Microbe-metabolite-host axis, two-way action in the pathogenesis and treatment of human autoimmunity
Tài liệu tham khảo
Blumberg, 2012, Microbiota, disease, and back to health: a metastable journey, Sci Transl Med, 4, 137rv7, 10.1126/scitranslmed.3004184
Turner, 2009, Intestinal mucosal barrier function in health and disease, Nat Rev Immunol, 9, 799, 10.1038/nri2653
Ley, 2006, Ecological and evolutionary forces shaping microbial diversity in the human intestine, Cell, 124, 837, 10.1016/j.cell.2006.02.017
Qin, 2010, A human gut microbial gene catalogue established by metagenomic sequencing, Nature, 464, 59, 10.1038/nature08821
Eckburg, 2005, Diversity of the human intestinal microbial flora, Science, 308, 1635, 10.1126/science.1110591
Ward, 1990, 16S rRNA sequences reveal numerous uncultured microorganisms in a natural community, Nature, 345, 63, 10.1038/345063a0
Sanger, 1977, DNA sequencing with chain-terminating inhibitors, Proc Natl Acad Sci U S A, 74, 5463, 10.1073/pnas.74.12.5463
Sanger, 1975, A rapid method for determining sequences in DNA by primed synthesis with DNA polymerase, J Mol Biol, 94, 441, 10.1016/0022-2836(75)90213-2
Dominguez-Bello, 2011, Development of the human gastrointestinal microbiota and insights from high-throughput sequencing, Gastroenterology, 140, 1713, 10.1053/j.gastro.2011.02.011
Quince, 2017, Shotgun metagenomics, from sampling to analysis, Nat Biotechnol, 35, 833, 10.1038/nbt.3935
Cox, 2017, A fast and robust protocol for metataxonomic analysis using RNAseq data, Microbiome, 5, 7, 10.1186/s40168-016-0219-5
Jiang, 2017, DACE: a scalable DP-means algorithm for clustering extremely large sequence data, Bioinformatics, 33, 834, 10.1093/bioinformatics/btw722
Kuczynski, 2011, Experimental and analytical tools for studying the human microbiome, Nat Rev Genet, 13, 47, 10.1038/nrg3129
Mandal, 2015, Metagenomic surveys of gut microbiota, Genomics Proteomics Bioinformatics, 13, 148, 10.1016/j.gpb.2015.02.005
Holmes, 2012, Gut microbiota composition and activity in relation to host metabolic phenotype and disease risk, Cell Metab, 16, 559, 10.1016/j.cmet.2012.10.007
Clemente, 2012, The impact of the gut microbiota on human health: an integrative view, Cell, 148, 1258, 10.1016/j.cell.2012.01.035
Sharon, 2014, Specialized metabolites from the microbiome in health and disease, Cell Metab, 20, 719, 10.1016/j.cmet.2014.10.016
Nicholson, 2012, Host-gut microbiota metabolic interactions, Science, 336, 1262, 10.1126/science.1223813
Koh, 2016, From Dietary Fiber to Host Physiology: Short-Chain Fatty Acids as Key Bacterial Metabolites, Cell, 165, 1332, 10.1016/j.cell.2016.05.041
Cho, 2012, The human microbiome: at the interface of health and disease, Nat Rev Genet, 13, 260, 10.1038/nrg3182
Brestoff, 2013, Commensal bacteria at the interface of host metabolism and the immune system, Nat Immunol, 14, 676, 10.1038/ni.2640
Mu, 2017, Control of lupus nephritis by changes of gut microbiota, Microbiome, 5, 73, 10.1186/s40168-017-0300-8
Atarashi, 2013, Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota, Nature, 500, 232, 10.1038/nature12331
Tanoue, 2016, Development and maintenance of intestinal regulatory T cells, Nat Rev Immunol, 16, 295, 10.1038/nri.2016.36
Arpaia, 2013, Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation, Nature, 504, 451, 10.1038/nature12726
Smith, 2013, The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis, Science, 341, 569, 10.1126/science.1241165
Round, 2010, Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of the intestinal microbiota, Proc Natl Acad Sci U S A, 107, 12204, 10.1073/pnas.0909122107
Huber, 2012, Life, death, and miracles: Th17 cells in the intestine, Eur J Immunol, 42, 2238, 10.1002/eji.201242619
Rothschild, 2018, Environment dominates over host genetics in shaping human gut microbiota, Nature, 555, 210, 10.1038/nature25973
Mattila, 2012, Fecal transplantation, through colonoscopy, is effective therapy for recurrent Clostridium difficile infection, Gastroenterology, 142, 490, 10.1053/j.gastro.2011.11.037
Ivanov, 2009, Induction of intestinal Th17 cells by segmented filamentous bacteria, Cell, 139, 485, 10.1016/j.cell.2009.09.033
Panea, 2015, Intestinal monocyte-derived macrophages control commensal-specific Th17 responses, Cell Rep, 12, 1314, 10.1016/j.celrep.2015.07.040
Goto, 2014, Segmented filamentous bacteria antigens presented by intestinal dendritic cells drive mucosal Th17 cell differentiation, Immunity, 40, 594, 10.1016/j.immuni.2014.03.005
Kumar, 2016, Intestinal Interleukin-17 Receptor Signaling Mediates Reciprocal Control of the Gut Microbiota and Autoimmune Inflammation, Immunity, 44, 659, 10.1016/j.immuni.2016.02.007
Atarashi, 2011, Induction of colonic regulatory T cells by indigenous Clostridium species, Science, 331, 337, 10.1126/science.1198469
de Pablo, 2008, Association of periodontal disease and tooth loss with rheumatoid arthritis in the US population, J Rheumatol, 35, 70
Chen, 2013, Association between a history of periodontitis and the risk of rheumatoid arthritis: a nationwide, population-based, case-control study, Ann Rheum Dis, 72, 1206, 10.1136/annrheumdis-2012-201593
Quirke, 2014, Heightened immune response to autocitrullinated Porphyromonas gingivalis peptidylarginine deiminase: a potential mechanism for breaching immunologic tolerance in rheumatoid arthritis, Ann Rheum Dis, 73, 263, 10.1136/annrheumdis-2012-202726
McGraw, 1999, Purification, characterization, and sequence analysis of a potential virulence factor from Porphyromonas gingivalis, peptidylarginine deiminase, Infect Immun, 67, 3248, 10.1128/IAI.67.7.3248-3256.1999
Veith, 2014, Porphyromonas gingivalis outer membrane vesicles exclusively contain outer membrane and periplasmic proteins and carry a cargo enriched with virulence factors, J Proteome Res, 13, 2420, 10.1021/pr401227e
Gui, 2016, Spheres of influence: Porphyromonas gingivalis outer membrane vesicles, Mol Oral Microbiol, 31, 365, 10.1111/omi.12134
Vossenaar, 2003, PAD, a growing family of citrullinating enzymes: genes, features and involvement in disease, BioEssays, 25, 1106, 10.1002/bies.10357
Maresz, 2013, Porphyromonas gingivalis facilitates the development and progression of destructive arthritis through its unique bacterial peptidylarginine deiminase (PAD), PLoS Pathog, 9, 10.1371/journal.ppat.1003627
Avouac, 2006, Diagnostic and predictive value of anti-cyclic citrullinated protein antibodies in rheumatoid arthritis: a systematic literature review, Ann Rheum Dis, 65, 845, 10.1136/ard.2006.051391
Schellekens, 2000, The diagnostic properties of rheumatoid arthritis antibodies recognizing a cyclic citrullinated peptide, Arthritis Rheum, 43, 155, 10.1002/1529-0131(200001)43:1<155::AID-ANR20>3.0.CO;2-3
Rosenstein, 2004, Hypothesis: the humoral immune response to oral bacteria provides a stimulus for the development of rheumatoid arthritis, Inflammation, 28, 311, 10.1007/s10753-004-6641-z
Kwon, 2016, Pep19 drives epitope spreading in periodontitis and periodontitis-associated autoimmune diseases, J Periodontal Res, 51, 381, 10.1111/jre.12318
Jeong, 2012, Predominant immunoreactivity of Porphyromonas gingivalis heat shock protein in autoimmune diseases, Periodontal Res, 47, 811, 10.1111/j.1600-0765.2012.01501.x
Cummings, 1983, Fermentation in the human large intestine: evidence and implications for health, The Lancet, 321, 1206, 10.1016/S0140-6736(83)92478-9
Macia, 2012, Microbial influences on epithelial integrity and immune function as a basis for inflammatory diseases, Immunol Rev, 245, 164, 10.1111/j.1600-065X.2011.01080.x
Cummings, 1987, Short chain fatty acids in human large intestine, portal, hepatic and venous blood, Gut, 28, 1221, 10.1136/gut.28.10.1221
Qiao, 2014, Butyrate protects liver against ischemia reperfusion injury by inhibiting nuclear factor kappa B activation in Kupffer cells, J Surg Res, 187, 653, 10.1016/j.jss.2013.08.028
Liu, 2014, Butyrate protects rat liver against total hepatic ischemia reperfusion injury with bowel congestion, PLoS One, 9, 10.1371/journal.pone.0106184
Haghikia, 2015, Dietary fatty acids directly impact central nervous system autoimmunity via the small intestine, Immunity, 43, 817, 10.1016/j.immuni.2015.09.007
Berndt, 2012, Butyrate increases IL-23 production by stimulated dendritic cells, Am J Physiol Gastrointest Liver Physiol, 303, G1384, 10.1152/ajpgi.00540.2011
Chang, 2014, The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition, Proc Natl Acad Sci U S A, 111, 2247, 10.1073/pnas.1322269111
Blad, 2012, G protein-coupled receptors for energy metabolites as new therapeutic targets, Nature reviews Nat Rev Drug Discov, 11, 603, 10.1038/nrd3777
Singh, 2014, Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis, Immunity, 40, 128, 10.1016/j.immuni.2013.12.007
Maslowski, 2009, Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43, Nature, 461, 1282, 10.1038/nature08530
Vieira, 2015, A Role for Gut Microbiota and the Metabolite-Sensing Receptor GPR43 in a Murine Model of Gout, Arthritis Rheumatol, 67, 1646, 10.1002/art.39107
Macia L, Tan J, Vieira AT, Leach K, Stanley D, Luong S, et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat Commun 2015;6(undefined):6734.
Nowarski, 2015, Epithelial IL-18 Equilibrium Controls Barrier Function in Colitis, Cell, 163, 1444, 10.1016/j.cell.2015.10.072
Kim MH, Kang SG, Park JH, Yanagisawa M, Kim CH. Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology 2013;145(2):396–406.e1–10.
Trompette, 2014, Gut microbiota metabolism of dietary fiber influences allergic airway disease and hematopoiesis, Nat Med, 20, 159, 10.1038/nm.3444
Seljeset, 2012, Receptor-specific regulation of ERK1/2 activation by members of the "free fatty acid receptor" family, J Recept Signal Transduct Res, 32, 196, 10.3109/10799893.2012.692118
Nastasi C, Candela M, Bonefeld CM, Geisler C, Hansen M, Krejsgaard T, et al. The effect of short-chain fatty acids on human monocyte-derived dendritic cells. Sci Rep 2015;5(undefined):16148.
Thangaraju, 2008, Sodium-coupled transport of the short chain fatty acid butyrate by SLC5A8 and its relevance to colon cancer, J Gastrointest Surg, 12, 1773, 10.1007/s11605-008-0573-0
Gurav, 2015, Slc5a8, a Na+-coupled high-affinity transporter for short-chain fatty acids, is a conditional tumour suppressor in colon that protects against colitis and colon cancer under low-fibre dietary conditions, Biochem J, 469, 267, 10.1042/BJ20150242
Singh, 2010, Blockade of dendritic cell development by bacterial fermentation products butyrate and propionate through a transporter (Slc5a8)-dependent inhibition of histone deacetylases, J Biol Chem, 285, 27601, 10.1074/jbc.M110.102947
Furusawa, 2013, Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells, Nature, 504, 446, 10.1038/nature12721
Kespohl M, Vachharajani N, Luu M, Harb H, Pautz S, Wolff S, et al. The microbial metabolite butyrate induces expression of Th1-associated factors in CD4 T cells. Front Immunol 2017;8(undefined):1036.
Fellows, 2018, Microbiota derived short chain fatty acids promote histone crotonylation in the colon through histone deacetylases, Nat Commun, 9, 105, 10.1038/s41467-017-02651-5
Liu L, Li L, Min J, Wang J, Wu H, Zeng Y, et al. Butyrate interferes with the differentiation and function of human monocyte-derived dendritic cells. Cell Immunol 2012;277(null):66–73.
Park, 2015, Short-chain fatty acids induce both effector and regulatory T cells by suppression of histone deacetylases and regulation of the mTOR-S6K pathway, Mucosal Immunol, 8, 80, 10.1038/mi.2014.44
Kim, 2016, Gut microbial metabolites fuel host antibody responses, Cell Host Microbe, 20, 202, 10.1016/j.chom.2016.07.001
Kim, 2016, B cell-helping functions of gut microbial metabolites, Microbial cell, 3, 529, 10.15698/mic2016.10.536
Fukuda, 2011, Bifidobacteria can protect from enteropathogenic infection through production of acetate, Nature, 469, 543, 10.1038/nature09646
Wrzosek L, Miquel S, Noordine ML, Bouet S, Joncquel Chevalier-Curt M, Robert V, et al. Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent. BMC Biol 2013;11(undefined):61.
Huuskonen, 2004, Regulation of microglial inflammatory response by sodium butyrate and short-chain fatty acids, Br J Pharmacol, 141, 874, 10.1038/sj.bjp.0705682
Erny, 2015, Host microbiota constantly control maturation and function of microglia in the CNS, Nat Neurosci, 18, 965, 10.1038/nn.4030
Zelante, 2013, Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22, Immunity, 39, 372, 10.1016/j.immuni.2013.08.003
Schroeder, 2016, Signals from the gut microbiota to distant organs in physiology and disease, Nat Med, 22, 1079, 10.1038/nm.4185
Lamas, 2016, CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands, Nat Med, 22, 598, 10.1038/nm.4102
Stockinger B, Di Meglio P, Gialitakis M, Duarte JH. The aryl hydrocarbon receptor: multitasking in the immune system. Annu Rev Immunol 2014;32(undefined):403–32.
Levy, 2015, Microbiota-modulated metabolites shape the intestinal microenvironment by regulating NLRP6 inflammasome signaling, Cell, 163, 1428, 10.1016/j.cell.2015.10.048
Ridlon, 2016, Consequences of bile salt biotransformations by intestinal bacteria, Gut microbes, 7, 22, 10.1080/19490976.2015.1127483
Parks, 1999, Bile acids: natural ligands for an orphan nuclear receptor, Science, 284, 1365, 10.1126/science.284.5418.1365
Gadaleta, 2011, Farnesoid X receptor activation inhibits inflammation and preserves the intestinal barrier in inflammatory bowel disease, Gut, 60, 463, 10.1136/gut.2010.212159
Makishima, 1999, Identification of a nuclear receptor for bile acids, Science, 284, 1362, 10.1126/science.284.5418.1362
Liu, 2016, Cholesterol 7α-hydroxylase protects the liver from inflammation and fibrosis by maintaining cholesterol homeostasis, J Lipid Res, 57, 1831, 10.1194/jlr.M069807
Cao W, Kayama H, Chen ML, Delmas A, Sun A, Kim SY, et al. The xenobiotic transporter Mdr1 enforces T cell homeostasis in the presence of intestinal bile acids. Immunity 2017;47(6):1182–96.e10.
Mazmanian, 2008, A microbial symbiosis factor prevents intestinal inflammatory disease, Nature, 453, 620, 10.1038/nature07008
Dasgupta, 2014, Plasmacytoid dendritic cells mediate anti-inflammatory responses to a gut commensal molecule via both innate and adaptive mechanisms, Cell Host Microbe, 15, 413, 10.1016/j.chom.2014.03.006
Round, 2011, The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota, Science, 332, 974, 10.1126/science.1206095
Telesford, 2015, A commensal symbiotic factor derived from Bacteroides fragilis promotes human CD39(+)Foxp3(+) T cells and Treg function, Gut microbes, 6, 234, 10.1080/19490976.2015.1056973
Yaron, 2015, K(+) regulates Ca(2+) to drive inflammasome signaling: dynamic visualization of ion flux in live cells, Cell Death Dis, 6, e1954, 10.1038/cddis.2015.277
Perruzza, 2017, T Follicular Helper Cells Promote a Beneficial Gut Ecosystem for Host Metabolic Homeostasis by Sensing Microbiota-Derived Extracellular ATP, Cell Rep, 18, 2566, 10.1016/j.celrep.2017.02.061
Faas, 2017, Extracellular ATP and adenosine: the Yin and Yang in immune responses?, Mol Aspects Med, 55, 9, 10.1016/j.mam.2017.01.002
Atarashi, 2008, ATP drives lamina propria T(H)17 cell differentiation, Nature, 455, 808, 10.1038/nature07240
Kusu, 2013, Ecto-nucleoside triphosphate diphosphohydrolase 7 controls Th17 cell responses through regulation of luminal ATP in the small intestine, J Immunol, 190, 774, 10.4049/jimmunol.1103067
Van Praet, 2015, Commensal microbiota influence systemic autoimmune responses, EMBO J, 34, 466, 10.15252/embj.201489966
Yurkovetskiy, 2013, Gender bias in autoimmunity is influenced by microbiota, Immunity, 39, 400, 10.1016/j.immuni.2013.08.013
Molodecky NA, Soon IS, Rabi DM, Ghali WA, Ferris M, Chernoff G, et al. Increasing incidence and prevalence of the inflammatory bowel diseases with time, based on systematic review. Gastroenterology 2012;142(1):46–54.e42; quiz e30.
Zhao, 2013, First prospective, population-based inflammatory bowel disease incidence study in mainland of China: the emergence of "western" disease, Inflamm Bowel Dis, 19, 1839
Navaneethan, 2010, Hepatopancreatobiliary manifestations and complications associated with inflammatory bowel disease, Inflamm Bowel Dis, 16, 1598, 10.1002/ibd.21219
Liu, 2015, Association analyses identify 38 susceptibility loci for inflammatory bowel disease and highlight shared genetic risk across populations, Nat Genet, 47, 979, 10.1038/ng.3359
Jostins, 2012, Host-microbe interactions have shaped the genetic architecture of inflammatory bowel disease, Nature, 491, 119, 10.1038/nature11582
Ananthakrishnan, 2018, Environmental triggers in IBD: a review of progress and evidence, Nat Rev Gastroenterol Hepatol, 15, 39, 10.1038/nrgastro.2017.136
Lewis, 2015, Inflammation, antibiotics, and diet as environmental stressors of the gut microbiome in pediatric crohn's disease, Cell Host Microbe, 18, 489, 10.1016/j.chom.2015.09.008
Ahmed, 2016, Microbiome, metabolome and inflammatory bowel disease, Microorganisms, 4, 10.3390/microorganisms4020020
Frank, 2007, Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases, Proc Natl Acad Sci U S A, 104, 13780, 10.1073/pnas.0706625104
Manichanh, 2006, Reduced diversity of faecal microbiota in Crohn's disease revealed by a metagenomic approach, Gut, 55, 205, 10.1136/gut.2005.073817
Takahashi, 2016, Reduced abundance of butyrate-producing bacteria species in the fecal microbial community in crohn's disease, Digestion, 93, 59, 10.1159/000441768
Joossens, 2011, Dysbiosis of the faecal microbiota in patients with Crohn's disease and their unaffected relatives, Gut, 60, 631, 10.1136/gut.2010.223263
Fujimoto, 2013, Decreased abundance of Faecalibacterium prausnitzii in the gut microbiota of Crohn's disease, J Gastroenterol Hepatol, 28, 613, 10.1111/jgh.12073
Magnusson, 2016, Anti-TNF therapy response in patients with ulcerative colitis is associated with colonic antimicrobial peptide expression and microbiota composition, J Crohns Colitis, 10, 943, 10.1093/ecco-jcc/jjw051
Hall, 2017, A novel Ruminococcus gnavus clade enriched in inflammatory bowel disease patients, Genome Med, 9, 103, 10.1186/s13073-017-0490-5
Mondot, 2011, Highlighting new phylogenetic specificities of Crohn's disease microbiota, Inflamm Bowel Dis, 17, 185, 10.1002/ibd.21436
Machiels, 2014, A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis, Gut, 63, 1275, 10.1136/gutjnl-2013-304833
Chen, 2014, Characteristics of fecal and mucosa-associated microbiota in Chinese patients with inflammatory bowel disease, Medicine, 93, 10.1097/MD.0000000000000051
Zhang T, Ding C, Zhao M, Dai X, Yang J, Li Y, et al. Sodium butyrate reduces colitogenic immunoglobulin a-coated bacteria and modifies the composition of microbiota in IL-10 deficient mice. Nutrients 2016;8(12):undefined.
Duboc, 2013, Connecting dysbiosis, bile-acid dysmetabolism and gut inflammation in inflammatory bowel diseases, Gut, 62, 531, 10.1136/gutjnl-2012-302578
Pavlidis, 2015, Systematic review: bile acids and intestinal inflammation-luminal aggressors or regulators of mucosal defence?, Aliment Pharmacol Ther, 42, 802, 10.1111/apt.13333
Wellman, 2017, Intestinal epithelial sirtuin 1 regulates intestinal inflammation during aging in mice by altering the intestinal microbiota, Gastroenterology, 153, 772, 10.1053/j.gastro.2017.05.022
Jing, 2014, Peptidoglycan recognition protein 3 and Nod2 synergistically protect mice from dextran sodium sulfate-induced colitis, J Immunol, 193, 3055, 10.4049/jimmunol.1301548
Schirmer, 2018, Dynamics of metatranscription in the inflammatory bowel disease gut microbiome, Nat Microbiol, 3, 337, 10.1038/s41564-017-0089-z
Huang, 2015, Using corticosteroids to reshape the gut microbiome: implications for inflammatory bowel diseases, Inflamm Bowel Dis, 21, 963, 10.1097/MIB.0000000000000332
Ebringer, 2010, Rheumatoid arthritis, Proteus, anti-CCP antibodies and Karl Popper, Autoimmun Rev, 9, 216, 10.1016/j.autrev.2009.10.006
Bogdanos DP, Smyk DS, Rigopoulou EI, Mytilinaiou MG, Heneghan MA, Selmi C, et al. Twin studies in autoimmune disease: genetics, gender and environment. J Autoimmun 2012;38(null):J156–69.
Wellcome Trust Case Control Consortium, 2007, Genome-wide association study of 14,000 cases of seven common diseases and 3,000 shared controls, Nature, 447, 661, 10.1038/nature05911
Eleftherohorinou, 2011, Pathway-driven gene stability selection of two rheumatoid arthritis GWAS identifies and validates new susceptibility genes in receptor mediated signalling pathways, Hum Mol Genet, 20, 3494, 10.1093/hmg/ddr248
Firestein, 2017, Immunopathogenesis of rheumatoid arthritis, Immunity, 46, 183, 10.1016/j.immuni.2017.02.006
Newkirk, 2010, Distinct bacterial colonization patterns of Escherichia coli subtypes associate with rheumatoid factor status in early inflammatory arthritis, Rheumatology (Oxford), 49, 1311, 10.1093/rheumatology/keq088
Chen, 2016, An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis, Genome Med, 8, 43, 10.1186/s13073-016-0299-7
Vaahtovuo, 2008, Fecal microbiota in early rheumatoid arthritis, J Rheumatol, 35, 1500
Liu, 2013, Analysis of fecal Lactobacillus community structure in patients with early rheumatoid arthritis, Curr Microbiol, 67, 170, 10.1007/s00284-013-0338-1
Scher, 2013, Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis, eLife, 2, 10.7554/eLife.01202
Jubair, 2018, Modulation of inflammatory arthritis in mice by gut microbiota through mucosal inflammation and autoantibody generation, Arthritis Rheumatol, 70, 1220, 10.1002/art.40490
Gomez, 2012, Loss of sex and age driven differences in the gut microbiome characterize arthritis-susceptible 0401 mice but not arthritis-resistant 0402 mice, PLoS One, 7, 10.1371/journal.pone.0036095
Al-Katma, 2007, Control of periodontal infection reduces the severity of active rheumatoid arthritis, J Clin Rheumatol, 13, 134, 10.1097/RHU.0b013e3180690616
Li, 2017, Rheumatoid arthritis and periodontal disease: What are the similarities and differences?, Int J Rheum Dis, 20, 1887, 10.1111/1756-185X.13240
Scher, 2012, Periodontal disease and the oral microbiota in new-onset rheumatoid arthritis, Arthritis Rheum, 64, 3083, 10.1002/art.34539
Sato, 2017, Aggravation of collagen-induced arthritis by orally administered Porphyromonas gingivalis through modulation of the gut microbiota and gut immune system, Sci Rep, 7, 6955, 10.1038/s41598-017-07196-7
Marchesan, 2013, Porphyromonas gingivalis oral infection exacerbates the development and severity of collagen-induced arthritis, Arthritis Res Ther, 15, R186, 10.1186/ar4376
Pischon, 2009, Effects of Porphyromonas gingivalis on cell cycle progression and apoptosis of primary human chondrocytes, Ann Rheum Dis, 68, 1902, 10.1136/ard.2008.102392
Wegner, 2010, Peptidylarginine deiminase from Porphyromonas gingivalis citrullinates human fibrinogen and α-enolase: implications for autoimmunity in rheumatoid arthritis, Arthritis Rheum, 62, 2662, 10.1002/art.27552
Okada, 2013, Periodontal treatment decreases levels of antibodies to Porphyromonas gingivalis and citrulline in patients with rheumatoid arthritis and periodontitis, J Periodontol, 84, e74, 10.1902/jop.2013.130079
Wu HJ, Ivanov II, Darce J, Hattori K, Shima T, Umesaki Y, et al. Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells. Immunity 2010;32(6):815–27.
Block, 2016, Gut Microbiota Regulates K/BxN Autoimmune Arthritis through Follicular Helper T but Not Th17 Cells, J Immunol, 196, 1550, 10.4049/jimmunol.1501904
Taneja, 2017, Microbiome in 2016: T follicular helper cells and the gut microbiome in arthritis, Nat Rev Rheumatol, 13, 72, 10.1038/nrrheum.2016.222
Doria A, Gatto M, Zen M, Iaccarino L, Punzi L. Optimizing outcome in SLE: treating-to-target and definition of treatment goals. Autoimmun Rev 2014;13(7):770–7.
Doria A, Zen M, Iaccarino L. Remission in SLE: the duration depends on multiple factors, including the definition. Ann Rheum Dis 2016;75(12):e77-e.
Wang, 2016, Elevated expression of miR-142-3p is related to the pro-inflammatory function of monocyte-derived dendritic cells in SLE, Arthritis Res Ther, 18, 263, 10.1186/s13075-016-1158-z
Rees, 2016, The incidence and prevalence of systemic lupus erythematosus in the UK, 1999-2012, Ann Rheum Dis, 75, 136, 10.1136/annrheumdis-2014-206334
Zhang, 2014, Dynamics of gut microbiota in autoimmune lupus, Appl Environ Microbiol, 80, 7551, 10.1128/AEM.02676-14
Luo, 2018, Gut microbiota in human systemic lupus erythematosus and a mouse model of lupus, Appl Environ Microbiol, 84, 10.1128/AEM.02288-17
Macanovic, 1996, The treatment of systemic lupus erythematosus (SLE) in NZB/W F1 hybrid mice; studies with recombinant murine DNase and with dexamethasone, Clin Exp Immunol, 106, 243, 10.1046/j.1365-2249.1996.d01-839.x
Hevia, 2014, Intestinal dysbiosis associated with systemic lupus erythematosus, mBio, 5, 10.1128/mBio.01548-14
He, 2016, Alterations of the gut microbiome in Chinese patients with systemic lupus erythematosus, Gut Pathog, 8, 64, 10.1186/s13099-016-0146-9
Rojo, 2015, Ranking the impact of human health disorders on gut metabolism: systemic lupus erythematosus and obesity as study cases, Sci Rep, 5, 8310, 10.1038/srep08310
López, 2016, Th17 responses and natural IgM antibodies are related to gut microbiota composition in systemic lupus erythematosus patients, Sci Rep, 6, 24072, 10.1038/srep24072
Kosiewicz, 2014, Relationship between gut microbiota and development of T cell associated disease, FEBS Lett, 588, 4195, 10.1016/j.febslet.2014.03.019
Johnson, 2015, Impact of dietary deviation on disease progression and gut microbiome composition in lupus-prone SNF1 mice, Clin Exp Immunol, 181, 323, 10.1111/cei.12609
Fanaro, 2003, Intestinal microflora in early infancy: composition and development, Acta Paediatr Suppl, 91, 48
Karlsson, 2011, The pioneer gut microbiota in human neonates vaginally born at term-a pilot study, Pediatr Res, 70, 282, 10.1203/PDR.0b013e318225f765
Perez, 2007, Bacterial imprinting of the neonatal immune system: lessons from maternal cells?, Pediatrics, 119, e724, 10.1542/peds.2006-1649
Satokari, 2009, Bifidobacterium and Lactobacillus DNA in the human placenta, Lett Appl Microbiol, 48, 8, 10.1111/j.1472-765X.2008.02475.x
Battersby, 2013, The gut mucosal immune system in the neonatal period, Pediatr Allergy Immunol, 24, 414, 10.1111/pai.12079
Mora, 2004, Role of L-selectin in the development of autoimmune diabetes in non-obese diabetic mice, Int Immunol, 16, 257, 10.1093/intimm/dxh036
Mullaney, 2018, Type 1 diabetes susceptibility alleles are associated with distinct alterations in the gut microbiota, Microbiome, 6, 35, 10.1186/s40168-018-0417-4
Round, 2009, The gut microbiota shapes intestinal immune responses during health and disease, Nat Rev Immunol, 9, 313, 10.1038/nri2515
Cerf-Bensussan, 2010, The immune system and the gut microbiota: friends or foes?, Nat Rev Immunol, 10, 735, 10.1038/nri2850
Brugman, 2006, Antibiotic treatment partially protects against type 1 diabetes in the Bio-Breeding diabetes-prone rat. Is the gut flora involved in the development of type 1 diabetes?, Diabetologia, 49, 2105, 10.1007/s00125-006-0334-0
Markle, 2014, Microbiome manipulation modifies sex-specific risk for autoimmunity, Gut microbes, 5, 485, 10.4161/gmic.29795
Alam, 2011, Effects of a germ-free environment on gut immune regulation and diabetes progression in non-obese diabetic (NOD) mice, Diabetologia, 54, 1398, 10.1007/s00125-011-2097-5
Gülden E, Chao C, Tai N, Pearson JA, Peng J, Majewska-Szczepanik M, et al. TRIF deficiency protects non-obese diabetic mice from type 1 diabetes by modulating the gut microbiota and dendritic cells. J Autoimmun 2018;93(undefined):57–65.
Hänninen, 2018, induces gut microbiota remodelling and controls islet autoimmunity in NOD mice, Gut, 67, 1445, 10.1136/gutjnl-2017-314508
Hansen, 2012, Early life treatment with vancomycin propagates Akkermansia muciniphila and reduces diabetes incidence in the NOD mouse, Diabetologia, 55, 2285, 10.1007/s00125-012-2564-7
Sofi, 2014, pH of drinking water influences the composition of gut microbiome and type 1 diabetes incidence, Diabetes, 63, 632, 10.2337/db13-0981
Wolf, 2014, Consumption of acidic water alters the gut microbiome and decreases the risk of diabetes in NOD mice, J Histochem Cytochem, 62, 237, 10.1369/0022155413519650
Giongo, 2011, Toward defining the autoimmune microbiome for type 1 diabetes, ISME J, 5, 82, 10.1038/ismej.2010.92
Brown, 2011, Gut microbiome metagenomics analysis suggests a functional model for the development of autoimmunity for type 1 diabetes, PLoS One, 6, 10.1371/journal.pone.0025792
Tormo-Badia, 2014, Antibiotic treatment of pregnant non-obese diabetic mice leads to altered gut microbiota and intestinal immunological changes in the offspring, Scand J Immunol, 80, 250, 10.1111/sji.12205
Sun, 2015, Pancreatic β-cells limit autoimmune diabetes via an immunoregulatory antimicrobial peptide expressed under the influence of the gut microbiota, Immunity, 43, 304, 10.1016/j.immuni.2015.07.013
Wen, 2017, Dietary short-chain fatty acids protect against type 1 diabetes, Nat Immunol, 18, 484, 10.1038/ni.3730
Mariño, 2017, Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes, Nat Immunol, 18, 552, 10.1038/ni.3713
Maynard, 2012, Reciprocal interactions of the intestinal microbiota and immune system, Nature, 489, 231, 10.1038/nature11551
Shen, 2012, Outer membrane vesicles of a human commensal mediate immune regulation and disease protection, Cell Host Microbe, 12, 509, 10.1016/j.chom.2012.08.004
Wen, 2008, Innate immunity and intestinal microbiota in the development of Type 1 diabetes, Nature, 455, 1109, 10.1038/nature07336
Alkanani, 2014, Induction of diabetes in the RIP-B7.1 mouse model is critically dependent on TLR3 and MyD88 pathways and is associated with alterations in the intestinal microbiome, Diabetes, 63, 619, 10.2337/db13-1007
Rouxel, 2017, Cytotoxic and regulatory roles of mucosal-associated invariant T cells in type 1 diabetes, Nat Immunol, 18, 1321, 10.1038/ni.3854
Kriegel, 2011, Naturally transmitted segmented filamentous bacteria segregate with diabetes protection in nonobese diabetic mice, Proc Natl Acad Sci U S A, 108, 11548, 10.1073/pnas.1108924108
Knip, 2017, Modulation of Type 1 Diabetes Risk by the Intestinal Microbiome, Curr Diab Rep, 17, 105, 10.1007/s11892-017-0933-9
Vatanen, 2018, The human gut microbiome in early-onset type 1 diabetes from the TEDDY study, Nature, 562, 589, 10.1038/s41586-018-0620-2
Lorscheider J, Buzzard K, Jokubaitis V, Spelman T, Havrdova E, Horakova D, et al. Defining secondary progressive multiple sclerosis. Brain 2016;139(null):2395–405.
Itoh, 2018, Cell-specific and region-specific transcriptomics in the multiple sclerosis model: Focus on astrocytes, Proc Natl Acad Sci U S A, 115, 10.1073/pnas.1716032115
Ben-Nun, 2014, From classic to spontaneous and humanized models of multiple sclerosis: impact on understanding pathogenesis and drug development, J Autoimmun, 54, 33, 10.1016/j.jaut.2014.06.004
Hindson, 2017, Multiple sclerosis: A possible link between multiple sclerosis and gut microbiota, Nat Rev Neurol, 13, 705, 10.1038/nrneurol.2017.142
Wekerle, 2015, Nature plus nurture: the triggering of multiple sclerosis, Swiss Med Wkly, 145, w14189
Cao, 2017, Long-term consumption of caffeine-free high sucrose cola beverages aggravates the pathogenesis of EAE in mice, Cell Discov, 3, 17020, 10.1038/celldisc.2017.20
Yadav, 2017, Gut dysbiosis breaks immunological tolerance toward the central nervous system during young adulthood, Proc Natl Acad Sci U S A, 114, 10.1073/pnas.1615715114
Cani, 2008, Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice, Diabetes, 57, 1470, 10.2337/db07-1403
Veldhoen, 2008, The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins, Nature, 453, 106, 10.1038/nature06881
Ochoa-Repáraz, 2018, The gut microbiome and multiple sclerosis, Cold Spring Harb Perspect Med, 8, 10.1101/cshperspect.a029017
Camara-Lemarroy, 2018, The intestinal barrier in multiple sclerosis: implications for pathophysiology and therapeutics, Brain, 141, 1900, 10.1093/brain/awy131
Zhu, 2017, Microbiota-gut-brain axis and the central nervous system, Oncotarget, 8, 53829, 10.18632/oncotarget.17754
Kamada, 2013, Role of the gut microbiota in immunity and inflammatory disease, Nat Rev Immunol, 13, 321, 10.1038/nri3430
Cekanaviciute, 2017, Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models, Proc Natl Acad Sci U S A, 114, 10713, 10.1073/pnas.1711235114
Berer, 2017, Gut microbiota from multiple sclerosis patients enables spontaneous autoimmune encephalomyelitis in mice, Proc Natl Acad Sci U S A, 114, 10719, 10.1073/pnas.1711233114
Cree, 2016, Gut microbiome analysis in neuromyelitis optica reveals overabundance of Clostridium perfringens, Ann Neurol, 80, 443, 10.1002/ana.24718
Jangi, 2016, Alterations of the human gut microbiome in multiple sclerosis, Nat Commun, 7, 12015, 10.1038/ncomms12015
Tremlett, 2016, Gut microbiota in early pediatric multiple sclerosis: a case-control study, Eur J Neurol, 23, 1308, 10.1111/ene.13026
Branton, 2016, Brain microbiota disruption within inflammatory demyelinating lesions in multiple sclerosis, Sci Rep, 6, 37344, 10.1038/srep37344
Chen, 2016, Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls, Sci Rep, 6, 28484, 10.1038/srep28484
Rothhammer, 2016, Type I interferons and microbial metabolites of tryptophan modulate astrocyte activity and central nervous system inflammation via the aryl hydrocarbon receptor, Nat Med, 22, 586, 10.1038/nm.4106
Berer, 2011, Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination, Nature, 479, 538, 10.1038/nature10554
Lee, 2011, Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis, Proc Natl Acad Sci U S A, 108, 4615, 10.1073/pnas.1000082107
Choi, 2016, A diet mimicking fasting promotes regeneration and reduces autoimmunity and multiple sclerosis symptoms, Cell Rep, 15, 2136, 10.1016/j.celrep.2016.05.009
Peruzzotti-Jametti L, Bernstock JD, Vicario N, Costa ASH, Kwok CK, Leonardi T, et al. Macrophage-derived extracellular succinate licenses neural stem cells to suppress chronic neuroinflammation. Cell Stem Cell 2018;22(3):355–68.e13.
Brandt, 2012, Long-term follow-up of colonoscopic fecal microbiota transplant for recurrent Clostridium difficile infection, Am J Gastroenterol, 107, 1079, 10.1038/ajg.2012.60
Bennet, 1989, Treatment of ulcerative colitis by implantation of normal colonic flora, The Lancet, 333, 164, 10.1016/S0140-6736(89)91183-5
Drew, 2016, Microbiota: reseeding the gut, Nature, 540, 10.1038/540S109a
Vermeire, 2016, Donor species richness determines faecal microbiota transplantation success in inflammatory bowel disease, J Crohns Colitis, 10, 387, 10.1093/ecco-jcc/jjv203
Angelberger, 2013, Temporal bacterial community dynamics vary among ulcerative colitis patients after fecal microbiota transplantation, Am J Gastroenterol, 108, 1620, 10.1038/ajg.2013.257
Kump, 2013, Alteration of intestinal dysbiosis by fecal microbiota transplantation does not induce remission in patients with chronic active ulcerative colitis, Inflamm Bowel Dis, 19, 2155, 10.1097/MIB.0b013e31829ea325
Kunde, 2013, Safety, tolerability, and clinical response after fecal transplantation in children and young adults with ulcerative colitis, J Pediatr Gastroenterol Nutr, 56, 597, 10.1097/MPG.0b013e318292fa0d
Suskind, 2015, Fecal microbial transplant via nasogastric tube for active pediatric ulcerative colitis, J Pediatr Gastroenterol Nutr, 60, 27, 10.1097/MPG.0000000000000544
Damman, 2015, Low level engraftment and improvement following a single colonoscopic administration of fecal microbiota to patients with ulcerative colitis, PloS one, 10, 10.1371/journal.pone.0133925
Cui, 2015, Step-up fecal microbiota transplantation strategy: a pilot study for steroid-dependent ulcerative colitis, J Transl Med, 13, 298, 10.1186/s12967-015-0646-2
Rossen, 2015, Findings from a randomized controlled trial of fecal transplantation for patients with ulcerative colitis, Gastroenterology, 149, 110, 10.1053/j.gastro.2015.03.045
Moayyedi, 2015, Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial, Gastroenterology, 149, 102, 10.1053/j.gastro.2015.04.001
Wei, 2016, Pectin enhances the effect of fecal microbiota transplantation in ulcerative colitis by delaying the loss of diversity of gut flora, BMC Microbiol, 16, 255, 10.1186/s12866-016-0869-2
Ishikawa, 2017, Changes in Intestinal Microbiota Following Combination Therapy with Fecal Microbial Transplantation and Antibiotics for Ulcerative Colitis, Inflamm Bowel Dis, 23, 116, 10.1097/MIB.0000000000000975
Nishida, 2017, Efficacy and safety of single fecal microbiota transplantation for Japanese patients with mild to moderately active ulcerative colitis, J Gastroenterol, 52, 476, 10.1007/s00535-016-1271-4
Paramsothy, 2017, Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial, Lancet, 389, 1218, 10.1016/S0140-6736(17)30182-4
Suskind, 2015, Fecal microbial transplant effect on clinical outcomes and fecal microbiome in active Crohn's disease, Inflamm Bowel Dis, 21, 556, 10.1097/MIB.0000000000000307
Cui, 2015, Fecal microbiota transplantation through mid-gut for refractory Crohn's disease: safety, feasibility, and efficacy trial results, J Gastroenterol Hepatol, 30, 51, 10.1111/jgh.12727
Vaughn, 2016, Increased Intestinal Microbial Diversity Following Fecal Microbiota Transplant for Active Crohn's Disease, Inflamm Bowel Dis, 22, 2182, 10.1097/MIB.0000000000000893
Costello, 2017, Systematic review with meta-analysis: faecal microbiota transplantation for the induction of remission for active ulcerative colitis, Aliment Pharmacol Ther, 46, 213, 10.1111/apt.14173
Qazi, 2017, The risk of inflammatory bowel disease flares after fecal microbiota transplantation: Systematic review and meta-analysis, Gut microbes, 8, 574, 10.1080/19490976.2017.1353848
Zhang, 2017, Cost-effectiveness analysis of fecal microbiota transplantation for inflammatory bowel disease, Oncotarget, 8, 88894, 10.18632/oncotarget.21491
Rossen, 2015, Fecal microbiota transplantation as novel therapy in gastroenterology: A systematic review, World J Gastroenterol, 21, 5359, 10.3748/wjg.v21.i17.5359
Colman, 2014, Fecal microbiota transplantation as therapy for inflammatory bowel disease: a systematic review and meta-analysis, J Crohns Colitis, 8, 1569, 10.1016/j.crohns.2014.08.006
Peng, 2014, Long term effect of gut microbiota transfer on diabetes development, J Autoimmun, 53, 85, 10.1016/j.jaut.2014.03.005
Bafeta, 2017, Methods and reporting studies assessing fecal microbiota transplantation: a systematic review, Ann Intern Med, 167, 34, 10.7326/M16-2810
Bojanova, 2016, Fecal Transplants: What Is Being Transferred?, PLoS Biol, 14, 10.1371/journal.pbio.1002503
Wekerle, 2017, Brain Autoimmunity and Intestinal Microbiota: 100 Trillion Game Changers, Trends Immunol, 38, 483, 10.1016/j.it.2017.03.008
Lee, 2016, Frozen vs fresh fecal microbiota transplantation and clinical resolution of diarrhea in patients with recurrent clostridium difficile Infection: a randomized clinical trial, JAMA, 315, 142, 10.1001/jama.2015.18098
Calvo-Barreiro, 2018, Combined therapies to treat complex diseases: The role of the gut microbiota in multiple sclerosis, Autoimmun Rev, 17, 165, 10.1016/j.autrev.2017.11.019
Olesen, 2018, Searching for superstool: maximizing the therapeutic potential of FMT, Nat Rev Gastroenterol Hepatol, 15, 387, 10.1038/s41575-018-0019-4
Hill, 2014, Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic, Nat Rev Gastroenterol Hepatol, 11, 506, 10.1038/nrgastro.2014.66
Kruis, 1997, Double-blind comparison of an oral Escherichia coli preparation and mesalazine in maintaining remission of ulcerative colitis, Aliment Pharmacol Ther, 11, 853, 10.1046/j.1365-2036.1997.00225.x
Rembacken, 1999, Non-pathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis: a randomised trial, Lancet, 354, 635, 10.1016/S0140-6736(98)06343-0
Kruis, 2004, Maintaining remission of ulcerative colitis with the probiotic Escherichia coli Nissle 1917 is as effective as with standard mesalazine, Gut, 53, 1617, 10.1136/gut.2003.037747
Matthes, 2010, Clinical trial: probiotic treatment of acute distal ulcerative colitis with rectally administered Escherichia coli Nissle 1917 (EcN), BMC Complement Altern Med, 10, 13, 10.1186/1472-6882-10-13
Petersen, 2014, Ciprofloxacin and probiotic Escherichia coli Nissle add-on treatment in active ulcerative colitis: a double-blind randomized placebo controlled clinical trial, J Crohns Colitis, 8, 1498, 10.1016/j.crohns.2014.06.001
Nagasaki, 2010, Ulcerative colitis with multidrug-resistant Pseudomonas aeruginosa infection successfully treated with bifidobacterium, Digestion, 81, 204, 10.1159/000236042
Kato, 2004, Randomized placebo-controlled trial assessing the effect of bifidobacteria-fermented milk on active ulcerative colitis, Aliment Pharmacol Ther, 20, 1133, 10.1111/j.1365-2036.2004.02268.x
Tamaki, 2016, Efficacy of probiotic treatment with Bifidobacterium longum 536 for induction of remission in active ulcerative colitis: A randomized, double-blinded, placebo-controlled multicenter trial, Dig Endosc, 28, 67, 10.1111/den.12553
Zocco, 2006, Efficacy of Lactobacillus GG in maintaining remission of ulcerative colitis, Aliment Pharmacol Ther, 23, 1567, 10.1111/j.1365-2036.2006.02927.x
Wildt, 2011, A randomised double-blind placebo-controlled trial with Lactobacillus acidophilus La-5 and Bifidobacterium animalis subsp. lactis BB-12 for maintenance of remission in ulcerative colitis, J Crohns Colitis, 5, 115, 10.1016/j.crohns.2010.11.004
Oliva, 2012, Randomised clinical trial: the effectiveness of Lactobacillus reuteri ATCC 55730 rectal enema in children with active distal ulcerative colitis, Aliment Pharmacol Ther, 35, 327, 10.1111/j.1365-2036.2011.04939.x
Prantera, 2002, Ineffectiveness of probiotics in preventing recurrence after curative resection for Crohn's disease: a randomised controlled trial with Lactobacillus GG, Gut, 51, 405, 10.1136/gut.51.3.405
Marteau, 2006, Ineffectiveness of Lactobacillus johnsonii LA1 for prophylaxis of postoperative recurrence in Crohn's disease: a randomised, double blind, placebo controlled GETAID trial, Gut, 55, 842, 10.1136/gut.2005.076604
Van Gossum, 2007, Multicenter randomized-controlled clinical trial of probiotics (Lactobacillus johnsonii, LA1) on early endoscopic recurrence of Crohn's disease after lleo-caecal resection, Inflamm Bowel Dis, 13, 135, 10.1002/ibd.20063
Garcia Vilela, 2008, Influence of Saccharomyces boulardii on the intestinal permeability of patients with Crohn's disease in remission, Scand J Gastroenterol, 43, 842, 10.1080/00365520801943354
Bourreille, 2013, Saccharomyces boulardii does not prevent relapse of Crohn's disease, Clin Gastroenterol Hepatol, 11, 982, 10.1016/j.cgh.2013.02.021
Tsuda, 2007, Clinical effectiveness of probiotics therapy (BIO-THREE) in patients with ulcerative colitis refractory to conventional therapy, Scand J Gastroenterol, 42, 1306, 10.1080/00365520701396091
Yoshimatsu, 2015, Effectiveness of probiotic therapy for the prevention of relapse in patients with inactive ulcerative colitis, World J Gastroenterol, 21, 5985, 10.3748/wjg.v21.i19.5985
Venturi, 1999, Impact on the composition of the faecal flora by a new probiotic preparation: preliminary data on maintenance treatment of patients with ulcerative colitis, Aliment Pharmacol Ther, 13, 1103, 10.1046/j.1365-2036.1999.00560.x
Mimura, 2004, Once daily high dose probiotic therapy (VSL#3) for maintaining remission in recurrent or refractory pouchitis, Gut, 53, 108, 10.1136/gut.53.1.108
Bibiloni, 2005, VSL#3 probiotic-mixture induces remission in patients with active ulcerative colitis, Am J Gastroenterol, 100, 1539, 10.1111/j.1572-0241.2005.41794.x
Pronio, 2008, Probiotic administration in patients with ileal pouch-anal anastomosis for ulcerative colitis is associated with expansion of mucosal regulatory cells, Inflamm Bowel Dis, 14, 662, 10.1002/ibd.20369
Miele, 2009, Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis, Am J Gastroenterol, 104, 437, 10.1038/ajg.2008.118
Sood, 2009, The probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately active ulcerative colitis, Clin Gastroenterol Hepatol, 7, 1202, 10.1016/j.cgh.2009.07.016
Ng, 2010, Immunosuppressive effects via human intestinal dendritic cells of probiotic bacteria and steroids in the treatment of acute ulcerative colitis, Inflamm Bowel Dis, 16, 1286, 10.1002/ibd.21222
Tursi, 2010, Treatment of relapsing mild-to-moderate ulcerative colitis with the probiotic VSL#3 as adjunctive to a standard pharmaceutical treatment: a double-blind, randomized, placebo-controlled study, Am J Gastroenterol, 105, 2218, 10.1038/ajg.2010.218
Fedorak, 2015, The probiotic VSL#3 has anti-inflammatory effects and could reduce endoscopic recurrence after surgery for Crohn's disease, Clin Gastroenterol Hepatol, 13, 928, 10.1016/j.cgh.2014.10.031
Ishikawa, 2003, Randomized controlled trial of the effect of bifidobacteria-fermented milk on ulcerative colitis, J Am Coll Nutr, 22, 56, 10.1080/07315724.2003.10719276
Hughes, 2017, Bifidobacterium breve reduces apoptotic epithelial cell shedding in an exopolysaccharide and MyD88-dependent manner, Open Biol, 7, 10.1098/rsob.160155
Seo, 2012, Construction of recombinant E. coli Nissle 1917 (EcN) strains for the expression and secretion of defensins, Int J Med Microbiol, 302, 276, 10.1016/j.ijmm.2012.05.002
Hering, 2014, TcpC protein from E. coli Nissle improves epithelial barrier function involving PKCζ and ERK1/2 signaling in HT-29/B6 cells, Mucosal Immunol, 7, 369, 10.1038/mi.2013.55
Liu, 2016, Anti-inflammatory effects of Lactobacillus brevis K65 on RAW 264.7 cells and in mice with dextran sulphate sodium-induced ulcerative colitis, Benef Microbes, 7, 387, 10.3920/BM2015.0109
Chapman, 2006, VSL#3 probiotic mixture: a review of its use in chronic inflammatory bowel diseases, Drugs, 66, 1371, 10.2165/00003495-200666100-00006
Guslandi, 2000, Saccharomyces boulardii in maintenance treatment of Crohn's disease, Dig Dis Sci, 45, 1462, 10.1023/A:1005588911207
Shen, 2014, Effect of probiotics on inducing remission and maintaining therapy in ulcerative colitis, Crohn's disease, and pouchitis: meta-analysis of randomized controlled trials, Inflamm Bowel Dis, 20, 21, 10.1097/01.MIB.0000437495.30052.be
Ganji-Arjenaki, 2018, Probiotics are a good choice in remission of inflammatory bowel diseases: A meta analysis and systematic review, J Cell Physiol, 233, 2091, 10.1002/jcp.25911
Derwa, 2017, Systematic review with meta-analysis: the efficacy of probiotics in inflammatory bowel disease, Aliment Pharmacol Ther, 46, 389, 10.1111/apt.14203
Mardini, 2014, Probiotic mix VSL#3 is effective adjunctive therapy for mild to moderately active ulcerative colitis: a meta-analysis, Inflamm Bowel Dis, 20, 1562, 10.1097/MIB.0000000000000084
Hudan Pan, 2017, Whether Probiotic Supplementation Benefits Rheumatoid Arthritis Patients: A Systematic Review and Meta-Analysis, Engineering, 3, 115, 10.1016/J.ENG.2017.01.006
Lavasani, 2010, A novel probiotic mixture exerts a therapeutic effect on experimental autoimmune encephalomyelitis mediated by IL-10 producing regulatory T cells, PloS one, 5, 10.1371/journal.pone.0009009
Devine, 2015, Modulation of host responses by oral commensal bacteria, J Oral Microbiol, 7, 26941, 10.3402/jom.v7.26941
Timmerman, 2004, Monostrain, multistrain and multispecies probiotics--A comparison of functionality and efficacy, Int J Food Microbiol, 96, 219, 10.1016/j.ijfoodmicro.2004.05.012
Plaza-Diaz, 2014, Modulation of immunity and inflammatory gene expression in the gut, in inflammatory diseases of the gut and in the liver by probiotics, World J Gastroenterol, 20, 15632, 10.3748/wjg.v20.i42.15632
Gibson, 2017, Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics, Nat Rev Gastroenterol Hepatol, 14, 491, 10.1038/nrgastro.2017.75
Hafer A, Krämer S, Duncker S, Krüger M, Manns MP, Bischoff SC. Effect of oral lactulose on clinical and immunohistochemical parameters in patients with inflammatory bowel disease: a pilot study. BMC Gastroenterol 2007;7(undefined):36.
Ishikawa, 2011, Beneficial effects of probiotic bifidobacterium and galacto-oligosaccharide in patients with ulcerative colitis: a randomized controlled study, Digestion, 84, 128, 10.1159/000322977
Benjamin, 2011, Randomised, double-blind, placebo-controlled trial of fructo-oligosaccharides in active Crohn's disease, Gut, 60, 923, 10.1136/gut.2010.232025
Furrie, 2005, Synbiotic therapy (Bifidobacterium longum/Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: a randomised controlled pilot trial, Gut, 54, 242, 10.1136/gut.2004.044834
Chermesh, 2007, Failure of Synbiotic 2000 to prevent postoperative recurrence of Crohn's disease, Dig Dis Sci, 52, 385, 10.1007/s10620-006-9549-7
Gibson, 1995, Selective stimulation of bifidobacteria in the human colon by oligofructose and inulin, Gastroenterology, 108, 975, 10.1016/0016-5085(95)90192-2
Rastall, 2015, Recent developments in prebiotics to selectively impact beneficial microbes and promote intestinal health, Curr Opin Biotechnol, 32, 42, 10.1016/j.copbio.2014.11.002
Fujimori, 2009, A randomized controlled trial on the efficacy of synbiotic versus probiotic or prebiotic treatment to improve the quality of life in patients with ulcerative colitis, Nutrition, 25, 520, 10.1016/j.nut.2008.11.017
Waldman, 2018, Guanylate cyclase-C as a therapeutic target in gastrointestinal disorders, Gut, 67, 1543, 10.1136/gutjnl-2018-316029
Cirstea, 2018, Good Bug, Bad Bug: Breaking through Microbial Stereotypes, Cell Host Microbe, 23, 10, 10.1016/j.chom.2017.12.008
Walsh, 2018, Drug-gut microbiota interactions: implications for neuropharmacology, Br J Pharmacol, 175, 4415, 10.1111/bph.14366
Maier, 2018, Extensive impact of non-antibiotic drugs on human gut bacteria, Nature, 555, 623, 10.1038/nature25979