Defined microbiota transplant restores Th17/RORγt+ regulatory T cell balance in mice colonized with inflammatory bowel disease microbiotas

Significance Composition of gut microbiota is altered in many human diseases, including inflammatory bowel disease. Some hope that restoring microbiota to a healthy state could help treat such diseases. We have used mice colonized with microbiotas from humans with inflammatory bowel disease to study what happens when these mice receive a microbiota transplant from a set of healthy humans. We find that the mouse gut immune system is changed by microbiota transplants, becoming broadly less inflammatory and protecting mice from colitis. By culturing bacteria from these microbiotas, we identify one strain that induces inflammatory responses in mice and show that it is modified by microbiota transplant. We also show that increases in the density of microbiota following transplant may be antiinflammatory. The building evidence for the contribution of microbiota to human disease has spurred an effort to develop therapies that target the gut microbiota. This is particularly evident in inflammatory bowel diseases (IBDs), where clinical trials of fecal microbiota transplantation have shown some efficacy. To aid the development of novel microbiota-targeted therapies and to better understand the biology underpinning such treatments, we have used gnotobiotic mice to model microbiota manipulations in the context of microbiotas from humans with inflammatory bowel disease. Mice colonized with IBD donor-derived microbiotas exhibit a stereotypical set of phenotypes, characterized by abundant mucosal Th17 cells, a deficit in the tolerogenic RORγt+ regulatory T (Treg) cell subset, and susceptibility to disease in colitis models. Transplanting healthy donor-derived microbiotas into mice colonized with human IBD microbiotas led to induction of RORγt+ Treg cells, which was associated with an increase in the density of the microbiotas following transplant. Microbiota transplant reduced gut Th17 cells in mice colonized with a microbiota from a donor with Crohn’s disease. By culturing strains from this microbiota and screening them in vivo, we identified a specific strain that potently induces Th17 cells. Microbiota transplants reduced the relative abundance of this strain in the gut microbiota, which was correlated with a reduction in Th17 cells and protection from colitis.

[1]  Jinfang Zhu Faculty Opinions recommendation of Microbiotas from Humans with Inflammatory Bowel Disease Alter the Balance of Gut Th17 and RORγt+ Regulatory T Cells and Exacerbate Colitis in Mice. , 2021, Faculty Opinions – Post-Publication Peer Review of the Biomedical Literature.

[2]  I. Barshack,et al.  Abstract CT042: Fecal microbiota transplantation (FMT) and re-induction of anti-PD-1 therapy in refractory metastatic melanoma patients - preliminary results from a phase I clinical trial (NCT03353402) , 2019, Clinical Trials.

[3]  D. Haller,et al.  Complex Bacterial Consortia Reprogram the Colitogenic Activity of Enterococcus faecalis in a Gnotobiotic Mouse Model of Chronic, Immune-Mediated Colitis , 2019, Front. Immunol..

[4]  J. Raes,et al.  Quantitative microbiome profiling disentangles inflammation- and bile duct obstruction-associated microbiota alterations across PSC/IBD diagnoses , 2019, Nature Microbiology.

[5]  Louis J. Cohen,et al.  Genetic Factors and the Intestinal Microbiome Guide Development of Microbe-Based Therapies for Inflammatory Bowel Diseases. , 2019, Gastroenterology.

[6]  J. Faith,et al.  Strain-level differences in gut microbiome composition determine fecal IgA levels and are modifiable by gut microbiota manipulation , 2019, bioRxiv.

[7]  E. Kuijper,et al.  Clinical Application and Potential of Fecal Microbiota Transplantation. , 2019, Annual review of medicine.

[8]  D. Gevers,et al.  Microbiotas from Humans with Inflammatory Bowel Disease Alter the Balance of Gut Th17 and ROR&ggr;t+ Regulatory T Cells and Exacerbate Colitis in Mice , 2019, Immunity.

[9]  J. Andrews,et al.  Effect of Fecal Microbiota Transplantation on 8-Week Remission in Patients With Ulcerative Colitis: A Randomized Clinical Trial , 2019, JAMA.

[10]  B. Helmink,et al.  Fecal microbiota transplantation for refractory immune checkpoint inhibitor-associated colitis , 2018, Nature Medicine.

[11]  S. Lynch,et al.  The gut microbiome: Relationships with disease and opportunities for therapy , 2018, The Journal of experimental medicine.

[12]  M. Defernez,et al.  Microbial taxonomic and metabolic alterations during faecal microbiota transplantation to treat Clostridium difficile infection. , 2018, The Journal of infection.

[13]  Judy H. Cho,et al.  Gut microbiota density influences host physiology and is shaped by host and microbial factors , 2018, bioRxiv.

[14]  Richard Bonneau,et al.  c-Maf-dependent regulatory T cells mediate immunological tolerance to a gut pathobiont , 2018, Nature.

[15]  Jun Wang,et al.  Quantitative microbiome profiling links gut community variation to microbial load , 2017, Nature.

[16]  J. Segre,et al.  Ectopic colonization of oral bacteria in the intestine drives TH1 cell induction and inflammation , 2017, Science.

[17]  A. Sivignon,et al.  Bacteriophages Targeting Adherent Invasive Escherichia coli Strains as a Promising New Treatment for Crohn’s Disease , 2017, Journal of Crohn's & colitis.

[18]  N. Ajami,et al.  Single Delivery of High-Diversity Fecal Microbiota Preparation by Colonoscopy Is Safe and Effective in Increasing Microbial Diversity in Active Ulcerative Colitis , 2017, Inflammatory bowel diseases.

[19]  Sophie J. Weiss,et al.  Changes in microbial ecology after fecal microbiota transplantation for recurrent C. difficile infection affected by underlying inflammatory bowel disease , 2017, Microbiome.

[20]  M. Kamm,et al.  Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: a randomised placebo-controlled trial , 2017, The Lancet.

[21]  C. Benoist,et al.  Mining the Human Gut Microbiota for Immunomodulatory Organisms , 2017, Cell.

[22]  N. Ajami,et al.  IgA-coated E. coli enriched in Crohn’s disease spondyloarthritis promote TH17-dependent inflammation , 2017, Science Translational Medicine.

[23]  C. Benoist,et al.  Identifying species of symbiont bacteria from the human gut that, alone, can induce intestinal Th17 cells in mice , 2016, Proceedings of the National Academy of Sciences.

[24]  T. Vatanen,et al.  Increased Intestinal Microbial Diversity Following Fecal Microbiota Transplant for Active Crohn's Disease , 2016, Inflammatory bowel diseases.

[25]  Lu Wei,et al.  Short-Term Surveillance of Cytokines and C-Reactive Protein Cannot Predict Efficacy of Fecal Microbiota Transplantation for Ulcerative Colitis , 2016, PloS one.

[26]  U. Panzer,et al.  RORγt(+)Foxp3(+) Cells are an Independent Bifunctional Regulatory T Cell Lineage and Mediate Crescentic GN. , 2016, Journal of the American Society of Nephrology : JASN.

[27]  M. Hattori,et al.  Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells , 2015, Cell.

[28]  Liza Konnikova,et al.  Individual intestinal symbionts induce a distinct population of RORγ+ regulatory T cells , 2015, Science.

[29]  D. Voehringer,et al.  The microbiota regulates type 2 immunity through RORγt+ T cells , 2015, Science.

[30]  P. Oliver Faculty Opinions recommendation of MUCOSAL IMMUNOLOGY. The microbiota regulates type 2 immunity through RORγt⁺ T cells. , 2015 .

[31]  M. Beckstette,et al.  Foxp3+ T cells expressing RORγt represent a stable regulatory T-cell effector lineage with enhanced suppressive capacity during intestinal inflammation , 2015, Mucosal Immunology.

[32]  E. Zoetendal,et al.  Findings From a Randomized Controlled Trial of Fecal Transplantation for Patients With Ulcerative Colitis. , 2015, Gastroenterology.

[33]  M. Surette,et al.  Fecal Microbiota Transplantation Induces Remission in Patients With Active Ulcerative Colitis in a Randomized Controlled Trial. , 2015, Gastroenterology.

[34]  A. Khoruts,et al.  Species and genus level resolution analysis of gut microbiota in Clostridium difficile patients following fecal microbiota transplantation , 2014, Microbiome.

[35]  Jonathan L. Linehan,et al.  Focused Specificity of Intestinal Th17 Cells towards Commensal Bacterial Antigens , 2014, Nature.

[36]  M. Hattori,et al.  Characterization of the 17 strains of regulatory T cell-inducing human-derived Clostridia , 2014, Gut microbes.

[37]  J. Faith,et al.  Identifying Gut Microbe–Host Phenotype Relationships Using Combinatorial Communities in Gnotobiotic Mice , 2014, Science Translational Medicine.

[38]  Shashank Garg,et al.  Microbiota Dynamics in Patients Treated with Fecal Microbiota Transplantation for Recurrent Clostridium difficile Infection , 2013, PloS one.

[39]  J. Gordon,et al.  Gnotobiotic mouse model of phage–bacterial host dynamics in the human gut , 2013, Proceedings of the National Academy of Sciences.

[40]  Bernard Henrissat,et al.  Effects of Diet on Resource Utilization by a Model Human Gut Microbiota Containing Bacteroides cellulosilyticus WH2, a Symbiont with an Extensive Glycobiome , 2013, PLoS biology.

[41]  M. Hattori,et al.  Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota , 2013, Nature.

[42]  E. Zoetendal,et al.  Duodenal infusion of donor feces for recurrent Clostridium difficile. , 2013, The New England journal of medicine.

[43]  T. Borody,et al.  Fecal microbiota transplantation: techniques, applications, and issues. , 2012, Gastroenterology clinics of North America.

[44]  J. Kolls,et al.  Targeting IL-17 and TH17 cells in chronic inflammation , 2012, Nature Reviews Drug Discovery.

[45]  Timothy L. Tickle,et al.  Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment , 2012, Genome Biology.

[46]  K. Honda,et al.  Induction of Colonic Regulatory T Cells by Indigenous Clostridium Species , 2011, Science.

[47]  Dan R. Littman,et al.  Induction of Intestinal Th17 Cells by Segmented Filamentous Bacteria , 2009, Cell.

[48]  J. Doré,et al.  Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients , 2008, Proceedings of the National Academy of Sciences.

[49]  Martin Wiedmann,et al.  Culture independent analysis of ileal mucosa reveals a selective increase in invasive Escherichia coli of novel phylogeny relative to depletion of Clostridiales in Crohn's disease involving the ileum , 2007, The ISME Journal.

[50]  N. Pace,et al.  Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases , 2007, Proceedings of the National Academy of Sciences.

[51]  C. Manichanh,et al.  Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach , 2005, Gut.

[52]  Laurent Beaugerie,et al.  High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn's disease. , 2004, Gastroenterology.

[53]  G. Lichtenstein,et al.  Fecal Microbiota Transplantation , 2019, Clinics in Colon and Rectal Surgery.

[54]  S. Hampson Randomised, placebo-controlled trial , 2002 .

[55]  E. Glaser The randomized clinical trial. , 1972, The New England journal of medicine.