Non-steroidal anti-inflammatory drugs dampen the cytokine and antibody response to SARS-CoV-2 infection

Jennifer S Chen, Mia Madel Alfajaro, Ryan D Chow, Jin Wei, Renata B Filler, Stephanie C Eisenbarth, Craig B Wilen, Jennifer S Chen, Mia Madel Alfajaro, Ryan D Chow, Jin Wei, Renata B Filler, Stephanie C Eisenbarth, Craig B Wilen

Abstract

Identifying drugs that regulate severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and its symptoms has been a pressing area of investigation during the coronavirus disease 2019 (COVID-19) pandemic. Nonsteroidal anti-inflammatory drugs (NSAIDs), which are frequently used for the relief of pain and inflammation, could modulate both SARS-CoV-2 infection and the host response to the virus. NSAIDs inhibit the enzymes cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2), which mediate the production of prostaglandins (PGs). As PGs play diverse biological roles in homeostasis and inflammatory responses, inhibiting PG production with NSAIDs could affect COVID-19 pathogenesis in multiple ways, including: (1) altering susceptibility to infection by modifying expression of angiotensin-converting enzyme 2 (ACE2), the cell entry receptor for SARS-CoV-2; (2) regulating replication of SARS-CoV-2 in host cells; and (3) modulating the immune response to SARS-CoV-2. Here, we investigate these potential roles. We demonstrate that SARS-CoV-2 infection upregulates COX-2 in diverse human cell culture and mouse systems. However, suppression of COX-2 by two commonly used NSAIDs, ibuprofen and meloxicam, had no effect on ACE2 expression, viral entry, or viral replication. In contrast, in a mouse model of SARS-CoV-2 infection, NSAID treatment reduced production of pro-inflammatory cytokines and impaired the humoral immune response to SARS-CoV-2 as demonstrated by reduced neutralizing antibody titers. Our findings indicate that NSAID treatment may influence COVID-19 outcomes by dampening the inflammatory response and production of protective antibodies rather than modifying susceptibility to infection or viral replication.ImportancePublic health officials have raised concerns about the use of nonsteroidal anti-inflammatory drugs (NSAIDs) for treating symptoms of coronavirus disease 2019 (COVID-19). NSAIDs inhibit the enzymes cyclooxygenase-1 (COX-1) and cyclooxygenase-2 (COX-2), which are critical for the generation of prostaglandins - lipid molecules with diverse roles in homeostasis and inflammation. Inhibition of prostaglandin production by NSAIDs could therefore have multiple effects on COVID-19 pathogenesis. Here, we demonstrate that NSAID treatment reduced both the antibody and pro-inflammatory cytokine response to SARS-CoV-2 infection. The ability of NSAIDs to modulate the immune response to SARS-CoV-2 infection has important implications for COVID-19 pathogenesis in patients. Whether this occurs in humans and whether it is beneficial or detrimental to the host remains an important area of future investigation. This also raises the possibility that NSAIDs may alter the immune response to SARS-CoV-2 vaccination.

Copyright © 2021, American Society for Microbiology.

Figures

FIG 1
FIG 1
SARS-CoV-2 infection induces PTGS2 expression in human cells and mice. (A) Calu-3 cells were infected with SARS-CoV-2 at an MOI of 0.05. PTGS2 expression was measured at 2 dpi, normalized to ACTB. (B and C) PTGS2 expression in Calu-3 (B) and ACE2-overexpressing A549 (A549-ACE2) (C) cells following SARS-CoV-2 infection. The data are from GSE147507 (26). (D) Huh7.5 cells were infected with SARS-CoV-2 at an MOI of 0.05. PTGS2 expression was measured at 2 dpi, normalized to ACTB. (E) HBECs were cultured at an air-liquid interface and then infected at the apical surface with 104 PFU of SARS-CoV-2. Cells were collected at 1, 2, and 3 dpi for single-cell RNA sequencing (scRNA-seq) (28). A volcano plot of differentially expressed genes in infected versus bystander ciliated cells pooled from all time points is shown. PTGS2 is highlighted. (F) K18-hACE2 mice were infected intranasally with 1.2 × 106 PFU of SARS-CoV-2. Ptgs2 expression in the lung was measured at 0, 2, 4, and 7 dpi. (G) Ptgs2 expression in the lung of K18-hACE2 mice following intranasal SARS-CoV-2 infection. The data are from GSE154104 (36). All data points in this figure are presented as means ± the standard errors of the mean (SEM). Data were analyzed by Welch’s two-tailed, unpaired t test (A, D, and F); Student two-tailed, unpaired t test (B, C, and G); and two-sided Mann-Whitney U test with continuity and Benjamini-Hochberg correction (E). *, P < 0.05; ***, P < 0.001; ****, P < 0.0001. Data in panels A and D are representative of two independent experiments with three replicates per condition.
FIG 2
FIG 2
NSAID treatment does not affect ACE2 expression in human cells and mice. (A and B) Calu-3 (A) and Huh7.5 (B) cells were treated with different concentrations of ibuprofen or meloxicam for 48 h. Cell viability was measured and calculated as a percentage of no treatment. (C) Calu-3 cells were treated with DMSO, 50 μM ibuprofen, or 50 μM meloxicam for 48 h. The levels of prostaglandin E2 (PGE2) were measured in the supernatant. The dotted line represents the limit of detection. (D and E) Calu-3 (D) and Huh7.5 (E) cells were treated with DMSO, 50 μM ibuprofen, or 50 μM meloxicam for 24 h. ACE2 expression was measured and normalized to ACTB. (F to I) C57BL/6 mice were treated intraperitoneally with DMSO, 30 mg/kg ibuprofen, or 1 mg/kg meloxicam daily for 4 days. Ace2 expression was measured in the lung (F), heart (G), kidney (H), and ileum (I), normalized to Actb. All data points in this figure are presented as means ± the SEM. Data were analyzed by Welch’s two-tailed, unpaired t test (C to I). **, P < 0.01; ns, not significant. Data in panels A to E are representative of two independent experiments with three replicates per condition; data in panels F to I are pooled from two independent experiments with a total of four to six mice per condition.
FIG 3
FIG 3
NSAID treatment does not affect SARS-CoV-2 entry or replication in vitro. (A and B) Calu-3 (A) and Huh7.5 (B) cells were pretreated with DMSO, 50 μM ibuprofen, or 50 μM meloxicam for 24 h and then infected with SARS2-VSVpp or G-VSVpp expressing Renilla luciferase. Luminescence was measured at 24 h postinfection (hpi) and normalized to DMSO for each infection. (C and D) Calu-3 (C) and Huh7.5 (D) cells were pretreated with DMSO, 50 μM ibuprofen, or 50 μM meloxicam for 24 h and then infected with mNeonGreen reporter replication-competent SARS-CoV-2 (icSARS-CoV-2-mNG) at an MOI of 1. The frequency of infected cells was measured by mNeonGreen expression at 1, 2, and 3 dpi. All data points in this figure are presented as means ± the SEM. Data were analyzed by Student two-tailed, unpaired t test (A and B) and two-way ANOVA (C and D). ns, not significant. Data in panels A and B are representative of two independent experiments with four replicates per condition; data in panels C and D are representative of two independent experiments with five replicates per condition.
FIG 4
FIG 4
NSAID treatment does not affect SARS-CoV-2-induced weight loss or lung viral burden in mice. (A and B) K18-hACE2 mice were treated intraperitoneally with DMSO or 1 mg/kg meloxicam daily for 7 days starting 1 day prior to infection. K18-hACE2 mice were infected intranasally with 103 PFU of SARS-CoV-2 or left uninfected and monitored daily. (A) Weight change expressed as a percentage of initial weight. (B) Viral burden in the lungs at 6 dpi measured by plaque assay. All data points in this figure are presented as means ± the SEM. Data were analyzed by two-way ANOVA (A) and Student two-tailed, unpaired t test (B). **, P < 0.01; ****, P < 0.0001; ns, not significant. Data in panels A and B are pooled from two independent experiments with a total of six mice per condition.
FIG 5
FIG 5
NSAID treatment does not affect innate immune cell activation in the lungs of SARS-CoV-2-infected mice. (A to H) K18-hACE2 mice were treated intraperitoneally with DMSO or 1 mg/kg meloxicam daily for 7 days starting 1 day prior to infection. K18-hACE2 mice were infected intranasally with 103 PFU of SARS-CoV-2 or left uninfected. Flow cytometric analysis of the lungs at 6 dpi for alveolar macrophage (MΦ) counts (A) and expression of CD86 (B), neutrophil counts (C), activated CD69+ natural killer (NK) cell counts (D), and Ly6C+ monocyte/macrophage (Mo/MΦ) counts (E) and expression of CD86 (F), MHCII (G), and CD64 (H) results are shown. All data points in this figure are presented as means ± the SEM. Data were analyzed by two-tailed Mann-Whitney test (A to H). *, P < 0.05; **, P < 0.01; ****, P < 0.0001; ns, not significant. Data in panels A to H are pooled from two independent experiments with a total of six mice per condition.
FIG 6
FIG 6
NSAID treatment impairs systemic neutralizing antibody responses but not adaptive immune cell activation in the lungs of SARS-CoV-2-infected mice. (A to G) K18-hACE2 mice were treated intraperitoneally with DMSO or 1 mg/kg meloxicam daily for 7 days starting 1 day prior to infection. K18-hACE2 mice were infected intranasally with 103 PFU of SARS-CoV-2 or left uninfected. Flow cytometric analysis of the lungs at 6 dpi for activated CD44+CD69+ CD4+ T cells (A), CD44+ CD69+ CD8+ T cells (B), CD44+ CD69+ γδ T cells (C), and B cells (D) was performed. (E and F) Spike (S)-specific IgM (E) and IgG (F) titers in the serum at 6 dpi. (G) Neutralizing antibody titers in the serum at 6 dpi measured by SARS2-VSVpp pseudovirus neutralization assay. Data points in panels A to F are presented as means ± the SEM. Data points in panel G are presented as boxplots. Data were analyzed by two-tailed Mann-Whitney test (A to D, G) and Student two-tailed, unpaired t test (E and F). *, P < 0.05; **, P < 0.01; ns, not significant. Data in panels A to G are pooled from two independent experiments with a total of four to six mice per condition.
FIG 7
FIG 7
NSAID treatment dampens the induction of proinflammatory cytokines that are upregulated by SARS-CoV-2 infection in mice. (A to D) K18-hACE2 mice were treated intraperitoneally with DMSO or 1 mg/kg meloxicam daily for 7 days starting 1 day prior to infection. K18-hACE2 mice were infected intranasally with 103 PFU of SARS-CoV-2 or left uninfected. Cytokine levels were measured in lung homogenates at 6 dpi. (A to C) Volcano plots detailing the differential abundance of cytokines in lung homogenates from infected versus uninfected mice treated with DMSO (A), uninfected mice treated with meloxicam versus DMSO (B), and infected mice treated with meloxicam versus DMSO (C). Significantly upregulated (red) and downregulated (blue) cytokines are labeled. (D) Levels of proinflammatory cytokines in lung homogenates from uninfected mice treated with DMSO, uninfected mice treated with meloxicam, infected mice treated with DMSO, and infected mice treated with meloxicam. The dotted line represents the upper limit of quantification. Data points in panel D are presented as means ± the SEM. Data were analyzed by two-tailed Mann–Whitney test (A to D). *, P < 0.05; **, P < 0.01; ns, not significant. Data in panels A to D are pooled from two independent experiments with a total of six mice per condition. Additional cytokine data are shown in Fig. S2 in the supplemental material.

References

    1. Day M. 2020. Covid-19: ibuprofen should not be used for managing symptoms, say doctors and scientists. BMJ 368:m1086.
    1. Powis S. 2020. Novel coronavirus: anti-inflammatory medications. Medicines and Healthcare Products Regulatory Agency, London, United Kingdom.
    1. Ricciotti E, FitzGerald GA. 2011. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol 31:986–1000. doi:10.1161/ATVBAHA.110.207449.
    1. Zhou P, Yang X-L, Wang X-G, Hu B, Zhang L, Zhang W, Si H-R, Zhu Y, Li B, Huang C-L, Chen H-D, Chen J, Luo Y, Guo H, Jiang R-D, Liu M-Q, Chen Y, Shen X-R, Wang X, Zheng X-S, Zhao K, Chen Q-J, Deng F, Liu L-L, Yan B, Zhan F-X, Wang Y-Y, Xiao G-F, Shi Z-L. 2020. A pneumonia outbreak associated with a new coronavirus of probable bat origin. Nature 579:270–273. doi:10.1038/s41586-020-2012-7.
    1. Fang L, Karakiulakis G, Roth M. 2020. Are patients with hypertension and diabetes mellitus at increased risk for COVID-19 infection? Lancet Respir Med 8:e21. doi:10.1016/S2213-2600(20)30116-8.
    1. Raaben M, Einerhand AW, Taminiau LJ, van Houdt M, Bouma J, Raatgeep RH, Büller HA, de Haan CA, Rossen JW. 2007. Cyclooxygenase activity is important for efficient replication of mouse hepatitis virus at an early stage of infection. Virol J 4:55. doi:10.1186/1743-422X-4-55.
    1. Qiao W, Wang C, Chen B, Zhang F, Liu Y, Lu Q, Guo H, Yan C, Sun H, Hu G, Yin X. 2015. Ibuprofen attenuates cardiac fibrosis in streptozotocin-induced diabetic rats. Cardiology 131:97–106. doi:10.1159/000375362.
    1. Miyoshi H, VanDussen KL, Malvin NP, Ryu SH, Wang Y, Sonnek NM, Lai C-W, Stappenbeck TS. 2017. Prostaglandin E2 promotes intestinal repair through an adaptive cellular response of the epithelium. EMBO J 36:5–24. doi:10.15252/embj.201694660.
    1. Alfajaro MM, Choi J-S, Kim D-S, Seo J-Y, Kim J-Y, Park J-G, Soliman M, Baek Y-B, Cho E-H, Kwon J, Kwon H-J, Park S-J, Lee WS, Kang M-I, Hosmillo M, Goodfellow I, Cho K-O. 2017. Activation of COX-2/PGE2 promotes sapovirus replication via the inhibition of nitric oxide production. J Virol 91:e01656-16. doi:10.1128/JVI.01656-16.
    1. Alfajaro MM, Cho E-H, Park J-G, Kim J-Y, Soliman M, Baek Y-B, Kang M-I, Park S-I, Cho K-O. 2018. Feline calicivirus- and murine norovirus-induced COX-2/PGE2 signaling pathway has proviral effects. PLoS One 13:e0200726. doi:10.1371/journal.pone.0200726.
    1. Lu R, Zhao X, Li J, Niu P, Yang B, Wu H, Wang W, Song H, Huang B, Zhu N, Bi Y, Ma X, Zhan F, Wang L, Hu T, Zhou H, Hu Z, Zhou W, Zhao L, Chen J, Meng Y, Wang J, Lin Y, Yuan J, Xie Z, Ma J, Liu WJ, Wang D, Xu W, Holmes EC, Gao GF, Wu G, Chen W, Shi W, Tan W. 2020. Genomic characterization and epidemiology of 2019 novel coronavirus: implications for virus origins and receptor binding. Lancet 395:565–574. doi:10.1016/S0140-6736(20)30251-8.
    1. Yan X, Hao Q, Mu Y, Timani KA, Ye L, Zhu Y, Wu J. 2006. Nucleocapsid protein of SARS-CoV activates the expression of cyclooxygenase-2 by binding directly to regulatory elements for nuclear factor-κB and CCAAT/enhancer binding protein. Int J Biochem Cell Biol 38:1417–1428. doi:10.1016/j.biocel.2006.02.003.
    1. Liu M, Yang Y, Gu C, Yue Y, Wu KK, Wu J, Zhu Y. 2007. Spike protein of SARS-CoV stimulates cyclooxygenase-2 expression via both calcium-dependent and calcium-independent protein kinase C pathways. FASEB J 21:1586–1596. doi:10.1096/fj.06-6589com.
    1. Fajgenbaum DC, June CH. 2020. Cytokine storm. N Engl J Med 383:2255–2273. doi:10.1056/NEJMra2026131.
    1. Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, Linsell L, Staplin N, Brightling C, Ustianowski A, Elmahi E, Prudon B, Green C, Felton T, Chadwick D, Rege K, Fegan C, Chappell LC, Faust SN, Jaki T, Jeffery K, Montgomery A, Rowan K, Juszczak E, Baillie JK, Haynes R, Landray MJ, RECOVERY Collaborative Group . 2020. Dexamethasone in hospitalized patients with Covid-19: preliminary report. N Engl J Med doi:10.1056/NEJMoa2021436.
    1. Lucas C, Wong P, Klein J, Castro TBR, Silva J, Sundaram M, Ellingson MK, Mao T, Oh JE, Israelow B, Takahashi T, Tokuyama M, Lu P, Venkataraman A, Park A, Mohanty S, Wang H, Wyllie AL, Vogels CBF, Earnest R, Lapidus S, Ott IM, Moore AJ, Muenker MC, Fournier JB, Campbell M, Odio CD, Casanovas-Massana A, Herbst R, Shaw AC, Medzhitov R, Schulz WL, Grubaugh ND, Cruz CD, Farhadian S, Ko AI, Omer SB, Iwasaki A, Yale IMPACT Team . 2020. Longitudinal analyses reveal immunological misfiring in severe COVID-19. 7821. Nature 584:463–469. doi:10.1038/s41586-020-2588-y.
    1. Mann ER, Menon M, Knight SB, Konkel JE, Jagger C, Shaw TN, Krishnan S, Rattray M, Ustianowski A, Bakerly ND, Dark P, Lord GM, Simpson A, Felton T, Ho L-P, Trc NR, Feldmann M, CIRCO, Grainger JR, Hussell T. 2020. Longitudinal immune profiling reveals key myeloid signatures associated with COVID-19. Sci Immunol 5:eabd6197. doi:10.1126/sciimmunol.abd6197.
    1. Robb CT, Goepp M, Rossi AG, Yao C. 2020. Non-steroidal anti-inflammatory drugs, prostaglandins, and COVID-19. Br J Pharmacol 177:4899–4920. doi:10.1111/bph.15206.
    1. Yao C, Narumiya S. 2019. Prostaglandin-cytokine crosstalk in chronic inflammation. Br J Pharmacol 176:337–354. doi:10.1111/bph.14530.
    1. Zhao J, Zhao J, Legge K, Perlman S. 2011. Age-related increases in PGD2 expression impair respiratory DC migration, resulting in diminished T cell responses upon respiratory virus infection in mice. J Clin Invest 121:4921–4930. doi:10.1172/JCI59777.
    1. Coulombe F, Jaworska J, Verway M, Tzelepis F, Massoud A, Gillard J, Wong G, Kobinger G, Xing Z, Couture C, Joubert P, Fritz JH, Powell WS, Divangahi M. 2014. Targeted prostaglandin E2 inhibition enhances antiviral immunity through induction of type I interferon and apoptosis in macrophages. Immunity 40:554–568. doi:10.1016/j.immuni.2014.02.013.
    1. Vijay R, Fehr AR, Janowski AM, Athmer J, Wheeler DL, Grunewald M, Sompallae R, Kurup SP, Meyerholz DK, Sutterwala FS, Narumiya S, Perlman S. 2017. Virus-induced inflammasome activation is suppressed by prostaglandin D2/DP1 signaling. Proc Natl Acad Sci U S A 114:E5444–E5453. doi:10.1073/pnas.1704099114.
    1. Hashimoto K, Graham BS, Geraci MW, FitzGerald GA, Egan K, Zhou W, Goleniewska K, O’Neal JF, Morrow JD, Durbin RK, Wright PF, Collins RD, Suzutani T, Peebles RS. 2004. Signaling through the prostaglandin I2 receptor IP protects against respiratory syncytial virus-induced illness. J Virol 78:10303–10309. doi:10.1128/JVI.78.19.10303-10309.2004.
    1. Bernard MP, Bancos S, Chapman TJ, Ryan EP, Treanor JJ, Rose RC, Topham DJ, Phipps RP. 2010. Chronic inhibition of cyclooxygenase-2 attenuates antibody responses against vaccinia infection. Vaccine 28:1363–1372. doi:10.1016/j.vaccine.2009.11.005.
    1. Arvin AM, Fink K, Schmid MA, Cathcart A, Spreafico R, Havenar-Daughton C, Lanzavecchia A, Corti D, Virgin HW. 2020. A perspective on potential antibody-dependent enhancement of SARS-CoV-2. 7821. Nature 584:353–363. doi:10.1038/s41586-020-2538-8.
    1. Blanco-Melo D, Nilsson-Payant BE, Liu W-C, Uhl S, Hoagland D, Møller R, Jordan TX, Oishi K, Panis M, Sachs D, Wang TT, Schwartz RE, Lim JK, Albrecht RA, tenOever BR. 2020. Imbalanced host response to SARS-CoV-2 drives development of COVID-19. Cell 181:1036–1045. doi:10.1016/j.cell.2020.04.026.
    1. Leung C, Wadsworth SJ, Yang SJ, Dorscheid DR. 2020. Structural and functional variations in human bronchial epithelial cells cultured in air-liquid interface using different growth media. Am J Physiol Lung Cell Mol Physiol 318:L1063–L1073. doi:10.1152/ajplung.00190.2019.
    1. Ravindra NG, Alfajaro MM, Gasque V, Habet V, Wei J, Filler RB, Huston NC, Wan H, Szigeti-Buck K, Wang B, Wang G, Montgomery RR, Eisenbarth SC, Williams A, Pyle AM, Iwasaki A, Horvath TL, Foxman EF, Pierce RW, van Dijk D, Wilen CB. 2020. Single-cell longitudinal analysis of SARS-CoV-2 infection in human airway epithelium. bioRxiv doi:10.1101/2020.05.06.081695.
    1. McCray PB, Pewe L, Wohlford-Lenane C, Hickey M, Manzel L, Shi L, Netland J, Jia HP, Halabi C, Sigmund CD, Meyerholz DK, Kirby P, Look DC, Perlman S. 2007. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J Virol 81:813–821. doi:10.1128/JVI.02012-06.
    1. Bao L, Deng W, Huang B, Gao H, Liu J, Ren L, Wei Q, Yu P, Xu Y, Qi F, Qu Y, Li F, Lv Q, Wang W, Xue J, Gong S, Liu M, Wang G, Wang S, Song Z, Zhao L, Liu P, Zhao L, Ye F, Wang H, Zhou W, Zhu N, Zhen W, Yu H, Zhang X, Guo L, Chen L, Wang C, Wang Y, Wang X, Xiao Y, Sun Q, Liu H, Zhu F, Ma C, Yan L, Yang M, Han J, Xu W, Tan W, Peng X, Jin Q, Wu G, Qin C. 2020. The pathogenicity of SARS-CoV-2 in hACE2 transgenic mice. Nature 583:830–833. doi:10.1038/s41586-020-2312-y.
    1. Jiang R-D, Liu M-Q, Chen Y, Shan C, Zhou Y-W, Shen X-R, Li Q, Zhang L, Zhu Y, Si H-R, Wang Q, Min J, Wang X, Zhang W, Li B, Zhang H-J, Baric RS, Zhou P, Yang X-L, Shi Z-L. 2020. Pathogenesis of SARS-CoV-2 in transgenic mice expressing human angiotensin-converting enzyme 2. Cell 182:50–58. doi:10.1016/j.cell.2020.05.027.
    1. Hassan AO, Case JB, Winkler ES, Thackray LB, Kafai NM, Bailey AL, McCune BT, Fox JM, Chen RE, Alsoussi WB, Turner JS, Schmitz AJ, Lei T, Shrihari S, Keeler SP, Fremont DH, Greco S, McCray PB, Perlman S, Holtzman MJ, Ellebedy AH, Diamond MS. 2020. A SARS-CoV-2 infection model in mice demonstrates protection by neutralizing antibodies. Cell 182:744–753. doi:10.1016/j.cell.2020.06.011.
    1. Sun J, Zhuang Z, Zheng J, Li K, Wong RL-Y, Liu D, Huang J, He J, Zhu A, Zhao J, Li X, Xi Y, Chen R, Alshukairi AN, Chen Z, Zhang Z, Chen C, Huang X, Li F, Lai X, Chen D, Wen L, Zhuo J, Zhang Y, Wang Y, Huang S, Dai J, Shi Y, Zheng K, Leidinger MR, Chen J, Li Y, Zhong N, Meyerholz DK, McCray PB, Perlman S, Zhao J. 2020. Generation of a broadly useful model for COVID-19 pathogenesis, vaccination, and treatment. Cell 182:734–743. doi:10.1016/j.cell.2020.06.010.
    1. Israelow B, Song E, Mao T, Lu P, Meir A, Liu F, Alfajaro MM, Wei J, Dong H, Homer RJ, Ring A, Wilen CB, Iwasaki A. 2020. Mouse model of SARS-CoV-2 reveals inflammatory role of type I interferon signaling. J Exp Med 217:e20201241. doi:10.1084/jem.20201241.
    1. Sun S-H, Chen Q, Gu H-J, Yang G, Wang Y-X, Huang X-Y, Liu S-S, Zhang N-N, Li X-F, Xiong R, Guo Y, Deng Y-Q, Huang W-J, Liu Q, Liu Q-M, Shen Y-L, Zhou Y, Yang X, Zhao T-Y, Fan C-F, Zhou Y-S, Qin C-F, Wang Y-C. 2020. A mouse model of SARS-CoV-2 infection and pathogenesis. Cell Host Microbe 28:124–133. doi:10.1016/j.chom.2020.05.020.
    1. Winkler ES, Bailey AL, Kafai NM, Nair S, McCune BT, Yu J, Fox JM, Chen RE, Earnest JT, Keeler SP, Ritter JH, Kang L-I, Dort S, Robichaud A, Head R, Holtzman MJ, Diamond MS. 2020. SARS-CoV-2 infection of human ACE2-transgenic mice causes severe lung inflammation and impaired function. Nat Immunol 21:1470–1479. doi:10.1038/s41590-020-0794-2.
    1. Park MK, Kang SH, Son JY, Lee MK, Ju JS, Bae YC, Ahn DK. 2019. Co-administered low doses of ibuprofen and dexamethasone produce synergistic antinociceptive effects on neuropathic mechanical allodynia in rats. J Pain Res 12:2959–2968. doi:10.2147/JPR.S222095.
    1. Tubbs JT, Kissling GE, Travlos GS, Goulding DR, Clark JA, King-Herbert AP, Blankenship-Paris TL. 2011. Effects of buprenorphine, meloxicam, and flunixin meglumine as postoperative analgesia in mice. J Am Assoc Lab Anim Sci 50:185–191.
    1. Santos ARS, Vedana EMA, De Freitas GAG. 1998. Antinociceptive effect of meloxicam, in neurogenic and inflammatory nociceptive models in mice. Inflammation Res 47:302–307. doi:10.1007/s000110050333.
    1. Laird JMA, Herrero JF, Garcia de la Rubia P, Cervero F. 1997. Analgesic activity of the novel COX-2 preferring NSAID, meloxicam in mono-arthritic rats: central and peripheral components. Inflamm Res 46:203–210. doi:10.1007/s000110050174.
    1. Avanzato VA, Oguntuyo KY, Escalera-Zamudio M, Gutierrez B, Golden M, Pond SLK, Pryce R, Walter TS, Seow J, Doores KJ, Pybus OG, Munster VJ, Lee B, Bowden TA. 2019. A structural basis for antibody-mediated neutralization of Nipah virus reveals a site of vulnerability at the fusion glycoprotein apex. Proc Natl Acad Sci U S A 116:25057–25067. doi:10.1073/pnas.1912503116.
    1. Wei J, Alfajaro MM, DeWeirdt PC, Hanna RE, Lu-Culligan WJ, Cai WL, Strine MS, Zhang S-M, Graziano VR, Schmitz CO, Chen JS, Mankowski MC, Filler RB, Ravindra NG, Gasque V, de Miguel FJ, Patil A, Chen H, Oguntuyo KY, Abriola L, Surovtseva YV, Orchard RC, Lee B, Lindenbach BD, Politi K, van Dijk D, Kadoch C, Simon MD, Yan Q, Doench JG, Wilen CB. 2021. Genome-wide CRISPR screens reveal host factors critical for SARS-CoV-2 infection. Cell 184:76–91. doi:10.1016/j.cell.2020.10.028.
    1. Sander WJ, O’Neill HG, Pohl CH. 2017. Prostaglandin E2 as a modulator of viral infections. Front Physiol 8:89. doi:10.3389/fphys.2017.00089.
    1. Luczak M, Gumulka W, Szmigielski S, Korbecki M. 1975. Inhibition of multiplication of parainfluenza 3 virus in prostaglandin-treated WISH cells. Arch Virol 49:377–380. doi:10.1007/BF01318248.
    1. Xie X, Muruato A, Lokugamage KG, Narayanan K, Zhang X, Zou J, Liu J, Schindewolf C, Bopp NE, Aguilar PV, Plante KS, Weaver SC, Makino S, LeDuc JW, Menachery VD, Shi P-Y. 2020. An infectious cDNA clone of SARS-CoV-2. Cell Host Microbe 27:841–848. doi:10.1016/j.chom.2020.04.004.
    1. Kindler E, Thiel V, Weber F. 2016. Interaction of SARS and MERS coronaviruses with the antiviral interferon response. Adv Virus Res 96:219–243.
    1. Dudek SE, Nitzsche K, Ludwig S, Ehrhardt C. 2016. Influenza A viruses suppress cyclooxygenase-2 expression by affecting its mRNA stability. 1. Sci Rep 6:27275. doi:10.1038/srep27275.
    1. Sumpter R, Loo Y-M, Foy E, Li K, Yoneyama M, Fujita T, Lemon SM, Gale M. 2005. Regulating intracellular antiviral defense and permissiveness to hepatitis C virus RNA replication through a cellular RNA helicase, RIG-I. J Virol 79:2689–2699. doi:10.1128/JVI.79.5.2689-2699.2005.
    1. Fink K. 2012. Origin and function of circulating plasmablasts during acute viral infections. Front Immunol 3:78. doi:10.3389/fimmu.2012.00078.
    1. Ryan EP, Pollock SJ, Pollack SJ, Murant TI, Bernstein SH, Felgar RE, Phipps RP. 2005. Activated human B lymphocytes express cyclooxygenase-2 and cyclooxygenase inhibitors attenuate antibody production. J Immunol 174:2619–2626. doi:10.4049/jimmunol.174.5.2619.
    1. Bernard MP, Phipps RP. 2007. CpG oligodeoxynucleotides induce cyclooxygenase-2 in human B lymphocytes: implications for adjuvant activity and antibody production. Clin Immunol 125:138–148. doi:10.1016/j.clim.2007.07.006.
    1. Bancos S, Bernard MP, Topham DJ, Phipps RP. 2009. Ibuprofen and other widely used non-steroidal anti-inflammatory drugs inhibit antibody production in human cells. Cell Immunol 258:18–28. doi:10.1016/j.cellimm.2009.03.007.
    1. Bernard MP, Phipps RP. 2010. Inhibition of cyclooxygenase-2 impairs the expression of essential plasma cell transcription factors and human B-lymphocyte differentiation. Immunology 129:87–96. doi:10.1111/j.1365-2567.2009.03152.x.
    1. Zheng J, Wong L-YR, Li K, Verma AK, Ortiz M, Wohlford-Lenane C, Leidinger MR, Knudson CM, Meyerholz DK, McCray PB, Perlman S. 2020. COVID-19 treatments and pathogenesis including anosmia in K18-hACE2 mice. Nature doi:10.1038/s41586-020-2943-z.
    1. Tipton DA, Flynn JC, Stein SH, Dabbous MK. 2003. Cyclooxygenase-2 inhibitors decrease interleukin-1β-stimulated prostaglandin E2 and IL-6 production by human gingival fibroblasts. J Periodontol 74:1754–1763. doi:10.1902/jop.2003.74.12.1754.
    1. Cheng Q, Li N, Chen M, Zheng J, Qian Z, Wang X, Huang C, Xu S, Shi G. 2013. Cyclooxygenase-2 promotes hepatocellular apoptosis by interacting with TNF-α and IL-6 in the pathogenesis of nonalcoholic steatohepatitis in rats. Dig Dis Sci 58:2895–2902. doi:10.1007/s10620-013-2823-6.
    1. Kaur J, Sanyal SN. 2011. Diclofenac, a selective COX-2 inhibitor, inhibits DMH-induced colon tumorigenesis through suppression of MCP-1, MIP-1α, and VEGF. Mol Carcinog 50:707–718. doi:10.1002/mc.20736.
    1. Lisboa FA, Bradley MJ, Hueman MT, Schobel SA, Gaucher BJ, Styrmisdottir EL, Potter BK, Forsberg JA, Elster EA. 2017. Nonsteroidal anti-inflammatory drugs may affect cytokine response and benefit healing of combat-related extremity wounds. Surgery 161:1164–1173. doi:10.1016/j.surg.2016.10.011.
    1. Anderson GD, Hauser SD, McGarity KL, Bremer ME, Isakson PC, Gregory SA. 1996. Selective inhibition of cyclooxygenase (COX)-2 reverses inflammation and expression of COX-2 and interleukin 6 in rat adjuvant arthritis. J Clin Invest 97:2672–2679. doi:10.1172/JCI118717.
    1. Aoki T, Frȍsen J, Fukuda M, Bando K, Shioi G, Tsuji K, Ollikainen E, Nozaki K, Laakkonen J, Narumiya S. 2017. Prostaglandin E2–EP2–NF-κB signaling in macrophages as a potential therapeutic target for intracranial aneurysms. Sci Signal 10:eaah6037. doi:10.1126/scisignal.aah6037.
    1. Zhou Y, Fu B, Zheng X, Wang D, Zhao C, Qi Y, Sun R, Tian Z, Xu X, Wei H. 2020. Pathogenic T cells and inflammatory monocytes incite inflammatory storms in severe COVID-19 patients. Natl Sci Rev 7:998–1002. doi:10.1093/nsr/nwaa041.
    1. Lam JKP, Hui KF, Ning RJ, Xu XQ, Chan KH, Chiang AKS. 2018. Emergence of CD4+ and CD8+ polyfunctional T cell responses against immunodominant lytic and latent EBV antigens in children with primary EBV infection. Front Microbiol 9:416. doi:10.3389/fmicb.2018.00416.
    1. Seo SH, Webster RG. 2002. Tumor necrosis factor alpha exerts powerful anti-influenza virus effects in lung epithelial cells. J Virol 76:1071–1076. doi:10.1128/jvi.76.3.1071-1076.2002.
    1. Karki R, Sharma BR, Tuladhar S, Williams EP, Zalduondo L, Samir P, Zheng M, Sundaram B, Banoth B, Malireddi RKS, Schreiner P, Neale G, Vogel P, Webby R, Jonsson CB, Kanneganti T-D. 2021. Synergism of TNF-α and IFN-γ triggers inflammatory cell death, tissue damage, and mortality in SARS-CoV-2 infection and cytokine shock syndromes. Cell 184:149–168. doi:10.1016/j.cell.2020.11.025.
    1. Huang E, Jordan SC. 2020. Tocilizumab for Covid-19: the ongoing search for effective therapies. N Engl J Med 383:2387–2388. doi:10.1056/NEJMe2032071.
    1. Imam Z, Odish F, Gill I, O’Connor D, Armstrong J, Vanood A, Ibironke O, Hanna A, Ranski A, Halalau A. 2020. Older age and comorbidity are independent mortality predictors in a large cohort of 1305 COVID-19 patients in Michigan, United States. J Intern Med 288:469–476. doi:10.1111/joim.13119.
    1. Rinott E, Kozer E, Shapira Y, Bar-Haim A, Youngster I. 2020. Ibuprofen use and clinical outcomes in COVID-19 patients. Clin Microbiol Infect 26:1259.e5–1259-e7. doi:10.1016/j.cmi.2020.06.003.
    1. Wong AY, MacKenna B, Morton C, Schultze A, Walker AJ, Bhaskaran K, Brown J, Rentsch CT, Williamson E, Drysdale H, Croker R, Bacon S, Hulme W, Bates C, Curtis HJ, Mehrkar A, Evans D, Inglesby P, Cockburn J, McDonald H, Tomlinson L, Mathur R, Wing K, Forbes H, Parry J, Hester F, Harper S, Evans S, Smeeth L, Douglas I, Goldacre B. 2020. OpenSAFELY: do adults prescribed Non-steroidal anti-inflammatory drugs have an increased risk of death from COVID-19? medRxiv doi:10.1101/2020.08.12.20171405.
    1. Abu Esba LC, Alqahtani RA, Thomas A, Shamas N, Alswaidan L, Mardawi G. 2020. Ibuprofen and NSAID use in COVID-19 infected patients is not associated with worse outcomes: a prospective cohort study. Infect Dis Ther doi:10.1007/s40121-020-00363-w.
    1. Bruce E, Barlow-Pay F, Short R, Vilches-Moraga A, Price A, McGovern A, Braude P, Stechman MJ, Moug S, McCarthy K, Hewitt J, Carter B, Myint PK. 2020. Prior routine use of non-steroidal anti-inflammatory drugs (NSAIDs) and important outcomes in hospitalized patients with COVID-19. J Clin Med 9:2586. doi:10.3390/jcm9082586.
    1. Lund LC, Kristensen KB, Reilev M, Christensen S, Thomsen RW, Christiansen CF, Støvring H, Johansen NB, Brun NC, Hallas J, Pottegård A. 2020. Adverse outcomes and mortality in users of non-steroidal anti-inflammatory drugs who tested positive for SARS-CoV-2: a Danish nationwide cohort study. PLoS Med 17:e1003308. doi:10.1371/journal.pmed.1003308.
    1. Love MI, Huber W, Anders S. 2014. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 15:550. doi:10.1186/s13059-014-0550-8.
    1. Decker JM, Bibollet-Ruche F, Wei X, Wang S, Levy DN, Wang W, Delaporte E, Peeters M, Derdeyn CA, Allen S, Hunter E, Saag MS, Hoxie JA, Hahn BH, Kwong PD, Robinson JE, Shaw GM. 2005. Antigenic conservation and immunogenicity of the HIV coreceptor binding site. J Exp Med 201:1407–1419. doi:10.1084/jem.20042510.

Source: PubMed

3
Iratkozz fel