A newly synthesized flavone avoids COMT-catalyzed methylation and mitigates myocardial ischemia/reperfusion injury in H9C2 cells via JNK and P38 pathways

Document Type : Original Article

Authors

1 School of Medical and Health Engineering, Changzhou University, Changzhou 213164, P.R. China

2 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore

3 Food Science and Technology Program, Department of Chemistry, Faculty of Science, National University of Singapore, Singapore 117597, Singapore

4 National University of Singapore (Suzhou) Research Institute, Suzhou, Jiangsu 215123, P.R. China

5 Changzhou Second People’s Hospital, Changzhou 213000, P.R. China

Abstract

Objective(s): Luteolin is a flavone that provides defense against myocardial ischemia/reperfusion (I/R) injury. However, this compound is subjected to methylation mediated by catechol-O-methyltransferase (COMT), thus influencing its pharmacological effect. To synthesize a new flavone from luteolin that avoids COMT-catalyzed methylation and find out the protective mechanism of LUA in myocardial I/R injury.
Materials and Methods: Luteolin and 2,2’-azobis (2-amidinopropane) dihydrochloride (AAPH) were used to synthesize the new flavone known as LUAAPH-1 (LUA). Then, the myocardial ischemia/reperfusion injury cell model was established using H9c2 cells to detect the effect in myocardial ischemia/reperfusion regulation and to identify the underlying mechanism. 
Results: Pretreatment with LUA (20 μmol/l) substantially increased cell viability while reducing cell apoptosis rate and caspase-3 expression induced by I/R, and the protective effect of LUA on cell viability was stronger than diosmetin, which is the major methylated metabolite of luteolin. In addition, intracellular reactive oxygen species (ROS) production and calcium accumulation were both inhibited by LUA. Furthermore, we identified that LUA markedly relieved the promotive effects of I/R stimulation upon JNK and p38 phosphorylation.
Conclusion: LUT pretreatment conveys significant cardioprotective effects after myocardial I/R injury, and JNK and p38 MAPK signaling pathway may be involved.

Keywords

Main Subjects


1. Zhang CX, Cheng Y, Liu DZ, Liu M, Cui H, Zhang BL, et al. Mitochondria-targeted cyclosporin A delivery system to treat myocardial ischemia reperfusion injury of rats. J Nanobiotechnology 2019;17:18-23.
2. Lopez-Lazaro M. Distribution and biological activities of the flavonoid luteolin. Mini Rev Med Chem 2009; 9:31-59.
3. Wang IC, Lin JH, Lee WS, Liu CH, Lin TY, Yang KT. Baicalein and luteolin inhibit ischemia/reperfusion-induced ferroptosis in rat cardiomyocytes. Int J Cardiol 2023; 375:74-86.
4. Wei B, Lin Q, Ji YG, Zhao YC, Ding LN, Zhou WJ, et al. Luteolin ameliorates rat myocardial ischaemia-reperfusion injury through activation of peroxiredoxin II. Br J Pharmacol 2018; 175:3315-3332.
5. Hu Y, Zhang C, Zhu H, Wang S, Zhou Y, Zhao J, et al. Luteolin modulates SERCA2a via Sp1 upregulation to attenuate myocardial ischemia/reperfusion injury in mice. Sci Rep 2020; 10:15407-15418.
6. Wang L, Chen Q, Zhu L, Li Q, Zeng X, Lu L, et al. Metabolic disposition of luteolin is mediated by the interplay of udp-glucuronosyltransferases and catechol-o-methyltransferases in rats. Drug Metab Dispos 2017; 45:306-315.
7. Chen ZJ, Dai YQ, Kong SS, Song FF, Li LP, Ye JF, et al. Luteolin is a rare substrate of human catechol-O-methyltransferase favoring a para-methylation. Mol Nutr Food Res 2013; 57:877-885.
8. Chen Z, Tu M, Sun S, Kong S, Wang Y, Ye J, et al. The exposure of luteolin is much lower than that of apigenin in oral administration of Flos Chrysanthemi extract to rats. Drug Metab Pharmacokinet 2012; 27:162-168.
9. Ha SK, Lee JA, Cho EJ, Choi I. Effects of catechol O-methyl transferase inhibition on anti-inflammatory activity of luteolin metabolites. J Food Sci 2017; 82:545-552.
10. Ye L, Xin Y, Wu ZY, Sun HJ, Huang DJ, Sun ZQ. A newly synthesized flavone from luteolin escapes from COMT-Catalyzed methylation and inhibits lipopolysaccharide-induced inflammation in RAW264.7 macrophages via JNK, p38 and NF-kappaB signaling pathways. J Microbiol Biotechnol 2022; 32:15-26.
11. Zhai M, Li B, Duan W, Jing L, Zhang B, Zhang M, et al. Melatonin ameliorates myocardial ischemia reperfusion injury through SIRT3-dependent regulation of oxidative stress and apoptosis. J Pineal Res 2017; 63: e12419.
12. Binu P, Soman R, Zakhariah Hisham O, Narayanan SP, Nair RH. Acute promyelocytic leukemia drug - arsenic trioxide in the presence of eugenol shows differential action on leukemia cells (HL-60) and cardiomyocytes (H9c2) - inference from NMR study. Toxicol Mech Methods 2021; 31:457-466.
13. Chen Z, Chen M, Pan H, Sun S, Li L, Zeng S, et al. Role of catechol-O-methyltransferase in the disposition of luteolin in rats. Drug Metab Dispos 2011; 39:667-674.
14. Xu Y, Yang J, Lu Y, Qian LL, Yang ZY, Han RM, et al. Copper (II) coordination and translocation in luteolin and effect on radical scavenging. J Phys Chem B 2020; 124:380-388.
15. Horvathova K, Novotny L, Tothova D, Vachalkova A. Determination of free radical scavenging activity of quercetin, rutin, luteolin and apigenin in H2O2-treated human ML cells K562. Neoplasma 2004; 51:395-399.
16. Huang D, Ou B, Hampsch-Woodill M, Flanagan JA, Prior RL. High-throughput assay of oxygen radical absorbance capacity (ORAC) using a multichannel liquid handling system coupled with a microplate fluorescence reader in 96-well format. J Agric Food Chem 2002; 50:4437-4444.
17. Gonzalez-Montero J, Brito R, Gajardo AI, Rodrigo R. Myocardial reperfusion injury and oxidative stress: Therapeutic opportunities. World J Cardiol 2018; 10:74-86.
18. Chen Y, Liu C, Zhou P, Li J, Zhao X, Wang Y, et al. Coronary endothelium no-reflow injury is associated with ROS-modified mitochondrial fission through the JNK-Drp1 signaling pathway. Oxid Med Cell Longev 2021; 2021:6699516.
19. Yang JT, Wang J, Zhou XR, Xiao C, Lou YY, Tang LH, et al. Luteolin alleviates cardiac ischemia/reperfusion injury in the hypercholesterolemic rat via activating Akt/Nrf2 signaling. Naunyn Schmiedebergs Arch Pharmacol 2018; 391:719-728.
20. Zhao L, Zhou Z, Zhu C, Fu Z, Yu D. Luteolin alleviates myocardial ischemia reperfusion injury in rats via Siti1/NLRP3/NF-kappaB pathway. Int Immunopharmacol 2020; 85:106680.
21. Zhang RQ, Li DY, Xu TD, Zhu SS, Pan HJ, Fang F, et al. Anti-oxidative effect of luteolin pretreatment on simulated ischemia/reperfusion injury in cardiomyocyte and perfused rat heart. Chin J Integr Med 2017; 23:518-527.
22. Shi M, Wang J, Bi F, Bai Z. Diosmetin alleviates cerebral ischemia-reperfusion injury through Keap1-mediated Nrf2/ARE signaling pathway activation and NLRP3 inflammasome inhibition. Environ Toxicol 2022; 37:1529-1542.
23. Mei Z, Du L, Liu X, Chen X, Tian H, Deng Y, et al. Diosmetin alleviated cerebral ischemia/reperfusion injury in vivo and in vitro by inhibiting oxidative stress via the SIRT1/Nrf2 signaling pathway. Food Funct 2022; 13:198-212.
24. Li H, Xia Z, Chen Y, Qi D, Zheng H. Mechanism and therapies of oxidative stress-mediated cell death in ischemia reperfusion injury. Oxid Med Cell Longev 2018; 2018:2910643.
25. Pena E, Brito J, El Alam S, Siques P. Oxidative stress, kinase activity and inflammatory implications in right ventricular hypertrophy and heart failure under hypobaric hypoxia. Int J Mol Sci 2020; 21:6421-6437.
26. Wang X, Cao Y, Chen S, Lin J, Bian J, Huang D. Anti-inflammation activity of flavones and their structure-activity relationship. J Agric Food Chem 2021; 69:7285-7302.
27. Matsuda H, Morikawa T, Ando S, Toguchida I, Yoshikawa M. Structural requirements of flavonoids for nitric oxide production inhibitory activity and mechanism of action. Bioorg Med Chem 2003; 11:1995-2000.
28. Du Y, Liu P, Xu T, Pan D, Zhu H, Zhai N, et al. Luteolin modulates SERCA2a leading to attenuation of myocardial ischemia/ reperfusion injury via sumoylation at lysine 585 in mice. Cell Physiol Biochem 2018; 45:883-898.
29. Zhu S, Xu T, Luo Y, Zhang Y, Xuan H, Ma Y, et al. Luteolin enhances sarcoplasmic reticulum Ca2+-ATPase activity through p38 MAPK signaling thus Improving rat cardiac function after ischemia/reperfusion. Cell Physiol Biochem 2017; 41:999-1010.
30. Zhu H, Jin Q, Li Y, Ma Q, Wang J, Li D, et al. Melatonin protected cardiac microvascular endothelial cells against oxidative stress injury via suppression of IP3R-[Ca (2+)]c/VDAC-[Ca (2+)]m axis by activation of MAPK/ERK signaling pathway. Cell Stress Chaperones 2018; 23:101-113.
31. Ma L, Zou R, Shi W, Zhou N, Chen S, Zhou H, et al. SGLT2 inhibitor dapagliflozin reduces endothelial dysfunction and microvascular damage during cardiac ischemia/reperfusion injury through normalizing the XO-SERCA2-CaMKII-coffilin pathways. Theranostics 2022; 12:5034-5050.
32. Li J, Miller EJ, Ninomiya-Tsuji J, Russell RR, 3rd, Young LH. AMP-activated protein kinase activates p38 mitogen-activated protein kinase by increasing recruitment of p38 MAPK to TAB1 in the ischemic heart. Circ Res 2005; 97:872-879.
33. Yu D, Li M, Tian Y, Liu J, Shang J. Luteolin inhibits ROS-activated MAPK pathway in myocardial ischemia/reperfusion injury. Life Sci 2015; 122:15-25.