Microbiota Composition in Diverticular Disease: Implications for Therapy

Antonio Tursi, Valerio Papa, Loris Riccardo Lopetuso, Carlo Romano Settanni, Antonio Gasbarrini, Alfredo Papa, Antonio Tursi, Valerio Papa, Loris Riccardo Lopetuso, Carlo Romano Settanni, Antonio Gasbarrini, Alfredo Papa

Abstract

Gut microbiota (GM) composition and its imbalance are crucial in the pathogenesis of several diseases, mainly those affecting the gastrointestinal tract. Colon diverticulosis and its clinical manifestations (diverticular disease, DD) are among the most common digestive disorders in developed countries. In recent literature, the role of GM imbalance in the onset of the different manifestations within the clinical spectrum of DD has been highlighted. This narrative review aims to summarize and critically analyze the current knowledge on GM dysbiosis in diverticulosis and DD by comparing the available data with those found in inflammatory bowel disease (IBD). The rationale for using probiotics to rebalance dysbiosis in DD is also discussed.

Keywords: acute diverticulitis; diverticular disease; diverticulosis; gut microbiota; probiotics.

Conflict of interest statement

The authors declare no conflict of interest.

References

    1. Tursi A., Scarpignato C., Strate L.L., Lanas A., Kruis W., Lahat A., Danese S. Colonic diverticular disease. Nat. Rev. Dis. Prim. 2020;6:20. doi: 10.1038/s41572-020-0153-5.
    1. Tursi A., Elisei W., Franceschi M., Picchio M., Di Mario F., Brandimarte G. The prevalence of symptomatic uncomplicated diverticular disease could be lower than expected: A single-center colonoscopy-based cohort study. Eur. J. Gastroenterol. Hepatol. 2021;33((Suppl. S1)):e478–e483. doi: 10.1097/MEG.0000000000002142.
    1. Tursi A., Franceschi M., Elisei W., Picchio M., Mario F.D., Brandimarte G. The natural history of symptomatic uncomplicated diverticular disease: A long-term follow-up study. Ann. Gastroenterol. 2021;34:208–213. doi: 10.20524/aog.2020.0560.
    1. Shahedi K., Fuller G., Bolus R., Cohen E., Vu M., Shah R., Agarwal N., Kaneshiro M., Atia M., Sheen V., et al. Long-term risk of acute diverticulitis among patients with incidental diverticulosis found during colonoscopy. Clin. Gastroenterol. Hepatol. 2013;11:1609–1613. doi: 10.1016/j.cgh.2013.06.020.
    1. Giorgetti G., Brandimarte G., Fabiocchi F., Ricci S., Flamini P., Sandri G., Trotta M.C., Elisei W., Penna A., Lecca P.G., et al. Interactions between Innate Immunity, Microbiota, and Probiotics. J. Immunol. Res. 2015;2015:501361. doi: 10.1155/2015/501361.
    1. Ni J., Wu G.D., Albenberg L., Tomov V.T. Gut microbiota and IBD: Causation or correlation? Nat. Rev. Gastroenterol. Hepatol. 2017;14:573–584. doi: 10.1038/nrgastro.2017.88.
    1. Jones R.B., Fodor A.A., Peery A.F., Tsilimigras M.C.B., Winglee K., McCoy A., Sioda M., Sandler R.S., Keku T.O. An Aberrant Microbiota is not Strongly Associated with Incidental Colonic Diverticulosis. Sci. Rep. 2018;8:4951. doi: 10.1038/s41598-018-23023-z.
    1. van Rossen T.M., Ooijevaar R.E., Kuyvenhoven J.P., Eck A., Bril H., Buijsman R., Boermeester M.A., Stockmann H.B.A.C., de Korte N., Budding A.E. Microbiota composition and mucosal immunity in patients with asymptomatic diverticulosis and controls. PLoS ONE. 2021;16:e0256657. doi: 10.1371/journal.pone.0256657.
    1. Ponziani F.R., Scaldaferri F., Petito V., Paroni Sterbini F., Pecere S., Lopetuso L.R., Palladini A., Gerardi V., Masucci L., Pompili M., et al. The Role of Antibiotics in Gut Microbiota Modulation: The Eubiotic Effects of Rifaximin. Dig. Dis. 2016;34:269–278. doi: 10.1159/000443361.
    1. Tursi A., Mastromarino P., Capobianco D., Elisei W., Miccheli A., Capuani G., Tomassini A., Campagna G., Picchio M., Giorgetti G., et al. Assessment of Fecal Microbiota and Fecal Metabolome in Symptomatic Uncomplicated Diverticular Disease of the Colon. J. Clin. Gastroenterol. 2016;50((Suppl. S1)):S9–S12. doi: 10.1097/MCG.0000000000000626.
    1. Barbara G., Scaioli E., Barbaro M.R., Biagi E., Laghi L., Cremon C., Marasco G., Colecchia A., Picone G., Salfi N., et al. Gut microbiota, metabolome and immune signatures in patients with uncomplicated diverticular disease. Gut. 2017;66:1252–1261. doi: 10.1136/gutjnl-2016-312377.
    1. Kvasnovsky C.L., Leong L.E.X., Choo J.M., Abell G.C.J., Papagrigoriadis S., Bruce K.D., Rogers G.B. Clinical and symptom scores are significantly correlated with fecal microbiota features in patients with symptomatic uncomplicated diverticular disease: A pilot study. Eur. J. Gastroenterol. Hepatol. 2018;30:107–112. doi: 10.1097/MEG.0000000000000995.
    1. Lopetuso L.R., Petito V., Graziani C., Schiavoni E., Paroni Sterbini F., Poscia A., Gaetani E., Franceschi F., Cammarota G., Sanguinetti M., et al. Gut Microbiota in Health, Diverticular Disease, Irritable Bowel Syndrome, and Inflammatory Bowel Diseases: Time for Microbial Marker of Gastrointestinal Disorders. Dig. Dis. 2018;36:56–65. doi: 10.1159/000477205.
    1. Linninge C., Roth B., Erlanson-Albertsson C., Molin G., Toth E., Ohlsson B. Abundance of Enterobacteriaceae in the colon mucosa in diverticular disease. World J. Gastrointest. Pathophysiol. 2018;9:18–27. doi: 10.4291/wjgp.v9.i1.18.
    1. Laghi L., Mastromarino P., Elisei W., Capobianco D., Zhu C.L., Picchio M., Giorgetti G., Brandimarte G., Tursi A. Impact of treatments on fecal microbiota and fecal metabolome in symptomatic uncomplicated diverticular disease of the colon: A pilot study. J. Biol. Regul. Homeost. Agents. 2018;32:1421–1432.
    1. 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;63:1275–1283. doi: 10.1136/gutjnl-2013-304833.
    1. Ponziani F.R., Scaldaferri F., De Siena M., Mangiola F., Matteo M.V., Pecere S., Petito V., Sterbini F.P., Lopetuso L.R., Masucci L., et al. Increased Faecalibacterium abundance is associated with clinical improvement in patients receiving rifaximin treatment. Benef. Microbes. 2020;11:519–525. doi: 10.3920/BM2019.0171.
    1. Tursi A., Mastromarino P., Capobianco D., Elisei W., Campagna G., Picchio M., Giorgetti G., Fabiocchi F., Brandimarte G. Faecalibacterium prausnitzii is not decreased in symptomatic uncomplicated diverticular disease of the colon. Biosci. Microbiota Food Health. 2022:2022–2046. doi: 10.12938/bmfh.2022-046.
    1. Gueimonde M., Ouwehand A., Huhtinen H., Salminen E., Salminen S. Qualitative and quantitative analyses of the bifidobacterial microbiota in the colonic mucosa of patients with colorectal cancer, diverticulitis, and inflammatory bowel disease. World J. Gastroenterol. 2007;13:3985–3989. doi: 10.3748/wjg.v13.i29.3985.
    1. Daniels L., Budding A.E., de Korte N., Eck A., Bogaards J.A., Stockmann H.B., Consten E.C., Savelkoul P.H., Boermeester M.A. Fecal microbiome analysis as a diagnostic test for diverticulitis. Eur. J. Clin. Microbiol. Infect. Dis. 2014;33:1927–1936. doi: 10.1007/s10096-014-2162-3.
    1. Schieffer K.M., Sabey K., Wright J.R., Toole D.R., Drucker R., Tokarev V., Harris L.R., Deiling S., Eshelman M.A., Hegarty J.P., et al. The Microbial Ecosystem Distinguishes Chronically Diseased Tissue from Adjacent Tissue in the Sigmoid Colon of Chronic, Recurrent Diverticulitis Patients. Sci. Rep. 2017;7:8467. doi: 10.1038/s41598-017-06787-8.
    1. O’Grady M.J., Turner G.A., Sulit A., Frizelle F.A., Purcell R. Distinct changes in the colonic microbiome associated with acute diverticulitis. Color. Dis. 2022:1–11. doi: 10.1111/codi.16271.
    1. Hayashi A., Sato T., Kamada N., Mikami Y., Matsuoka K., Hisamatsu T., Hibi T., Roers A., Yagita H., Ohteki T., et al. A single strain of Clostridium butyricum induces intestinal IL-10-producing macrophages to suppress acute experimental colitis in mice. Cell Host Microbe. 2013;13:711–722. doi: 10.1016/j.chom.2013.05.013.
    1. Atarashi K., Tanoue T., Oshima K., Suda W., Nagano Y., Nishikawa H., Fukuda S., Saito T., Narushima S., Hase K., et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013;500:232–236. doi: 10.1038/nature12331.
    1. Rodriguez-Nogales A., Algieri F., Garrido-Mesa J., Vezza T., Utrilla M.P., Chueca N., Garcia F., Olivares M., Rodríguez-Cabezas M.E., Gálvez J. Differential intestinal anti-inflammatory effects of Lactobacillus fermentum and Lactobacillus salivarius in DSS mouse colitis: Impact of microRNAs expression and microbiota composition. Mol. Nutr. Food Res. 2017;61:1700144. doi: 10.1002/mnfr.201700144.
    1. Knox N.C., Forbes J.D., Peterson C.L., Van Domselaar G., Bernstein C.N. The Gut Microbiome in Inflammatory Bowel Disease: Lessons Learned From Other Immune-Mediated Inflammatory Diseases. Am. J. Gastroenterol. 2019;114:1051–1070. doi: 10.14309/ajg.0000000000000305.
    1. Lopez-Siles M., Enrich-Capo N., Aldeguer X., Sabat-Mir M., Duncan S.H., Garcia-Gil L.J., Martinez-Medina M. Alterations in the Abundance and Co-occurrence of Akkermansia muciniphila and Faecalibacterium prausnitzii in the Colonic Mucosa of Inflammatory Bowel Disease Subjects. Front. Cell. Infect. Microbiol. 2018;8:281. doi: 10.3389/fcimb.2018.00281.
    1. Belzer C., de Vos W.M. Microbes inside—From diversity to function: The case of Akkermansia. ISME J. 2012;6:1449–1458. doi: 10.1038/ismej.2012.6.
    1. Alam A., Leoni G., Quiros M., Wu H., Desai C., Nishio H., Jones R.M., Nusrat A., Neish A.S. The microenvironment of injured murine gut elicits a local pro-restitutive microbiota. Nat. Microbiol. 2016;1:15021. doi: 10.1038/nmicrobiol.2015.21.
    1. Derrien M., Vaughan E.E., Plugge C.M., de Vos W.M. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 2004;54:1469–1476. doi: 10.1099/ijs.0.02873-0.
    1. Van Passel M.W., Kant R., Zoetendal E.G., Plugge C.M., Derrien M., Malfatti S.A., Chain P.S., Woyke T., Palva A., de Vos W.M., et al. The genome of Akkermansia muciniphila, a dedicated intestinal mucin degrader, and its use in exploring intestinal metagenomes. PLoS ONE. 2011;6:e16876. doi: 10.1371/journal.pone.0016876.
    1. Everard A., Belzer C., Geurts L., Ouwerkerk J.P., Druart C., Bindels L.B., Guiot Y., Derrien M., Muccioli G.G., Delzenne N.M., et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA. 2013;110:9066–9071. doi: 10.1073/pnas.1219451110.
    1. Macchione I.G., Lopetuso L.R., Ianiro G., Napoli M., Gibiino G., Rizzatti G., Petito V., Gasbarrini A., Scaldaferri F. Akkermansia muciniphila: Key player in metabolic and gastrointestinal disorders. Eur. Rev. Med. Pharmacol. Sci. 2019;23:8075–8083.
    1. Lopetuso L.R., Quagliariello A., Schiavoni M., Petito V., Russo A., Reddel S., del Chierico F., Ianiro G., Scaldaferri F., Neri M., et al. Towards a disease-associated common trait of gut microbiota dysbiosis: The pivotal role of Akkermansia muciniphila. Dig. Liver Dis. 2020;52:1002–1010. doi: 10.1016/j.dld.2020.05.020.
    1. Hall A.B., Yassour M., Sauk J., Garner A., Jiang X., Arthur T., Lagoudas G.K., Vatanen T., Fornelos N., Wilson R., et al. A novel Ruminococcus gnavus clade enriched in inflammatory bowel disease patients. Genome Med. 2017;9:103. doi: 10.1186/s13073-017-0490-5.
    1. Tursi A., Elisei W., Brandimarte G., Giorgetti G.M., Inchingolo C.D., Nenna R., Picchio M., Giorgio F., Ierardi E. Musosal tumor necrosis factor α in diverticular disease of the colon is overexpressed with disease severity. Color. Dis. 2012;14:e258–e263. doi: 10.1111/j.1463-1318.2012.02926.x.
    1. Humes D.J., Simpson J., Smith J., Sutton P., Zaitoun A., Bush D., Bennett A., Scholefield J.H., Spiller R.C. Visceral hypersensitivity in symptomatic diverticular disease and the role of neuropeptides and low grade inflammation. Neurogastroenterol. Motil. 2012;24:318-e163. doi: 10.1111/j.1365-2982.2011.01863.x.
    1. Tursi A., Elisei W., Inchingolo C.D., Nenna R., Picchio M., Ierardi E., Brandimarte G. Chronic diverticulitis and Crohn’s disease share the same expression of basic fibroblastic growth factor, syndecan 1 and tumor necrosis factor-α. J. Clin. Pathol. 2014;67:844–846. doi: 10.1136/jclinpath-2013-202137.
    1. Connelly T.M., Choi C.S., Berg A.S., Harris L., 3rd., Coble J., Koltun W.A. Diverticulitis and Crohn’s disease have distinct but overlapping tumor necrosis superfamily 15 haplotypes. J. Surg. Res. 2017;214:262–269. doi: 10.1016/j.jss.2017.02.030.
    1. Dai L., King D.W., Perera D.S., Lubowski D.Z., Burcher E., Liu L. Inverse expression of prostaglandin E2-related enzymes highlights differences between diverticulitis and inflammatory bowel disease. Dig. Dis. Sci. 2015;60:1236–1246. doi: 10.1007/s10620-014-3478-7.
    1. Li M.C., He S.H. IL-10 and its related cytokines for treatment of inflammatory bowel disease. World J. Gastroenterol. 2004;10:620–625. doi: 10.3748/wjg.v10.i5.620.
    1. Tursi A., Mastromarino P., Capobianco D., Elisei W., Picchio M., Brandimarte G. No changes in Interleukin-10 expression in symptomatic uncomplicated diverticular disease of the colon. J. Gastrointest. Liver Dis. 2018;27:476–477. doi: 10.15403/jgld.2014.1121.274.ink.
    1. Turco F., Andreozzi P., Palumbo I., Zito F.P., Cargiolli M., Fiore W., Gennarelli N., De Palma G.D., Sarnelli G., Cuomo R. Bacterial stimuli activate nitric oxide colonic mucosal production in diverticular disease. Protective effects of L. casei DG® (Lactobacillus paracasei CNCM I-1572) United Eur. Gastroenterol. J. 2017;5:715–724. doi: 10.1177/2050640616684398.
    1. O’Toole P.W., Jeffery I.B. Gut microbiota and aging. Science. 2015;350:1214–1215. doi: 10.1126/science.aac8469.
    1. Bischoff S.C. Microbiota and aging. Curr. Opin. Clin. Nutr. Metab. Care. 2016;19:26–30. doi: 10.1097/MCO.0000000000000242.
    1. Ottman N., Smidt H., de Vos W.M., Belzer C. The function of our microbiota: Who is out there and what do they do? Front. Cell. Infect. Microbiol. 2012;2:104. doi: 10.3389/fcimb.2012.00104.
    1. Biagi E., Nylund L., Candela M., Ostan R., Bucci L., Pini E., Nikkïla J., Monti D., Satokari R., Franceschi C., et al. Through ageing, and beyond: Gut microbiota and inflammatory status in seniors and centenarians. PLoS ONE. 2010;5:e10667. doi: 10.1371/annotation/df45912f-d15c-44ab-8312-e7ec0607604d.
    1. Heinritz S.N., Weiss E., Eklund M., Aumiller T., Heyer C.M., Messner S., Rings A., Louis S., Bischoff S.C., Mosenthin R. Impact of a high-fat or high-fiber diet on intestinal microbiota and metabolic markers in a pig model. Nutrients. 2016;8:317. doi: 10.3390/nu8050317.
    1. Dominianni C., Sinha R., Goedert J.J., Pei Z., Yang L., Hayes R.B., Ahn J. Sex, body mass index, and dietary fiber intake influence the human gut microbiome. PLoS ONE. 2015;10:e0124599. doi: 10.1371/journal.pone.0124599.
    1. Chen H.M., Yu Y.N., Wang J.L., Lin Y.W., Kong X., Yang C.Q., Yang L., Liu Z.J., Yuan Y.Z., Liu F., et al. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal carcinoma. Am. J. Clin. Nutr. 2013;97:1044–1052. doi: 10.3945/ajcn.112.046607.
    1. Macia L., Tan J., Vieira A.T., Leach K., Stanley D., Luong S., Maruya M., McLenzie I., Hijkata A., Wong C., et al. Metabolite-sensing receptors GPR43 and GPR109A facilitate dietary fibre-induced gut homeostasis through regulation of the inflammasome. Nat. Commun. 2015;6:6734. doi: 10.1038/ncomms7734.
    1. Zhu Y., Shi X., Lin X., Ye K., Xu X., Li C., Zhou G. Beef, chicken, and soy proteins in diets induce different gut microbiota and metabolites in rats. Front. Microbiol. 2017;8:1395. doi: 10.3389/fmicb.2017.01395.
    1. Engen P.A., Green S.J., Voigt R.M., Forsyth C.B., Keshavarzian A. The gastrointestinal microbiome: Alcohol effects on the composition of intestinal microbiota. Alcohol Res. 2015;37:223–236.
    1. Mentella M.C., Scaldaferri F., Pizzoferrato M., Gasbarrini A., Miggiano G.A.D. Nutrition, IBD and Gut Microbiota: A Review. Nutrients. 2020;12:944. doi: 10.3390/nu12040944.
    1. Sugihara K., Kamada N. Diet-Microbiota Interactions in Inflammatory Bowel Disease. Nutrients. 2021;13:1533. doi: 10.3390/nu13051533.
    1. Ananthakrishnan A.N., Khalili H., Song M., Higuchi L.M., Richter J.M., Chan A.T. Zinc intake and risk of Crohn’s disease and ulcerative colitis: A prospective cohort study. Int. J. Epidemiol. 2015;44:1995–2005. doi: 10.1093/ije/dyv301.
    1. Ananthakrishnan A.N., Khalili H., Konijeti G.G., Higuchi L.M., de Silva P., Korzenik J.R., Fuchs C.S., Willett W.C., Richter J.M., Chan A.T. A prospective study of long-term intake of dietary fiber and risk of Crohn’s disease and ulcerative colitis. Gastroenterology. 2013;145:970–977. doi: 10.1053/j.gastro.2013.07.050.
    1. Tursi A., Brandimarte G., Di Mario F., Elisei W., Picchio M., Allegretta L., Annunziata M.L., Bafutto M., Bassotti G., Bianco M.A., et al. An international, multicentre, prospective cohort study is the prognostic performance of the ‘DICA’ endoscopic classification and the ‘CODA’ score in predicting clinical outcomes of diverticular disease. Gut. 2022;71:1350–1358. doi: 10.1136/gutjnl-2021-325574.
    1. Ticinesi A., Milani C., Lauretani F., Nouvenne A., Mancabelli L., Lugli G.A., Turroni F., Duranti S., Mangifesta M., Viappiani A., et al. Gut microbiota composition is associated with polypharmacy in elderly hospitalized patients. Sci. Rep. 2017;7:11102. doi: 10.1038/s41598-017-10734-y.
    1. Jackson M.A., Verdi S., Maxan M.E., Shin C.M., Zierer J., Bowyer R.C.E., Martin T., Williams F.M.K., Menni C., Bell J.T., et al. Gut microbiota associations with common diseases and prescription medications in a population-based cohort. Nat. Commun. 2018;9:2655. doi: 10.1038/s41467-018-05184-7.
    1. Zhuang X., Tian Z., Feng R., Li M., Li T., Zhou G., Qiu Y., Chen B., He Y., Chen M., et al. Fecal Microbiota Alterations Associated With Clinical and Endoscopic Response to Infliximab Therapy in Crohn’s Disease. Inflamm. Bowel Dis. 2020;26:1636–1647. doi: 10.1093/ibd/izaa253.
    1. Chen L., Lu Z., Kang D., Feng Z., Li G., Sun M., Liu Z., Wu W., Fang L. Distinct alterations of fecal microbiota refer to the efficacy of adalimumab in Crohn’s disease. Front. Pharmacol. 2022;13:913720. doi: 10.3389/fphar.2022.913720.
    1. Ribaldone D.G., Caviglia G.P., Abdulle A., Pellicano R., Ditto M.C., Morino M., Fusaro E., Saracco G.M., Bugianesi E., Astegiano M. Adalimumab Therapy Improves Intestinal Dysbiosis in Crohn’s Disease. J. Clin. Med. 2019;8:1646. doi: 10.3390/jcm8101646.
    1. Radhakrishnan S.T., Alexander J.L., Mullish B.H., Gallagher K.I., Powell N., Hicks L.C., Hart A.L., Li J.V., Marchesi J.R., Williams H.R.T. Systematic review: The association between the gut microbiota and medical therapies in inflammatory bowel disease. Aliment. Pharmacol. Ther. 2022;55:26–48. doi: 10.1111/apt.16656.
    1. Montalban-Arques A., De Schryver P., Bossier P., Gorkiewicz G., Mulero V., Gatlin D.M., 3rd., Galindo-Villegas J. Selective Manipulation of the Gut Microbiota Improves Immune Status in Vertebrates. Front. Immunol. 2015;6:512. doi: 10.3389/fimmu.2015.00512.
    1. Scarpignato C., Bertelé A., Tursi A. Probiotics for the Treatment of Symptomatic Uncomplicated Diverticular Disease: Rationale and Current Evidence. J. Clin. Gastroenterol. 2016;50((Suppl. S1)):S70–S73. doi: 10.1097/MCG.0000000000000641.
    1. Rondanelli M., Faliva M.A., Perna S., Giacosa A., Peroni G., Castellazzi A.M. Using probiotics in clinical practice: Where are we now? A review of existing meta-analyses. Gut Microbes. 2017;8:521–543. doi: 10.1080/19490976.2017.1345414.
    1. Ticinesi A., Nouvenne A., Corrente V., Tana C., Di Mario F., Meschi T. Diverticular Disease: A Gut Microbiota Perspective. J. Gastrointest. Liver Dis. 2019;28:327–337. doi: 10.15403/jgld-277.
    1. Kvasnovsky C.L., Bjarnason I., Donaldson A.N., Sherwood R.A., Papagrigoriadis S. A randomized double-blind placebo-controlled trial of a multi-strain probiotic in treatment of symptomatic uncomplicated diverticular disease. Inflammopharmacology. 2017;25:499–509. doi: 10.1007/s10787-017-0363-y.
    1. Tursi A., Brandimarte G., Elisei W., Picchio M., Forti G., Pianese G., Rodino S., D’Amico T., Sacca N., Portincasa P., et al. Randomised clinical trial: Mesalazine and/or probiotics in maintaining remission of symptomatic uncomplicated diverticular disease: A double-blind, randomised, placebo-controlled study. Aliment. Pharmacol. Ther. 2013;38:741–751. doi: 10.1111/apt.12463.
    1. Bretto E., D’Amico F., Fiore W., Tursi A., Danese S. Lactobacillus paracasei CNCM I 1572: A Promising Candidate for Management of Colonic Diverticular Disease. J. Clin. Med. 2022;11:1916. doi: 10.3390/jcm11071916.
    1. Petruzziello C., Migneco A., Cardone S., Covino M., Saviano A., Franceschi F., Ojetti V. Supplementation with Lactobacillus reuteri ATCC PTA 4659 in patients affected by acute uncomplicated diverticulitis: A randomized double-blind placebo controlled trial. Int. J. Color. Dis. 2019;34:1087–1109. doi: 10.1007/s00384-019-03295-1.
    1. Ojetti V., Saviano A., Brigida M., Petruzziello C., Caronna M., Gayani G., Franceschi F. Randomized control trial on the efficacy of Limosilactobacillus reuteri ATCC PTA 4659 in reducing inflammatory markers in acute uncomplicated diverticulitis. Eur. J. Gastroenterol. Hepatol. 2022;34:496–502. doi: 10.1097/MEG.0000000000002342.

Source: PubMed

3
Subscribe