1,882
Views
5
CrossRef citations to date
0
Altmetric
Research Paper

Dysbiotic but nonpathogenic shift in the fecal mycobiota of patients with rheumatoid arthritis

, ORCID Icon, , , , , & ORCID Icon show all
Article: 2149020 | Received 25 Aug 2022, Accepted 10 Nov 2022, Published online: 06 Dec 2022

References

  • Epstein FH, Harris ,ED. Rheumatoid arthritis: pathophysiology and implications for therapy. New Engl J Med. 1990;322(18):1277–16. doi:10.1056/NEJM199005033221805.
  • Klareskog L, Catrina AI, Paget S. Rheumatoid arthritis. Lancet. 2009;373:659–672.
  • Smolen JS, Aletaha D, Barton A, Burmester GR, Emery P, Firestein GS, Kavanaugh A, McInnes IB, Solomon DH, Strand V, et al. Rheumatoid arthritis. Nat Rev Dis Primers. 2018;4:18001.
  • Tsuchiya Y, Takayanagi N, Sugiura H, Miyahara Y, Tokunaga D, Kawabata Y, Sugita Y. Lung diseases directly associated with rheumatoid arthritis and their relationship to outcome. Eur Respir J. 2011;37:1411–1417.
  • Solomon DH, Goodson NJ, Katz JN, Weinblatt ME, Avorn J, Setoguchi S, Canning C, Schneeweiss S. Patterns of cardiovascular risk in rheumatoid arthritis. Ann Rheum Dis. 2006;65:1608–1612.
  • He J, Ding Y, Feng M, Guo J, Sun X, Zhao J, Yu D, Li Z. Characteristics of Sjögren’s syndrome in rheumatoid arthritis. Rheumatology. 2013;52:1084–1089.
  • Haugeberg G, Uhlig T, Falch JA, Halse JI, Kvien TK. Bone mineral density and frequency of osteoporosis in female patients with rheumatoid arthritis: results from 394 patients in the Oslo county rheumatoid arthritis register. Arthritis Rheumatol. 2000;43:522–530.
  • McInnes IB, Schett G. The pathogenesis of rheumatoid arthritis. New Engl J Med. 2011;365:2205–2219.
  • Stahl EA, Raychaudhuri S, Remmers EF, Xie G, Eyre S, Thomson BP, Li Y, Kurreeman FA, Zhernakova A, Hinks A. Genome-wide association study meta-analysis identifies seven new rheumatoid arthritis risk loci. Nat Genet. 2010;42:508.
  • Bae S-C. Epidemiology and etiology of rheumatoid arthritis. J Korean Med Assoc. 2010;53:843–852.
  • Okada Y, Wu D, Trynka G, Raj T, Terao C, Ikari K, Kochi Y, Ohmura K, Suzuki A, Yoshida S. Genetics of rheumatoid arthritis contributes to biology and drug discovery. Nature. 2014;506:376–381.
  • Li S, Yu Y, Yue Y, Zhang Z, Su K. Microbial infection and rheumatoid arthritis. J Clin Cell Immunol. 2013;4(6):1–6.
  • Zhang X, Zhang D, Jia H, Feng Q, Wang D, Liang D, Wu X, Li J, Tang L, Li Y. The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat Med. 2015;21:895–905.
  • H-J W, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y, DR L, Benoist C, Mathis D. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity. 2010;32:815–827.
  • Maeda Y, Takeda K. Host–microbiota interactions in rheumatoid arthritis. Exp Mol Med. 2019;51:1–6.
  • Maeda Y, Kurakawa T, Umemoto E, Motooka D, Ito Y, Gotoh K, Hirota K, Matsushita M, Furuta Y, Narazaki M. Dysbiosis contributes to arthritis development via activation of autoreactive T cells in the intestine. Arthritis Rheumatol. 2016;68:2646–2661.
  • Picchianti-Diamanti A, Panebianco C, Salemi S, Sorgi ML, Di Rosa R, Tropea A, Sgrulletti M, Salerno G, Terracciano F, D’amelio R. Analysis of gut microbiota in rheumatoid arthritis patients: disease-related dysbiosis and modifications induced by etanercept. Int J Mol Sci. 2018;19:2938.
  • Chen J, Wright K, Davis JM, Jeraldo P, Marietta EV, Murray J, Nelson H, Matteson EL, Taneja V. An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis. Genome Med. 2016;8:1–14.
  • Rooney CM, Mankia K, Mitra S, Moura IB, Emery P, Wilcox MH. Perturbations of the gut microbiome in anti-CCP positive individuals at risk of developing rheumatoid arthritis. Rheumatology. 2021;60:3380–3387.
  • Chiang H-I, J-R L, Liu -C-C, Liu P-Y, Chen -H-H, Chen Y-M, Lan J-L, Chen D-Y. An association of gut microbiota with different phenotypes in Chinese patients with rheumatoid arthritis. J Clin Med. 2019;8:1770.
  • Yoshitomi H, Sakaguchi N, Kobayashi K, Brown GD, Tagami T, Sakihama T, Hirota K, Tanaka S, Nomura T, Miki I. A role for fungal β-glucans and their receptor Dectin-1 in the induction of autoimmune arthritis in genetically susceptible mice. J Exp Med. 2005;201:949–960.
  • Sun X, Wang Y, Li X, Wang M, Dong J, Tang W, Lei Z, Guo Y, Li M, Li Y. Alterations of gut fungal microbiota in patients with rheumatoid arthritis. PeerJ. 2022;10:e13037.
  • Artacho A, Isaac S, Nayak R, Flor‐Duro A, Alexander M, Koo I, Manasson J, Smith PB, Rosenthal P, Homsi Y. The pretreatment gut microbiome is associated with lack of response to methotrexate in new‐onset rheumatoid arthritis. Arthritis Rheumatol. 2021;73:931–942.
  • Jeong Y, Kim J-W, You HJ, Park S-J, Lee J, Ju JH, Park MS, Jin H, Cho M-L, Kwon B. Gut microbial composition and function are altered in patients with early rheumatoid arthritis. J Clin Med. 2019;8:693.
  • Liu Z, Wu Y, Luo Y, Wei S, Lu C, Zhou Y, Wang J, Miao T, Lin H, Zhao Y. Self-balance of intestinal flora in spouses of patients with rheumatoid arthritis. Front Med. 2020;7:538–549.
  • Sam QH, Chang MW, Chai LYA. The fungal mycobiome and its interaction with gut bacteria in the host. Int J Mol Sci. 2017;18:330.
  • Deveau A, Bonito G, Uehling J, Paoletti M, Becker M, Bindschedler S, Hacquard S, Hervé V, Labbé J, Lastovetsky OA. Bacterial–fungal interactions: ecology, mechanisms and challenges. FEMS Microbiol Rev. 2018;42:335–352.
  • van Tilburg Bernardes E, Pettersen VK, Gutierrez MW, Laforest-Lapointe I, Jendzjowsky NG, Cavin J-B, Vicentini FA, Keenan CM, Ramay HR, Samara J. Intestinal fungi are causally implicated in microbiome assembly and immune development in mice. Nat Commun. 2020;11:1–16.
  • Kumar A, Asthana M, Gupta A, Nigam D, Mahajan S. 2018. Secondary metabolism and antimicrobial metabolites of Penicillium. In: Gupta V, editor. New and future developments in microbial biotechnology and bioengineering. Amsterdam, The Netherlands: Elsevier; p. 47–68.
  • Mangalea MR, Paez-Espino D, Kieft K, Chatterjee A, Chriswell ME, Seifert JA, Feser ML, Demoruelle MK, Sakatos A, Anantharaman K. Individuals at risk for rheumatoid arthritis harbor differential intestinal bacteriophage communities with distinct metabolic potential. Cell Host Microbe. 2021;29:726–739. e725.
  • Nash AK, Auchtung TA, Wong MC, Smith DP, Gesell JR, Ross MC, Stewart CJ, Metcalf GA, Muzny DM, Gibbs RA. The gut mycobiome of the human microbiome project healthy cohort. Microbiome. 2017;5:1–13.
  • Wu X, Xia Y, He F, Zhu C, Ren W. Intestinal mycobiota in health and diseases: from a disrupted equilibrium to clinical opportunities. Microbiome. 2021;9:1–18.
  • Hallen-Adams HE, Suhr MJ. Fungi in the healthy human gastrointestinal tract. Virulence. 2017;8:352–358.
  • Suhr MJ, Banjara N, Hallen‐Adams HE. Sequence‐based methods for detecting and evaluating the human gut mycobiome. Lett Appl Microbiol. 2016;62:209–215.
  • Motooka D, Fujimoto K, Tanaka R, Yaguchi T, Gotoh K, Maeda Y, Furuta Y, Kurakawa T, Goto N, Yasunaga T. Fungal ITS1 deep-sequencing strategies to reconstruct the composition of a 26-species community and evaluation of the gut mycobiota of healthy Japanese individuals. Front Microbiol. 2017;8:238.
  • Qiu X, Zhao X, Cui X, Mao X, Tang N, Jiao C, Wang D, Zhang Y, Ye Z, Zhang H. Characterization of fungal and bacterial dysbiosis in young adult Chinese patients with Crohn’s disease. Therap Adv Gastroenterol. 2020;13:1756284820971202.
  • Gurleen K, Savio R. Prevalence of candida in diarrhoeal stools. IOSR J Dent and Med Sci. 2016;15:47–49.
  • Limon JJ, Skalski JH, Underhill DM. Commensal fungi in health and disease. Cell Host Microbe. 2017;22:156–165.
  • Li Q, Wang C, Tang C, He Q, Li N, Li J. Dysbiosis of gut fungal microbiota is associated with mucosal inflammation in Crohn’s disease. J Clin Gastroenterol. 2014;48:513.
  • Sokol H, Leducq V, Aschard H, Pham H-P, Jegou S, Landman C, Cohen D, Liguori G, Bourrier A, Nion-Larmurier I. Fungal microbiota dysbiosis in IBD. Gut. 2017;66:1039–1048.
  • Imai T, Inoue R, Kawada Y, Morita Y, Inatomi O, Nishida A, Bamba S, Kawahara M, Andoh A. Characterization of fungal dysbiosis in Japanese patients with inflammatory bowel disease. J Gastroenterol. 2019;54:149–159.
  • Bishu S, Su EW, Wilkerson ER, Reckley KA, Jones DM, McGeachy MJ, Gaffen SL, Levesque MC. Rheumatoid arthritis patients exhibit impaired Candida albicans-specific Th17 responses. Arthrit Res Ther. 2014;16:1–9.
  • Aletaha D, Neogi T, Silman AJ, Funovits J, Felson DT, Bingham IIICO, Birnbaum NS, Burmester GR, Bykerk VP, Cohen MD. rheumatoid arthritis classification criteria: an American college of rheumatology/European league against rheumatism collaborative initiative. Arthritis Rheum. 2010;62:2569–2581.
  • Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJA, Holmes SP. DADA2: high-resolution sample inference from Illumina amplicon data. Nat Methods. 2016;13:581–583.
  • Rognes T, Flouri T, Nichols B, Quince C, Mahé F. VSEARCH: a versatile open source tool for metagenomics. PeerJ. 2016;4:e2584.
  • Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Yarza P, Peplies J, Glöckner FO. The SILVA ribosomal RNA gene database project: improved data processing and web-based tools. Nucleic Acids Res. 2012;41:D590–596.
  • Edgar RC, Haas BJ, Clemente JC, Quince C, Knight R. UCHIME improves sensitivity and speed of chimera detection. Bioinformatics. 2011;27:2194–2200.
  • Nilsson RH, Tedersoo L, Ryberg M, Kristiansson E, Hartmann M, Unterseher M, Porter TM, Bengtsson-Palme J, Walker DM, De Sousa F. A comprehensive, automatically updated fungal ITS sequence dataset for reference-based chimera control in environmental sequencing efforts. Microbes Environ. 2015;30:145–150.
  • Bokulich NA, Kaehler BD, Rideout JR, Dillon M, Bolyen E, Knight R, Huttley GA, Caporaso JG. Optimizing taxonomic classification of marker-gene amplicon sequences with QIIME 2ʹs q2-feature-classifier plugin. Microbiome. 2018;6:1–17.
  • Abarenkov K, Henrik Nilsson R, Larsson KH, Alexander IJ, Eberhardt U, Erland S, Høiland K, Kjøller R, Larsson E, Pennanen T. The UNITE database for molecular identification of fungi–recent updates and future perspectives. New Phytol. 2010;186:281–285.
  • McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One. 2013;8:e61217.
  • R-Core-Team. R: a Language and environment for statistical computing. Vienna: R Foundation for Statistical Computing; 2017.
  • Paulson JN, Stine OC, Bravo HC, Pop M. Differential abundance analysis for microbial marker-gene surveys. Nat Methods. 2013;10:1200–1202.
  • Oksanen J, Blanchet FG, Kindt R, Legendre P, Minchin PR, O’Hara RB, Simpson GL, Solymos P, Stevens MHH, Wagner H. vegan: community ecology package. R package version 2.5-3. 2018.
  • Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, Huttenhower C. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12:R60.
  • Friedman J, Alm EJ. Inferring correlation networks from genomic survey data. PLoS Comput Biol. 2012;8:e1002687.
  • Kurtz ZD, Müller CL, Miraldi ER, Littman DR, Blaser MJ, Bonneau RA. Sparse and compositionally robust inference of microbial ecological networks. PLoS Comput Biol. 2015;11:e1004226.
  • Bastian M, Heymann S, Jacomy M. 2009. Gephi: an open source software for exploring and manipulating networks. Proceedings of the international AAAI conference on web and social media; San Jose, California. 361–362.
  • Durán P, Thiergart T, Garrido-Oter R, Agler M, Kemen E, Schulze-Lefert P, Hacquard S. Microbial interkingdom interactions in roots promote Arabidopsis survival. Cell. 2018;175:973–983. e914.
  • Csardi G, Nepusz T. The igraph software package for complex network research. Int J Complex Syst. 2006;1695:1–9.
  • Frykman PK, Nordenskjöld A, Kawaguchi A, Hui TT, Granström AL, Cheng Z, Tang J, Underhill DM, Iliev I, Funari VA, et al. Characterization of bacterial and fungal microbiome in children with Hirschsprung disease with and without a history of enterocolitis: a multicenter study. PLoS One. 2015;10:e0124172–e0124172.