1,646
Views
0
CrossRef citations to date
0
Altmetric
Research Paper

Microbiota and mucosal gene expression of fecal microbiota transplantation or placebo treated patients with chronic pouchitis

ORCID Icon, , , , , , & show all
Article: 2295445 | Received 18 Aug 2023, Accepted 12 Dec 2023, Published online: 12 Jan 2024

References

  • Kamada N, Seo S-U, Chen GY, Núñez G. Role of the gut microbiota in immunity and inflammatory disease. Nat Rev Immunol. 2013;13(5):321–20. doi:10.1038/nri3430.
  • Lin L, Zhang J. Role of intestinal microbiota and metabolites on gut homeostasis and human diseases. BMC Immunol. 2017;18(2):1–25. doi:10.1186/s12865-016-0187-3.
  • Chen Y, Cui W, Li X, Yang H. Interaction between commensal bacteria, immune response and the intestinal barrier in inflammatory bowel disease. Front Immunol. 2021;12:761981. doi:10.3389/fimmu.2021.761981.
  • Biondi A, Zoccali M, Costa S, Troci A, Contessini-Avesani E, Fichera A. Surgical treatment of ulcerative colitis in the biologic therapy era. WJG. 2012;18(16):1861–1870. doi:10.3748/wjg.v18.i16.1861.
  • Lightner AL, Mathis KL, Dozois EJ, Hahnsloser D, Loftus EVJ, Raffals LE, Pemberton JH. Results at up to 30 years after ileal pouch–anal anastomosis for chronic ulcerative colitis. Inflamm Bowel Dis. 2017;23(5):781–790. doi:10.1097/MIB.0000000000001061.
  • Kayal M, Plietz M, Rizvi A, Radcliffe M, Riggs A, Yzet C, Tixier E, Trivedi P, Ungaro RC, Khaitov S, et al. Inflammatory pouch conditions are common after ileal pouch anal anastomosis in ulcerative colitis patients. Inflamm Bowel Dis. 2020;26(7):1079–1086. doi:10.1093/ibd/izz227.
  • Bresteau C, Amiot A, Kirchgesner J, de’Angelis N, Lefevre JH, Bouhnik Y, Panis Y, Beaugerie L, Allez M, Brouquet A, et al. Chronic pouchitis and Crohn’s disease of the pouch after ileal pouch-anal anastomosis: incidence and risk factors. Digestive Liver Dis. 2021;53(9):1128–1135. doi:10.1016/j.dld.2021.03.027.
  • Shen B. Pouchitis: what every gastroenterologist needs to know. Clin Gastroenterol Hepatol. 2013;11(12):1538–1549. doi:10.1016/j.cgh.2013.03.033.
  • Segal JP, Ding NS, Worley G, Mclaughlin S, Preston S, Faiz OD, Clark SK, Hart AL. Systematic review with meta-analysis: the management of chronic refractory pouchitis with an evidence-based treatment algorithm. Aliment Pharmacol Ther. 2017;45(5):581–592. doi:10.1111/apt.13905.
  • Kousgaard SJ, Michaelsen TY, Nielsen HL, Kirk KF, Albertsen M, Thorlacius-Ussing O. The Microbiota Profile in Inflamed and Non-Inflamed Ileal Pouch–Anal Anastomosis. Microorganisms. 2020;8(10):1611. doi:10.3390/microorganisms8101611.
  • Palmieri O, Castellana S, Biscaglia G, Panza A, Latiano A, Fontana R, Guerra M, Corritore G, Latiano T, Martino G, et al. Microbiome analysis of mucosal ileoanal pouch in ulcerative colitis patients revealed impairment of the pouches immunometabolites. Cells. 2021;10(11):3243. doi:10.3390/cells10113243.
  • Segal JP, Oke S, Hold GL, Clark SK, Faiz OD, Hart AL. Systematic review: ileoanal pouch microbiota in health and disease. Aliment Pharmacol Ther. 2018;47(4):466–477. doi:10.1111/apt.14454.
  • Sriranganathan D, Kilic Y, Nabil Quraishi M, Segal JP. Prevalence of pouchitis in both ulcerative colitis and familial adenomatous polyposis: a systematic review and meta‐analysis. Colorectal Dis. 2021;24(1):27–39. doi:10.1111/codi.15995.
  • Huang Y, Dalal S, Antonopoulos D, Hubert N, Raffals LH, Dolan K, Weber C, Messer JS, Jabri B, Bendelac A, et al. Early transcriptomic changes in the ileal pouch provide insight into the molecular pathogenesis of pouchitis and ulcerative colitis. Inflamm Bowel Dis. 2017;23(3):366–378. doi:10.1097/MIB.0000000000001027.
  • LeBlanc J-F, Segal JP, de Campos Braz LM, Hart AL. The microbiome as a therapy in pouchitis and ulcerative colitis. Nutrients. 2021;13(6):1780. doi:10.3390/nu13061780.
  • Mimura T, Rizzello F, Helwig U, Poggioli G, Schreiber S, Talbot IC, Nicholls RJ, Gionchetti P, Campieri M, Kamm MA. Once daily high dose probiotic therapy (VSL#3) for maintaining remission in recurrent or refractory pouchitis. Gut. 2004;53(1):108–114. doi:10.1136/gut.53.1.108.
  • van Nood E, Vrieze A, Nieuwdorp M, Fuentes S, Zoetendal EG, de Vos WM, Visser CE, Kuijper EJ, Bartelsman JFWM, Tijssen JGP, et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med. 2013;368(5):407–415. doi:10.1056/NEJMoa1205037.
  • Mattila E, Uusitalo–Seppälä R, Wuorela M, Lehtola L, Nurmi H, Ristikankare M, Moilanen V, Salminen K, Seppälä M, Mattila PS, et al. Fecal transplantation, through colonoscopy, is effective therapy for recurrent Clostridium difficile infection. Gastroenterology. 2012;142(3):490–496. doi:10.1053/j.gastro.2011.11.037.
  • Khoruts A, Sadowsky MJ. Understanding the mechanisms of faecal microbiota transplantation. Nat Rev Gastroenterol Hepatol. 2016;13(9):508–516. doi:10.1038/nrgastro.2016.98.
  • Mocanu V, Rajaruban S, Dang J, Kung JY, Deehan EC, Madsen KL. Repeated fecal microbial transplantations and antibiotic pre-treatment are linked to improved clinical response and remission in inflammatory bowel disease: a systematic review and pooled proportion meta-analysis. JCM. 2021;10(5):959. doi:10.3390/jcm10050959.
  • Karjalainen EK, Renkonen-Sinisalo L, Satokari R, Mustonen H, Ristimäki A, Arkkila P, Lepistö AH. Fecal microbiota transplantation in chronic pouchitis: a randomized, parallel, double-blinded clinical trial. Inflamm Bowel Dis. 2021;27(11):1766–1772. doi:10.1093/ibd/izab001.
  • Cammarota G, Ianiro G, Tilg H, Rajilić-Stojanović M, Kump P, Satokari R, Sokol H, Arkkila P, Pintus C, Hart A, et al. European consensus conference on faecal microbiota transplantation in clinical practice. Gut. 2017;66(4):569–580. doi:10.1136/gutjnl-2016-313017.
  • Satokari R, Mattila E, Kainulainen V, Arkkila PET. Simple faecal preparation and efficacy of frozen inoculum in faecal microbiota transplantation for recurrent Clostridium difficile infection - an observational cohort study. Aliment Pharmacol Ther. 2015;41(1):46–53. doi:10.1111/apt.13009.
  • Costea PI, Zeller G, Sunagawa S, Pelletier E, Alberti A, Levenez F, Tramontano M, Driessen M, Hercog R, Jung F-E, et al. Towards standards for human fecal sample processing in metagenomic studies. Nat Biotechnol. 2017;35(11):1069–1076. doi:10.1038/nbt.3960.
  • Salonen A, Nikkilä J, Jalanka-Tuovinen J, Immonen O, Rajilić-Stojanović M, Kekkonen RA, Palva A, de Vos WM. Comparative analysis of fecal DNA extraction methods with phylogenetic microarray: effective recovery of bacterial and archaeal DNA using mechanical cell lysis. J Microbiol Methods. 2010;81(2):127–134. doi:10.1016/j.mimet.2010.02.007.
  • Kalliomäki M, Satokari R, Lähteenoja H, Vähämiko S, Grönlund J, Routi T, Salminen S. Expression of microbiota, toll-like receptors, and their regulators in the small intestinal mucosa in celiac disease. J Pediatr Gastroenterol Nutr. 2012;54(6):727–732. doi:10.1097/MPG.0b013e318241cfa8.
  • Klindworth A, Pruesse E, Schweer T, Peplies J, Quast C, Horn M, Glöckner FO. Evaluation of general 16S ribosomal RNA gene PCR primers for classical and next-generation sequencing-based diversity studies. Nucleic Acids Res. 2013;41(1):e1. doi:10.1093/nar/gks808.
  • Korpela K. Mare: microbiota analysis in R easily. R package version 1.0. 2016. doi:10.5281/zenodo.50310.
  • R Core Team. R: a language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing; 2021. https://www.R-project.org/.
  • Edgar RC. Search and clustering orders of magnitude faster than BLAST. Bioinformatics. 2010;26(19):2460–2461. doi:10.1093/bioinformatics/btq461.
  • Cole JR, Wang Q, Fish JA, Chai B, McGarrell DM, Sun Y, Brown CT, Porras-Alfaro A, Kuske CR, Tiedje JM. Ribosomal database project: data and tools for high throughput rRNA analysis. Nucl Acids Res. 2014;42(D1):D633–D642. doi:10.1093/nar/gkt1244.
  • Oksanen J, Blanchet FG, Friendly M, Kindt R, Legendre P, McGlinn D, Minchin PR, O’Hara RB, Simpson GL, Solymos P, et al. Vegan: community ecology package. R package version 2.5-7. 2020. https://CRAN.R-project.org/package=vegan
  • Venables W, Ripley B. Modern applied statistics with S. 4th ed. New York: Springer; 2002.
  • Pinheiro J, Bates D, DebRoy S, Sarkar D. nlme: Linear and Nonlinear Mixed Effects Models. R package version 3.1-155. 2022. https://CRAN.R-project.org/package=nlme
  • Wickham H. ggplot2: elegant graphics for data analysis. New York: Springer-Verlag; 2016. https://ggplot2.tidyverse.org.
  • Freitag TL, Podojil JR, Pearson RM, Fokta FJ, Sahl C, Messing M, Andersson LC, Leskinen K, Saavalainen P, Hoover LI, et al. Gliadin nanoparticles induce immune tolerance to gliadin in mouse models of celiac disease. Gastroenterology. 2020;158(6):1667–1681. doi:10.1053/j.gastro.2020.01.045.
  • Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, Tirosh I, Bialas AR, Kamitaki N, Martersteck EM, et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell. 2015;161(5):1202–1214. doi:10.1016/j.cell.2015.05.002.
  • Robinson MD, McCarthy DJ, Smyth GK. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–140. doi:10.1093/bioinformatics/btp616.
  • Krämer A, Green J, Pollard J Jr, Tugendreich S. Causal analysis approaches in ingenuity pathway analysis. Bioinformatics. 2014;30(4):523–530. doi:10.1093/bioinformatics/btt703.
  • Benjamini Y, Hochberg Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J R Stat Soc. 1995;57(1):289–300. doi:10.1111/j.2517-6161.1995.tb02031.x.
  • Landy J, Walker AW, Li JV, Al-Hassi HO, Ronde E, English NR, Mann ER, Bernardo D, McLaughlin SD, Parkhill J, et al. Variable alterations of the microbiota, without metabolic or immunological change, following faecal microbiota transplantation in patients with chronic pouchitis. Sci Rep. 2015;5(1):12955. doi:10.1038/srep12955.
  • Selvig D, Piceno Y, Terdiman J, Zydek M, Umetsu SE, Balitzer D, Fadrosh D, Lynch K, Lamere B, Leith T, et al. Fecal microbiota transplantation in pouchitis: clinical, endoscopic, histologic, and microbiota results from a pilot study. Dig Dis Sci. 2020;65(4):1099–1106. doi:10.1007/s10620-019-05715-2.
  • Stallmach A, Lange K, Buening J, Sina C, Vital M, Pieper DH. Fecal microbiota transfer in patients with chronic antibiotic-refractory pouchitis. Am J Gastroenterol. 2016;111(3):441–443. doi:10.1038/ajg.2015.436.
  • Angriman I, Scarpa M, Castagliuolo I. Relationship between pouch microbiota and pouchitis following restorative proctocolectomy for ulcerative colitis. WJG. 2014;20(29):9665–9674. doi:10.3748/wjg.v20.i29.9665.
  • Nishida A, Inoue R, Inatomi O, Bamba S, Naito Y, Andoh A. Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol. 2018;11(1):1–10. doi:10.1007/s12328-017-0813-5.
  • Guo X, Huang C, Xu J, Xu H, Liu L, Zhao H, Wang J, Huang W, Peng W, Chen Y, et al. Gut microbiota is a potential biomarker in inflammatory bowel disease. Front Nutr. 2022;8:818902. doi:10.3389/fnut.2021.818902.
  • Dubinsky V, Reshef L, Rabinowitz K, Yadgar K, Godny L, Zonensain K, Wasserberg N, Dotan I, Gophna U. Dysbiosis in metabolic genes of the gut microbiomes of patients with an ileo-anal pouch resembles that observed in Crohn’s disease. mSystems. 2021;6(2):e00984–20. doi:10.1128/mSystems.00984-20.
  • Morgan XC, Kabakchiev B, Waldron L, Tyler AD, Tickle TL, Milgrom R, Stempak JM, Gevers D, Xavier RJ, Silverberg MS, et al. Associations between host gene expression, the mucosal microbiome, and clinical outcome in the pelvic pouch of patients with inflammatory bowel disease. Genome Biol. 2015;16(1):67. doi:10.1186/s13059-015-0637-x.
  • Li J, Li Y, Zhou Y, Wang C, Wu B, Wan J. Actinomyces and alimentary tract diseases: a review of its biological functions and pathology. Biomed Res Int. 2018;2018:1–8. doi:10.1155/2018/3820215.
  • Martinez C, Antolin M, Santos J, Torrejon A, Casellas F, Borruel N, Guarner F, Malagelada J-R. Unstable composition of the fecal microbiota in ulcerative colitis during clinical remission. Am J Gastroenterol. 2008;103(3):643–648. doi:10.1111/j.1572-0241.2007.01592.x.
  • Herfarth H, Barnes EL, Long MD, Isaacs KL, Leith T, Silverstein M, Gerardin Y, Kassam Z. Combined endoscopic and oral fecal microbiota transplantation in patients with antibiotic-dependent pouchitis: low clinical efficacy due to low donor microbial engraftment. Inflamm Intest Dis. 2019;4(1):1–6. doi:10.1159/000497042.
  • Kousgaard SJ, Michaelsen TY, Nielsen HL, Kirk KF, Brandt J, Albertsen M, Thorlacius-Ussing O. Clinical results and microbiota changes after faecal microbiota transplantation for chronic pouchitis: a pilot study. Scand J Gastroenterol. 2020;55(4):421–429. doi:10.1080/00365521.2020.1748221.
  • Rigottier-Gois L. Dysbiosis in inflammatory bowel diseases: the oxygen hypothesis. ISME J. 2013;7(7):1256–1261. doi:10.1038/ismej.2013.80.
  • Moayyedi P, Surette MG, Kim PT, Libertucci J, Wolfe M, Onischi C, Armstrong D, Marshall JK, Kassam Z, Reinisch W, et al. Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial. Gastroenterology. 2015;149(1):102–109.e6. doi:10.1053/j.gastro.2015.04.001.
  • Rossen NG, Fuentes S, van der Spek MJ, Tijssen JG, Hartman JHA, Duflou A, Löwenberg M, van den Brink GR, Mathus-Vliegen EMH, de Vos WM, et al. Findings from a randomized controlled trial of fecal transplantation for patients with ulcerative colitis. Gastroenterology. 2015;149(1):110–118.e4. doi:10.1053/j.gastro.2015.03.045.
  • Li SS, Zhu A, Benes V, Costea PI, Hercog R, Hildebrand F, Huerta-Cepas J, Nieuwdorp M, Salojärvi J, Voigt AY, et al. Durable coexistence of donor and recipient strains after fecal microbiota transplantation. Sci. 2016;352(6285):586–589. doi:10.1126/science.aad8852.
  • Jalanka J, Mattila E, Jouhten H, Hartman J, de Vos WM, Arkkila P, Satokari R. Long-term effects on luminal and mucosal microbiota and commonly acquired taxa in faecal microbiota transplantation for recurrent Clostridium difficile infection. BMC Med. 2016;14(1):155. doi:10.1186/s12916-016-0698-z.
  • Smillie CS, Sauk J, Gevers D, Friedman J, Sung J, Youngster I, Hohmann EL, Staley C, Khoruts A, Sadowsky MJ, et al. Strain tracking reveals the determinants of bacterial engraftment in the human gut following fecal microbiota transplantation. Cell Host & Microbe. 2018;23(2):229–240.e5. doi:10.1016/j.chom.2018.01.003.
  • Staley C, Kaiser T, Vaughn BP, Graiziger C, Hamilton MJ, Kabage AJ, Khoruts A, Sadowsky MJ, Pettigrew MM. Durable long-term bacterial engraftment following encapsulated fecal microbiota transplantation to treat Clostridium difficile infection. mBio. 2019;10(4):e01586–19. doi:10.1128/mBio.01586-19.
  • Lahtinen P, Jalanka J, Hartikainen A, Mattila E, Hillilä M, Punkkinen J, Koskenpato J, Anttila V-J, Tillonen J, Satokari R, et al. Randomised clinical trial: faecal microbiota transplantation versus autologous placebo administered via colonoscopy in irritable bowel syndrome. Aliment Pharmacol Ther. 2020;51(12):1321–1331. doi:10.1111/apt.15740.
  • El-Salhy M, Hatlebakk JG, Gilja OH, Bråthen Kristoffersen A, Hausken T. Efficacy of faecal microbiota transplantation for patients with irritable bowel syndrome in a randomised, double-blind, placebo-controlled study. Gut. 2020;69(5):859–867. doi:10.1136/gutjnl-2019-319630.
  • Wilson BC, Vatanen T, Cutfield WS, O’Sullivan JM. The super-donor phenomenon in fecal microbiota transplantation. Front Cell Infect Microbiol. 2019;9:2. doi:10.3389/fcimb.2019.00002.
  • Tett A, Pasolli E, Masetti G, Ercolini D, Segata N. Prevotella diversity, niches and interactions with the human host. Nat Rev Microbiol. 2021;19(9):585–599. doi:10.1038/s41579-021-00559-y.
  • Alpizar-Rodriguez D, Lesker TR, Gronow A, Gilbert B, Raemy E, Lamacchia C, Gabay C, Finckh A, Strowig T. Prevotella copri in individuals at risk for rheumatoid arthritis. Ann Rheum Dis. 2019;78(5):590–593. doi:10.1136/annrheumdis-2018-214514.
  • Larsen JM. The immune response to Prevotella bacteria in chronic inflammatory disease. Immunology. 2017;151(4):363–374. doi:10.1111/imm.12760.
  • Iljazovic A, Roy U, Gálvez EJC, Lesker TR, Zhao B, Gronow A, Amend L, Will SE, Hofmann JD, Pils MC, et al. Perturbation of the gut microbiome by Prevotella spp. enhances host susceptibility to mucosal inflammation. Mucosal Immunol. 2021;14(1):113–124. doi:10.1038/s41385-020-0296-4.
  • Sokol H, Pigneur B, Watterlot L, Lakhdari O, Bermúdez-Humarán LG, Gratadoux J-J, Blugeon S, Bridonneau C, Furet J-P, Corthier G, et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci USA. 2008;105(43):16731–16736. doi:10.1073/pnas.0804812105.
  • Sokol H, Seksik P, Furet JP, Firmesse O, Nion-Larmurier I, Beaugerie L, Cosnes J, Corthier G, Marteau P, Doré J. Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflamm Bowel Dis. 2009;15(8):1183–1189. doi:10.1002/ibd.20903.
  • Miquel S, Martín R, Rossi O, Bermúdez-Humarán L, Chatel JM, Sokol H, Thomas M, Wells JM, Langella P. Faecalibacterium prausnitzii and human intestinal health. Curr Opin Microbiol. 2013;16(3):255–261. doi:10.1016/j.mib.2013.06.003.
  • Machiels K, Joossens M, Sabino J, De Preter V, Arijs I, Eeckhaut V, Ballet V, Claes K, Van Immerseel F, Verbeke K, et al. A decrease of the butyrate-producing species Roseburia hominis and Faecalibacterium prausnitzii defines dysbiosis in patients with ulcerative colitis. Gut. 2014;63(8):1275–1283. doi:10.1136/gutjnl-2013-304833.
  • Reshef L, Kovacs A, Ofer A, Yahav L, Maharshak N, Keren N, Konikoff FM, Tulchinsky H, Gophna U, Dotan I. Pouch inflammation is associated with a decrease in specific bacterial taxa. Gastroenterology. 2015;149(3):718–727. doi:10.1053/j.gastro.2015.05.041.
  • Landy J, Al-Hassi HO, McLaughlin SD, Knight SC, Ciclitira PJ, Nicholls RJ, Clark SK, Hart AL. Etiology of pouchitis. Inflamm Bowel Dis. 2012;18(6):1146–1155. doi:10.1002/ibd.21911.
  • Zlotnik A, Yoshie O. The chemokine superfamily revisited. Immunity. 2012;36(5):705–716. doi:10.1016/j.immuni.2012.05.008.
  • García-Cuesta EM, Santiago CA, Vallejo-Díaz J, Juarranz Y, Rodríguez-Frade JM, Mellado M. The role of the CXCL12/CXCR4/ACKR3 Axis in Autoimmune diseases. Front Endocrinol. 2019;10:585. doi:10.3389/fendo.2019.00585.
  • Dotan I, Werner L, Vigodman S, Weiss S, Brazowski E, Maharshak N, Chen O, Tulchinsky H, Halpern Z, Guzner-Gur H. CXCL12 is a constitutive and inflammatory chemokine in the intestinal immune system. Inflamm Bowel Dis. 2010;16(4):583–592. doi:10.1002/ibd.21106.
  • Mikami S, Nakase H, Yamamoto S, Takeda Y, Yoshino T, Kasahara K, Ueno S, Uza N, Oishi S, Fujii N, et al. Blockade of CXCL12/CXCR4 Axis Ameliorates Murine Experimental Colitis. J Pharmacol Exp Ther. 2008;327(2):383–392. doi:10.1124/jpet.108.141085.
  • Xia X-M, Wang F-Y, Xu W-A, Wang Z-K, Liu J, Lu Y-K, Jin X-X, Lu H, Shen Y-Z. CXCR4 antagonist AMD3100 attenuates colonic damage in mice with experimental colitis. WJG. 2010;16(23):2873–2880. doi:10.3748/wjg.v16.i23.2873.