We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×
Skip main navigation
Aging Health
Bioelectronics in Medicine
Biomarkers in Medicine
Breast Cancer Management
CNS Oncology
Colorectal Cancer
Concussion
Epigenomics
Future Cardiology
Future Medicine AI
Future Microbiology
Future Neurology
Future Oncology
Future Rare Diseases
Future Virology
Hepatic Oncology
HIV Therapy
Immunotherapy
International Journal of Endocrine Oncology
International Journal of Hematologic Oncology
Journal of 3D Printing in Medicine
Lung Cancer Management
Melanoma Management
Nanomedicine
Neurodegenerative Disease Management
Pain Management
Pediatric Health
Personalized Medicine
Pharmacogenomics
Regenerative Medicine

Biomarkers of human gut microbiota diversity and dysbiosis

    Alina M Rüb

    Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany

    ,
    Anastasia Tsakmaklis

    Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany

    ,
    Stefanie K Gräfe

    Department I of Internal Medicine, University Hospital Cologne, Cologne, Germany

    ,
    Marie-Christine Simon

    Department of Nutrition & Food Sciences, Nutrition & Microbiota, University of Bonn, Bonn, Germany

    ,
    Maria JGT Vehreschild

    *Author for correspondence:

    E-mail Address: Maria.Vehreschild@kgu.de

    Department of Internal Medicine, Infectious Diseases, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main, Germany

    &
    Irene Wuethrich

    Department of Biosystems Science & Engineering, ETH Zurich, Basel, Switzerland

    Published Online:https://doi.org/10.2217/bmm-2020-0353

    The association of gut microbiota dysbiosis with various human diseases is being substantiated with increasing evidence. Metabolites derived from both, microbiota and the human host play a central role in disease susceptibility and disease progression by extensively modulating host physiology and metabolism. Several of these metabolites have the potential to serve as diagnostic biomarkers for monitoring disease states in conjunction with intestinal microbiota dysbiosis. In this narrative review we evaluate the potential of trimethylamine-N-oxide, short-chain fatty acids, 3-indoxyl sulfate, p-cresyl sulfate, secondary bile acids, hippurate, human β-defensin-2, chromogranin A, secreted immunoglobulins and zonulin to serve as biomarkers for metabolite profiling and diagnostic suitability for dysbiosis and disease.

    Papers of special note have been highlighted as: • of interest; •• of considerable interest

    References

    • 1. Petersen C, Round JL. Defining dysbiosis and its influence on host immunity and disease. Cell. Microbiol. 16(7), 1024–1033 (2014). •• This review article is a wonderful introduction into the topic of dysbiosis of the microbial ecosystem, how it is triggered by external influences and which effects dysbiosis has on hosts health and immune regulation.
    • 2. Branchereau M, Burcelin R, Heymes C. The gut microbiome and heart failure: a better gut for a better heart. Rev. Endocr. Metab. Disord. 20(4), 407–414 (2019).
    • 3. Brown EG, Tanner CM, Goldman SM. The microbiome in neurodegenerative disease. Curr. Geriatr. Rep. 7(2), 81–91 (2018).
    • 4. Chen J, Chia N, Kalari KR et al. Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls. Sci. Rep. 6(1), 1–10 (2016).
    • 5. Round JL, Mazmanian SK. The gut microbiota shapes intestinal immune responses during health and disease. Nat. Rev. Immunol. 9(5), 313–323 (2009).
    • 6. Tilg H, Kaser A. Gut microbiome, obesity, and metabolic dysfunction. J. Clin. Invest. 121(6), 2126–2132 (2011).
    • 7. Clarke SF, Murphy EF, O'sullivan O et al. Exercise and associated dietary extremes impact on gut microbial diversity. Gut 63(12), 1913–1920 (2014).
    • 8. David LA, Maurice CF, Carmody RN et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 505(7484), 559–563 (2014).
    • 9. Koeth RA, Wang Z, Levison BS et al. Intestinal microbiota metabolism of L-carnitine, a nutrient in red meat, promotes atherosclerosis. Nat. Med. 19(5), 576–585 (2013).
    • 10. Wang Z, Klipfell E, Bennett BJ et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 472(7341), 57–63 (2011).
    • 11. Wang Z, Roberts AB, Buffa JA et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell 163(7), 1585–1595 (2015).
    • 12. Al-Obaide MA, Singh R, Datta P et al. Gut microbiota-dependent trimethylamine-N-oxide and serum biomarkers in patients with T2DM and advanced CKD. J. Clin. Med. 6(9), 86 (2017).
    • 13. Cho CE, Taesuwan S, Malysheva OV et al. Trimethylamine-N-oxide (TMAO) response to animal source foods varies among healthy young men and is influenced by their gut microbiota composition: a randomized controlled trial. Mol. Nutr. Food Res. 61(1), 1600324 (2017).
    • 14. Janeiro MH, Ramírez MJ, Milagro FI, Martínez JA, Solas M. Implication of trimethylamine N-oxide (TMAO) in disease: potential biomarker or new therapeutic target. Nutrients 10(10), 1398 (2018). • Presents the endogenous synthesis of trimethylamine-N-oxide within microbial metabolism. It explains the potential effects of trimethylamine-N-oxide on immune functions and the etiology of cardiovascular as well as neurological disorders and is therefore an excellent introduction to the topic.
    • 15. Velasquez MT, Ramezani A, Manal A, Raj DS. Trimethylamine N-oxide: the good, the bad and the unknown. Toxins 8(11), 326 (2016).
    • 16. Sanchez-Alcoholado L, Castellano-Castillo D, Jordán-Martínez L et al. Role of gut microbiota on cardio-metabolic parameters and immunity in coronary artery disease patients with and without type-2 diabetes mellitus. Front. Microbiol. 8, 1936 (2017).
    • 17. Xu K-Y, Xia G-H, Lu J-Q et al. Impaired renal function and dysbiosis of gut microbiota contribute to increased trimethylamine-N-oxide in chronic kidney disease patients. Sci. Rep. 7(1), 1–12 (2017).
    • 18. Yin J, Liao SX, He Y et al. Dysbiosis of gut microbiota with reduced trimethylamine-N-oxide level in patients with large-artery atherosclerotic stroke or transient ischemic attack. J. Am. Heart Assoc. 4(11), e002699 (2015).
    • 19. Falony G, Vieira-Silva S, Raes J. Microbiology meets big data: the case of gut microbiota–derived trimethylamine. Annu. Rev. Microbiol. 69, 305–321 (2015).
    • 20. Ríos-Covián D, Ruas-Madiedo P, Margolles A, Gueimonde M, De Los Reyes-Gavilán CG, Salazar N. Intestinal short chain fatty acids and their link with diet and human health. Front. Microbiol. 7, 185 (2016).
    • 21. Zeng H, Lazarova DL, Bordonaro M. Mechanisms linking dietary fiber, gut microbiota and colon cancer prevention. World J. Gastrointest. Oncol. 6(2), 41 (2014).
    • 22. Sorbara MT, Dubin K, Littmann ER et al. Inhibiting antibiotic-resistant Enterobacteriaceae by microbiota-mediated intracellular acidification. J. Exp. Med. 216(1), 84–98 (2019). •• This article is particularly relevant in the context of understanding colonization resistance and the process of colonization with multiresistant pathogens. It reveals that commensal microbiota-derived metabolites, such as short-chain fatty acids, are involved in immunity against pathogenic organisms.
    • 23. La Cuesta-Zuluaga D, Mueller NT, Álvarez-Quintero R et al. Higher fecal short-chain fatty acid levels are associated with gut microbiome dysbiosis, obesity, hypertension and cardiometabolic disease risk factors. Nutrients 11(1), 51 (2019).
    • 24. Vogt JA, Wolever TM. Fecal acetate is inversely related to acetate absorption from the human rectum and distal colon. J. Nutr. 133(10), 3145–3148 (2003).
    • 25. Topping DL, Clifton PM. Short-chain fatty acids and human colonic function: roles of resistant starch and nonstarch polysaccharides. Physiol. Rev. 81, 1031–1064 (2001). •• Topping and Clifton provide here a very detailed and interesting overview of the importance of short-chain fatty acids for GI tract functions and its effects on physiological functions. This article is particularly suitable for nutritionists but also for other scientists interested in the metabolic functions of the intestinal microbiome.
    • 26. Sun Q, Jia Q, Song L, Duan L. Alterations in fecal short-chain fatty acids in patients with irritable bowel syndrome: a systematic review and meta-analysis. Medicine 98(7), e14513 (2019).
    • 27. Devaraj S, Hemarajata P, Versalovic J. The human gut microbiome and body metabolism: implications for obesity and diabetes. Clin. Chem. 59(4), 617–628 (2013).
    • 28. Hinnebusch BF, Meng S, Wu JT, Archer SY, Hodin RA. The effects of short-chain fatty acids on human colon cancer cell phenotype are associated with histone hyperacetylation. J. Nutr. 132(5), 1012–1017 (2002).
    • 29. Kelly CJ, Zheng L, Campbell EL et al. Crosstalk between microbiota-derived short-chain fatty acids and intestinal epithelial HIF augments tissue barrier function. Cell Host Microbe 17(5), 662–671 (2015).
    • 30. Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nat. Rev. Microbiol. 12(10), 661–672 (2014).
    • 31. Velazquez OC, Lederer HM, Rombeau JL. Butyrate and the colonocyte. In: Dietary Fiber in Health and Disease. Springer, MA, USA, 123–134 (1997).
    • 32. Yan J, Herzog JW, Tsang K et al. Gut microbiota induce IGF-1 and promote bone formation and growth. Proc. Natl Acad. Sci. USA 113(47), E7554–E7563 (2016).
    • 33. Wang T, Cai G, Qiu Y et al. Structural segregation of gut microbiota between colorectal cancer patients and healthy volunteers. ISME J. 6(2), 320–329 (2012).
    • 34. Weir TL, Manter DK, Sheflin AM, Barnett BA, Heuberger AL, Ryan EP. Stool microbiome and metabolome differences between colorectal cancer patients and healthy adults. PLoS ONE 8(8), e70803 (2013).
    • 35. Sze MA, Topçuoğlu BD, Lesniak NA, Ruffin MT, Schloss PD. Fecal short-chain fatty acids are not predictive of colonic tumor status and cannot be predicted based on bacterial community structure. mBio 10(4), e01454–e01419 (2019).
    • 36. Vrieze A, Van Nood E, Holleman F et al. Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome. Gastroenterology 143(4), 913–916.e917 (2012).
    • 37. Müller M, Hernández MaG, Goossens GH et al. Circulating but not faecal short-chain fatty acids are related to insulin sensitivity, lipolysis and GLP-1 concentrations in humans. Sci. Rep. 9(1), 1–9 (2019).
    • 38. Rowland I, Gibson G, Heinken A et al. Gut microbiota functions: metabolism of nutrients and other food components. Eur. J. Nutr. 57(1), 1–24 (2018).
    • 39. Banoglu E, King R. Sulfation of indoxyl by human and rat aryl (phenol) sulfotransferases to form indoxyl sulfate. Eur. J. Drug Metab. Pharmacokinet. 27(2), 135–140 (2002).
    • 40. Weber D, Oefner PJ, Hiergeist A et al. Low urinary indoxyl sulfate levels early after transplantation reflect a disrupted microbiome and are associated with poor outcome. Blood 126(14), 1723–1728 (2015).
    • 41. Gryp T, Vanholder R, Vaneechoutte M, Glorieux G. p-Cresyl sulfate. Toxins 9(2), 52 (2017).
    • 42. Lin C-J, Wu V, Wu P-C, Wu C-J. Meta-analysis of the associations of p-cresyl sulfate (PCS) and indoxyl sulfate (IS) with cardiovascular events and all-cause mortality in patients with chronic renal failure. PLoS ONE 10(7), e0132589 (2015).
    • 43. Vanholder R, Schepers E, Pletinck A, Nagler EV, Glorieux G. The uremic toxicity of indoxyl sulfate and p-cresyl sulfate: a systematic review. J. Am. Soc. Nephrol. 25(9), 1897–1907 (2014).
    • 44. Wong J, Piceno YM, Desantis TZ, Pahl M, Andersen GL, Vaziri ND. Expansion of urease-and uricase-containing, indole-and p-cresol-forming and contraction of short-chain fatty acid-producing intestinal microbiota in ESRD. Am. J. Nephrol. 39(3), 230–237 (2014).
    • 45. Monte MJ, Marin JJ, Antelo A, Vazquez-Tato J. Bile acids: chemistry, physiology, and pathophysiology. World J. Gastroenterol. 15(7), 804 (2009).
    • 46. Staels B, Fonseca VA. Bile acids and metabolic regulation: mechanisms and clinical responses to bile acid sequestration. Diabetes Care 32(Suppl. 2), S237–S245 (2009).
    • 47. Ridlon JM, Harris SC, Bhowmik S, Kang D-J, Hylemon PB. Consequences of bile salt biotransformations by intestinal bacteria. Gut Microbes 7(1), 22–39 (2016).
    • 48. Wang C, Zhu C, Shao L, Ye J, Shen Y, Ren Y. Role of bile acids in dysbiosis and treatment of nonalcoholic fatty liver disease. Mediators Inflammation 2019, 13 (2019).
    • 49. Chávez-Talavera O, Tailleux A, Lefebvre P, Staels B. Bile acid control of metabolism and inflammation in obesity, Type 2 diabetes, dyslipidemia, and nonalcoholic fatty liver disease. Gastroenterology 152(7), 1679–1694. e1673 (2017).
    • 50. Gottlieb A, Canbay A. Why bile acids are so important in non-alcoholic fatty liver disease (NAFLD) progression. Cells 8(11), 1358 (2019).
    • 51. Lajczak NK, Saint-Criq V, O'dwyer AM et al. Bile acids deoxycholic acid and ursodeoxycholic acid differentially regulate human β-defensin-1 and-2 secretion by colonic epithelial cells. FASEB J. 31(9), 3848–3857 (2017).
    • 52. Corbin KD, Zeisel SH. Choline metabolism provides novel insights into non-alcoholic fatty liver disease and its progression. Curr. Opin. Gastroenterol. 28(2), 159 (2012).
    • 53. Begley M, Gahan CG, Hill C. The interaction between bacteria and bile. FEMS Microbiol. Rev. 29(4), 625–651 (2005).
    • 54. Antharam VC, Li EC, Ishmael A et al. Intestinal dysbiosis and depletion of butyrogenic bacteria in Clostridium difficile infection and nosocomial diarrhea. J. Clin. Microbiol. 51(9), 2884–2892 (2013).
    • 55. Buffie CG, Bucci V, Stein RR et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature 517(7533), 205–208 (2015). • Using mathematical modeling and various statistical analyses, it is shown here that secondary bile acids could be a potential approach to Clostridioides difficile infection recurrence prophylaxis and which microorganisms are important in this context.
    • 56. Khanna S, Pardi DS, Kelly CR et al. A novel microbiome therapeutic increases gut microbial diversity and prevents recurrent Clostridium difficile infection. J. Infect. Dis. 214(2), 173–181 (2016).
    • 57. Theriot CM, Bowman AA, Young VB. Antibiotic-induced alterations of the gut microbiota alter secondary bile acid production and allow for Clostridium difficile spore germination and outgrowth in the large intestine. MSphere 1(1), e00045–e00115 (2016).
    • 58. Pallister T, Jackson MA, Martin TC et al. Hippurate as a metabolomic marker of gut microbiome diversity: modulation by diet and relationship to metabolic syndrome. Sci. Rep. 7(1), 1–9 (2017). •• Provides an extensive profiling of various metabolic markers and identifies hippurate as marker of microbial diversity and possible marker of metabolic disorders. Many important findings of this work are summarized in our review article.
    • 59. Gatley SJ, Sherratt HSA. The localization of hippurate synthesis in the matrix of rat liver mitochondria. Biochem Soc Trans 4(3), 525–526 (1976).
    • 60. Dall'asta M, Calani L, Tedeschi M, Jechiu L, Brighenti F, Del Rio D. Identification of microbial metabolites derived from in vitro fecal fermentation of different polyphenolic food sources. Nutrition 28(2), 197–203 (2012).
    • 61. Lees HJ, Swann JR, Wilson ID, Nicholson JK, Holmes E. Hippurate: the natural history of a mammalian–microbial cometabolite. J. Proteome Res. 12(4), 1527–1546 (2013).
    • 62. Edberg SC, Samuels S. Rapid, colorimetric test for the determination of hippurate hydrolysis by group B Streptococcus. J. Clin. Microbiol. 3(1), 49–50 (1976).
    • 63. Ziegler P, Kutzner H. Hippurate hydrolysis as a taxonomic criterion in the genus Streptomyces (order Actinomycetales). Zeitschrift für allgemeine Mikrobiologie 13(3), 265–272 (1973).
    • 64. Swann JR, Tuohy KM, Lindfors P et al. Variation in antibiotic-induced microbial recolonization impacts on the host metabolic phenotypes of rats. J. Proteome Res. 10(8), 3590–3603 (2011).
    • 65. Yap IK, Li JV, Saric J et al. Metabonomic and microbiological analysis of the dynamic effect of vancomycin-induced gut microbiota modification in the mouse. J. Proteome Res. 7(9), 3718–3728 (2008).
    • 66. Joossens M, Huys G, Cnockaert M et al. Dysbiosis of the faecal microbiota in patients with Crohn’s disease and their unaffected relatives. Gut 60(5), 631–637 (2011).
    • 67. Williams HR, Cox IJ, Walker DG et al. Differences in gut microbial metabolism are responsible for reduced hippurate synthesis in Crohn’s disease. BMC Gastroenterol. 10(1), 108 (2010).
    • 68. Schröder J-M, Harder J. Human beta-defensin-2. Int. J. Biochem. Cell Biol. 31(6), 645–651 (1999).
    • 69. Niyonsaba F, Ogawa H, Nagaoka I. Human β-defensin-2 functions as a chemotactic agent for tumour necrosis factor-α-treated human neutrophils. Immunology 111(3), 273–281 (2004).
    • 70. Takahashi A, Wada A, Ogushi K-I et al. Production of β-defensin-2 by human colonic epithelial cells induced by Salmonella enteritidis flagella filament structural protein. FEBS Lett. 508(3), 484–488 (2001).
    • 71. Voss E, Wehkamp J, Wehkamp K, Stange EF, Schröder JM, Harder J. NOD2/CARD15 mediates induction of the antimicrobial peptide human beta-defensin-2. J. Biol. Chem. 281(4), 2005–2011 (2006).
    • 72. Schlee M, Harder J, Köten B, Stange E, Wehkamp J, Fellermann K. Probiotic lactobacilli and VSL# 3 induce enterocyte β-defensin 2. Clin. Exp. Immunol. 151(3), 528–535 (2008).
    • 73. Wehkamp J, Schmidt K, Herrlinger K et al. Defensin pattern in chronic gastritis: HBD-2 is differentially expressed with respect to Helicobacter pylori status. J. Clin. Pathol. 56(5), 352–357 (2003).
    • 74. Wehkamp J, Harder J, Wehkamp K et al. NF-κB-and AP-1-mediated induction of human beta defensin-2 in intestinal epithelial cells by Escherichia coli Nissle 1917: a novel effect of a probiotic bacterium. Infect. Immun. 72(10), 5750–5758 (2004).
    • 75. Möndel M, Schroeder B, Zimmermann K et al. Probiotic E. coli treatment mediates antimicrobial human β-defensin synthesis and fecal excretion in humans. Mucosal Immunol. 2(2), 166–172 (2009).
    • 76. Kapel N, Benahmed N, Morali A et al. Fecal β-defensin-2 in children with inflammatory bowel diseases. J. Pediatr. Gastroenterol. Nutr. 48(1), 117–120 (2009).
    • 77. Coretti L, Natale A, Cuomo M et al. The interplay between defensins and microbiota in Crohn’s disease. Mediators Inflammation 2017, 8 (2017).
    • 78. Gutierrez A, Holler E, Zapater P et al. Antimicrobial peptide response to blood translocation of bacterial DNA in Crohn’s disease is affected by NOD2/CARD15 genotype. Inflamm. Bowel Dis. 17(8), 1641–1650 (2011).
    • 79. D'amico MA, Ghinassi B, Izzicupo P, Manzoli L, Di Baldassarre A. Biological function and clinical relevance of chromogranin A and derived peptides. Endocr. Connect. 3(2), R45–R54 (2014).
    • 80. Sundin J, Stridsberg M, Tap J et al. Fecal chromogranins and secretogranins are linked to the fecal and mucosal intestinal bacterial composition of IBS patients and healthy subjects. Sci. Rep. 8(1), 1–13 (2018).
    • 81. Shooshtarizadeh P, Zhang D, Chich J-F et al. The antimicrobial peptides derived from chromogranin/secretogranin family, new actors of innate immunity. Regul. Pept. 165(1), 102–110 (2010).
    • 82. Conlon JM, Demandt A, Nielsen PF, Leprince J, Vaudry H, Woodhams DC. The alyteserins: two families of antimicrobial peptides from the skin secretions of the midwife toad Alytes obstetricans (Alytidae). Peptides 30(6), 1069–1073 (2009).
    • 83. O'toole D, Grossman A, Gross D et al. ENETS consensus guidelines for the standards of care in neuroendocrine tumors: biochemical markers. Neuroendocrinology 90(2), 194–202 (2009).
    • 84. Bartolomucci A, Pasinetti G, Salton S. Granins as disease-biomarkers: translational potential for psychiatric and neurological disorders. Neuroscience 170(1), 289–297 (2010).
    • 85. Ottesen AH, Christensen G, Omland T, Røsjø H. Glycosylated chromogranin A: potential role in the pathogenesis of heart failure. Curr. Heart Fail. Rep. 14(6), 478–488 (2017).
    • 86. Zhernakova A, Kurilshikov A, Bonder MJ et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 352(6285), 565–569 (2016).
    • 87. Ghavami SB, Rostami E, Sephay AA et al. Alterations of the human gut Methanobrevibacter smithii as a biomarker for inflammatory bowel diseases. Microb. Pathog. 117, 285–289 (2018).
    • 88. Cerutti A. The regulation of IgA class switching. Nature Rev. Immunol. 8(6), 421–434 (2008).
    • 89. Mantis NJ, Rol N, Corthésy B. Secretory IgA’s complex roles in immunity and mucosal homeostasis in the gut. Mucosal Immunol. 4(6), 603–611 (2011).
    • 90. Peterson DA, Mcnulty NP, Guruge JL, Gordon JI. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe 2(5), 328–339 (2007).
    • 91. Maes M, Mihaylova I, Leunis J-C. Increased serum IgA and IgM against LPS of enterobacteria in chronic fatigue syndrome (CFS): indication for the involvement of gram-negative enterobacteria in the etiology of CFS and for the presence of an increased gut–intestinal permeability. J. Affect. Disord. 99(1–3), 237–240 (2007).
    • 92. Bischoff SC, Barbara G, Buurman W et al. Intestinal permeability–a new target for disease prevention and therapy. BMC Gastroenterol. 14(1), 189 (2014).
    • 93. Chang J, Leong RW, Wasinger VC, Ip M, Yang M, Phan TG. Impaired intestinal permeability contributes to ongoing bowel symptoms in patients with inflammatory bowel disease and mucosal healing. Gastroenterology 153(3), 723–731. e721 (2017).
    • 94. Maes M, Kubera M, Leunis J-C, Berk M. Increased IgA and IgM responses against gut commensals in chronic depression: further evidence for increased bacterial translocation or leaky gut. J. Affect. Disord. 141(1), 55–62 (2012).
    • 95. Sjögren YM, Tomicic S, Lundberg A et al. Influence of early gut microbiota on the maturation of childhood mucosal and systemic immune responses: gut microbiota and immune responses. Clin. Exp. Allergy 39(12), 1842–1851 (2009).
    • 96. Spencer SP, Fragiadakis GK, Sonnenburg JL. Pursuing human-relevant gut microbiota-immune interactions. Immunity 51(2), 225–239 (2019).
    • 97. Fasano A. Zonulin, regulation of tight junctions, and autoimmune diseases. Ann. NY Acad. Sci. 1258(1), 25 (2012).
    • 98. El Asmar R, Panigrahi P, Bamford P et al. Host-dependent zonulin secretion causes the impairment of the small intestine barrier function after bacterial exposure. Gastroenterology 123(5), 1607–1615 (2002).
    • 99. Vanuytsel T, Vermeire S, Cleynen I. The role of haptoglobin and its related protein, zonulin, in inflammatory bowel disease. Tissue Barriers 1(5), e27321 (2013).
    • 100. Sturgeon C, Fasano A. Zonulin, a regulator of epithelial and endothelial barrier functions, and its involvement in chronic inflammatory diseases. Tissue Barriers 4(4), e1251384 (2016).
    • 101. Klaus DA, Motal MC, Burger-Klepp U et al. Increased plasma zonulin in patients with sepsis. Biochem. Med. 23(1), 107–111 (2013).
    • 102. Li X, Atkinson MA. The role for gut permeability in the pathogenesis of Type 1 diabetes–a solid or leaky concept? Pediatr. Diabetes 16(7), 485–492 (2015).
    • 103. Boulangé CL, Neves AL, Chilloux J, Nicholson JK, Dumas M-E. Impact of the gut microbiota on inflammation, obesity, and metabolic disease. Genome Med. 8(1), 1–12 (2016).
    • 104. Durazzo M, Ferro A, Gruden G. Gastrointestinal microbiota and Type 1 diabetes mellitus: the state of art. J. Clin. Med. 8(11), 1843 (2019).
    • 105. Frati F, Salvatori C, Incorvaia C et al. The role of the microbiome in asthma: the gut–lung axis. Int. J. Mol. Sci. 20(1), 123 (2019).
    • 106. Iacob S, Iacob DG. Infectious threats, the intestinal barrier, and its Trojan horse: dysbiosis. Front.Microbiol. 10, 10 (2019).
    • 107. Mörkl S, Lackner S, Meinitzer A et al. Gut microbiota, dietary intakes and intestinal permeability reflected by serum zonulin in women. Eur. J. Nutr. 57(8), 2985–2997 (2018).
    • 108. Liu Z-H, Huang M-J, Zhang X-W et al. The effects of perioperative probiotic treatment on serum zonulin concentration and subsequent postoperative infectious complications after colorectal cancer surgery: a double-center and double-blind randomized clinical trial. Am. J. Clin. Nutr. 97(1), 117–126 (2013).