PDF
Abstract
Background: The aim was to elucidate the function of IL-37 in middle east respiratory syndrome coronavirus (MERS-CoV) infection, thereby providing a novel therapeutic strategy for managing the clinical treatment of inflammatory response caused by respiratory virus infection.
Methods: We investigated the development of MERS by infecting hDPP4 mice with hCoV-EMC (107 TCID50 [50% tissue culture infectious dose]) intranasally. We infected A549 cells with MERS-CoV, which concurrently interfered with IL-37, detecting the viral titer, viral load, and cytokine expression at certain points postinfection. Meanwhile, we administered IL-37 (12.5 μg/kg) intravenously to hDPP4 mice 2 h after MERS-CoV-2 infection and collected the serum and lungs 5 days after infection to investigate the efficacy of IL-37 in MERS-CoV infection.
Results: The viral titer of MERS-CoV-infected A549 cells interfering with IL-37 was significantly reduced by 4.7-fold, and the viral load of MERS-CoV-infected hDPP4 mice was decreased by 59-fold in lung tissue. Furthermore, the administration of IL-37 suppressed inflammatory cytokine and chemokine (monocyte chemoattractant protein 1, interferon-γ, and IL-17A) expression and ameliorated the infiltration of inflammatory cells in hDPP4 mice.
Conclusion: IL-37 exhibits protective properties in severe pneumonia induced by MERS-CoV infection. This effect is achieved through attenuation of lung viral load, suppression of inflammatory cytokine secretion, reduction in inflammatory cell infiltration, and mitigation of pulmonary injury.
Keywords
anti-inflammation
/
interleukin-37
/
MERS-CoV
/
pneumonia
Cite this article
Download citation ▾
Feifei Qi, Yiwei Yan, Qi Lv, Mingya Liu, Ming Liu, Fengdi Li, Ran Deng, Xujian Liang, Shuyue Li, Guocui Mou, Linlin Bao.
IL-37 possesses both anti-inflammatory and antiviral effects against Middle East respiratory syndrome coronavirus infection.
Animal Models and Experimental Medicine, 2025, 8(3): 483-492 DOI:10.1002/ame2.12435
| [1] |
http://www.who.int/emergencies/mers-cov/en/
|
| [2] |
Chan JF, Chan KH, Kao RY, et al. Broad-spectrum antivirals for the emerging Middle East respiratory syndrome coronavirus. J Infect. 2013; 67(6): 606-616.
|
| [3] |
Chan JF, Yao Y, Yeung ML, et al. Treatment with lopinavir/ritonavir or interferon-β1b improves outcome of MERS-CoV infection in a nonhuman primate model of common marmoset. J Infect Dis. 2015; 212(12): 1904-1913.
|
| [4] |
Falzarano D, de Wit E, Rasmussen AL, et al. Treatment with interferon-α2b and ribavirin improves outcome in MERS-CoV-infected rhesus macaques. Nat Med. 2013; 19(10): 1313-1317.
|
| [5] |
Arabi YM, Asiri AY, Assiri AM, et al. Interferon Beta-1b and lopinavir-ritonavir for Middle East respiratory syndrome. N Engl J Med. 2020; 383(17): 1645-1656.
|
| [6] |
Ma D, Wang X, Li M, Hu C, Tang L. Reconsideration of interferon treatment for viral diseases: lessons from SARS, MERS, and COVID-19. Int Immunopharmacol. 2023; 121: 110485.
|
| [7] |
Al-Tawfiq JA, Momattin H, Dib J, Memish ZA. Ribavirin and interferon therapy in patients infected with the Middle East respiratory syndrome coronavirus: an observational study. Int J Infect Dis. 2014; 20: 42-46.
|
| [8] |
Omrani AS, Saad MM, Baig K, et al. Ribavirin and interferon alfa-2a for severe Middle East respiratory syndrome coronavirus infection: a retrospective cohort study. Lancet Infect Dis. 2014; 14(11): 1090-1095.
|
| [9] |
Kim ES, Choe PG, Park WB, et al. Clinical progression and cytokine profiles of Middle East respiratory syndrome coronavirus infection. J Korean Med Sci. 2016; 31(11): 1717-1725.
|
| [10] |
Mubarak A, Alrfaei B, Aljurayyan A, et al. In vivo and in vitro evaluation of cytokine expression profiles during Middle East respiratory syndrome coronavirus (MERS-CoV) infection. J Inflamm Res. 2021; 14: 2121-2131.
|
| [11] |
Alosaimi B, Hamed ME, Naeem A, et al. MERS-CoV infection is associated with downregulation of genes encoding Th1 and Th2 cytokines/chemokines and elevated inflammatory innate immune response in the lower respiratory tract. Cytokine. 2020; 126: 154895.
|
| [12] |
Kim SY, Park SJ, Cho SY, et al. Viral RNA in blood as indicator of severe outcome in Middle East respiratory syndrome coronavirus infection. Emerg Infect Dis. 2016; 22(10): 1813-1816.
|
| [13] |
Alhetheel A, Albarrag A, Shakoor Z, Somily A, Barry M, Altalhi H. Bakhrebah, ls among patients with Middle East respiratory syndrome coronavirus infection. Vaccine. 2023; 11(6): 1048.
|
| [14] |
Min CK, Cheon S, Ha NY, et al. Comparative and kinetic analysis of viral shedding and immunological responses in MERS patients representing a broad spectrum of disease severity. Sci Rep. 2016; 6: 25359.
|
| [15] |
Huang KJ, Su IJ, Theron M, et al. An interferon-gamma-related cytokine storm in SARS patients. J Med Virol. 2005; 75(2): 185-194.
|
| [16] |
Chen N, Zhou M, Dong X, et al. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet (London, England). 2020; 395(10223): 507-513.
|
| [17] |
Yin SW, Zhou Z, Wang JL, Deng YF, Jing H, Qiu Y. Viral loads, lymphocyte subsets and cytokines in asymptomatic, mildly and critical symptomatic patients with SARS-CoV-2 infection: a retrospective study. Virol J. 2021; 18(1): 126.
|
| [18] |
Cui L, Qin X, Fu T, et al. Attenuated airways inflammation and remodeling in IL-37a and IL-37b transgenic mice with an ovalbumin-induced chronic asthma. Cell Immunol. 2023; 391-392: 104759.
|
| [19] |
Xiao S, Song X, Zheng M, et al. Interleukin-37 ameliorates atherosclerosis by regulating autophagy-mediated endothelial cell apoptosis and inflammation. Int Immunopharmacol. 2023; 118: 110098.
|
| [20] |
Qi F, Liu M, Li F, et al. Interleukin-37 ameliorates influenza pneumonia by attenuating macrophage cytokine production in a MAPK-dependent manner. Front Microbiol. 2019; 10: 2482.
|
| [21] |
Xu J, Chen J, Li W, et al. Additive therapeutic effects of mesenchymal stem cells and IL-37 for systemic lupus erythematosus. J Am Soc Nephrol. 2020; 31(1): 54-65.
|
| [22] |
Cavalli G, Koenders M, Kalabokis V, et al. Treating experimental arthritis with the innate immune inhibitor interleukin-37 reduces joint and systemic inflammation. Rheumatology (Oxford). 2017; 56(12): 2256.
|
| [23] |
An B, Liu X, Li G, Yuan H. Interleukin-37 ameliorates coxsackievirus B3-induced viral myocarditis by modulating the Th17/regulatory T cell immune response. J Cardiovasc Pharmacol. 2017; 69(5): 305-313.
|
| [24] |
Li A, Ling Y, Song Z, et al. Correlation between early plasma interleukin 37 responses with low inflammatory cytokine levels and benign clinical outcomes in severe acute respiratory syndrome coronavirus 2 infection. J Infect Dis. 2021; 223(4): 568-580.
|
| [25] |
Gong S, Qi F, Li F, et al. Human-derived a/Guangdong/Th005/2017 (H7N9) exhibits extremely high replication in the lungs of ferrets and is highly pathogenic in chickens. Viruses. 2019; 11(6): 494.
|
| [26] |
Zhang Y, Lv Q, Qi F, et al. Comparison of the replication and neutralization of different SARS-CoV-2 omicron subvariants in vitro. Animal Model Exp Med. 2023; 6(1): 51-56.
|
| [27] |
Reed J, Muench H. A simple method of estimating fifty per cent endpoint. Am J Epidemiol. 1938; 27(3): 493-497.
|
| [28] |
Sethi GS, Gracias DT, Gupta RK, et al. Anti-CD3 inhibits circulatory and tissue-resident memory CD4 T cells that drive asthma exacerbations in mice. Allergy. 2023; 78(8): 2168-2180.
|
| [29] |
Jiang T, Sheng J, Qin Y, et al. Enhanced proliferation and defective activation-induced cell death of CD4+ T cells in childhood asthma. Asian Pac J Allergy Immunol. 2014; 32(1): 75-83.
|
| [30] |
Schaller MA, Lundy SK, Huffnagle GB, Lukacs NW. CD8+ T cell contributions to allergen induced pulmonary inflammation and airway hyperreactivity. Eur J Immunol. 2005; 35(7): 2061-2070.
|
| [31] |
O'Sullivan S, Cormican L, Faul JL, et al. Activated, cytotoxic CD8+ T lymphocytes contribute to the pathology of asthma death. Am J Respir Crit Care Med. 2001; 164(4): 560-564.
|
| [32] |
Coleman CM, Sisk JM, Halasz G, et al. CD8+ T cells and macrophages regulate pathogenesis in a mouse model of Middle East respiratory syndrome. J Virol. 2016; 91(1): e01825-16.
|
| [33] |
Zhao L, Yang W, Yang X, et al. Chemerin suppresses murine allergic asthma by inhibiting CCL2 production and subsequent airway recruitment of inflammatory dendritic cells. Allergy. 2014; 69(6): 763-774.
|
| [34] |
Dong J, Wong CK, Cai Z, Jiao D, Chu M, Lam CW. Amelioration of allergic airway inflammation in mice by regulatory IL-35 through dampening inflammatory dendritic cells. Allergy. 2015; 70(8): 921-932.
|
| [35] |
Zhu Y, Yu J, Zhu X, et al. Experimental observation of the effect of immunotherapy on CD4+ T cells and Th1/Th2 cytokines in mice with allergic rhinitis. Sci Rep. 2023; 13(1): 5273.
|
| [36] |
Glück J, Rogala B, Rogala E, Oleś E. Allergen immunotherapy in intermittent allergic rhinitis reduces the intracellular expression of IL-4 by CD8+ T cells. Vaccine. 2007; 26(1): 77-81.
|
| [37] |
Wu W, Wang W, Wang Y, et al. IL-37b suppresses T cell priming by modulating dendritic cell maturation and cytokine production via dampening ERK/NF-κB/S6K signalings. Acta Biochim Biophys Sin. 2015; 47(8): 597-603.
|
| [38] |
Luo Y, Cai X, Liu S, et al. Suppression of antigen-specific adaptive immunity by IL-37 via induction of tolerogenic dendritic cells. Proc Natl Acad Sci USA. 2014; 111(42): 15178-15183.
|
| [39] |
Landolina N, Mariotti FR, Pelosi A, et al. The anti-inflammatory cytokine IL-37 improves the NK cell-mediated anti-tumor response. Onco Targets Ther. 2023; 13(1): 2297504.
|
| [40] |
Hou T, Sun X, Zhu J, et al. IL-37 ameliorating allergic inflammation in atopic dermatitis through regulating microbiota and AMPK-mTOR signaling pathway-modulated autophagy mechanism. Front Immunol. 2020; 11: 752.
|
| [41] |
Cao J, Hou S, Chen Z, et al. Interleukin-37 relieves PM2.5-triggered lung injury by inhibiting autophagy through the AKT/mTOR signaling pathway in vivo and in vitro. Ecotoxicol Environ Saf. 2024; 269: 115816.
|
| [42] |
Verma G, Dhawan M, Saied AA, Kaur G, Kumar R, Emran TB. Immunomodulatory approaches in managing lung inflammation in COVID-19: a double-edge sword. Immun Inflamm Dis. 2023; 11(9): e1020.
|
| [43] |
Li L, Li J, Li S, Chen H, Wu Y, Qiu Y. IL-37 alleviates alveolar bone resorption and inflammatory response through the NF-κB/NLRP3 signaling pathway in male mice with periodontitis. Arch Oral Biol. 2023; 147: 105629.
|
RIGHTS & PERMISSIONS
2024 The Author(s). Animal Models and Experimental Medicine published by John Wiley & Sons Australia, Ltd on behalf of The Chinese Association for Laboratory Animal Sciences.