The role of bacterial metabolites derived from aromatic amino acids in non-alcoholic fatty liver disease

Cover Page


Cite item

Full Text

Abstract

The review deals with the role of aromatic amino acids and their microbial metabolites in the development and progression of non-alcoholic fatty liver disease (NAFLD). Pathological changes typical for NAFLD, as well as abnormal composition and/or functional activity of gut microbiota, results in abnormal aromatic amino acid metabolism. The authors discuss the potential of these amino acids and their bacterial metabolites to produce both negative and positive impact on the main steps of NAFLD pathophysiology, such as lipogenesis and inflammation, as well as on the liver functions through regulation of the intestinal barrier and microbiota-gut-liver axis signaling. The review gives detailed description of the mechanism of biological activity of tryptophan and its derivatives (indole, tryptamine, indole-lactic, indole-propyonic, indole-acetic acids, and indole-3-aldehyde) through the activation of aryl hydrocarbon receptor (AhR), preventing the development of liver steatosis. Bacteria-produced phenyl-alanine metabolites could promote liver steatosis (phenyl acetic and phenyl lactic acids) or, on the contrary, could reduce liver inflammation and increase insulin sensitivity (phenyl propionic acid). Tyramine, para-cumarate, 4-hydroxyphenylacetic acids, being by-products of bacterial catabolism of tyrosine, can prevent NAFLD, whereas para-cresol and phenol accelerate the progression of NAFLD by damaging the barrier properties of intestinal epithelium. Abnormalities in bacterial catabolism of tyrosine, leading to its excess, stimulate fatty acid synthesis and promote lipid infiltration of the liver. The authors emphasize a close interplay between bacterial metabolism of aromatic amino acids by gut microbiota and the functioning of the human body. They hypothesize that microbial metabolites of aromatic amino acids may represent not only therapeutic targets or non-invasive biomarkers, but also serve as bioactive agents for NAFLD treatment and prevention.

About the authors

E. S. Shcherbakova

State Research Institute of Especially Purified Bioproducts

Author for correspondence.
Email: elenka.shcherbakova@ya.ru
ORCID iD: 0000-0002-4268-8881

Elena S. Shcherbakova – Junior Research Fellow, Laboratory of Microbiology

7 Pudozhskaya ul., Saint Petersburg, 197110

Russian Federation

T. S. Sall

State Research Institute of Especially Purified Bioproducts

Email: miss_taty@mail.ru
ORCID iD: 0000-0002-5890-5641

Tatyana S. Sall – Junior Research Fellow, Laboratory of Microbiology

7 Pudozhskaya ul., Saint Petersburg, 197110

Russian Federation

S. I. Sitkin

State Research Institute of Especially Purified Bioproducts;
North Western State Medical University named after I.I. Mechnikov

Email: drsitkin@gmail.com
ORCID iD: 0000-0003-0331-0963

Stanislav I. Sitkin – MD, PhD, Leading Research Fellow, Laboratory of Microbiology State Research Institute of Especially Purified Bioproducts ; Associate Professor, Chair of Internal Medicine, Gastroenterology and Dietetics n.a. S.M. Ryss North Western State Medical University named after I.I. Mechnikov

7 Pudozhskaya ul., Saint Petersburg, 197110,

47 Piskarevskiy prospekt, Saint Petersburg, 195067

Russian Federation

T. Ya. Vakhitov

State Research Institute of Especially Purified Bioproducts

Email: tim-vakhitov@yandex.ru
ORCID iD: 0000-0001-8221-6910

Timur Ya. Vakhitov – PhD (in Biol.), Chief Research Fellow, Laboratory of Microbiology

7 Pudozhskaya ul., Saint Petersburg, 197110

Russian Federation

E. V. Demyanova

State Research Institute of Especially Purified Bioproducts

Email: lenna_22@mail.ru
ORCID iD: 0000-0002-1872-3464

Elena V. Demyanova – PhD (in Pharmacy), Head of Laboratory of Microbiology

7 Pudozhskaya ul., Saint Petersburg, 197110

Russian Federation

References

  1. Ji Y, Yin Y, Li Z, Zhang W. Gut Microbiota-Derived Components and Metabolites in the Progression of Non-Alcoholic Fatty Liver Disease (NAFLD). Nutrients. 2019;11(8):1712. doi: 10.3390/nu11081712.
  2. Jiang X, Zheng J, Zhang S, Wang B, Wu C, Guo X. Advances in the Involvement of Gut Microbiota in Pathophysiology of NAFLD. Front Med (Lausanne). 2020;7:361. doi: 10.3389/fmed.2020.00361.
  3. Philips CA, Augustine P, Yerol PK, Ramesh GN, Ahamed R, Rajesh S, George T, Kumbar S. Modulating the intestinal microbiota: Therapeutic opportunities in liver disease. J Clin Transl Hepatol. 2020;8(1):87–99. doi: 10.14218/JCTH.2019.00035.
  4. Krishnan S, Ding Y, Saedi N, Choi M, Sridharan GV, Sherr DH, Yarmush ML, Alaniz RC, Jayaraman A, Lee K. Gut Microbiota-Derived Tryptophan Metabolites Modulate Inflammatory Response in Hepatocytes and Macrophages. Cell Rep. 2018;23(4):1099–111. doi: 10.1016/j.celrep.2018.03.109.
  5. Aron-Wisnewsky J, Vigliotti C, Witjes J, Le P, Holleboom AG, Verheij J, Nieuwdorp M, Clément K. Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat Rev Gastroenterol Hepatol. 2020;17(5):279–97. doi: 10.1038/s41575-020-0269-9.
  6. Canfora EE, Meex RCR, Venema K, Blaak EE. Gut microbial metabolites in obesity, NAFLD and T2DM. Nat Rev Endocrinol. 2019;15(5):261–73. doi: 10.1038/s41574-019-0156-z.
  7. Lee G, You HJ, Bajaj JS, Joo SK, Yu J, Park S, Kang H, Park JH, Kim JH, Lee DH, Lee S, Kim W, Ko G. Distinct signatures of gut microbiome and metabolites associated with significant fibrosis in non-obese NAFLD. Nat Commun. 2020;11(1): 4982. doi: 10.1038/s41467-020-18754-5.
  8. Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of non-alcoholic fatty liver disease (NAFLD). Metabolism. 2016;65(8): 1038–48. doi: 10.1016/j.metabol.2015.12.012.
  9. Shao M, Ye Z, Qin Y, Wu T. Abnormal metabolic processes involved in the pathogenesis of non-alcoholic fatty liver disease (Review). Exp Ther Med. 2020;20(5):26. doi: 10.3892/etm.2020.9154.
  10. Liu Y, Hou Y, Wang G, Zheng X, Hao H. Gut microbial metabolites of aromatic amino acids as signals in host-microbe interplay. Trends Endocrinol Metab. 2020;31(11):818–34. doi: 10.1016/j.tem.2020.02.012.
  11. Agus A, Planchais J, Sokol H. Gut microbiota regulation of tryptophan metabolism in health and disease. Cell Host Microbe. 2018;23(6): 716–24. doi: 10.1016/j.chom.2018.05.003.
  12. Roager HM, Licht TR. Microbial tryptophan catabolites in health and disease. Nat Commun. 2018;9(1):3294. doi: 10.1038/s41467-018-05470-4.
  13. Шапошников АМ, Хальчицкий СЕ. Патохимия обмена фенилаланина, тирозина, триптофана и активность фенилаланингидроксилазы печени при вирусных гепатитах. Естественные и технические науки. 2007;(2):138–54.
  14. Delzenne NM, Bindels LB. Microbiome metabolomics reveals new drivers of human liver steatosis. Nat Med. 2018;24(7):906–7. doi: 10.1038/s41591-018-0126-3.
  15. Portune K, Beaumont M, Davila A, Tomé D, Blachier F, Sanz Y. Gut microbiota role in dietary protein metabolism and health-related outcomes: The two sides of the coin. Trends Food Sci Technol. 2016;57:213–32. doi: 10.1016/J.TIFS.2016.08.011.
  16. Oliphant K, Allen-Vercoe E. Macronutrient metabolism by the human gut microbiome: major fermentation by-products and their impact on host health. Microbiome. 2019;7(1): 91. doi: 10.1186/s40168-019-0704-8.
  17. Kalogirou M, Sinakos E. Treating nonalcoholic steatohepatitis with antidiabetic drugs: Will GLP-1 agonists end the struggle? World J Hepatol. 2018;10(11):790–4. doi: 10.4254/wjh.v10.i11.790.
  18. Ma L, Li H, Hu J, Zheng J, Zhou J, Botchlett R, Matthews D, Zeng T, Chen L, Xiao X, Athrey G, Threadgill DW, Li Q, Glaser S, Francis H, Meng F, Li Q, Alpini G, Wu C. Indole Alleviates Diet-Induced Hepatic Steatosis and Inflammation in a Manner Involving Myeloid Cell 6-Phosphofructo-2-Kinase/Fructose-2,6-Biphosphatase 3. Hepatology. 2020;72(4): 1191–203. doi: 10.1002/hep.31115.
  19. Fling RR, Doskey CM, Fader KA, Nault R, Zacharewski TR. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) dysregulates hepatic one carbon metabolism during the progression of steatosis to steatohepatitis with fibrosis in mice. Sci Rep. 2020;10(1):14831. doi: 10.1038/s41598-020-71795-0.
  20. Pan X, Wen SW, Kaminga AC, Liu A. Gut metabolites and inflammation factors in non-alcoholic fatty liver disease: A systematic review and meta-analysis. Sci Rep. 2020;10(1): 8848. doi: 10.1038/s41598-020-65051-8.
  21. Meng D, Sommella E, Salviati E, Campiglia P, Ganguli K, Djebali K, Zhu W, Walker WA. Indole-3-lactic acid, a metabolite of tryptophan, secreted by Bifidobacterium longum subspecies infantis is anti-inflammatory in the immature intestine. Pediatr Res. 2020;88(2): 209–17. doi: 10.1038/s41390-019-0740-x.
  22. Kliewer SA, Goodwin B, Willson TM. The nuclear pregnane X receptor: a key regulator of xenobiotic metabolism. Endocr Rev. 2002;23(5): 687–702. doi: 10.1210/er.2001-0038.
  23. Ларина СН, Чебышев НВ, Ших ЕВ. Модулирование действия ядерных рецепторов и регуляция биотрансформации лекарств. Антибиотики и химиотерапия. 2009;54(5– 6):69–75.
  24. Abildgaard A, Elfving B, Hokland M, Wegener G, Lund S. The microbial metabolite indole-3-propionic acid improves glucose metabolism in rats, but does not affect behaviour. Arch Physiol Biochem. 2018;124(4):306–12. doi: 10.1080/13813455.2017.1398262.
  25. Vacca M, Allison M, Griffin JL, Vidal-Puig A. Fatty acid and glucose sensors in hepatic lipid metabolism: Implications in NAFLD. Semin Liver Dis. 2015;35(3):250–61. doi: 10.1055/s0035-1562945.
  26. Zhao ZH, Xin FZ, Xue Y, Hu Z, Han Y, Ma F, Zhou D, Liu XL, Cui A, Liu Z, Liu Y, Gao J, Pan Q, Li Y, Fan JG. Indole-3-propionic acid inhibits gut dysbiosis and endotoxin leakage to attenuate steatohepatitis in rats. Exp Mol Med. 2019;51(9):1–14. doi: 10.1038/s12276-019-0304-5.
  27. Wlodarska M, Luo C, Kolde R, d'Hennezel E, Annand JW, Heim CE, Krastel P, Schmitt EK, Omar AS, Creasey EA, Garner AL, Mohammadi S, O'Connell DJ, Abubucker S, Arthur TD, Franzosa EA, Huttenhower C, Murphy LO, Haiser HJ, Vlamakis H, Porter JA, Xavier RJ. Indoleacrylic acid produced by commensal peptostreptococcus species suppresses inflammation. Cell Host Microbe. 2017;22(1): 25–37.e6. doi: 10.1016/j.chom.2017.06.007.
  28. Turner MD, Nedjai B, Hurst T, Pennington DJ. Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease. Biochim Biophys Acta. 2014;1843(11):2563–82. doi: 10.1016/j.bbamcr.2014.05.014.
  29. Ritze Y, Bárdos G, Hubert A, Böhle M, Bischoff SC. Effect of tryptophan supplementation on diet-induced non-alcoholic fatty liver disease in mice. Br J Nutr. 2014;112(1):1–7. doi: 10.1017/S0007114514000440.
  30. Вахитов ТЯ, Чалисова НИ, Ситкин СИ, Салль ТС, Шалаева ОН, Демьянова ЕВ, Моругина АС, Виноградова АФ, Петров АВ, Ноздрачев АД. Низкомолекулярные компоненты метаболома крови регулируют пролиферативную активность в клеточных и бактериальных культурах. Доклады Академии наук. 2017;472(4):491–3. doi: 10.7868/S0869565217040284.
  31. Rosario D, Benfeitas R, Bidkhori G, Zhang C, Uhlen M, Shoaie S, Mardinoglu A. Understanding the representative gut microbiota dysbiosis in metformin-treated Type 2 diabetes patients using genome-scale metabolic modeling. Front Physiol. 2018;9:775. doi: 10.3389/fphys.2018.00775.
  32. Liu G, Huang Y, Zhai L. Impact of nutritional and environmental factors on inflammation, oxidative stress, and the microbiome. Biomed Res Int. 2018;2018:5606845. doi: 10.1155/2018/5606845.
  33. Белобородова НВ. Интеграция метаболизма человека и его микробиома при критических состояниях. Общая реаниматология. 2012;8(4):42. doi: 10.15360/1813-9779-2012-4-42.
  34. Черневская ЕА, Белобородова НВ. Микробиота кишечника при критических состояниях (обзор). Общая реаниматология. 2018;14(5):96–119. doi: 10.15360/1813-9779-2018-5-96-119.
  35. Федотчева НИ, Теплова ВВ, Белобородова НВ. Влияние микробных метаболитов на функции митохондрий в условиях ацидоза и дефицита субстратов окисления. Биологические мембраны. 2019;36(1):44–52. doi: 10.1134/S0233475518060038.
  36. Оковитый СВ, Радько СВ. Митохондриальная дисфункция в патогенезе различных поражений печени. Доктор.Ру. Гастроэнтерология. 2015;(12):30–3.
  37. Léveillé M, Estall JL. Mitochondrial dysfunction in the transition from NASH to HCC. Metabolites. 2019;9(10):233. doi: 10.3390/metabo9100233.
  38. Rowland I, Gibson G, Heinken A, Scott K, Swann J, Thiele I, Tuohy K. Gut microbiota functions: metabolism of nutrients and other food components. Eur J Nutr. 2018;57(1): 1–24. doi: 10.1007/s00394-017-1445-8.
  39. Delzenne NM, Knudsen C, Beaumont M, Rodriguez J, Neyrinck AM, Bindels LB. Contribution of the gut microbiota to the regulation of host metabolism and energy balance: a focus on the gut-liver axis. Proc Nutr Soc. 2019;78(3): 319–28. doi: 10.1017/S0029665118002756.
  40. Sharpton SR, Ajmera V, Loomba R. Emerging Role of the Gut Microbiome in Nonalcoholic Fatty Liver Disease: From Composition to Function. Clin Gastroenterol Hepatol. 2019;17(2):296–306. doi: 10.1016/j.cgh.2018.08.065.
  41. Xie C, Halegoua-DeMarzio D. Role of probiotics in non-alcoholic fatty liver disease: does gut microbiota matter? Nutrients. 2019;11(11):2837. doi: 10.3390/nu11112837.
  42. Hoyles L, Fernández-Real JM, Federici M, Serino M, Abbott J, Charpentier J, Heymes C, Luque JL, Anthony E, Barton RH, Chilloux J, Myridakis A, Martinez-Gili L, Moreno-Navarrete JM, Benhamed F, Azalbert V, Blasco-Baque V, Puig J, Xifra G, Ricart W, Tomlinson C, Woodbridge M, Cardellini M, Davato F, Cardolini I, Porzio O, Gentileschi P, Lopez F, Foufelle F, Butcher SA, Holmes E, Nicholson JK, Postic C, Burcelin R, Dumas ME. Molecular phenomics and metagenomics of hepatic steatosis in non-diabetic obese women. Nat Med. 2018;24(7):1070–80. doi: 10.1038/s41591-018-0061-3.
  43. Wikoff WR, Anfora AT, Liu J, Schultz PG, Lesley SA, Peters EC, Siuzdak G. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A. 2009;106(10):3698–703. doi: 10.1073/pnas.0812874106.
  44. Cho S, Yang X, Won K, Leone V, Hubert N, Chang E, Chung E, Park J, Guzman G, Lee H, Jeong H. Phenylpropionc acid produced by gut microbiota alleviates acetaminophen-induced hepatotoxicity. bioRxiv. 2020; 811984. doi: 10.1101/811984.
  45. Sparks SM, Aquino C, Banker P, Collins JL, Cowan D, Diaz C, Dock ST, Hertzog DL, Liang X, Swiger ED, Yuen J, Chen G, Jayawickreme C, Moncol D, Nystrom C, Rash V, Rimele T, Roller S, Ross S. Exploration of phenylpropanoic acids as agonists of the free fatty acid receptor 4 (FFA4): Identification of an orally efficacious FFA4 agonist. Bioorg Med Chem Lett. 2017;27(5):1278–83. doi: 10.1016/j.bmcl.2017.01.034.
  46. Moniri NH. Free-fatty acid receptor-4 (GPR120): Cellular and molecular function and its role in metabolic disorders. Biochem Pharmacol. 2016;110–1:1–15. doi: 10.1016/j.bcp.2016.01.021.
  47. Melhem H, Kaya B, Ayata CK, Hruz P, Niess JH. Metabolite-Sensing G Protein-Coupled Receptors Connect the Diet-Microbiota-Metabolites Axis to Inflammatory Bowel Disease. Cells. 2019;8(5):450. doi: 10.3390/cells8050450.
  48. Colosimo DA, Kohn JA, Luo PM, Piscotta FJ, Han SM, Pickard AJ, Rao A, Cross JR, Cohen LJ, Brady SF. Mapping Interactions of microbial metabolites with human G-protein-coupled receptors. Cell Host Microbe. 2019;26(2):273– 82.e7. doi: 10.1016/j.chom.2019.07.002.
  49. Caussy C, Hsu C, Lo MT, Liu A, Bettencourt R, Ajmera VH, Bassirian S, Hooker J, Sy E, Richards L, Schork N, Schnabl B, Brenner DA, Sirlin CB, Chen CH, Loomba R; Genetics of NAFLD in Twins Consortium. Link between gut-microbiome derived metabolite and shared gene-effects with hepatic steatosis and fibrosis in NAFLD. Hepatology. 2018;68(3):918–32. doi: 10.1002/hep.29892.
  50. Maini Rekdal V, Bess EN, Bisanz JE, Turnbaugh PJ, Balskus EP. Discovery and inhibition of an interspecies gut bacterial pathway for Levodopa metabolism. Science. 2019;364(6445):eaau6323. doi: 10.1126/science.aau6323.
  51. Jin R, Banton S, Tran VT, Konomi JV, Li S, Jones DP, Vos MB. Amino acid metabolism is altered in adolescents with nonalcoholic fatty liver disease – An untargeted, high resolution metabolomics study. J Pediatr. 2016;172:14– 9.e5. doi: 10.1016/j.jpeds.2016.01.026.
  52. Patel A, Thompson A, Abdelmalek L, Adams-Huet B, Jialal I. The relationship between tyramine levels and inflammation in metabolic syndrome. Horm Mol Biol Clin Investig. 2019;40(1):/j/hmbci.2019.40.issue-1/hmbci-2019-0047/hmbci-2019-0047.xml. doi: 10.1515/hmbci-2019-0047.
  53. Ekinci Akdemir FN, Albayrak M, Çalik M, Bayir Y, Gülçin İ. The Protective Effects of p-coumaric acid on acute liver and kidney damages induced by cisplatin. Biomedicines. 2017;5(2): 18. doi: 10.3390/biomedicines5020018.
  54. Lima LC, Buss GD, Ishii-Iwamoto EL, Salgueiro-Pagadigorria C, Comar JF, Bracht A, Constantin J. Metabolic effects of p-coumaric acid in the perfused rat liver. J Biochem Mol Toxicol. 2006;20(1):18–26. doi: 10.1002/jbt.20114.
  55. Vily-Petit J, Soty-Roca M, Silva M, Raffin M, Gautier-Stein A, Rajas F, Mithieux G. Intestinal gluconeogenesis prevents obesity-linked liver steatosis and non-alcoholic fatty liver disease. Gut. 2020;69(12):2193–202. doi: 10.1136/gutjnl-2019-319745.
  56. Takahama U, Ansai T, Hirota S. Nitrogen oxides toxicology of the aerodigestive tract. In: Advances in Molecular Toxicology. Chapter IV. 2013;7:129–77.
  57. Brial F, Alzaid F, Sonomura K, Kamatani Y, Meneyrol K, Le Lay A, Péan N, Hedjazi L, Sato TA, Venteclef N, Magnan C, Lathrop M, Dumas ME, Matsuda F, Zalloua P, Gauguier D. The natural metabolite 4-cresol improves glucose homeostasis and enhances β-cell function. Cell Rep. 2020;30(7):2306–20.e5. doi: 10.1016/j.celrep.2020.01.066.
  58. Rebholz CM, Yu B, Zheng Z, Chang P, Tin A, Köttgen A, Wagenknecht LE, Coresh J, Boerwinkle E, Selvin E. Serum metabolomic profile of incident diabetes. Diabetologia. 2018;61(5):1046–54. doi: 10.1007/s00125-018-4573-7.
  59. Chernevskaya E, Beloborodova N, Klimenko N, Pautova A, Shilkin D, Gusarov V, Tyakht A. Serum and fecal profiles of aromatic microbial metabolites reflect gut microbiota disruption in critically ill patients: a prospective observational pilot study. Crit Care. 2020;24(1):312. doi: 10.1186/s13054-020-03031-0.
  60. Zhao H, Jiang Z, Chang X, Xue H, Yahefu W, Zhang X. 4-Hydroxyphenylacetic Acid Prevents Acute APAP-Induced Liver Injury by Increasing Phase II and Antioxidant Enzymes in Mice. Front Pharmacol. 2018;9:653. doi: 10.3389/fphar.2018.00653.
  61. Leung TM, Nieto N. CYP2E1 and oxidant stress in alcoholic and non-alcoholic fatty liver disease. J Hepatol. 2013;58(2):395–8. doi: 10.1016/j.jhep.2012.08.018.
  62. Ситкин СИ, Вахитов ТЯ, Ткаченко ЕИ, Лазебник ЛБ, Орешко ЛС, Жигалова ТН, Радченко ВГ, Авалуева ЕБ, Селиверстов ПВ, Утсаль ВА, Комличенко ЭВ. Нарушения микробного и эндогенного метаболизма при язвенном колите и целиакии: метаболомный подход к выявлению потенциальных биомаркеров хронического воспаления в кишечнике, связанного с дисбиозом. Экспериментальная и клиническая гастроэнтерология. 2017;(7):4–50.
  63. Selmer T, Andrei PI. p-Hydroxyphenylacetate decarboxylase from Clostridium difficile. A novel glycyl radical enzyme catalysing the formation of p-cresol. Eur J Biochem. 2001;268(5):1363–72. doi: 10.1046/j.1432-1327.2001.02001.x.
  64. Белобородова НВ, Мороз ВВ, Осипов АА, Бедова АЮ, Оленин АЮ, Гецина МЛ, Карпова ОВ, Оленина ЕГ. Нормальный уровень сепсис-ассоциированных фенилкарбоновых кислот в сыворотке крови человека. Биохимия. 2015;80(3):449–55. doi: 10.1134/S0006297915030128.
  65. Zheng D, Liwinski T, Elinav E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020;30(6):492–506. doi: 10.1038/s41422-020-0332-7.
  66. Ghazarian M, Revelo XS, Nøhr MK, Luck H, Zeng K, Lei H, Tsai S, Schroer SA, Park YJ, Chng MHY, Shen L, D'Angelo JA, Horton P, Chapman WC, Brockmeier D, Woo M, Engleman EG, Adeyi O, Hirano N, Jin T, Gehring AJ, Winer S, Winer DA. Type I interferon responses drive intrahepatic T cells to promote metabolic syndrome. Sci Immunol. 2017;2(10):eaai7616. doi: 10.1126/sciimmunol.aai7616.

Supplementary files

Supplementary Files
Action
1. JATS XML

Copyright (c) 2020 Shcherbakova E.S., Sall T.S., Sitkin S.I., Vakhitov T.Y., Demyanova E.V.

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies