51
|
Martinefski MR, Rodriguez MR, Buontempo F, Lucangioli SE, Bianciotti LG, Tripodi VP. Coenzyme Q 10 supplementation: A potential therapeutic option for the treatment of intrahepatic cholestasis of pregnancy. Eur J Pharmacol 2020; 882:173270. [PMID: 32534074 DOI: 10.1016/j.ejphar.2020.173270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/05/2020] [Accepted: 06/08/2020] [Indexed: 12/12/2022]
Abstract
Intrahepatic cholestasis of pregnancy (ICP) is a pregnancy specific liver disease characterized by pruritus, elevated serum bile acids and abnormal liver function that may be associated with severe adverse pregnancy outcomes. We previously reported that plasma coenzyme Q10 (CoQ10) is decreased in women with ICP as it is its analogue coenzyme Q9 (CoQ9) in rats with ethinyl estradiol (EE)-induced cholestasis. The aim of the present study was to evaluate the possible therapeutic role of CoQ10 in experimental hepatocellular cholestasis and to compare it with ursodeoxycholic acid (UDCA) supplementation. Bile acids, CoQ9, CoQ10, transaminases, alkaline phosphatase, retinol, α-tocopherol, ascorbic acid, thiobarbituric acid reactive substances, carbonyls, glutathione, superoxide dismutase and catalase were assessed in plasma, liver and/or hepatic mitochondria in control and cholestatic rats supplemented with CoQ10 (250 mg/kg) administered alone or combined with UDCA (25 mg/kg). CoQ10 supplementation prevented bile flow decline (P < 0.05) and the increase in serum alkaline phosphatase and bile acids, particularly lithocholic acid (P < 0.05) in cholestatic rats. Furthermore, it also improved oxidative stress parameters in the liver, increased both CoQ10 and CoQ9 plasma levels and partially prevented the fall in α-tocopherol (P < 0.05). UDCA also prevented cholestasis, but it was less efficient than CoQ10 to improve the liver redox environment. Combined administration of CoQ10 and UDCA resulted in additive effects. In conclusion, present findings show that CoQ10 supplementation attenuated EE-induced cholestasis by promoting a favorable redox environment in the liver, and further suggest that it may represent an alternative therapeutic option for ICP.
Collapse
Affiliation(s)
- Manuela R Martinefski
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Buenos Aires, Argentina
| | - Myrian R Rodriguez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, INIGEM, UBA-CONICET, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Argentina
| | - Fabián Buontempo
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Buenos Aires, Argentina
| | - Silvia E Lucangioli
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Argentina
| | - Liliana G Bianciotti
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, INIGEM, UBA-CONICET, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Argentina.
| | - Valeria P Tripodi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Tecnología Farmacéutica, Buenos Aires, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas, CONICET, Argentina.
| |
Collapse
|
52
|
Kuşaslan R, Ercan G, Ağcaoğlu O, Altınay S, Binboğa S, Altınel Y. A novel coenzyme-Q approach for the prevention of postsurgical adhesion. Turk J Surg 2020; 36:202-208. [PMID: 33015565 DOI: 10.5578/turkjsurg.4398] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Accepted: 12/30/2019] [Indexed: 11/15/2022]
Abstract
Objectives Postoperative intraperitoneal adhesions are an unsolved and important problem in abdominal surgery. In the present study, the probable preventive role of coenzyme-Q in the development of peritoneal adhesions was investigated. Material and Methods Sixteen Wistar Hannover male rats weighing 300-350 g were randomly separated into two groups of 8 rats each. The cecum was abraded with a sterile gauze until sub-serosal hemorrhage developed. A patch of peritoneum located opposite to the cecal abrasion was completely dissected. No treatment was given to Group 1. Group 2 received 30 mg/kg coenzyme-Q, which was injected 2 mL intraperitoneally. All the rats were sacrificed on the postoperative 21st day, and after adhesions were scored macroscopically, tissue specimens of the peritoneum and bowel were subjected to histopathological investigation. Tissue and blood specimens were also taken for biochemical analysis to investigate antioxidant efficiency. Results Adhesion scores were significantly different between the control group and the coenzyme-Q group (p= 0.001). According to the tissue levels of GSH-Px, MDA, and SOD levels, there was no significant difference between the study groups (p= 0.074, p= 0.208, p= 0.526). According to the plasma GSH-Px and SOD levels, there was significant difference between the groups (p= 0.002, p= 0.001), but the difference was not significant at MDA levels (p= 0.793). The differences between the pathological scores of the control and coenzyme-Q (p= 0.028 for fibrosis; p= 0.025 for inflammation) groups were statistically significant. Conclusion This study confirms that coenzyme-Q is the potential application in the prevention of early postoperative adhesions.
Collapse
Affiliation(s)
- Ramazan Kuşaslan
- Department of General Surgery, Bagcilar Training and Research Hospital, Istanbul, Turkey
| | - Gülçin Ercan
- Department of General Surgery, Bagcilar Training and Research Hospital, Istanbul, Turkey
| | - Orhan Ağcaoğlu
- Department of General Surgery, Koc University School of Medicine Hospital, Istanbul, Turkey
| | - Serdar Altınay
- Department of Pathology, Bagcilar Training and Research Hospital, Istanbul, Turkey
| | - Sinan Binboğa
- Department of General Surgery, Bagcilar Training and Research Hospital, Istanbul, Turkey
| | - Yüksel Altınel
- Department of General Surgery, Bagcilar Training and Research Hospital, Istanbul, Turkey
| |
Collapse
|
53
|
Ehrenhaus Masotta N, Höcht C, Contin M, Lucangioli S, Rojas AM, Tripodi VP. Bioavailability of coenzyme Q10 loaded in an oleogel formulation for oral therapy: Comparison with a commercial-grade solid formulation. Int J Pharm 2020; 582:119315. [DOI: 10.1016/j.ijpharm.2020.119315] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 03/30/2020] [Accepted: 04/06/2020] [Indexed: 12/01/2022]
|
54
|
|
55
|
Ibrahim Fouad G. Synergistic anti-atherosclerotic role of combined treatment of omega-3 and co-enzyme Q10 in hypercholesterolemia-induced obese rats. Heliyon 2020; 6:e03659. [PMID: 32258512 PMCID: PMC7118318 DOI: 10.1016/j.heliyon.2020.e03659] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Revised: 12/16/2019] [Accepted: 03/20/2020] [Indexed: 01/13/2023] Open
Abstract
Hypercholesterolemia is a metabolic disorder associated with atherosclerosis. This study aimed to investigate the effects of omega-3 and/or coenzyme Q10 (CoQ10) on hypercholesterolemia-induced atherosclerosis. Rats were divided into five groups; (1): served as the negative control, (2): served as hypercholesterolemic (HC) control, (3): HC-rats administrated omega-3 orally, (4): HC-rats administrated CoQ10 orally, and (5): HC-rats administered the combination treatment of both omega-3 and CoQ10. Lipid profile was assayed and cardiovascular risk indices were calculated. Serum levels of Adiponectin (APN) and creatine kinase (CK-MB) were determined using ELISA. Besides, oxidative stress markers, malondialdehyde (MDA), nitric oxide (NO) and glutathione (GSH) were assayed in the heart homogenate. Histopathological investigation of the aortae and heart tissues were investigated. The results revealed that atherogenic HC-rats demonstrated a significant elevation in lipid profiles, except for HDL-C, along with decreased levels of APN, but increased CK-MB activities. Hypercholesterolemia increased lipid peroxidation, reduced NO production, and decreased GSH content in the cardiac tissue. Treatment of atherogenic HC-rats with omega-3 and/or CoQ10 improved dyslipidemia and ameliorated most of the HC-induced biochemical and histopathological changes. The histological observations of aortae and cardiac tissues validated our biochemical results. We concluded that the combined treatment of nutraceuticals such as omega-3 and CoQ10 demonstrated the best outcome, demonstrating their anti-hyperlipidemic, cardioprotective, and atheroprotective potentials. Together, this study supports a beneficial role of dietary co-administration of omega-3 and CoQ10 in obese patients who are prone to develop cardiovascular disorders.
Collapse
Affiliation(s)
- Ghadha Ibrahim Fouad
- Department of Therapeutic Chemistry, National Research Centre, 33 El-Bohouth Street, Dokki, Cairo, 12622, Egypt
| |
Collapse
|
56
|
Comparative Bioavailability of Different Coenzyme Q10 Formulations in Healthy Elderly Individuals. Nutrients 2020; 12:nu12030784. [PMID: 32188111 PMCID: PMC7146408 DOI: 10.3390/nu12030784] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/05/2020] [Accepted: 03/11/2020] [Indexed: 12/19/2022] Open
Abstract
Coenzyme Q10 (CoQ10) plays a central role in mitochondrial oxidative phosphorylation. Several studies have shown the beneficial effects of dietary CoQ10 supplementation, particularly in relation to cardiovascular health. CoQ10 biosynthesis decreases in the elderly, and consequently, the beneficial effects of dietary supplementation in this population are of greater significance. However, most pharmacokinetic studies have been conducted on younger populations. The aim of this study was to investigate the single-dose bioavailability of different formulations of CoQ10 in a healthy geriatric population. A randomized, three-period, crossover bioavailability study was conducted on 21 healthy older adults (aged 65–74). The treatment was a single dose with a one-week washout period. Three different formulations containing the equivalent of 100 mg of CoQ10 were used: Q10Vital® water-soluble CoQ10 syrup (the investigational product—IP); ubiquinol capsules (the comparative product—CP); and ubiquinone capsules (the standard product—SP). Ubiquinone/ubiquinol was followed in the plasma for 48 h. An analysis of the ratio of the area under the baseline-corrected concentration curve (ΔAUC48) for total CoQ10 and a comparison to SP yielded the following: The bioavailability of CoQ10 in the IP was 2.4-fold higher (95% CI: 1.3–4.5; p = 0.002), while the bioavailability of ubiquinol (CP) was not significantly increased (1.7-fold; 95% CI: 0.9–3.1, p = 0.129). No differences in the redox status of the absorbed coenzyme Q10 were observed between formulations, showing that CoQ10 appeared in the blood mostly as ubiquinol, even if consumed as ubiquinone.
Collapse
|
57
|
Hargreaves IP, Mantle D. Supplementation with selenium and coenzyme Q10 in critically ill patients. Br J Hosp Med (Lond) 2020; 80:589-593. [PMID: 31589506 DOI: 10.12968/hmed.2019.80.10.589] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Multiple organ dysfunction and resultant mortality in critically ill patients has been linked with impaired cellular energy supply and oxidative stress. Clinical studies supplementing selenium, on the basis of its role as a key cofactor of antioxidant enzymes, have reported variable outcomes in critically ill patients. However, the synergistic interaction between selenium and coenzyme Q10, which has essential roles in cellular energy supply and as an antioxidant, has not been considered in such studies. This article reviews the link between selenium and coenzyme Q10, and the potential role of their co-supplementation in critical illness.
Collapse
Affiliation(s)
- I P Hargreaves
- Senior Lecturer, School of Pharmacy, Liverpool John Moores University, Liverpool L3 3AF
| | - D Mantle
- Consultant, Pharma Nord (UK) Ltd, Morpeth, Newcastle
| |
Collapse
|
58
|
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: 10.3] [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
|
59
|
Coenzyme Q10 and cerebral malaria in mice: Questionable interpretations, improbable usefulness in humans. Parasitol Int 2020; 74:101969. [DOI: 10.1016/j.parint.2019.101969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Accepted: 08/03/2019] [Indexed: 11/19/2022]
|
60
|
Pagano G, Pallardó FV, Porto B, Fittipaldi MR, Lyakhovich A, Trifuoggi M. Mitoprotective Clinical Strategies in Type 2 Diabetes and Fanconi Anemia Patients: Suggestions for Clinical Management of Mitochondrial Dysfunction. Antioxidants (Basel) 2020; 9:antiox9010082. [PMID: 31963742 PMCID: PMC7023409 DOI: 10.3390/antiox9010082] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/11/2022] Open
Abstract
Oxidative stress (OS) and mitochondrial dysfunction (MDF) occur in a number of disorders, and several clinical studies have attempted to counteract OS and MDF by providing adjuvant treatments against disease progression. The present review is aimed at focusing on two apparently distant diseases, namely type 2 diabetes (T2D) and a rare genetic disease, Fanconi anemia (FA). The pathogenetic links between T2D and FA include the high T2D prevalence among FA patients and the recognized evidence for OS and MDF in both disorders. This latter phenotypic/pathogenetic feature-namely MDF-may be regarded as a mechanistic ground both accounting for the clinical outcomes in both diseases, and as a premise to clinical studies aimed at counteracting MDF. In the case for T2D, the working hypothesis is raised of evaluating any in vivo decrease of mitochondrial cofactors, or mitochondrial nutrients (MNs) such as α-lipoic acid, coenzyme Q10, and l-carnitine, with possibly combined MN-based treatments. As for FA, the established knowledge of MDF, as yet only obtained from in vitro or molecular studies, prompts the requirement to ascertain in vivo MDF, and to design clinical studies aimed at utilizing MNs toward mitigating or delaying FA's clinical progression. Altogether, this paper may contribute to building hypotheses for clinical studies in a number of OS/MDF-related diseases.
Collapse
Affiliation(s)
- Giovanni Pagano
- Department of Chemical Sciences, Federico II Naples University, I-80126 Naples, Italy;
- Correspondence: ; Tel.: +39-335-790-7261
| | - Federico V. Pallardó
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-INCLIVA, CIBERER, E-46010 Valencia, Spain;
| | - Beatriz Porto
- Institute of Biomedical Sciences, ICBAS, University of Porto, 4099-030 Porto, Portugal;
| | - Maria Rosa Fittipaldi
- Internal Medicine Unit, San Francesco d’Assisi Hospital, I-84020 Oliveto Citra (SA), Italy;
| | - Alex Lyakhovich
- Vall d’Hebron Institut de Recerca, E-08035 Barcelona, Spain;
- Institute of Molecular Biology and Biophysics of the “Federal Research Center of Fundamental and Translational Medicine”, 630117 Novosibirsk, Russia
| | - Marco Trifuoggi
- Department of Chemical Sciences, Federico II Naples University, I-80126 Naples, Italy;
| |
Collapse
|
61
|
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: 0.3] [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
|
62
|
Suárez-Rivero JM, Pastor-Maldonado CJ, de la Mata M, Villanueva-Paz M, Povea-Cabello S, Álvarez-Córdoba M, Villalón-García I, Suárez-Carrillo A, Talaverón-Rey M, Munuera M, Sánchez-Alcázar JA. Atherosclerosis and Coenzyme Q 10. Int J Mol Sci 2019; 20:ijms20205195. [PMID: 31635164 PMCID: PMC6834161 DOI: 10.3390/ijms20205195] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 10/12/2019] [Accepted: 10/16/2019] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis is the most common cause of cardiac deaths worldwide. Classically, atherosclerosis has been explained as a simple arterial lipid deposition with concomitant loss of vascular elasticity. Eventually, this condition can lead to consequent blood flow reduction through the affected vessel. However, numerous studies have demonstrated that more factors than lipid accumulation are involved in arterial damage at the cellular level, such as inflammation, autophagy impairment, mitochondrial dysfunction, and/or free-radical overproduction. In order to consider the correction of all of these pathological changes, new approaches in atherosclerosis treatment are necessary. Ubiquinone or coenzyme Q10 is a multifunctional molecule that could theoretically revert most of the cellular alterations found in atherosclerosis, such as cholesterol biosynthesis dysregulation, impaired autophagy flux and mitochondrial dysfunction thanks to its redox and signaling properties. In this review, we will show the latest advances in the knowledge of the relationships between coenzyme Q10 and atherosclerosis. In addition, as atherosclerosis phenotype is closely related to aging, it is reasonable to believe that coenzyme Q10 supplementation could be beneficial for both conditions.
Collapse
Affiliation(s)
- Juan M Suárez-Rivero
- 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, 41013 Sevilla, Spain.
| | - Carmen J Pastor-Maldonado
- 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, 41013 Sevilla, Spain.
| | - Mario de la Mata
- 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, 41013 Sevilla, Spain.
| | - Marina Villanueva-Paz
- 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, 41013 Sevilla, Spain.
| | - Suleva Povea-Cabello
- 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, 41013 Sevilla, Spain.
| | - Mónica Álvarez-Córdoba
- 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, 41013 Sevilla, Spain.
| | - Irene Villalón-García
- 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, 41013 Sevilla, Spain.
| | - Alejandra Suárez-Carrillo
- 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, 41013 Sevilla, Spain.
| | - Marta Talaverón-Rey
- 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, 41013 Sevilla, Spain.
| | - Manuel Munuera
- 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, 41013 Sevilla, Spain.
| | - 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, 41013 Sevilla, Spain.
| |
Collapse
|
63
|
Ya F, Xu XR, Shi Y, Gallant RC, Song F, Zuo X, Zhao Y, Tian Z, Zhang C, Xu X, Ling W, Ni H, Yang Y. Coenzyme Q10 Upregulates Platelet cAMP/PKA Pathway and Attenuates Integrin αIIbβ3 Signaling and Thrombus Growth. Mol Nutr Food Res 2019; 63:e1900662. [PMID: 31512815 DOI: 10.1002/mnfr.201900662] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 08/22/2019] [Indexed: 12/11/2022]
Abstract
SCOPE Platelet integrin αIIbβ3 is the key mediator of atherothrombosis. Supplementation of coenzyme Q10 (CoQ10), a fat-soluble molecule that exists in various foods, exerts protective cardiovascular effects. This study aims to investigate whether and how CoQ10 acts on αIIbβ3 signaling and thrombosis, the major cause of cardiovascular diseases. METHODS AND RESULTS Using a series of platelet functional assays in vitro, it is demonstrated that CoQ10 reduces human platelet aggregation, granule secretion, platelet spreading, and clot retraction. It is further demonstrated that CoQ10 inhibits platelet integrin αIIbβ3 outside-in signaling. These inhibitory effects are mainly mediated by upregulating cAMP/PKA pathway, where CoQ10 stimulates the A2A adenosine receptor and decreases phosphodiesterase 3A phosphorylation. Moreover, CoQ10 attenuates murine thrombus growth and vessel occlusion in a ferric chloride (FeCl3 )-induced thrombosis model in vivo. Importantly, the randomized, double-blind, placebo-controlled clinical trial in dyslipidemic patients demonstrates that 24 weeks of CoQ10 supplementation increases platelet CoQ10 concentrations, enhances the cAMP/PKA pathway, and attenuates αIIbβ3 outside-in signaling, leading to decreased platelet aggregation and granule release. CONCLUSION Through upregulating the platelet cAMP/PKA pathway, and attenuating αIIbβ3 signaling and thrombus growth, CoQ10 supplementation may play an important protective role in patients with risks of cardiovascular diseases.
Collapse
Affiliation(s)
- Fuli Ya
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, 510080, China.,Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province, 510080, China.,Guangdong Province Engineering Laboratory for Nutrition Translation, Guangzhou, Guangdong Province, 510080, China
| | - Xiaohong Ruby Xu
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
| | - Yilin Shi
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, 510080, China.,Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province, 510080, China.,Guangdong Province Engineering Laboratory for Nutrition Translation, Guangzhou, Guangdong Province, 510080, China
| | - Reid C Gallant
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada
| | - Fenglin Song
- School of Food Science, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, 510006, China
| | - Xiao Zuo
- Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province, 510080, China.,Guangdong Province Engineering Laboratory for Nutrition Translation, Guangzhou, Guangdong Province, 510080, China.,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province, 510006, China
| | - Yimin Zhao
- Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province, 510080, China.,Guangdong Province Engineering Laboratory for Nutrition Translation, Guangzhou, Guangdong Province, 510080, China.,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province, 510006, China
| | - Zezhong Tian
- Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province, 510080, China.,Guangdong Province Engineering Laboratory for Nutrition Translation, Guangzhou, Guangdong Province, 510080, China.,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province, 510006, China
| | - Cheng Zhang
- Department of Clinical Laboratory, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong Province, 510120, China
| | - Xiping Xu
- National Clinical Research Center for Kidney Disease, Renal Division, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, 510515, China
| | - Wenhua Ling
- Department of Nutrition, School of Public Health, Sun Yat-sen University (Northern Campus), Guangzhou, Guangdong Province, 510080, China.,Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province, 510080, China.,Guangdong Province Engineering Laboratory for Nutrition Translation, Guangzhou, Guangdong Province, 510080, China
| | - Heyu Ni
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, Toronto, Ontario, M5B 1W8, Canada.,Canadian Blood Services Centre for Innovation, Toronto, Ontario, M5G 2M1, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, M5S 1A1, Canada.,Department of Medicine, University of Toronto, Toronto, Ontario, M5S 1A1, Canada.,Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A1, Canada
| | - Yan Yang
- Guangdong Provincial Key Laboratory for Food, Nutrition and Health, Guangzhou, Guangdong Province, 510080, China.,Guangdong Province Engineering Laboratory for Nutrition Translation, Guangzhou, Guangdong Province, 510080, China.,School of Public Health (Shenzhen), Sun Yat-sen University, Guangzhou, Guangdong Province, 510006, China
| |
Collapse
|
64
|
Deng Y, Chen X, Wang L, Peng X, Lin M. Characterization of unknown impurities in Coenzyme Q10 using LC–MS and NMR. J Pharm Biomed Anal 2019; 175:112771. [PMID: 31330281 DOI: 10.1016/j.jpba.2019.07.019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2019] [Revised: 07/08/2019] [Accepted: 07/09/2019] [Indexed: 01/01/2023]
|
65
|
Omidi G, Karimi SA, Shahidi S, Faraji N, Komaki A. Coenzyme Q10 supplementation reverses diabetes-related impairments in long-term potentiation induction in hippocampal dentate gyrus granular cells: An in vivo study. Brain Res 2019; 1726:146475. [PMID: 31560865 DOI: 10.1016/j.brainres.2019.146475] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 09/18/2019] [Accepted: 09/23/2019] [Indexed: 01/20/2023]
Abstract
Diabetes mellitus (DM) is associated with impaired hippocampal synaptic plasticity. Coenzyme Q10 (CoQ10) acts as an antioxidant and exerts neuroprotective effects. Accordingly, this study aimed at evaluating the effects of CoQ10 on hippocampal long-term potentiation (LTP) and paired-pulse facilitation (PPF) in streptozotocin (STZ)-induced diabetic rats. Male Wistar rats were randomly divided into six groups (n = 8 per group) as follows and treated for 90 days: the control, control + low dose of CoQ10 (100 mg/kg), control + high dose of CoQ10 (600 mg/kg), diabetic, diabetic + low dose of CoQ10, and diabetic + high dose of CoQ10 groups. Diabetes was induced by a single intraperitoneal injection of 50 mg/kg STZ. The population spike (PS) amplitude and slope of excitatory post synaptic potentials (EPSPs) were measured in dentate gyrus (DG) area in response to the stimulation applied to the perforant path (PP). The results showed that the STZ-induced diabetes impaired LTP induction in the PP-DG synapses. This finding is supported by the decreased EPSP slope and PS amplitude of LTP (P < 0.05). Both low- and high-dose CoQ10 supplementation in the control and diabetic animals enhanced EPSP slope and PS amplitude of LTP in the granular cells of DG (P < 0.05). PPF was affected by LTP induction in diabetic animals receiving the high dose of CoQ10 (P < 0.05). It is suggested that CoQ10 administration could attenuate deteriorative effect of STZ-induced diabetes on in vivo LTP in the DG. The enhanced transmitter release can be partly one of the possible underlying mechanism(s) responsible for the LTP induction in the diabetic animals treated with CoQ10.
Collapse
Affiliation(s)
- Ghazaleh Omidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Asaad Karimi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Nafiseh Faraji
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran. https://umsha.ac.ir
| |
Collapse
|
66
|
Díaz-Casado ME, Quiles JL, Barriocanal-Casado E, González-García P, Battino M, López LC, Varela-López A. The Paradox of Coenzyme Q 10 in Aging. Nutrients 2019; 11:nu11092221. [PMID: 31540029 PMCID: PMC6770889 DOI: 10.3390/nu11092221] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/06/2019] [Accepted: 09/08/2019] [Indexed: 12/14/2022] Open
Abstract
Coenzyme Q (CoQ) is an essential endogenously synthesized molecule that links different metabolic pathways to mitochondrial energy production thanks to its location in the mitochondrial inner membrane and its redox capacity, which also provide it with the capability to work as an antioxidant. Although defects in CoQ biosynthesis in human and mouse models cause CoQ deficiency syndrome, some animals models with particular defects in the CoQ biosynthetic pathway have shown an increase in life span, a fact that has been attributed to the concept of mitohormesis. Paradoxically, CoQ levels decline in some tissues in human and rodents during aging and coenzyme Q10 (CoQ10) supplementation has shown benefits as an anti-aging agent, especially under certain conditions associated with increased oxidative stress. Also, CoQ10 has shown therapeutic benefits in aging-related disorders, particularly in cardiovascular and metabolic diseases. Thus, we discuss the paradox of health benefits due to a defect in the CoQ biosynthetic pathway or exogenous supplementation of CoQ10.
Collapse
Affiliation(s)
- M Elena Díaz-Casado
- Institute of Biotechnology, Department of Physiology, Biomedical Research Center, University of Granada, Avda del Conocimiento sn, 18016 Granada, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 18016 Granada, Spain.
| | - José L Quiles
- Institute of Nutrition and Food Technology "José Mataix Verdú", Department of Physiology, Biomedical Research Center, University of Granada, Avda del Conocimiento sn, 18016 Granada, Spain.
| | - Eliana Barriocanal-Casado
- Institute of Biotechnology, Department of Physiology, Biomedical Research Center, University of Granada, Avda del Conocimiento sn, 18016 Granada, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 18016 Granada, Spain.
| | - Pilar González-García
- Institute of Biotechnology, Department of Physiology, Biomedical Research Center, University of Granada, Avda del Conocimiento sn, 18016 Granada, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 18016 Granada, Spain.
| | - Maurizio Battino
- Department of Clinical Sicences, Università Politecnica delle Marche, 60131 Ancona, Italy.
- Nutrition and Food Science Group, Department of Analytical and Food Chemistry, CITACA, CACTI, University of Vigo, 36310 Vigo, Spain.
- International Research Center for Food Nutrition and Safety, Jiangsu University, Zhenjiang 212013, China.
| | - Luis C López
- Institute of Biotechnology, Department of Physiology, Biomedical Research Center, University of Granada, Avda del Conocimiento sn, 18016 Granada, Spain.
- Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable (CIBERFES), 18016 Granada, Spain.
| | - Alfonso Varela-López
- Institute of Nutrition and Food Technology "José Mataix Verdú", Department of Physiology, Biomedical Research Center, University of Granada, Avda del Conocimiento sn, 18016 Granada, Spain.
| |
Collapse
|
67
|
Soleimani M, Golab F, Alizadeh A, Rigi S, Samani ZN, Vahabzadeh G, Peirovi T, Sarbishegi M, Katebi M, Azedi F. Evaluation of the neuroprotective effects of electromagnetic fields and coenzyme Q 10 on hippocampal injury in mouse. J Cell Physiol 2019; 234:18720-18730. [PMID: 30932191 DOI: 10.1002/jcp.28512] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/23/2018] [Accepted: 01/10/2019] [Indexed: 11/10/2022]
Abstract
Electromagnetic fields (EMFs) are reported to interfere with chemical reactions involving free radical production. Coenzyme Q10 (CoQ10) is a strong antioxidant with some neuroprotective activities. The purpose of this study was to examine and compare the neuroprotective effects of EMF and CoQ10 in a mouse model of hippocampal injury. Hippocampal injury was induced in mature female mice (25-30 g), using an intraperitoneal injection of trimethyltin hydroxide (TMT; 2.5 mg/kg). The experimental groups were exposed to EMF at a frequency of 50 Hz and intensity of 5.9 mT for 7 hr daily over 1 week or treated with CoQ10 (10 mg/kg) for 2 weeks following TMT injection. A Morris water maze apparatus was used to assess learning and spatial memory. Nissl staining and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) tests were also performed for the histopathological analysis of the hippocampus. Antiapoptotic genes were studied, using the Western blot technique. The water maze test showed memory improvement following treatment with CoQ10 and coadministration of CoQ10 + EMF. The Nissl staining and TUNEL tests indicated a decline in necrotic and apoptotic cell count following treatment with CoQ10 and coadministration of CoQ10 + EMF. The Western blot study indicated the upregulation of antiapoptotic genes in treatment with CoQ10, as well as coadministration. Also, treatment with EMF had no significant effects on reducing damage induced by TMT in the hippocampus. According to the results, EMF had no significant neuroprotective effects in comparison with CoQ10 on hippocampal injury in mice. Nevertheless, coadministration of EMF and CoQ10 could improve the neuroprotective effects of CoQ10.
Collapse
Affiliation(s)
- Mansoureh Soleimani
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Razi Drug Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Anatomical Sciences, Iran University of Medical Sciences, Tehran, Iran
| | - Fereshteh Golab
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Akram Alizadeh
- Department of Tissue Engineering, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sara Rigi
- Department of Anatomy, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Zeinab Nazarian Samani
- Department of Tissue Engineering, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Gelareh Vahabzadeh
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Peirovi
- Department of Anatomical Sciences, Urumia University of Medical Sciences, Urumia, Iran
| | - Maryam Sarbishegi
- Department of Anatomical Sciences, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Majid Katebi
- Department of Anatomical Sciences, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Fereshteh Azedi
- Department of Neuroscience, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
68
|
Omidi G, Karimi SA, Rezvani-Kamran A, Monsef A, Shahidi S, Komaki A. Effect of coenzyme Q10 supplementation on diabetes induced memory deficits in rats. Metab Brain Dis 2019; 34:833-840. [PMID: 30848472 DOI: 10.1007/s11011-019-00402-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 02/25/2019] [Indexed: 12/19/2022]
Abstract
The main objective of current work was to determine the effects of low and high dose supplementation with coenzyme Q10 (CoQ10) on spatial learning and memory in rats with streptozotocin (STZ)-induced diabetes. Male Wistar rats (weighing 220 ± 10) were randomly divided into six groups: (i) Control (Con, n = 8); (ii) Control+ Low dose of CoQ10 (100 mg/kg) (CLD, n = 10); (iii) Control+ high dose of CoQ10 (600 mg/kg) (CHD, n = 10); (iv) Diabetic (D, n = 10); (v) Diabetic + Low dose of CoQ10 (100 mg/kg) (DLD, n = 10); (vi) Diabetic + high dose of CoQ10 (600 mg/kg) (DHD, n = 10). Diabetes was induced by a single intraperitoneal injection of 50 mg/kg STZ. CoQ10 was administered intragastrically by gavage once a day for 90 days. After 90 days, Morris water maze (MWM) task was used to evaluate the spatial learning and memory in rats. Diabetic animals showed a slower rate of acquisition with respect to the control animals [F (1, 51) = 92.81, P < 0.0001, two-way ANOVA]. High dose (but no low dose) supplementation with CoQ10 could attenuate deteriorative effect of diabetes on memory acquisition. Diabetic animals which received CoQ10 (600 mg/kg) show a considerable decrease in escape latency and traveled distance compared to diabetic animals (p < 0.05, two-way ANOVA,). The present study has shown that low dose supplementation with CoQ10 in diabetic rats failed to improve deficits in cognitive function but high dose supplementation with CoQ10 reversed diabetes-related declines in spatial learning.
Collapse
Affiliation(s)
- Ghazaleh Omidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Asaad Karimi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Neuroscience, School of Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Arezoo Rezvani-Kamran
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Amirreza Monsef
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Siamak Shahidi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
- Department of Neuroscience, School of Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Neuroscience, School of Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Shahid Fahmideh Street, Hamadan, 65178/518, Iran.
| |
Collapse
|
69
|
Saboori S, Rad EY, Mardani M, Khosroshahi MZ, Nouri Y, Falahi E. Effect of Q10 supplementation on body weight and body mass index: A systematic review and meta-analysis of randomized controlled clinical trials. Diabetes Metab Syndr 2019; 13:1179-1185. [PMID: 31336462 DOI: 10.1016/j.dsx.2019.01.047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/23/2019] [Indexed: 01/05/2023]
Abstract
AIMS This meta-analysis study was carried out to assess the effects of coenzyme Q10 supplementation on body weight and body mass index of patients in randomized controlled clinical trial studies. MATERIALS AND METHODS A comprehensive systematic search of literature was performed through ISI web of sciences, PubMed, Scopus and Cochrane library databases up to February 2018 which was supplemented by manual search of the references list of included studies. From a total of 1579 identified articles, only 17 trials with 14 and 14 effect-sizes were included for pooling the effects of co-enzyme Q10 supplementation on body weight and body mass index, respectively. RESULTS Results of random-effect size meta-analysis showed that supplementation with coenzyme Q10 had no significant decreasing effects on body weight (WMD: 0.28 kg; 95% CI = -0.91, 1.47; P = 0.64) and BMI (WMD: -0.03; 95% CI = -0.4, 0.34; P = 0.86) of study participants. Subgroup analysis revealed that dosage of Q10 and trial duration could not differ the results of Q10 supplementation. CONCLUSION Results of this meta-analysis study failed to show any beneficial effect of coenzyme Q10 supplementation on body weight and BMI of patients in clinical trial studies.
Collapse
Affiliation(s)
- Somayeh Saboori
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Esmaeil Yousefi Rad
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mahnaz Mardani
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | | | - Yasaman Nouri
- Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Ebrahim Falahi
- Nutritional Health Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran.
| |
Collapse
|
70
|
Andalib S, Mashhadi-Mousapour M, Bijani S, Hosseini MJ. Coenzyme Q 10 Alleviated Behavioral Dysfunction and Bioenergetic Function in an Animal Model of Depression. Neurochem Res 2019; 44:1182-1191. [PMID: 30820817 DOI: 10.1007/s11064-019-02761-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 02/21/2019] [Accepted: 02/22/2019] [Indexed: 10/27/2022]
Abstract
Coenzyme Q10 (CoQ10) is a natural compound, is involved in the mitochondrial electron transfer chain (ETC) and plays an important pattern in adenosine triphosphate (ATP) production. Amelioration of ATP is related to abnormalities in cognitive function and psychiatric diseases. Previous studies have shown that depression is accompanied by the induction of inflammatory and oxidative stress pathways and amelioration of antioxidant status. In a recent study, we investigated the beneficial effects of CoQ10 on behavioral dysfunction and CoQ10 level in the rat brain. Therefore, intracerebroventricular (ICV) infusion of a single dose of streptozotocin (STZ, 0.2 mg/mouse) was used in adult male mice to induce depression. The behavioral data revealed a significant difference between the depression and control groups regarding the forced swim test (FST) and splash test results at 24 h following STZ treatment. Also, the validated and accurate high-performance liquid chromatography (HPLC) technique showed decreased CoQ10 level in the brain samples of the STZ group, compared to the controls. Our findings revealed that behavioral abnormalities due to STZ target mitochondria and affect energy metabolism and hemostasis, resulting in the initiation of oxidative damage in the brain. Besides, 4-week administration of CoQ10 could reverse the depressive like behavior and bioenergetic effects of STZ in the treated groups.
Collapse
Affiliation(s)
- Sina Andalib
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical sciences, Zanjan, Iran.,Departments of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, P. O. Box: 45139-56184, Iran
| | - Mobin Mashhadi-Mousapour
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical sciences, Zanjan, Iran.,Departments of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, P. O. Box: 45139-56184, Iran
| | - Soroush Bijani
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical sciences, Zanjan, Iran.,Departments of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, P. O. Box: 45139-56184, Iran
| | - Mir-Jamal Hosseini
- Zanjan Applied Pharmacology Research Center, Zanjan University of Medical sciences, Zanjan, Iran. .,Departments of Pharmacology and Toxicology, School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, P. O. Box: 45139-56184, Iran.
| |
Collapse
|
71
|
Shimizu Y, Mukumoto N, Idrus N, Akasaka H, Inubushi S, Yoshida K, Miyawaki D, Ishihara T, Okamoto Y, Yasuda T, Nakahana M, Sasaki R. Amelioration of Radiation Enteropathy by Dietary Supplementation With Reduced Coenzyme Q10. Adv Radiat Oncol 2019; 4:237-245. [PMID: 31011668 PMCID: PMC6460318 DOI: 10.1016/j.adro.2019.01.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2018] [Accepted: 01/23/2019] [Indexed: 01/08/2023] Open
Abstract
Purpose Effective methods to ameliorate radiation enteropathy have not been developed. To address this issue, we investigated the reduced form of coenzyme Q10 (rCoQ10) as a potential radioprotector in a mouse model. Methods and Materials rCoQ10 was added to a standard laboratory mouse diet at a final concentration of 1.0% 9 days before irradiation and 30 days thereafter or dissolved in corn oil and administered transorally. Accumulated amounts of coenzyme Q10 (CoQ10) or coenzyme Q9 in the intestine were measured by high-performance liquid chromatography. Reactive oxygen species (ROS), apoptosis, and morphologic changes in the intestine were assessed by immunohistochemistry after administration of 13 Gy of x-ray to the mouse abdomen. Body weight and survival were monitored for 30 days after irradiation. Cytotoxicity using 3 human cancer cell lines and the tumor growth–inhibiting effect in a xenograft were investigated to determine whether rCoQ10 interferes with radiation-specific cytotoxic effects on tumor growth. Results CoQ10 was greatly accumulated in all sections of the intestine after both massive transoral dosing and dietary administration, whereas coenzyme Q9 was not. Administration of rCoQ10 suppressed ROS production and inhibited apoptosis in the crypts, resulting in preservation of villi structures after irradiation. Notably, 92% of mice fed the rCoQ10-supplemented diet were healthy and alive 30 days after irradiation, whereas 50% of control mice died (P < .05). Moreover, rCoQ10 did not interfere with radiation-specific cytotoxic effects on tumors either in vitro or in vivo. Conclusions Administration of rCoQ10 led to its accumulation in the intestine and induced radioprotective effects by inhibiting ROS-mediated apoptosis, thereby preserving intestinal structures. Our results indicated that rCoQ10 supplementation effectively ameliorated radiation enteropathy.
Collapse
Affiliation(s)
- Yasuyuki Shimizu
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Naritoshi Mukumoto
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Nelly Idrus
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan.,Siloam Hospitals TB, Simatupang, Jakarta, Indonesia
| | - Hiroaki Akasaka
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Sachiko Inubushi
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Kenji Yoshida
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Daisuke Miyawaki
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Takeaki Ishihara
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Yoshiaki Okamoto
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan.,Department of Radiation Therapy, Osaka Police Hospital, Osaka, Japan
| | - Takahiro Yasuda
- Clinical & Translational Research Center, Kobe University Hospital, Hyogo, Japan
| | - Makiko Nakahana
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan
| | - Ryohei Sasaki
- Division of Radiation Oncology, Kobe University Graduate School of Medicine, Hyogo, Japan
| |
Collapse
|
72
|
Hargreaves IP, Mantle D. Coenzyme Q10 Supplementation in Fibrosis and Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1178:103-112. [DOI: 10.1007/978-3-030-25650-0_6] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
73
|
Affiliation(s)
| | - Iain P Hargreaves
- Senior Lecturer, School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University
| |
Collapse
|
74
|
Kuhlman AB, Morville T, Dohlmann TL, Hansen M, Kelly B, Helge JW, Dela F. Coenzyme Q10 does not improve peripheral insulin sensitivity in statin-treated men and women: the LIFESTAT study. Appl Physiol Nutr Metab 2018; 44:485-492. [PMID: 30273493 DOI: 10.1139/apnm-2018-0488] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Simvastatin is a cholesterol-lowering drug that is prescribed to lower the risk of cardiovascular disease following high levels of blood cholesterol. There is a possible risk of new-onset diabetes mellitus with statin treatment but the mechanisms behind are unknown. Coenzyme Q10 (CoQ10) supplementation has been found to improve glucose homeostasis in various patient populations and may increase muscle glucose transporter type 4 content. Our aim was to investigate if 8 weeks of CoQ10 supplementation can improve glucose homeostasis in simvastatin-treated subjects. Thirty-five men and women in treatment with a minimum of 40 mg of simvastatin daily were randomized to receive either 2 × 200 mg/day of CoQ10 supplementation or placebo for 8 weeks. Glucose homeostasis was investigated with fasting blood samples, oral glucose tolerance test (OGTT) and intravenous glucose tolerance test. Insulin sensitivity was assessed with the hyperinsulinemic-euglycemic clamp. Different indices were calculated from fasting samples and OGTT as secondary measures of insulin sensitivity. A muscle biopsy was obtained from the vastus lateralis muscle for muscle protein analyzes. There were no changes in body composition, fasting plasma insulin, fasting plasma glucose, or 3-h glucose with intervention, but glycated hemoglobin decreased with time. Glucose homeostasis measured as the area under the curve for glucose, insulin, and C-peptide during OGTT was unchanged after intervention. Insulin secretory capacity was also unaltered after CoQ10 supplementation. Insulin sensitivity was unchanged but hepatic insulin sensitivity increased. No changes in muscle GLUT4 content was observed after intervention. CoQ10 supplementation does not change muscle GLUT4 content, insulin sensitivity, or secretory capacity, but hepatic insulin sensitivity may improve.
Collapse
Affiliation(s)
- Anja Birk Kuhlman
- a Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Thomas Morville
- a Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Tine Lovsø Dohlmann
- a Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Maria Hansen
- a Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Bo Kelly
- a Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Jørn Wulff Helge
- a Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Flemming Dela
- a Xlab, Center for Healthy Aging, Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark.,b Department of Geriatrics, Bispebjerg University Hospital, 2400 Copenhagen, Denmark
| |
Collapse
|
75
|
Casagrande D, Waib PH, Jordão Júnior AA. Mechanisms of action and effects of the administration of Coenzyme Q10 on metabolic syndrome. JOURNAL OF NUTRITION & INTERMEDIARY METABOLISM 2018. [DOI: 10.1016/j.jnim.2018.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
|
76
|
Elbaky NAA, El-Orabi NF, Fadda LM, Abd-Elkader OH, Ali HM. Role of N-Acetylcysteine and Coenzyme Q10 in the Amelioration of Myocardial Energy Expenditure and Oxidative Stress, Induced by Carbon Tetrachloride Intoxication in Rats. Dose Response 2018; 16:1559325818790158. [PMID: 30116167 PMCID: PMC6088489 DOI: 10.1177/1559325818790158] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 06/13/2018] [Accepted: 06/19/2018] [Indexed: 12/20/2022] Open
Abstract
This study is designed to evaluate the potential impact of N-acetyl cysteine (NAC) and coenzyme Q10 (CoQ10) each alone or in combination against carbon tetrachloride (CCl4)-induced cardiac damage in rats. Animals were treated with CCl4 in single intraperitoneal dose of 1 mL/Kg body weight; CCl4-intoxicated animals were pretreated with 20 mg/kg/d NAC or pretreated with 200 mg/kg/d CoQ10 or NAC and CoQ10 with the same previously mentioned doses. Carbon tetrachloride-intoxicated rats showed a significant elevation in nitric oxide and lipid peroxides and downregulation in reduced glutathione level and calcium adenosine triphosphatase. Cardiac glycolytic enzymes levels such as lactate dehydrogenase, phosphofructokinase, and hexokinase were declined coupled with a reduction in glucose content after CCl4 treatment. Moreover, myocardial hydroxyproline level was significantly increased after CCl4-treatment indicating accumulation of interstitial collagen. N-acetyl cysteine and/or CoQ10 effectively alleviated the disturbances in myocardial oxidative stress and antioxidant markers. These antioxidants effectively upregulated the reduction in cardiac energetic biomarkers due to CCl4 treatment. N-acetyl cysteine and/or CoQ10 significantly decreased hydroxyproline level compared to that of CCl4-treated rats. The current data showed that the aforementioned antioxidants have a remarkable cardioprotective effect, suggesting that they may be useful as prophylactic agents against the detrimental effects of cardiotoxins.
Collapse
Affiliation(s)
- Nayira A. Abd Elbaky
- Department of Pharmacology, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology, Al-Azhar University, Cairo, Egypt
| | - Naglaa F. El-Orabi
- Department of Pharmacology, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, Suez Canal University, Ismailia, Egypt
| | - Laila M. Fadda
- Department of Pharmacology, King Saud University, Riyadh, Saudi Arabia
| | - Omar H. Abd-Elkader
- Department of Zoology, King Saud University, Riyadh, Saudi Arabia
- Electron Microscope and Thin Films Department, National Research Center, Cairo, Egypt
| | - Hanaa M. Ali
- Department of Genetics and Cytology, National Research Center, Cairo, Egypt
- King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
77
|
Pierce JD, Mahoney DE, Hiebert JB, Thimmesch AR, Diaz FJ, Smith C, Shen Q, Mudaranthakam DP, Clancy RL. Study protocol, randomized controlled trial: reducing symptom burden in patients with heart failure with preserved ejection fraction using ubiquinol and/or D-ribose. BMC Cardiovasc Disord 2018; 18:57. [PMID: 29606104 PMCID: PMC5879598 DOI: 10.1186/s12872-018-0796-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/20/2018] [Indexed: 12/15/2022] Open
Abstract
Background Heart failure (HF), the leading cause of morbidity and mortality in the US, affects 6.6 million adults with an estimated additional 3 million people by 2030. More than 50% of HF patients have heart failure with preserved left ventricular ejection fraction (HFpEF). These patients have impaired cardiac muscle relaxation and diastolic filling, which investigators have associated with cellular energetic impairment. Patients with HFpEF experience symptoms of: (1) fatigue; (2) shortness of breath; and (3) swelling (edema) of the lower extremities. However, current HF guidelines offer no effective treatment to address these underlying pathophysiologic mechanisms. Thus, we propose a biobehavioral symptom science study using ubiquinol and D-ribose (therapeutic interventions) to target mitochondrial bioenergetics to reduce the complex symptoms experienced by patients with HFpEF. Methods Using a randomized, double-blind, placebo-controlled design, the overall objective is to determine if administering ubiquinol and/or D-ribose to HFpEF patients for 12 weeks would decrease the severity of their complex symptoms and improve their cardiac function. The measures used to assess patients’ perceptions of their health status and level of vigor (energy) will be the Kansas City Cardiomyopathy Questionnaire (KCCQ) and Vigor subscale of the Profile of Mood States. The 6-min walk test will be used to test exercise tolerance. Left ventricular diastolic function will be assessed using innovative advanced echocardiography software called speckle tracking. We will measure B-type natriuretic peptides (secreted from ventricles in HF) and lactate/ATP ratio (measure of cellular energetics). Discussions Ubiquinol (active form of Coenzyme Q10) and D-ribose are two potential treatments that can positively affect cellular energetic impairment, the major underlying mechanism of HFpEF. Ubiquinol, the reduced form of CoQ10, is more effective in adults over the age of 50. In patients with HFpEF, mitochondrial deficiency of ubiquinol results in decreased adenosine triphosphate (ATP) synthesis and reduced scavenging of reactive oxygen species. D-ribose is a substrate required for ATP synthesis and when administered has been shown to improve impaired myocardial bioenergetics. Therefore, if the biological underpinning of deficient mitochondrial ATP in HFpEF is not addressed, patients will suffer major symptoms including lack of energy, fatigue, exertional dyspnea, and exercise intolerance. Trial registration ClinicalTrials.gov Identifier: NCT03133793; Data of Registration: April 28, 2017.
Collapse
Affiliation(s)
- Janet D Pierce
- School of Nursing, University of Kansas, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.,Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Diane E Mahoney
- School of Nursing, University of Kansas, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - John B Hiebert
- School of Nursing, University of Kansas, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Amanda R Thimmesch
- School of Nursing, University of Kansas, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA.
| | - Francisco J Diaz
- Department of Biostatistics, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Carol Smith
- School of Nursing, University of Kansas, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Qiuhua Shen
- School of Nursing, University of Kansas, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Dinesh Pal Mudaranthakam
- Department of Biostatistics, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| | - Richard L Clancy
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City, KS, 66160, USA
| |
Collapse
|
78
|
Huang H, Chi H, Liao D, Zou Y. Effects of coenzyme Q 10 on cardiovascular and metabolic biomarkers in overweight and obese patients with type 2 diabetes mellitus: a pooled analysis. Diabetes Metab Syndr Obes 2018; 11:875-886. [PMID: 30568475 PMCID: PMC6276825 DOI: 10.2147/dmso.s184301] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
BACKGROUND The potential effects of coenzyme Q10 (CoQ10) supplementation in overweight/obese patients with type 2 diabetes mellitus are not fully established. In this article, we aimed to perform a pooled analysis to investigate the effects of CoQ10 intervention on cardiovascular disease (CVD) risk factors in overweight/obese patients with type 2 diabetes mellitus (T2DM). METHODS MEDLINE, Embase, and Cochrane databases were searched for randomized controlled trials that evaluated the changes in CVD risk factors among overweight and obese patients with T2DM following CoQ10 supplementation. Two investigators independently assessed articles for inclusion, extracted data, and assessed risk of bias. Major endpoints were synthesized as weighted mean differences (WMDs) with 95% CIs. Subgroup analyses were performed to check the consistency of effect sizes across groups. Publication bias and sensitivity analysis were also performed. RESULTS Fourteen eligible trials with 693 overweight/obese diabetic subjects were included for pooling. CoQ10 interventions significantly reduced fasting blood glucose (FBG; -0.59 mmol/L; 95% CI, -1.05 to -0.12; P=0.01), hemoglobin A1c (HbA1c; -0.28%; 95% CI-0.53 to -0.03; P=0.03), and triglyceride (TG) levels (0.17 mmol/L; 95% CI, -0.32 to -0.03; P=0.02). Subgroup analysis also showed that low-dose consumption of CoQ10 (<200 mg/d) effectively reduces the values of FBG, HbA1c, fasting blood insulin, homeostatic model assessment of insulin resistance, and TG. CoQ10 treatment was well tolerated, and no drug-related adverse reactions were reported. CONCLUSION Our findings provide substantial evidence that daily CoQ10 supplementation has beneficial effects on glucose control and lipid management in overweight and obese patients with T2DM.
Collapse
Affiliation(s)
- Haohai Huang
- Department of Clinical Pharmacy, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China
| | - Honggang Chi
- Department of Traditional Chinese Medicine, Scientific Research Platform, The Second Clinical Medical College, Guangdong Medical University, Dongguan, China,
| | - Dan Liao
- Department of Gynaecology & Obstetrics, Dongguan Third People's Hospital, Affiliated Dongguan Shilong People's Hospital of Southern Medical University, Dongguan, Guangdong, China,
| | - Ying Zou
- Department of Traditional Chinese Medicine, Scientific Research Platform, The Second Clinical Medical College, Guangdong Medical University, Dongguan, China,
- Key Laboratory for Medical Molecular Diagnostics of Guangdong Province, Guangdong Medical University, Dongguan, Guangdong, China,
| |
Collapse
|
79
|
Ozer EK, Goktas MT, Kilinc I, Pehlivan S, Bariskaner H, Ugurluoglu C, Iskit AB. Coenzyme Q10 improves the survival, mesenteric perfusion, organs and vessel functions in septic rats. Biomed Pharmacother 2017; 91:912-919. [DOI: 10.1016/j.biopha.2017.05.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 05/02/2017] [Accepted: 05/04/2017] [Indexed: 01/16/2023] Open
|
80
|
Stepanović-Petrović R, Micov A, Tomić M, Pecikoza U. Levetiracetam synergizes with gabapentin, pregabalin, duloxetine and selected antioxidants in a mouse diabetic painful neuropathy model. Psychopharmacology (Berl) 2017; 234:1781-1794. [PMID: 28332005 DOI: 10.1007/s00213-017-4583-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Accepted: 02/25/2017] [Indexed: 12/29/2022]
Abstract
RATIONALE We have reported that levetiracetam, a novel anticonvulsant with analgesic properties, synergizes with ibuprofen/aspirin/paracetamol in a model of diabetic painful neuropathy (DPN). Most guidelines recommend gabapentin, pregabalin, and duloxetine as first- or second-line agents for DPN. OBJECTIVE We examined the effects of combination treatment of first-/second-line analgesics with levetiracetam in a model of DPN. Additionally, the levetiracetam's combinations with antioxidants, low dose of aspirin, coenzyme Q10, or α-lipoic acid were evaluated. METHODS Diabetes was induced in C57BL/6 mice with a single high dose of streptozotocin. The antinociceptive effects of orally administered levetiracetam, gabapentin, pregabalin, duloxetine (acute treatment) and aspirin, coenzyme Q10, and α-lipoic acid (preventive 7-day treatment), as well as combinations of levetiracetam with individual drugs were examined in the tail-flick test. In combination experiments, the drugs were coadministered in fixed-dose fractions of single-drug ED50; the type of interaction was determined by isobolographic analysis. RESULTS About 60-, 32-, 30-, 26-, 18-, and 6-fold reductions of doses of both drugs in levetiracetam combinations with pregabalin, gabapentin, coenzyme Q10, aspirin, duloxetine, and α-lipoic acid, respectively, were detected. CONCLUSIONS Combinations of levetiracetam with gabapentin/pregabalin/duloxetine that target different mechanisms/sites of action involved in DPN, as well as combinations of levetiracetam and low-dose aspirin/coenzyme Q10/α-lipoic acid that target underlying causes of DPN, produce marked synergistic interactions in reducing nociception in diabetic mice. This suggests that these combination treatments might be of great benefit for diabetic patients and should be explored further in clinical trials.
Collapse
Affiliation(s)
- Radica Stepanović-Petrović
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe, 450, POB 146, Belgrade, 11221, Serbia.
| | - Ana Micov
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe, 450, POB 146, Belgrade, 11221, Serbia
| | - Maja Tomić
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe, 450, POB 146, Belgrade, 11221, Serbia
| | - Uroš Pecikoza
- Department of Pharmacology, Faculty of Pharmacy, University of Belgrade, Vojvode Stepe, 450, POB 146, Belgrade, 11221, Serbia
| |
Collapse
|
81
|
Ki Y, Kim W, Kim YH, Kim D, Bae JS, Park D, Jeon H, Lee JH, Lee J, Nam J. Effect of Coenzyme Q10 on Radiation Nephropathy in Rats. J Korean Med Sci 2017; 32:757-763. [PMID: 28378548 PMCID: PMC5383607 DOI: 10.3346/jkms.2017.32.5.757] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Accepted: 02/05/2017] [Indexed: 11/20/2022] Open
Abstract
The kidney is one of the most radiosensitive organs in the abdominal cavity and is the dose-limiting structure in cancer patients receiving abdominal or total body irradiation. In the present study, the effect of coenzyme Q10 (CoQ10) on radiation nephropathy was evaluated in rats. A total of 72 rats were equally randomized into 4 groups: Control, CoQ10, irradiation with 10 Gy (RT) + placebo, or RT + CoQ10. The 2 RT groups received single 10 Gy of abdominal irradiation. The 2 CoQ10 groups were supplemented daily with 1 mL of soybean oil containing 10 mg/kg of CoQ10. The RT + placebo and control groups received same dose of soybean oil. After 24 weeks, laboratory and histopathologic findings were compared. The 2 RT groups showed significant increases in blood urea nitrogen (BUN) and creatinine levels and significant pathologic changes such as glomerulosclerosis and tubulointerstitial fibrosis. CoQ10 supplementation resulted in significant reductions of BUN and creatinine levels compared with the RT + placebo group (P < 0.001 and P = 0.038, respectively). CoQ10 treatment significantly attenuated glomerular and tubular changes of irradiated kidney in semiquantitative analysis (P < 0.001 for both). Administration of CoQ10 can alleviate the radiation-induced nephropathy.
Collapse
Affiliation(s)
- Yongkan Ki
- Department of Radiation Oncology, Biomedical Research Institute, Pusan National University Hospital and Pusan National University School of Medicine, Busan, Korea
| | - Wontaek Kim
- Department of Radiation Oncology, Biomedical Research Institute, Pusan National University Hospital and Pusan National University School of Medicine, Busan, Korea.
| | - Yong Ho Kim
- Department of Radiation Oncology, Biomedical Research Institute, Pusan National University Hospital and Pusan National University School of Medicine, Busan, Korea
| | - Donghyun Kim
- Department of Radiation Oncology, Biomedical Research Institute, Pusan National University Hospital and Pusan National University School of Medicine, Busan, Korea
| | - Jin Sook Bae
- Department of Radiation Oncology, Biomedical Research Institute, Pusan National University Hospital and Pusan National University School of Medicine, Busan, Korea
| | - Dahl Park
- Department of Radiation Oncology, Biomedical Research Institute, Pusan National University Hospital and Pusan National University School of Medicine, Busan, Korea
| | - Hosang Jeon
- Department of Radiation Oncology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Ju Hye Lee
- Department of Radiation Oncology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Jayoung Lee
- Department of Radiation Oncology, Pusan National University Yangsan Hospital, Yangsan, Korea
| | - Jiho Nam
- Department of Radiation Oncology, Pusan National University Yangsan Hospital, Yangsan, Korea
| |
Collapse
|
82
|
Cicero AFG, Colletti A, Fogacci F, Bove M, Rosticci M, Borghi C. Effects of a Combined Nutraceutical on Lipid Pattern, Glucose Metabolism and Inflammatory Parameters in Moderately Hypercholesterolemic Subjects: A Double-blind, Cross-over, Randomized Clinical Trial. High Blood Press Cardiovasc Prev 2017; 24:13-18. [PMID: 27339779 PMCID: PMC5315731 DOI: 10.1007/s40292-016-0163-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Accepted: 06/07/2016] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND There is an increasing interest for combined nutraceuticals that can act on several points of lipid and glucose metabolism with preventive purposes. However, the simple assemblage of nutraceuticals with potentially additive mechanism of action need to be clinically tested. METHODS To assess the effects of a combination of nutraceuticals based on artichoke, red yeast rice, banaba, and coenzyme Q10, we performed a double bind, cross-over designed trial versus placebo in 30 adults with LDL cholesterol suboptimal in primary prevention of cardiovascular disease. After a period of 3 weeks of dietary habits correction, patients began a period of 6 weeks of treatment with nutraceutical or placebo, followed by 2 weeks of washout and finally 6 weeks in cross-over. Data related to lipid pattern, insulin resistance, renal function, liver and CPK have been obtained at each visit. RESULTS In particular, the after the nutraceutical treatment the enrolled patients experienced a significant improvement in total cholesterol (-13.6 %), LDL-C (-18.2 %), non-HDL-C (-15 %), glutamic oxaloacetic transaminase (-10 %), glutamate-pyruvate transaminase (-30.9 %), and hs-CRP (-18.2 %) versus placebo. No changes have been observed in the other investigated parameters in both groups. CONCLUSIONS The tested combination of nutraceuticals has shown clinical efficacy in the reduction of total cholesterol, non-HDL, LDL and triglycerides, while improving the level of liver transaminases and high sensitivity C-reactive protein. Further confirmation are needed to verify these observations on the middle and long term with a larger number of subjects.
Collapse
Affiliation(s)
- Arrigo Francesco Giuseppe Cicero
- Department of Medicine and Surgery Sciences, University of Bologna, Bologna, Italy.
- Hypertension Research Center, Poliambulatorio Pad. 2, Via Albertoni, 15, 40138, Bologna, Italy.
| | - Alessandro Colletti
- Department of Medicine and Surgery Sciences, University of Bologna, Bologna, Italy
| | - Federica Fogacci
- Department of Medicine and Surgery Sciences, University of Bologna, Bologna, Italy
| | - Marilisa Bove
- Department of Medicine and Surgery Sciences, University of Bologna, Bologna, Italy
| | - Martina Rosticci
- Department of Medicine and Surgery Sciences, University of Bologna, Bologna, Italy
| | - Claudio Borghi
- Department of Medicine and Surgery Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
83
|
Yamada Y, Burger L, Kawamura E, Harashima H. Packaging of the Coenzyme Q 10 into a Liposome for Mitochondrial Delivery and the Intracellular Observation in Patient Derived Mitochondrial Disease Cells. Biol Pharm Bull 2017; 40:2183-2190. [DOI: 10.1248/bpb.b17-00609] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yuma Yamada
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Laila Burger
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Eriko Kawamura
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| | - Hideyoshi Harashima
- Laboratory for Molecular Design of Pharmaceutics, Faculty of Pharmaceutical Sciences, Hokkaido University
| |
Collapse
|
84
|
Lopez Sanchez M, Crowston J, Mackey D, Trounce I. Emerging Mitochondrial Therapeutic Targets in Optic Neuropathies. Pharmacol Ther 2016; 165:132-52. [DOI: 10.1016/j.pharmthera.2016.06.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Indexed: 12/14/2022]
|
85
|
Sharma A, Fonarow GC, Butler J, Ezekowitz JA, Felker GM. Coenzyme Q10 and Heart Failure: A State-of-the-Art Review. Circ Heart Fail 2016; 9:e002639. [PMID: 27012265 DOI: 10.1161/circheartfailure.115.002639] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/22/2015] [Indexed: 11/16/2022]
Abstract
Heart failure (HF) with either preserved or reduced ejection fraction is associated with increased morbidity and mortality. Evidence-based therapies are often limited by tolerability, hypotension, electrolyte disturbances, and renal dysfunction. Coenzyme Q10 (CoQ10) may represent a safe therapeutic option for patients with HF. CoQ10 is a highly lipophilic molecule with a chemical structure similar to vitamin K. Although being a common component of cellular membranes, CoQ10's most prominent role is to facilitate the production of adenosine triphosphate in the mitochondria by participating in redox reactions within the electron transport chain. Numerous trials during the past 30 years examining CoQ10 in patients with HF have been limited by small numbers and lack of contemporary HF therapies. The recent publication of the Q-SYMBIO randomized controlled trial demonstrated a reduction in major adverse cardiovascular events with CoQ10 supplementation in a contemporary HF population. Although having limitations, this study has renewed interest in evaluating CoQ10 supplementation in patients with HF. Current literature suggests that CoQ10 is relatively safe with few drug interactions and side effects. Furthermore, it is already widely available as an over-the-counter supplement. These findings warrant future adequately powered randomized controlled trials of CoQ10 supplementation in patients with HF. This state-of-the-art review summarizes the literature about the mechanisms, clinical data, and safety profile of CoQ10 supplementation in patients with HF.
Collapse
Affiliation(s)
- Abhinav Sharma
- From the Division of Cardiology, Duke University School of Medicine, Duke Heart Center, Durham, NC (A.S., G.M.F.); Division of Cardiology, Canadian VIGOUR Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton (A.S., J.A.E.); Ahmanson-UCLA Cardiomyopathy Center, Ronald Reagan UCLA Medical Center (G.C.F.); and Department of Medicine, Division of Cardiology, Stony Brook University, NY (J.B.)
| | - Gregg C Fonarow
- From the Division of Cardiology, Duke University School of Medicine, Duke Heart Center, Durham, NC (A.S., G.M.F.); Division of Cardiology, Canadian VIGOUR Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton (A.S., J.A.E.); Ahmanson-UCLA Cardiomyopathy Center, Ronald Reagan UCLA Medical Center (G.C.F.); and Department of Medicine, Division of Cardiology, Stony Brook University, NY (J.B.)
| | - Javed Butler
- From the Division of Cardiology, Duke University School of Medicine, Duke Heart Center, Durham, NC (A.S., G.M.F.); Division of Cardiology, Canadian VIGOUR Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton (A.S., J.A.E.); Ahmanson-UCLA Cardiomyopathy Center, Ronald Reagan UCLA Medical Center (G.C.F.); and Department of Medicine, Division of Cardiology, Stony Brook University, NY (J.B.)
| | - Justin A Ezekowitz
- From the Division of Cardiology, Duke University School of Medicine, Duke Heart Center, Durham, NC (A.S., G.M.F.); Division of Cardiology, Canadian VIGOUR Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton (A.S., J.A.E.); Ahmanson-UCLA Cardiomyopathy Center, Ronald Reagan UCLA Medical Center (G.C.F.); and Department of Medicine, Division of Cardiology, Stony Brook University, NY (J.B.)
| | - G Michael Felker
- From the Division of Cardiology, Duke University School of Medicine, Duke Heart Center, Durham, NC (A.S., G.M.F.); Division of Cardiology, Canadian VIGOUR Centre, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton (A.S., J.A.E.); Ahmanson-UCLA Cardiomyopathy Center, Ronald Reagan UCLA Medical Center (G.C.F.); and Department of Medicine, Division of Cardiology, Stony Brook University, NY (J.B.).
| |
Collapse
|
86
|
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: 51] [Impact Index Per Article: 6.4] [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
|
87
|
Tarry-Adkins JL, Fernandez-Twinn DS, Hargreaves IP, Neergheen V, Aiken CE, Martin-Gronert MS, McConnell JM, Ozanne SE. Coenzyme Q10 prevents hepatic fibrosis, inflammation, and oxidative stress in a male rat model of poor maternal nutrition and accelerated postnatal growth. Am J Clin Nutr 2016; 103:579-88. [PMID: 26718412 PMCID: PMC4733260 DOI: 10.3945/ajcn.115.119834] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Accepted: 11/11/2015] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND It is well established that low birth weight and accelerated postnatal growth increase the risk of liver dysfunction in later life. However, molecular mechanisms underlying such developmental programming are not well characterized, and potential intervention strategies are poorly defined. OBJECTIVES We tested the hypotheses that poor maternal nutrition and accelerated postnatal growth would lead to increased hepatic fibrosis (a pathological marker of liver dysfunction) and that postnatal supplementation with the antioxidant coenzyme Q10 (CoQ10) would prevent this programmed phenotype. DESIGN A rat model of maternal protein restriction was used to generate low-birth-weight offspring that underwent accelerated postnatal growth (termed "recuperated"). These were compared with control rats. Offspring were weaned onto standard feed pellets with or without dietary CoQ10 (1 mg/kg body weight per day) supplementation. At 12 mo, hepatic fibrosis, indexes of inflammation, oxidative stress, and insulin signaling were measured by histology, Western blot, ELISA, and reverse transcriptase-polymerase chain reaction. RESULTS Hepatic collagen deposition (diameter of deposit) was greater in recuperated offspring (mean ± SEM: 12 ± 2 μm) than in controls (5 ± 0.5 μm) (P < 0.001). This was associated with greater inflammation (interleukin 6: 38% ± 24% increase; P < 0.05; tumor necrosis factor α: 64% ± 24% increase; P < 0.05), lipid peroxidation (4-hydroxynonenal, measured by ELISA: 0.30 ± 0.02 compared with 0.19 ± 0.05 μg/mL per μg protein; P < 0.05), and hyperinsulinemia (P < 0.05). CoQ10 supplementation increased (P < 0.01) hepatic CoQ10 concentrations and ameliorated liver fibrosis (P < 0.001), inflammation (P < 0.001), some measures of oxidative stress (P < 0.001), and hyperinsulinemia (P < 0.01). CONCLUSIONS Suboptimal in utero nutrition combined with accelerated postnatal catch-up growth caused more hepatic fibrosis in adulthood, which was associated with higher indexes of oxidative stress and inflammation and hyperinsulinemia. CoQ10 supplementation prevented liver fibrosis accompanied by downregulation of oxidative stress, inflammation, and hyperinsulinemia.
Collapse
Affiliation(s)
- Jane L Tarry-Adkins
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, United Kingdom; and
| | - Denise S Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, United Kingdom; and
| | - Iain P Hargreaves
- Neurometabolic Unit, National Hospital, University College London, London, United Kingdom
| | - Viruna Neergheen
- Neurometabolic Unit, National Hospital, University College London, London, United Kingdom
| | - Catherine E Aiken
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, United Kingdom; and
| | - Malgorzata S Martin-Gronert
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, United Kingdom; and
| | - Josie M McConnell
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, United Kingdom; and
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge, United Kingdom; and
| |
Collapse
|
88
|
Tarry-Adkins JL, Fernandez-Twinn DS, Madsen R, Chen JH, Carpenter A, Hargreaves IP, McConnell JM, Ozanne SE. Coenzyme Q10 Prevents Insulin Signaling Dysregulation and Inflammation Prior to Development of Insulin Resistance in Male Offspring of a Rat Model of Poor Maternal Nutrition and Accelerated Postnatal Growth. Endocrinology 2015; 156. [PMID: 26214037 PMCID: PMC4869840 DOI: 10.1210/en.2015-1424] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Low birth weight and rapid postnatal growth increases the risk of developing insulin resistance and type 2 diabetes in later life. However, underlying mechanisms and potential intervention strategies are poorly defined. Here we demonstrate that male Wistar rats exposed to a low-protein diet in utero that had a low birth weight but then underwent postnatal catch-up growth (recuperated offspring) had reductions in the insulin signaling proteins p110-β (13% ± 6% of controls [P < .001]) and insulin receptor substrate-1 (39% ± 10% of controls [P < .05]) in adipose tissue. These changes were not accompanied by any change in expression of the corresponding mRNAs, suggesting posttranscriptional regulation. Recuperated animals displayed evidence of a proinflammatory phenotype of their adipose tissue with increased IL-6 (139% ± 8% [P < .05]) and IL1-β (154% ± 16% [P < .05]) that may contribute to the insulin signaling protein dysregulation. Postweaning dietary supplementation of recuperated animals with coenzyme Q (CoQ10) (1 mg/kg of body weight per day) prevented the programmed reduction in insulin receptor substrate-1 and p110-β and the programmed increased in IL-6. These findings suggest that postweaning CoQ10 supplementation has antiinflammatory properties and can prevent programmed changes in insulin-signaling protein expression. We conclude that CoQ10 supplementation represents an attractive intervention strategy to prevent the development of insulin resistance that results from suboptimal in utero nutrition.
Collapse
Affiliation(s)
- Jane L Tarry-Adkins
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit (J.L.T.-A., D.S.F.-T., R.M., J.-H.C., A.C., J.M.M., S.E.O.), Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge CB2 OQQ, United Kingdom; and Neurometabolic Unit (I.P.H.), National Hospital, University College London, London WC1N 3BG, United Kingdom
| | - Denise S Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit (J.L.T.-A., D.S.F.-T., R.M., J.-H.C., A.C., J.M.M., S.E.O.), Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge CB2 OQQ, United Kingdom; and Neurometabolic Unit (I.P.H.), National Hospital, University College London, London WC1N 3BG, United Kingdom
| | - Ralitsa Madsen
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit (J.L.T.-A., D.S.F.-T., R.M., J.-H.C., A.C., J.M.M., S.E.O.), Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge CB2 OQQ, United Kingdom; and Neurometabolic Unit (I.P.H.), National Hospital, University College London, London WC1N 3BG, United Kingdom
| | - Jian-Hua Chen
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit (J.L.T.-A., D.S.F.-T., R.M., J.-H.C., A.C., J.M.M., S.E.O.), Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge CB2 OQQ, United Kingdom; and Neurometabolic Unit (I.P.H.), National Hospital, University College London, London WC1N 3BG, United Kingdom
| | - Asha Carpenter
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit (J.L.T.-A., D.S.F.-T., R.M., J.-H.C., A.C., J.M.M., S.E.O.), Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge CB2 OQQ, United Kingdom; and Neurometabolic Unit (I.P.H.), National Hospital, University College London, London WC1N 3BG, United Kingdom
| | - Iain P Hargreaves
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit (J.L.T.-A., D.S.F.-T., R.M., J.-H.C., A.C., J.M.M., S.E.O.), Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge CB2 OQQ, United Kingdom; and Neurometabolic Unit (I.P.H.), National Hospital, University College London, London WC1N 3BG, United Kingdom
| | - Josie M McConnell
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit (J.L.T.-A., D.S.F.-T., R.M., J.-H.C., A.C., J.M.M., S.E.O.), Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge CB2 OQQ, United Kingdom; and Neurometabolic Unit (I.P.H.), National Hospital, University College London, London WC1N 3BG, United Kingdom
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and Medical Research Council Metabolic Diseases Unit (J.L.T.-A., D.S.F.-T., R.M., J.-H.C., A.C., J.M.M., S.E.O.), Wellcome Trust-Medical Research Council Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Cambridge CB2 OQQ, United Kingdom; and Neurometabolic Unit (I.P.H.), National Hospital, University College London, London WC1N 3BG, United Kingdom
| |
Collapse
|
89
|
Peel MM, Cooke M, Lewis-Peel HJ, Lea RA, Moyle W. A randomized controlled trial of coenzyme Q10 for fatigue in the late-onset sequelae of poliomyelitis. Complement Ther Med 2015; 23:789-93. [PMID: 26645517 DOI: 10.1016/j.ctim.2015.09.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 07/15/2015] [Accepted: 09/06/2015] [Indexed: 11/25/2022] Open
Abstract
OBJECTIVE To determine if coenzyme Q(10) alleviates fatigue in the late-onset sequelae of poliomyelitis. DESIGN Parallel-group, randomized, placebo-controlled trial. BACKGROUND SETTING Coenzyme Q(10) has been shown to boost muscle energy metabolism in post-polio subjects but it does not promote muscle strength, endurance or function in polio survivors with post-poliomyelitis syndrome. However, the collective increased energy metabolism might contribute to a reduction in post-polio fatigue. PARTICIPANTS Polio survivors from the Australian post-polio networks in Queensland and New South Wales who attribute a moderate to high level of fatigue to their diagnosed late-onset sequelae of poliomyelitis. Those with fatigue-associated comorbidities of diabetes, anaemia, hypothyroidism and fibromyalgia were excluded. METHOD Participants were assigned (1:1), with stratification of those who use energy-saving mobility aids, to receive 100 mg coenzyme Q(10) or matching placebo daily for 60 days. Participants and investigators were blinded to group allocation. Fatigue was assessed by the Multidimensional Assessment of Fatigue as the primary outcome and the Fatigue Severity Scale as secondary outcome. RESULTS Of 103 participants, 54 were assigned to receive coenzyme Q(10) and 49 to receive the placebo. The difference in the mean score reductions between the two groups was not statistically significant for either fatigue measure. Oral supplementation with coenzyme Q(10) was safe and well-tolerated. CONCLUSION A daily dose of 100 mg coenzyme Q(10) for 60 days does not alleviate the fatigue of the late-onset sequelae of poliomyelitis. The registration number for the clinical trial is ACTRN 12612000552886.
Collapse
Affiliation(s)
- Margaret M Peel
- NHMRC Centre for Research Excellence in Nursing Interventions, Menzies Health Institute Queensland, Centre for Health Practice Innovation, Griffith University, Queensland, Australia
| | - Marie Cooke
- NHMRC Centre for Research Excellence in Nursing Interventions, Menzies Health Institute Queensland, Centre for Health Practice Innovation, Griffith University, Queensland, Australia.
| | - Helen J Lewis-Peel
- NHMRC Centre for Research Excellence in Nursing Interventions, Menzies Health Institute Queensland, Centre for Health Practice Innovation, Griffith University, Queensland, Australia
| | - Rodney A Lea
- Institute of Health and Biomedical Innovation, Queensland University of Technology, Queensland, Australia
| | - Wendy Moyle
- NHMRC Centre for Research Excellence in Nursing Interventions, Menzies Health Institute Queensland, Centre for Health Practice Innovation, Griffith University, Queensland, Australia
| |
Collapse
|
90
|
Abstract
PURPOSE OF REVIEW To describe the beneficial effects of micronutrient supplementation on male fertility. RECENT FINDINGS Several micronutrients have beneficial effects on sperm quality, as well on male fertility (e.g. pregnancy rate). A deliberate use of micronutrients might be helpful for infertile patients. Healthcare providers should be aware that supplements contain the studied dose. SUMMARY Male sterility is becoming increasingly important because of various factors. In addition to the avoidable risk factors (alcohol and smoking), unchangeable factors are also likely involved in the genesis. Modern reproductive medicine methods help resulting in decent pregnancy rates in subfertile men. However, in addition to reproductive medicine methods, factors other than cessation of smoking and alcohol consumption can influence the fertility of men. Several studies have reported a significant increase in sperm quality and pregnancy rates when the men were supplemented by specific vitamins and micronutrients.The present review gives an overview of the study results and discusses specific legal requirements (e.g. 'upper limits').
Collapse
|
91
|
Ko EY, Sabanegh ES, Agarwal A. Male infertility testing: reactive oxygen species and antioxidant capacity. Fertil Steril 2014; 102:1518-27. [DOI: 10.1016/j.fertnstert.2014.10.020] [Citation(s) in RCA: 159] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/14/2014] [Accepted: 10/14/2014] [Indexed: 02/07/2023]
|
92
|
de Frutos F, Gea A, Hernandez-Estefania R, Rabago G. Prophylactic treatment with coenzyme Q10 in patients undergoing cardiac surgery: could an antioxidant reduce complications? A systematic review and meta-analysis. Interact Cardiovasc Thorac Surg 2014; 20:254-9. [DOI: 10.1093/icvts/ivu334] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
93
|
Swaminathan A, Jicha GA. Nutrition and prevention of Alzheimer's dementia. Front Aging Neurosci 2014; 6:282. [PMID: 25368575 PMCID: PMC4202787 DOI: 10.3389/fnagi.2014.00282] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 09/25/2014] [Indexed: 11/13/2022] Open
Abstract
A nutritional approach to prevent, slow, or halt the progression of disease is a promising strategy that has been widely investigated. Much epidemiologic data suggests that nutritional intake may influence the development and progression of Alzheimer’s dementia (AD). Modifiable, environmental causes of AD include potential metabolic derangements caused by dietary insufficiency and or excess that may be corrected by nutritional supplementation and or dietary modification. Many nutritional supplements contain a myriad of health promoting constituents (anti-oxidants, vitamins, trace minerals, flavonoids, lipids, …etc.) that may have novel mechanisms of action affecting cellular health and regeneration, the aging process itself, or may specifically disrupt pathogenic pathways in the development of AD. Nutritional modifications have the advantage of being cost effective, easy to implement, socially acceptable and generally safe and devoid of significant adverse events in most cases. Many nutritional interventions have been studied and continue to be evaluated in hopes of finding a successful agent, combination of agents, or dietary modifications that can be used for the prevention and or treatment of AD. The current review focuses on several key nutritional compounds and dietary modifications that have been studied in humans, and further discusses the rationale underlying their potential utility for the prevention and treatment of AD.
Collapse
Affiliation(s)
- Arun Swaminathan
- Department of Neurology and Sanders-Brown Center on Aging, College of Medicine, University of Kentucky Lexington, KY, USA
| | - Gregory A Jicha
- Department of Neurology and Sanders-Brown Center on Aging, College of Medicine, University of Kentucky Lexington, KY, USA
| |
Collapse
|
94
|
Tarry-Adkins JL, Fernandez-Twinn DS, Chen JH, Hargreaves IP, Martin-Gronert MS, McConnell JM, Ozanne SE. Nutritional programming of coenzyme Q: potential for prevention and intervention? FASEB J 2014; 28:5398-405. [PMID: 25172893 PMCID: PMC4232289 DOI: 10.1096/fj.14-259473] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Low birth weight and rapid postnatal growth increases risk of cardiovascular-disease (CVD); however, underlying mechanisms are poorly understood. Previously, we demonstrated that rats exposed to a low-protein diet in utero that underwent postnatal catch-up growth (recuperated) have a programmed deficit in cardiac coenzyme Q (CoQ) that was associated with accelerated cardiac aging. It is unknown whether this deficit occurs in all tissues, including those that are clinically accessible. We investigated whether aortic and white blood cell (WBC) CoQ is programmed by suboptimal early nutrition and whether postweaning dietary supplementation with CoQ could prevent programmed accelerated aging. Recuperated male rats had reduced aortic CoQ [22 d (35±8.4%; P<0.05); 12 m (53±8.8%; P<0.05)], accelerated aortic telomere shortening (P<0.01), increased DNA damage (79±13% increase in nei-endonucleaseVIII-like-1), increased oxidative stress (458±67% increase in NAPDH-oxidase-4; P<0.001), and decreased mitochondrial complex II-III activity (P<0.05). Postweaning dietary supplementation with CoQ prevented these detrimental programming effects. Recuperated WBCs also had reduced CoQ (74±5.8%; P<0.05). Notably, WBC CoQ levels correlated with aortic telomere-length (P<0.0001) suggesting its potential as a diagnostic marker of vascular aging. We conclude that early intervention with CoQ in at-risk individuals may be a cost-effective and safe way of reducing the global burden of CVDs.—Tarry-Adkins, J. L., Fernandez-Twinn, D. S., Chen, J.-H., Hargreaves, I. P., Martin-Gronert, M. S., McConnell, J. M., Ozanne, S. E. Nutritional programming of coenzyme Q: potential for prevention and intervention?
Collapse
Affiliation(s)
- Jane L Tarry-Adkins
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK; and
| | - Denise S Fernandez-Twinn
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK; and
| | - Jian-Hua Chen
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK; and
| | - Iain P Hargreaves
- Neurometabolic Unit, National Hospital, University College London, London, UK
| | - Malgorzata S Martin-Gronert
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK; and
| | - Josie M McConnell
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK; and
| | - Susan E Ozanne
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK; and
| |
Collapse
|
95
|
Garrido-Maraver J, Cordero MD, Oropesa-Ávila M, Fernández Vega A, de la Mata M, Delgado Pavón A, de Miguel M, Pérez Calero C, Villanueva Paz M, Cotán D, Sánchez-Alcázar JA. Coenzyme q10 therapy. Mol Syndromol 2014; 5:187-97. [PMID: 25126052 DOI: 10.1159/000360101] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
For a number of years, coenzyme Q10 (CoQ10) was known for its key role in mitochondrial bioenergetics; later studies demonstrated its presence in other subcellular fractions and in blood plasma, and extensively investigated its antioxidant role. These 2 functions constitute the basis for supporting the clinical use of CoQ10. Also, at the inner mitochondrial membrane level, CoQ10 is recognized as an obligatory cofactor for the function of uncoupling proteins and a modulator of the mitochondrial transition pore. Furthermore, recent data indicate that CoQ10 affects the expression of genes involved in human cell signaling, metabolism and transport, and some of the effects of CoQ10 supplementation may be due to this property. CoQ10 deficiencies are due to autosomal recessive mutations, mitochondrial diseases, aging-related oxidative stress and carcinogenesis processes, and also statin treatment. Many neurodegenerative disorders, diabetes, cancer, and muscular and cardiovascular diseases have been associated with low CoQ10 levels as well as different ataxias and encephalomyopathies. CoQ10 treatment does not cause serious adverse effects in humans and new formulations have been developed that increase CoQ10 absorption and tissue distribution. Oral administration of CoQ10 is a frequent antioxidant strategy in many diseases that may provide a significant symptomatic benefit.
Collapse
Affiliation(s)
- Juan Garrido-Maraver
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-Consejo Superior de Investigaciones Científicas, Sevilla, Spain
| | - Mario D Cordero
- Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Universidad Pablo de Olavide-Consejo Superior de Investigaciones Científicas, Sevilla, Spain ; Departamento de Citología e Histología Normal y Patológica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Manuel Oropesa-Ávila
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-Consejo Superior de Investigaciones Científicas, Sevilla, Spain
| | - Alejandro Fernández Vega
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-Consejo Superior de Investigaciones Científicas, Sevilla, Spain
| | - Mario de la Mata
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-Consejo Superior de Investigaciones Científicas, Sevilla, Spain
| | - Ana Delgado Pavón
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-Consejo Superior de Investigaciones Científicas, Sevilla, Spain
| | - Manuel de Miguel
- Departamento de Citología e Histología Normal y Patológica, Facultad de Medicina, Universidad de Sevilla, Sevilla, Spain
| | - Carmen Pérez Calero
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-Consejo Superior de Investigaciones Científicas, Sevilla, Spain
| | - Marina Villanueva Paz
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-Consejo Superior de Investigaciones Científicas, Sevilla, Spain
| | - David Cotán
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-Consejo Superior de Investigaciones Científicas, Sevilla, Spain
| | - José A Sánchez-Alcázar
- Centro Andaluz de Biología del Desarrollo (CABD), Universidad Pablo de Olavide-Consejo Superior de Investigaciones Científicas, Sevilla, Spain ; Centro de Investigación Biomédica en Red: Enfermedades Raras, Instituto de Salud Carlos III, Universidad Pablo de Olavide-Consejo Superior de Investigaciones Científicas, Sevilla, Spain
| |
Collapse
|
96
|
Sikorska M, Lanthier P, Miller H, Beyers M, Sodja C, Zurakowski B, Gangaraju S, Pandey S, Sandhu JK. Nanomicellar formulation of coenzyme Q10 (Ubisol-Q10) effectively blocks ongoing neurodegeneration in the mouse 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model: potential use as an adjuvant treatment in Parkinson's disease. Neurobiol Aging 2014; 35:2329-46. [PMID: 24775711 DOI: 10.1016/j.neurobiolaging.2014.03.032] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 03/25/2014] [Accepted: 03/28/2014] [Indexed: 12/13/2022]
Abstract
Although the support for the use of antioxidants, such as coenzyme Q(10) (CoQ(10)), to treat Parkinson's disease (PD) comes from the extensive scientific evidence, the results of conducted thus far clinical trials are inconclusive. It is assumed that the efficacy of CoQ(10) is hindered by insolubility, poor bioavailability, and lack of brain penetration. We have developed a nanomicellar formulation of CoQ(10) (Ubisol-Q(10)) with improved properties, including the brain penetration, and tested its effectiveness in mouse MPTP (1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine) model with the objectives to assess its potential use as an adjuvant therapy for PD. We used a subchronic MPTP model (5-daily MPTP injections), characterized by 50% loss of dopamine neurons over a period of 28 days. Ubisol-Q(10) was delivered in drinking water. Prophylactic application of Ubisol-Q(10), started 2 weeks before the MPTP exposure, significantly offset the neurotoxicity (approximately 50% neurons died in MPTP group vs. 17% in MPTP+ Ubisol-Q(10) group by day 28). Therapeutic application of Ubisol-Q(10), given after the last MPTP injection, was equally effective. At the time of intervention on day 5 nearly 25% of dopamine neurons were already lost, but the treatment saved the remaining 25% of cells, which otherwise would have died by day 28. This was confirmed by cell counts, analyses of striatal dopamine levels, and improved animals' motor skill on a beam walk test. Similar levels of neuroprotection were obtained with 3 different Ubisol-Q(10) concentrations tested, that is, 30 mg, 6 mg, or 3 mg CoQ(10)/kg body weight/day, showing clearly that high doses of CoQ(10) were not required to deliver these effects. Furthermore, the Ubisol-Q(10) treatments brought about a robust astrocytic activation in the brain parenchyma, indicating that astroglia played an active role in this neuroprotection. Thus, we have shown for the first time that Ubisol-Q(10) was capable of halting the neurodegeneration already in progress; however, to maintain it a continuous supplementation of Ubisol-Q(10) was required. The pathologic processes initiated by MPTP resumed if supplementation was withdrawn. We suggest that in addition to brain delivery of powerful antioxidants, Ubisol-Q(10) might have also supported subcellular oxidoreductase systems allowing them to maintain a favorable cellular redox status, especially in astroglia, facilitating their role in neuroprotection. Based on this data further clinical testing of this formulation in PD patients might be justifiable.
Collapse
Affiliation(s)
- Marianna Sikorska
- Department of Translational Bioscience, Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, Ontario, Canada
| | - Patricia Lanthier
- Department of Translational Bioscience, Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, Ontario, Canada
| | - Harvey Miller
- Department of Translational Bioscience, Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, Ontario, Canada
| | - Melissa Beyers
- Department of Translational Bioscience, Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, Ontario, Canada
| | - Caroline Sodja
- Department of Translational Bioscience, Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, Ontario, Canada
| | - Bogdan Zurakowski
- Department of Translational Bioscience, Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, Ontario, Canada
| | - Sandhya Gangaraju
- Department of Translational Bioscience, Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, Ontario, Canada
| | - Siyaram Pandey
- Department of Chemistry and Biochemistry, University of Windsor Essex Hall, Windsor, Ontario, Canada
| | - Jagdeep K Sandhu
- Department of Translational Bioscience, Human Health Therapeutics Portfolio, National Research Council Canada, Ottawa, Ontario, Canada.
| |
Collapse
|
97
|
Wang D, Yan X, Xia M, Yang Y, Li D, Li X, Song F, Ling W. Coenzyme Q10 promotes macrophage cholesterol efflux by regulation of the activator protein-1/miR-378/ATP-binding cassette transporter G1-signaling pathway. Arterioscler Thromb Vasc Biol 2014; 34:1860-70. [PMID: 24675662 DOI: 10.1161/atvbaha.113.302879] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Recent studies have shown the role of miRNAs in macrophage reverse cholesterol transport and atherogenesis. We hypothesized that coenzyme Q10 (CoQ10) may increase macrophage reverse cholesterol transport by regulating miRNA expression that contributes to the prevention of atherosclerosis. APPROACH AND RESULTS CoQ10 treatment suppressed oxidized low-density lipoprotein-induced macrophage foam cell formation by ameliorating the binding of activator protein-1 to the putative promoter region of miR-378 primary transcript, thus decreasing the miR-378 level and enhancing the ATP-binding cassette transporter G1-mediated macrophage cholesterol efflux to high-density lipoprotein. Subsequently, the axis of activator protein-1/miR-378/ATP-binding cassette transporter G1 cholesterol efflux was confirmed in peritoneal macrophages isolated from CoQ10-treated apolipoprotein E-deficient mice. Finally, CoQ10 consumption promoted macrophage reverse cholesterol transport and inhibited the progression of atherosclerosis in apolipoprotein E-deficient mice. CONCLUSIONS This study identified activator protein-1/miR-378/ATP-binding cassette transporter G1 as a novel cascade for CoQ10 in facilitating macrophage cholesterol efflux in vitro and in vivo. Our data thus imply that both CoQ10 and miR-378 are promising candidates for atherosclerosis prevention and treatment.
Collapse
Affiliation(s)
- Dongliang Wang
- From the Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., X.Y., M.X., Y.Y., D.L., X.L., F.S., W.L.); and Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., M.X., Y.Y., D.L., W.L.)
| | - Xiao Yan
- From the Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., X.Y., M.X., Y.Y., D.L., X.L., F.S., W.L.); and Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., M.X., Y.Y., D.L., W.L.)
| | - Min Xia
- From the Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., X.Y., M.X., Y.Y., D.L., X.L., F.S., W.L.); and Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., M.X., Y.Y., D.L., W.L.)
| | - Yan Yang
- From the Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., X.Y., M.X., Y.Y., D.L., X.L., F.S., W.L.); and Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., M.X., Y.Y., D.L., W.L.)
| | - Dan Li
- From the Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., X.Y., M.X., Y.Y., D.L., X.L., F.S., W.L.); and Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., M.X., Y.Y., D.L., W.L.)
| | - Xinrui Li
- From the Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., X.Y., M.X., Y.Y., D.L., X.L., F.S., W.L.); and Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., M.X., Y.Y., D.L., W.L.)
| | - Fenglin Song
- From the Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., X.Y., M.X., Y.Y., D.L., X.L., F.S., W.L.); and Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., M.X., Y.Y., D.L., W.L.)
| | - Wenhua Ling
- From the Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., X.Y., M.X., Y.Y., D.L., X.L., F.S., W.L.); and Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Department of Nutrition, School of Public Health, Sun Yat-Sen University (Northern Campus), Guangzhou, Guangdong Province, People's Republic of China (D.W., M.X., Y.Y., D.L., W.L.).
| |
Collapse
|
98
|
Lee JH, Hoang NH, Huong NL, Shrestha A, Park JW. Ultra-Performance Liquid Chromatography with Electrospray Ionization Mass Spectrometry for the Determination of Coenzyme Q10as an Anti-Aging Ingredient in Edible Cosmetics. ANAL LETT 2014. [DOI: 10.1080/00032719.2013.841175] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
99
|
Coenzyme Q10 as a therapy for mitochondrial disease. Int J Biochem Cell Biol 2014; 49:105-11. [PMID: 24495877 DOI: 10.1016/j.biocel.2014.01.020] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/14/2014] [Accepted: 01/26/2014] [Indexed: 01/11/2023]
Abstract
Treatment of mitochondrial respiratory chain (MRC) disorders is extremely difficult, however, coenzyme Q10 (CoQ10) and its synthetic analogues are the only agents which have shown some therapeutic benefit to patients. CoQ10 serves as an electron carrier in the MRC as well as functioning as a potent lipid soluble antioxidant. CoQ10 supplementation is fundamental to the treatment of patients with primary defects in the CoQ10 biosynthetic pathway. The efficacy of CoQ10 and its analogues in the treatment of patients with MRC disorders not associated with a CoQ10 deficiency indicates their ability to restore electron flow in the MRC and/or increase mitochondrial antioxidant capacity may also be important contributory factors to their therapeutic potential.
Collapse
|
100
|
Barakat A, Shegokar R, Dittgen M, Müller RH. Coenzyme Q10 oral bioavailability: effect of formulation type. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2013. [DOI: 10.1007/s40005-013-0101-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|