Journal Home Online First Current Issue Archive For Authors Journal Information 中文版

Engineering >> 2022, Volume 14, Issue 7 doi: 10.1016/j.eng.2020.12.013

Gut Microbiota-Controlled Tryptophan Metabolism Improves D-Gal/LPS-Induced Acute Liver Failure in C57BL/6 Mice

State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, China

# These authors contributed equally to this work.

Received: 2020-10-13 Revised: 2020-12-01 Accepted: 2020-12-28 Available online: 2021-02-07

Next Previous

Abstract

Acute liver failure (ALF) has an abrupt onset with a frequently fatal outcome. Previous studies have found that oral antibiotics prevent drug-induced liver injury in animal experiments, indicating that the gut microbiota plays a critical role in the pathophysiological process. However, the underlying mechanism has not been fully understood. This study explored the comprehensive role of the gut microbiota in ALF using multi-omics. A cocktail of broad-spectrum antibiotics (Abx) pretreatment by gavage for four weeks improved the survival of D-(+)-galactosamine hydrochloride (D-Gal)/lipopolysaccharide (LPS)-induced ALF in C57BL/6 mice. RNA sequencing showed that inflammatory responses were inhibited and metabolic pathways were upregulated in the liver of Abx-treated ALF mice. The 16S rRNA gene sequencing revealed that Abx reshaped the composition and function of the gut microbiota, with an increased proportion of tryptophan (Trp) metabolism. In addition, global metabolic profiling by ultra-performance liquid chromatography–mass spectrometry (UPLC–MS) indicated that the gut microbiota post-Abx intervention reduced Trp excretion, liberated more Trp to the host, and enhanced the kynurenine (Kyn) pathway with increased production of Kyn. As an endogenous aryl hydrocarbon receptor (AhR) ligand, Kyn has anti-inflammatory and immunosuppressive effects. Furthermore, AhR-targeted treatments affected the outcome of ALF mice with or without Abx pretreatment, indicating that AhR directly regulated susceptibility to ALF, at least in part. This study demonstrates that the gut microbiota-dependent control of the Trp metabolism could regulate host susceptibility to ALF by modulating the activity of AhR, and thus provides a promising target for better management of ALF.

SupplementaryMaterials

Figures

Fig. 1

Fig. 2

Fig. 3

Fig. 4

Fig. 5

Fig. 6

Fig. 7

Fig. 8

References

[ 1 ] Stravitz RT, Lee WM. Acute liver failure. Lancet 2019;394(10201):869–81. link1

[ 2 ] Nolan JP. The role of intestinal endotoxin in liver injury: a long and evolving history. Hepatology 2010;52(5):1829–35. link1

[ 3 ] Wei Y, Zeng B, Chen J, Cui G, Lu C, Wu W, et al. Enterogenous bacterial glycolipids are required for the generation of natural killer T cells mediated liver injury. Sci Rep 2016;6(1):36365. link1

[ 4 ] Sharon G, Garg N, Debelius J, Knight R, Dorrestein P, Mazmanian S. Specialized metabolites from the microbiome in health and disease. Cell Metab 2014;20 (5):719–30. link1

[ 5 ] Postler TS, Ghosh S. Understanding the holobiont: how microbial metabolites affect human health and shape the immune system. Cell Metab 2017;26 (1):110–30. link1

[ 6 ] Blacher E, Levy M, Tatirovsky E, Elinav E. Microbiome-modulated metabolites at the interface of host immunity. J Immunol 2017;198(2):572–80. link1

[ 7 ] Morgun A, Dzutsev A, Dong X, Greer RL, Sexton DJ, Ravel J, et al. Uncovering effects of antibiotics on the host and microbiota using transkingdom gene networks. Gut 2015;64(11):1732–43. link1

[ 8 ] Ubeda C, Pamer EG. Antibiotics, microbiota, and immune defense. Trends Immunol 2012;33(9):459–66. link1

[ 9 ] Zarrinpar A, Chaix A, Xu ZZ, Chang MW, Marotz CA, Saghatelian A, et al. Antibiotic-induced microbiome depletion alters metabolic homeostasis by affecting gut signaling and colonic metabolism. Nat Commun 2018;9 (1):2872. link1

[10] Clarke G, Grenham S, Scully P, Fitzgerald P, Moloney RD, Shanahan F, et al. The microbiome–gut–brain axis during early life regulates the hippocampal serotonergic system in a sex-dependent manner. Mol Psychiatry 2013;18 (6):666–73. link1

[11] Desbonnet L, Clarke G, Traplin A, O’Sullivan O, Crispie F, Moloney RD, et al. Gut microbiota depletion from early adolescence in mice: implications for brain and behaviour. Brain Behav Immun 2015;48:165–73. link1

[12] Agus A, Planchais J, Sokol H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe 2018;23(6):716–24. link1

[13] Cervenka I, Agudelo LZ, Ruas JL. Kynurenines: tryptophan’s metabolites in exercise, inflammation, and mental health. Science 2017;357(6349): eaaf9794. link1

[14] Clària J, Moreau R, Fenaille F, Amorós A, Junot C, Gronbaek H, et al; CANONIC Study Investigators of the EASL Clif Consortium, Grifols Chair and the European Foundation for the Study of Chronic Liver Failure (EF Clif). Orchestration of tryptophan–kynurenine pathway, acute decompensation, and acute-on-chronic liver failure in cirrhosis. Hepatology 2019;69 (4):1686–701. link1

[15] He B, Liu Y, Hoang TK, Tian X, Taylor CM, Luo M, et al. Antibiotic-modulated microbiome suppresses lethal inflammation and prolongs lifespan in Tregdeficient mice. Microbiome 2019;7(1):145. link1

[16] Nakamura K, Kageyama S, Ito T, Hirao H, Kadono K, Aziz A, et al. Antibiotic pretreatment alleviates liver transplant damage in mice and humans. J Clin Invest 2019;129(8):3420–34. link1

[17] Gehrke N, Hövelmeyer N, Waisman A, Straub BK, Weinmann-Menke J, Wörns MA, et al. Hepatocyte-specific deletion of IL1-RI attenuates liver injury by blocking IL-1 driven autoinflammation. J Hepatol 2018;68(5):986–95. link1

[18] Taniki N, Nakamoto N, Chu PS, Mikami Y, Amiya T, Teratani T, et al. Intestinal barrier regulates immune responses in the liver via IL-10-producing macrophages. JCI Insight 2018;3(12):e91980. link1

[19] Gong S, Lan T, Zeng L, Luo H, Yang X, Li N, et al. Gut microbiota mediates diurnal variation of acetaminophen induced acute liver injury in mice. J Hepatol 2018;69(1):51–9. link1

[20] Monteleone I, Rizzo A, Sarra M, Sica G, Sileri P, Biancone L, et al. Aryl hydrocarbon receptor-induced signals up-regulate IL-22 production and inhibit inflammation in the gastrointestinal tract. Gastroenterology 2011;141(1):237–48.e1. link1

[21] Wu W, Lv L, Shi D, Ye J, Fang D, Guo F, et al. Protective effect of Akkermansia muciniphila against immune-mediated liver injury in a mouse model. Front Microbiol 2017;8:1804. link1

[22] Cao H, Huang H, Xu W, Chen D, Yu J, Li J, et al. Fecal metabolome profiling of liver cirrhosis and hepatocellular carcinoma patients by ultra performance liquid chromatography–mass spectrometry. Anal Chim Acta 2011; 691(1–2):68–75. link1

[23] Huang Q, Tan Y, Yin P, Ye G, Gao P, Lu X, et al. Metabolic characterization of hepatocellular carcinoma using nontargeted tissue metabolomics. Cancer Res 2013;73(16):4992–5002. link1

[24] Simonato M, Fochi I, Vedovelli L, Giambelluca S, Carollo C, Padalino M, et al. Urinary metabolomics reveals kynurenine pathway perturbation in newborns with transposition of great arteries after surgical repair. Metabolomics 2019;15 (11):145. link1

[25] Comai S, Bertazzo A, Brughera M, Crotti S. Tryptophan in health and disease. Adv Clin Chem 2020;95:165–218. link1

[26] Hijikata Y, Hara K, Shiozaki Y, Murata K, Sameshima Y. Determination of free tryptophan in plasma and its clinical applications. J Clin Chem Clin Biochem 1984;22(4):291–9. link1

[27] Badawy A. Letter to the editor: the kynurenine pathway in hepatic cirrhosis. Hepatology 2019;70(3):1077–8. link1

[28] Günther J, Fallarino F, Fuchs D, Wirthgen E. Editorial: immunomodulatory roles of tryptophan metabolites in inflammation and cancer. Front Immunol 2020;11:1497. link1

[29] Sorgdrager FJH, Naudé PJW, Kema IP, Nollen EA, Deyn PPD. Tryptophan metabolism in inflammaging: from biomarker to therapeutic target. Front Immunol 2019;10:2565. link1

[30] Gutiérrez-Vázquez C, Quintana FJ. Regulation of the immune response by the aryl hydrocarbon receptor. Immunity 2018;48(1):19–33. link1

[31] Yin J, Sheng B, Qiu Y, Yang K, Xiao W, Yang H. Role of AhR in positive regulation of cell proliferation and survival. Cell Prolif 2016;49(5):554–60. link1

[32] Maes M, Vinken M, Jaeschke H. Experimental models of hepatotoxicity related to acute liver failure. Toxicol Appl Pharmacol 2016;290:86–97. link1

[33] Gegner JA, Ulevitch RJ, Tobias PS. Lipopolysaccharide (LPS) signal transduction and clearance dual roles for LPS binding protein and membrane CD14. J Biol Chem 1995;270(10):5320–5. link1

[34] Tripathi A, Debelius J, Brenner DA, Karin M, Loomba R, Schnabl B, et al. The gut–liver axis and the intersection with the microbiome. Nat Rev Gastroenterol Hepatol 2018;15(7):397–411. link1

[35] Heymann F, Tacke F. Immunology in the liver—from homeostasis to disease. Nat Rev Gastroenterol Hepatol 2016;13(2):88–110. link1

[36] Lee JH, Lee J. Indole as an intercellular signal in microbial communities. FEMS Microbiol Rev 2010;34(4):426–44. link1

[37] Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nat Commun 2018;9(1):3294. link1

[38] Mándi Y, Vécsei L. The kynurenine system and immunoregulation. J Neural Transm 2012;119(2):197–209. link1

[39] Frumento G, Rotondo R, Tonetti M, Damonte G, Benatti U, Ferrara GB. Tryptophan-derived catabolites are responsible for inhibition of T and natural killer cell proliferation induced by indoleamine 2,3-dioxygenase. J Exp Med 2002;196(4):459–68. link1

[40] Orabona C, Puccetti P, Vacca C, Bicciato S, Luchini A, Fallarino F, et al. Toward the identification of a tolerogenic signature in IDO-competent dendritic cells. Blood 2006;107(7):2846–54. link1

[41] Belladonna ML, Puccetti P, Orabona C, Fallarino F, Vacca C, Volpi C, et al. Immunosuppression via tryptophan catabolism: the role of kynurenine pathway enzymes. Transplantation 2007;84(1 Suppl):S17–20. link1

[42] Hill M, Tanguy-Royer S, Royer P, Chauveau C, Asghar K, Tesson L, et al. IDO expands human CD4+ CD25high regulatory T cells by promoting maturation of LPS-treated dendritic cells. Eur J Immunol 2007;37 (11):3054–62. link1

[43] Manni G, Mondanelli G, Scalisi G, Pallotta MT, Nardi D, Padiglioni E, et al. Pharmacologic induction of endotoxin tolerance in dendritic cells by lkynurenine. Front Immunol 2020;11:292. link1

[44] Esser C. The aryl hydrocarbon receptor in immunity: tools and potential. Methods Mol Biol 2016;1371:239–57. link1

[45] Neavin DR, Liu D, Ray B, Weinshilboum RM. The role of the aryl hydrocarbon receptor (AHR) in immune and inflammatory diseases. Int J Mol Sci 2018;19 (12):3851. link1

[46] Natividad JM, Agus A, Planchais J, Lamas B, Jarry AC, Martin R, et al. Impaired aryl hydrocarbon receptor ligand production by the gut microbiota is a key factor in metabolic syndrome. Cell Metab 2018;28(5):737–49.e4. link1

[47] Krishnan S, Ding Y, Saedi N, Choi M, Sridharan GV, Sherr DH, et al. Gut microbiota-derived tryptophan metabolites modulate inflammatory response in hepatocytes and macrophages. Cell Reports 2018;23 (4):1099–111. link1

[48] Hankinson O. Role of coactivators in transcriptional activation by the aryl hydrocarbon receptor. Arch Biochem Biophys 2005;433(2):379–86. link1

[49] Bessede A, Gargaro M, Pallotta MT, Matino D, Servillo G, Brunacci C, et al. Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature 2014;511(7508):184–90. link1

[50] Quintana FJ. LeA(H)Rning self-control. Cell Res 2014;24(10):1155–6. link1

[51] Beaumont M, Neyrinck AM, Olivares M, Rodriguez J, Rocca Serra A, Roumain M, et al. The gut microbiota metabolite indole alleviates liver inflammation in mice. FASEB J 2018;32(12):6681–93. link1

[52] Gutsmann T, Muller M, Carroll SF, MacKenzie RC, Wiese A, Seydel U. Dual role of lipopolysaccharide (LPS)-binding protein in neutralization of LPS and enhancement of LPS-induced activation of mononuclear cells. Infect Immun 2001;69(11):6942–50. link1

[53] Yan J, Tung HC, Li S, Niu Y, Garbacz WG, Lu P, et al. Aryl hydrocarbon receptor signaling prevents activation of hepatic stellate cells and liver fibrogenesis in mice. Gastroenterology 2019;157(3):793–806.e14. link1

Related Research