메뉴 건너뛰기




Volumn 30, Issue 1, 2017, Pages 191-231

Gleaning insights from fecal microbiota transplantation and probiotic studies for the rational design of combination microbial therapies

Author keywords

Clostridium difficile; Fecal microbiota transplantation; Microbiota; Probiotics; Ulcerative colitis

Indexed keywords

AMINOPENICILLIN; BACTERIAL TOXIN; BETA CATENIN; CEPHALOSPORIN; CHEMOKINE RECEPTOR CCR2; CLINDAMYCIN; EPITHELIAL DERIVED NEUTROPHIL ACTIVATING FACTOR 78; FORDIN; GAMMA INTERFERON; GLIADIN; GRANULOCYTE CHEMOTACTIC PROTEIN 2; MITOGEN ACTIVATED PROTEIN KINASE 1; MITOGEN ACTIVATED PROTEIN KINASE 3; MONOCYTE CHEMOTACTIC PROTEIN; MUCIN 1; MUCIN 2; OCCLUDIN; PEPTIDES AND PROTEINS; PROBIOTIC AGENT; PROTEIN P130; PROTON PUMP INHIBITOR; SECONDARY LYMPHOID TISSUE CHEMOKINE; SYMPLEKIN; TOLL LIKE RECEPTOR; TOLL LIKE RECEPTOR 2; TOLL LIKE RECEPTOR 4; TUMOR NECROSIS FACTOR; UNCLASSIFIED DRUG; UNINDEXED DRUG; UVOMORULIN; VANCOMYCIN;

EID: 85007188919     PISSN: 08938512     EISSN: 10986618     Source Type: Journal    
DOI: 10.1128/CMR.00049-16     Document Type: Review
Times cited : (62)

References (386)
  • 2
    • 84971378706 scopus 로고    scopus 로고
    • Fecal microbiota transplantation: Current applications, effectiveness, and future perspectives
    • Choi HH, Cho Y. 2016. Fecal microbiota transplantation: current applications, effectiveness, and future perspectives. Clin Endosc 49: 257-265. https://doi.org/10.5946/ce.2015.117.
    • (2016) Clin Endosc , vol.49 , pp. 257-265
    • Choi, H.H.1    Cho, Y.2
  • 3
    • 84874737205 scopus 로고    scopus 로고
    • High-throughput DNA sequence analysis reveals stable engraftment of gut microbiota following transplantation of previously frozen fecal bacteria
    • Hamilton MJ, Weingarden AR, Unno T, Khoruts A, Sadowsky MJ. 2013. High-throughput DNA sequence analysis reveals stable engraftment of gut microbiota following transplantation of previously frozen fecal bacteria. Gut Microbes 4: 125-135. https://doi.org/10.4161/gmic.23571.
    • (2013) Gut Microbes , vol.4 , pp. 125-135
    • Hamilton, M.J.1    Weingarden, A.R.2    Unno, T.3    Khoruts, A.4    Sadowsky, M.J.5
  • 4
    • 84966420793 scopus 로고    scopus 로고
    • Optimal screening and donor management in a public stool bank
    • Kazerouni A, Burgess J, Burns LJ, Wein LM. 2015. Optimal screening and donor management in a public stool bank. Microbiome 3: 75. https:// doi.org/10.1186/s40168-015-0140-3.
    • (2015) Microbiome , vol.3 , pp. 75
    • Kazerouni, A.1    Burgess, J.2    Burns, L.J.3    Wein, L.M.4
  • 6
    • 84881148330 scopus 로고    scopus 로고
    • Norovirus gastroenteritis after fecal microbiota transplantation for treatment of Clostridium difficile infection despite asymptomatic donors and lack of sick contacts
    • Schwartz M, Gluck M, Koon S. 2013. Norovirus gastroenteritis after fecal microbiota transplantation for treatment of Clostridium difficile infection despite asymptomatic donors and lack of sick contacts. Am J Gastroenterol 108: 1367. https://doi.org/10.1038/ajg.2013.164.
    • (2013) Am J Gastroenterol , vol.108 , pp. 1367
    • Schwartz, M.1    Gluck, M.2    Koon, S.3
  • 8
    • 84930375791 scopus 로고    scopus 로고
    • Fecal microbiota transplantation (FMT) for Clostridium difficile infection: Focus on immunocompromised patients
    • Di Bella S, Gouliouris T, Petrosillo N. 2015. Fecal microbiota transplantation (FMT) for Clostridium difficile infection: focus on immunocompromised patients. J Infect Chemother 21: 230-237. https://doi.org/10.1016/j.jiac.2015.01.011.
    • (2015) J Infect Chemother , vol.21 , pp. 230-237
    • Di Bella, S.1    Gouliouris, T.2    Petrosillo, N.3
  • 9
    • 84880601378 scopus 로고    scopus 로고
    • Transient flare of ulcerative colitis after fecal microbiota transplantation for recurrent Clostridium difficile infection
    • De Leon LM, Watson JB, Kelly CR. 2013. Transient flare of ulcerative colitis after fecal microbiota transplantation for recurrent Clostridium difficile infection. Clin Gastroenterol Hepatol 11: 1036-1038. https:// doi.org/10.1016/j.cgh.2013.04.045.
    • (2013) Clin Gastroenterol Hepatol , vol.11 , pp. 1036-1038
    • De Leon, L.M.1    Watson, J.B.2    Kelly, C.R.3
  • 11
    • 84978328160 scopus 로고    scopus 로고
    • Weight gain after fecal microbiota transplantation
    • ofv004
    • Alang N, Kelly CR. 2015. Weight gain after fecal microbiota transplantation. Open Forum Infect Dis 2: ofv004. https://doi.org/10.1093/ofid/ ofv004.
    • (2015) Open Forum Infect Dis , vol.2
    • Alang, N.1    Kelly, C.R.2
  • 13
    • 84975125454 scopus 로고    scopus 로고
    • Banking on stool despite an uncertain future
    • Amirtha T. 2016. Banking on stool despite an uncertain future. Science 352: 1261-1262. https://doi.org/10.1126/science.352.6291.1261.
    • (2016) Science , vol.352 , pp. 1261-1262
    • Amirtha, T.1
  • 14
    • 84920911290 scopus 로고    scopus 로고
    • Fecal transplant policy and legislation
    • Vyas D, Aekka A, Vyas A. 2015. Fecal transplant policy and legislation. World J Gastroenterol 21: 6-11. https://doi.org/10.3748/wjg.v21.i1.6.
    • (2015) World J Gastroenterol , vol.21 , pp. 6-11
    • Vyas, D.1    Aekka, A.2    Vyas, A.3
  • 15
    • 33645455337 scopus 로고    scopus 로고
    • Meta-analysis of probiotics for the prevention of antibiotic associated diarrhea and the treatment of Clostridium difficile disease
    • McFarland L. 2006. Meta-analysis of probiotics for the prevention of antibiotic associated diarrhea and the treatment of Clostridium difficile disease. Am J Gastroenterol 101: 812-822. https://doi.org/10.1111/ j.1572-0241.2006.00465.x.
    • (2006) Am J Gastroenterol , vol.101 , pp. 812-822
    • McFarland, L.1
  • 16
    • 84930475582 scopus 로고    scopus 로고
    • Systematic review of randomized controlled trials of probiotics, prebiotics, and synbiotics in inflammatory bowel disease
    • Ghouri YA, Richards DM, Rahimi EF, Krill JT, Jelinek KA, DuPont AW. 2014. Systematic review of randomized controlled trials of probiotics, prebiotics, and synbiotics in inflammatory bowel disease. Clin Exp Gastroenterol 7: 473-487. https://doi.org/10.2147/CEG.S27530.
    • (2014) Clin Exp Gastroenterol , vol.7 , pp. 473-487
    • Ghouri, Y.A.1    Richards, D.M.2    Rahimi, E.F.3    Krill, J.T.4    Jelinek, K.A.5    DuPont, A.W.6
  • 18
    • 59749088586 scopus 로고    scopus 로고
    • Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis
    • Miele E, Pascarella F, Giannetti E, Quaglietta L, Baldassano RN, Staiano A. 2009. Effect of a probiotic preparation (VSL#3) on induction and maintenance of remission in children with ulcerative colitis. Am J Gastroenterol 104: 437-443. https://doi.org/10.1038/ajg.2008.118.
    • (2009) Am J Gastroenterol , vol.104 , pp. 437-443
    • Miele, E.1    Pascarella, F.2    Giannetti, E.3    Quaglietta, L.4    Baldassano, R.N.5    Staiano, A.6
  • 19
    • 58949102552 scopus 로고    scopus 로고
    • Lactobacillus phylogenomics-towards a reclassification of the genus
    • Claesson MJ, van Sinderen D, O’Toole PW. 2008. Lactobacillus phylogenomics-towards a reclassification of the genus. Int J Syst Evol Microbiol 58: 2945-2954. https://doi.org/10.1099/ijs.0.65848-0.
    • (2008) Int J Syst Evol Microbiol , vol.58 , pp. 2945-2954
    • Claesson, M.J.1    van Sinderen, D.2    O’Toole, P.W.3
  • 21
    • 84973442912 scopus 로고    scopus 로고
    • Phylogenetic analysis of the Bifidobacterium genus using glycolysis enzyme sequences
    • Brandt K, Barrangou R. 2016. Phylogenetic analysis of the Bifidobacterium genus using glycolysis enzyme sequences. Front Microbiol 7: 657. https://doi.org/10.3389/fmicb.2016.00657.
    • (2016) Front Microbiol , vol.7 , pp. 657
    • Brandt, K.1    Barrangou, R.2
  • 22
    • 80455140368 scopus 로고    scopus 로고
    • Ulcerative colitis
    • Danese S, Fiocchi C. 2011. Ulcerative colitis. N Engl J Med 365: 1713-1725. https://doi.org/10.1056/NEJMra1102942.
    • (2011) N Engl J Med , vol.365 , pp. 1713-1725
    • Danese, S.1    Fiocchi, C.2
  • 23
    • 67649391053 scopus 로고    scopus 로고
    • Clostridium difficile infection: New developments in epidemiology and pathogenesis
    • Rupnik M, Wilcox MH, Gerding DN. 2009. Clostridium difficile infection: new developments in epidemiology and pathogenesis. Nat Rev Microbiol 7: 526-536. https://doi.org/10.1038/nrmicro2164.
    • (2009) Nat Rev Microbiol , vol.7 , pp. 526-536
    • Rupnik, M.1    Wilcox, M.H.2    Gerding, D.N.3
  • 26
    • 0024545882 scopus 로고
    • Nosocomial acquisition of Clostridium difficile infection
    • McFarland L, Mulligan M, Kwok R, Stamm W. 1989. Nosocomial acquisition of Clostridium difficile infection. N Engl J Med 320: 204-210. https://doi.org/10.1056/NEJM198901263200402.
    • (1989) N Engl J Med , vol.320 , pp. 204-210
    • McFarland, L.1    Mulligan, M.2    Kwok, R.3    Stamm, W.4
  • 27
    • 0019497353 scopus 로고
    • Isolation of Clostridium difficile from the environment and contacts of patients with antibiotic-associated colitis
    • Kim KH, Fekety R, Batts DH, Brown D, Cudmore M, Silva J, Waters D. 1981. Isolation of Clostridium difficile from the environment and contacts of patients with antibiotic-associated colitis. J Infect Dis 143: 42-50. https://doi.org/10.1093/infdis/143.1.42.
    • (1981) J Infect Dis , vol.143 , pp. 42-50
    • Kim, K.H.1    Fekety, R.2    Batts, D.H.3    Brown, D.4    Cudmore, M.5    Silva, J.6    Waters, D.7
  • 29
    • 41549142775 scopus 로고    scopus 로고
    • Bile salts and glycine as cogerminants for Clostridium difficile spores
    • Sorg JA, Sonenshein AL. 2008. Bile salts and glycine as cogerminants for Clostridium difficile spores. J Bacteriol 190: 2505-2512. https://doi.org/10.1128/JB.01765-07.
    • (2008) J Bacteriol , vol.190 , pp. 2505-2512
    • Sorg, J.A.1    Sonenshein, A.L.2
  • 30
    • 77951026738 scopus 로고    scopus 로고
    • Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the Society for Healthcare Epidemiology of America (SHEA) and the Infectious Diseases Society of America (IDSA)
    • Cohen SH, Gerding DN, Johnson S, Kelly CP, Loo VG, McDonald LC, Pepin J, Wilcox MH. 2010. Clinical practice guidelines for Clostridium difficile infection in adults: 2010 update by the Society for Healthcare Epidemiology of America (SHEA) and the Infectious Diseases Society of America (IDSA). Infect Control Hosp Epidemiol 31: 431-455. https:// doi.org/10.1086/651706.
    • (2010) Infect Control Hosp Epidemiol , vol.31 , pp. 431-455
    • Cohen, S.H.1    Gerding, D.N.2    Johnson, S.3    Kelly, C.P.4    Loo, V.G.5    McDonald, L.C.6    Pepin, J.7    Wilcox, M.H.8
  • 31
    • 84884510057 scopus 로고    scopus 로고
    • Use of multilocus variable number of tandem repeats analysis genotyping to determine the role of asymptomatic carriers in Clostridium difficile transmission
    • Curry SR, Muto CA, Schlackman JL, Pasculle AW, Shutt KA, Marsh JW, Harrison LH. 2013. Use of multilocus variable number of tandem repeats analysis genotyping to determine the role of asymptomatic carriers in Clostridium difficile transmission. Clin Infect Dis 57: 1094-1102. https://doi.org/10.1093/cid/cit475.
    • (2013) Clin Infect Dis , vol.57 , pp. 1094-1102
    • Curry, S.R.1    Muto, C.A.2    Schlackman, J.L.3    Pasculle, A.W.4    Shutt, K.A.5    Marsh, J.W.6    Harrison, L.H.7
  • 32
    • 0034628426 scopus 로고    scopus 로고
    • Asymptomatic carriage of Clostridium difficile and serum levels of IgG antibody against toxin A
    • Kyne L, Warny M, Qamar A, Kelly CP. 2000. Asymptomatic carriage of Clostridium difficile and serum levels of IgG antibody against toxin A. N Engl J Med 342: 390-397. https://doi.org/10.1056/NEJM200002103420604.
    • (2000) N Engl J Med , vol.342 , pp. 390-397
    • Kyne, L.1    Warny, M.2    Qamar, A.3    Kelly, C.P.4
  • 33
    • 84855510455 scopus 로고    scopus 로고
    • The role of toxin A and toxin B in the virulence of Clostridium difficile
    • Carter GP, Rood JI, Lyras D. 2012. The role of toxin A and toxin B in the virulence of Clostridium difficile. Trends Microbiol 20: 21-29. https:// doi.org/10.1016/j.tim.2011.11.003.
    • (2012) Trends Microbiol , vol.20 , pp. 21-29
    • Carter, G.P.1    Rood, J.I.2    Lyras, D.3
  • 35
    • 84891353755 scopus 로고    scopus 로고
    • Importance of toxin A, toxin B, and CDT in virulence of an epidemic Clostridium difficile strain
    • Kuehne SA, Collery MM, Kelly ML, Cartman ST, Cockayne A, Minton NP. 2014. Importance of toxin A, toxin B, and CDT in virulence of an epidemic Clostridium difficile strain. J Infect Dis 209: 83-86. https:// doi.org/10.1093/infdis/jit426.
    • (2014) J Infect Dis , vol.209 , pp. 83-86
    • Kuehne, S.A.1    Collery, M.M.2    Kelly, M.L.3    Cartman, S.T.4    Cockayne, A.5    Minton, N.P.6
  • 36
    • 33845580482 scopus 로고    scopus 로고
    • Toxin A-negative, toxin B-positive Clostridium difficile
    • Drudy D, Fanning S, Kyne L. 2007. Toxin A-negative, toxin B-positive Clostridium difficile. Int J Infect Dis 11: 5-10. https://doi.org/10.1016/ j.ijid.2006.04.003.
    • (2007) Int J Infect Dis , vol.11 , pp. 5-10
    • Drudy, D.1    Fanning, S.2    Kyne, L.3
  • 37
    • 84930965989 scopus 로고    scopus 로고
    • Identification of an epithelial cell receptor responsible for Clostridium difficile TcdB-induced cytotoxicity
    • LaFrance ME, Farrow M, Chandrasekaran R, Sheng J, Rubin DH, Lacy DB. 2015. Identification of an epithelial cell receptor responsible for Clostridium difficile TcdB-induced cytotoxicity. Proc Natl Acad Sci U S A 112: 7073-7078. https://doi.org/10.1073/pnas.1500791112.
    • (2015) Proc Natl Acad Sci U S A , vol.112 , pp. 7073-7078
    • LaFrance, M.E.1    Farrow, M.2    Chandrasekaran, R.3    Sheng, J.4    Rubin, D.H.5    Lacy, D.B.6
  • 38
    • 46449130378 scopus 로고    scopus 로고
    • gp96 is a human colonocyte plasma membrane binding protein for Clostridium difficile toxin A
    • Na X, Kim H, Moyer MP, Pothoulakis C, LaMont JT. 2008. gp96 is a human colonocyte plasma membrane binding protein for Clostridium difficile toxin A. Infect Immun 76: 2862-2871. https://doi.org/10.1128/ IAI.00326-08.
    • (2008) Infect Immun , vol.76 , pp. 2862-2871
    • Na, X.1    Kim, H.2    Moyer, M.P.3    Pothoulakis, C.4    LaMont, J.T.5
  • 39
    • 0026078094 scopus 로고
    • Toxin A of Clostridium difficile binds to the human carbohydrate antigens I, X, and Y
    • Tucker KD, Wilkins TD. 1991. Toxin A of Clostridium difficile binds to the human carbohydrate antigens I, X, and Y. Infect Immun 59: 73-78.
    • (1991) Infect Immun , vol.59 , pp. 73-78
    • Tucker, K.D.1    Wilkins, T.D.2
  • 41
    • 0029054398 scopus 로고
    • Glucosylation of Rho proteins by Clostridium difficile toxin B
    • Just I, Selzer J, Wilm M, von Eichel-Streiber C, Mann M, Aktories K. 1995. Glucosylation of Rho proteins by Clostridium difficile toxin B. Nature 375: 500-503. https://doi.org/10.1038/375500a0.
    • (1995) Nature , vol.375 , pp. 500-503
    • Just, I.1    Selzer, J.2    Wilm, M.3    von Eichel-Streiber, C.4    Mann, M.5    Aktories, K.6
  • 42
    • 0029011449 scopus 로고
    • The enterotoxin from Clostridium difficile (ToxA) monoglucosylates the Rho proteins
    • Just I, Wilm M, Selzer J, Rex G, Von Eichel-Streiber C, Mann M, Aktories K. 1995. The enterotoxin from Clostridium difficile (ToxA) monoglucosylates the Rho proteins. J Biol Chem 270: 13932-13936. https://doi.org/10.1074/jbc.270.23.13932.
    • (1995) J Biol Chem , vol.270 , pp. 13932-13936
    • Just, I.1    Wilm, M.2    Selzer, J.3    Rex, G.4    Von Eichel-Streiber, C.5    Mann, M.6    Aktories, K.7
  • 43
    • 0025367873 scopus 로고
    • Interaction of Clostridium difficile toxin A with cultured cells: Cytoskeletal changes and nuclear polarization
    • Fiorentini C, Malorni W, Paradisi S, Giuliano M, Mastrantonio P, Donelli G. 1990. Interaction of Clostridium difficile toxin A with cultured cells: cytoskeletal changes and nuclear polarization. Infect Immun 58: 2329-2336.
    • (1990) Infect Immun , vol.58 , pp. 2329-2336
    • Fiorentini, C.1    Malorni, W.2    Paradisi, S.3    Giuliano, M.4    Mastrantonio, P.5    Donelli, G.6
  • 45
    • 84891773348 scopus 로고    scopus 로고
    • Clostridium difficile binary toxin CDT: Mechanism, epidemiology, and potential clinical importance
    • Gerding DN, Johnson S, Rupnik M, Aktories K. 2014. Clostridium difficile binary toxin CDT: mechanism, epidemiology, and potential clinical importance. Gut Microbes 5: 15-27. https://doi.org/10.4161/gmic.26854.
    • (2014) Gut Microbes , vol.5 , pp. 15-27
    • Gerding, D.N.1    Johnson, S.2    Rupnik, M.3    Aktories, K.4
  • 46
    • 0023747353 scopus 로고
    • Actin-specific ADPribosyltransferase produced by a Clostridium difficile strain
    • Popoff MR, Rubin EJ, Gill DM, Boquet P. 1988. Actin-specific ADPribosyltransferase produced by a Clostridium difficile strain. Infect Immun 56: 2299-2306.
    • (1988) Infect Immun , vol.56 , pp. 2299-2306
    • Popoff, M.R.1    Rubin, E.J.2    Gill, D.M.3    Boquet, P.4
  • 47
    • 0024204607 scopus 로고
    • Clostridium difficile toxin A perturbs cytoskeletal structure and tight junction permeability of cultured human intestinal epithelial monolayers
    • Hecht G, Pothoulakis C, LaMont JT, Madara JL. 1988. Clostridium difficile toxin A perturbs cytoskeletal structure and tight junction permeability of cultured human intestinal epithelial monolayers. J Clin Invest 82: 1516-1524. https://doi.org/10.1172/JCI113760.
    • (1988) J Clin Invest , vol.82 , pp. 1516-1524
    • Hecht, G.1    Pothoulakis, C.2    LaMont, J.T.3    Madara, J.L.4
  • 48
    • 0035112067 scopus 로고    scopus 로고
    • Clostridium difficile toxins disrupt epithelial barrier function by altering membrane microdomain localization of tight junction proteins
    • Nusrat A, Turner JR, Verkade P, Madara L, Parkos CA. 2001. Clostridium difficile toxins disrupt epithelial barrier function by altering membrane microdomain localization of tight junction proteins. Infect Immun 69: 1329-1336. https://doi.org/10.1128/IAI.69.3.1329-1336.2001.
    • (2001) Infect Immun , vol.69 , pp. 1329-1336
    • Nusrat, A.1    Turner, J.R.2    Verkade, P.3    Madara, L.4    Parkos, C.A.5
  • 49
    • 84898978251 scopus 로고    scopus 로고
    • Clostridium difficile toxins facilitate bacterial colonization by modulating the fence and gate function of colonic epithelium
    • Kasendra M, Barrile R, Leuzzi R, Soriani M. 2014. Clostridium difficile toxins facilitate bacterial colonization by modulating the fence and gate function of colonic epithelium. J Infect Dis 209: 1095-1104. https:// doi.org/10.1093/infdis/jit617.
    • (2014) J Infect Dis , vol.209 , pp. 1095-1104
    • Kasendra, M.1    Barrile, R.2    Leuzzi, R.3    Soriani, M.4
  • 50
    • 84862770598 scopus 로고    scopus 로고
    • Interaction between the intestinal microbiota and host in Clostridium difficile colonization resistance
    • Britton RA, Young VB. 2012. Interaction between the intestinal microbiota and host in Clostridium difficile colonization resistance. Trends Microbiol 20: 313-319. https://doi.org/10.1016/j.tim.2012.04.001.
    • (2012) Trends Microbiol , vol.20 , pp. 313-319
    • Britton, R.A.1    Young, V.B.2
  • 51
    • 84964247557 scopus 로고    scopus 로고
    • Dynamics of the fecal microbiome in patients with recurrent and nonrecurrent Clostridium difficile infection
    • Seekatz A, Rao A, Santhosh K, Young V. 2016. Dynamics of the fecal microbiome in patients with recurrent and nonrecurrent Clostridium difficile infection. Genome Med 8: 47. https://doi.org/10.1186/s13073-016-0298-8.
    • (2016) Genome Med , vol.8 , pp. 47
    • Seekatz, A.1    Rao, A.2    Santhosh, K.3    Young, V.4
  • 52
    • 84979998476 scopus 로고    scopus 로고
    • Gut microbiome predictors of treatment response and recurrence in primary Clostridium difficile infection
    • Khanna S, Montassier E, Schmidt B, Patel R, Knights D, Pardi D, Kashyap P. 2016. Gut microbiome predictors of treatment response and recurrence in primary Clostridium difficile infection. Aliment Pharmacol Ther 44: 715-727. https://doi.org/10.1111/apt.13750.
    • (2016) Aliment Pharmacol Ther , vol.44 , pp. 715-727
    • Khanna, S.1    Montassier, E.2    Schmidt, B.3    Patel, R.4    Knights, D.5    Pardi, D.6    Kashyap, P.7
  • 53
    • 0032726458 scopus 로고    scopus 로고
    • Antibiotics and Clostridium difficile
    • Freeman J, Wilcox MH. 1999. Antibiotics and Clostridium difficile. Microbes Infect 1: 377-384. https://doi.org/10.1016/S1286-4579(99)80054-9.
    • (1999) Microbes Infect , vol.1 , pp. 377-384
    • Freeman, J.1    Wilcox, M.H.2
  • 54
    • 38049084783 scopus 로고    scopus 로고
    • Update on the changing epidemiology of Clostridium difficile-associated disease
    • McFarland L. 2008. Update on the changing epidemiology of Clostridium difficile-associated disease. Nat Clin Pract Gastroenterol Hepatol 5: 40-48. https://doi.org/10.1038/ncpgasthep1029.
    • (2008) Nat Clin Pract Gastroenterol Hepatol , vol.5 , pp. 40-48
    • McFarland, L.1
  • 56
    • 66549102207 scopus 로고    scopus 로고
    • Reproducible community dynamics of the gastrointestinal microbiota following antibiotic perturbation
    • Antonopoulos DA, Huse SM, Morrison HG, Schmidt TM, Sogin ML, Young VB. 2009. Reproducible community dynamics of the gastrointestinal microbiota following antibiotic perturbation. Infect Immun 77: 2367-2375. https://doi.org/10.1128/IAI.01520-08.
    • (2009) Infect Immun , vol.77 , pp. 2367-2375
    • Antonopoulos, D.A.1    Huse, S.M.2    Morrison, H.G.3    Schmidt, T.M.4    Sogin, M.L.5    Young, V.B.6
  • 57
    • 84857067037 scopus 로고    scopus 로고
    • Profound alterations of intestinal microbiota following a single dose of clindamycin results in sustained susceptibility to Clostridium difficile-induced colitis
    • Buffie CG, Jarchum I, Equinda M, Lipuma L, Gobourne A, Viale A, Ubeda C, Xavier J, Pamer EG. 2012. Profound alterations of intestinal microbiota following a single dose of clindamycin results in sustained susceptibility to Clostridium difficile-induced colitis. Infect Immun 80: 62-73. https://doi.org/10.1128/IAI.05496-11.
    • (2012) Infect Immun , vol.80 , pp. 62-73
    • Buffie, C.G.1    Jarchum, I.2    Equinda, M.3    Lipuma, L.4    Gobourne, A.5    Viale, A.6    Ubeda, C.7    Xavier, J.8    Pamer, E.G.9
  • 58
    • 84953837329 scopus 로고    scopus 로고
    • Identification of key taxa that favor intestinal colonization of Clostridium difficile in an adult Chinese population
    • Gu S, Chen Y, Zhang X, Lu H, Lv T, Shen P, Lv L, Zheng B, Jiang X, Li L. 2016. Identification of key taxa that favor intestinal colonization of Clostridium difficile in an adult Chinese population. Microbes Infect 18: 30-38. https://doi.org/10.1016/j.micinf.2015.09.008.
    • (2016) Microbes Infect , vol.18 , pp. 30-38
    • Gu, S.1    Chen, Y.2    Zhang, X.3    Lu, H.4    Lv, T.5    Shen, P.6    Lv, L.7    Zheng, B.8    Jiang, X.9    Li, L.10
  • 59
    • 84961291897 scopus 로고    scopus 로고
    • Insight into alteration of gut microbiota in Clostridium difficile infection and asymptomatic C. difficile colonization
    • Zhang L, Dong D, Jiang C, Li Z, Wang X, Peng Y. 2015. Insight into alteration of gut microbiota in Clostridium difficile infection and asymptomatic C. difficile colonization. Anaerobe 34: 1-7. https:// doi.org/10.1016/j.anaerobe.2015.03.008.
    • (2015) Anaerobe , vol.34 , pp. 1-7
    • Zhang, L.1    Dong, D.2    Jiang, C.3    Li, Z.4    Wang, X.5    Peng, Y.6
  • 60
    • 79952756960 scopus 로고    scopus 로고
    • Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation
    • Dethlefsen L, Relman DA. 2011. Incomplete recovery and individualized responses of the human distal gut microbiota to repeated antibiotic perturbation. Proc Natl Acad Sci U S A 108: 4554-4561. https://doi.org/10.1073/pnas.1000087107.
    • (2011) Proc Natl Acad Sci U S A , vol.108 , pp. 4554-4561
    • Dethlefsen, L.1    Relman, D.A.2
  • 61
    • 84983048071 scopus 로고    scopus 로고
    • Loss of microbiota-mediated colonization resistance to Clostridium difficile infection with oral vancomycin compared with metronidazole
    • Lewis BB, Buffie CG, Carter RA, Leiner I, Toussaint NC, Miller LC, Gobourne A, Ling L, Pamer EG. 2015. Loss of microbiota-mediated colonization resistance to Clostridium difficile infection with oral vancomycin compared with metronidazole. J Infect Dis 212: 1656-1665. https:// doi.org/10.1093/infdis/jiv256.
    • (2015) J Infect Dis , vol.212 , pp. 1656-1665
    • Lewis, B.B.1    Buffie, C.G.2    Carter, R.A.3    Leiner, I.4    Toussaint, N.C.5    Miller, L.C.6    Gobourne, A.7    Ling, L.8    Pamer, E.G.9
  • 63
    • 84964221805 scopus 로고    scopus 로고
    • The role of Clostridium difficile in the paediatric and neonatal gut-a narrative review
    • Lees EA, Miyajima F, Pirmohamed M, Carrol ED. 2016. The role of Clostridium difficile in the paediatric and neonatal gut-a narrative review. Eur J Clin Microbiol Infect Dis 35: 1047-1057. https://doi.org/10.1007/s10096-016-2639-3.
    • (2016) Eur J Clin Microbiol Infect Dis , vol.35 , pp. 1047-1057
    • Lees, E.A.1    Miyajima, F.2    Pirmohamed, M.3    Carrol, E.D.4
  • 64
    • 84888146253 scopus 로고    scopus 로고
    • Clostridium difficile infection in the elderly
    • Keller JM, Surawicz CM. 2014. Clostridium difficile infection in the elderly. Clin Geriatr Med 30: 79-93. https://doi.org/10.1016/ j.cger.2013.10.008.
    • (2014) Clin Geriatr Med , vol.30 , pp. 79-93
    • Keller, J.M.1    Surawicz, C.M.2
  • 66
    • 33644523410 scopus 로고    scopus 로고
    • Clostridium difficile infection in patients discharged from US short-stay hospitals, 1996-2003
    • McDonald LC, Owings M, Jernigan DB. 2006. Clostridium difficile infection in patients discharged from US short-stay hospitals, 1996-2003. Emerg Infect Dis 12: 409-415. https://doi.org/10.3201/eid1203.051064.
    • (2006) Emerg Infect Dis , vol.12 , pp. 409-415
    • McDonald, L.C.1    Owings, M.2    Jernigan, D.B.3
  • 67
    • 0036241355 scopus 로고    scopus 로고
    • Changes in predominant bacterial populations in human faeces with age and with Clostridium difficile infection
    • Hopkins MJ, Macfarlane GT. 2002. Changes in predominant bacterial populations in human faeces with age and with Clostridium difficile infection. J Med Microbiol 51: 448-454. https://doi.org/10.1099/0022-1317-51-5-448.
    • (2002) J Med Microbiol , vol.51 , pp. 448-454
    • Hopkins, M.J.1    Macfarlane, G.T.2
  • 69
    • 0027963265 scopus 로고
    • Immunoglobulin G directed against toxins A and B of Clostridium difficile in the general population and patients with antibiotic associated diarrhea
    • Bacon AEI, Fekety R. 1994. Immunoglobulin G directed against toxins A and B of Clostridium difficile in the general population and patients with antibiotic associated diarrhea. Diagn Microbiol Infect Dis 18: 205-209. https://doi.org/10.1016/0732-8893(94)90021-3.
    • (1994) Diagn Microbiol Infect Dis , vol.18 , pp. 205-209
    • Bacon, A.E.I.1    Fekety, R.2
  • 71
    • 0026527192 scopus 로고
    • Human colonic aspirates containing immunoglobulin A antibody to Clostridium difficile toxin A inhibit toxin A-receptor binding
    • Kelly C, Pothoulakis C, Orellana J, LaMont J. 1992. Human colonic aspirates containing immunoglobulin A antibody to Clostridium difficile toxin A inhibit toxin A-receptor binding. Gastroenterology 102: 35-40. https://doi.org/10.1016/0016-5085(92)91781-X.
    • (1992) Gastroenterology , vol.102 , pp. 35-40
    • Kelly, C.1    Pothoulakis, C.2    Orellana, J.3    LaMont, J.4
  • 72
    • 0025727765 scopus 로고
    • Treatment with intravenously administered gamma globulin of chronic relapsing colitis induced by Clostridium difficile toxin
    • Leung DY, Kelly CP, Boguniewicz M, Pothoulakis C, LaMont JT, Flores A. 1991. Treatment with intravenously administered gamma globulin of chronic relapsing colitis induced by Clostridium difficile toxin. J Pediatr 118: 633-637. https://doi.org/10.1016/S0022-3476(05)83393-1.
    • (1991) J Pediatr , vol.118 , pp. 633-637
    • Leung, D.Y.1    Kelly, C.P.2    Boguniewicz, M.3    Pothoulakis, C.4    LaMont, J.T.5    Flores, A.6
  • 73
    • 0028911365 scopus 로고
    • Gamma globulin administration in relapsing Clostridium difficile-induced pseudomembranous colitis with a defective antibody response to toxin A
    • Warny M, Denie C, Delmee M, Lefebvre C. 1995. Gamma globulin administration in relapsing Clostridium difficile-induced pseudomembranous colitis with a defective antibody response to toxin A. Acta Clin Belg 50: 36-39. https://doi.org/10.1080/17843286.1995.11718419.
    • (1995) Acta Clin Belg , vol.50 , pp. 36-39
    • Warny, M.1    Denie, C.2    Delmee, M.3    Lefebvre, C.4
  • 74
    • 0035915688 scopus 로고    scopus 로고
    • Association between antibody response to toxin A and protection against recurrent Clostridium difficile diarrhoea
    • Kyne L, Warny M, Qamar A, Kelly CP. 2001. Association between antibody response to toxin A and protection against recurrent Clostridium difficile diarrhoea. Lancet 357: 189-193. https://doi.org/10.1016/S0140-6736(00)03592-3.
    • (2001) Lancet , vol.357 , pp. 189-193
    • Kyne, L.1    Warny, M.2    Qamar, A.3    Kelly, C.P.4
  • 75
    • 0026453188 scopus 로고
    • Systemic and mucosal antibody responses to toxin A in patients infected with Clostridium difficile
    • Johnson S, Gerding DN, Janoff EN. 1992. Systemic and mucosal antibody responses to toxin A in patients infected with Clostridium difficile. J Infect Dis 166: 1287-1294. https://doi.org/10.1093/infdis/166.6.1287.
    • (1992) J Infect Dis , vol.166 , pp. 1287-1294
    • Johnson, S.1    Gerding, D.N.2    Janoff, E.N.3
  • 76
    • 0028089161 scopus 로고
    • Human antibody response to Clostridium difficile toxin A in relation to clinical course of infection
    • Warny M, Vaerman JP, Avesani V, Delmée M. 1994. Human antibody response to Clostridium difficile toxin A in relation to clinical course of infection. Infect Immun 62: 384-389.
    • (1994) Infect Immun , vol.62 , pp. 384-389
    • Warny, M.1    Vaerman, J.P.2    Avesani, V.3    Delmée, M.4
  • 77
  • 78
    • 0028997769 scopus 로고
    • Recurrent Clostridium difficile infection in a patient with selective IgG1 deficiency treated with intravenous immune globulin and Saccharomyces boulardii
    • Hassett J, Meyers S, McFarland L, Mulligan ME. 1995. Recurrent Clostridium difficile infection in a patient with selective IgG1 deficiency treated with intravenous immune globulin and Saccharomyces boulardii. Clin Infect Dis 20(Suppl 2): S266-S268. https://doi.org/10.1093/ clinids/20.Supplement_2.S266.
    • (1995) Clin Infect Dis , vol.20 , pp. S266-S268
    • Hassett, J.1    Meyers, S.2    McFarland, L.3    Mulligan, M.E.4
  • 79
    • 2442705389 scopus 로고    scopus 로고
    • Descriptive study of intravenous immunoglobulin for the treatment of recurrent Clostridium difficile diarrhoea
    • Wilcox M. 2004. Descriptive study of intravenous immunoglobulin for the treatment of recurrent Clostridium difficile diarrhoea. J Antimicrob Chemother 53: 882-884. https://doi.org/10.1093/jac/dkh176.
    • (2004) J Antimicrob Chemother , vol.53 , pp. 882-884
    • Wilcox, M.1
  • 80
    • 84901440669 scopus 로고    scopus 로고
    • Intravenous immunoglobulin in the treatment of severe Clostridium difficile colitis
    • Shah N, Shaaban H, Spira R, Slim J, Boghossian J. 2014. Intravenous immunoglobulin in the treatment of severe Clostridium difficile colitis. J Glob Infect Dis 6: 82-85. https://doi.org/10.4103/0974-777X.132053.
    • (2014) J Glob Infect Dis , vol.6 , pp. 82-85
    • Shah, N.1    Shaaban, H.2    Spira, R.3    Slim, J.4    Boghossian, J.5
  • 82
    • 33646582618 scopus 로고    scopus 로고
    • Intravenous immunoglobulin for the treatment of severe, refractory, and recurrent Clostridium difficile diarrhea
    • McPherson S, Rees CJ, Ellis R, Soo S, Panter SJ. 2006. Intravenous immunoglobulin for the treatment of severe, refractory, and recurrent Clostridium difficile diarrhea. Dis Colon Rectum 49: 640-645. https:// doi.org/10.1007/s10350-006-0511-8.
    • (2006) Dis Colon Rectum , vol.49 , pp. 640-645
    • McPherson, S.1    Rees, C.J.2    Ellis, R.3    Soo, S.4    Panter, S.J.5
  • 84
    • 84921767408 scopus 로고    scopus 로고
    • Diagnosis and treatment of Clostridium difficile in adults
    • Bagdasarian N, Rao K, Malani PN. 2015. Diagnosis and treatment of Clostridium difficile in adults. JAMA 313: 398-408. https://doi.org/10.1001/jama.2014.17103.
    • (2015) JAMA , vol.313 , pp. 398-408
    • Bagdasarian, N.1    Rao, K.2    Malani, P.N.3
  • 86
    • 78651111353 scopus 로고    scopus 로고
    • Clostridium difficile infection: Epidemiology, risk factors and management
    • Ananthakrishnan AN. 2011. Clostridium difficile infection: epidemiology, risk factors and management. Nat Rev Gastroenterol Hepatol 8: 17-26. https://doi.org/10.1038/nrgastro.2010.190.
    • (2011) Nat Rev Gastroenterol Hepatol , vol.8 , pp. 17-26
    • Ananthakrishnan, A.N.1
  • 87
    • 84868546746 scopus 로고    scopus 로고
    • Can we identify patients at high risk of recurrent Clostridium difficile infection?
    • Kelly CP. 2012. Can we identify patients at high risk of recurrent Clostridium difficile infection? Clin Microbiol Infect 18(Suppl 6): 21-27. https://doi.org/10.1111/1469-0691.12046.
    • (2012) Clin Microbiol Infect , vol.18 , pp. 21-27
    • Kelly, C.P.1
  • 90
    • 84863644993 scopus 로고    scopus 로고
    • Fidaxomicin preserves the intestinal microbiome during and after treatment of Clostridium difficile infection (CDI) and reduces both toxin reexpression and recurrence of CDI
    • Louie TJ, Cannon K, Byrne B, Emery J, Ward L, Eyben M, Krulicki W. 2012. Fidaxomicin preserves the intestinal microbiome during and after treatment of Clostridium difficile infection (CDI) and reduces both toxin reexpression and recurrence of CDI. Clin Infect Dis 55: S132-S142. https://doi.org/10.1093/cid/cis338.
    • (2012) Clin Infect Dis , vol.55 , pp. S132-S142
    • Louie, T.J.1    Cannon, K.2    Byrne, B.3    Emery, J.4    Ward, L.5    Eyben, M.6    Krulicki, W.7
  • 93
    • 84903954202 scopus 로고    scopus 로고
    • Recovery of the gut microbiome following fecal microbiota transplantation
    • Seekatz AM, Aas J, Gessert CE, Rubin TA, Saman DM, Bakken JS, Young VB. 2014. Recovery of the gut microbiome following fecal microbiota transplantation. mBio 5: e00893-14. https://doi.org/10.1128/mBio.00893-14.
    • (2014) mBio , vol.5 , pp. e00893-e00914
    • Seekatz, A.M.1    Aas, J.2    Gessert, C.E.3    Rubin, T.A.4    Saman, D.M.5    Bakken, J.S.6    Young, V.B.7
  • 94
    • 77951700268 scopus 로고    scopus 로고
    • Changes in the composition of the human fecal microbiome after bacteriotherapy for recurrent Clostridium difficile-associated diarrhea
    • Khoruts A, Dicksved J, Jansson JK, Sadowsky MJ. 2010. Changes in the composition of the human fecal microbiome after bacteriotherapy for recurrent Clostridium difficile-associated diarrhea. J Clin Gastroenterol 44: 354-360. https://doi.org/10.1097/MCG.0b013e3181c87e02.
    • (2010) J Clin Gastroenterol , vol.44 , pp. 354-360
    • Khoruts, A.1    Dicksved, J.2    Jansson, J.K.3    Sadowsky, M.J.4
  • 98
    • 84892629673 scopus 로고    scopus 로고
    • Faecal microbiota transplantation and bacteriotherapy for recurrent Clostridium difficile infection: A retrospective evaluation of 31 patients
    • Emanuelsson F, Claesson BEB, Ljungström L, Tvede M, Ung K-A. 2014. Faecal microbiota transplantation and bacteriotherapy for recurrent Clostridium difficile infection: a retrospective evaluation of 31 patients. Scand J Infect Dis 46: 89-97. https://doi.org/10.3109/00365548.2013.858181.
    • (2014) Scand J Infect Dis , vol.46 , pp. 89-97
    • Emanuelsson, F.1    Claesson, B.E.B.2    Ljungström, L.3    Tvede, M.4    Ung, K.-A.5
  • 99
    • 0024312284 scopus 로고
    • Bacteriotherapy for chronic relapsing Clostridium difficile diarrhoea in six patients
    • Tvede M, Rask-Madsen J. 1989. Bacteriotherapy for chronic relapsing Clostridium difficile diarrhoea in six patients. Lancet 333: 1156-1160. https://doi.org/10.1016/S0140-6736(89)92749-9.
    • (1989) Lancet , vol.333 , pp. 1156-1160
    • Tvede, M.1    Rask-Madsen, J.2
  • 102
    • 84938903548 scopus 로고    scopus 로고
    • Fecal microbiota transplantation and bacterial consortium transplantation have comparable effects on the re-establishment of mucosal barrier function in mice with intestinal dysbiosis
    • Li M, Liang P, Li Z, Wang Y, Zhang G, Gao H, Wen S, Tang L. 2015. Fecal microbiota transplantation and bacterial consortium transplantation have comparable effects on the re-establishment of mucosal barrier function in mice with intestinal dysbiosis. Front Microbiol 6: 692. https:// doi.org/10.3389/fmicb.2015.00692.
    • (2015) Front Microbiol , vol.6 , pp. 692
    • Li, M.1    Liang, P.2    Li, Z.3    Wang, Y.4    Zhang, G.5    Gao, H.6    Wen, S.7    Tang, L.8
  • 103
    • 84959483325 scopus 로고    scopus 로고
    • Probiotics are effective at preventing Clostridium difficile-associated diarrhea: A systematic review and metaanalysis
    • Lau CSM, Chamberlain RS. 2016. Probiotics are effective at preventing Clostridium difficile-associated diarrhea: a systematic review and metaanalysis. Int J Gen Med 9: 27-37. https://doi.org/10.2147/IJGM.S98280.
    • (2016) Int J Gen Med , vol.9 , pp. 27-37
    • Lau, C.S.M.1    Chamberlain, R.S.2
  • 105
    • 22744446859 scopus 로고    scopus 로고
    • Probiotic therapy for the prevention and treatment of Clostridium difficile-associated diarrhea: A systematic review
    • Dendukuri N, Costa V, McGregor M, Brophy JM. 2005. Probiotic therapy for the prevention and treatment of Clostridium difficile-associated diarrhea: a systematic review. CMAJ 173: 167-170. https://doi.org/10.1503/cmaj.050350.
    • (2005) CMAJ , vol.173 , pp. 167-170
    • Dendukuri, N.1    Costa, V.2    McGregor, M.3    Brophy, J.M.4
  • 106
    • 84887390865 scopus 로고    scopus 로고
    • Lactobacilli and bifidobacteria in the prevention of antibiotic-associated diarrhoea and Clostridium difficile diarrhoea in older inpatients (PLACIDE): A randomised, double-blind, placebo-controlled, multicentre trial
    • Allen SJ, Wareham K, Wang D, Bradley C, Hutchings H, Harris W, Dhar A, Brown H, Foden A, Gravenor MB, Mack D. 2013. Lactobacilli and bifidobacteria in the prevention of antibiotic-associated diarrhoea and Clostridium difficile diarrhoea in older inpatients (PLACIDE): a randomised, double-blind, placebo-controlled, multicentre trial. Lancet 382: 1249-1257. https://doi.org/10.1016/S0140-6736(13)61218-0.
    • (2013) Lancet , vol.382 , pp. 1249-1257
    • Allen, S.J.1    Wareham, K.2    Wang, D.3    Bradley, C.4    Hutchings, H.5    Harris, W.6    Dhar, A.7    Brown, H.8    Foden, A.9    Gravenor, M.B.10    Mack, D.11
  • 107
    • 33846042589 scopus 로고    scopus 로고
    • Inappropriate use of meta-analysis to estimate efficacy of probiotics
    • Dendukuri N, Brophy J. 2007. Inappropriate use of meta-analysis to estimate efficacy of probiotics. Am J Gastroenterol 102: 201. https:// doi.org/10.1111/j.1572-0241.2007.00916.x.
    • (2007) Am J Gastroenterol , vol.102 , pp. 201
    • Dendukuri, N.1    Brophy, J.2
  • 108
    • 33846047427 scopus 로고    scopus 로고
    • Response to "Meta-analysis of probiotics for the prevention of antibiotic-associated diarrhea and the treatment of Clostridium difficile disease". Am J Gastroenterol 2006;101: 812-822
    • Lewis S. 2007. Response to "Meta-analysis of probiotics for the prevention of antibiotic-associated diarrhea and the treatment of Clostridium difficile disease". Am J Gastroenterol 2006;101: 812-822. Am J Gastroenterol 102: 201-202. https://doi.org/10.1111/j.1572-0241.2007.00916.x.
    • (2007) Am J Gastroenterol , vol.102 , pp. 201-202
    • Lewis, S.1
  • 109
    • 42449115458 scopus 로고    scopus 로고
    • Probiotics for treatment of Clostridium difficileassociated colitis in adults
    • Pillai A, Nelson R. 2008. Probiotics for treatment of Clostridium difficileassociated colitis in adults. Cochrane Database Syst Rev 1: CD004611. https://doi.org/10.1002/14651858.CD004611.pub2.
    • (2008) Cochrane Database Syst Rev , vol.1
    • Pillai, A.1    Nelson, R.2
  • 111
    • 80051717743 scopus 로고    scopus 로고
    • The interplay between microbiome dynamics and pathogen dynamics in a murine model of Clostridium difficile infection
    • Reeves AE, Theriot CM, Bergin IL, Huffnagle GB, Schloss PD, Young VB. 2011. The interplay between microbiome dynamics and pathogen dynamics in a murine model of Clostridium difficile infection. Gut Microbes 2: 145-158. https://doi.org/10.4161/gmic.2.3.16333.
    • (2011) Gut Microbes , vol.2 , pp. 145-158
    • Reeves, A.E.1    Theriot, C.M.2    Bergin, I.L.3    Huffnagle, G.B.4    Schloss, P.D.5    Young, V.B.6
  • 112
    • 84940868812 scopus 로고    scopus 로고
    • Antibiotic-induced alterations of the murine gut microbiota and subsequent effects on colonization resistance against Clostridium difficile
    • Schubert AM, Sinani H, Schloss PD. 2015. Antibiotic-induced alterations of the murine gut microbiota and subsequent effects on colonization resistance against Clostridium difficile. mBio 6: e00974-15. https:// doi.org/10.1128/mBio.00974-15.
    • (2015) mBio , vol.6 , pp. e00974-e001015
    • Schubert, A.M.1    Sinani, H.2    Schloss, P.D.3
  • 114
    • 78650370258 scopus 로고    scopus 로고
    • Comparative metagenomic study of alterations to the intestinal microbiota and risk of nosocomial Clostridum difficileassociated disease
    • Manges AR, Labbe A, Loo VG, Atherton JK, Behr MA, Masson L, Tellis PA, Brousseau R. 2010. Comparative metagenomic study of alterations to the intestinal microbiota and risk of nosocomial Clostridum difficileassociated disease. J Infect Dis 202: 1877-1884. https://doi.org/10.1086/657319.
    • (2010) J Infect Dis , vol.202 , pp. 1877-1884
    • Manges, A.R.1    Labbe, A.2    Loo, V.G.3    Atherton, J.K.4    Behr, M.A.5    Masson, L.6    Tellis, P.A.7    Brousseau, R.8
  • 115
    • 84904012195 scopus 로고    scopus 로고
    • Microbiome data distinguish patients with Clostridium difficile infection and non-C. difficile-associated diarrhea from healthy controls
    • Schubert AM, Rogers MA, Ring C, Mogle J, Petrosino JP, Young VB, Aronoff DM, Schloss PD. 2014. Microbiome data distinguish patients with Clostridium difficile infection and non-C. difficile-associated diarrhea from healthy controls. mBio 5: e01021-14. https://doi.org/10.1128/ mBio.01021-14.
    • (2014) mBio , vol.5 , pp. e01021-e01114
    • Schubert, A.M.1    Rogers, M.A.2    Ring, C.3    Mogle, J.4    Petrosino, J.P.5    Young, V.B.6    Aronoff, D.M.7    Schloss, P.D.8
  • 116
    • 84868361276 scopus 로고    scopus 로고
    • Toward an understanding of changes in diversity associated with fecal microbiome transplantation based on 16S rRNA gene deep sequencing
    • Shahinas D, Silverman M, Sittler T, Chiu C, Kim P, Allen-Vercoe E, Weese S, Wong A, Pillai DR. 2012. Toward an understanding of changes in diversity associated with fecal microbiome transplantation based on 16S rRNA gene deep sequencing. mBio 3: e00338-12. https://doi.org/10.1128/mBio.00338-12.
    • (2012) mBio , vol.3 , pp. e00338-e00412
    • Shahinas, D.1    Silverman, M.2    Sittler, T.3    Chiu, C.4    Kim, P.5    Allen-Vercoe, E.6    Weese, S.7    Wong, A.8    Pillai, D.R.9
  • 117
    • 84896708902 scopus 로고    scopus 로고
    • Microbiota dynamics in patients treated with fecal microbiota transplantation for recurrent Clostridium difficile infection
    • Song Y, Garg S, Girotra M, Maddox C, von Rosenvinge EC, Dutta A, Dutta S, Fricke WF. 2013. Microbiota dynamics in patients treated with fecal microbiota transplantation for recurrent Clostridium difficile infection. PLoS One 8: e81330. https://doi.org/10.1371/journal.pone.0081330.
    • (2013) PLoS One , vol.8
    • Song, Y.1    Garg, S.2    Girotra, M.3    Maddox, C.4    von Rosenvinge, E.C.5    Dutta, A.6    Dutta, S.7    Fricke, W.F.8
  • 118
    • 84882779621 scopus 로고    scopus 로고
    • Intestinal dysbiosis and depletion of butyrogenic bacteria in Clostridium difficile infection and nosocomial diarrhea
    • Antharam VC, Li EC, Ishmael A, Sharma A, Mai V, Rand KH, Wang GP. 2013. Intestinal dysbiosis and depletion of butyrogenic bacteria in Clostridium difficile infection and nosocomial diarrhea. J Clin Microbiol 51: 2884-2892. https://doi.org/10.1128/JCM.00845-13.
    • (2013) J Clin Microbiol , vol.51 , pp. 2884-2892
    • Antharam, V.C.1    Li, E.C.2    Ishmael, A.3    Sharma, A.4    Mai, V.5    Rand, K.H.6    Wang, G.P.7
  • 120
    • 84883861314 scopus 로고    scopus 로고
    • Finding a sugary foothold: How antibiotics pave the way for enteric pathogens
    • Stecher B. 2013. Finding a sugary foothold: how antibiotics pave the way for enteric pathogens. Cell Host Microbe 14: 225-227. https:// doi.org/10.1016/j.chom.2013.08.008.
    • (2013) Cell Host Microbe , vol.14 , pp. 225-227
    • Stecher, B.1
  • 124
    • 84920621547 scopus 로고    scopus 로고
    • Gut microbiota-produced succinate promotes C. difficile infection after antibiotic treatment or motility disturbance
    • Ferreyra JA, Wu KJ, Hryckowian AJ, Bouley DM, Weimer BC, Sonnenburg JL. 2014. Gut microbiota-produced succinate promotes C. difficile infection after antibiotic treatment or motility disturbance. Cell Host Microbe 16: 770-777. https://doi.org/10.1016/j.chom.2014.11.003.
    • (2014) Cell Host Microbe , vol.16 , pp. 770-777
    • Ferreyra, J.A.1    Wu, K.J.2    Hryckowian, A.J.3    Bouley, D.M.4    Weimer, B.C.5    Sonnenburg, J.L.6
  • 125
    • 0032574193 scopus 로고    scopus 로고
    • Primary symptomless colonisation by Clostridium difficile and decreased risk of subsequent diarrhoea
    • Shim JK, Johnson S, Samore MH, Bliss DZ, Gerding DN. 1998. Primary symptomless colonisation by Clostridium difficile and decreased risk of subsequent diarrhoea. Lancet 351: 633-636. https://doi.org/10.1016/ S0140-6736(97)08062-8.
    • (1998) Lancet , vol.351 , pp. 633-636
    • Shim, J.K.1    Johnson, S.2    Samore, M.H.3    Bliss, D.Z.4    Gerding, D.N.5
  • 126
    • 84885915939 scopus 로고    scopus 로고
    • Nontoxigenic Clostridium difficile protects hamsters against challenge with historic and epidemic strains of toxigenic BI/NAP1/027 C. difficile
    • Nagaro KJ, Phillips ST, Cheknis AK, Sambol SP, Zukowski WE, Johnson S, Gerding DN. 2013. Nontoxigenic Clostridium difficile protects hamsters against challenge with historic and epidemic strains of toxigenic BI/NAP1/027 C. difficile. Antimicrob Agents Chemother 57: 5266-5270. https://doi.org/10.1128/AAC.00580-13.
    • (2013) Antimicrob Agents Chemother , vol.57 , pp. 5266-5270
    • Nagaro, K.J.1    Phillips, S.T.2    Cheknis, A.K.3    Sambol, S.P.4    Zukowski, W.E.5    Johnson, S.6    Gerding, D.N.7
  • 127
    • 0037115014 scopus 로고    scopus 로고
    • Colonization for the prevention of Clostridium difficile disease in hamsters
    • Sambol SP, Merrigan MM, Tang JK, Johnson S, Gerding DN. 2002. Colonization for the prevention of Clostridium difficile disease in hamsters. J Infect Dis 186: 1781-1789. https://doi.org/10.1086/345676.
    • (2002) J Infect Dis , vol.186 , pp. 1781-1789
    • Sambol, S.P.1    Merrigan, M.M.2    Tang, J.K.3    Johnson, S.4    Gerding, D.N.5
  • 128
    • 84866313203 scopus 로고    scopus 로고
    • Evaluation of an oral suspension of VP20621, spores of nontoxigenic Clostridium difficile strain M3, in healthy subjects
    • Villano SA, Seiberling M, Tatarowicz W, Monnot-Chase E, Gerding DN. 2012. Evaluation of an oral suspension of VP20621, spores of nontoxigenic Clostridium difficile strain M3, in healthy subjects. Antimicrob Agents Chemother 56: 5224-5229. https://doi.org/10.1128/AAC.00913-12.
    • (2012) Antimicrob Agents Chemother , vol.56 , pp. 5224-5229
    • Villano, S.A.1    Seiberling, M.2    Tatarowicz, W.3    Monnot-Chase, E.4    Gerding, D.N.5
  • 130
    • 84886255455 scopus 로고    scopus 로고
    • Horizontal gene transfer converts non-toxigenic Clostridium difficile strains into toxin producers
    • Brouwer MSM, Roberts AP, Hussain H, Williams RJ, Allan E, Mullany P. 2013. Horizontal gene transfer converts non-toxigenic Clostridium difficile strains into toxin producers. Nat Commun 4: 2601. https://doi.org/10.1038/ncomms3601.
    • (2013) Nat Commun , vol.4 , pp. 2601
    • Brouwer, M.S.M.1    Roberts, A.P.2    Hussain, H.3    Williams, R.J.4    Allan, E.5    Mullany, P.6
  • 131
    • 33244467651 scopus 로고    scopus 로고
    • Bile salt biotransformations by human intestinal bacteria
    • Ridlon JM. 2005. Bile salt biotransformations by human intestinal bacteria. J Lipid Res 47: 241-259. https://doi.org/10.1194/jlr.R500013-JLR200.
    • (2005) J Lipid Res , vol.47 , pp. 241-259
    • Ridlon, J.M.1
  • 132
    • 0020554353 scopus 로고
    • Efficiency of various bile salt preparations for stimulation of Clostridium difficile spore germination
    • Wilson KH. 1983. Efficiency of various bile salt preparations for stimulation of Clostridium difficile spore germination. J Clin Microbiol 18: 1017-1019.
    • (1983) J Clin Microbiol , vol.18 , pp. 1017-1019
    • Wilson, K.H.1
  • 133
    • 77649282955 scopus 로고    scopus 로고
    • Metabolism of bile salts in mice influences spore germination in Clostridium difficile
    • Giel JL, Sorg JA, Sonenshein AL, Zhu J. 2010. Metabolism of bile salts in mice influences spore germination in Clostridium difficile. PLoS One 5: e8740. https://doi.org/10.1371/journal.pone.0008740.
    • (2010) PLoS One , vol.5
    • Giel, J.L.1    Sorg, J.A.2    Sonenshein, A.L.3    Zhu, J.4
  • 134
    • 84903785994 scopus 로고    scopus 로고
    • Metabolomics analysis identifies intestinal microbiota-derived biomarkers of colonization resistance in clindamycin-treated mice
    • Jump RLP, Polinkovsky A, Hurless K, Sitzlar B, Eckart K, Tomas M, Deshpande A, Nerandzic MM, Donskey CJ. 2014. Metabolomics analysis identifies intestinal microbiota-derived biomarkers of colonization resistance in clindamycin-treated mice. PLoS One 9: e101267. https:// doi.org/10.1371/journal.pone.0101267.
    • (2014) PLoS One , vol.9
    • Jump, R.L.P.1    Polinkovsky, A.2    Hurless, K.3    Sitzlar, B.4    Eckart, K.5    Tomas, M.6    Deshpande, A.7    Nerandzic, M.M.8    Donskey, C.J.9
  • 137
    • 84875624651 scopus 로고    scopus 로고
    • In vitro inhibition of Clostridium difficile and Clostridium perfringens by commercial probiotic strains
    • Schoster A, Kokotovic B, Permin A, Pedersen PD, Bello FD, Guardabassi L. 2013. In vitro inhibition of Clostridium difficile and Clostridium perfringens by commercial probiotic strains. Anaerobe 20: 36-41. https://doi.org/10.1016/j.anaerobe.2013.02.006.
    • (2013) Anaerobe , vol.20 , pp. 36-41
    • Schoster, A.1    Kokotovic, B.2    Permin, A.3    Pedersen, P.D.4    Bello, F.D.5    Guardabassi, L.6
  • 138
    • 84867330378 scopus 로고    scopus 로고
    • In vitro evaluation of the antimicrobial activity of a range of probiotics against pathogens: Evidence for the effects of organic acids
    • Tejero-Sariñena S, Barlow J, Costabile A, Gibson GR, Rowland I. 2012. In vitro evaluation of the antimicrobial activity of a range of probiotics against pathogens: evidence for the effects of organic acids. Anaerobe 18: 530-538. https://doi.org/10.1016/j.anaerobe.2012.08.004.
    • (2012) Anaerobe , vol.18 , pp. 530-538
    • Tejero-Sariñena, S.1    Barlow, J.2    Costabile, A.3    Gibson, G.R.4    Rowland, I.5
  • 139
    • 33748302984 scopus 로고    scopus 로고
    • Inhibition of Clostridium difficile growth and adhesion to enterocytes by Bifidobacterium supernatants
    • Trejo FM, Minnaard J, Perez PF, De Antoni GL. 2006. Inhibition of Clostridium difficile growth and adhesion to enterocytes by Bifidobacterium supernatants. Anaerobe 12: 186-193. https://doi.org/10.1016/ j.anaerobe.2006.03.004.
    • (2006) Anaerobe , vol.12 , pp. 186-193
    • Trejo, F.M.1    Minnaard, J.2    Perez, P.F.3    De Antoni, G.L.4
  • 140
    • 84937426631 scopus 로고    scopus 로고
    • Bacillus amyloliquefaciens as prophylactic treatment for Clostridium difficile-associated disease in a mouse model
    • Geeraerts S, Ducatelle R, Haesebrouck F, Van Immerseel F. 2015. Bacillus amyloliquefaciens as prophylactic treatment for Clostridium difficile-associated disease in a mouse model. J Gastroenterol Hepatol 30: 1275-1280. https://doi.org/10.1111/jgh.12957.
    • (2015) J Gastroenterol Hepatol , vol.30 , pp. 1275-1280
    • Geeraerts, S.1    Ducatelle, R.2    Haesebrouck, F.3    Van Immerseel, F.4
  • 141
    • 34447250021 scopus 로고    scopus 로고
    • Antimicrobial activity of lacticin 3147 against clinical Clostridium difficile strains
    • Rea MC, Clayton E, O’Connor PM, Shanahan F, Kiely B, Ross RP, Hill C. 2007. Antimicrobial activity of lacticin 3147 against clinical Clostridium difficile strains. J Med Microbiol 56: 940-946. https://doi.org/10.1099/ jmm.0.47085-0.
    • (2007) J Med Microbiol , vol.56 , pp. 940-946
    • Rea, M.C.1    Clayton, E.2    O’Connor, P.M.3    Shanahan, F.4    Kiely, B.5    Ross, R.P.6    Hill, C.7
  • 142
    • 0029670684 scopus 로고    scopus 로고
    • An application in cheddar cheese manufacture for a strain of Lactococcus lactis producing a novel broad-spectrum bacteriocin, lacticin 3147
    • Ryan MP, Rea MC, Hill C, Ross RP. 1996. An application in cheddar cheese manufacture for a strain of Lactococcus lactis producing a novel broad-spectrum bacteriocin, lacticin 3147. Appl Environ Microbiol 62: 612-619.
    • (1996) Appl Environ Microbiol , vol.62 , pp. 612-619
    • Ryan, M.P.1    Rea, M.C.2    Hill, C.3    Ross, R.P.4
  • 145
    • 10544226189 scopus 로고    scopus 로고
    • Saccharomyces boulardii protease inhibits Clostridium difficile toxin A effects in the rat ileum
    • Castagliuolo I, LaMont JT, Nikulasson ST, Pothoulakis C. 1996. Saccharomyces boulardii protease inhibits Clostridium difficile toxin A effects in the rat ileum. Infect Immun 64: 5225-5232.
    • (1996) Infect Immun , vol.64 , pp. 5225-5232
    • Castagliuolo, I.1    LaMont, J.T.2    Nikulasson, S.T.3    Pothoulakis, C.4
  • 146
    • 0032919855 scopus 로고    scopus 로고
    • Saccharomyces boulardii protease inhibits the effects of Clostridium difficile toxins A and B in human colonic mucosa
    • Castagliuolo I, Riegler MF, Valenick L, LaMont JT, Pothoulakis C. 1999. Saccharomyces boulardii protease inhibits the effects of Clostridium difficile toxins A and B in human colonic mucosa. Infect Immun 67: 302-307.
    • (1999) Infect Immun , vol.67 , pp. 302-307
    • Castagliuolo, I.1    Riegler, M.F.2    Valenick, L.3    LaMont, J.T.4    Pothoulakis, C.5
  • 147
    • 0026523844 scopus 로고
    • Effect of oral Saccharomyces boulardii treatment on the activity of Clostridium difficile toxins in mouse digestive tract
    • Corthier G, Lucas F, Jouvert S, Castex F. 1992. Effect of oral Saccharomyces boulardii treatment on the activity of Clostridium difficile toxins in mouse digestive tract. Toxicon 30: 1583-1589. https://doi.org/10.1016/0041-0101(92)90030-9.
    • (1992) Toxicon , vol.30 , pp. 1583-1589
    • Corthier, G.1    Lucas, F.2    Jouvert, S.3    Castex, F.4
  • 148
    • 0035077712 scopus 로고    scopus 로고
    • Saccharomyces boulardii stimulates intestinal immunoglobulin A immune response to Clostridium difficile toxin A in mice
    • Qamar A, Aboudola S, Warny M, Michetti P, Pothoulakis C, LaMont JT, Kelly CP. 2001. Saccharomyces boulardii stimulates intestinal immunoglobulin A immune response to Clostridium difficile toxin A in mice. Infect Immun 69: 2762-2765. https://doi.org/10.1128/IAI.69.4.2762-2765.2001.
    • (2001) Infect Immun , vol.69 , pp. 2762-2765
    • Qamar, A.1    Aboudola, S.2    Warny, M.3    Michetti, P.4    Pothoulakis, C.5    LaMont, J.T.6    Kelly, C.P.7
  • 150
    • 84936873049 scopus 로고    scopus 로고
    • A combination of three fully human toxin A-and toxin B-specific monoclonal antibodies protects against challenge with highly virulent epidemic strains of Clostridium difficile in the hamster model
    • Anosova NG, Cole LE, Li L, Zhang J, Brown AM, Mundle S, Zhang J, Ray S, Ma F, Garrone P, Bertraminelli N, Kleanthous H, Anderson SF. 2015. A combination of three fully human toxin A-and toxin B-specific monoclonal antibodies protects against challenge with highly virulent epidemic strains of Clostridium difficile in the hamster model. Clin Vaccine Immunol 22: 711-725. https://doi.org/10.1128/CVI.00763-14.
    • (2015) Clin Vaccine Immunol , vol.22 , pp. 711-725
    • Anosova, N.G.1    Cole, L.E.2    Li, L.3    Zhang, J.4    Brown, A.M.5    Mundle, S.6    Zhang, J.7    Ray, S.8    Ma, F.9    Garrone, P.10    Bertraminelli, N.11    Kleanthous, H.12    Anderson, S.F.13
  • 152
    • 0025181832 scopus 로고
    • Stimulation of secretory IgA and secretory component of immunoglobulins in small intestine of rats treated with Saccharomyces boulardii
    • Buts JP, Bernasconi P, Vaerman JP, Dive C. 1990. Stimulation of secretory IgA and secretory component of immunoglobulins in small intestine of rats treated with Saccharomyces boulardii. Dig Dis Sci 35: 251-256. https://doi.org/10.1007/BF01536771.
    • (1990) Dig Dis Sci , vol.35 , pp. 251-256
    • Buts, J.P.1    Bernasconi, P.2    Vaerman, J.P.3    Dive, C.4
  • 153
    • 69449102096 scopus 로고    scopus 로고
    • Comparative study of Bifidobacterium animalis, Escherichia coli, Lactobacillus casei and Saccharomyces boulardii probiotic properties
    • Martins FS, Silva AA, Vieira AT, Barbosa FH, Arantes RM, Teixeira MM, Nicoli JR. 2009. Comparative study of Bifidobacterium animalis, Escherichia coli, Lactobacillus casei and Saccharomyces boulardii probiotic properties. Arch Microbiol 191: 623-630. https://doi.org/10.1007/ s00203-009-0491-x.
    • (2009) Arch Microbiol , vol.191 , pp. 623-630
    • Martins, F.S.1    Silva, A.A.2    Vieira, A.T.3    Barbosa, F.H.4    Arantes, R.M.5    Teixeira, M.M.6    Nicoli, J.R.7
  • 155
    • 80052540085 scopus 로고    scopus 로고
    • Protection against increased intestinal permeability and bacterial translocation induced by intestinal obstruction in mice treated with viable and heat-killed Saccharomyces boulardii
    • Generoso SV, Viana ML, Santos RG, Arantes RME, Martins FS, Nicoli JR, Machado JAN, Correia MITD, Cardoso VN. 2011. Protection against increased intestinal permeability and bacterial translocation induced by intestinal obstruction in mice treated with viable and heat-killed Saccharomyces boulardii. Eur J Nutr 50: 261-269. https://doi.org/10.1007/s00394-010-0134-7.
    • (2011) Eur J Nutr , vol.50 , pp. 261-269
    • Generoso, S.V.1    Viana, M.L.2    Santos, R.G.3    Arantes, R.M.E.4    Martins, F.S.5    Nicoli, J.R.6    Machado, J.A.N.7    Correia, M.I.T.D.8    Cardoso, V.N.9
  • 156
    • 0347986893 scopus 로고    scopus 로고
    • Prebiotic inulin enriched with oligofructose in combination with the probiotics Lactobacillus rhamnosus and Bifidobacterium lactis modulates intestinal immune functions in rats
    • Roller M, Rechkemmer G, Watzl B. 2004. Prebiotic inulin enriched with oligofructose in combination with the probiotics Lactobacillus rhamnosus and Bifidobacterium lactis modulates intestinal immune functions in rats. J Nutr 134: 153-156.
    • (2004) J Nutr , vol.134 , pp. 153-156
    • Roller, M.1    Rechkemmer, G.2    Watzl, B.3
  • 157
    • 84904276018 scopus 로고    scopus 로고
    • Improvement in Th1/Th2 immune homeostasis, antioxidative status and resistance to pathogenic E. coli on consumption of probiotic Lactobacillus rhamnosus fermented milk in aging mice
    • Sharma R, Kapila R, Dass G, Kapila S. 2014. Improvement in Th1/Th2 immune homeostasis, antioxidative status and resistance to pathogenic E. coli on consumption of probiotic Lactobacillus rhamnosus fermented milk in aging mice. Age 36: 9686. https://doi.org/10.1007/ s11357-014-9686-4.
    • (2014) Age , vol.36 , pp. 9686
    • Sharma, R.1    Kapila, R.2    Dass, G.3    Kapila, S.4
  • 158
    • 33644928317 scopus 로고    scopus 로고
    • The probiotic bacterium Lactobacillus casei induces activation of the gut mucosal immune system through innate immunity
    • Galdeano CM, Perdigo G. 2006. The probiotic bacterium Lactobacillus casei induces activation of the gut mucosal immune system through innate immunity. Clin Vaccine Immunol 13: 219-226. https://doi.org/10.1128/CVI.13.2.219-226.2006.
    • (2006) Clin Vaccine Immunol , vol.13 , pp. 219-226
    • Galdeano, C.M.1    Perdigo, G.2
  • 159
    • 77958170401 scopus 로고    scopus 로고
    • Intestinal goblet cells and mucins in health and disease: Recent insights and progress
    • Kim YS, Ho SB. 2010. Intestinal goblet cells and mucins in health and disease: recent insights and progress. Curr Gastroenterol Rep 12: 319-330. https://doi.org/10.1007/s11894-010-0131-2.
    • (2010) Curr Gastroenterol Rep , vol.12 , pp. 319-330
    • Kim, Y.S.1    Ho, S.B.2
  • 160
    • 54449083567 scopus 로고    scopus 로고
    • The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria
    • Johansson M, Phillipson M, Petersson J, Velcich A, Holm L, Hansson GC. 2008. The inner of the two Muc2 mucin-dependent mucus layers in colon is devoid of bacteria. Proc Natl Acad Sci U S A 105: 15064-15069. https://doi.org/10.1073/pnas.0803124105.
    • (2008) Proc Natl Acad Sci U S A , vol.105 , pp. 15064-15069
    • Johansson, M.1    Phillipson, M.2    Petersson, J.3    Velcich, A.4    Holm, L.5    Hansson, G.C.6
  • 161
    • 79952748335 scopus 로고    scopus 로고
    • The two mucus layers of colon are organized by the MUC2 mucin, whereas the outer layer is a legislator of host-microbial interactions
    • Johansson M, Larsson JMH, Hansson GC. 2011. The two mucus layers of colon are organized by the MUC2 mucin, whereas the outer layer is a legislator of host-microbial interactions. Proc Natl Acad Sci U S A 108: 4659-4665. https://doi.org/10.1073/pnas.1006451107.
    • (2011) Proc Natl Acad Sci U S A , vol.108 , pp. 4659-4665
    • Johansson, M.1    Larsson, J.M.H.2    Hansson, G.C.3
  • 163
    • 0027537914 scopus 로고
    • Identification and characterization of adhesive factors of Clostridium difficile involved in adhesion to human colonic enterocyte-like Caco-2 and mucus-secreting HT29 cells in culture
    • Eveillard M, Fourel V, Barc MC, Kernéis S, Coconnier MH, Karjalainen T, Bourlioux P, Servin A. 1993. Identification and characterization of adhesive factors of Clostridium difficile involved in adhesion to human colonic enterocyte-like Caco-2 and mucus-secreting HT29 cells in culture. Mol Microbiol 7: 371-381. https://doi.org/10.1111/j.1365-2958.1993.tb01129.x.
    • (1993) Mol Microbiol , vol.7 , pp. 371-381
    • Eveillard, M.1    Fourel, V.2    Barc, M.C.3    Kernéis, S.4    Coconnier, M.H.5    Karjalainen, T.6    Bourlioux, P.7    Servin, A.8
  • 164
    • 0036785598 scopus 로고    scopus 로고
    • Binding of Clostridium difficile surface layer proteins to gastrointestinal tissues
    • Calabi E, Calabi F, Phillips AD, Fairweather NF, Neil F. 2002. Binding of Clostridium difficile surface layer proteins to gastrointestinal tissues. Infect Immun 70: 5770-5778. https://doi.org/10.1128/IAI.70.10.5770-5778.2002.
    • (2002) Infect Immun , vol.70 , pp. 5770-5778
    • Calabi, E.1    Calabi, F.2    Phillips, A.D.3    Fairweather, N.F.4    Neil, F.5
  • 165
    • 0022525082 scopus 로고
    • Cell surface binding site for Clostridium difficile enterotoxin: Evidence for a glycoconjugate containing the sequence Gala1-3Galb1-4GlcNAc
    • Krivan HC, Clark GF, Smith DF, Wilkins TD. 1986. Cell surface binding site for Clostridium difficile enterotoxin: evidence for a glycoconjugate containing the sequence Gala1-3Galb1-4GlcNAc. Infect Immun 53: 573-581.
    • (1986) Infect Immun , vol.53 , pp. 573-581
    • Krivan, H.C.1    Clark, G.F.2    Smith, D.F.3    Wilkins, T.D.4
  • 166
    • 0030730630 scopus 로고    scopus 로고
    • Clostridium difficile toxin A binding to human intestinal epithelial cells
    • Smith J, Cooke DL, Hyde S, Borriello SP, Long RG. 1997. Clostridium difficile toxin A binding to human intestinal epithelial cells. J Med Microbiol 46: 953-958. https://doi.org/10.1099/00222615-46-11-953.
    • (1997) J Med Microbiol , vol.46 , pp. 953-958
    • Smith, J.1    Cooke, D.L.2    Hyde, S.3    Borriello, S.P.4    Long, R.G.5
  • 169
    • 0038237678 scopus 로고    scopus 로고
    • Extracellular MUC3 mucin secretion follows adherence of Lactobacillus strains to intestinal epithelial cells in vitro
    • Mack D, Ahrne S, Hyde L, Wei S, Hollingsworth M. 2003. Extracellular MUC3 mucin secretion follows adherence of Lactobacillus strains to intestinal epithelial cells in vitro. Gut 52: 827-833. https://doi.org/10.1136/gut.52.6.827.
    • (2003) Gut , vol.52 , pp. 827-833
    • Mack, D.1    Ahrne, S.2    Hyde, L.3    Wei, S.4    Hollingsworth, M.5
  • 170
    • 84904512621 scopus 로고    scopus 로고
    • Activation of epidermal growth factor receptor mediates mucin production stimulated by p40, a Lactobacillus rhamnosus GG-derived protein
    • Wang L, Cao H, Liu L, Wang B, Walker WA, Acra SA, Yan F. 2014. Activation of epidermal growth factor receptor mediates mucin production stimulated by p40, a Lactobacillus rhamnosus GG-derived protein. J Biol Chem 289: 20234-20244. https://doi.org/10.1074/ jbc.M114.553800.
    • (2014) J Biol Chem , vol.289 , pp. 20234-20244
    • Wang, L.1    Cao, H.2    Liu, L.3    Wang, B.4    Walker, W.A.5    Acra, S.A.6    Yan, F.7
  • 171
    • 84928534091 scopus 로고    scopus 로고
    • A selected Lactobacillus rhamnosus strain oromotes EGFR-independent Akt activation in an enterotoxigenic Escherichia coli K88-infected IPEC-J2 cell model
    • Zhang W, Zhu Y, Yang J, Yang G, Zhou D, Wang J. 2015. A selected Lactobacillus rhamnosus strain oromotes EGFR-independent Akt activation in an enterotoxigenic Escherichia coli K88-infected IPEC-J2 cell model. PLoS One 10: e0125717. https://doi.org/10.1371/ journal.pone.0125717.
    • (2015) PLoS One , vol.10
    • Zhang, W.1    Zhu, Y.2    Yang, J.3    Yang, G.4    Zhou, D.5    Wang, J.6
  • 172
    • 1642274741 scopus 로고    scopus 로고
    • Functional modulation of enterocytes by gram-positive and gram-negative microorganisms
    • Otte J-M, Podolsky DK. 2004. Functional modulation of enterocytes by gram-positive and gram-negative microorganisms. Am J Physiol Gastrointest Liver Physiol 286: G613-G626. https://doi.org/10.1152/ ajpgi.00341.2003.
    • (2004) Am J Physiol Gastrointest Liver Physiol , vol.286 , pp. G613-G626
    • Otte, J.-M.1    Podolsky, D.K.2
  • 173
    • 33846175058 scopus 로고    scopus 로고
    • The VSL#3 probiotic formula induces mucin gene expression and secretion in colonic epithelial cells
    • Caballero-Franco C, Keller K, Simone C, Chadee K. 2007. The VSL#3 probiotic formula induces mucin gene expression and secretion in colonic epithelial cells. Am J Physiol Gastrointest Liver Physiol 292: G315-G322.
    • (2007) Am J Physiol Gastrointest Liver Physiol , vol.292 , pp. G315-G322
    • Caballero-Franco, C.1    Keller, K.2    Simone, C.3    Chadee, K.4
  • 174
    • 79953680466 scopus 로고    scopus 로고
    • Saccharomyces cerevisiae modulates immune gene expressions and inhibits ETEC-mediated ERK1/2 and p38 signaling pathways in intestinal epithelial cells
    • Zanello G, Berri M, Dupont J, Sizaret PY, D’Inca R, Salmon H, Meurens F. 2011. Saccharomyces cerevisiae modulates immune gene expressions and inhibits ETEC-mediated ERK1/2 and p38 signaling pathways in intestinal epithelial cells. PLoS One 6: e18573. https://doi.org/10.1371/ journal.pone.0018573.
    • (2011) PLoS One , vol.6
    • Zanello, G.1    Berri, M.2    Dupont, J.3    Sizaret, P.Y.4    D’Inca, R.5    Salmon, H.6    Meurens, F.7
  • 175
    • 30644475117 scopus 로고    scopus 로고
    • Effects of Saccharomyces boulardii on fecal short-chain fatty acids and microflora in patients on long-term total enteral nutrition
    • Schneider S-M, Girard-Pipau F, Filippi J, Hebuterne X, Moyse D, Hinojosa G-C, Pompei A, Rampal P. 2005. Effects of Saccharomyces boulardii on fecal short-chain fatty acids and microflora in patients on long-term total enteral nutrition. World J Gastroenterol 11: 6165-6169. https:// doi.org/10.3748/wjg.v11.i39.6165.
    • (2005) World J Gastroenterol , vol.11 , pp. 6165-6169
    • Schneider, S.-M.1    Girard-Pipau, F.2    Filippi, J.3    Hebuterne, X.4    Moyse, D.5    Hinojosa, G.-C.6    Pompei, A.7    Rampal, P.8
  • 176
    • 66549123465 scopus 로고    scopus 로고
    • Butyrate enemas upregulate Muc genes expression but decrease adherent mucus thickness in mice colon
    • Gaudier E, Rival M, Buisine MP, Robineau I, Hoebler C. 2009. Butyrate enemas upregulate Muc genes expression but decrease adherent mucus thickness in mice colon. Physiol Res 58: 111-119.
    • (2009) Physiol Res , vol.58 , pp. 111-119
    • Gaudier, E.1    Rival, M.2    Buisine, M.P.3    Robineau, I.4    Hoebler, C.5
  • 177
    • 0033032723 scopus 로고    scopus 로고
    • Adhesion of four Bifidobacterium strains to human intestinal mucus from subjects in different age groups
    • Ouwehand A, Isolauri E, Kirjavainen PV, Salminen SJ. 1999. Adhesion of four Bifidobacterium strains to human intestinal mucus from subjects in different age groups. FEMS Microbiol Lett 172: 61-64. https://doi.org/10.1111/j.1574-6968.1999.tb13450.x.
    • (1999) FEMS Microbiol Lett , vol.172 , pp. 61-64
    • Ouwehand, A.1    Isolauri, E.2    Kirjavainen, P.V.3    Salminen, S.J.4
  • 178
    • 0034054767 scopus 로고    scopus 로고
    • The mucus binding of Bifidobacterium lactis Bb12 is enhanced in the presence of Lactobacillus GG and Lact. delbrueckii subsp. bulgaricus
    • Ouwehand A, Isolauri E, Kirjavainen PV, Tölkko S, Salminen SJ. 2000. The mucus binding of Bifidobacterium lactis Bb12 is enhanced in the presence of Lactobacillus GG and Lact. delbrueckii subsp. bulgaricus. Lett Appl Microbiol 30: 10-13. https://doi.org/10.1046/j.1472-765x.2000.00590.x.
    • (2000) Lett Appl Microbiol , vol.30 , pp. 10-13
    • Ouwehand, A.1    Isolauri, E.2    Kirjavainen, P.V.3    Tölkko, S.4    Salminen, S.J.5
  • 179
    • 0000752197 scopus 로고
    • Junctional complexes in various epithelia
    • Farquhar MG, Palade GE. 1963. Junctional complexes in various epithelia. J Cell Biol 17: 375-412. https://doi.org/10.1083/jcb.17.2.375.
    • (1963) J Cell Biol , vol.17 , pp. 375-412
    • Farquhar, M.G.1    Palade, G.E.2
  • 180
    • 0032491391 scopus 로고    scopus 로고
    • The tight junction protein ZO-1 establishes a link between the transmembrane protein occludin and the actin cytoskeleton
    • Fanning AS, Jameson BJ, Lynne A, Anderson JM, Jesaitis LA, Melvin J. 1998. The tight junction protein ZO-1 establishes a link between the transmembrane protein occludin and the actin cytoskeleton. J Biol Chem 273: 29745-29753. https://doi.org/10.1074/jbc.273.45.29745.
    • (1998) J Biol Chem , vol.273 , pp. 29745-29753
    • Fanning, A.S.1    Jameson, B.J.2    Lynne, A.3    Anderson, J.M.4    Jesaitis, L.A.5    Melvin, J.6
  • 181
    • 0023176321 scopus 로고
    • Intestinal absorptive cell tight junctions are linked to cytoskeleton
    • Madara JL. 1987. Intestinal absorptive cell tight junctions are linked to cytoskeleton. Am J Physiol 253: C171-C175.
    • (1987) Am J Physiol , vol.253 , pp. C171-C175
    • Madara, J.L.1
  • 182
    • 33750459738 scopus 로고    scopus 로고
    • Catenins: Keeping cells from getting their signals crossed
    • Perez-Moreno M, Fuchs E. 2006. Catenins: keeping cells from getting their signals crossed. Dev Cell 11: 601-612. https://doi.org/10.1016/ j.devcel.2006.10.010.
    • (2006) Dev Cell , vol.11 , pp. 601-612
    • Perez-Moreno, M.1    Fuchs, E.2
  • 183
    • 52949134856 scopus 로고    scopus 로고
    • Physiological concentrations of short-chain fatty acids immediately suppress colonic epithelial permeability
    • Suzuki T, Yoshida S, Hara H. 2008. Physiological concentrations of short-chain fatty acids immediately suppress colonic epithelial permeability. Br J Nutr 100: 297-305. https://doi.org/10.1017/ S0007114508888733.
    • (2008) Br J Nutr , vol.100 , pp. 297-305
    • Suzuki, T.1    Yoshida, S.2    Hara, H.3
  • 184
    • 69549114504 scopus 로고    scopus 로고
    • Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers
    • Peng L, Li Z-R, Green RS, Holzman IR, Lin J. 2009. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J Nutr 139: 1619-1625. https://doi.org/10.3945/jn.109.104638.
    • (2009) J Nutr , vol.139 , pp. 1619-1625
    • Peng, L.1    Li, Z.-R.2    Green, R.S.3    Holzman, I.R.4    Lin, J.5
  • 185
    • 84881005790 scopus 로고    scopus 로고
    • Strain-dependent augmentation of tight-junction barrier function in human primary epidermal keratinocytes by Lactobacillus and Bifidobacterium lysates
    • Sultana R, McBain AJ, O’Neill CA. 2013. Strain-dependent augmentation of tight-junction barrier function in human primary epidermal keratinocytes by Lactobacillus and Bifidobacterium lysates. Appl Environ Microbiol 79: 4887-4894. https://doi.org/10.1128/AEM.00982-13.
    • (2013) Appl Environ Microbiol , vol.79 , pp. 4887-4894
    • Sultana, R.1    McBain, A.J.2    O’Neill, C.A.3
  • 186
    • 0033806709 scopus 로고    scopus 로고
    • Saccharomyces boulardii preserves the barrier function and modulates the signal transduction pathway induced in enteropathogenic Escherichia coli-infected T84 cells
    • Czerucka D, Dahan S, Mograbi B, Rossi B, Rampal P. 2000. Saccharomyces boulardii preserves the barrier function and modulates the signal transduction pathway induced in enteropathogenic Escherichia coli-infected T84 cells. Infect Immun 68: 5998-6004. https://doi.org/10.1128/IAI.68.10.5998-6004.2000.
    • (2000) Infect Immun , vol.68 , pp. 5998-6004
    • Czerucka, D.1    Dahan, S.2    Mograbi, B.3    Rossi, B.4    Rampal, P.5
  • 188
    • 84964211538 scopus 로고    scopus 로고
    • Oral administration of Saccharomyces boulardii ameliorates carbon tetrachloride-induced liver fibrosis in rats via reducing intestinal permeability and modulating gut microbial composition
    • Li M, Zhu L, Xie A, Yuan J. 2015. Oral administration of Saccharomyces boulardii ameliorates carbon tetrachloride-induced liver fibrosis in rats via reducing intestinal permeability and modulating gut microbial composition. Inflammation 38: 170-179. https://doi.org/10.1007/ s10753-014-0019-7.
    • (2015) Inflammation , vol.38 , pp. 170-179
    • Li, M.1    Zhu, L.2    Xie, A.3    Yuan, J.4
  • 190
    • 41549123221 scopus 로고    scopus 로고
    • Saccharomyces boulardii interferes with Shigella pathogenesis by postinvasion signaling events
    • Mumy KL, Chen X, Kelly CP, McCormick BA. 2008. Saccharomyces boulardii interferes with Shigella pathogenesis by postinvasion signaling events. Am J Physiol Gastrointest Liver Physiol 294: G599-G609. https://doi.org/10.1152/ajpgi.00391.2007.
    • (2008) Am J Physiol Gastrointest Liver Physiol , vol.294 , pp. G599-G609
    • Mumy, K.L.1    Chen, X.2    Kelly, C.P.3    McCormick, B.A.4
  • 192
    • 84980413569 scopus 로고    scopus 로고
    • Strengthening of the intestinal epithelial tight junction by Bifidobacterium bifidum
    • Hsieh C-Y, Osaka T, Moriyama E, Date Y, Kikuchi J, Tsuneda S. 2015. Strengthening of the intestinal epithelial tight junction by Bifidobacterium bifidum. Physiol Rep 3: e12327. https://doi.org/10.14814/ phy2.12327.
    • (2015) Physiol Rep , vol.3
    • Hsieh, C.-Y.1    Osaka, T.2    Moriyama, E.3    Date, Y.4    Kikuchi, J.5    Tsuneda, S.6
  • 193
    • 82555194144 scopus 로고    scopus 로고
    • Lactobacillus plantarum prevents bacterial translocation in rats following ischemia and reperfusion injury
    • Wang B, Huang Q, Zhang W, Li N, Li J. 2011. Lactobacillus plantarum prevents bacterial translocation in rats following ischemia and reperfusion injury. Dig Dis Sci 56: 3187-3194. https://doi.org/10.1007/s10620-011-1747-2.
    • (2011) Dig Dis Sci , vol.56 , pp. 3187-3194
    • Wang, B.1    Huang, Q.2    Zhang, W.3    Li, N.4    Li, J.5
  • 194
    • 84868203487 scopus 로고    scopus 로고
    • Immune responses to Clostridium difficile infection
    • Madan R, Petri WA. 2012. Immune responses to Clostridium difficile infection. Trends Mol Med 18: 658-666. https://doi.org/10.1016/ j.molmed.2012.09.005.
    • (2012) Trends Mol Med , vol.18 , pp. 658-666
    • Madan, R.1    Petri, W.A.2
  • 196
    • 84876417481 scopus 로고    scopus 로고
    • Lactobacillus bacteremia associated with probiotic use in a pediatric patient with ulcerative colitis
    • Vahabnezhad E, Mochon A, Wozniak L, Ziring D. 2013. Lactobacillus bacteremia associated with probiotic use in a pediatric patient with ulcerative colitis. J Clin Gastroenterol 47: 437-439. https://doi.org/10.1097/MCG.0b013e318279abf0.
    • (2013) J Clin Gastroenterol , vol.47 , pp. 437-439
    • Vahabnezhad, E.1    Mochon, A.2    Wozniak, L.3    Ziring, D.4
  • 197
    • 2442563304 scopus 로고    scopus 로고
    • Clinical epidemiology of inflammatory bowel disease: Incidence, prevalence, and environmental influences
    • Loftus E. 2004. Clinical epidemiology of inflammatory bowel disease: incidence, prevalence, and environmental influences. Gastroenterology 126: 1504-1517. https://doi.org/10.1053/j.gastro.2004.01.063.
    • (2004) Gastroenterology , vol.126 , pp. 1504-1517
    • Loftus, E.1
  • 200
    • 84920901942 scopus 로고    scopus 로고
    • Differential diagnosis in inflammatory bowel disease colitis: State of the art and future perspectives
    • Tontini GE, Vecchi M, Pastorelli L, Neurath MF, Neumann H. 2015. Differential diagnosis in inflammatory bowel disease colitis: state of the art and future perspectives. World J Gastroenterol 21: 21-46. https:// doi.org/10.3748/wjg.v21.i1.21.
    • (2015) World J Gastroenterol , vol.21 , pp. 21-46
    • Tontini, G.E.1    Vecchi, M.2    Pastorelli, L.3    Neurath, M.F.4    Neumann, H.5
  • 201
    • 70449482735 scopus 로고    scopus 로고
    • Diagnostic standards in the pathology of inflammatory bowel disease
    • Loddenkemper C. 2009. Diagnostic standards in the pathology of inflammatory bowel disease. Dig Dis 27: 576-583. https://doi.org/10.1159/000233301.
    • (2009) Dig Dis , vol.27 , pp. 576-583
    • Loddenkemper, C.1
  • 203
    • 33847052752 scopus 로고    scopus 로고
    • Meta-analysis: Cancer risk of low-grade dysplasia in chronic ulcerative colitis
    • Thomas T, Abrams KA, Robinson RJ, Mayberry JF. 2007. Meta-analysis: cancer risk of low-grade dysplasia in chronic ulcerative colitis. Aliment Pharmacol Ther 25: 657-668. https://doi.org/10.1111/j.1365-2036.2007.03241.x.
    • (2007) Aliment Pharmacol Ther , vol.25 , pp. 657-668
    • Thomas, T.1    Abrams, K.A.2    Robinson, R.J.3    Mayberry, J.F.4
  • 204
    • 0035044890 scopus 로고    scopus 로고
    • The prevalence of extraintestinal diseases in inflammatory bowel disease: A populationbased study
    • Bernstein CN, Blanchard JF, Rawsthorne P, Yu N. 2001. The prevalence of extraintestinal diseases in inflammatory bowel disease: a populationbased study. Am J Gastroenterol 96: 1116-1122. https://doi.org/10.1111/j.1572-0241.2001.03756.x.
    • (2001) Am J Gastroenterol , vol.96 , pp. 1116-1122
    • Bernstein, C.N.1    Blanchard, J.F.2    Rawsthorne, P.3    Yu, N.4
  • 205
    • 0030794276 scopus 로고    scopus 로고
    • Pre-illness dietary factors in inflammatory bowel disease
    • Reif S, Klein I, Lubin F, Farbstein M, Hallak A, Gilat T. 1997. Pre-illness dietary factors in inflammatory bowel disease. Gut 40: 754-760. https:// doi.org/10.1136/gut.40.6.754.
    • (1997) Gut , vol.40 , pp. 754-760
    • Reif, S.1    Klein, I.2    Lubin, F.3    Farbstein, M.4    Hallak, A.5    Gilat, T.6
  • 206
    • 3342988481 scopus 로고    scopus 로고
    • Inflammatory bowel disease: The role of environmental factors
    • Danese S, Sans M, Fiocchi C. 2004. Inflammatory bowel disease: the role of environmental factors. Autoimmun Rev 3: 394-400. https://doi.org/10.1016/j.autrev.2004.03.002.
    • (2004) Autoimmun Rev , vol.3 , pp. 394-400
    • Danese, S.1    Sans, M.2    Fiocchi, C.3
  • 207
    • 0035869415 scopus 로고    scopus 로고
    • Appendectomy and protection against ulcerative colitis
    • Andersson RE, Olaison G, Tysk C, Ekbom A. 2001. Appendectomy and protection against ulcerative colitis. N Engl J Med 344: 808-814. https:// doi.org/10.1056/NEJM200103153441104.
    • (2001) N Engl J Med , vol.344 , pp. 808-814
    • Andersson, R.E.1    Olaison, G.2    Tysk, C.3    Ekbom, A.4
  • 208
    • 0001213646 scopus 로고
    • Non-smoking: A feature of ulcerative colitis
    • Harries AD, Baird A, Rhodes J. 1982. Non-smoking: a feature of ulcerative colitis. Br Med J 284: 706. https://doi.org/10.1136/ bmj.284.6317.706.
    • (1982) Br Med J , vol.284 , pp. 706
    • Harries, A.D.1    Baird, A.2    Rhodes, J.3
  • 209
    • 33745775434 scopus 로고    scopus 로고
    • Mechanisms of disease: Pathogenesis of Crohn’s disease and ulcerative colitis
    • Sartor RB. 2006. Mechanisms of disease: pathogenesis of Crohn’s disease and ulcerative colitis. Nat Clin Pract Gastroenterol Hepatol 3: 390-407. https://doi.org/10.1038/ncpgasthep0528.
    • (2006) Nat Clin Pract Gastroenterol Hepatol , vol.3 , pp. 390-407
    • Sartor, R.B.1
  • 210
    • 84951919076 scopus 로고    scopus 로고
    • Immunopathogenesis of IBD: Current state of the art
    • de Souza HSP, Fiocchi C. 2016. Immunopathogenesis of IBD: current state of the art. Nat Rev Gastroenterol Hepatol 13: 13-27. https:// doi.org/10.1038/nrgastro.2015.186.
    • (2016) Nat Rev Gastroenterol Hepatol , vol.13 , pp. 13-27
    • de Souza, H.S.P.1    Fiocchi, C.2
  • 211
    • 84947461191 scopus 로고    scopus 로고
    • Intestinal permeability in inflammatory bowel disease: Pathogenesis, clinical evaluation, and therapy of leaky gut
    • Michielan A, D’Incà R. 2015. Intestinal permeability in inflammatory bowel disease: pathogenesis, clinical evaluation, and therapy of leaky gut. Mediators Inflamm 2015: 628157. https://doi.org/10.1155/2015/628157.
    • (2015) Mediators Inflamm , vol.2015 , pp. 628157
    • Michielan, A.1    D’Incà, R.2
  • 212
    • 70649103789 scopus 로고    scopus 로고
    • Genome-wide association study of ulcerative colitis identifies three new susceptibility loci, including the HNF4A region
    • UK IBD Genetics Consortium, Wellcome Trust Case Control Consortium 2. 2009. Genome-wide association study of ulcerative colitis identifies three new susceptibility loci, including the HNF4A region. Nat Genet 41: 1330-1334. https://doi.org/10.1038/ng.483.
    • (2009) Nat Genet , vol.41 , pp. 1330-1334
  • 213
    • 14944341671 scopus 로고    scopus 로고
    • Allelic variations of the multidrug resistance gene determine susceptibility and disease behavior in ulcerative colitis
    • Ho G-T, Nimmo ER, Tenesa A, Fennell J, Drummond H, Mowat C, Arnott ID, Satsangi J. 2005. Allelic variations of the multidrug resistance gene determine susceptibility and disease behavior in ulcerative colitis. Gastroenterology 128: 288-296. https://doi.org/10.1053/ j.gastro.2004.11.019.
    • (2005) Gastroenterology , vol.128 , pp. 288-296
    • Ho, G.-T.1    Nimmo, E.R.2    Tenesa, A.3    Fennell, J.4    Drummond, H.5    Mowat, C.6    Arnott, I.D.7    Satsangi, J.8
  • 222
    • 79955935457 scopus 로고    scopus 로고
    • Intestinal microbiota in inflammatory bowel disease: Friend of foe?
    • Fava F, Danese S. 2011. Intestinal microbiota in inflammatory bowel disease: friend of foe? World J Gastroenterol 17: 557-566. https:// doi.org/10.3748/wjg.v17.i5.557.
    • (2011) World J Gastroenterol , vol.17 , pp. 557-566
    • Fava, F.1    Danese, S.2
  • 223
    • 84936946364 scopus 로고    scopus 로고
    • Impacts of infection with different toxigenic Clostridium difficile strains on faecal microbiota in children
    • Ling Z, Liu X, Jia X, Cheng Y, Luo Y, Yuan L, Wang Y, Zhao C, Guo S, Li L, Xu X, Xiang C. 2014. Impacts of infection with different toxigenic Clostridium difficile strains on faecal microbiota in children. Sci Rep 4: 7485. https://doi.org/10.1038/srep07485.
    • (2014) Sci Rep , vol.4 , pp. 7485
    • Ling, Z.1    Liu, X.2    Jia, X.3    Cheng, Y.4    Luo, Y.5    Yuan, L.6    Wang, Y.7    Zhao, C.8    Guo, S.9    Li, L.10    Xu, X.11    Xiang, C.12
  • 224
    • 84871630781 scopus 로고    scopus 로고
    • Intestinal microbes in inflammatory bowel diseases
    • Sartor RB, Mazmanian SK. 2012. Intestinal microbes in inflammatory bowel diseases. Am J Gastroenterol Suppl 1: 15-21. https://doi.org/10.1038/ajgsup.2012.4.
    • (2012) Am J Gastroenterol Suppl , vol.1 , pp. 15-21
    • Sartor, R.B.1    Mazmanian, S.K.2
  • 225
    • 0026597788 scopus 로고
    • Virulence properties of Escherichia coli strains isolated from patients with inflammatory bowel disease
    • Giaffer M, Holdsworth C, Duerden B. 1992. Virulence properties of Escherichia coli strains isolated from patients with inflammatory bowel disease. Gut 33: 646-650. https://doi.org/10.1136/gut.33.5.646.
    • (1992) Gut , vol.33 , pp. 646-650
    • Giaffer, M.1    Holdsworth, C.2    Duerden, B.3
  • 226
    • 4544348026 scopus 로고    scopus 로고
    • Comparative composition of bacteria in the human intestinal microflora during remission and active ulcerative colitis
    • Bullock NR, Booth JCL, Gibson GR. 2004. Comparative composition of bacteria in the human intestinal microflora during remission and active ulcerative colitis. Curr Issues Intest Microbiol 5: 59-64.
    • (2004) Curr Issues Intest Microbiol , vol.5 , pp. 59-64
    • Bullock, N.R.1    Booth, J.C.L.2    Gibson, G.R.3
  • 228
    • 67349250428 scopus 로고    scopus 로고
    • The gut microbiota shapes intestinal immune responses during health and disease
    • Round JL, Mazmanian SK. 2009. The gut microbiota shapes intestinal immune responses during health and disease. Nat Rev Immunol 9: 313-324. https://doi.org/10.1038/nri2515.
    • (2009) Nat Rev Immunol , vol.9 , pp. 313-324
    • Round, J.L.1    Mazmanian, S.K.2
  • 229
    • 0032412057 scopus 로고    scopus 로고
    • Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice
    • Sellon RK, Tonkonogy S, Schultz M, Dieleman LA, Grenther W, Balish E, Rennick DM, Sartor RB. 1998. Resident enteric bacteria are necessary for development of spontaneous colitis and immune system activation in interleukin-10-deficient mice. Infect Immun 66: 5224-5231.
    • (1998) Infect Immun , vol.66 , pp. 5224-5231
    • Sellon, R.K.1    Tonkonogy, S.2    Schultz, M.3    Dieleman, L.A.4    Grenther, W.5    Balish, E.6    Rennick, D.M.7    Sartor, R.B.8
  • 230
    • 0031012926 scopus 로고    scopus 로고
    • T cell receptor-alpha beta-deficient mice fail to develop colitis in the absence of a microbial environment
    • Dianda L, Hanby A, Wright N, Sebesteny A, Hayday A, Owen MJ. 1997. T cell receptor-alpha beta-deficient mice fail to develop colitis in the absence of a microbial environment. Am J Pathol 150: 91-97.
    • (1997) Am J Pathol , vol.150 , pp. 91-97
    • Dianda, L.1    Hanby, A.2    Wright, N.3    Sebesteny, A.4    Hayday, A.5    Owen, M.J.6
  • 232
    • 0021276878 scopus 로고
    • Intestinal lymphocyte subpopulations in inflammatory bowel disease: An analysis by immunohistological and cell isolation techniques
    • Selby WS, Janossy G, Bofill M, Jewell DP. 1984. Intestinal lymphocyte subpopulations in inflammatory bowel disease: an analysis by immunohistological and cell isolation techniques. Gut 25: 32-40. https:// doi.org/10.1136/gut.25.1.32.
    • (1984) Gut , vol.25 , pp. 32-40
    • Selby, W.S.1    Janossy, G.2    Bofill, M.3    Jewell, D.P.4
  • 233
    • 47949116890 scopus 로고    scopus 로고
    • Chemokines and chemokine receptors in mucosal homeostasis at the intestinal epithelial barrier in inflammatory bowel disease
    • Zimmerman NP, Vongsa RA, Wendt MK, Dwinell MB. 2008. Chemokines and chemokine receptors in mucosal homeostasis at the intestinal epithelial barrier in inflammatory bowel disease. Inflamm Bowel Dis 14: 1000-1011. https://doi.org/10.1002/ibd.20480.
    • (2008) Inflamm Bowel Dis , vol.14 , pp. 1000-1011
    • Zimmerman, N.P.1    Vongsa, R.A.2    Wendt, M.K.3    Dwinell, M.B.4
  • 235
    • 79551558403 scopus 로고    scopus 로고
    • CCL25/CCR9 interactions regulate large intestinal inflammation in a murine model of acute colitis
    • Wurbel M, McIntire MG, Dwyer P, Fiebiger E. 2011. CCL25/CCR9 interactions regulate large intestinal inflammation in a murine model of acute colitis. PLoS One 6: e16442. https://doi.org/10.1371/ journal.pone.0016442.
    • (2011) PLoS One , vol.6
    • Wurbel, M.1    McIntire, M.G.2    Dwyer, P.3    Fiebiger, E.4
  • 236
    • 84880074086 scopus 로고    scopus 로고
    • Cytokine networks in ulcerative colitis
    • Roda G, Marocchi M, Sartini A, Roda E. 2011. Cytokine networks in ulcerative colitis. Ulcers 2011: 391787. https://doi.org/10.1155/2011/391787.
    • (2011) Ulcers , vol.2011
    • Roda, G.1    Marocchi, M.2    Sartini, A.3    Roda, E.4
  • 237
    • 34447650384 scopus 로고    scopus 로고
    • The role of TNF-alpha in ulcerative colitis
    • Sands BE, Kaplan GG. 2007. The role of TNF-alpha in ulcerative colitis. J Clin Pharmacol 47: 930-941. https://doi.org/10.1177/0091270007301623.
    • (2007) J Clin Pharmacol , vol.47 , pp. 930-941
    • Sands, B.E.1    Kaplan, G.G.2
  • 240
    • 84880692710 scopus 로고    scopus 로고
    • IL-16 induces intestinal inflammation via PepT1 upregulation in a pufferfish model: New insights into the molecular mechanism of inflammatory bowel disease
    • Wang P, Lu Y, Wen Y, Yu D, Ge L, Dong W, Xiang L, Shao J. 2013. IL-16 induces intestinal inflammation via PepT1 upregulation in a pufferfish model: new insights into the molecular mechanism of inflammatory bowel disease. J Immunol 191: 1413-1427. https://doi.org/10.4049/ jimmunol.1202598.
    • (2013) J Immunol , vol.191 , pp. 1413-1427
    • Wang, P.1    Lu, Y.2    Wen, Y.3    Yu, D.4    Ge, L.5    Dong, W.6    Xiang, L.7    Shao, J.8
  • 243
    • 84940843952 scopus 로고    scopus 로고
    • Th17 cells as potential probiotic therapeutic targets in inflammatory bowel diseases
    • Owaga E, Hsieh R-H, Mugendi B, Masuku S, Shih C-K, Chang J-S. 2015. Th17 cells as potential probiotic therapeutic targets in inflammatory bowel diseases. Int J Mol Sci 16: 20841-20858. https://doi.org/10.3390/ ijms160920841.
    • (2015) Int J Mol Sci , vol.16 , pp. 20841-20858
    • Owaga, E.1    Hsieh, R.-H.2    Mugendi, B.3    Masuku, S.4    Shih, C.-K.5    Chang, J.-S.6
  • 244
    • 84886951561 scopus 로고    scopus 로고
    • Innate and adaptive immunity in inflammatory bowel disease
    • Geremia A, Biancheri P, Allan P, Corazza GR, Di Sabatino A. 2014. Innate and adaptive immunity in inflammatory bowel disease. Autoimmun Rev 13: 3-10. https://doi.org/10.1016/j.autrev.2013.06.004.
    • (2014) Autoimmun Rev , vol.13 , pp. 3-10
    • Geremia, A.1    Biancheri, P.2    Allan, P.3    Corazza, G.R.4    Di Sabatino, A.5
  • 246
    • 65549098640 scopus 로고    scopus 로고
    • CD4 T-cell differentiation and inflammatory bowel disease
    • Zenewicz LA, Antov A, Flavell RA. 2009. CD4 T-cell differentiation and inflammatory bowel disease. Trends Mol Med 15: 199-207. https:// doi.org/10.1016/j.molmed.2009.03.002.
    • (2009) Trends Mol Med , vol.15 , pp. 199-207
    • Zenewicz, L.A.1    Antov, A.2    Flavell, R.A.3
  • 249
    • 77952222918 scopus 로고    scopus 로고
    • The increased mucosal mRNA expressions of complement C3 and interleukin-17 in inflammatory bowel disease
    • Sugihara T, Kobori A, Imaeda H, Tsujikawa T, Amagase K, Takeuchi K, Fujiyama Y, Andoh A. 2010. The increased mucosal mRNA expressions of complement C3 and interleukin-17 in inflammatory bowel disease. Clin Exp Immunol 160: 386-393. https://doi.org/10.1111/j.1365-2249.2010.04093.x.
    • (2010) Clin Exp Immunol , vol.160 , pp. 386-393
    • Sugihara, T.1    Kobori, A.2    Imaeda, H.3    Tsujikawa, T.4    Amagase, K.5    Takeuchi, K.6    Fujiyama, Y.7    Andoh, A.8
  • 250
    • 0842324613 scopus 로고    scopus 로고
    • Neutralization of interleukin-17 aggravates dextran sulfate sodium-induced colitis in mice
    • Ogawa A, Andoh A, Araki Y, Bamba T, Fujiyama Y. 2004. Neutralization of interleukin-17 aggravates dextran sulfate sodium-induced colitis in mice. Clin Immunol 110: 55-62. https://doi.org/10.1016/ j.clim.2003.09.013.
    • (2004) Clin Immunol , vol.110 , pp. 55-62
    • Ogawa, A.1    Andoh, A.2    Araki, Y.3    Bamba, T.4    Fujiyama, Y.5
  • 251
    • 33646804892 scopus 로고    scopus 로고
    • Critical role of IL-17 receptor signaling in acute TNBS-induced colitis
    • Zhang Z, Zheng M, Bindas J, Schwarzenberger P, Kolls JK. 2006. Critical role of IL-17 receptor signaling in acute TNBS-induced colitis. Inflamm Bowel Dis 12: 382-388. https://doi.org/10.1097/01.MIB.0000218764.06959.91.
    • (2006) Inflamm Bowel Dis , vol.12 , pp. 382-388
    • Zhang, Z.1    Zheng, M.2    Bindas, J.3    Schwarzenberger, P.4    Kolls, J.K.5
  • 252
    • 77953201731 scopus 로고    scopus 로고
    • 4SC-101, a novel immunosuppressive drug, inhibits IL-17 and attenuates colitis in two murine models of inflammatory bowel disease
    • Fitzpatrick LR, Deml L, Hofmann C, Small JS, Groeppel M, Hamm S, Lemstra S, Leban J, Ammendola A. 2010. 4SC-101, a novel immunosuppressive drug, inhibits IL-17 and attenuates colitis in two murine models of inflammatory bowel disease. Inflamm Bowel Dis 16: 1763-1777. https://doi.org/10.1002/ibd.21264.
    • (2010) Inflamm Bowel Dis , vol.16 , pp. 1763-1777
    • Fitzpatrick, L.R.1    Deml, L.2    Hofmann, C.3    Small, J.S.4    Groeppel, M.5    Hamm, S.6    Lemstra, S.7    Leban, J.8    Ammendola, A.9
  • 253
    • 85029716515 scopus 로고    scopus 로고
    • Vidofludimus inhibits IL-17 and improves hapten-induced colitis in young rats by a unique dual mode of action
    • Fitzpatrick LR, Small J, Doblhofer R, Ammendola A. 2012. Vidofludimus inhibits IL-17 and improves hapten-induced colitis in young rats by a unique dual mode of action. J Crohns Colitis 6: S15-S16. https://doi.org/10.1016/S1873-9946(12)60033-6.
    • (2012) J Crohns Colitis , vol.6 , pp. S15-S16
    • Fitzpatrick, L.R.1    Small, J.2    Doblhofer, R.3    Ammendola, A.4
  • 254
    • 0033849872 scopus 로고    scopus 로고
    • A reproducible grading scale for histological assessment of inflammation in ulcerative colitis
    • Geboes K, Riddell R, Ost A, Jensfelt B, Persson T, Löfberg R. 2000. A reproducible grading scale for histological assessment of inflammation in ulcerative colitis. Gut 47: 404-409. https://doi.org/10.1136/ gut.47.3.404.
    • (2000) Gut , vol.47 , pp. 404-409
    • Geboes, K.1    Riddell, R.2    Ost, A.3    Jensfelt, B.4    Persson, T.5    Löfberg, R.6
  • 255
    • 0023839955 scopus 로고
    • Diminished neutrophil function in Crohn’s disease and ulcerative colitis identified by decreased oxidative metabolism and low superoxide dismutase content
    • Verspaget HW, Peña A, Weterman IT, Lamers CB. 1988. Diminished neutrophil function in Crohn’s disease and ulcerative colitis identified by decreased oxidative metabolism and low superoxide dismutase content. Gut 29: 223-228. https://doi.org/10.1136/gut.29.2.223.
    • (1988) Gut , vol.29 , pp. 223-228
    • Verspaget, H.W.1    Peña, A.2    Weterman, I.T.3    Lamers, C.B.4
  • 256
    • 4644303010 scopus 로고    scopus 로고
    • Relationship between fecal calprotectin, intestinal inflammation, and peripheral blood neutrophils in patients with active ulcerative colitis
    • Hanai H, Takeuchi K, Iida T, Kashiwagi N, Saniabadi AR, Matsushita I, Sato Y, Kasuga N, Nakamura T. 2004. Relationship between fecal calprotectin, intestinal inflammation, and peripheral blood neutrophils in patients with active ulcerative colitis. Dig Dis Sci 49: 1438-1443. https:// doi.org/10.1023/B: DDAS.0000042243.47279.87.
    • (2004) Dig Dis Sci , vol.49 , pp. 1438-1443
    • Hanai, H.1    Takeuchi, K.2    Iida, T.3    Kashiwagi, N.4    Saniabadi, A.R.5    Matsushita, I.6    Sato, Y.7    Kasuga, N.8    Nakamura, T.9
  • 257
    • 84864861276 scopus 로고    scopus 로고
    • The role of neutrophils during intestinal inflammation
    • Fournier BM, Parkos CA. 2012. The role of neutrophils during intestinal inflammation. Mucosal Immunol 5: 354-366. https://doi.org/10.1038/ mi.2012.24.
    • (2012) Mucosal Immunol , vol.5 , pp. 354-366
    • Fournier, B.M.1    Parkos, C.A.2
  • 258
    • 0037442137 scopus 로고    scopus 로고
    • IL-17, produced by lymphocytes and neutrophils, is necessary for lipopolysaccharide-induced airway neutrophilia: IL-15 as a possible trigger
    • Ferretti S, Bonneau O, Dubois GR, Jones CE, Trifilieff A. 2003. IL-17, produced by lymphocytes and neutrophils, is necessary for lipopolysaccharide-induced airway neutrophilia: IL-15 as a possible trigger. J Immunol 170: 2106-2112. https://doi.org/10.4049/ jimmunol.170.4.2106.
    • (2003) J Immunol , vol.170 , pp. 2106-2112
    • Ferretti, S.1    Bonneau, O.2    Dubois, G.R.3    Jones, C.E.4    Trifilieff, A.5
  • 259
    • 84875442814 scopus 로고    scopus 로고
    • Neutrophil recruitment and function in health and inflammation
    • Kolaczkowska E, Kubes P. 2013. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol 13: 159-175. https:// doi.org/10.1038/nri3399.
    • (2013) Nat Rev Immunol , vol.13 , pp. 159-175
    • Kolaczkowska, E.1    Kubes, P.2
  • 260
    • 79551628086 scopus 로고    scopus 로고
    • Upregulation of Gr1+ CD11b+ population in spleen of dextran sulfate sodium administered mice works to repair colitis
    • Zhang R, Ito S, Nishio N, Cheng Z, Suzuki H, Isobe K. 2011. Upregulation of Gr1+ CD11b+ population in spleen of dextran sulfate sodium administered mice works to repair colitis. Inflamm Allergy Drug Targets 10: 39-46. https://doi.org/10.2174/187152811794352114.
    • (2011) Inflamm Allergy Drug Targets , vol.10 , pp. 39-46
    • Zhang, R.1    Ito, S.2    Nishio, N.3    Cheng, Z.4    Suzuki, H.5    Isobe, K.6
  • 263
    • 0027484754 scopus 로고
    • Inhibition of apoptosis and prolongation of neutrophil functional longevity by inflammatory mediators
    • Lee A, Whyte MK, Haslett C. 1993. Inhibition of apoptosis and prolongation of neutrophil functional longevity by inflammatory mediators. J Leukoc Biol 54: 283-288.
    • (1993) J Leukoc Biol , vol.54 , pp. 283-288
    • Lee, A.1    Whyte, M.K.2    Haslett, C.3
  • 264
    • 0029948263 scopus 로고    scopus 로고
    • IL-10 enhances resolution of pulmonary inflammation in vivo by promoting apoptosis of neutrophils
    • Cox G. 1996. IL-10 enhances resolution of pulmonary inflammation in vivo by promoting apoptosis of neutrophils. Am J Physiol 271: L566-L571.
    • (1996) Am J Physiol , vol.271 , pp. L566-L571
    • Cox, G.1
  • 266
    • 0035185891 scopus 로고    scopus 로고
    • Neutrophil transmigration in inflammatory bowel disease is associated with differential expression of epithelial intercellular junction proteins
    • Kucharzik T, Walsh SV, Chen J, Parkos CA, Nusrat A. 2001. Neutrophil transmigration in inflammatory bowel disease is associated with differential expression of epithelial intercellular junction proteins. Am J Pathol 159: 2001-2009. https://doi.org/10.1016/S0002-9440(10)63051-9.
    • (2001) Am J Pathol , vol.159 , pp. 2001-2009
    • Kucharzik, T.1    Walsh, S.V.2    Chen, J.3    Parkos, C.A.4    Nusrat, A.5
  • 267
    • 0026529032 scopus 로고
    • Faecal elastase reflects disease activity in active ulcerative colitis
    • Adeyemi E, Hodgson H. 1992. Faecal elastase reflects disease activity in active ulcerative colitis. Scand J Gastroenterol 27: 139-142. https:// doi.org/10.3109/00365529209165434.
    • (1992) Scand J Gastroenterol , vol.27 , pp. 139-142
    • Adeyemi, E.1    Hodgson, H.2
  • 268
    • 33847707327 scopus 로고    scopus 로고
    • Drug insight: Aminosalicylates for the treatment of IBD
    • Nielsen OH, Munck LK. 2007. Drug insight: aminosalicylates for the treatment of IBD. Nat Clin Pract Gastroenterol Hepatol 4: 160-170. https://doi.org/10.1038/ncpgasthep0696.
    • (2007) Nat Clin Pract Gastroenterol Hepatol , vol.4 , pp. 160-170
    • Nielsen, O.H.1    Munck, L.K.2
  • 269
    • 77949271455 scopus 로고    scopus 로고
    • Ulcerative colitis practice guidelines in adults: American College of Gastroenterology Practice Parameters Committee
    • Kornbluth A, Sachar DB, Practice Parameters Committee of the American College of Gastroenterology. 2010. Ulcerative colitis practice guidelines in adults: American College of Gastroenterology Practice Parameters Committee. Am J Gastroenterol 105: 501-523. https:// doi.org/10.1038/ajg.2009.727.
    • (2010) Am J Gastroenterol , vol.105 , pp. 501-523
    • Kornbluth, A.1    Sachar, D.B.2
  • 270
    • 0036278519 scopus 로고    scopus 로고
    • The efficacy of azathioprine for the treatment of inflammatory bowel disease: A 30 year review
    • Fraser AG, Orchard TR, Jewell DP. 2002. The efficacy of azathioprine for the treatment of inflammatory bowel disease: a 30 year review. Gut 50: 485-489. https://doi.org/10.1136/gut.50.4.485.
    • (2002) Gut , vol.50 , pp. 485-489
    • Fraser, A.G.1    Orchard, T.R.2    Jewell, D.P.3
  • 271
    • 0029817846 scopus 로고    scopus 로고
    • The long-term outcome of ulcerative colitis treated with 6-mercaptopurine
    • George J, Present DH, Pou R, Bodian C, Rubin PH. 1996. The long-term outcome of ulcerative colitis treated with 6-mercaptopurine. Am J Gastroenterol 91: 1711-1714.
    • (1996) Am J Gastroenterol , vol.91 , pp. 1711-1714
    • George, J.1    Present, D.H.2    Pou, R.3    Bodian, C.4    Rubin, P.H.5
  • 275
    • 12344329448 scopus 로고    scopus 로고
    • Ciclosporin use in acute ulcerative colitis: A long-term experience
    • Campbell S, Travis S, Jewell D. 2005. Ciclosporin use in acute ulcerative colitis: a long-term experience. Eur J Gastroenterol Hepatol 17: 79-84. https://doi.org/10.1097/00042737-200501000-00016.
    • (2005) Eur J Gastroenterol Hepatol , vol.17 , pp. 79-84
    • Campbell, S.1    Travis, S.2    Jewell, D.3
  • 276
    • 84873020600 scopus 로고    scopus 로고
    • Azathioprine and 6-mercaptopurine for maintenance of remission in ulcerative colitis
    • Timmer A, McDonald J, Tsoulis D, Macdonald J. 2012. Azathioprine and 6-mercaptopurine for maintenance of remission in ulcerative colitis. Cochrane Database Syst Rev 9: CD000478. https://doi.org/10.1002/14651858.CD000478.pub3.
    • (2012) Cochrane Database Syst Rev , vol.9
    • Timmer, A.1    McDonald, J.2    Tsoulis, D.3    Macdonald, J.4
  • 277
    • 77957887624 scopus 로고    scopus 로고
    • Timing and indications for colectomy in chronic ulcerative colitis: Surgical consideration
    • Cima R. 2010. Timing and indications for colectomy in chronic ulcerative colitis: surgical consideration. Dig Dis 28: 501-507. https://doi.org/10.1159/000320409.
    • (2010) Dig Dis , vol.28 , pp. 501-507
    • Cima, R.1
  • 278
    • 73849116788 scopus 로고    scopus 로고
    • Secondary pouchitis: Those with identifiable etiopathogenetic or triggering factors
    • Navaneethan U, Shen B. 2010. Secondary pouchitis: those with identifiable etiopathogenetic or triggering factors. Am J Gastroenterol 105: 51-64. https://doi.org/10.1038/ajg.2009.530.
    • (2010) Am J Gastroenterol , vol.105 , pp. 51-64
    • Navaneethan, U.1    Shen, B.2
  • 279
    • 70450169730 scopus 로고    scopus 로고
    • Pouchitis and pouch dysfunction
    • Wu H, Shen B. 2010. Pouchitis and pouch dysfunction. Med Clin North Am 94: 75-92. https://doi.org/10.1016/j.mcna.2009.08.014.
    • (2010) Med Clin North Am , vol.94 , pp. 75-92
    • Wu, H.1    Shen, B.2
  • 281
    • 84885455975 scopus 로고    scopus 로고
    • Curbing our enthusiasm for fecal transplantation in ulcerative colitis
    • Rubin DT. 2013. Curbing our enthusiasm for fecal transplantation in ulcerative colitis. Am J Gastroenterol 108: 1631-1633. https://doi.org/10.1038/ajg.2013.279.
    • (2013) Am J Gastroenterol , vol.108 , pp. 1631-1633
    • Rubin, D.T.1
  • 282
    • 84957886668 scopus 로고    scopus 로고
    • Fecal microbiota transplantation: In perspective
    • Gupta S, Allen-Vercoe E, Petrof EO. 2016. Fecal microbiota transplantation: in perspective. Therap Adv Gastroenterol 9: 229-239. https://doi.org/10.1177/1756283X15607414.
    • (2016) Therap Adv Gastroenterol , vol.9 , pp. 229-239
    • Gupta, S.1    Allen-Vercoe, E.2    Petrof, E.O.3
  • 283
    • 84904128204 scopus 로고    scopus 로고
    • Fecal microbiota transplantation inducing remission in Crohn’s colitis and the associated changes in fecal microbial profile
    • Kao D, Hotte N, Gillevet P, Madsen K. 2014. Fecal microbiota transplantation inducing remission in Crohn’s colitis and the associated changes in fecal microbial profile. J Clin Gastroenterol 48: 625-628. https:// doi.org/10.1097/MCG.0000000000000131.
    • (2014) J Clin Gastroenterol , vol.48 , pp. 625-628
    • Kao, D.1    Hotte, N.2    Gillevet, P.3    Madsen, K.4
  • 286
    • 84918584474 scopus 로고    scopus 로고
    • Fecal microbiota transplantation as therapy for inflammatory bowel disease: A systematic review and meta-analysis
    • Colman RJ, Rubin DT. 2014. Fecal microbiota transplantation as therapy for inflammatory bowel disease: a systematic review and meta-analysis. J Crohns Colitis 8: 1569-1581. https://doi.org/10.1016/ j.crohns.2014.08.006.
    • (2014) J Crohns Colitis , vol.8 , pp. 1569-1581
    • Colman, R.J.1    Rubin, D.T.2
  • 287
    • 35148850257 scopus 로고    scopus 로고
    • Clinical effectiveness of probiotics therapy (BIO-THREE) in patients with ulcerative colitis refractory to conventional therapy
    • Tsuda Y, Yoshimatsu Y, Aoki H, Nakamura K, Irie M, Fukuda K, Hosoe N, Takada N, Shirai K, Suzuki Y. 2007. Clinical effectiveness of probiotics therapy (BIO-THREE) in patients with ulcerative colitis refractory to conventional therapy. Scand J Gastroenterol 42: 1306-1311. https:// doi.org/10.1080/00365520701396091.
    • (2007) Scand J Gastroenterol , vol.42 , pp. 1306-1311
    • Tsuda, Y.1    Yoshimatsu, Y.2    Aoki, H.3    Nakamura, K.4    Irie, M.5    Fukuda, K.6    Hosoe, N.7    Takada, N.8    Shirai, K.9    Suzuki, Y.10
  • 288
    • 0037309595 scopus 로고    scopus 로고
    • Randomized controlled trial of the effect of bifidobacteria-fermented milk on ulcerative colitis
    • Ishikawa H, Akedo I, Umesaki Y, Tanaka R, Imaoka A, Otani T. 2003. Randomized controlled trial of the effect of bifidobacteria-fermented milk on ulcerative colitis. J Am Coll Nutr 22: 56-63. https://doi.org/10.1080/07315724.2003.10719276.
    • (2003) J Am Coll Nutr , vol.22 , pp. 56-63
    • Ishikawa, H.1    Akedo, I.2    Umesaki, Y.3    Tanaka, R.4    Imaoka, A.5    Otani, T.6
  • 290
    • 39049130131 scopus 로고    scopus 로고
    • Effect of dietary intervention with different pre-and probiotics on intestinal bacterial enzyme activities
    • De Preter V, Raemen H, Cloetens L, Houben E, Rutgeerts P, Verbeke K. 2008. Effect of dietary intervention with different pre-and probiotics on intestinal bacterial enzyme activities. Eur J Clin Nutr 62: 225-231. https://doi.org/10.1038/sj.ejcn.1602706.
    • (2008) Eur J Clin Nutr , vol.62 , pp. 225-231
    • De Preter, V.1    Raemen, H.2    Cloetens, L.3    Houben, E.4    Rutgeerts, P.5    Verbeke, K.6
  • 291
    • 51049116216 scopus 로고    scopus 로고
    • Molecular analysis of the digestive microbiota in a gnotobiotic mouse model during antibiotic treatment: Influence of Saccharomyces boulardii
    • Barc MC, Charrin-Sarnel C, Rochet V, Bourlioux F, Sandré C, Boureau H, Doré J, Collignon A. 2008. Molecular analysis of the digestive microbiota in a gnotobiotic mouse model during antibiotic treatment: influence of Saccharomyces boulardii. Anaerobe 14: 229-233. https:// doi.org/10.1016/j.anaerobe.2008.04.003.
    • (2008) Anaerobe , vol.14 , pp. 229-233
    • Barc, M.C.1    Charrin-Sarnel, C.2    Rochet, V.3    Bourlioux, F.4    Sandré, C.5    Boureau, H.6    Doré, J.7    Collignon, A.8
  • 294
    • 84903975395 scopus 로고    scopus 로고
    • Saccharomyces boulardii administration changes gut microbiota and reduces hepatic steatosis, low-grade inflammation, and fat mass in obese and type 2 diabetic db/db mice
    • Everard A, Matamoros S, Geurts L, Delzenne NM, Cani PD. 2014. Saccharomyces boulardii administration changes gut microbiota and reduces hepatic steatosis, low-grade inflammation, and fat mass in obese and type 2 diabetic db/db mice. mBio 5: e01011-14. https://doi.org/10.1128/mBio.01011-14.
    • (2014) mBio , vol.5 , pp. e01011-e01114
    • Everard, A.1    Matamoros, S.2    Geurts, L.3    Delzenne, N.M.4    Cani, P.D.5
  • 295
    • 63849337600 scopus 로고    scopus 로고
    • Mechanism of cytokine modulation of epithelial tight junction barrier
    • Al-Sadi R, Boivin M, Ma T. 2009. Mechanism of cytokine modulation of epithelial tight junction barrier. Front Biosci (Landmark Ed) 14: 2765-2778.
    • (2009) Front Biosci (Landmark Ed) , vol.14 , pp. 2765-2778
    • Al-Sadi, R.1    Boivin, M.2    Ma, T.3
  • 297
    • 70049095676 scopus 로고    scopus 로고
    • Prevention of TNBS-induced colitis by different Lactobacillus and Bifidobacterium strains is associated with an expansion of gamma delta T and regulatory T cells of intestinal intraepithelial lymphocytes
    • Roselli M, Finamore A, Nuccitelli S, Carnevali P, Brigidi P, Vitali B, Nobili F, Rami R, Garaguso I, Mengheri E. 2009. Prevention of TNBS-induced colitis by different Lactobacillus and Bifidobacterium strains is associated with an expansion of gamma delta T and regulatory T cells of intestinal intraepithelial lymphocytes. Inflamm Bowel Dis 15: 1526-1536. https://doi.org/10.1002/ibd.20961.
    • (2009) Inflamm Bowel Dis , vol.15 , pp. 1526-1536
    • Roselli, M.1    Finamore, A.2    Nuccitelli, S.3    Carnevali, P.4    Brigidi, P.5    Vitali, B.6    Nobili, F.7    Rami, R.8    Garaguso, I.9    Mengheri, E.10
  • 298
    • 0036431805 scopus 로고    scopus 로고
    • Role of IL-10 in hepatocyte tight junction alteration in mouse model of experimental colitis
    • Mazzon E, Puzzolo D, Caputi AP, Cuzzocrea S. 2002. Role of IL-10 in hepatocyte tight junction alteration in mouse model of experimental colitis. Mol Med 8: 353-366.
    • (2002) Mol Med , vol.8 , pp. 353-366
    • Mazzon, E.1    Puzzolo, D.2    Caputi, A.P.3    Cuzzocrea, S.4
  • 299
    • 84941770102 scopus 로고    scopus 로고
    • A multihit model: Colitis lessons from the interleukin-10-deficient mouse
    • Keubler LM, Buettner M, Häger C, Bleich A. 2015. A multihit model: colitis lessons from the interleukin-10-deficient mouse. Inflamm Bowel Dis 21: 1967-1975. https://doi.org/10.1097/MIB.0000000000000468.
    • (2015) Inflamm Bowel Dis , vol.21 , pp. 1967-1975
    • Keubler, L.M.1    Buettner, M.2    Häger, C.3    Bleich, A.4
  • 300
    • 0027521572 scopus 로고
    • Interleukin-10-deficient mice develop chronic enterocolitis
    • Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W. 1993. Interleukin-10-deficient mice develop chronic enterocolitis. Cell 75: 263-274. https://doi.org/10.1016/0092-8674(93)80068-P.
    • (1993) Cell , vol.75 , pp. 263-274
    • Kuhn, R.1    Lohler, J.2    Rennick, D.3    Rajewsky, K.4    Muller, W.5
  • 301
    • 79958136845 scopus 로고    scopus 로고
    • Lactobacillus rhamnosus GG and Streptococcus thermophilus induce suppressor of cytokine signalling 3 (SOCS3) gene expression directly and indirectly via interleukin-10 in human primary macrophages
    • Latvala S, Miettinen M, Kekkonen RA, Korpela R, Julkunen I. 2011. Lactobacillus rhamnosus GG and Streptococcus thermophilus induce suppressor of cytokine signalling 3 (SOCS3) gene expression directly and indirectly via interleukin-10 in human primary macrophages. Clin Exp Immunol 165: 94-103. https://doi.org/10.1111/j.1365-2249.2011.04408.x.
    • (2011) Clin Exp Immunol , vol.165 , pp. 94-103
    • Latvala, S.1    Miettinen, M.2    Kekkonen, R.A.3    Korpela, R.4    Julkunen, I.5
  • 303
    • 80052604385 scopus 로고    scopus 로고
    • Regulation of the IL-10/IL-12 axis in human dendritic cells with probiotic bacteria
    • Gad M, Ravn P, Soborg DA, Lund-Jensen K, Ouwehand AC, Jensen SS. 2011. Regulation of the IL-10/IL-12 axis in human dendritic cells with probiotic bacteria. FEMS Immunol Med Microbiol 63: 93-107. https:// doi.org/10.1111/j.1574-695X.2011.00835.x.
    • (2011) FEMS Immunol Med Microbiol , vol.63 , pp. 93-107
    • Gad, M.1    Ravn, P.2    Soborg, D.A.3    Lund-Jensen, K.4    Ouwehand, A.C.5    Jensen, S.S.6
  • 304
    • 48749108299 scopus 로고    scopus 로고
    • Bifidobacterium infantis suppresses proinflammatory interleukin-17 production in murine splenocytes and dextran sodium sulfate-induced intestinal inflammation
    • Tanabe S, Kinuta Y, Saito Y. 2008. Bifidobacterium infantis suppresses proinflammatory interleukin-17 production in murine splenocytes and dextran sodium sulfate-induced intestinal inflammation. Int J Mol Med 22: 181-185.
    • (2008) Int J Mol Med , vol.22 , pp. 181-185
    • Tanabe, S.1    Kinuta, Y.2    Saito, Y.3
  • 305
    • 37549072218 scopus 로고    scopus 로고
    • Lactobacillus fermentum ACA-DC 179 displays probiotic potential in vitro and protects against trinitrobenzene sulfonic acid (TNBS)-induced colitis and Salmonella infection in murine models
    • Zoumpopoulou G, Foligne B, Christodoulou K, Grangetteb C, Pot B, Tsakalidou E. 2008. Lactobacillus fermentum ACA-DC 179 displays probiotic potential in vitro and protects against trinitrobenzene sulfonic acid (TNBS)-induced colitis and Salmonella infection in murine models. Int J Food Microbiol 121: 18-26. https://doi.org/10.1016/ j.ijfoodmicro.2007.10.013.
    • (2008) Int J Food Microbiol , vol.121 , pp. 18-26
    • Zoumpopoulou, G.1    Foligne, B.2    Christodoulou, K.3    Grangetteb, C.4    Pot, B.5    Tsakalidou, E.6
  • 306
    • 85046979518 scopus 로고    scopus 로고
    • Flexible cytokine production by macrophages and T cells in response to probiotic bacteria: A possible mechanism by which probiotics exert multifunctional immune regulatory activities
    • Shida K, Nanno M, Nagata S. 2011. Flexible cytokine production by macrophages and T cells in response to probiotic bacteria: a possible mechanism by which probiotics exert multifunctional immune regulatory activities. Gut Microbes 2: 109-114. https://doi.org/10.4161/ gmic.2.2.15661.
    • (2011) Gut Microbes , vol.2 , pp. 109-114
    • Shida, K.1    Nanno, M.2    Nagata, S.3
  • 307
    • 0036136282 scopus 로고    scopus 로고
    • Lactobacilli differentially modulate expression of cytokines and maturation surface markers in murine dendritic cells
    • Christensen HR, Frøkiaer H, Pestka JJ. 2002. Lactobacilli differentially modulate expression of cytokines and maturation surface markers in murine dendritic cells. J Immunol 168: 171-178. https://doi.org/10.4049/jimmunol.168.1.171.
    • (2002) J Immunol , vol.168 , pp. 171-178
    • Christensen, H.R.1    Frøkiaer, H.2    Pestka, J.J.3
  • 308
    • 33645549167 scopus 로고    scopus 로고
    • Lactic acid bacteria inducing a weak interleukin-12 and tumor necrosis factor alpha response in human dendritic cells inhibit strongly stimulating lactic acid bacteria but act synergistically with Gram-negative bacteria
    • Zeuthen LH, Christensen HR. 2006. Lactic acid bacteria inducing a weak interleukin-12 and tumor necrosis factor alpha response in human dendritic cells inhibit strongly stimulating lactic acid bacteria but act synergistically with Gram-negative bacteria. Clin Vaccine Immunol 13: 365-375. https://doi.org/10.1128/CVI.13.3.365-375.2006.
    • (2006) Clin Vaccine Immunol , vol.13 , pp. 365-375
    • Zeuthen, L.H.1    Christensen, H.R.2
  • 309
    • 84901236147 scopus 로고    scopus 로고
    • Yeast modulation of human dendritic cell cytokine secretion: An in vitro study
    • Smith IM, Christensen JE, Arneborg N, Jespersen L. 2014. Yeast modulation of human dendritic cell cytokine secretion: an in vitro study. PLoS One 9: e96595. https://doi.org/10.1371/journal.pone.0096595.
    • (2014) PLoS One , vol.9
    • Smith, I.M.1    Christensen, J.E.2    Arneborg, N.3    Jespersen, L.4
  • 310
    • 61849185743 scopus 로고    scopus 로고
    • Saccharomyces boulardii inhibits lipopolysaccharide-induced activation of human dendritic cells and T cell proliferation
    • Thomas S, Przesdzing I, Metzke D, Schmitz J, Radbruch A, Baumgart DC. 2009. Saccharomyces boulardii inhibits lipopolysaccharide-induced activation of human dendritic cells and T cell proliferation. Clin Exp Immunol 156: 78-87. https://doi.org/10.1111/j.1365-2249.2009.03878.x.
    • (2009) Clin Exp Immunol , vol.156 , pp. 78-87
    • Thomas, S.1    Przesdzing, I.2    Metzke, D.3    Schmitz, J.4    Radbruch, A.5    Baumgart, D.C.6
  • 312
    • 21744449027 scopus 로고    scopus 로고
    • In vitro screening of probiotic properties of Saccharomyces cerevisiae var. boulardii and food-borne Saccharomyces cerevisiae strains
    • van der Aa Kühle A, Skovgaard K, Jespersen L. 2005. In vitro screening of probiotic properties of Saccharomyces cerevisiae var. boulardii and food-borne Saccharomyces cerevisiae strains. Int J Food Microbiol 101: 29-39. https://doi.org/10.1016/j.ijfoodmicro.2004.10.039.
    • (2005) Int J Food Microbiol , vol.101 , pp. 29-39
    • Van Der Aa Kühle, A.1    Skovgaard, K.2    Jespersen, L.3
  • 313
    • 84921464952 scopus 로고    scopus 로고
    • In vitro characterization of the digestive stress response and immunomodulatory properties of microorganisms isolated from smear-ripened cheese
    • Adouard N, Foligné B, Dewulf J, Bouix M, Picque D, Bonnarme P. 2015. In vitro characterization of the digestive stress response and immunomodulatory properties of microorganisms isolated from smear-ripened cheese. Int J Food Microbiol 197: 98-107. https://doi.org/10.1016/ j.ijfoodmicro.2014.12.011.
    • (2015) Int J Food Microbiol , vol.197 , pp. 98-107
    • Adouard, N.1    Foligné, B.2    Dewulf, J.3    Bouix, M.4    Picque, D.5    Bonnarme, P.6
  • 315
    • 38549171792 scopus 로고    scopus 로고
    • Probiotic lactobacilli and VSL#3 induce enterocyte β-defensin 2
    • Schlee M, Harder J, Köten B, Stange EF, Wehkamp J, Fellermann K. 2008. Probiotic lactobacilli and VSL#3 induce enterocyte β-defensin 2. Clin Exp Immunol 151: 528-535. https://doi.org/10.1111/j.1365-2249.2007.03587.x.
    • (2008) Clin Exp Immunol , vol.151 , pp. 528-535
    • Schlee, M.1    Harder, J.2    Köten, B.3    Stange, E.F.4    Wehkamp, J.5    Fellermann, K.6
  • 316
    • 47849097120 scopus 로고    scopus 로고
    • Anti-inflammatory activity of probiotic Bifidobacterium: Enhancement of IL-10 production in peripheral blood mononuclear cells from ulcerative colitis patients and inhibition of IL-8 secretion in HT-29 cells
    • Imaoka A, Shima T, Kato K, Mizuno S, Uehara T, Matsumoto S, Setoyana H, Hara T, Umesaki Y. 2008. Anti-inflammatory activity of probiotic Bifidobacterium: enhancement of IL-10 production in peripheral blood mononuclear cells from ulcerative colitis patients and inhibition of IL-8 secretion in HT-29 cells. World J Gastroenterol 14: 2511-2516. https:// doi.org/10.3748/wjg.14.2511.
    • (2008) World J Gastroenterol , vol.14 , pp. 2511-2516
    • Imaoka, A.1    Shima, T.2    Kato, K.3    Mizuno, S.4    Uehara, T.5    Matsumoto, S.6    Setoyana, H.7    Hara, T.8    Umesaki, Y.9
  • 317
    • 84868136017 scopus 로고    scopus 로고
    • Epigenetic imprinting by commensal probiotics inhibits the IL-23/IL-17 axis in an in vitro model of the intestinal mucosal immune system
    • Ghadimi D, Helwig U, Schrezenmeir J, Heller KJ, de Vrese M. 2012. Epigenetic imprinting by commensal probiotics inhibits the IL-23/IL-17 axis in an in vitro model of the intestinal mucosal immune system. J Leukoc Biol 92: 895-911. https://doi.org/10.1189/jlb.0611286.
    • (2012) J Leukoc Biol , vol.92 , pp. 895-911
    • Ghadimi, D.1    Helwig, U.2    Schrezenmeir, J.3    Heller, K.J.4    de Vrese, M.5
  • 318
    • 84892531568 scopus 로고    scopus 로고
    • Bifidobacterium longum alleviates dextran sulfate sodium-induced colitis by suppressing IL-17A response: Involvement of intestinal epithelial costimulatory molecules
    • Miyauchi E, Ogita T, Miyamoto J, Kawamoto S, Morita H, Ohno H, Suzuki T, Tanabe S. 2013. Bifidobacterium longum alleviates dextran sulfate sodium-induced colitis by suppressing IL-17A response: involvement of intestinal epithelial costimulatory molecules. PLoS One 8: e79735. https://doi.org/10.1371/journal.pone.0079735.
    • (2013) PLoS One , vol.8
    • Miyauchi, E.1    Ogita, T.2    Miyamoto, J.3    Kawamoto, S.4    Morita, H.5    Ohno, H.6    Suzuki, T.7    Tanabe, S.8
  • 319
    • 84929340454 scopus 로고    scopus 로고
    • Lactobacillus acidophilus suppresses colitis-associated activation of the IL-23/Th17 axis
    • Chen L, Zou Y, Peng J, Lu F, Yin Y, Li F, Yang J. 2015. Lactobacillus acidophilus suppresses colitis-associated activation of the IL-23/Th17 axis. J Immunol Res 2015: 909514. https://doi.org/10.1155/2015/909514.
    • (2015) J Immunol Res , vol.2015 , pp. 909514
    • Chen, L.1    Zou, Y.2    Peng, J.3    Lu, F.4    Yin, Y.5    Li, F.6    Yang, J.7
  • 320
    • 80053117777 scopus 로고    scopus 로고
    • Suppression of Th17 response by Streptococcus thermophilus ST28 through induction of IFN-γ
    • Ogita T, Tanii Y, Morita H, Suzuki T, Tanabe S. 2011. Suppression of Th17 response by Streptococcus thermophilus ST28 through induction of IFN-γ. Int J Mol Med 28: 817-822. https://doi.org/10.3892/ ijmm.2011.755.
    • (2011) Int J Mol Med , vol.28 , pp. 817-822
    • Ogita, T.1    Tanii, Y.2    Morita, H.3    Suzuki, T.4    Tanabe, S.5
  • 321
    • 70349509222 scopus 로고    scopus 로고
    • Lactobacillus fermentum CECT 5716 prevents and reverts intestinal damage on TNBS-induced colitis in mice
    • Mañé J, Lorén V, Pedrosa E, Ojanguren I, Xaus J, Cabré E, Domènech E, Gassull MA. 2009. Lactobacillus fermentum CECT 5716 prevents and reverts intestinal damage on TNBS-induced colitis in mice. Inflamm Bowel Dis 15: 1155-1163. https://doi.org/10.1002/ibd.20908.
    • (2009) Inflamm Bowel Dis , vol.15 , pp. 1155-1163
    • Mañé, J.1    Lorén, V.2    Pedrosa, E.3    Ojanguren, I.4    Xaus, J.5    Cabré, E.6    Domènech, E.7    Gassull, M.A.8
  • 322
    • 84902972002 scopus 로고    scopus 로고
    • A probiotic preparation Duolac-Gold ameliorates dextran sulphate sodium-induced mouse colitis by downregulating the expression of IL-6
    • Yoon H, Yoon Y, Kim M, Chung M, Yum D. 2014. A probiotic preparation Duolac-Gold ameliorates dextran sulphate sodium-induced mouse colitis by downregulating the expression of IL-6. Toxicol Res 30: 27-32. https://doi.org/10.5487/TR.2014.30.1.027.
    • (2014) Toxicol Res , vol.30 , pp. 27-32
    • Yoon, H.1    Yoon, Y.2    Kim, M.3    Chung, M.4    Yum, D.5
  • 323
    • 40149084454 scopus 로고    scopus 로고
    • Lactic acid bacteria inhibit proinflammatory cytokine expression and bacterial glycosaminoglycan degradation activity in dextran sulfate sodiuminduced colitic mice
    • Lee HS, Han SY, Bae EA, Huh CS, Ahn YT, Lee JH, Kim DH. 2008. Lactic acid bacteria inhibit proinflammatory cytokine expression and bacterial glycosaminoglycan degradation activity in dextran sulfate sodiuminduced colitic mice. Int Immunopharmacol 8: 574-580. https://doi.org/10.1016/j.intimp.2008.01.009.
    • (2008) Int Immunopharmacol , vol.8 , pp. 574-580
    • Lee, H.S.1    Han, S.Y.2    Bae, E.A.3    Huh, C.S.4    Ahn, Y.T.5    Lee, J.H.6    Kim, D.H.7
  • 324
    • 84894266945 scopus 로고    scopus 로고
    • Probiotic Lactobacillus rhamnosus inhibits the formation of neutrophil extracellular traps
    • Vong L, Lorentz RJ, Assa A, Glogauer M, Sherman PM. 2014. Probiotic Lactobacillus rhamnosus inhibits the formation of neutrophil extracellular traps. J Immunol 192: 1870-1877. https://doi.org/10.4049/ jimmunol.1302286.
    • (2014) J Immunol , vol.192 , pp. 1870-1877
    • Vong, L.1    Lorentz, R.J.2    Assa, A.3    Glogauer, M.4    Sherman, P.M.5
  • 325
    • 77950797408 scopus 로고    scopus 로고
    • Clinical trial: Probiotic treatment of acute distal ulcerative colitis with rectally administered Escherichia coli Nissle 1917 (EcN)
    • Matthes H, Krummenerl T, Giensch M, Wolff C, Schulze J. 2010. Clinical trial: probiotic treatment of acute distal ulcerative colitis with rectally administered Escherichia coli Nissle 1917 (EcN). BMC Complement Altern Med 10: 13. https://doi.org/10.1186/1472-6882-10-13.
    • (2010) BMC Complement Altern Med , vol.10 , pp. 13
    • Matthes, H.1    Krummenerl, T.2    Giensch, M.3    Wolff, C.4    Schulze, J.5
  • 326
    • 0033592203 scopus 로고    scopus 로고
    • Non-pathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis: A randomised trial
    • Rembacken BJ, Snelling AM, Hawkey PM, Chalmers DM, Axon ATR. 1999. Non-pathogenic Escherichia coli versus mesalazine for the treatment of ulcerative colitis: a randomised trial. Lancet 354: 635-639. https://doi.org/10.1016/S0140-6736(98)06343-0.
    • (1999) Lancet , vol.354 , pp. 635-639
    • Rembacken, B.J.1    Snelling, A.M.2    Hawkey, P.M.3    Chalmers, D.M.4    Axon, A.T.R.5
  • 327
    • 84887803390 scopus 로고    scopus 로고
    • Animal models of ulcerative colitis and their application in drug research
    • Low D, Nguyen DD, Mizoguchi E. 2013. Animal models of ulcerative colitis and their application in drug research. Drug Des Devel Ther 7: 1341-1357. https://doi.org/10.2147/DDDT.S40107.
    • (2013) Drug Des Devel Ther , vol.7 , pp. 1341-1357
    • Low, D.1    Nguyen, D.D.2    Mizoguchi, E.3
  • 328
    • 84897028931 scopus 로고    scopus 로고
    • Small animal models for the study of Clostridium difficile disease pathogenesis
    • Hutton ML, Mackin KE, Chakravorty A, Lyras D. 2014. Small animal models for the study of Clostridium difficile disease pathogenesis. FEMS Microbiol Lett 352: 140-149. https://doi.org/10.1111/1574-6968.12367.
    • (2014) FEMS Microbiol Lett , vol.352 , pp. 140-149
    • Hutton, M.L.1    Mackin, K.E.2    Chakravorty, A.3    Lyras, D.4
  • 329
    • 84922888723 scopus 로고    scopus 로고
    • Predictive values of models of Clostridium difficile infection
    • Chilton C, Freeman J. 2015. Predictive values of models of Clostridium difficile infection. Infect Dis Clin North Am 29: 163-177. https://doi.org/10.1016/j.idc.2014.11.011.
    • (2015) Infect Dis Clin North Am , vol.29 , pp. 163-177
    • Chilton, C.1    Freeman, J.2
  • 332
    • 70349430938 scopus 로고    scopus 로고
    • Bile acids: Regulation of synthesis
    • Chiang JY. 2009. Bile acids: regulation of synthesis. J Lipid Res 50: 1955-1966. https://doi.org/10.1194/jlr.R900010-JLR200.
    • (2009) J Lipid Res , vol.50 , pp. 1955-1966
    • Chiang, J.Y.1
  • 334
    • 34447630803 scopus 로고    scopus 로고
    • Use of probiotic Lactobacillus preparation to prevent diarrhoea associated with antibiotics: Randomised double blind placebo controlled trial
    • Hickson M, D’Souza AL, Muthu N, Rogers TR, Want S, Rajkumar C, Bulpitt CJ. 2007. Use of probiotic Lactobacillus preparation to prevent diarrhoea associated with antibiotics: randomised double blind placebo controlled trial. BMJ 335: 80. https://doi.org/10.1136/ bmj.39231.599815.55.
    • (2007) BMJ , vol.335 , pp. 80
    • Hickson, M.1    D’Souza, A.L.2    Muthu, N.3    Rogers, T.R.4    Want, S.5    Rajkumar, C.6    Bulpitt, C.J.7
  • 335
    • 77954424086 scopus 로고    scopus 로고
    • Dose-response efficacy of a proprietary probiotic formula of Lactobacillus acidophilus CL1285 and Lactobacillus casei LBC80R for antibiotic-associated diarrhea and Clostridium difficile-associated diarrhea prophylaxis in adult patients
    • Gao XW, Mubasher M, Fang CY, Reifer C, Miller LE. 2010. Dose-response efficacy of a proprietary probiotic formula of Lactobacillus acidophilus CL1285 and Lactobacillus casei LBC80R for antibiotic-associated diarrhea and Clostridium difficile-associated diarrhea prophylaxis in adult patients. Am J Gastroenterol 105: 1636-1641. https://doi.org/10.1038/ ajg.2010.11.
    • (2010) Am J Gastroenterol , vol.105 , pp. 1636-1641
    • Gao, X.W.1    Mubasher, M.2    Fang, C.Y.3    Reifer, C.4    Miller, L.E.5
  • 336
    • 0034839847 scopus 로고    scopus 로고
    • Lack of effect of Lactobacillus GG on antibiotic-associated diarrhea: A randomized, placebo-controlled trial
    • Thomas MR, Litin SC, Osmon DR, Corr AP, Weaver AL, Lohse CM. 2001. Lack of effect of Lactobacillus GG on antibiotic-associated diarrhea: a randomized, placebo-controlled trial. Mayo Clin Proc 76: 883-889. https://doi.org/10.4065/76.9.883.
    • (2001) Mayo Clin Proc , vol.76 , pp. 883-889
    • Thomas, M.R.1    Litin, S.C.2    Osmon, D.R.3    Corr, A.P.4    Weaver, A.L.5    Lohse, C.M.6
  • 337
    • 2942704185 scopus 로고    scopus 로고
    • Clostridium difficile pilot study: Effects of probiotic supplementation on the incidence of C. difficile diarrhoea
    • Plummer S, Weaver MA, Harris JC, Dee P, Huter J. 2004. Clostridium difficile pilot study: effects of probiotic supplementation on the incidence of C. difficile diarrhoea. Int Microbiol 7: 59-62.
    • (2004) Int Microbiol , vol.7 , pp. 59-62
    • Plummer, S.1    Weaver, M.A.2    Harris, J.C.3    Dee, P.4    Huter, J.5
  • 338
    • 24044452865 scopus 로고    scopus 로고
    • Probiotics for recurrent Clostridium difficile disease
    • Lawrence SJ, Korzenik JR, Mundy LM. 2005. Probiotics for recurrent Clostridium difficile disease. J Med Microbiol 54: 905-906. https:// doi.org/10.1099/jmm.0.46096-0.
    • (2005) J Med Microbiol , vol.54 , pp. 905-906
    • Lawrence, S.J.1    Korzenik, J.R.2    Mundy, L.M.3
  • 339
    • 34648846061 scopus 로고    scopus 로고
    • Probiotics as prophylactic agents against antibiotic-associated diarrhea in hospitalized patients
    • Stein G, Nanim R, Karniel E, Moskowitz I, Zeidman A. 2007. Probiotics as prophylactic agents against antibiotic-associated diarrhea in hospitalized patients. Harefuah 146: 520-522, 575.
    • (2007) Harefuah , vol.146
    • Stein, G.1    Nanim, R.2    Karniel, E.3    Moskowitz, I.4    Zeidman, A.5
  • 340
    • 38649108836 scopus 로고    scopus 로고
    • Effect of a fermented milk combining Lactobacillus acidophilus CL1285 and Lactobacillus casei in the prevention of antibiotic-associated diarrhea: A randomized, double-blind, placebocontrolled trial
    • Beausoleil M, Fortier N, Guénette S, L’Ecuyer A, Savoie M, Franco M, Lachaîne J, Weiss K. 2007. Effect of a fermented milk combining Lactobacillus acidophilus CL1285 and Lactobacillus casei in the prevention of antibiotic-associated diarrhea: a randomized, double-blind, placebocontrolled trial. Can J Gastroenterol 21: 732-736. https://doi.org/10.1155/2007/720205.
    • (2007) Can J Gastroenterol , vol.21 , pp. 732-736
    • Beausoleil, M.1    Fortier, N.2    Guénette, S.3    L’Ecuyer, A.4    Savoie, M.5    Franco, M.6    Lachaîne, J.7    Weiss, K.8
  • 341
    • 55949121333 scopus 로고    scopus 로고
    • Feasibility and tolerability of probiotics for prevention of antibiotic-associated diarrhoea in hospitalized US military veterans
    • Safdar N, Barigala R, Said A, McKinley L. 2008. Feasibility and tolerability of probiotics for prevention of antibiotic-associated diarrhoea in hospitalized US military veterans. J Clin Pharm Ther 33: 663-668. https:// doi.org/10.1111/j.1365-2710.2008.00980.x.
    • (2008) J Clin Pharm Ther , vol.33 , pp. 663-668
    • Safdar, N.1    Barigala, R.2    Said, A.3    McKinley, L.4
  • 342
    • 0042922857 scopus 로고    scopus 로고
    • Lactobacillus plantarum 299v for the treatment of recurrent Clostridium difficile-associated diarrhoea: A double-blind, placebo-controlled trial
    • Wullt M, Hagslätt M-LJ, Odenholt I. 2003. Lactobacillus plantarum 299v for the treatment of recurrent Clostridium difficile-associated diarrhoea: a double-blind, placebo-controlled trial. Scand J Infect Dis 35: 365-367. https://doi.org/10.1080/00365540310010985.
    • (2003) Scand J Infect Dis , vol.35 , pp. 365-367
    • Wullt, M.1    Hagslätt, M.-L.J.2    Odenholt, I.3
  • 343
    • 77949370649 scopus 로고    scopus 로고
    • Efficacy of BIO K+ CL1285 in the reduction of antibiotic-associated diarrhea-a placebo controlled double-blind randomized, multi-center study
    • Sampalis J, Psaradellis E, Rampakakis E. 2010. Efficacy of BIO K+ CL1285 in the reduction of antibiotic-associated diarrhea-a placebo controlled double-blind randomized, multi-center study. Arch Med Sci 6: 56-64. https://doi.org/10.5114/aoms.2010.13508.
    • (2010) Arch Med Sci , vol.6 , pp. 56-64
    • Sampalis, J.1    Psaradellis, E.2    Rampakakis, E.3
  • 345
    • 15044347921 scopus 로고    scopus 로고
    • Saccharomyces boulardii in the prevention of antibiotic-associated diarrhoea in children: A randomized double-blind placebo-controlled trial
    • Kotowska M, Albrecht P, Szajewska H. 2005. Saccharomyces boulardii in the prevention of antibiotic-associated diarrhoea in children: a randomized double-blind placebo-controlled trial. Aliment Pharmacol Ther 21: 583-590. https://doi.org/10.1111/j.1365-2036.2005.02356.x.
    • (2005) Aliment Pharmacol Ther , vol.21 , pp. 583-590
    • Kotowska, M.1    Albrecht, P.2    Szajewska, H.3
  • 346
    • 0024562960 scopus 로고
    • Prevention of antibiotic-associated diarrhea by Saccharomyces boulardii: A prospective study
    • Surawicz CM, Elmer GW, Speelman P, McFarland LV, Chinn J, van Belle G. 1989. Prevention of antibiotic-associated diarrhea by Saccharomyces boulardii: a prospective study. Gastroenterology 96: 981-988. https:// doi.org/10.1016/0016-5085(89)91613-2.
    • (1989) Gastroenterology , vol.96 , pp. 981-988
    • Surawicz, C.M.1    Elmer, G.W.2    Speelman, P.3    McFarland, L.V.4    Chinn, J.5    van Belle, G.6
  • 347
    • 0024317452 scopus 로고
    • Treatment of recurrent Clostridium difficile colitis with vancomycin and Saccharomyces boulardii
    • Surawicz CM, McFarland L, Elmer G, Chinn J. 1989. Treatment of recurrent Clostridium difficile colitis with vancomycin and Saccharomyces boulardii. Am J Gastroenterol 84: 1285-1287.
    • (1989) Am J Gastroenterol , vol.84 , pp. 1285-1287
    • Surawicz, C.M.1    McFarland, L.2    Elmer, G.3    Chinn, J.4
  • 349
    • 0000650388 scopus 로고    scopus 로고
    • The lack of therapeutic effect of Saccharomyces boulardii in the prevention of antibiotic-related diarrhoea in elderly patients
    • Lewis S, Potts L, Barry R. 1998. The lack of therapeutic effect of Saccharomyces boulardii in the prevention of antibiotic-related diarrhoea in elderly patients. J Infect 36: 171-174. https://doi.org/10.1016/ S0163-4453(98)80008-X.
    • (1998) J Infect , vol.36 , pp. 171-174
    • Lewis, S.1    Potts, L.2    Barry, R.3
  • 350
    • 33645544410 scopus 로고    scopus 로고
    • Prophylactic Saccharomyces boulardii in the prevention of antibiotic-associated diarrhea: A prospective study
    • Can M, Beşirbellioglu BA, Avci IY, Beker CM, Pahsa A. 2006. Prophylactic Saccharomyces boulardii in the prevention of antibiotic-associated diarrhea: a prospective study. Med Sci Monit 12: PI19-PI22.
    • (2006) Med Sci Monit , vol.12 , pp. PI19-PI22
    • Can, M.1    Beşirbellioglu, B.A.2    Avci, I.Y.3    Beker, C.M.4    Pahsa, A.5
  • 351
    • 61349161828 scopus 로고    scopus 로고
    • Lactobacillus GG treatment ameliorates alcohol-induced intestinal oxidative stress, gut leakiness, and liver injury in a rat model of alcoholic steatohepatitis
    • Forsyth CB, Farhadi A, Jakate SM, Tang Y, Shaikh M, Keshavarzian A. 2009. Lactobacillus GG treatment ameliorates alcohol-induced intestinal oxidative stress, gut leakiness, and liver injury in a rat model of alcoholic steatohepatitis. Alcohol 43: 163-172. https://doi.org/10.1016/ j.alcohol.2008.12.009.
    • (2009) Alcohol , vol.43 , pp. 163-172
    • Forsyth, C.B.1    Farhadi, A.2    Jakate, S.M.3    Tang, Y.4    Shaikh, M.5    Keshavarzian, A.6
  • 352
    • 84893170319 scopus 로고    scopus 로고
    • Lactobacillus GG restoration of the gliadin induced epithelial barrier disruption: The role of cellular polyamines
    • Orlando A, Linsalata M, Notarnicola M, Tutino V, Russo F. 2014. Lactobacillus GG restoration of the gliadin induced epithelial barrier disruption: the role of cellular polyamines. BMC Microbiol 14: 19. https://doi.org/10.1186/1471-2180-14-19.
    • (2014) BMC Microbiol , vol.14 , pp. 19
    • Orlando, A.1    Linsalata, M.2    Notarnicola, M.3    Tutino, V.4    Russo, F.5
  • 353
    • 42049098409 scopus 로고    scopus 로고
    • Probiotics ameliorate the hydrogen peroxide-induced epithelial barrier disruption by a PKC-and MAP kinase-dependent mechanism
    • Seth A, Yan F, Polk DB, Rao RK. 2008. Probiotics ameliorate the hydrogen peroxide-induced epithelial barrier disruption by a PKC-and MAP kinase-dependent mechanism. Am J Physiol Gastrointest Liver Physiol 294: G1060-G1069. https://doi.org/10.1152/ajpgi.00202.2007.
    • (2008) Am J Physiol Gastrointest Liver Physiol , vol.294 , pp. G1060-G1069
    • Seth, A.1    Yan, F.2    Polk, D.B.3    Rao, R.K.4
  • 354
    • 0038460949 scopus 로고    scopus 로고
    • Live probiotics protect intestinal epithelial cells from the effects of infection with enteroinvasive Escherichia coli (EIEC)
    • Resta-Lenert S, Barrett KE. 2003. Live probiotics protect intestinal epithelial cells from the effects of infection with enteroinvasive Escherichia coli (EIEC). Gut 52: 988-997. https://doi.org/10.1136/gut.52.7.988.
    • (2003) Gut , vol.52 , pp. 988-997
    • Resta-Lenert, S.1    Barrett, K.E.2
  • 355
    • 84896975706 scopus 로고    scopus 로고
    • Effect of Lactobacillus plantarum on diarrhea and intestinal barrier function of young piglets challenged with enterotoxigenic Escherichia coli K88
    • Yang KM, Jiang ZY, Zheng CT, Wang L, Yang XF. 2014. Effect of Lactobacillus plantarum on diarrhea and intestinal barrier function of young piglets challenged with enterotoxigenic Escherichia coli K88. J Anim Sci 92: 1496-1503. https://doi.org/10.2527/jas.2013-6619.
    • (2014) J Anim Sci , vol.92 , pp. 1496-1503
    • Yang, K.M.1    Jiang, Z.Y.2    Zheng, C.T.3    Wang, L.4    Yang, X.F.5
  • 356
    • 0042236701 scopus 로고    scopus 로고
    • Oral and intestinal microflora in 5-fluorouracil treated rats, translocation to cervical and mesenteric lymph nodes and effects of probiotic bacteria
    • Von Bültzingslöwen I, Adlerberth I, Wold A, Dahlén G, Jontell M. 2003. Oral and intestinal microflora in 5-fluorouracil treated rats, translocation to cervical and mesenteric lymph nodes and effects of probiotic bacteria. Oral Microbiol Immunol 18: 278-284. https://doi.org/10.1034/ j.1399-302X.2003.00075.x.
    • (2003) Oral Microbiol Immunol , vol.18 , pp. 278-284
    • Von Bültzingslöwen, I.1    Adlerberth, I.2    Wold, A.3    Dahlén, G.4    Jontell, M.5
  • 359
    • 33750364231 scopus 로고    scopus 로고
    • Probiotics prevent bacterial translocation and improve intestinal barrier function in rats following chronic psychological stress
    • Zareie M, Johnson-Henry K, Jury J, Yang P-C, Ngan B-Y, McKay DM, Soderholm JD, Perdue MH, Sherman PM. 2006. Probiotics prevent bacterial translocation and improve intestinal barrier function in rats following chronic psychological stress. Gut 55: 1553-1560. https:// doi.org/10.1136/gut.2005.080739.
    • (2006) Gut , vol.55 , pp. 1553-1560
    • Zareie, M.1    Johnson-Henry, K.2    Jury, J.3    Yang, P.-C.4    Ngan, B.-Y.5    McKay, D.M.6    Soderholm, J.D.7    Perdue, M.H.8    Sherman, P.M.9
  • 360
    • 0037304989 scopus 로고    scopus 로고
    • Saccharomyces boulardii interferes with enterohemorrhagic Escherichia coli-induced signaling pathways in T84 cells
    • Dahan S, Dalmasso G, Imbert V, Peyron J, Rampal P, Czerucka D. 2003. Saccharomyces boulardii interferes with enterohemorrhagic Escherichia coli-induced signaling pathways in T84 cells. Infect Immun 71: 766-773. https://doi.org/10.1128/IAI.71.2.766-773.2003.
    • (2003) Infect Immun , vol.71 , pp. 766-773
    • Dahan, S.1    Dalmasso, G.2    Imbert, V.3    Peyron, J.4    Rampal, P.5    Czerucka, D.6
  • 361
    • 33747699728 scopus 로고    scopus 로고
    • Saccharomyces boulardii inhibits ERK1/2 mitogen-activated protein kinase activation both in vitro and in vivo and protects against Clostridium difficile toxin A-induced enteritis
    • Chen X, Kokkotou EG, Mustafa N, Bhaskar KR, Sougioultzis S, O’Brien M, Pothoulakis C, Kelly CP. 2006. Saccharomyces boulardii inhibits ERK1/2 mitogen-activated protein kinase activation both in vitro and in vivo and protects against Clostridium difficile toxin A-induced enteritis. J Biol Chem 281: 24449-24454. https://doi.org/10.1074/jbc.M605200200.
    • (2006) J Biol Chem , vol.281 , pp. 24449-24454
    • Chen, X.1    Kokkotou, E.G.2    Mustafa, N.3    Bhaskar, K.R.4    Sougioultzis, S.5    O’Brien, M.6    Pothoulakis, C.7    Kelly, C.P.8
  • 362
    • 0040699797 scopus 로고    scopus 로고
    • Immunological and trophical effects of Saccharomyces boulardi on the small intestine in healthy human volunteers
    • Jahn H-U, Ullrich R, Schneider T, Liehr R-M, Schieferdecker HL, Holst H, Zeitz M. 1996. Immunological and trophical effects of Saccharomyces boulardi on the small intestine in healthy human volunteers. Digestion 57: 95-104.
    • (1996) Digestion , vol.57 , pp. 95-104
    • Jahn, H.-U.1    Ullrich, R.2    Schneider, T.3    Liehr, R.-M.4    Schieferdecker, H.L.5    Holst, H.6    Zeitz, M.7
  • 364
    • 12344249654 scopus 로고    scopus 로고
    • Synbiotic therapy (Bifidobacterium longum/ Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: A randomised controlled pilot trial
    • Furrie E, Macfarlane S, Kennedy A, Cummings JH, Walsh SV, O’Neil DA, Macfarlane GT. 2005. Synbiotic therapy (Bifidobacterium longum/ Synergy 1) initiates resolution of inflammation in patients with active ulcerative colitis: a randomised controlled pilot trial. Gut 54: 242-249. https://doi.org/10.1136/gut.2004.044834.
    • (2005) Gut , vol.54 , pp. 242-249
    • Furrie, E.1    Macfarlane, S.2    Kennedy, A.3    Cummings, J.H.4    Walsh, S.V.5    O’Neil, D.A.6    Macfarlane, G.T.7
  • 365
    • 0030611915 scopus 로고    scopus 로고
    • Double-blind comparison of an oral Escherichia coli preparation and mesalazine in maintaining remission of ulcerative colitis
    • Kruis W, Schütz E, Fric P, Fixa B, Judmaier G, Stolte M. 1997. Double-blind comparison of an oral Escherichia coli preparation and mesalazine in maintaining remission of ulcerative colitis. Aliment Pharmacol Ther 11: 853-858. https://doi.org/10.1046/j.1365-2036.1997.00225.x.
    • (1997) Aliment Pharmacol Ther , vol.11 , pp. 853-858
    • Kruis, W.1    Schütz, E.2    Fric, P.3    Fixa, B.4    Judmaier, G.5    Stolte, M.6
  • 368
    • 72049119604 scopus 로고    scopus 로고
    • The probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately active ulcerative colitis
    • Sood A, Midha V, Makharia GK, Ahuja V, Singal D, Goswami P, Tandon RK. 2009. The probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately active ulcerative colitis. Clin Gastroenterol Hepatol 7: 1202-1209. https://doi.org/10.1016/j.cgh.2009.07.016.
    • (2009) Clin Gastroenterol Hepatol , vol.7 , pp. 1202-1209
    • Sood, A.1    Midha, V.2    Makharia, G.K.3    Ahuja, V.4    Singal, D.5    Goswami, P.6    Tandon, R.K.7
  • 369
    • 0032815979 scopus 로고    scopus 로고
    • Impact on the composition of the faecal flora by a new probiotic preparation: Preliminary data on maintenance treatment of patients with ulcerative colitis
    • Venturi A, Gionchetti P, Rizzello F, Johansson R, Zucconi E, Brigidi P, Matteuzzi D, Campieri M. 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: 1103-1108. https://doi.org/10.1046/j.1365-2036.1999.00560.x.
    • (1999) Aliment Pharmacol Ther , vol.13 , pp. 1103-1108
    • Venturi, A.1    Gionchetti, P.2    Rizzello, F.3    Johansson, R.4    Zucconi, E.5    Brigidi, P.6    Matteuzzi, D.7    Campieri, M.8
  • 370
    • 84866254110 scopus 로고    scopus 로고
    • The effect of bifid triple viable on immune function of patients with ulcerative colitis
    • Li G, Zeng S, Liao W, Lv N. 2012. The effect of bifid triple viable on immune function of patients with ulcerative colitis. Gastroenterol Res Pract 2012: 404752. https://doi.org/10.1155/2012/404752.
    • (2012) Gastroenterol Res Pract , vol.2012 , pp. 404752
    • Li, G.1    Zeng, S.2    Liao, W.3    Lv, N.4
  • 371
    • 0037783259 scopus 로고    scopus 로고
    • A pilot trial of Saccharomyces boulardii in ulcerative colitis
    • Guslandi M, Giollo P, Testoni PA. 2003. A pilot trial of Saccharomyces boulardii in ulcerative colitis. Eur J Gastroenterol Hepatol 15: 697-698. https://doi.org/10.1097/00042737-200306000-00017.
    • (2003) Eur J Gastroenterol Hepatol , vol.15 , pp. 697-698
    • Guslandi, M.1    Giollo, P.2    Testoni, P.A.3
  • 373
    • 9144236376 scopus 로고    scopus 로고
    • Low-dose balsalazide plus a high-potency probiotic preparation is more effective than balsalazide alone or mesalazine in the treatment of acute mild-to-moderate ulcerative colitis
    • Tursi A, Brandimarte G, Giorgetti GM, Forti G, Modeo ME, Gigliobianco A. 2004. Low-dose balsalazide plus a high-potency probiotic preparation is more effective than balsalazide alone or mesalazine in the treatment of acute mild-to-moderate ulcerative colitis. Med Sci Monit 10: PI126-PI131.
    • (2004) Med Sci Monit , vol.10 , pp. PI126-PI131
    • Tursi, A.1    Brandimarte, G.2    Giorgetti, G.M.3    Forti, G.4    Modeo, M.E.5    Gigliobianco, A.6
  • 374
    • 77955761858 scopus 로고    scopus 로고
    • Immunosuppressive effects via human intestinal dendritic cells of probiotic bacteria and steroids in the treatment of acute ulcerative colitis
    • Ng SC, Plamondon S, Kamm MA, Hart AL, Al-Hassi HO, Guenther T, Stagg AJ, Knight SC. 2010. Immunosuppressive effects via human intestinal dendritic cells of probiotic bacteria and steroids in the treatment of acute ulcerative colitis. Inflamm Bowel Dis 16: 1286-1298. https://doi.org/10.1002/ibd.21222.
    • (2010) Inflamm Bowel Dis , vol.16 , pp. 1286-1298
    • Ng, S.C.1    Plamondon, S.2    Kamm, M.A.3    Hart, A.L.4    Al-Hassi, H.O.5    Guenther, T.6    Stagg, A.J.7    Knight, S.C.8
  • 376
    • 79953069242 scopus 로고    scopus 로고
    • 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
    • Wildt S, Nordgaard I, Hansen U, Brockmann E, Rumessen JJ. 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: 115-121. https://doi.org/10.1016/j.crohns.2010.11.004.
    • (2011) J Crohns Colitis , vol.5 , pp. 115-121
    • Wildt, S.1    Nordgaard, I.2    Hansen, U.3    Brockmann, E.4    Rumessen, J.J.5
  • 377
    • 33847109825 scopus 로고    scopus 로고
    • A comparative study of the preventative effects exerted by two probiotics, Lactobacillus reuteri and Lactobacillus fermentum, in the trinitrobenzenesulfonic acid model of rat colitis
    • Peran L, Sierra S, Comalada M, Lara-Villoslada F, Bailón E, Nieto A, Concha A, Olivares M, Zarzuelo A, Xaus J, Gálvez J. 2007. A comparative study of the preventative effects exerted by two probiotics, Lactobacillus reuteri and Lactobacillus fermentum, in the trinitrobenzenesulfonic acid model of rat colitis. Br J Nutr 97: 96-103. https://doi.org/10.1017/S0007114507257770.
    • (2007) Br J Nutr , vol.97 , pp. 96-103
    • Peran, L.1    Sierra, S.2    Comalada, M.3    Lara-Villoslada, F.4    Bailón, E.5    Nieto, A.6    Concha, A.7    Olivares, M.8    Zarzuelo, A.9    Xaus, J.10    Gálvez, J.11
  • 378
    • 84904648536 scopus 로고    scopus 로고
    • Amelioration of colitis in mouse model by exploring antioxidative potentials of an indigenous probiotic strain of Lactobacillus fermentum Lf1
    • Chauhan R, Vasanthakumari AS, Panwar H, Mallapa RH, Duary RK, Batish VK, Grover S. 2014. Amelioration of colitis in mouse model by exploring antioxidative potentials of an indigenous probiotic strain of Lactobacillus fermentum Lf1. Biomed Res Int 2014: 206732. https://doi.org/10.1155/2014/206732.
    • (2014) Biomed Res Int , vol.2014 , pp. 206732
    • Chauhan, R.1    Vasanthakumari, A.S.2    Panwar, H.3    Mallapa, R.H.4    Duary, R.K.5    Batish, V.K.6    Grover, S.7
  • 379
    • 65349153174 scopus 로고    scopus 로고
    • Lactobacillus reuteri prevents colitis by reducing P-selectin-associated leukocyte-and platelet-endothelial cell interactions
    • Schreiber O, Petersson J, Phillipson M, Perry M, Roos S, Holm L. 2009. Lactobacillus reuteri prevents colitis by reducing P-selectin-associated leukocyte-and platelet-endothelial cell interactions. Am J Physiol Gastrointest Liver Physiol 296: G534-G542. https://doi.org/10.1152/ ajpgi.90470.2008.
    • (2009) Am J Physiol Gastrointest Liver Physiol , vol.296 , pp. G534-G542
    • Schreiber, O.1    Petersson, J.2    Phillipson, M.3    Perry, M.4    Roos, S.5    Holm, L.6
  • 380
    • 84920111658 scopus 로고    scopus 로고
    • Bifidobacterium infantis attenuates colitis by regulating T cell subset responses
    • Zuo L, Yuan K, Yu L, Meng Q, Chung PC, Yang D. 2014. Bifidobacterium infantis attenuates colitis by regulating T cell subset responses. World J Gastroenterol 20: 18316-18329. https://doi.org/10.3748/ wjg.v20.i48.18316.
    • (2014) World J Gastroenterol , vol.20 , pp. 18316-18329
    • Zuo, L.1    Yuan, K.2    Yu, L.3    Meng, Q.4    Chung, P.C.5    Yang, D.6
  • 381
    • 59449084229 scopus 로고    scopus 로고
    • The effect of Saccharomyces boulardii on human colon cells and inflammation in rats with trinitrobenzene sulfonic acid-induced colitis
    • Lee SK, Kim YW, Chi SG, Joo YS, Kim HJ. 2009. The effect of Saccharomyces boulardii on human colon cells and inflammation in rats with trinitrobenzene sulfonic acid-induced colitis. Dig Dis Sci 54: 255-263. https://doi.org/10.1007/s10620-008-0357-0.
    • (2009) Dig Dis Sci , vol.54 , pp. 255-263
    • Lee, S.K.1    Kim, Y.W.2    Chi, S.G.3    Joo, Y.S.4    Kim, H.J.5
  • 383
    • 33744827083 scopus 로고    scopus 로고
    • Saccharomyces boulardii prevents TNF-alpha-induced apoptosis in EHEC-infected T84 cells
    • Dalmasso G, Loubat A, Dahan S, Calle G, Rampal P, Czerucka D. 2006. Saccharomyces boulardii prevents TNF-alpha-induced apoptosis in EHEC-infected T84 cells. Res Microbiol 157: 456-465. https://doi.org/10.1016/j.resmic.2005.11.007.
    • (2006) Res Microbiol , vol.157 , pp. 456-465
    • Dalmasso, G.1    Loubat, A.2    Dahan, S.3    Calle, G.4    Rampal, P.5    Czerucka, D.6
  • 384
    • 26644452974 scopus 로고    scopus 로고
    • Inducible nitric oxide synthase involvement in the mechanism of action of Saccharomyces boulardii in castor oil-induced diarrhoea in rats
    • Girard P, Pansart Y, Gillardin JM. 2005. Inducible nitric oxide synthase involvement in the mechanism of action of Saccharomyces boulardii in castor oil-induced diarrhoea in rats. Nitric Oxide 13: 163-169. https:// doi.org/10.1016/j.niox.2005.06.001.
    • (2005) Nitric Oxide , vol.13 , pp. 163-169
    • Girard, P.1    Pansart, Y.2    Gillardin, J.M.3


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.