The transcriptome of MHV-infected RAW264.7 cells offers an alternative model for macrophage innate immunity research

Xue Wang , Chunyou Ning , Xingyi Cheng , Zhengzhong Wu , Dongbo Wu , Xuemei Ding , Cunxiang Ju , Zhihang Zhou , Lingfeng Wan , Wei Zhao , Peiliang Shi

Animal Models and Experimental Medicine ›› 2025, Vol. 8 ›› Issue (1) : 114 -125.

PDF (9482KB)
Animal Models and Experimental Medicine ›› 2025, Vol. 8 ›› Issue (1) : 114 -125. DOI: 10.1002/ame2.12506
ORIGINAL ARTICLE

The transcriptome of MHV-infected RAW264.7 cells offers an alternative model for macrophage innate immunity research

Author information +
History +
PDF (9482KB)

Abstract

Background: The emerging incidence of pathogenic liver conditions is turning into a major concern for global health. Induction of pyroptosis in hepatocytes instigates cellular disintegration, which in turn liberates substantial quantities of pro-inflammatory intracellular substances, thereby accelerating the advancement of liver fibrosis. Consequently, directing therapeutic efforts towards inhibiting pyroptosis could potentially serve as an innovative approach in managing inflammation related chronic hepatic disorders.

Methods: GSDMD-NTki/wt mice and Alb-creki/wt mice were generated using CRISPR/9 technology. After crossing the two strains together, we induced conditional cell death by doxycycline to construct a mouse model of liver fibrosis. We analyzed differentially expressed genes by RNA sequencing and explored their biological functions. The efficacy of obeticholic acid (OCA) in the treatment of liver fibrosis was assessed.

Results: Doxycycline-treated GSDMD-NTki/wt×Alb-creki/wt mice showed severe liver damage, vacuolation of hepatocytes, increased collagen fibers, and accumulation of lipid droplets. The expression of liver fibrosis related genes was greatly increased in the doxycycline-treated mouse liver compared with untreated mouse liver. RNA-sequencing showed that upregulated differentially expressed genes were involved in inflammatory responses, cell activation, and metabolic processes. Treatment with OCA alleviated the liver fibrosis, with reduced ALT and AST levels seen in the GSDMD-NTki/wt×Alb-creki/wt mice.

Conclusions: We successfully constructed a novel mouse model for liver fibrosis. This GSDMD-NT-induced fibrosis may be mediated by abnormal lipid metabolism. Our results demonstrated that we successfully constructed a mouse model of liver fibrosis, and GSDMD-NT induced fibrosis by mediating lipid metabolism.

Keywords

GSDMD-NT / lipid metabolism / liver fibrosis / NASH / pyroptosis

Cite this article

Download citation ▾
Xue Wang, Chunyou Ning, Xingyi Cheng, Zhengzhong Wu, Dongbo Wu, Xuemei Ding, Cunxiang Ju, Zhihang Zhou, Lingfeng Wan, Wei Zhao, Peiliang Shi. The transcriptome of MHV-infected RAW264.7 cells offers an alternative model for macrophage innate immunity research. Animal Models and Experimental Medicine, 2025, 8(1): 114-125 DOI:10.1002/ame2.12506

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Tripodi A, Mannucci PM. The coagulopathy of chronic liver disease. N Engl J Med. 2011;365:147-156.

[2]

Xuan LJ, Bin LB, Fen ZL, Yu YH, Hospital C. Relationship between lipid metabolism and liver fibrosis: an update. Acad J Second Mil Univ. 2018;39:531-534.

[3]

Hwang S, Chung KW. Targeting fatty acid metabolism for fibrotic disorders. Arch Pharm Res. 2021;44:839-856.

[4]

Mehal W. Mechanisms of liver fibrosis in metabolic syndrome. eGastroenterology. 2023;1:e100015.

[5]

Kaufmann B, Reca A, Wang B, Friess H, Feldstein AE, Hartmann D. Mechanisms of nonalcoholic fatty liver disease and implications for surgery. Langenbeck’s Arch Surg. 2021;406:1-17.

[6]

Wang W, Tan J, Liu X, et al. Cytoplasmic endonuclease G promotes nonalcoholic fatty liver disease via mTORC2-AKT-ACLY and endoplasmic reticulum stress. Nat Commun. 2023;14:6201.

[7]

Seitz HK, Bataller R, Cortez-Pinto H, et al. Alcoholic liver disease. Nat Rev Dis Primers. 2018;4:16.

[8]

Kardashian A, Serper M, Terrault N, Nephew LD. Health disparities in chronic liver disease. Hepatology. 2023;77:1382-1403.

[9]

Bugianesi E, Petta S. Nafld/Nash. J Hepatol. 2022;77:549-550.

[10]

Harrison SA, Allen AM, Dubourg J, Noureddin M, Alkhouri N. Challenges and opportunities in NASH drug development. Nat Med. 2023;29:562-573.

[11]

Chapman RW, Lynch KD. Obeticholic acid—a new therapy in PBC and NASH. Br Med Bull. 2020;133:95-104.

[12]

Zhou J, Huang N, Guo Y, et al. Combined obeticholic acid and apoptosis inhibitor treatment alleviates liver fibrosis. Acta Pharm Sin B. 2019;9:526-536.

[13]

Younossi ZM, Ratziu V, Loomba R, et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 2019;394:2184-2196.

[14]

Shi J, Zhao Y, Wang K, et al. Cleavage of GSDMD by inflammatory pases determines pyroptotic cell death. Nature. 2015;526:660-665.

[15]

Csak T, Pillai A, Ganz M, et al. Both bone marrow-derived and non-bone marrow-derived cells contribute to AIM2 and NLRP3 inflammasome activation in a MyD88-dependent manner in dietary steatohepatitis. Liver Int. 2014;34:1402-1413.

[16]

Wree A, Eguchi A, McGeough MD, et al. NLRP3 inflammasome activation results in hepatocyte pyroptosis, liver inflammation, and fibrosis in mice. Hepatology. 2014;59:898-910.

[17]

Szabo G, Petrasek J. Inflammasome activation and function in liver disease. Nat Rev Gastroenterol Hepatol. 2015;12:387-400.

[18]

Wang K, Sun Q, Zhong X, et al. Structural mechanism for GSDMD targeting by autoprocessed pases in pyroptosis. Cell. 2020;180:941-955. e20.

[19]

Karmakar M, Minns M, Greenberg EN, et al. N-GSDMD trafficking to neutrophil organelles facilitates IL-1β release independently of plasma membrane pores and pyroptosis. Nat Commun. 2020;11:2212.

[20]

Xiao Y, Zhao C, Tai Y, et al. STING mediates hepatocyte pyroptosis in liver fibrosis by epigenetically activating the NLRP3 inflammasome. Redox Biol. 2023;62:102691.

[21]

Gaul S, Leszczynska A, Alegre F, et al. Hepatocyte pyroptosis and release of inflammasome particles induce stellate cell activation and liver fibrosis. J Hepatol. 2021;74:156-167.

[22]

de Carvalho Ribeiro M, Szabo G. Role of the inflammasome in liver disease. Annu Rev Pathol. 2022;17:345-365.

[23]

Shi J, Gao W, Shao F. Pyroptosis: Gasdermin-mediated programmed necrotic cell death. Trends Biochem Sci. 2017;42:245-254.

[24]

Man SM, Kanneganti TD. Gasdermin D: the long-awaited executioner of pyroptosis. Cell Res. 2015;25:1183-1184.

[25]

Chen TX, Meng Y, Zhou ZH, et al. GAS5 protects against nonalcoholic fatty liver disease via miR-28a-5p/MARCH7/NLRP3 axis-mediated pyroptosis. Cell Death Differ. 2023;30:1829-1848.

[26]

Kim D, Langmead B, Salzberg SL. HISAT: a fast spliced aligner with low memory requirements. Nat Methods. 2015;12:357-360.

[27]

Pertea M, Pertea GM, Antonescu CM, Chang TC, Mendell JT, Salzberg SL. StringTie enables improved reconstruction of a transcriptome from RNA-seq reads. Nat Biotechnol. 2015;33:290-295.

[28]

Robinson MD, McCarthy DJ, Smyth GK. edgeR: a bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics (Oxford, England). 2010;26:139-140.

[29]

Wu T, Hu E, Xu S, et al. clusterProfiler 4.0:A universal enrichment tool for interpreting omics data. Innovation (Camb). 2021;2(3):100141.

[30]

Govaere O, Cockell S, Tiniakos D, et al. Transcriptomic profiling across the nonalcoholic fatty liver disease spectrum reveals gene signatures for steatohepatitis and fibrosis. Sci Transl Med. 2020;12:572.

[31]

Parola M, Pinzani M. Liver fibrosis: pathophysiology, pathogenetic targets and clinical issues. Mol Asp Med. 2019;65:37-55.

[32]

Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol. 2021;18:151-166.

[33]

Smith A, Baumgartner K, Bositis C. Cirrhosis: diagnosis and management. Am Fam Physician. 2019;100:759-770.

[34]

Wallach D, Kang TB, Dillon CP, Green DR. Programmed necrosis in inflammation: toward identification of the effector molecules. Science (New York, NY). 2016;352:aaf2154.

[35]

Kayagaki N, Stowe IB, Lee BL, et al. pase-11 cleaves gasdermin D for non-canonical inflammasome signalling. Nature. 2015;526:666-671.

[36]

Xiao J, Wang C, Yao JC, et al. Gasdermin D mediates the pathogenesis of neonatal-onset multisystem inflammatory disease in mice. PLoS Biol. 2018;16:e3000047.

[37]

Liu X, Xia S, Zhang Z, Wu H, Lieberman J. Channelling inflammation: gasdermins in physiology and disease. Nat Rev Drug Discov. 2021;20:384-405.

[38]

Wang Y, Gao W, Shi X, et al. Chemotherapy drugs induce pyroptosis through pase-3 cleavage of a gasdermin. Nature. 2017;547:99-103.

[39]

Humphries F, Shmuel-Galia L, Ketelut-Carneiro N, et al. Succination inactivates gasdermin D and blocks pyroptosis. Science (New York, NY). 2020;369:1633-1637.

[40]

Hirsova P, Ibrahim SH, Krishnan A, et al. Lipid-induced signaling causes release of inflammatory extracellular vesicles from hepatocytes. Gastroenterology. 2016;150:956-967.

[41]

Petrasek J, Bala S, Csak T, et al. IL-1 receptor antagonist ameliorates inflammasome-dependent alcoholic steatohepatitis in mice. J Clin Invest. 2012;122:3476-3489.

[42]

Miura K, Kodama Y, Inokuchi S, et al. Toll-like receptor 9 promotes steatohepatitis by induction of interleukin-1beta in mice. Gastroenterology. 2010;139:323-334.

[43]

Zhang X, Han J, Man K, et al. CXC chemokine receptor 3 promotes steatohepatitis in mice through mediating inflammatory cytokines, macrophages and autophagy. J Hepatol. 2016;64:160-170.

[44]

Amano SU, Cohen JL, Vangala P, et al. Local proliferation of macrophages contributes to obesity-associated adipose tissue inflammation. Cell Metab. 2014;19:162-171.

[45]

Hu C, Xin Z, Sun X, et al. Activation of ACLY by SEC63 deploys metabolic reprogramming to facilitate hepatocellular carcinoma metastasis upon endoplasmic reticulum stress. J Exp Clin Cancer Res. 2023;42:108.

[46]

Loomba R, Mohseni R, Lu KJ, et al. TVB-2640 (FASN inhibitor) for the treatment of nonalcoholic Steatohepatitis: FASCINATE-1, a randomized, placebo-controlled phase 2a trial. Gastroenterology. 2021;161:1475-1486.

[47]

Hua YQ, Zeng Y, Xu J, Xu XL. Naringenin alleviates nonalcoholic steatohepatitis in middle-aged Apoe(−/−)mice: role of SIRT1. Phytomedicine. 2021;81:153412.

[48]

Zhao J, Zhang Q, Zou G, Gao G, Yue Q. Arenobufagin, isolated from toad venom, inhibited epithelial-to-mesenchymal transition and suppressed migration and invasion of lung cancer cells via targeting IKKβ/NFκB signal cade. J Ethnopharmacol. 2020;250:112492.

[49]

Mann JP, Hoare M. A minority of somatically mutated genes in pre-existing fatty liver disease have prognostic importance in the development of NAFLD. Liver Int. 2022;42:1823-1835.

[50]

Peng L, Piekos S, Guo GL, Zhong XB. Role of farnesoid X receptor in establishment of ontogeny of phase-I drug metabolizing enzyme genes in mouse liver. Acta Pharm Sin B. 2016;6:453-459.

[51]

Zhu Y, Liu H, Zhang M, Guo GL. Fatty liver diseases, bile acids, and FXR. Acta Pharm Sin B. 2016;6:409-412.

[52]

Zhou J, Cui S, He Q, et al. SUMOylation inhibitors synergize with FXR agonists in combating liver fibrosis. Nat Commun. 2020;11:240.

[53]

Chiang JYL, Ferrell JM. Bile acid receptors FXR and TGR5 signaling in fatty liver diseases and therapy. Am J Physiol Gastrointest Liver Physiol. 2020;318:G554-g573.

[54]

Fiorucci S, Antonelli E, Rizzo G, et al. The nuclear receptor SHP mediates inhibition of hepatic stellate cells by FXR and protects against liver fibrosis. Gastroenterology. 2004;127:1497-1512.

[55]

Clifford BL, Sedgeman LR, Williams KJ, et al. FXR activation protects against NAFLD via bile-acid-dependent reductions in lipid absorption. Cell Metab. 2021;33:1671-1684.e4.

[56]

Lleo A, Wang GQ, Gershwin ME, Hirschfield GM. Primary biliary cholangitis. Lancet (London, England). 2020;396:1915-1926.

[57]

Rinella ME, Dufour JF, Anstee QM, et al. Non-invasive evaluation of response to obeticholic acid in patients with NASH: results from the REGENERATE study. J Hepatol. 2022;76:536-548.

[58]

Verbeke L, Farre R, Trebicka J, et al. Obeticholic acid, a farnesoid X receptor agonist, improves portal hypertension by two distinct pathways in cirrhotic rats. Hepatology (Baltimore, Md). 2014;59:2286-2298.

[59]

Huang S, Wu Y, Zhao Z, et al. A new mechanism of obeticholic acid on NASH treatment by inhibiting NLRP3 inflammasome activation in macrophage. Metab Clin Exp. 2021;120:154797.

[60]

Kumar S, Duan Q, Wu R, Harris EN, Su Q. Pathophysiological communication between hepatocytes and non-parenchymal cells in liver injury from NAFLD to liver fibrosis. Adv Drug Deliv Rev. 2021;176:113869.

[61]

Huby T, Gautier EL. Immune cell-mediated features of non-alcoholic steatohepatitis. Nat Rev Immunol. 2022;22:429-443.

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.

AI Summary AI Mindmap
PDF (9482KB)

203

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/