Monascin ameliorate inflammation in the lipopolysaccharide-induced BV-2 microglial cells via suppressing the NF-κB/p65 pathway

Document Type : Original Article

Authors

Department of Orthopedics Surgery, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, 310009, Zhejiang, People’s Republic of China

Abstract

Objective(s): The pathophysiology of neurodegenerative diseases is complicated, in which inflammatory reactions play a vital role. Microglia cells activation, an essential process of neuroinflammation, can produce neurotoxic molecules and neurotrophic factors, which aggravate inflammation and neuronal injury. Monascin, a major component of red yeast rice, is an azaphilonoid pigment with potential anti-inflammatory effects; however, the effects in central nervous system have not been evaluated. Our goal in this project was to explore the therapeutic effect and the underlying mechanism of Monascin, which may be via anti-inflammatory action.
Materials and Methods: We used lipopolysaccharide to induce BV-2 microglial cells in order to form an inflammation model in vitro. The anti-inflammatory effects of Monascin were measured by enzyme-linked immunosorbent assay (ELISA), real time-polymerase chain reaction (RT-PCR), Western Blot and Immunofluorescent staining.
Results: Our data indicated that inflammatory cytokines including interleukin-1β (IL-1β), IL-6, tumor necrosis factor-alpha (TNF-α) and nitric oxide were suppressed by Monascin treatment. Furthermore, the related pro-inflammatory genes were inhibited consistent with the results of ELISA assay. Western blotting results showed that the phosphorylation of nuclear factor kappa B (NF-κB/p65) was reduced by Monascin treatment may be through suppressing the activation of IκB. Furthermore, immunofluorescence staining showed that the translocation of NF-κB/p65 to the cellular nuclear was blockaded after Monascin treatment.
Conclusion: Taken together, Monascin exerts anti-inflammatory effect and suppressed microglia activation, which suggested its potential therapeutic effect for inflammation-related diseases.

Keywords


1. Davies CL,  Patir A,  McColl BW. Myeloid Cell and Transcriptome Signatures Associated With Inflammation Resolution in a Model of Self-Limiting Acute Brain Inflammation. Front Immunol. 2019;10:1048.
2. Schropp V, Rohde J, Rovituso DM, Jabari S, Bharti R, Kuerten S. Contribution of LTi and T17 cells to B cell aggregate formation in the central nervous system in a mouse model of multiple sclerosis. J Neuroinflammation 2019; 16:111.
3. Glass CK, Saijo K, Winner B, Marchetto MC, Gage FH. Mechanisms underlying inflammation in neurodegeneration. Cell 2010; 140:918-34.
4. Block ML, Hong JS. Microglia and inflammation-mediated neurodegeneration: multiple triggers with a common mechanism. Prog Neurobiol 2005; 76:77-98.
5. Wang C, Wang Q, Lou Y, Xu J, Feng Z, Chen Y, et al. Salidroside attenuates neuroinflammation and improves functional recovery after spinal cord injury through microglia polarization regulation. J Cell Mol Med 2018; 02:22
6. Bhalala US, Koehler RC, Kannan S. Neuroinflammation and neuroimmune dysregulation after acute hypoxic-ischemic injury of developing brain. Frontiers in Pediatrics 2015; 2:144.
7. Fudong, Louise, McCullough. Inflammatory responses in hypoxic ischemic encephalopathy. Acta Pharmacol Sin 2013; 34:1121-30.
8. Shukla V, Shakya AK, Perez-Pinzon MA, Dave KR. Cerebral ischemic damage in diabetes: an inflammatory perspective. J Neuroinflammation 2017; 14:21.
9. Vidale S, Consoli A, Arnaboldi M, Consoli D. Postischemic Inflammation in Acute Stroke. J Clin Neurol 2017; 13:1-9.
10. Tak PP, Firestein GS. NF-κB: a key role in inflammatory diseases. The Journal of clinical investigation 2001; 107:7-11.
11. Shih VF, Tsui R, Caldwell A, Hoffmann A. A single NFκB system for both canonical and non-canonical signaling. Cell Res 2011; 21:86-102.
12. Luo Q, Yan X, Bobrovskaya L, Ji M, Yuan H, Lou H, et al. Anti-neuroinflammatory effects of grossamide from hemp seed via suppression of TLR-4-mediated NF-κB signaling pathways in lipopolysaccharide-stimulated BV2 microglia cells. Mol Cell Biochem 2017; 428:129-37.
13. Fan H, Wu PF, Zhang L, Hu ZL, Wang W, Guan XL, et al. Methionine sulfoxide reductase A negatively controls microglia-mediated neuroinflammation via inhibiting ROS/MAPKs/NF-κB signaling pathways through a catalytic antioxidant function. Antioxidants & redox signaling 2015; 22:832-47.
14. Xu J, Zhou L, Ji L, Chen F, Fortmann K, Zhang K, et al. The REG [gamma]-proteasome forms a regulatory circuit with I [kappa] B [epsiv] and NF [kappa] B in experimental colitis. Nature communications 2016; 7.
15. Puneet P, Yap CT, Wong L, Yulin L, Koh DR, Moochhala S, et al. SphK1 regulates proinflammatory responses associated with endotoxin and polymicrobial sepsis. Science 2010; 328:1290-4.
16. Chang YY, Hsu WH, Pan TM. Monascus secondary metabolites monascin and ankaflavin inhibit activation of RBL-2H3 cells. J Agric Food Chem 2015; 63:192-9.
17. Hsu LC, Liang YH, Hsu YW, Kuo YH, Pan TM,. Anti-inflammatory Properties of Yellow and Orange Pigments from Monascus purpureus NTU 568. J Agric Food Chem 2013; 61:2796-802.
18. Hsu WH, Chen TH, Lee BH, Hsu YW, Pan TM. Monascin and ankaflavin act as natural AMPK activators with PPARα agonist activity to down-regulate nonalcoholic steatohepatitis in high-fat diet-fed C57BL/6 mice. Food & Chemical Toxicology An International Journal Published for the British Industrial Biological Research Association 2014; 64:94-103.
19. Hsu WH, Lee BH, Pan TM. Monascin attenuates oxidative stress-mediated lung inflammation via peroxisome proliferator-activated receptor-gamma (PPAR-γ) and nuclear factor-erythroid 2 related factor 2 (Nrf-2) modulation. J Agric Food Chem 2014; 62:5337-44.
20. Hsu WH, Lu SS, Lee BH, Hsu YW, Pan TM. Monacolin K and monascin attenuated pancreas impairment and hyperglycemia induced by advanced glycation endproducts in BALB/c mice. Food Funct 2013; 4:1742-50.
21. Hsu WH, Pan TM. A novel PPARgamma agonist monascin’s potential application in diabetes prevention. Food Funct 2014; 5:1334-40.
22. Hsu WH, Lee BH, Liao TH, Hsu YW, Pan TM. Monascus -fermented metabolite monascin suppresses inflammation via PPAR-γ regulation and JNK inactivation in THP-1 monocytes. Food Chem Toxicol 2012; 50:1178-86.
23. Zheng G, Zhan Y, Tang Q, Chen T, Zheng F, Wang H, et al. Monascin inhibits IL-1β induced catabolism in mouse chondrocytes and ameliorates murine osteoarthritis. Food Funct 2018; 9: 1454-1464
24. Hsu WH, Lee BH, Lu IJ, Pan TM. Ankaflavin and monascin regulate endothelial adhesion molecules and endothelial NO synthase (eNOS) expression induced by tumor necrosis factor-α (TNF-α) in human umbilical vein endothelial cells (HUVECs). J Agric Food Chem 2012; 60:1666-72.
25. Wang C, Wang M, Xu T, Zhang X, Lin C, Gao W, et al. Engineering Bioactive Self-Healing Antibacterial Exosomes Hydrogel for Promoting Chronic Diabetic Wound Healing and Complete Skin Regeneration. Theranostics 2019; 9:65-76.
26. Kim BW, Koppula S, Hong SS, Jeon SB, Kwon JH, Hwang BY, et al. Regulation of microglia activity by glaucocalyxin-A: attenuation of lipopolysaccharide-stimulated neuroinflammation through NF-κB and p38 MAPK signaling pathways. PLoS One 2013; 8:e55792.
27. Rivest S. Molecular insights on the cerebral innate immune system. Brain Behavior & Immunity 2003; 17:13-9.
28. Michal S, Ravid S. Systemic inflammatory cells fight off neurodegenerative disease. Nature Reviews Neurology 2010; 6:405-10.
29. Gebicke-Haerter PJ. Microglia in neurodegeneration: molecular aspects. Microsc Res Tech 2010; 54:47.
30. Liang X, Wu L, Wang Q, Hand T, Bilak M, McCullough L, et al. Function of COX-2 and prostaglandins in neurological disease. J Mol Neurosci 2007; 33:94-9.
31. Wang W, Ji P, Dow KE. Corticotropin-releasing hormone induces proliferation and TNF-alpha release in cultured rat microglia via MAP kinase signalling pathways. J Neurochem 2003; 84:189-95.
32. Dheen S, Kaur C, Ea. Microglial activation and its implications in the brain diseases. Curr Med Chem 2007; 14.
33. Rothwell N. Interleukin-1 and neuronal injury: mechanisms, modification, and therapeutic potential. Brain Behavior & Immunity 2003; 17:152-7.
34. Allagnat F, Fukaya M, Nogueira TC, Delaroche D, Welsh N, Marselli L, et al. C/EBP homologous protein contributes to cytokine-induced pro-inflammatory responses and apoptosis in β-cells. Cell Death Differ 2012; 19:1836-46.
35. Lim JY, Won TJ, Hwang BY, Kim HR, Hwang KW, Sul D, et al. The new diterpene isodojaponin D inhibited LPS-induced microglial activation through NF-kappaB and MAPK signaling pathways. Eur J Pharmacol 2010; 642:10-8.
36. Giovannini M, Scali C, Prosperi C, Bellucci A, Pepeu G, Casamenti F. Experimental brain inflammation and neurodegeneration as model of Alzheimer’s disease: protective effects of selective COX-2 inhibitors. Int J Immunopathol Pharmacol 2003; 16:31-40.
37. Miller RL, James-Kracke M, Sun GY, Sun AY. Oxidative and inflammatory pathways in Parkinson’s disease. Neurochem Res 2009; 34:55-65.
38. Spooren A, Kooijman R, Lintermans B, Van Craenenbroeck K, Vermeulen L, Haegeman G, et al. Cooperation of NFκB and CREB to induce synergistic IL-6 expression in astrocytes. Cell Signal 2010; 22:871-81.
39. Vaillancourt F, Morquette B, Shi Q, Fahmi H, Lavigne P, Di Battista JA, et al. Differential regulation of cyclooxygenase‐2 and inducible nitric oxide synthase by 4‐hydroxynonenal in human osteoarthritic chondrocytes through ATF‐2/CREB‐1 transactivation and concomitant inhibition of NF‐κB signaling cascade. J Cell Biochem 2007; 100:1217-31.