DOI QR코드

DOI QR Code

NRF2 activation by 2-methoxycinnamaldehyde attenuates inflammatory responses in macrophages via enhancing autophagy flux

  • Kim, Bo-Sung (Department of Korean Medical Science, School of Korean Medicine, Pusan National University) ;
  • Shin, Minwook (RNA Therapeutics Institute, University of Massachusetts Chan Medical School) ;
  • Kim, Kyu-Won (College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University) ;
  • Ha, Ki-Tae (Department of Korean Medical Science, School of Korean Medicine, Pusan National University) ;
  • Bae, Sung-Jin (Department of Molecular Biology and Immunology, Kosin University College of Medicine)
  • Received : 2022.04.11
  • Accepted : 2022.06.15
  • Published : 2022.08.31

Abstract

A well-controlled inflammatory response is crucial for the recovery from injury and maintenance of tissue homeostasis. The anti-inflammatory response of 2-methoxycinnamaldehyde (2-MCA), a natural compound derived from cinnamon, has been studied; however, the underlying mechanism on macrophage has not been fully elucidated. In this study, LPS-stimulated production of TNF-α and NO was reduced by 2-MCA in macrophages. 2-MCA significantly activated the NRF2 pathway, and expression levels of autophagy-associated proteins in macrophages, including LC3 and P62, were enhanced via NRF2 activation regardless of LPS treatment, suggesting the occurrence of 2-MCA-mediated autophagy. Moreover, evaluation of autophagy flux using luciferase-conjugated LC3 revealed that incremental LC3 and P62 levels are coupled to enhanced autophagy flux. Finally, reduced expression levels of TNF-α and NOS2 by 2-MCA were reversed by autophagy inhibitors, such as bafilomycin A1 and NH4Cl, in LPS-stimulated macrophages. In conclusion, 2-MCA enhances autophagy flux in macrophages via NRF2 activation and consequently reduces LPS-induced inflammation.

Keywords

Acknowledgement

M.S., S.-J.B. and K.-W.K. conceived and designed the research. M.S. and B.-S.K. performed the experiments and collected the data. B.-S.K. and S.-J.B. wrote the manuscript. M.S., K.-W.K. and K.-T.H. analyzed the data and revised the manuscript. K.-T.H. and S.-J.B. provided guidance on the study and supervised the project. All the authors reviewed the manuscript and agreed to the submission. This work was supported by the National Research Foundation of Korea (NRF) grants funded by the Korean government (MIST; NRF-2021R1A2B5B01002223 to Kyu-Won Kim, NRF-2020R1C1C1003703 to Sung-Jin Bae, and NRF-2021R1A4A1025662 to Sung-Jin Bae and Ki-Tae Ha).

References

  1. Medzhitov R (2008) Origin and physiological roles of inflammation. Nature 454, 428-435 https://doi.org/10.1038/nature07201
  2. Ley K, Laudanna C, Cybulsky MI and Nourshargh S (2007) Getting to the site of inflammation: the leukocyte adhesion cascade updated. Nat Rev Immunol 7, 678-689 https://doi.org/10.1038/nri2156
  3. Guzik TJ, Korbut R and Adamek-Guzik T (2003) Nitric oxide and superoxide in inflammation and immune regulation. J Physiol Pharmacol 54, 469-487
  4. Kalliolias GD and Ivashkiv LB (2016) TNF biology, pathogenic mechanisms and emerging therapeutic strategies. Nat Rev Rheumatol 12, 49-62 https://doi.org/10.1038/nrrheum.2015.169
  5. Rao PV and Gan SH (2014) Cinnamon: a multifaceted medicinal plant. Evid Based Complement Alternat Med 2014, 642942
  6. Kim NY, Trinh NT, Ahn SG and Kim SA (2020) Cinnamaldehyde protects against oxidative stress and inhibits the TNFalpha induced inflammatory response in human umbilical vein endothelial cells. Int J Mol Med 46, 449-457
  7. Tsuji-Naito K (2008) Aldehydic components of cinnamon bark extract suppresses RANKL-induced osteoclastogenesis through NFATc1 downregulation. Bioorg Med Chem 16, 9176-9183 https://doi.org/10.1016/j.bmc.2008.09.036
  8. Yamakawa D, Kidoya H, Sakimoto S, Jia W and Takakura N (2011) 2-Methoxycinnamaldehyde inhibits tumor angiogenesis by suppressing Tie2 activation. Biochem Biophys Res Commun 415, 174-180 https://doi.org/10.1016/j.bbrc.2011.10.053
  9. Tsai KD, Cherng J, Liu YH et al (2016) Cinnamomum verum component 2-methoxycinnamaldehyde: a novel antiproliferative drug inducing cell death through targeting both topoisomerase I and II in human colorectal adenocarcinoma COLO 205 cells. Food Nutr Res 60, 31607
  10. Kim SY, Koo YK, Koo JY et al (2010) Platelet anti-aggregation activities of compounds from Cinnamomum cassia. J Med Food 13, 1069-1074 https://doi.org/10.1089/jmf.2009.1365
  11. Kim GY, Jeong H, Yoon HY et al (2020) Anti-inflammatory mechanisms of suppressors of cytokine signaling target ROS via NRF-2/thioredoxin induction and inflammasome activation in macrophages. BMB Rep 53, 640-645 https://doi.org/10.5483/BMBRep.2020.53.12.161
  12. Ma Q (2013) Role of Nrf2 in oxidative stress and toxicity. Annu Rev Pharmacol Toxicol 53, 401-426 https://doi.org/10.1146/annurev-pharmtox-011112-140320
  13. Bellezza I, Giambanco I, Minelli A and Donato R (2018) Nrf2-Keap1 signaling in oxidative and reductive stress. Biochim Biophys Acta Mol Cell Res 1865, 721-733 https://doi.org/10.1016/j.bbamcr.2018.02.010
  14. Baird L and Yamamoto M (2020) The molecular mechanisms regulating the KEAP1-NRF2 pathway. Mol Cell Biol 40, e00099-00020
  15. Jain A, Lamark T, Sjottem E et al (2010) p62/SQSTM1 is a target gene for transcription factor NRF2 and creates a positive feedback loop by inducing antioxidant response element-driven gene transcription. J Biol Chem 285, 22576-22591 https://doi.org/10.1074/jbc.M110.118976
  16. Pankiv S, Clausen TH, Lamark T et al (2007) p62/SQSTM1 binds directly to Atg8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 282, 24131-24145 https://doi.org/10.1074/jbc.M702824200
  17. Hwa JS, Jin YC, Lee YS et al (2012) 2-methoxycinnamaldehyde from Cinnamomum cassia reduces rat myocardial ischemia and reperfusion injury in vivo due to HO-1 induction. J Ethnopharmacol 139, 605-615 https://doi.org/10.1016/j.jep.2011.12.001
  18. Jiang T, Harder B, Rojo de la Vega M, Wong PK, Chapman E and Zhang DD (2015) p62 links autophagy and Nrf2 signaling. Free Radic Biol Med 88, 199-204 https://doi.org/10.1016/j.freeradbiomed.2015.06.014
  19. Frias DP, Gomes RLN, Yoshizaki K et al (2020) Nrf2 positively regulates autophagy antioxidant response in human bronchial epithelial cells exposed to diesel exhaust particles. Sci Rep 10, 3704
  20. Palmieri M, Impey S, Kang H et al (2011) Characterization of the CLEAR network reveals an integrated control of cellular clearance pathways. Hum Mol Genet 20, 3852-3866 https://doi.org/10.1093/hmg/ddr306
  21. Park GT, Yoon JW, Yoo SB et al (2021) Echinochrome A treatment alleviates fibrosis and inflammation in bleomycin-induced scleroderma. Mar Drugs 19, 237
  22. Mao K, Chen S, Chen M et al (2013) Nitric oxide suppresses NLRP3 inflammasome activation and protects against LPS-induced septic shock. Cell Res 23, 201-212 https://doi.org/10.1038/cr.2013.6
  23. Choi H and Shin EC (2022) Hyper-inflammatory responses in COVID-19 and anti-inflammatory therapeutic approaches. BMB Rep 55, 11-19 https://doi.org/10.5483/BMBRep.2022.55.1.152
  24. Matsuura M (2013) Structural modifications of bacterial lipopolysaccharide that facilitate gram-negative bacteria evasion of host innate immunity. Front Immunol 4, 109
  25. Kwon DJ, Bae YS, Ju SM, Youn GS, Choi SY and Park J (2014) Salicortin suppresses lipopolysaccharide-stimulated inflammatory responses via blockade of NF-kappaB and JNK activation in RAW 264.7 macrophages. BMB Rep 47, 318-323 https://doi.org/10.5483/BMBRep.2014.47.6.200
  26. Harada K, Ohira S, Isse K et al (2003) Lipopolysaccharide activates nuclear factor-kappaB through toll-like receptors and related molecules in cultured biliary epithelial cells. Lab Invest 83, 1657-1667 https://doi.org/10.1097/01.LAB.0000097190.56734.FE
  27. Hattori Y, Hattori S and Kasai K (2003) Lipopolysaccharide activates Akt in vascular smooth muscle cells resulting in induction of inducible nitric oxide synthase through nuclear factor-kappa B activation. Eur J Pharmacol 481, 153-158 https://doi.org/10.1016/j.ejphar.2003.09.034
  28. Wink DA, Hines HB, Cheng RY et al (2011) Nitric oxide and redox mechanisms in the immune response. J Leukoc Biol 89, 873-891 https://doi.org/10.1189/jlb.1010550
  29. Parameswaran N and Patial S (2010) Tumor necrosis factor-alpha signaling in macrophages. Crit Rev Eukaryot Gene Expr 20, 87-103 https://doi.org/10.1615/CritRevEukarGeneExpr.v20.i2.10
  30. Gabay C, Lamacchia C and Palmer G (2010) IL-1 pathways in inflammation and human diseases. Nat Rev Rheumatol 6, 232-241 https://doi.org/10.1038/nrrheum.2010.4
  31. Chousterman BG, Swirski FK and Weber GF (2017) Cytokine storm and sepsis disease pathogenesis. Semin Immunopathol 39, 517-528 https://doi.org/10.1007/s00281-017-0639-8
  32. Titheradge MA (1999) Nitric oxide in septic shock. Biochim Biophys Acta 1411, 437-455 https://doi.org/10.1016/S0005-2728(99)00031-6
  33. Farkas T and Jaattela M (2017) Renilla luciferase-LC3 based reporter assay for measuring autophagic flux. Methods Enzymol 588, 1-13 https://doi.org/10.1016/bs.mie.2016.09.072
  34. Kim JE, Kalimuthu S and Ahn BC (2015) In vivo cell tracking with bioluminescence imaging. Nucl Med Mol Imaging 49, 3-10 https://doi.org/10.1007/s13139-014-0309-x
  35. Zeng M, Sang W, Chen S et al (2017) 4-PBA inhibits LPS-induced inflammation through regulating ER stress and autophagy in acute lung injury models. Toxicol Lett 271, 26-37 https://doi.org/10.1016/j.toxlet.2017.02.023
  36. Feng Y, Liu B, Zheng X, Chen L, Chen W and Fang Z (2019) The protective role of autophagy in sepsis. Microb Pathog 131, 106-111 https://doi.org/10.1016/j.micpath.2019.03.039