Novel CoQ10 antidiabetic mechanisms underlie its positive effect: modulation of insulin and adiponectine receptors, Tyrosine kinase, PI3K, glucose transporters, sRAGE and visfatin in insulin resistant/diabetic rats

Mohamed M Amin, Gihan F Asaad, Rania M Abdel Salam, Hanan S El-Abhar, Mahmoud S Arbid, Mohamed M Amin, Gihan F Asaad, Rania M Abdel Salam, Hanan S El-Abhar, Mahmoud S Arbid

Abstract

As a nutritional supplement, coenzyme Q10 (CoQ10) was tested previously in several models of diabetes and/or insulin resistance (IR); however, its exact mechanisms have not been profoundly explicated. Hence, the objective of this work is to verify some of the possible mechanisms that underlie its therapeutic efficacy. Moreover, the study aimed to assess the potential modulatory effect of CoQ10 on the antidiabetic action of glimebiride. An insulin resistance/type 2 diabetic model was adopted, in which rats were fed high fat/high fructose diet (HFFD) for 6 weeks followed by a single sub-diabetogenic dose of streptozotocin (35 mg/kg, i.p.). At the end of the 7(th) week animals were treated with CoQ10 (20 mg/kg, p.o) and/or glimebiride (0.5 mg/kg, p.o) for 2 weeks. CoQ10 alone opposed the HFFD effect and increased the hepatic/muscular content/activity of tyrosine kinase (TK), phosphatidylinositol kinase (PI3K), and adiponectin receptors. Conversely, it decreased the content/activity of insulin receptor isoforms, myeloperoxidase and glucose transporters (GLUT4; 2). Besides, it lowered significantly the serum levels of glucose, insulin, fructosamine and HOMA index, improved the serum lipid panel and elevated the levels of glutathione, sRAGE and adiponectin. On the other hand, CoQ10 lowered the serum levels of malondialdehyde, visfatin, ALT and AST. Surprisingly, CoQ10 effect surpassed that of glimepiride in almost all the assessed parameters, except for glucose, fructosamine, TK, PI3K, and GLUT4. Combining CoQ10 with glimepiride enhanced the effect of the latter on the aforementioned parameters.

Conclusion: These results provided a new insight into the possible mechanisms by which CoQ10 improves insulin sensitivity and adjusts type 2 diabetic disorder. These mechanisms involve modulation of insulin and adiponectin receptors, as well as TK, PI3K, glucose transporters, besides improving lipid profile, redox system, sRAGE, and adipocytokines. The study also points to the potential positive effect of CoQ10 as an adds- on to conventional antidiabetic therapies.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. The glucose tolerance curve depicts…
Figure 1. The glucose tolerance curve depicts the changes in glucose (2 g/kg, p.o) response in serum of normal control group (NFD) and non-treated insulin-resistant rats (HFFD), after 6 weeks of food manipulation at 0, 30, 60, 90, and 120 min.
Values are means ± S.E of 6 animals; as compared to the normal control group (*) (one-way ANOVA followed by Tukey-Kramer post hoc test), P<0.05.
Figure 2. Effect of diabetes and 2…
Figure 2. Effect of diabetes and 2 weeks oral administration of CoQ10 (20 mg/kg) and/or glimepiride (0.5 mg/kg) on the hepatic (A, B) and muscular (C, D) insulin receptor isoforms (high affinity, [HAIR, fmol/mg protein] and low affinity [LAIR, pmol/mg protein] insulin receptor).
Values are means of 8–10 animals ± S.E.M. As compared with normal control (*), diabetic control (#), glimepiride treated ($) and CoQ10 treated (@) groups (one-way ANOVA followed by Tukey-Kramer post hoc test), P<0.05.
Figure 3. Effect of diabetes and 2…
Figure 3. Effect of diabetes and 2 weeks oral administration of CoQ10 (20 mg/kg) and/or glimepiride (0.5 mg/kg) on the hepatic (A, B) and muscular (C, D) activity of tyrosine kinase (ng/mg protein) and PI3K (µg/mg protein).
Values are means of 8–10 animals ± S.E.M. As compared with normal control (*), diabetic control (#), glimepiride treated ($) and CoQ10 treated (@) groups (one-way ANOVA followed by Tukey-Kramer post hoc test), P<0.05.
Figure 4. Effect of diabetes and 2…
Figure 4. Effect of diabetes and 2 weeks oral administration of CoQ10 (20 mg/kg) and/or glimepiride (0.5 mg/kg) on hepatic (A, B) and muscular (C, D) adiponectin receptors (Adipo-R1, Adipo-R2 [ng/mg protein]).
Values are means of 8–10 animals ± S.E.M. As compared with normal control (*), diabetic control (#), glimepiride treated ($) and CoQ10 treated (@) groups (one-way ANOVA followed by Tukey-Kramer post hoc test), P<0.05.
Figure 5. Effect of diabetes and 2…
Figure 5. Effect of diabetes and 2 weeks oral administration of CoQ10 (20 mg/kg) and/or glimepiride (0.5 mg/kg) on the hepatic (A) and muscular (C) glucose transporters [ng/mg protein], and (B, D) myeloperoxidase activity [MPO, U/mg].
Values are means of 8–10 animals ± S.E.M. As compared with normal control (*), diabetic control (#), glimepiride treated ($) and CoQ10 treated (@) groups (one-way ANOVA followed by Tukey-Kramer post hoc test), P<0.05.

References

    1. Keller KB, Lemberg L (2003) Obesity and the metabolic syndrome. Am J Crit Care. 12: 167–170.
    1. Schaalan M, El-Abhar H, Barakat M, El-Denshary ES (2009) Westernized-like-diet-fed rats: effect on glucose homeostasis, lipid profile, and adipocyte hormones and their modulation by rosiglitazone and glimepiride. J Diabetes Complications 23: 199–208.
    1. Lamson DW, Plaza SM (2002) Mitochondrial factors in the pathogenesis of diabetes: a hypothesis for treatment. Altern Med Rev 7: 94–111.
    1. Van de Weijer T, Sparks LM, Phielix E, Meex RC, van Herpen NA, et al. (2013) Relationships between mitochondrial function and metabolic flexibility in type 2 diabetes mellitus. PLoS One 8: e51648.
    1. Lee HM, Kim JJ, Kim HJ, Shong M, Ku BJ, et al. (2013) Up-regulated NLRP3 inflammasome activation in patients with Type 2 Diabetes. Diabetes 62: 194–204.
    1. Sourris KC, Harcourt BE, Tang PH, Morley AL, Huynh K, et al. (2012) Ubiquinone (coenzyme Q10) prevents renal mitochondrial dysfunction in an experimental model of type 2 diabetes. Free Radic Biol Med 52: 716–723.
    1. Eriksson J, Lindström J, Valle T, Aunola S, Hämäläinen H, et al. (1999) Prevention of type II diabetes in subjects with impaired glucose tolerance: the Diabetes Prevention Study (DPS) in Finland: Study design and 1-year interim report on the feasibility of the lifestyle intervention programme. Diabetologia 42: 793–801.
    1. Sena CM, Nunes E, Gomes A, Santos MS, Proença T, et al. (2008) Supplementation of coenzyme Q10 and alpha-tocopherol lowers glycated hemoglobin level and lipid peroxidation in pancreas of diabetic rats. Nutr Res 28: 113–121.
    1. Villalba JM, Parrado C, Santos-Gonzalez M, Alcain FJ (2010) Therapeutic use of coenzyme Q10 and coenzyme Q10-related compounds and formulations. Expert Opin Investig Drugs 19: 535–554.
    1. Molyneux SL, Florkowski CM, George PM, Pilbrow AP, Frampton CM, et al. (2008) Coenzyme Q10: an independent predictor of mortality in chronic heart failure. J Am Coll Cardiol 52: 1435–1441.
    1. Chew GT, Watts GF (2004) Coenzyme Q10 and diabetic endotheliopathy: oxidative stress and the ‘recoupling hypothesis’. QJM. 97: 537–548.
    1. Sohet FM, Neyrinck AM, Pachikian BD, de Backer FC, Bindels LB, et al. (2009) Coenzyme Q10 supplementation lowers hepatic oxidative stress and inflammation associated with diet-induced obesity in mice. Biochem Pharmacol 78: 1391–1400.
    1. Ahmadvand H, Tavafi M, Khosrowbeygi A (2012) Amelioration of altered antioxidant enzymes activity and glomerulosclerosis by coenzyme Q10 in alloxan-induced diabetic rats. J Diabetes Complications 26: 476–482.
    1. Andersen CB, Henriksen JE, Hother-Nielsen O, Vaag A, Mortensen SA, et al. (1997) The effect of coenzyme Q10 on blood glucose and insulin requirement in patients with insulin dependent diabetes mellitus. Mol Aspects Med 18 Suppl: S307–309
    1. Hodgson JM, Watts GF, Playford DA, Burke V, Croft KD (2002) Coenzyme Q10 improves blood pressure and glycaemic control: a controlled trial in subjects with type 2 diabetes. Eur J Clin Nutr 56: 1137–1142.
    1. Mezawa M, Takemoto M, Onishi S, Ishibashi R, Ishikawa T, et al. (2012) The reduced form of coenzyme Q10 improves glycemic control in patients with type 2 diabetes: an open label pilot study. Biofactors 38: 416–421.
    1. Bauerova K, Paulovicova E, Mihalova D, Drafi F, Strosova M, et al. (2010) Combined methotrexate and coenzyme Q10 therapy in adjuvant-induced arthritis evaluated using parameters of inflammation and oxidative stress. Acta Biochimica Polonica 57: 347–354.
    1. Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, et al. (1985) Homeostasis model assessment: Insulin resistance and b-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 28: 412–419.
    1. El-Abhar HS, Schaalan MF (2012) Topiramate-induced modulation of hepatic molecular mechanisms: An aspect for its anti-insulin resistant effect. PLoS ONE 7: e37757 doi:
    1. Bradford MM (1976) A rapid and sensitive method for the quantitation of microgram quantities of protein utilizing the principle of protein-dye binding. Anal Biochem 72: 248–254.
    1. Corin RE, Donner DB (1982) Insulin receptors convert to a higher affinity state subsequent to hormone binding. A two state model for the insulin receptors. J Biol Chem 275: 104–110.
    1. Miller JL, Lynn CH, Shuster J, Driscoll DJ (2011) Carnitine and coenzyme Q10 levels in individuals with Prader-Willi syndrome. Am J Med Genet A 155 A: 569–573.
    1. Dzugkoev SG, Kaloeva MB, Dzugkoeva FS (2012) Effect of combination therapy with coenzyme Q10 on functional and metabolic parameters in patients with type 1 diabetes mellitus. Bull Exp Biol Med 152: 364–366.78.
    1. Mc Carty MF (1999) Can correction of sub-optimal coenzyme Q status improve beta-cell function in type II diabetics? Med Hypotheses 52: 397–400.
    1. Crescenzo R, Bianco F, Falcone I, Tsalouhidou S, Yepuri G, et al. (2012) Hepatic mitochondrialenergetics during catch-up fat with high-fat diets rich in lard or safflower oil. Obesity (SilverSpring) 20: 1763–1772.
    1. Ernster L, Dallner G (1995) Biochemical, physiological and medical aspects of ubiquinone function. Biochim Biophys Acta 1271: 195–204.
    1. Moore MC, Cherrington AD, Mann SL, Davis SN (2000) Acute fructose administration decreases the glycemic response to an oral glucose tolerance test in normal adults. J Clin Endocrinol Metab 85: 4515–4519.
    1. Avramoglu RK, Basciano H, Adeli K (2006) Lipid and lipoprotein dysregulation in insulin resistant states. Clin Chim Acta 368: 1–19.
    1. Julius U (2003) Influence of plasma free fatty acids on lipoprotein synthesis and diabetic dyslipidemia. Exp Clin Endocrinol Diabetes 111: 246–250. Review.
    1. Taskinen MR, Beltz WF, Harper I, Fields RM, Schonfeld G, et al. (1986) Effects of NIDDM on very-low-density lipoprotein triglyceride and apo-lipoprotein B metabolism. Studies before andafter sulfonylurea therapy. Diabetes 35: 1268–1277.
    1. Song H, Shojima N, Sakoda H, Ogihara T, Fujishiro M, et al. (2002) Resistin is regulated by C/EBPs, PPARs, and signal-transducing molecules. Biochem Biophys Res Commun 29: 291–298.
    1. Marshall S, Bacote V, Traxinger RR (1991) Discovery of a metabolic pathway mediating glucose-induced desensitization of the glucose transport system. Role of hexosamine biosynthesis in the induction of insulin resistance. J Biol Chem 266: 4706–4712.
    1. McClain DA (2002) Hexosamines as mediators of nutrient sensing and regulation in diabetes. J Diabetes Complications 16: 72–80.
    1. Ueno M, Bezerra RM, Silva MS, Tavares DQ, Carvalho CR, et al. (2000) A high-fructose diet induces changes in pp185 phosphorylation in muscle and liver of rats. Braz J Med Biol Res 33: 1421–1427.
    1. Mosthab L, Vogt B, Haring HU, Ullrich A (1991) Altered expression of insulin receptor types A and B in the skeletal muscle of noninsulin-dependent diabetes mellitus. Proc Natl Acad Sci USA 88: 4728–4730.
    1. Kellerer M, Sesti G, Seffer E, Obermaier-Kusser B, Pongratz DE, et al. (1993) Altered pattern of insulin receptor isotypes in skeletal muscle membranes of typeII (noninsulin-dependent) diabetic subjects. Diabetologia 36: 628–632.
    1. Sbraccia P, D’Adamo M, Leonetti F, Caiola S, Iozzo P, et al. (1996) Chronic primary hyperinsulinemia is associated with altered insulin receptor mRNA splicing in muscle of patients with insulinoma. Diabetologia 39: 220–225.
    1. Krauss H, Grzymisławski M, Koźlik J, Sosnowski P, Piatek J, et al. (2004) The influence of glimepiride on the binding kinetics of insulin with its skeletal muscle and liver receptors in rats with short term and prolonged hyperglycemia induced by streptozotocin. Med Sci Monit 10: BR11–16.
    1. Baron AD, Zhu JS, Weldon H, Maianu L, Garvey WT (1995) Glucosamine induces insulin resistance in vivo by affecting GLUT-4 translocation in skeletal muscle: Implications for glucose toxicity. J Clin Invest 96: 2792–2801.
    1. Stephens JM, Lee J, Pilch PF (1997) Tumor necrosis factor-alpha-induced insulin resistance in 3T3-L1 adipocytes is accompanied by a loss of insulin receptor substrate-1 and GLUT4 expression without a loss of insulin receptor-mediated signal transduction. J Biol Chem 272: 971–976.
    1. Leguisamo NM, Lehnen AM, Machado UF, Okamoto MM, Markoski MM, et al. (2012) GLUT4 content decreases along with insulin resistance and high levels of inflammatory markers in rats with metabolic syndrome. Cardiovasc Diabetol 11: 100.
    1. Bernat-Karpińska M, Piątkiewicz P, Czech A, Wierzbicki P (2012) The expression of particular glucose transporters and insulin resistance indicators in the risk groups of type 2 diabetes–a two year follow-up. Endokrynol Pol 63: 212–219. [Abstract].
    1. Ho YJ, Chen WP, Chi TC, Chang Chien CC, Lee AS, et al. (2013) 1 Caffeic acid phenethyl amide improves glucose homeostasis and attenuates the progression of vascular dysfunction in Streptozotocin-induced diabetic rats. Cardiovasc Diabetol 12: 99.
    1. Kruszynska YT, Worrall DS, Ofrecio J, Frias JP, Macaraeg G, et al. (2002) Fatty acid-induced insulin resistance: decreased muscle PI3K activation but unchanged Akt phosphorylation. J Clin Endocrinol Metab 87: 226–234.
    1. Choi H, Park HH, Lee KY, Choi NY, Yu HJ, et al. (2013) Coenzyme Q10 restores amyloid beta-inhibited proliferation of neural stem cells by activating the PI3K pathway. Stem Cells Dev. 22: 2112–2120.
    1. Ma P, Xiong W, Liu H, Ma J, Gu B, et al. (2011) Extrapancreatic roles of glimepiride on osteoblasts from rat manibular bone in vitro: Regulation of cytodifferentiation through PI3-kinases/Akt signalling pathway. Arch Oral Biol. 56: 307–316.
    1. Oka Y, Asano T, Shibasaki Y, Lin JL, Tsukuda K, et al. (1990) Increased liver glucose transporter protein and mRNA in streptozocin-induced diabetic rats. Diabetes 39: 441–446.
    1. Burcelin R, Eddouks M, Kande J, Assan R, Girard J (1992) Evidence that GLUT-2 mRNA and protein concentrations are decreased by hyperinsulinaemia and increased by hyperglycaemia in liver of diabetic rats. Biochem J 288: 675–679.
    1. Postic C, Burcelin R, Rencurel F, Pegorier JP, Loizeau M, et al. (1993) Evidence for a transient inhibitory effect of insulin on GLUT2 expression in the liver: studies in vivo and in vitro. Biochem J 293: 119–124.
    1. Bernsmeier C, Heim MH (2009) Insulin resistance in chronic hepatitis C: mechanisms and clinical relevance. Swiss Med Wkly 139: 678–84.
    1. Koyama H, Yamamoto H, Nishizawa Y (2007) Endogenous secretory RAGE as a novel biomarker for metabolic syndrome and cardiovascular diseases. Biomark Insights 2: 331–339.
    1. Tan KC, Shiu SW, Wong Y, Tam X (2011) Serum advanced glycation end products (AGEs) are associated with insulin resistance. Diabetes Metab Res Rev 27: 488–492.
    1. Lu L, Peng W, Wang W, Wang L, Chen Q, et al. (2011) Effects of atorvastatin on progression of diabetic nephropathy and local RAGE and soluble RAGE expressions in rats. J Zhejiang Univ Sci B 2: 652–659.
    1. Koyama H, Shoji T, Yokoyama H, Motoyama K, Mori K, et al. (2005) Plasma level of endogenous secretory RAGE is associated with components of the metabolic syndrome and atherosclerosis. Arterioscler Thromb Vasc Biol 25: 2587–2593.
    1. Devangelio E, Santilli F, Formoso G, Ferroni P, Bucciarelli L, et al. (2007) Soluble RAGE in type 2 diabetes: association with oxidative stress. Free Radic Biol Med 43: 511–518.
    1. Anderson JW, Gowri MS, Turner J, Nichols L, Diwadkar VA, et al. (1999) Antioxidant supplementation effects on low-density lipoprotein oxidation for individuals with type 2 diabetes mellitus. J Am Coll Nutr 18: 451–461.
    1. El-ghoroury EA, Raslan HM, Badawy EA, El-Saaid GS, Agybi MH, et al. (2009) Malondialdehyde and coenzyme Q10 in platelets and serum in type 2 diabetes mellitus: correlation with glycemic control. Blood Coagul Fibrinolysis 20: 248–251.
    1. Dzugkoev SG, Kaloeva MB, Dzugkoeva FS (2012) Effect of combination therapy with coenzyme Q10 on functional and metabolic parameters in patients with type 1 diabetes mellitus. Bull Exp Biol Med 152: 364–366.
    1. Bhagavan HN, Chopra RK (2007) Plasma coenzyme Q10 response to oral ingestion of coenzyme Q10 formulations. Mitochondrion 7: S78–88.
    1. Crane FL (2001) Biochemical functions of coenzyme Q10. J Am Coll Nutr 20: 591–598.
    1. Kunitomo M, Yamaguchi Y, Kagota S, Otsubo K (2008) Beneficial effect of coenzyme Q10 on increased oxidative and nitrative stress and inflammation and individual metabolic components developing in a rat model of metabolic syndrome. J Pharmacol Sci 107: 128–137.
    1. Tsai KL, Chen LH, Chiou SH, Chiou GY, Chen YC, et al. (2012) Coenzyme Q10 suppresses oxLDL-induced endothelial oxidative injuries by the modulation of LOX-1-mediated ROS generation via the AMPK/PKC/NADPH oxidase signaling pathway. Mol Nutr Food Res 55: S227–240.
    1. Milan G, Granzotto M, Scarda A, Calcagno A, Pagano C, et al. (2002) Resistin and adiponectin expression in visceral fat of obese rats: effect of weight loss. Obes Res 10: 1095–1103.
    1. Lu JY, Huang KC, Chang LC, Huang YS, Chi YC, et al. (2008) Adiponectin: a biomarker of obesity-induced insulin resistance in adipose tissue and beyond. J Biomed Sci 15: 565–576.
    1. Krssak M, Falk Petersen K, Dresner A, Di Pietro L, Vogel SM, et al. (1999) Intramyocellular lipid concentrations are correlated with insulin sensitivity in humans: a 1H NMR spectroscopy study. Diabetologia 42: 113–116.
    1. Shulman GI (2000) Cellular mechanisms of insulin resistance. J Clin Invest 106: 171–176.
    1. Kadowaki T, Yamauchi T (2005) Adiponectin and Adiponectin Receptors. Endocrine Reviews 26: 439–451.
    1. Yamauchi T, Nio Y, Maki T, Kobayashi M, Takazawa T, et al. (2007) Targeted disruption of AdipoR1 and AdipoR2 causes abrogation of adiponectin binding and metabolic actions. Nat Med 13: 332–339.
    1. Haider DG, Schaller G, Kapiotis S, Maier C, Luger A, et al. (2006) The release of the adipocytokine visfatin is regulated by glucose and insulin. Diabetologia 49: 1909–1914.
    1. Lee WJ, Wu CS, Lin H, Lee IT, Wu CM, et al... (2009) Visfatin-induced expression of inflammatory mediators in human endothelial cells through the NF-κB pathway. Int J Obes 33: 30 465–472.
    1. Fukuhara A, Matsuda M, Nishizawa M, Segawa K, Tanaka M, et al. (2005) Visfatin: a protein secreted by visceral fat that mimics the effects of insulin. Science 307: 426–430.
    1. Moschen AR, Kaser A, Enrich B, Mosheimer B, Theurl M, et al. (2007) Visfatin, an adipocytokine with proinflammatory and immunomodulating properties. J Immunol 178: 1748–1758.
    1. Esposito E, Impellizzeri D, Mazzon E, Fakhfouri G, Rahimian R, et al. (2012) The NAMPT inhibitor FK866 reverts the damage in spinal cord injury. J Neuroinflammation 9: 66.
    1. Kim SR, Bae YH, Bae SK, Choi KS, Yoon KH, et al. (2008) Visfatin enhances ICAM-1 and VCAM-1 expression through ROS-dependent NF-kappaB activation in endothelial cells. Biochim Biophys Acta 1783: 886–895.
    1. Vozarova B, Stefan N, Lindsay RS, Saremi A, Pratley RE, et al. (2002) High alanine aminotransferase is associated with decreased hepatic insulin sensitivity and predicts the development of type 2 diabetes. Diabetes 51: 1889–1895.

Source: PubMed

3
Subscribe