DOI QR코드

DOI QR Code

Anti-inflammatory effects of N-cyclooctyl-5-methylthiazol-2-amine hydrobromide on lipopolysaccharide-induced inflammatory response through attenuation of NLRP3 activation in microglial cells

  • Kim, Eun-A (Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine) ;
  • Hwang, Kyouk (Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine) ;
  • Kim, Ji-Eun (Department of Biomedical Laboratory Science, Konyang University) ;
  • Ahn, Jee-Yin (Department of Molecular Cell Biology and Single Cell Network Research Center, Sungkyunkwan University School of Medicine) ;
  • Choi, Soo Young (Department of Biomedical Science and Research Institute for Bioscience and Biotechnology, Hallym University) ;
  • Yang, Seung-Ju (Department of Biomedical Laboratory Science, Konyang University) ;
  • Cho, Sung-Woo (Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine)
  • Received : 2021.06.28
  • Accepted : 2021.07.22
  • Published : 2021.11.30

Abstract

Microglial activation is closely associated with neuroinflammatory pathologies. The nucleotide-binding and oligomerization domain-like receptor containing a pyrin domain 3 (NLRP3) inflammasomes are highly organized intracellular sensors of neuronal alarm signaling. NLRP3 inflammasomes activate nuclear factor kappa-B (NF-κB) and reactive oxygen species (ROS), which induce inflammatory responses. Moreover, NLRP3 dysfunction is a common feature of chronic inflammatory diseases. The present study investigated the effect of a novel thiazol derivative, N-cyclooctyl-5-methylthiazol-2-amine hydrobromide (KHG26700), on inflammatory responses in lipopolysaccharide (LPS)-treated BV-2 microglial cells. KHG26700 significantly attenuated the expression of several pro-inflammatory cytokines, including tumor necrosis factor-α, interleukin-1β, and interleukin-6, in these cells, as well as the LPS-induced increases in NLRP3, NF-κB, and phospho-IkBα levels. KHG26700 also suppressed the LPS-induced increases in protein levels of autophagy protein 5 (ATG5), microtubule-associated protein 1 light chain 3 (LC3), and beclin-1, as well as downregulating the LPS-enhanced levels of ROS, lipid peroxidation, and nitric oxide. These results suggest that the anti-inflammatory effects of KHG26700 may be due, at least in part, to the regulation of the NLRP3-mediated signaling pathway during microglial activation.

Keywords

Acknowledgement

This work was supported by the National Research Foundation of Korea (NRF) grant funded by the Korean government (MSIT) (2018R1A2B6001743 and 2021R1F1A1051920), and by a Student Research Grant from the University of Ulsan College of Medicine, Seoul, Korea.

References

  1. Lee CM, Lee DS, Jung WK et al (2016) Benzyl isothiocyanate inhibits inflammasome activation in E. coli LPSstimulated BV2 cells. Int Mol Med 38, 912-918 https://doi.org/10.3892/ijmm.2016.2667
  2. Budai MM, Varga A, Milesz S et al (2013) Aloe vera downregulates LPS-induced inflammatory cytokine production and expression of NLRP3 inflammasome in human macrophages. Mol Immunol 56, 471-479 https://doi.org/10.1016/j.molimm.2013.05.005
  3. inarello CA (2009) Immunological and inflammatory functions of the interleukin-1 family. Annu Rev Immunol 27, 519-550 https://doi.org/10.1146/annurev.immunol.021908.132612
  4. Jeng KCG, Hou RCW, Wang JC et al (2005) Sesamin inhibits lipopolysaccharide-induced cytokine production by suppression of p38 mitogen-activated protein kinase and nuclear factor-kappaB. Immunol Lett 97, 101-106 https://doi.org/10.1016/j.imlet.2004.10.004
  5. Fu YY, Zhang F, Zhang L et al (2014) Mangiferin regulates interleukin-6 and cystathionine-b-synthase in lipopolysaccharide-induced brain injury. Cell Mol Neurobiol 34, 651-657 https://doi.org/10.1007/s10571-014-0039-8
  6. De Nardo D and Latz E (2011) NLRP3 inflammasomes link inflammation and metabolic disease. Trends Immunol 32, 373-379 https://doi.org/10.1016/j.it.2011.05.004
  7. Yang F, Wang Z, Wei X et al (2014) NLRP3 deficiency ameliorates neurovascular damage in experimental ischemic stroke. J Cereb Blood Flow Metab 34, 660-667 https://doi.org/10.1038/jcbfm.2013.242
  8. Chen C, Wei YZ, He XM et al (2019) Naringenin produces neuroprotection against LPS-induced dopamine neurotoxicity via the inhibition of microglial NLRP3 inflammasome activation. Front Immunol 10, 936 https://doi.org/10.3389/fimmu.2019.00936
  9. Kim J, Kim SM, Na JM et al (2016) Protective effect of 3-(naphthalen-2-yl(propoxy)methyl)azetidine hydrochloride on hypoxia-induced toxicity by suppressing microglial activation in BV-2 cells. BMB Rep 49, 687-692 https://doi.org/10.5483/BMBRep.2016.49.12.169
  10. Kim EA, Na JM, Kim J et al (2017) Neuroprotective effect of 3-(naphthalen-2-yl(propoxy)methyl)azetidine hydrochloride on brain ischaemia/reperfusion injury. J Neuroimmune Pharmacol 12, 447-461 https://doi.org/10.1007/s11481-017-9733-x
  11. Kim EA, Kim H, Ahn JY et al (2010) Suppression of lipopolysaccharide-induced microglial activation by a benzothiazole derivative. Mol Cells 30, 51-57 https://doi.org/10.1007/s10059-010-0087-y
  12. Sorbara MT and Girardin SE (2011) Mitochondrial ROS fuel the inflammasome. Cell Res 21, 558-560 https://doi.org/10.1038/cr.2011.20
  13. Yang JW, Yang SJ, Na JM et al (2018) 3-(Naphthalen-2-yl (propoxy)methyl)azetidine hydrochloride attenuates NLRP3 inflammasome-mediated signaling pathway in lipopolysaccharide-stimulated BV2 microglial cells. Biochem Biophys Res Commun 495, 151-156 https://doi.org/10.1016/j.bbrc.2017.10.131
  14. Ha JS, Choi HR, Kim IS et al (2021) Hypoxia-induced S100A8 expression activates microglial inflammation and promotes neuronal apoptosis. Int J Mol Sci 22, 1205 https://doi.org/10.3390/ijms22031205
  15. Yang SJ, Han AR, Choi HR et al (2020) N-Adamantyl4-methylthiazol-2-amine suppresses glutamate-induced auto-phagic cell death via PI3K/Akt/mTOR signaling pathways in cortical neurons. BMB Rep 53, 527-532 https://doi.org/10.5483/BMBRep.2020.53.10.059
  16. Levine B, Mizushima N and Virgin HW (2011) Autophagy in immunity and inflammation. Nature 469, 323-335 https://doi.org/10.1038/nature09782
  17. Kim J, Kwak HJ, Cha JY et al (2014) Metformin suppresses lipopolysaccharide (LPS)-induced inflammatory response in murine macrophages via activating transcription factor-3 (ATF-3) induction. J Biol Chem 289, 23246-23255 https://doi.org/10.1074/jbc.M114.577908
  18. Kim H, Choi J, Ryu J et al (2009) Activation of autophagy during glutamate-induced HT22 cell death. Biochem Biophys Res Commun 388, 339-344 https://doi.org/10.1016/j.bbrc.2009.08.007
  19. Yin WY, Ye Q and Huang HJ (2016) Salidroside protects cortical neurons against glutamate-induced cytotoxicity by inhibiting autophagy. Mol Cell Biochem 419, 53-64 https://doi.org/10.1007/s11010-016-2749-3
  20. Kim EA, Han AR, Choi J et al (2014) Anti-inflammatory mechanisms of N-adamantyl-4-methylthiazol-2-amine in lipopolysaccharide-stimulated BV-2 microglial cells. Int Immunopharmacol 22, 73-83 https://doi.org/10.1016/j.intimp.2014.06.022
  21. Murad F (1994) The nitric oxide-cyclic GMP signal transduction system for intracellular and intercellular communication. Recent Prog Horm Res 49, 239-248
  22. Possel H, Noack H, Putzke J et al (2000) Selective upregulation of inducible nitric oxide synthase (iNOS) by lipopolysaccharide (LPS) and cytokines in microglia: in vitro and in vivo studies. Glia 32, 51-59 https://doi.org/10.1002/1098-1136(200010)32:1<51::AID-GLIA50>3.0.CO;2-4
  23. Chew LJ, Takanohashi A and Bell M (2006) Microglia and inflammation: impact on developmental brain injuries. Ment Retard Dev Disabil Res Rev 12, 105-112 https://doi.org/10.1002/mrdd.20102
  24. Cho CH, Kim EA, Kim J et al (2016) N-Adamantyl-4-methylthiazol-2-amine suppresses amyloid β-induced neuronal oxidative damage in cortical neurons. Free Radic Res 50, 678-690 https://doi.org/10.3109/10715762.2016.1167277
  25. Urabe T, Yamasaki Y, Hattori N et al (2000) Accumulation of 4-hydroxynonenal-modified proteins in hippocampal CA1 pyramidal neurons precedes delayed neuronal damage in the gerbil brain. Neuroscience 100, 241-250 https://doi.org/10.1016/S0306-4522(00)00264-5
  26. Ha SC, Han AR, Kim DW et al (2013) Neuroprotective effects of the antioxidant action of 2-cyclopropylimino-3-methyl-1,3-thiazoline hydrochloride against ischemic neuronal damage in the brain. BMB Rep 46, 370-375 https://doi.org/10.5483/BMBRep.2013.46.7.018
  27. Kim SJ, Cha JY, Kang HS et al (2016) Corosolic acid ameliorates acute inflammation through inhibition of IRAK-1 phosphorylation in macrophages. BMB Rep 49, 276-281 https://doi.org/10.5483/BMBRep.2016.49.5.241
  28. Lee KJ, Kim YK, Krupa M et al (2016) Crotamine stimulates phagocytic activity by inducing nitric oxide and TNF-α via p38 and NFκ-B signaling in RAW 264.7 macrophages. BMB Rep 49, 185-190 https://doi.org/10.5483/BMBRep.2016.49.3.271
  29. Kim H, Youn GS, An SY et al (2016) 2,3-Dimethoxy-2'-hydroxychalcone ameliorates TNF-α-induced ICAM-1 expression and subsequent monocyte adhesiveness via NF-kappaB inhibition and HO-1 induction in HaCaT cells. BMB Rep 49, 57-62 https://doi.org/10.5483/BMBRep.2016.49.1.141
  30. Kim EA, Choi J, Han AR et al (2013) Anti-oxidative and anti-inflammatory effects of 2-cyclopropylimino-3-methyl-1,3-thiazoline hydrochloride on glutamate-induced neurotoxicity in rat brain. Neurotoxicology 38, 106-114 https://doi.org/10.1016/j.neuro.2013.07.001
  31. Yang SJ, Lee WJ, Kim EA et al (2014) Effects of N-adamantyl-4-methylthiazol-2-amine on hyperglycemia, hyperlipidemia and oxidative stress in streptozotocin-induced diabetic rats. Eur J Pharmacol 736, 26-34 https://doi.org/10.1016/j.ejphar.2014.04.031
  32. Han AR, Yang JW, Na JM et al (2019) Protective effects of N,4,5-trimethylthiazol-2-amine hydrochloride on hypoxia-induced β-amyloid production in SH-SY5Y cells. BMB Rep 52, 439-444 https://doi.org/10.5483/bmbrep.2019.52.7.231