Babaodan controls excessive immune responses and may represent a cytokine-targeted agent suitable for COVID-19 treatment

Jing Qian, Hangdi Xu, Dongqing Lv, Wei Liu, Enguo Chen, Yong Zhou, Yi Wang, Kejing Ying, Xiaohui Fan, Jing Qian, Hangdi Xu, Dongqing Lv, Wei Liu, Enguo Chen, Yong Zhou, Yi Wang, Kejing Ying, Xiaohui Fan

Abstract

It has become evident that the actions of pro-inflammatory cytokines and/or the development of a cytokine storm are responsible for the occurrence of severe COVID-19 during SARS-CoV-2 infection. Although immunomodulatory mechanisms vary among viruses, the activation of multiple TLRs that occurs primarily through the recruitment of adapter proteins such as MyD88 and TRIF contributes to the induction of a cytokine storm. Based on this, controlling the robust production of pro-inflammatory cytokines by macrophages may be applicable as a cellular approach to investigate potential cytokine-targeted therapies against COVID-19. In the current study, we utilized TLR2/MyD88 and TLR3/TRIF co-activated macrophages and evaluated the anti-cytokine storm effect of the traditional Chinese medicine (TCM) formula Babaodan (BBD). An RNA-seq-based transcriptomic approach was used to determine the molecular mode of action. Additionally, we evaluated the anti-inflammatory activity of BBD in vivo using a mouse model of post-viral bacterial infection-induced pneumonia and seven severely ill COVID-19 patients. Our study reveals the protective role of BBD against excessive immune responses in macrophages, where the underlying mechanisms involve the inhibition of the NF-κB and MAPK signaling pathways. In vivo, BBD significantly inhibited the release of IL-6, thus resulting in increased survival rates in mice. Based on limited data, we demonstrated that severely ill COVID-19 patients benefited from BBD treatment due to a reduction in the overproduction of IL-6. In conclusion, our study indicated that BBD controls excessive immune responses and may thus represent a cytokine-targeted agent that could be considered to treating COVID-19.

Keywords: Babaodan; COVID-19; Cytokine storm; Inflammation; Traditional Chinese medicine.

Conflict of interest statement

The authors declare that they have no known competing financial interests or personal relationships that could have influenced the work reported in this paper.

Copyright © 2021 The Authors. Published by Elsevier Masson SAS.. All rights reserved.

Figures

Graphical abstract
Graphical abstract
Fig. 1
Fig. 1
Babaodan (BBD) inhibits TLR1/2 and TLR3 dual stimulation-induced cytokine expression in Raw 264.7 macrophages. Raw 264.7 cells were grown on 12-well plates and incubated with different doses of BBD for 24 h, and this was followed by treatment with a combination of Poly(I:C) (10 μg/mL) and Pam3CSK4 (100 ng/mL) for 24 h. Supernatants were collected, and TNF-α and IL-6 concentrations were analyzed using the corresponding ELISA kits. *P < 0.05, **P < 0.01,***P < 0.001 vs. model group.
Fig. 2
Fig. 2
BBD abolishes IL-6 and TNF-α production after optimized TLR1/2 and TLR3 dual activation of Raw264.7 macrophages. (A) Raw 264.7 cells were grown on 12-well plates and incubated with different combinations of Poly(I:C) and Pam3CSK4 for 24 h. Supernatants were collected, and TNF-α and IL-6 concentrations were determined using the corresponding ELISA kits. (B) Raw 264.7 cells were grown on 6-well plates and incubated with 1 mg/mL of BBD for 24 h, and this was followed by treatment with a combination of Poly(I:C) (10 μg/mL) and Pam3CSK4 (100 ng/mL) for 8 h. IL-6 and TNF-α mRNA levels were detected by RT-qPCR. *** P < 0.001 vs. model group.
Fig. 3
Fig. 3
Gene expression profiling by RNA-seq. Raw 264.7 cells grown on 6-well plates were treated with a combination of 10 μg/mL Poly(I:C) and 100 ng/mL Pam3CSK4 (model) for 8 h with or without pre-treatment with 1 mg/mL of Babaodan (BBD) for 24 h. RNA samples were collected for RNA-seq analysis. Two samples were analyzed per group. Differentially expressed genes (DEG) are identified by absolute log2 (FC) > 2 and P < 0.05 values. (A) Number of up- and downregulated genes in the model and treatment groups. (B) Venn diagram summary of DEGs between the control vs. model and model vs. BBD groups. (C) Top 15 KEGG pathway enrichment scores of co-regulated DEGs in the model vs. control and BBD vs. model groups.
Fig. 4
Fig. 4
Transcriptional profiling changes in inflammatory DEG result from BBD treatment. (A) Heatmap of relative gene expression (log2[FC]) for the 124 inflammatory genes that were upregulated and the 37 inflammatory genes that were downregulated in the model vs. control groups. BBD regulates the expression of 84 and 9 of these categories of genes, respectively, compared to the control group. Blue, downregulation; red, upregulation. (B) Transcription factor (TF) enrichment analysis was performed for the 84 downregulated inflammatory DEGs, and the top 10 TFs with the highest importance are listed.
Fig. 5
Fig. 5
BBD inhibits NF-κB and MAPK signaling in macrophages promoting excessive inflammatory processes. Raw 264.7 cells were grown on 6-well plates and incubated with 1 mg/mL BBD for 24 h, and this was followed by treatment with a combination of Poly(I:C) (10 μg/mL) and Pam3CSK4 (100 ng/mL) (P2P) for 15, 30, 60, and 120 min. The cells were harvested at the indicated time points after stimulation. Cell lysates were resolved by SDS-PAGE, blotted, and analyzed using the respective antibodies. One representative experiment of three is shown.
Fig. 6
Fig. 6
BBD attenuates lung inflammation in mice with acute lung injury. C57BL/6 mice were infected with 200 PFU of influenza A PR/8/34 or vehicle for 5 days before being challenged with Staphylococcus aureus (SA) at the indicated CFU in 50 μL: 5 × 107 for A-B and 2.5 × 107 for C-D. (A) Survival curves; n = 16, model group; n = 17, BBD treatment group. (B) Representative histological sections of lungs at 12 h post SA challenge (n = 4–5 for each group). (C) CFU counts in LH and BAL samples (n = 4–5 for each group). (D) Concentrations of TNF-α and IL-6 in BALF (n = 3–5 for each group). * P < 0.05 vs. model group. CFU, colony-forming units; PFU, plaque-forming units; BALF, bronchoalveolar lavage fluid.
Fig. 7
Fig. 7
Outcomes of CT scan image and inflammation analyses in a severely ill COVID-19 patient (PT-7) following BBD treatment. (A) Lung CT scan images for the patient at 2, 5, 7, and 11 days after BBD treatment prior to being discharged. While the bilateral pneumonia continued to develop at 2, 5, and 7 days after BBD treatment, improvement occurred on the 11th day after BBD treatment. (B-C) The dynamics of serum IL-6 and CRP levels were monitored.

References

    1. Wiersinga W.J., Rhodes A., Cheng A.C., Peacock S.J., Prescott H.C. Pathophysiology, transmission, diagnosis, and treatment of coronavirus disease 2019 (COVID-19): a review. JAMA. 2020;324:782. doi: 10.1001/jama.2020.12839.
    1. Huang C., Wang Y., Li X., Ren L., Zhao J., Hu Y., Zhang L., Fan G., Xu J., Gu X., Cheng Z., Yu T., Xia J., Wei Y., Wu W., Xie X., Yin W., Li H., Liu M., Xiao Y., Gao H., Guo L., Xie J., Wang G., Jiang R., Gao Z., Jin Q., Wang J., Cao B. Clinical features of patients infected with 2019 novel coronavirus in Wuhan, China. Lancet. 2020;395:497–506. doi: 10.1016/S0140-6736(20)30183-5.
    1. Qin C., Zhou L., Hu Z., Zhang S., Yang S., Tao Y., Xie C., Ma K., Shang K., Wang W., Tian D.S. Dysregulation of immune response in patients with coronavirus 2019 (COVID-19) in Wuhan, China. Clin. Infect. Dis. 2020;71:762–768. doi: 10.1093/cid/ciaa248.
    1. Xu Z., Shi L., Wang Y., Zhang J., Huang L., Zhang C., Liu S., Zhao P., Liu H., Zhu L., Tai Y., Bai C., Gao T., Song J., Xia P., Dong J., Zhao J., Wang F.S. Pathological findings of COVID-19 associated with acute respiratory distress syndrome. Lancet Respir. Med. 2020 doi: 10.1016/S2213-2600(20)30076-X.
    1. Chen N., Zhou M., Dong X., Qu J., Gong F., Han Y., Qiu Y., Wang J., Liu Y., Wei Y., Xia J., Yu T., Zhang X., Zhang L. Epidemiological and clinical characteristics of 99 cases of 2019 novel coronavirus pneumonia in Wuhan, China: a descriptive study. Lancet. 2020;395:507–513. doi: 10.1016/S0140-6736(20)30211-7.
    1. Pedersen S.F., Ho Y.C. SARS-CoV-2: a storm is raging. J. Clin. Investig. 2020;130:2202–2205. doi: 10.1172/JCI137647.
    1. Cao X. COVID-19: immunopathology and its implications for therapy. Nat. Rev. Immunol. 2020;20:269–270. doi: 10.1038/s41577-020-0308-3.
    1. Catanzaro M., Fagiani F., Racchi M., Corsini E., Govoni S., Lanni C. Immune response in COVID-19: addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2. Signal Transduct. Target. Ther. 2020;5:84. doi: 10.1038/s41392-020-0191-1.
    1. Uccellini M.B., García-Sastre A. ISRE-reporter mouse reveals high basal and induced type I IFN responses in inflammatory monocytes. Cell Rep. 2018;25:2784–2796. doi: 10.1016/j.celrep.2018.11.030.
    1. Zhang D., Guo R., Lei L., Liu H., Wang Y., Wang Y., Dai T., Zhang T., Lai Y., Wang J., Liu Z., He A., O’Dwyer M., Hu J. COVID-19 infection induces readily detectable morphological and inflammation-related phenotypic changes in peripheral blood monocytes, the severity of which correlate with patient outcome. MedRxiv. 2020 doi: 10.1101/2020.03.24.20042655. 2020.03.24.20042655.
    1. Wen W., Su W., Tang H., Le W., Zhang X., Zheng Y., Liu X., Xie L., Li J., Ye J., Dong L., Cui X., Miao Y., Wang D., Dong J., Xiao C., Chen W., Wang H. Immune cell profiling of COVID-19 patients in the recovery stage by single-cell sequencing. Cell Discov. 2020 doi: 10.1038/s41421-020-0168-9.
    1. M. Cascella, M. Rajnik, A. Cuomo, S.C. Dulebohn, R. Di Napoli, Features, Evaluation and Treatment Coronavirus (COVID-19), StatPearls, Treasure island (FL), 2020.
    1. Roschewski M., Lionakis M.S., Sharman J.P., Roswarski J., Goy A., Monticelli M.A., Roshon M., Wrzesinski S.H., Desai J.V., Zarakas M.A., Collen J., Rose K.M., Hamdy A., Izumi R., Wright G.W., Chung K.K., Baselga J., Staudt L.M., Wilson W.H. Inhibition of Bruton tyrosine kinase in patients with severe COVID-19. Sci. Immunol. 2020;5 doi: 10.1126/SCIIMMUNOL.ABD0110.
    1. Tian X., Xu F., Lung W.Y., Meyerson C., Ghaffari A.A., Cheng G., Deng J.C. Poly I:C enhances susceptibility to secondary pulmonary infections by gram-positive bacteria. PLoS One. 2012;7 doi: 10.1371/journal.pone.0041879.
    1. Lee B., Robinson K.M., McHugh K.J., Scheller E.V., Mandalapu S., Chen C., Di Y.P., Clay M.E., Enelow R.I., Dubin P.J., Alcorn J.F. Influenza-induced type I interferon enhances susceptibility to gram-negative and gram-positive bacterial pneumonia in mice. Am. J. Physiol. -Lung Cell. Mol. Physiol. 2015;309:L158–L167. doi: 10.1152/ajplung.00338.2014.
    1. Shepardson K.M., Larson K., Morton R.V., Prigge J.R., Schmidt E.E., Huber V.C., Rynda-Apple A. Differential type I interferon signaling is a master regulator of susceptibility to postinfluenza bacterial superinfection. MBio. 2016;7 doi: 10.1128/mBio.00506-16.
    1. Ting Tan R.S., Lin B., Liu Q., Tucker-Kellogg L., Ho B., Leung B.P., Ding J.L. The synergy in cytokine production through MyD88-TRIF pathways is co-ordinated with ERK phosphorylation in macrophages. Immunol. Cell Biol. 2013;91:377–387. doi: 10.1038/icb.2013.13.
    1. Lin B., Dutta B., Fraser I.D.C. Systematic investigation of multi-TLR sensing identifies regulators of sustained gene activation in macrophages. Cell Syst. 2017;5:25–37. doi: 10.1016/j.cels.2017.06.014.
    1. Conti P., Ronconi G., Caraffa A., Gallenga C.E., Ross R., Frydas I., Kritas S.K. Induction of pro-inflammatory cytokines (IL-1 and IL-6) and lung inflammation by coronavirus-19 (COVI-19 or SARS-CoV-2): anti-inflammatory strategies. J. Biol. Regul. Homeost. Agents. 2020 doi: 10.23812/CONTI-E.
    1. Runfeng L., Yunlong H., Jicheng H., Weiqi P., Qinhai M., Yongxia S., Chufang L., Jin Z., Zhenhua J., Haiming J., Kui Z., Shuxiang H., Jun D., Xiaobo L., Xiaotao H., Lin W., Nanshan Z., Zifeng Y. Lianhuaqingwen exerts anti-viral and anti-inflammatory activity against novel coronavirus (SARS-CoV-2) Pharmacol. Res. 2020;156 doi: 10.1016/j.phrs.2020.104761.
    1. Li Q., Pang P., Zheng K., Sun L., Wang J., Chen X. Xin-Jia-Xiang-Ru-Yin alleviated H1N1-induced acute lung injury and inhibited the IFN-gamma-related regulatory pathway in summer flu. Biomed. Pharmacother. 2018;108:201–207. doi: 10.1016/j.biopha.2018.09.022.
    1. Guo C., Xie S., Chi Z., Zhang J., Liu Y., Zhang L., Zheng M., Zhang X., Xia D., Ke Y., Lu L., Wang D. Bile acids control inflammation and metabolic disorder through inhibition of NLRP3 inflammasome. Immunity. 2016;45:802–816. doi: 10.1016/j.immuni.2016.09.008.
    1. Yu Y., Tian Z.Q., Liang L., Yang X., Sheng D.D., Zeng J.X., Li X.Y., Shi R.Y., Han Z.P., Wei L.X. Babao Dan attenuates acute ethanol-induced liver injury via Nrf2 activation and autophagy. Cell Biosci. 2019;9:80. doi: 10.1186/s13578-019-0343-6.
    1. Sheng D., Zhao S., Gao L., Zheng H., Liu W., Hou J., Jin Y., Ye F., Zhao Q., Li R., Zhao N., Zhang L., Han Z., Wei L. BabaoDan attenuates high-fat diet-induced non-alcoholic fatty liver disease via activation of AMPK signaling. Cell Biosci. 2019;9:77. doi: 10.1186/s13578-019-0339-2.
    1. Lu L., Wu C., Lu B.J., Xie D., Wang Z., Azami L., An Y.T., Wang H.J., Ye G., Sun M.Y. Babaodan cures hepatic encephalopathy by decreasing ammonia levels and alleviating inflammation. J. Ethnopharmacol. 2019 doi: 10.1016/j.jep.2019.112301.
    1. Gong W., Liu L., Li M., Wang L., Zhang M., Luo Z., Sridhar S., Woo P.C.Y., Wang L. Evaluation of antiviral efficacy of Chinese traditional medicine Babao Dan in rabbits infected with hepatitis E virus. J. Gen. Virol. 2018;99:1036–1043. doi: 10.1099/jgv.0.001089.
    1. Liang L., Yang X., Yu Y., Li X., Wu Y., Shi R., Jiang J., Gao L., Ye F., Zhao Q., Li R., Wei L., Han Z. Babao Dan attenuates hepatic fibrosis by inhibiting hepatic stellate cells activation and proliferation via TLR4 signaling pathway. Oncotarget. 2016;7:82554–82566. doi: 10.18632/oncotarget.12783.
    1. Wang Q., Liu Z., Du K., Liang M., Zhu X., Yu Z., Chen R., Qin L., Li Y., Zheng Y. Babaodan inhibits cell growth by inducing autophagy through the PI3K/AKT/mTOR pathway and enhances antitumor effects of cisplatin in NSCLC cells. Am. J. Transl. Res. 2019;11:5272–5283.
    1. Du Y., Xin L., Shi Y., Zhang T.H., Wu N.C., Dai L., Gong D., Brar G., Shu S., Luo J., Reiley W., Tseng Y.W., Bai H., Wu T.T., Wang J., Shu Y., Sun R. Genome-wide identification of interferon-sensitive mutations enables influenza vaccine design. Science. 2018;359:290–296. doi: 10.1126/science.aan8806. (80-.)
    1. Liu Y., Hong Z., Qian J., Wang Y., Wang S. Protective effect of Jie-Geng-Tang against Staphylococcus aureus induced acute lung injury in mice and discovery of its effective constituents. J. Ethnopharmacol. 2019;243 doi: 10.1016/j.jep.2019.112076.
    1. Qian J., Hu Y., Zhao L., Xia J., Li C., Shi L., Xu F. Protective role of adipose-derived stem cells in staphylococcus aureus-induced lung injury is mediated by RegIIIgamma secretion. Stem Cells. 2016;34:1947–1956. doi: 10.1002/stem.2337.
    1. Smith A.M., Adler F.R., Ribeiro R.M., Gutenkunst R.N., McAuley J.L., McCullers J.A., Perelson A.S. Kinetics of coinfection with influenza A virus and Streptococcus pneumoniae. PLoS Pathog. 2013;9 doi: 10.1371/journal.ppat.1003238.
    1. Wang J., Li F., Sun R., Gao X., Wei H., Li L.J., Tian Z. Bacterial colonization dampens influenza-mediated acute lung injury via induction of M2 alveolar macrophages. Nat. Commun. 2013;4:2106. doi: 10.1038/ncomms3106.
    1. Kim D., Paggi J.M., Park C., Bennett C., Salzberg S.L. Graph-based genome alignment and genotyping with HISAT2 and HISAT-genotype. Nat. Biotechnol. 2019;37:907–915. doi: 10.1038/s41587-019-0201-4.
    1. Li B., Dewey C.N. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMC Bioinform. 2011;12:323. doi: 10.1186/1471-2105-12-323.
    1. Yu G., Wang L.G., Han Y., He Q.Y. clusterProfiler: an R package for comparing biological themes among gene clusters. OMICS. 2012;16:284–287. doi: 10.1089/omi.2011.0118.
    1. Gotea V., Ovcharenko I. DiRE: identifying distant regulatory elements of co-expressed genes. Nucleic Acids Res. 2008;36:W133–W139. doi: 10.1093/nar/gkn300.
    1. Schmolke M., Viemann D., Roth J., Ludwig S. Essential impact of NF-kappaB signaling on the H5N1 influenza A virus-induced transcriptome. J. Immunol. 2009;183:5180–5189. doi: 10.4049/jimmunol.0804198.
    1. Droebner K., Reiling S.J., Planz O. Role of hypercytokinemia in NF-kappaB p50-deficient mice after H5N1 influenza A virus infection. J. Virol. 2008;82:11461–11466. doi: 10.1128/JVI.01071-08.
    1. Li Y., Yu J., Zhao Y.H., An Y.T., Xue D., Chai Y.F., Zhang G.Q., Zhang H. UHPLC-Q-TOF-MS in analyzing chemical constituents of traditional Chinese herbal preparation Babao Dan. Acad. J. Second Mil. Univ. 2016;37:1548–1554. doi: 10.16781/j.0258-879x.2016.12.1548.
    1. Guo C., Chi Z., Jiang D., Xu T., Yu W., Wang Z., Chen S., Zhang L., Liu Q., Guo X., Zhang X., Li W., Lu L., Wu Y., Song B.L., Wang D. Cholesterol homeostatic regulator SCAP-SREBP2 integrates NLRP3 inflammasome activation and cholesterol biosynthetic signaling in macrophages. Immunity. 2018;49:842–856. doi: 10.1016/j.immuni.2018.08.021.
    1. Guo C., Xie S., Chi Z., Zhang J., Liu Y., Zhang L., Zheng M., Zhang X., Xia D., Ke Y., Lu L., Wang D. Bile acids control inflammation and metabolic disorder through inhibition of NLRP3 inflammasome. Immunity. 2016;45:802–816. doi: 10.1016/j.immuni.2016.09.008.
    1. Li Y., Huang B., Ye T., Wang Y., Xia D., Qian J. Physiological concentrations of bilirubin control inflammatory response by inhibiting NF-κB and inflammasome activation. Int. Immunopharmacol. 2020;84 doi: 10.1016/j.intimp.2020.106520.

Source: PubMed

3
Předplatit