Single-cell RNA sequencing reveals reduced intercellular adhesion molecule crosstalk between activated hepatic stellate cells and neutrophils alleviating liver fibrosis in hepatitis B virus transgenic mice post menstrual blood-derived mesenchymal stem cell transplantation

Lijun Chen , Yuqi Huang , Ning Zhang , Jingjing Qu , Yangxin Fang , Jiamin Fu , Yin Yuan , Qi Zhang , Hang Li , Zuoshi Wen , Li Yuan , Lu Chen , Zhenyu Xu , Yifei Li , Huadong Yan , Hiromi Izawa , Lanjuan Li , Charlie Xiang

MedComm ›› 2024, Vol. 5 ›› Issue (8) : e654

PDF
MedComm ›› 2024, Vol. 5 ›› Issue (8) : e654 DOI: 10.1002/mco2.654
ORIGINAL ARTICLE

Single-cell RNA sequencing reveals reduced intercellular adhesion molecule crosstalk between activated hepatic stellate cells and neutrophils alleviating liver fibrosis in hepatitis B virus transgenic mice post menstrual blood-derived mesenchymal stem cell transplantation

Author information +
History +
PDF

Abstract

Liver fibrosis can cause hepatitis B virus (HBV)-associated hepatocellular carcinoma. Menstrual blood-derived mesenchymal stem cells (MenSCs) can ameliorate liver fibrosis through paracrine. Single-cell RNA sequencing (scRNA-seq) may be used to explore the roadmap of activated hepatic stellate cell (aHSC) inactivation to target liver fibrosis. This study established HBV transgenic (HBV-Tg) mouse model of carbon tetrachloride (CCl4)-induced liver fibrosis and demonstrated that MenSCs migrated to the injured liver to improve serological indices and reduce fibrotic accumulation. RNA-bulk analysis revealed that MenSCs mediated extracellular matrix accumulation and cell adhesion. Liver parenchymal cells and nonparenchymal cells were identified by scRNA-seq in the control, CCl4, and MenSC groups, revealing the heterogeneity of fibroblasts/HSCs. A CellChat analysis revealed that diminished intercellular adhesion molecule (ICAM) signaling is vital for MenSC therapy. Specifically, Icam1 in aHSCs acted on Itgal/Itgb2 and Itgam/Itgb2 in neutrophils, causing decreased adhesion. The expression of Itgal, Itgam, and Itgb2 was higher in CCl4 group than in the control group and decreased after MenSC therapy in neutrophil clusters. The Lcn2, Pglyrp1, Wfdc21, and Mmp8 had high expression and may be potential targets in neutrophils. This study highlights interacting cells, corresponding molecules, and underlying targets for MenSCs in treating HBV-associated liver fibrosis.

Keywords

hepatic stellate cell (HSC) / HSC and neutrophil crosstalk / liver fibrosis in HBV-Tg mouse / menstrual blood-derived MSC / single-cell RNA sequencing

Cite this article

Download citation ▾
Lijun Chen, Yuqi Huang, Ning Zhang, Jingjing Qu, Yangxin Fang, Jiamin Fu, Yin Yuan, Qi Zhang, Hang Li, Zuoshi Wen, Li Yuan, Lu Chen, Zhenyu Xu, Yifei Li, Huadong Yan, Hiromi Izawa, Lanjuan Li, Charlie Xiang. Single-cell RNA sequencing reveals reduced intercellular adhesion molecule crosstalk between activated hepatic stellate cells and neutrophils alleviating liver fibrosis in hepatitis B virus transgenic mice post menstrual blood-derived mesenchymal stem cell transplantation. MedComm, 2024, 5(8): e654 DOI:10.1002/mco2.654

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

Devarbhavi H, Asrani SK, Arab JP, et al. Global burden of liver disease: 2023 update. J Hepatol. 2023; 79: 516-537.

[2]

Xiao J, Wang F, Wong NK, et al. Global liver disease burdens and research trends: analysis from a Chinese perspective. J Hepatol. 2019; 71: 212-221.

[3]

Bram Y, Nguyen DT, Gupta V, et al. Cell and tissue therapy for the treatment of chronic liver disease. Annu Rev Biomed Eng. 2021; 23: 517-546.

[4]

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

[5]

Weng S-C, Zhou J, Zhong X-Y, et al. Assessing the diagnostic accuracy of SD BIOLINE for detection of hepatitis C virus: a systematic review and meta-analysis. Infectious Microbes and Diseases. 2023; 5: 76-82.

[6]

Hernandez-Gea V, Friedman SL. Pathogenesis of liver fibrosis. Annu Rev Pathol. 2011; 6: 425-456.

[7]

Huang DQ, Mathurin P, Cortez-Pinto H, et al. Global epidemiology of alcohol-associated cirrhosis and HCC: trends, projections and risk factors. Nat Rev Gastroenterol Hepatol. 2023; 20: 37-49.

[8]

Altamirano-Barrera A, Barranco-Fragoso B, Mendez-Sanchez N. Management strategies for liver fibrosis. Ann Hepatol. 2017; 16: 48-56.

[9]

Huppert SS, Schwartz RE. Multiple facets of cellular homeostasis and regeneration of the mammalian liver. Annu Rev Physiol. 2023; 85: 469-493.

[10]

Lua I, Li Y, Zagory JA, et al. Characterization of hepatic stellate cells, portal fibroblasts, and mesothelial cells in normal and fibrotic livers. J Hepatol. 2016; 64: 1137-1146.

[11]

Kim HY, Sakane S, Eguileor A, et al. The origin and fate of liver myofibroblasts. Cell Mol Gastroenterol Hepatol. 2024; 17: 93-106.

[12]

Iwaisako K, Jiang C, Zhang M, et al. Origin of myofibroblasts in the fibrotic liver in mice. Proc Natl Acad Sci USA. 2014; 111: E3297-3305.

[13]

Nishio T, Koyama Y, Liu X, et al. Immunotherapy-based targeting of MSLN(+) activated portal fibroblasts is a strategy for treatment of cholestatic liver fibrosis. Proc Natl Acad Sci USA. 2021; 118: e2101270118.

[14]

Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol. 2017; 14: 397-411.

[15]

Higashi T, Friedman SL, Hoshida Y. Hepatic stellate cells as key target in liver fibrosis. Adv Drug Deliv Rev. 2017; 121: 27-42.

[16]

Fabre T, Molina MF, Soucy G, et al. Type 3 cytokines IL-17A and IL-22 drive TGF-beta-dependent liver fibrosis. Sci Immunol. 2018; 3: eaar7754.

[17]

Mantovani A, Cassatella MA, Costantini C, et al. Neutrophils in the activation and regulation of innate and adaptive immunity. Nat Rev Immunol. 2011; 11: 519-531.

[18]

Que H, Fu Q, Lan T, et al. Tumor-associated neutrophils and neutrophil-targeted cancer therapies. Biochim Biophys Acta Rev Cancer. 2022; 1877: 188762.

[19]

Tang S, Zhang J, Zhang L, et al. Knockdown of CXCL1 improves ACLF by reducing neutrophil recruitment to attenuate ROS production and hepatocyte apoptosis. Hepatol Commun. 2023; 7: e0257.

[20]

Liu K, Wang FS, Xu R. Neutrophils in liver diseases: pathogenesis and therapeutic targets. Cell Mol Immunol. 2021; 18: 38-44.

[21]

Zhou T, Kiran M, Lui KO, et al. Decoding liver fibrogenesis with single-cell technologies. Life Med. 2022; 1: 333-344.

[22]

Zhou J, Feng X, Zhu J, et al. Mesenchymal stem cell treatment restores liver macrophages homeostasis to alleviate mouse acute liver injury revealed by single-cell analysis. Pharmacol Res. 2022; 179: 106229.

[23]

Zou J, Li J, Zhong X, et al. Liver in infections: a single-cell and spatial transcriptomics perspective. J Biomed Sci. 2023; 30: 53.

[24]

Lin P, Yan X, Jing S, et al. Single-cell and spatially resolved transcriptomics for liver biology. Hepatology. 2023. doi:10.1097/HEP.0000000000000387. Epub ahead of print.

[25]

Cheng S, Zou Y, Zhang M, et al. Single-cell RNA sequencing reveals the heterogeneity and intercellular communication of hepatic stellate cells and macrophages during liver fibrosis. MedComm. 2023; 4: e378.

[26]

Jiang P, Jia H, Qian X, et al. Single-cell RNA sequencing reveals the immunoregulatory roles of PegIFN-alpha in patients with chronic hepatitis B. Hepatology. 2024; 79: 167-182.

[27]

El Agha E, Kramann R, Schneider RK, et al. Mesenchymal stem cells in fibrotic disease. Cell Stem Cell. 2017; 21: 166-177.

[28]

Krenkel O, Hundertmark J, Ritz TP, et al. Single cell RNA sequencing identifies subsets of hepatic stellate cells and myofibroblasts in liver fibrosis. Cells. 2019; 8: 503.

[29]

Yang W, He H, Wang T, et al. Single-cell transcriptomic analysis reveals a hepatic stellate cell-activation roadmap and myofibroblast origin during liver fibrosis in mice. Hepatology. 2021; 74: 2774-2790.

[30]

Bai YM, Yang F, Luo P, et al. Single-cell transcriptomic dissection of the cellular and molecular events underlying the triclosan-induced liver fibrosis in mice. Mil Med Res. 2023; 10: 7.

[31]

Zhang W, Conway SJ, Liu Y, et al. Heterogeneity of hepatic stellate cells in fibrogenesis of the liver: insights from single-cell transcriptomic analysis in liver injury. Cells. 2021; 10: 2129.

[32]

Zisser A, Ipsen DH, Tveden-Nyborg P. Hepatic stellate cell activation and inactivation in NASH-fibrosis-roles as putative treatment targets? Biomedicines. 2021; 9: 365.

[33]

Rosenthal SB, Liu X, Ganguly S, et al. Heterogeneity of HSCs in a mouse model of NASH. Hepatology. 2021; 74: 667-685.

[34]

Liu X, Xu J, Rosenthal S, et al. Identification of lineage-specific transcription factors that prevent activation of hepatic stellate cells and promote fibrosis resolution. Gastroenterology. 2020; 158: 1728-1744. e1714.

[35]

Zhou Z, Xu MJ, Cai Y, et al. Neutrophil-hepatic stellate cell interactions promote fibrosis in experimental steatohepatitis. Cell Mol Gastroenterol Hepatol. 2018; 5: 399-413.

[36]

Bui TM, Wiesolek HL, Sumagin R. ICAM-1: a master regulator of cellular responses in inflammation, injury resolution, and tumorigenesis. J Leukoc Biol. 2020; 108: 787-799.

[37]

Li N, Yang H, Wang M, et al. Ligand-specific binding forces of LFA-1 and Mac-1 in neutrophil adhesion and crawling. Mol Biol Cell. 2018; 29: 408-418.

[38]

LaFlamme SE, Mathew-Steiner S, Singh N, et al. Integrin and microtubule crosstalk in the regulation of cellular processes. Cell Mol Life Sci. 2018; 75: 4177-4185.

[39]

de Jesus AA, Chen G, Yang D, et al. Constitutively active Lyn kinase causes a cutaneous small vessel vasculitis and liver fibrosis syndrome. Nat Commun. 2023; 14: 1502.

[40]

Brown C, McKee C, Bakshi S, et al. Mesenchymal stem cells: cell therapy and regeneration potential. J Tissue Eng Regen Med. 2019; 13: 1738-1755.

[41]

Wang Y, Fang J, Liu B, et al. Reciprocal regulation of mesenchymal stem cells and immune responses. Cell Stem Cell. 2022; 29: 1515-1530.

[42]

Yang X, Li Q, Liu W, et al. Mesenchymal stromal cells in hepatic fibrosis/cirrhosis: from pathogenesis to treatment. Cell Mol Immunol. 2023; 20: 583-599.

[43]

Chen L, Zhang N, Huang Y, et al. Multiple dimensions of using mesenchymal stem cells for treating liver diseases: from bench to beside. Stem Cell Rev Rep. 2023; 19: 2192-2224.

[44]

Li TT, Wang ZR, Yao WQ, et al. Stem cell therapies for chronic liver diseases: progress and challenges. Stem Cells Transl Med. 2022; 11: 900-911.

[45]

Chen L, Qu J, Cheng T, et al. Menstrual blood-derived stem cells: toward therapeutic mechanisms, novel strategies, and future perspectives in the treatment of diseases. Stem Cell Res Ther. 2019; 10: 406.

[46]

Meng X, Ichim TE, Zhong J, et al. Endometrial regenerative cells: a novel stem cell population. J Transl Med. 2007; 5: 57.

[47]

Chen L, Zhang C, Chen L, et al. Human menstrual blood-derived stem cells ameliorate liver fibrosis in mice by targeting hepatic stellate cells via paracrine mediators. Stem Cells Transl Med. 2017; 6: 272-284.

[48]

Tang J, Zhang J, Zhang G, et al. Stat3 activation-triggered transcriptional networks govern the early stage of HBV-induced hepatic inflammation. mBio. 2024; 15: e0306823.

[49]

Bedossa P, Poitou C, Veyrie N, et al. Histopathological algorithm and scoring system for evaluation of liver lesions in morbidly obese patients. Hepatology. 2012; 56: 1751-1759.

[50]

He XL, Chen JY, Feng YL, et al. Single-cell RNA sequencing deciphers the mechanism of sepsis-induced liver injury and the therapeutic effects of artesunate. Acta Pharmacol Sin. 2023; 44: 1801-1814.

[51]

Halpern KB, Shenhav R, Matcovitch-Natan O, et al. Single-cell spatial reconstruction reveals global division of labour in the mammalian liver. Nature. 2017; 542: 352-356.

[52]

Liang Y, Kaneko K, Xin B, et al. Temporal analyses of postnatal liver development and maturation by single-cell transcriptomics. Dev Cell. 2022; 57: 398-414. e395.

[53]

Buechler MB, Pradhan RN, Krishnamurty AT, et al. Cross-tissue organization of the fibroblast lineage. Nature. 2021; 593: 575-579.

[54]

Hautz T, Salcher S, Fodor M, et al. Immune cell dynamics deconvoluted by single-cell RNA sequencing in normothermic machine perfusion of the liver. Nat Commun. 2023; 14: 2285.

[55]

Jung SH, Hwang BH, Shin S, et al. Spatiotemporal dynamics of macrophage heterogeneity and a potential function of Trem2(hi) macrophages in infarcted hearts. Nat Commun. 2022; 13: 4580.

[56]

Zhang X, Zeng Y, Zhao L, et al. Targeting hepatic stellate cell death to reverse hepatic fibrosis. Curr Drug Targets. 2023; 24: 568-583.

[57]

Ma L, Yang X, Wei R, et al. MicroRNA-214 promotes hepatic stellate cell activation and liver fibrosis by suppressing Sufu expression. Cell Death Dis. 2018; 9: 718.

[58]

Ma L, Wei J, Zeng Y, et al. Mesenchymal stem cell-originated exosomal circDIDO1 suppresses hepatic stellate cell activation by miR-141-3p/PTEN/AKT pathway in human liver fibrosis. Drug Deliv. 2022; 29: 440-453.

[59]

Lin Y, Yan M, Bai Z, et al. Huc-MSC-derived exosomes modified with the targeting peptide of aHSCs for liver fibrosis therapy. J Nanobiotechnol. 2022; 20: 432.

[60]

Shi M, Li YY, Xu RN, et al. Mesenchymal stem cell therapy in decompensated liver cirrhosis: a long-term follow-up analysis of the randomized controlled clinical trial. Hepatol Int. 2021; 15: 1431-1441.

[61]

Khoury M, Alcayaga-Miranda F, Illanes SE, et al. The promising potential of menstrual stem cells for antenatal diagnosis and cell therapy. Front Immunol. 2014; 5: 205.

[62]

Chen L, Qu J, Xiang C. The multi-functional roles of menstrual blood-derived stem cells in regenerative medicine. Stem Cell Res Ther. 2019; 10: 1.

[63]

Du J, Jiang Y, Liu X, et al. HGF secreted by menstrual blood-derived endometrial stem cells ameliorates non-alcoholic fatty liver disease through downregulation of hepatic Rnf186. Stem Cells. 2023; 41: 153-168.

[64]

Lv H, Hu Y, Cui Z, et al. Human menstrual blood: a renewable and sustainable source of stem cells for regenerative medicine. Stem Cell Res Ther. 2018; 9: 325.

[65]

Xu X, Jiang W, Chen L, et al. Evaluation of the safety and efficacy of using human menstrual blood-derived mesenchymal stromal cells in treating severe and critically ill COVID-19 patients: an exploratory clinical trial. Clin Transl Med. 2021; 11: e297.

[66]

Fathi-Kazerooni M, Fattah-Ghazi S, Darzi M, et al. Safety and efficacy study of allogeneic human menstrual blood stromal cells secretome to treat severe COVID-19 patients: clinical trial phase I & II. Stem Cell Res Ther. 2022; 13: 96.

[67]

Xu X, Poulsen KL, Wu L, et al. Targeted therapeutics and novel signaling pathways in non-alcohol-associated fatty liver/steatohepatitis (NAFL/NASH). Signal Transduct Target Ther. 2022; 7: 287.

[68]

Nishio T, Hu R, Koyama Y, et al. Activated hepatic stellate cells and portal fibroblasts contribute to cholestatic liver fibrosis in MDR2 knockout mice. J Hepatol. 2019; 71: 573-585.

[69]

Hammerich L, Tacke F. Hepatic inflammatory responses in liver fibrosis. Nat Rev Gastroenterol Hepatol. 2023; 20: 633-646.

[70]

Hume DA, Millard SM, Pettit AR. Macrophage heterogeneity in the single-cell era: facts and artifacts. Blood. 2023; 142: 1339-1347.

[71]

Yang M, Ong J, Meng F, et al. Spatiotemporal insight into early pregnancy governed by immune-featured stromal cells. Cell. 2023; 186: 4271-4288. e4224.

[72]

Domcke S, Shendure J. A reference cell tree will serve science better than a reference cell atlas. Cell. 2023; 186: 1103-1114.

[73]

De Smet V, Eysackers N, Merens V, et al. Initiation of hepatic stellate cell activation extends into chronic liver disease. Cell Death Dis. 2021; 12: 1110.

[74]

Noom A, Sawitzki B, Knaus P, et al. A two-way street—cellular metabolism and myofibroblast contraction. NPJ Regen Med. 2024; 9: 15.

[75]

Wei Y, Zhang E, Yu L, et al. Dissecting embryonic and extraembryonic lineage crosstalk with stem cell co-culture. Cell. 2023; 186: 5859-5875. e5824.

[76]

Taylor NJ, Manakkat Vijay GK, Abeles RD, et al. The severity of circulating neutrophil dysfunction in patients with cirrhosis is associated with 90-day and 1-year mortality. Aliment Pharmacol Ther. 2014; 40: 705-715.

[77]

Hu J, Lin YY, Chen PJ, et al. Cell and animal models for studying hepatitis B virus infection and drug development. Gastroenterology. 2019; 156: 338-354.

[78]

Gulati GS, Sikandar SS, Wesche DJ, et al. Single-cell transcriptional diversity is a hallmark of developmental potential. Science. 2020; 367: 405-411.

[79]

Afonso MB, Marques V, van Mil SWC, et al. Human liver organoids: from generation to applications. Hepatology. 2024; 79: 1432-1451.

[80]

Guan H, Zhu N, Tang G, et al. DNA methyltransferase 1 knockdown reverses PTEN and VDR by mediating demethylation of promoter and protects against renal injuries in hepatitis B virus-associated glomerulonephritis. Cell Biosci. 2022; 12: 98.

[81]

Zhou S, Liu Y, Zhang Q, et al. Human menstrual blood-derived stem cells reverse sorafenib resistance in hepatocellular carcinoma cells through the hyperactivation of mitophagy. Stem Cell Res Ther. 2023; 14: 58.

[82]

Chen X, Wu Y, Wang Y, et al. Human menstrual blood-derived stem cells mitigate bleomycin-induced pulmonary fibrosis through anti-apoptosis and anti-inflammatory effects. Stem Cell Res Ther. 2020; 11: 477.

[83]

Robinson MD, Oshlack A. A scaling normalization method for differential expression analysis of RNA-seq data. Genome Biol. 2010; 11: R25.

[84]

Do VH, Canzar S. A generalization of t-SNE and UMAP to single-cell multimodal omics. Genome Biol. 2021; 22: 130.

[85]

Korsunsky I, Millard N, Fan J, et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat Methods. 2019; 16: 1289-1296.

[86]

Harris MA, Clark J, Ireland A, et al. The Gene Ontology (GO) database and informatics resource. Nucleic Acids Res. 2004; 32: D258-261.

[87]

Kanehisa M, Goto S, Hattori M, et al. From genomics to chemical genomics: new developments in KEGG. Nucleic Acids Res. 2006; 34: D354-357.

RIGHTS & PERMISSIONS

2024 The Author(s). MedComm published by Sichuan International Medical Exchange & Promotion Association (SCIMEA) and John Wiley & Sons Australia, Ltd.

AI Summary AI Mindmap
PDF

193

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/