomal system, attenuated Aβ toxicity by reducing the levels of carbonylated proteins, tau phosphorylation and the level/distribution/activity of CatD. CONCLUSION Our study reveals that increased cytosolic level/activity of CatD play an important role in determining neuronal vulnerability in AD. Additionally, native PLGA can protect neurons against Aβ toxicity by restoring lysosomal membrane integrity, thus signifying its implication in attenuating AD.
Collapse
Affiliation(s)
- Y Wang
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada.,Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - Q Wu
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - B G Anand
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - G Karthivashan
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - G Phukan
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - J Yang
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| | - G Thinakaran
- Department of Molecular Medicine, and Byrd Alzheimer's Institute, University of South Florida, Tampa, FL, 33613, USA
| | - D Westaway
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada.,Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - S Kar
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada.,Department of Medicine, University of Alberta, Edmonton, Alberta, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
4
|
Liu Y, Eaton ED, Wills TE, McCann SK, Antonic A, Howells DW. Human Ischaemic Cascade Studies Using SH-SY5Y Cells: a Systematic Review and Meta-Analysis. Transl Stroke Res 2018; 9:564-574. [PMID: 29572690 PMCID: PMC6208743 DOI: 10.1007/s12975-018-0620-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/03/2018] [Accepted: 03/06/2018] [Indexed: 01/21/2023]
Abstract
Low translational yield for stroke may reflect the focus of discovery science on rodents rather than humans. Just how little is known about human neuronal ischaemic responses is confirmed by systematic review and meta-analysis revealing that data for the most commonly used SH-SY5Y human cells comprises only 84 papers. Oxygen-glucose deprivation, H2O2, hypoxia, glucose-deprivation and glutamate excitotoxicity yielded - 58, - 61, - 29, - 45 and - 49% injury, respectively, with a dose-response relationship found only for H2O2 injury (R2 = 29.29%, p < 0.002). Heterogeneity (I2 = 99.36%, df = 132, p < 0.0001) was largely attributable to the methods used to detect injury (R2 = 44.77%, p < 0.000) with cell death assays detecting greater injury than survival assays (- 71 vs - 47%, R2 = 28.64%, p < 0.000). Seventy-four percent of publications provided no description of differentiation status, but in the 26% that did, undifferentiated cells were susceptible to greater injury (R2 = 4.13%, p < 0.047). One hundred and sixty-nine interventions improved average survival by 34.67% (p < 0.0001). Eighty-eight comparisons using oxygen-glucose deprivation found both benefit and harm, but studies using glutamate and H2O2 injury reported only improvement. In studies using glucose deprivation, intervention generally worsened outcome. There was insufficient data to rank individual interventions, but of the studies reporting greatest improvement (> 90% effect size), 7/13 were of herbal medicine constituents (24.85% of the intervention dataset). We conclude that surprisingly little is known of the human neuronal response to ischaemic injury, and that the large impact of methodology on outcome indicates that further model validation is required. Lack of evidence for randomisation, blinding or power analysis suggests that the intervention data is at substantial risk of bias.
Collapse
Affiliation(s)
- Ye Liu
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade, The University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Emma D Eaton
- School of Medicine, Faculty of Health, University of Tasmania, Medical Sciences Precinct, 17 Liverpool Street, Hobart, TAS, 7000, Australia
| | - Taryn E Wills
- Melbourne Brain Centre, Florey Institute of Neuroscience and Mental Health, 245 Burgundy St, Heidelberg, VIC, 3084, Australia
| | - Sarah K McCann
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, EH16 4SB, UK
| | - Ana Antonic
- Department of Neuroscience, Monash University, Melbourne, VIC, 3004, Australia
| | - David W Howells
- School of Medicine, Faculty of Health, University of Tasmania, Medical Sciences Precinct, 17 Liverpool Street, Hobart, TAS, 7000, Australia.
- School of Medicine, Faculty of Health, University of Tasmania, Medical Sciences Precinct, 17 Liverpool Street, Hobart, TAS, 7000, Australia.
| |
Collapse
|
5
|
De Benedetto F, Pastorelli R, Ferrario M, de Blasio F, Marinari S, Brunelli L, Wouters EFM, Polverino F, Celli BR. Supplementation with Qter ® and Creatine improves functional performance in COPD patients on long term oxygen therapy. Respir Med 2018; 142:86-93. [PMID: 30170808 DOI: 10.1016/j.rmed.2018.08.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 08/01/2018] [Accepted: 08/05/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Skeletal muscle dysfunction and poor functional capacity are important extra-pulmonary manifestations of chronic obstructive pulmonary disease (COPD), especially in COPD patients on long-term O2 therapy (LTOT). Beside the role of pulmonary rehabilitation, the effect of nutritional interventions is still controversial, and there are knowledge gaps on the effective role of nutraceutical supplementation on hard endpoints. The aim of this study was to investigate the effects of nutritional supplementation with Coenzyme Q10 (QTer®) - a powerful antioxidant with the potential to reduce oxidative stress and improve mitochondrial function - and Creatine on functional, nutritional, and metabolomic profile in COPD patients on long-term O2 therapy. METHODS One-hundred and eight patients with COPD from 9 Italian hospitals were enrolled in this double-blinded randomized placebo-controlled clinical study. At baseline and after 2 months of therapy, the patients underwent spirometry, 6-minute walk test (6MWT), bioelectrical impedance analysis, and activities of daily living questionnaire (ADL). Also, dyspnea scores and BODE index were calculated. At both time points, plasma concentration of CoQ10 and metabolomic profiling were measured. FINDINGS Ninety patients, who randomly received supplementation with QTer® and Creatine or placebo, completed the study. Compared with placebo, supplemented patients showed improvements in 6MWT (51 ± 69 versus 15 ± 91 m, p < 0.05), body cell mass and phase angle, sodium/potassium ratio, dyspnea indices and ADL score. The CoQ10 plasma concentration increased in the supplementation group whereas it did not change in the placebo group. The metabolomics profile also differed between groups. Adverse events were similar in both groups. INTERPRETATION These results show that in patients with COPD, dietary supplementation with CoQ10 and Creatine improves functional performance, body composition and perception of dyspnea. A systemic increase in some anti-inflammatory metabolites supports a pathobiological mechanism as a reason for these benefits. Further trials should help clarifying the role of QTer® and Creatine supplementation in patients with COPD.
Collapse
Affiliation(s)
| | - Roberta Pastorelli
- Department of Environmental Health Science, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | - Manuela Ferrario
- Department of Electronics, Information, and Bioengineering (DEIB), Politecnico di Milano, Milan, Italy
| | | | | | - Laura Brunelli
- Department of Environmental Health Science, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, Milano, Italy
| | - Emiel F M Wouters
- Department of Respiratory Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | | | - Bartolome R Celli
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | | |
Collapse
|
6
|
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
|
7
|
Kalatanova AV, Makarov VG, Faustova NM, Gushchin YI, Makarova MN. [Evaluation of the cardioprotective effect of ubiquinol on the model of reperfusion injury of rat myocardium]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2018; 64:188-194. [PMID: 29723149 DOI: 10.18097/pbmc20186402188] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The cardioprotective effect of ubiquinol on the model of myocardium reperfusion injury in rats was investigated. The study was carried out using mature males of outbred rats. Myocardial ischemia-reperfusion injury was performed after 30-minute ligation of the left coronary artery followed by reperfusion. The main criteria for assessing the development of pathology included the results of electrocardiography, biochemical analysis of blood plasma, histological and histochemical study of the myocardium. Development of the reperfusion damage of the myocardium caused specific changes in non-treated animals. The best therapeutic effect on biochemical indices was provided by a drug with the known cardioprotective activity - Mexidolâ and the tested object ubiquinol at doses of 2-6 mg/kg. Evaluation of the results of electrocardiography allowed to confirm the development of ischemic myocardial damage in all groups. The results of histochemical and histological examination of the myocardium suggest a high cardioprotective activity of ubiquinol at a dose of 3 mg/kg and a potential cardioprotective effect of ubiquinol in doses closest to the therapeutic doses of 2 and 6 mg/kg. Ubiquinol is a dose 9 mg/kg showed signs of prooxidant activity, manifested in the form of aggravation of reperfusion injury of the myocardium. The most effective in the conditions of experimental pathology is 1% solution of ubiquinol, at a dose of 3 mg/kg, whose cardioprotective effect is comparable or higher than that for the reference drug Mexidolâ at the therapeutic dose. In doses that are greater than therapeutic ubiquinol is able to act as a pro-oxidant.
Collapse
Affiliation(s)
- A V Kalatanova
- ICJSC "Saint-Petersburg Institute of Pharmacy", Kuzmolovskiy, Leningradskaya reg., Vsevolozhskiy distr., Russia
| | - V G Makarov
- ICJSC "Saint-Petersburg Institute of Pharmacy", Kuzmolovskiy, Leningradskaya reg., Vsevolozhskiy distr., Russia
| | - N M Faustova
- CJSC "Institute of experimental pharmacology", Leningradskaya reg., Russia
| | - Ya I Gushchin
- JSC "Research-and-manufacturing company "HOME OF PHARMACY"", Leningradskaya reg., Russia
| | - M N Makarova
- JSC "Research-and-manufacturing company "HOME OF PHARMACY"", Leningradskaya reg., Russia
| |
Collapse
|
8
|
The Combination of Physical Exercise with Muscle-Directed Antioxidants to Counteract Sarcopenia: A Biomedical Rationale for Pleiotropic Treatment with Creatine and Coenzyme Q10. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7083049. [PMID: 29123615 PMCID: PMC5632475 DOI: 10.1155/2017/7083049] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 08/13/2017] [Accepted: 08/23/2017] [Indexed: 12/21/2022]
Abstract
Sarcopenia represents an increasing public health risk due to the rapid aging of the world's population. It is characterized by both low muscle mass and function and is associated with mobility disorders, increased risk of falls and fractures, loss of independence, disabilities, and increased risk of death. Despite the urgency of the problem, the development of treatments for sarcopenia has lagged. Increased reactive oxygen species (ROS) production and decreased antioxidant (AO) defences seem to be important factors contributing to muscle impairment. Studies have been conducted to verify whether physical exercise and/or AOs could prevent and/or delay sarcopenia through a normalization of the etiologically relevant ROS imbalance. Despite the strong rationale, the results obtained were contradictory, particularly with regard to the effects of the tested AOs. A possible explanation might be that not all the agents included in the general heading of "AOs" could fulfill the requisites to counteract the complex series of events causing/accelerating sarcopenia: the combination of the muscle-directed antioxidants creatine and coenzyme Q10 with physical exercise as a biomedical rationale for pleiotropic prevention and/or treatment of sarcopenia is discussed.
Collapse
|
9
|
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
|
10
|
Rybalka E, Timpani CA, Stathis CG, Hayes A, Cooke MB. Metabogenic and Nutriceutical Approaches to Address Energy Dysregulation and Skeletal Muscle Wasting in Duchenne Muscular Dystrophy. Nutrients 2015; 7:9734-67. [PMID: 26703720 PMCID: PMC4690050 DOI: 10.3390/nu7125498] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 10/29/2015] [Accepted: 11/13/2015] [Indexed: 12/21/2022] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is a fatal genetic muscle wasting disease with no current cure. A prominent, yet poorly treated feature of dystrophic muscle is the dysregulation of energy homeostasis which may be associated with intrinsic defects in key energy systems and promote muscle wasting. As such, supplementative nutriceuticals that target and augment the bioenergetical expansion of the metabolic pathways involved in cellular energy production have been widely investigated for their therapeutic efficacy in the treatment of DMD. We describe the metabolic nuances of dystrophin-deficient skeletal muscle and review the potential of various metabogenic and nutriceutical compounds to ameliorate the pathological and clinical progression of the disease.
Collapse
Affiliation(s)
- Emma Rybalka
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne 8001, Australia.
- Institute of Sport, Exercise & Healthy Living, Victoria University, Melbourne 8001, Australia.
- Australian Institute of Musculoskeletal Science, Western Health, Melbourne 3021, Australia.
| | - Cara A Timpani
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne 8001, Australia.
- Institute of Sport, Exercise & Healthy Living, Victoria University, Melbourne 8001, Australia.
| | - Christos G Stathis
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne 8001, Australia.
- Institute of Sport, Exercise & Healthy Living, Victoria University, Melbourne 8001, Australia.
- Australian Institute of Musculoskeletal Science, Western Health, Melbourne 3021, Australia.
| | - Alan Hayes
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne 8001, Australia.
- Institute of Sport, Exercise & Healthy Living, Victoria University, Melbourne 8001, Australia.
- Australian Institute of Musculoskeletal Science, Western Health, Melbourne 3021, Australia.
| | - Matthew B Cooke
- Centre for Chronic Disease, College of Health & Biomedicine, Victoria University, Melbourne 8001, Australia.
- Institute of Sport, Exercise & Healthy Living, Victoria University, Melbourne 8001, Australia.
- Australian Institute of Musculoskeletal Science, Western Health, Melbourne 3021, Australia.
| |
Collapse
|
11
|
Coenzyme Q10 protects human endothelial cells from β-amyloid uptake and oxidative stress-induced injury. PLoS One 2014; 9:e109223. [PMID: 25272163 PMCID: PMC4182835 DOI: 10.1371/journal.pone.0109223] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 08/31/2014] [Indexed: 01/30/2023] Open
Abstract
Neuropathological symptoms of Alzheimer's disease appear in advances stages, once neuronal damage arises. Nevertheless, recent studies demonstrate that in early asymptomatic stages, ß-amyloid peptide damages the cerebral microvasculature through mechanisms that involve an increase in reactive oxygen species and calcium, which induces necrosis and apoptosis of endothelial cells, leading to cerebrovascular dysfunction. The goal of our work is to study the potential preventive effect of the lipophilic antioxidant coenzyme Q (CoQ) against ß-amyloid-induced damage on human endothelial cells. We analyzed the protective effect of CoQ against Aβ-induced injury in human umbilical vein endothelial cells (HUVECs) using fluorescence and confocal microscopy, biochemical techniques and RMN-based metabolomics. Our results show that CoQ pretreatment of HUVECs delayed Aβ incorporation into the plasma membrane and mitochondria. Moreover, CoQ reduced the influx of extracellular Ca2+, and Ca2+ release from mitochondria due to opening the mitochondrial transition pore after β-amyloid administration, in addition to decreasing O2.− and H2O2 levels. Pretreatment with CoQ also prevented ß-amyloid-induced HUVECs necrosis and apoptosis, restored their ability to proliferate, migrate and form tube-like structures in vitro, which is mirrored by a restoration of the cell metabolic profile to control levels. CoQ protected endothelial cells from Aβ-induced injury at physiological concentrations in human plasma after oral CoQ supplementation and thus could be a promising molecule to protect endothelial cells against amyloid angiopathy.
Collapse
|
12
|
Banerjee P, Sahoo A, Anand S, Ganguly A, Righi G, Bovicelli P, Saso L, Chakrabarti S. Multiple mechanisms of iron-induced amyloid beta-peptide accumulation in SHSY5Y cells: protective action of negletein. Neuromolecular Med 2014; 16:787-98. [PMID: 25249289 DOI: 10.1007/s12017-014-8328-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 09/17/2014] [Indexed: 10/24/2022]
Abstract
The increased accumulation of iron in the brain in Alzheimer's disease (AD) is well documented, and excess iron is strongly implicated in the pathogenesis of the disease. The adverse effects of accumulated iron in AD brain may include the oxidative stress, altered amyloid beta-metabolism and the augmented toxicity of metal-bound amyloid beta 42. In this study, we have shown that exogenously added iron in the form of ferric ammonium citrate (FAC) leads to considerable accumulation of amyloid precursor protein (APP) without a corresponding change in the concerned gene expression in cultured SHSY5Y cells during exposure up to 48 h. This phenomenon is also associated with increased β-secretase activity and augmented release of amyloid beta 42 in the medium. Further, the increase in β-secretase activity, in SHSY5Y cells, upon exposure to iron apparently involves reactive oxygen species (ROS) and NF-κB activation. The synthetic flavone negletein (5,6-dihydroxy-7-methoxyflavone), which is a known chelator for iron, can significantly prevent the effects of FAC on APP metabolism in SHSY5Y cells. Further, this compound inhibits the iron-dependent formation of ROS and also blocks the iron-induced oligomerization of amyloid beta 42 in vitro. In concentrations used in this study, negletein alone appears to have only marginal toxic effects on cell viability, but, on the other hand, the drug is capable of ameliorating the iron-induced loss of cell viability considerably. Our results provide the initial evidence of potential therapeutic effects of negletein, which should be explored in suitable animal models of AD.
Collapse
Affiliation(s)
- Priyanjalee Banerjee
- Department of Biochemistry, Institute of Postgraduate Medical Education and Research, 244, AJC Bose Road, Kolkata, 700020, India
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Aboul-Fotouh S. Coenzyme Q10 displays antidepressant-like activity with reduction of hippocampal oxidative/nitrosative DNA damage in chronically stressed rats. Pharmacol Biochem Behav 2013; 104:105-12. [PMID: 23313551 DOI: 10.1016/j.pbb.2012.12.027] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 12/23/2012] [Accepted: 12/26/2012] [Indexed: 11/16/2022]
Abstract
UNLABELLED Multiple evidences suggest that depression is accompanied by an induction of oxidative/nitrosative stress (O&NS) pathways and by a reduced antioxidant status. Coenzyme Q10 (CoQ10) is an essential cofactor in the mitochondrial electron transport pathway and has a powerful antioxidant capacity. METHODS This study investigated the effect of chronic treatment with CoQ10 (25, 50, 100 and 150 mg/kg/day, i.p. for 3 weeks) on depressive-like behavior and hippocampal, O&NS, and DNA damage, induced by chronic restraint stress (CRS), an experimental model of depression, in rats. RESULTS CoQ10 showed a significant antidepressant effect, as evidenced by amelioration of CRS-induced behavioral aberrations in forced swimming and open field tests, elevated corticosterone level and body weight loss. Moreover, CoQ10 dose-dependently restored the hippocampal catalase, glutathione peroxidase and reduced glutathione and decreased the hippocampal malondialdehyde, nitric oxide and 8-hydroxy-2'-deoxyguanosine levels, which indicated a potential protective effect of CoQ10 against hippocampal O&NS lipid peroxidation and DNA damage. CONCLUSION CoQ10 possesses antidepressant activity and can protect against CRS-induced hippocampal DNA damage which could be mediated in part by maintaining mitochondrial function and its well documented antioxidant properties. Therefore, CoQ10 may have a potential therapeutic value for the management of depressive disorders. However, further research, is still required to characterize the mechanism of the antidepressant effect of CoQ10 and extend these results before the safe application in humans.
Collapse
Affiliation(s)
- Sawsan Aboul-Fotouh
- Department of Pharmacology, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
14
|
Ivanov A, Gorodetskaya E, Kalenikova E, Medvedev O. Single intravenous injection of CoQ<sub>10</sub> reduces infarct size in a rat model of ischemia and reperfusion injury. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/wjcd.2013.35a001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
15
|
Chang Y, Huang SK, Wang SJ. Coenzyme Q10 inhibits the release of glutamate in rat cerebrocortical nerve terminals by suppression of voltage-dependent calcium influx and mitogen-activated protein kinase signaling pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:11909-11918. [PMID: 23167655 DOI: 10.1021/jf302875k] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
This study investigates the effects and possible mechanism of coenzyme Q10 (CoQ10) on endogenous glutamate release in the cerebral cortex nerve terminals of rats. CoQ10 inhibited the release of glutamate evoked by the K+ channel blocker 4-aminopyridine (4-AP). CoQ10 reduced the depolarization-induced increase in cytosolic [Ca2+]c but did not alter the 4-AP-mediated depolarization. The effect of CoQ10 on evoked glutamate release was abolished by blocking the Cav2.2 (N-type) and Cav2.1 (P/Q-type) Ca2+ channels and mitogen-activated protein kinase kinase (MEK). In addition, CoQ10 decreased the 4-AP-induced phosphorylation of extracellular signal-regulated kinase 1 and 2 (ERK1/2) and synaptic vesicle-associated protein synapsin I, a major presynaptic substrate for ERK. Moreover, the inhibition of glutamate release by CoQ10 was strongly attenuated in mice without synapsin I. These results suggest that CoQ10 inhibits glutamate release from cortical synaptosomes in rats through the suppression of the presynaptic voltage-dependent Ca2+ entry and ERK/synapsin I signaling pathway.
Collapse
Affiliation(s)
- Yi Chang
- School of Medicine, Fu Jen Catholic University, and Department of Anesthesiology, Far-EAstern Memorial Hospital, No. 510 Zhongzheng Road, Xinzhuang District, New Taipei City, Taiwan 24205
| | | | | |
Collapse
|
16
|
Lee HJ, Lyu DH, Koo U, Lee SJ, Hong SS, Kim K, Kim KH, Lee D, Mar W. Inhibitory effect of 2-arylbenzofurans from the Mori Cortex Radicis (Moraceae) on oxygen glucose deprivation (OGD)-induced cell death of SH-SY5Y cells. Arch Pharm Res 2011; 34:1373-80. [PMID: 21910060 DOI: 10.1007/s12272-011-0818-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 04/11/2011] [Accepted: 05/24/2011] [Indexed: 10/17/2022]
Abstract
Three known 2-arylbenzofurans, moracin P (1), moracin O (2) and mulberrofuran Q (3) were isolated from the MeOH extract of the Mori Cortex Radicis. These compounds 1-3 enhanced cell viability in dose-dependent manner against oxygen-glucose deprivation (OGD)-induced cell death in neuroblastoma SH-SY5Y cells, which was measured by MTT reduction assay. (EC(50) values of 10.4, 12.6, and 15.9 μM, respectively). In addition, the compounds 1-3 were examined for their inhibitory effect on OGD-induced ROS production by FACS analysis. We observed these compounds reduced ROS production in OGD-induced cell death (IC(50) values of 1.9, 0.3 and 12.1 μM, respectively). Consequently, reactive oxygen species (ROS) were overexpressed in OGD-induced cells and all three compounds reduced ROS induced by OGD in dosedependent manner. Taken together, compounds 1-3 might protect neuronal cell death against the oxidative stress induced by OGD, though further studies in vitro and in vivo models are necessary.
Collapse
Affiliation(s)
- Hak Ju Lee
- Korea Forest Research Institute, Seoul 130-712, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Genova ML, Lenaz G. New developments on the functions of coenzyme Q in mitochondria. Biofactors 2011; 37:330-54. [PMID: 21989973 DOI: 10.1002/biof.168] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2011] [Accepted: 04/06/2011] [Indexed: 12/12/2022]
Abstract
The notion of a mobile pool of coenzyme Q (CoQ) in the lipid bilayer has changed with the discovery of respiratory supramolecular units, in particular the supercomplex comprising complexes I and III; in this model, the electron transfer is thought to be mediated by tunneling or microdiffusion, with a clear kinetic advantage on the transfer based on random collisions. The CoQ pool, however, has a fundamental function in establishing a dissociation equilibrium with bound quinone, besides being required for electron transfer from other dehydrogenases to complex III. The mechanism of CoQ reduction by complex I is analyzed regarding recent developments on the crystallographic structure of the enzyme, also in relation to the capacity of complex I to generate superoxide. Although the mechanism of the Q-cycle is well established for complex III, involvement of CoQ in proton translocation by complex I is still debated. Some additional roles of CoQ are also examined, such as the antioxidant effect of its reduced form and the capacity to bind the permeability transition pore and the mitochondrial uncoupling proteins. Finally, a working hypothesis is advanced on the establishment of a vicious circle of oxidative stress and supercomplex disorganization in pathological states, as in neurodegeneration and cancer.
Collapse
|
18
|
Maes M, Galecki P, Chang YS, Berk M. A review on the oxidative and nitrosative stress (O&NS) pathways in major depression and their possible contribution to the (neuro)degenerative processes in that illness. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:676-92. [PMID: 20471444 DOI: 10.1016/j.pnpbp.2010.05.004] [Citation(s) in RCA: 813] [Impact Index Per Article: 58.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2010] [Revised: 04/17/2010] [Accepted: 05/03/2010] [Indexed: 02/06/2023]
Abstract
This paper reviews the body of evidence that major depression is accompanied by a decreased antioxidant status and by induction of oxidative and nitrosative (IO&NS) pathways. Major depression is characterized by significantly lower plasma concentrations of a number of key antioxidants, such as vitamin E, zinc and coenzyme Q10, and a lowered total antioxidant status. Lowered antioxidant enzyme activity, e.g. glutathione peroxidase (GPX), is another hallmark of depression. The abovementioned lowered antioxidant capacity may impair protection against reactive oxygen species (ROS), causing damage to fatty acids, proteins and DNA by oxidative and nitrosative stress (O&NS). Increased ROS in depression is demonstrated by increased levels of plasma peroxides and xanthine oxidase. Damage caused by O&NS is shown by increased levels of malondialdehyde (MDA), a by-product of polyunsaturated fatty acid peroxidation and arachidonic acid; and increased 8-hydroxy-2-deoxyguanosine, indicating oxidative DNA damage. There is also evidence in major depression, that O&NS may have changed inactive autoepitopes to neoantigens, which have acquired immunogenicity and serve as triggers to bypass immunological tolerance, causing (auto)immune responses. Thus, depression is accompanied by increased levels of plasma IgG antibodies against oxidized LDL; and increased IgM-mediated immune responses against membrane fatty acids, like phosphatidyl inositol (Pi); oleic, palmitic, and myristic acid; and NO modified amino-acids, e.g. NO-tyrosine, NO-tryptophan and NO-arginine; and NO-albumin. There is a significant association between depression and polymorphisms in O&NS genes, like manganese superoxide dismutase, catalase, and myeloperoxidase. Animal models of depression very consistently show lowered antioxidant defences and activated O&NS pathways in the peripheral blood and the brain. In animal models of depression, antidepressants consistently increase lowered antioxidant levels and normalize the damage caused by O&NS processes. Antioxidants, such as N-acetyl-cysteine, compounds that mimic GPX activity, and zinc exhibit antidepressive effects. This paper reviews the pathways by which lowered antioxidants and O&NS may contribute to depression, and the (neuro)degenerative processes that accompany that illness. It is concluded that aberrations in O&NS pathways are--together with the inflammatory processes--key components of depression. All in all, the results suggest that depression belongs to the spectrum of (neuro)degenerative disorders.
Collapse
|
19
|
Unlocking the Door to Neuronal Woes in Alzheimer's Disease: Aβ and Mitochondrial Permeability Transition Pore. Pharmaceuticals (Basel) 2010; 3:1936-1948. [PMID: 27713335 PMCID: PMC4033960 DOI: 10.3390/ph3061936] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Revised: 06/10/2010] [Accepted: 06/14/2010] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial dysfunction occurs early in the progression of Alzheimer’s disease. Amyloid-β peptide has deleterious effects on mitochondrial function and contributes to energy failure, respiratory chain impairment, neuronal apoptosis, and generation of reactive oxygen species in Alzheimer’s disease. The mechanisms underlying amyloid-β induced mitochondrial stress remain unclear. Emerging evidence indicates that mitochondrial permeability transition pore is important for maintenance of mitochondrial and neuronal function in aging and neurodegenerative disease. Cyclophilin D (Cyp D) plays a central role in opening mitochondrial permeability transition pore, ultimately leading to cell death. Interaction of amyloid-β with cyclophilin D triggers or enhances the formation of mitochondrial permeability transition pores, consequently exacerbating mitochondrial and neuronal dysfunction, as shown by decreased mitochondrial membrane potential, impaired mitochondrial respiration function, and increased oxidative stress and cytochrome c release. Blockade of cyclophilin D by genetic abrogation or pharmacologic inhibition protects mitochondria and neurons from amyloid-β induced toxicity, suggesting that cyclophilin D dependent mitochondrial transition pore is a therapeutic target for Alzheimer’s disease.
Collapse
|
20
|
Beneficial effects of a Q-ter based nutritional mixture on functional performance, mitochondrial function, and oxidative stress in rats. PLoS One 2010; 5:e10572. [PMID: 20485503 PMCID: PMC2868025 DOI: 10.1371/journal.pone.0010572] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Accepted: 04/16/2010] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Mitochondrial dysfunction and oxidative stress are central mechanisms underlying the aging process and the pathogenesis of many age-related diseases. Selected antioxidants and specific combinations of nutritional compounds could target many biochemical pathways that affect both oxidative stress and mitochondrial function and, thereby, preserve or enhance physical performance. METHODOLOGY/PRINCIPAL FINDINGS In this study, we evaluated the potential anti-aging benefits of a Q-ter based nutritional mixture (commercially known as Eufortyn) mainly containing the following compounds: terclatrated coenzyme Q(10) (Q-ter), creatine and a standardized ginseng extract. We found that Eufortyn supplementation significantly ameliorated the age-associated decreases in grip strength and gastrocnemius subsarcolemmal mitochondria Ca(2+) retention capacity when initiated in male Fischer344 x Brown Norway rats at 21 months, but not 29 months, of age. Moreover, the increases in muscle RNA oxidation and subsarcolemmal mitochondrial protein carbonyl levels, as well as the decline of total urine antioxidant power, which develop late in life, were mitigated by Eufortyn supplementation in rats at 29 months of age. CONCLUSIONS/SIGNIFICANCE These data imply that Eufortyn is efficacious in reducing oxidative damage, improving the age-related mitochondrial functional decline, and preserving physical performance when initiated in animals at early midlife (21 months). The efficacy varied, however, according to the age at which the supplementation was provided, as initiation in late middle age (29 months) was incapable of restoring grip strength and mitochondrial function. Therefore, the Eufortyn supplementation may be particularly beneficial when initiated prior to major biological and functional declines that appear to occur with advancing age.
Collapse
|
21
|
López-Lluch G, Rodríguez-Aguilera JC, Santos-Ocaña C, Navas P. Is coenzyme Q a key factor in aging? Mech Ageing Dev 2010; 131:225-35. [PMID: 20193705 DOI: 10.1016/j.mad.2010.02.003] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2009] [Revised: 01/19/2010] [Accepted: 02/20/2010] [Indexed: 01/28/2023]
Abstract
Coenzyme Q (Q) is a key component for bioenergetics and antioxidant protection in the cell. During the last years, research on diseases linked to Q-deficiency has highlighted the essential role of this lipid in cell physiology. Q levels are also affected during aging and neurodegenerative diseases. Therefore, therapies based on dietary supplementation with Q must be considered in cases of Q deficiency such as in aging. However, the low bioavailability of dietary Q for muscle and brain obligates to design new mechanisms to increase the uptake of this compound in these tissues. In the present review we show a complete picture of the different functions of Q in cell physiology and their relationship to age and age-related diseases. Furthermore, we describe the problems associated with dietary Q uptake and the mechanisms currently used to increase its uptake or even its biosynthesis in cells. Strategies to increase Q levels in tissues are indicated.
Collapse
Affiliation(s)
- Guillermo López-Lluch
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide, CIBERER-Instituto de Salud Carlos III, Carretera de Utrera, Km 1, 41013 Sevilla, Spain
| | | | | | | |
Collapse
|
22
|
Moreira PI, Zhu X, Wang X, Lee HG, Nunomura A, Petersen RB, Perry G, Smith MA. Mitochondria: a therapeutic target in neurodegeneration. Biochim Biophys Acta Mol Basis Dis 2009; 1802:212-20. [PMID: 19853657 DOI: 10.1016/j.bbadis.2009.10.007] [Citation(s) in RCA: 218] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2009] [Revised: 10/08/2009] [Accepted: 10/13/2009] [Indexed: 01/24/2023]
Abstract
Mitochondrial dysfunction has long been associated with neurodegenerative disease. Therefore, mitochondrial protective agents represent a unique direction for the development of drug candidates that can modify the pathogenesis of neurodegeneration. This review discusses evidence showing that mitochondrial dysfunction has a central role in the pathogenesis of Alzheimer's, Parkinson's and Huntington's diseases and amyotrophic lateral sclerosis. We also debate the potential therapeutic efficacy of metabolic antioxidants, mitochondria-directed antioxidants and Szeto-Schiller (SS) peptides. Since these compounds preferentially target mitochondria, a major source of oxidative damage, they are promising therapeutic candidates for neurodegenerative diseases. Furthermore, we will briefly discuss the novel action of the antihistamine drug Dimebon on mitochondria.
Collapse
Affiliation(s)
- Paula I Moreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Oral administration of coenzyme Q10 prevents cytochrome c release from mitochondria induced by 1-methyl-4-phenylpyridinium ion in mouse brain synaptosomes. Neurosci Lett 2009; 463:22-5. [DOI: 10.1016/j.neulet.2009.07.072] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2009] [Revised: 07/21/2009] [Accepted: 07/23/2009] [Indexed: 11/22/2022]
|
24
|
Abstract
A large body of evidence from postmortem brain tissue and genetic analysis in humans and biochemical and pathological studies in animal models (transgenic and toxin) of neurodegeneration suggest that mitochondrial dysfunction is a common pathological mechanism. Mitochondrial dysfunction from oxidative stress, mitochondrial DNA deletions, pathological mutations, altered mitochondrial morphology, and interaction of pathogenic proteins with mitochondria leads to neuronal demise. Therefore, therapeutic approaches targeting mitochondrial dysfunction and oxidative damage hold great promise in neurodegenerative diseases. This review discusses the potential therapeutic efficacy of creatine, coenzyme Q10, idebenone, synthetic triterpenoids, and mitochondrial targeted antioxidants (MitoQ) and peptides (SS-31) in in vitro studies and in animal models of Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Alzheimer's disease. We have also reviewed the current status of clinical trials of creatine, coenzyme Q10, idebenone, and MitoQ in neurodegenerative disorders. Further, we discuss newly identified therapeutic targets, including peroxisome proliferator-activated receptor-gamma-coactivator and sirtuins, which provide promise for future therapeutic developments in neurodegenerative disorders.
Collapse
Affiliation(s)
- Rajnish K Chaturvedi
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York, NY 10065, USA
| | | |
Collapse
|
25
|
Spindler M, Beal MF, Henchcliffe C. Coenzyme Q10 effects in neurodegenerative disease. Neuropsychiatr Dis Treat 2009; 5:597-610. [PMID: 19966907 PMCID: PMC2785862 DOI: 10.2147/ndt.s5212] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Indexed: 12/13/2022] Open
Abstract
Coenzyme Q10 (CoQ10) is an essential cofactor in the mitochondrial respiratory chain, and as a dietary supplement it has recently gained attention for its potential role in the treatment of neurodegenerative disease. Evidence for mitochondrial dysfunction in neurodegenerative disorders derives from animal models, studies of mitochondria from patients, identification of genetic defects in patients with neurodegenerative disease, and measurements of markers of oxidative stress. Studies of in vitro models of neuronal toxicity and animal models of neurodegenerative disorders have demonstrated potential neuroprotective effects of CoQ10. With this data in mind, several clinical trials of CoQ10 have been performed in Parkinson's disease and atypical Parkinson's syndromes, Huntington's disease, Alzheimer disease, Friedreich's ataxia, and amyotrophic lateral sclerosis, with equivocal findings. CoQ10 is widely available in multiple formulations and is very well tolerated with minimal adverse effects, making it an attractive potential therapy. Phase III trials of high-dose CoQ10 in large sample sizes are needed to further ascertain the effects of CoQ10 in neurodegenerative diseases.
Collapse
Affiliation(s)
- Meredith Spindler
- Department of Neurology, Weill Medical, College of Cornell University, 525 east 68th Street, Suite F610, New York, NY, USA.
| | | | | |
Collapse
|
26
|
Coenzyme Q10 attenuates beta-amyloid pathology in the aged transgenic mice with Alzheimer presenilin 1 mutation. J Mol Neurosci 2008; 34:165-71. [PMID: 18181031 DOI: 10.1007/s12031-007-9033-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Accepted: 12/11/2007] [Indexed: 10/22/2022]
Abstract
One of the neuropathological features of Alzheimer's disease (AD) is the deposition of senile plaques containing beta-amyloid (A beta). There is limited evidence for the treatment to arrest A beta pathology of AD. In our present study, we tested the effect of coenzyme Q10 (CoQ10), an endogenous antioxidant and a powerful free radical scavenger, on A beta in the aged transgenic mice overexpressing Alzheimer presenilin 1-L235P (leucine-to-proline mutation at codon 235, 16-17 months old). The treatment by feeding the transgenic mice with CoQ10 for 60 days (1,200 mg kg(-1) day(-1)) partially attenuated A beta overproduction and intracellular A beta deposit in the cortex of the transgenic mice compared with the age-matched untreated transgenic mice. Meanwhile, an increased oxidative stress reaction was detected as evidenced by elevated level of malondialdehyde (MDA) and decreased activity of superoxide dismutase (SOD) in the transgenic mice relative to the wild-type mice, and supplementation of CoQ10 partially decreased MDA level and upregulated the activity of SOD. The results indicate that oxidative stress is enhanced in the brain of the transgenic mice, that this enhancement may further promote A beta 42 overproduction in a vicious formation, and that CoQ10 would be beneficial for the therapy of AD.
Collapse
|
27
|
Li G, Jack CR, Yang XF, Yang ES. Diet supplement CoQ10 delays brain atrophy in aged transgenic mice with mutations in the amyloid precursor protein: an in vivo volume MRI study. Biofactors 2008; 32:169-78. [PMID: 19096113 DOI: 10.1002/biof.5520320120] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
We tested the hypotheses that supplemental intake of the diet supplement Coenzyme Q10 (CoQ10) could delay brain atrophy in double transgenic amyloid precursor protein (APP) / presenilin 1 (PS1), single transgenic APP and PS1 as well as wild type mice by volume MR image in vivo. One hundred and twelve mice (28 APP/PS1, 28 APP, 28 PS1 and 28 wild types) were studied. Half of each genotype group (n = 14 per group) was treated with CoQ10 2400 mg/kg/day, and the other half with placebo for 60 days. Magnetic resonance (MR) images were used to obtain the volumes of the hemispheres and hippocampi. APP / PS1, APP, PS1 and wild type mice treated with CoQ10 exhibited significantly less atrophy in hemisphere and hippocampus than those receiving placebo. The neuro-protective effect of the CoQ10 on hemispheric volume, and hippocampal volume was related to genotype; greater in APP/PS1 than APP and PS1 mice and less in wild type mice. Our result indicated that CoQ10 may have therapeutic potential in the prevention and treatment of MCI and AD.
Collapse
Affiliation(s)
- Geng Li
- Hong Kong Applied Science and Technology Research Institute Company Limited, Hong Kong, HKSAR, China.
| | | | | | | |
Collapse
|