569
Views
4
CrossRef citations to date
0
Altmetric
Editorial

Microflora in Crohn’s disease: the emergence of adherent and invasive Escherichia coli

&
Pages 133-137 | Published online: 10 Jan 2014

Crohn’s disease (CD) is a chronic, relapsing, transmural, inflammatory bowel disease (IBD) with symptoms including diarrhea, abdominal pain, weight loss and perianal fistula or fissures. Although the exact etiology is still unknown, it is generally considered a consequence of uncontrolled intestinal inflammation in response to a combination of elusive environmental, enteric microbial and immuno-regulatory factors in genetically susceptible individuals Citation[1]. A diverse spectrum of pathogenic bacteria, including Listeria, Streptococcus, Enterococcus, Enterobacteriaceae, Bacteroides, Clostridium, Yersinia and Mycobacterium avium subsp. paratuberculosis (MAP) Citation[1–8], have been implicated in the development of inflammation, but no single agent has emerged as a consistent cause. In the absence of a defined cause, therapy is symptomatic and supportive and achieves remission rather than cure.

The experimental literature now provides a clear indication that the resident enteric microflora, rather than typically recognized pathogenic bacteria, is involved in the pathogenesis of IBD in rodent models. For example, Enterobacteriaceae, particularly Escherichia coli, are the dominant microflora in the inflamed intestines of IL-2-/- mice, and the ceca of HLA-B27 β2-microglobulin transgenic rats Citation[9,10]. Resident E. coli and Entero-coccus faecalis have also been shown to induce colitis and cecal inflammation in monoinfected gnotobiotic IL-10-/- mice Citation[11]. Similarly, evidence implicating the resident enteric microflora in the pathogenesis of spontaneous IBD in people is provided by the clinical responses of CD to fecal stream diversion Citation[12] and anti-microbials Citation[2,13], the increased circulating and intraluminal humoral and T-cell responses to enteric commensal bacteria, and imbalances in the ratio of beneficial to harmful bacteria (termed ‘dysbiosis’) observed in CD Citation[13–18]. Complementing this is the discovery of mutations in genes regulating microflora-sensing ability (NOD2/CARD15 and Toll-like receptor [TLR]4) of patients with CD, which provides mechanisms to explain individual susceptibility to the resident microflora Citation[19,20] that, in the presence of the enteric microflora and appropriate triggers, may lead to upregulated mucosal cytokine production, delayed bacterial clearance and increased bacterial translocation, thereby promoting and perpetuating intestinal inflammation Citation[1,21,22].

Culture-independent analysis of CD mucosa reveals a selective increase in E. coli & depletion in Clostridiales

Advances in molecular microbiology have led to a new awareness of the diversity and complexity of the enteric flora. Culture--independent analyses of 16S rDNA libraries reveal that only 30% of the fecal flora appears cultivable, and there is significant variation in the flora in different gastrointestinal segments and luminal contents versus the mucosa of healthy individuals Citation[23–25]. Analysis of 16S rDNA libraries constructed from endoscopic biopsies of the ileum from patients with Crohn’s ileitis (a disease phenotype that occurs in approximately 70% of individuals with CD), demonstrates selective enrichment of ileal mucosa in E. coli and relative depletion in Clostridiales compared with both patients with CD restricted to the colon and to healthy individuals Citation[26]. PCR of ileal DNA yielded no evidence of MAP, Listeria or Shigella and confirmed the increase in E. coli Citation[26]. Similarly, 16S rDNA library-based evaluation of surgically resected CD ileum shows an increase in Proteobacteria (this phylum includes Enterobacteriaceae), decrease in Firmicutes (this phylum contains Clostridiales) and the absence of DNA from MAP in CD mucosa Citation[27]. The relative decrease in Clostridiales in CD ileal mucosa echoes findings in studies of feces and colonic mucosa from patients with CD, and suggests that this clostridial dys-biosis may be a feature of CD in general, rather than a marker of CD ileitis Citation[28–30].

The in-depth knowledge of microfloral composition provided by 16S sequence analysis can be used to guide in situ examination of the spatial distribution of the mucosal flora interacting most closely with the innate immune system. Fluorescence in situ hybridization (FISH) with labeled oligonucleo-tide probes targeted to specific 16S ribosomal RNA sequences (for example Citation[31]) allows spatial localization of different microbial subsets within histological samples. FISH with a probe restricted to E. coli/Shigella reveals that the ileal mucosa of CD patients harbors significantly more E. coli than normal ileum, and mucosally invasive E. coli are only detected in CD mucosa Citation[26]. In patients with Crohn’s ileitis, E. coli represent approximately 3.5% of the mucosal flora labeled with the panbacterial FISH probe EUB338 and approximately 40% of the Enterobacteriaceae recognized by the enterobacterial FISH probe 1531 Citation[26]. This contrasts with culture-based studies reporting that E. coli represent more than 50% of the cultivable mucosal flora in 40–58% of patients with ileal CD Citation[32,33]. However, this discrepancy is readily explained by the inability to culture the vast majority of enteric mucosal flora and supports the use of culture-independent analysis. Furthermore, the number of E. coli visualized in ileal biopsies by Baumgart et al. Citation[26], but not bacterial colonization in general, correlates with histological and endoscopic disease activity, which suggests E. coli could be specifically involved in the inflammatory process Citation[26]. These findings complement previous studies that have documented E. coli antigens and DNA in granulomas and peri-ulcerative lesions in CD Citation[6,34], and provide a rational explanation for the higher prevalence of antibodies directed against E. coli outer membrane porin C (OmpC) and flagellin in patients with CD involving the ileum versus colon-restricted CD Citation[16,35].

The association of adherent & invasive E. coli with CD

Culture-based analysis of E. coli isolated from the mucosa of patients with CD has revealed that many strains display pathogen-like behavior in cultured cells Citation[26,36–38]. The best-characterized CD-associated E. coli strain, LF82 (isolated by Darfeuille-Michaud from the ileum of a patient from Lille, France), adheres to, invades, persists and replicates in cultured intestinal epithelial cells Citation[36]. Invasion is associated with trigger-type endocytosis and utilizes host cell microfilaments and microtubules Citation[39]. LF82 is also able to replicate in J477-A1 macrophages, promote the elaboration of disease-relevant cytokines, such as TNF-a and IL-8, in cultured cells and elicit granuloma formation in vitro Citation[8,39–41]. LF82 is distinct from known pathogenic E. coli and is the prototypical strain of a putative new pathogroup of E. coli, adherent and invasive E. coli (AIEC).

Adherent and invasive E. coli have been isolated from the ileum of 36–38% of patients with Crohn’s ileitis compared with 6% from the ileum of healthy individuals Citation[26,32]. Conversely, in colonic specimens, AIEC strains were found in only 3.7% of CD patients, 0% of ulcerative colitis (UC) patients and 1.9% of controls Citation[8,32], suggesting that AIEC may be preferentially involved in ileal, rather than colonic inflammation. The predominance of AIEC in the inflamed ileum has recently been addressed by Barnich et al. who found that AIEC strains adhere to the brush border of primary ileal enterocytes isolated from CD patients but not controls without IBD Citation[42]. AIEC adhesion is dependent on both bacterial surface expression of type I pili and on expression of carcinoembryonic antigen-related cell adhesion molecule (CEACAM)6 on the apical surface of ileal epithelial cells, an interaction that can be inhibited by mannose Citation[42]. These in vitro findings are highly consistent with recent in situ observations that E. coli colonization correlates with disease activity Citation[26]. It is intriguing to think that CEACAM6 may be the proverbial door through which AIEC gain entry into the mucosa, whereupon they are eradicated in nonsusceptible individuals, but evade ineffectual host defenses in susceptible individuals, resulting in chronic intestinal inflammation.

While AIEC are generally associated with ileal colonization, invasive AIEC-like E. coli strains have also been isolated from the colonic mucosa of patients with CD, UC and colon cancer Citation[37,38,43]. However, CD-associated E. coli strains, especially the strains from inflamed CD tissue, have been shown to be much more invasive than UC-associated strains, and E. coli strains from inflamed CD tissue induced significantly higher TNF-a expression in macrophage cell lines than UC-associated E. coli strains Citation[38]. CEACAM6 is expressed in the colonic mucosa of patients with CD and UC Citation[8,42], and could be a receptor for colonic E. coli strains that display AIEC-like activity in cultured epithelial cells. However, this possibility has to be reconciled with the results of in situ analysis that show marked increases in mucosally adherent Bacteroides rather than E. coli in CD and UC colonic mucosa Citation[44].

AIEC are associated with intestinal inflammation in dogs

Granulomatous colitis of Boxer dogs (GCB) is a severe disease of unknown etiology that typically affects Boxer dogs Citation[45]. GCB has features in common with UC (macroscopic appearance, regional distribution and immunopathology), Whipple’s disease (PAS-positive macrophages) and CD (granulomatous inflammation, bacteria within macrophages and may respond to fluroquinolones) in humans but it is not identical to any of these disease syndromes Citation[46–49]. 16S rDNA libraries generated from GCB dogs are dominated by sequences for Enterobacteriaceae, predominantly E. coli and Shigella and FISH probes recognize invasive E. coli within the colonic mucosa (in the lamina propria and macrophages) of Boxer dogs with GCB but not unaffected controls, and non-GCB colitis. E. coli strains isolated from affected Boxer dogs display an AIEC pathotype in cultured cells, and resemble LF82 in phylo-geny and virulence genes Citation[45]. Subsequent studies of GCB-affected dogs have shown that antibiotic-induced clinical remission correlates with eradication of invasive E. coli. Thus, GCB is a curable enteropathy associated with mucosally invasive E. coli.

AIEC strains may share common pathoadaptive determinants of virulence that promote intestinal inflammation across species

Bacterial virulence is regulated typically by genes that promote the invasion or replication of organisms in target cells. In contrast to pathogenic E. coli, such as O157:H7, AIEC strains differ in serotype, overall genotype Citation[26,45] and ribotype Citation[50] and are not a single virulent clone. By use of PCR-based virulence screening and genome subtraction, we discovered that AIEC strains with unique multilocus sequence type sequences and distinct phylogenetic backgrounds (A, B1, B2 and D) harbor chromosomal and episomal elements that are homologous to those described in extraintestinal pathogenic E. coli (uropathogenic E. coli [UPEC] and avian pathogenic E. coli), and pathogenic Enterobacteriaceae, such as Salmonella and Yersinia Citation[26,45], and support the possibility that AIEC strains share common pathoadaptive determinants of virulence. For example, nucleotide sequences resembling ratA in Salmonella typhimurium , which is associated with colonization of the cecum and Peyer’s patches Citation[51], and another resembling a hemolysin coregulated protein (hcp) secreted by a type VI secretion system associated with virulence in Vibrio Citation[52], are potentially disease-relevant genes present in AIEC from CD and GCB. A plasmid resembling Yersinia pestis pMT1 is present in strain LF82, but not other AIEC strains, which has important implications as LF82 is regarded as the prototype AIEC strain, and it may not be representative of AIEC strains in general Citation[8,39].

While studies to date reinforce the high degree of diversity in E. coli as a species and its propensity for acquiring DNA from distantly related organisms, they provide only a glimpse of the potential genetic armory of AIEC. Genome sequencing will be required to resolve whether AIEC strains have acquired common virulence determinants in parallel, or have adopted the ‘mix-and-match’ approach described for UPEC Citation[54], and to aid in the identification of additional candidate genes associated with virulence. It is also important to consider that differences in gene regulation, rather than presence or absence of a locus or molecular selection, may influence virulence of AIEC. This possibility is reinforced by recent studies of type I pili, flagella and OmpC in AIEC LF82 Citation[8,53].

Conclusion

There is mounting evidence, derived from independent studies utilizing an array of culture-based and culture-independent methodologies that links E. coli to CD. The correlation between the number of E. coli visualized in ileal biopsies and disease activity, coupled with the ability of CD-associated E. coli to display pathogen-like adherent and invasive behavior in cultured cells, supports a role for E. coli in the inflammatory process. The remarkable similarity between E. coli strains isolated from Crohn’s ileitis and GCB raises the possibility that AIEC are a novel group of pathogenic E. coli that share common pathoadaptive determinants of virulence that promote intestinal inflammation. The isolation of AIEC from healthy and inflamed mucosa indicates that their mere presence alone is insufficient to cause disease. The increased numbers of mucosa-associated and invasive E. coli relative to other bacteria observed in patients with Crohn’s ileitis suggest that AIEC, with the appropriate stimulus, are most likely opportunistic pathogens that can exploit the mucosal environment of a Crohn’s-susceptible individual who has, for example, reduced defensin production and delayed bacterial clearance associated with functionally important polymorphisms in NOD2/CARD15 Citation[1,22]. Alternatively, AIEC may permissively proliferate subsequent to a depletion of normal flora, such as Faecalibacterium, or alterations in bacterial products like butyrate, which is important for colonic health and ileal regeneration Citation[55,56], or a combination of these possibilities. Resolving these issues will provide unique insights into the biology of CD and opportunities for therapeutic intervention.

Financial & competing interests disclosure

Belgin Dogan is supported by a grant from the Jill Roberts Center for Inflammatory Bowel Disease, Weill Medical College of Cornell University, NY 10021, USA. The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

  • Sartor RB. Mechanisms of disease: pathogenesis of Crohn’s disease and ulcerative colitis. Nat. Clin. Pract. Gastroenterol. Hepatol.3(7), 390–407 (2006).
  • Marteau P, Lepage P, Mangin I et al. Review article: gut flora and inflammatory bowel disease. Aliment. Pharmacol. Ther.20(Suppl. 4), 18–23 (2004).
  • Cartun RW, Van Kruiningen HJ, Pedersen CA, Berman MM. An immunocytochemical search for infectious agents in Crohn’s disease. Mod. Pathol.6(2), 212–219 (1993).
  • Bulois P, Desreumaux P, Neut C, Darfeuille-Michaud A, Cortot A, Colombel JF. Infectious agents and Crohn’s disease. Clin. Microbiol. Infect.5(10), 601–604 (1999).
  • Lamps LW, Madhusudhan KT, Havens JM et al. Pathogenic Yersinia DNA is detected in bowel and mesenteric lymph nodes from patients with Crohn’s disease. Am. J. Surg. Pathol.27(2), 220–227 (2003).
  • Liu Y, van Kruiningen HJ, West AB, Cartun RW, Cortot A, Colombel JF. Immunocytochemical evidence of Listeria, Escherichia coli, and Streptococcus antigens in Crohn’s disease. Gastroenterology108(5), 1396–1404 (1995).
  • Shanahan F, O’Mahony J. The mycobacteria story in Crohn’s disease. Am. J. Gastroenterol.100(7), 1537–1538 (2005).
  • Barnich N, Darfeuille-Michaud A. Adherent-invasive Escherichia coli and Crohn’s disease. Curr. Opin. Gastroenterol.23(1), 16–20 (2007).
  • Onderdonk AB, Richardson JA, Hammer RE, Taurog JD. Correlation of cecal microflora of HLA-B27 transgenic rats with inflammatory bowel disease. Infect. Immun.66(12), 6022–6023 (1998).
  • Schuppler M, Lotzsch K, Waidmann M, Autenrieth IB. An abundance of Escherichia coli is harbored by the mucosa-associated bacterial flora of interleukin-2-deficient mice. Infect. Immun.72(4), 1983–1990 (2004).
  • Kim SC, Tonkonogy SL, Albright CA et al. Variable phenotypes of enterocolitis in interleukin 10-deficient mice monoassociated with two different commensal bacteria. Gastroenterology128(4), 891–906 (2005).
  • Rutgeerts P, Goboes K, Peeters M et al. Effect of faecal stream diversion on recurrence of Crohn’s disease in the neoterminal ileum. Lancet338(8770), 771–774 (1991).
  • Mow WS, Landers CJ, Steinhart AH et al. High-level serum antibodies to bacterial antigens are associated with antibiotic-induced clinical remission in Crohn’s disease: a pilot study. Dig. Dis. Sci.49(7–8), 1280–1286 (2004).
  • Seksik P, Sokol H, Lepage P et al. Review article: the role of bacteria in onset and perpetuation of inflammatory bowel disease. Aliment. Pharmacol. Ther.24(Suppl. 3), 11–18 (2006).
  • Tamboli CP, Neut C, Desreumaux P, Colombel JF. Dysbiosis in inflammatory bowel disease. Gut53(1), 1–4 (2004).
  • Arnott ID, Landers CJ, Nimmo EJ et al. Sero-reactivity to microbial components in Crohn's disease is associated with disease severity and progression, but not NOD2/CARD15 genotype. Am. J. Gastroenterol.99(12), 2376–2384 (2004).
  • Furrie E, Macfarlane S, Cummings JH, Macfarlane GT. Systemic antibodies towards mucosal bacteria in ulcerative colitis and Crohn’s disease differentially activate the innate immune response. Gut53(1), 91–98 (2004).
  • Lodes MJ, Cong Y, Elson CO et al. Bacterial flagellin is a dominant antigen in Crohn disease. J. Clin. Invest.113(9), 1296–1306 (2004).
  • Inohara N, Ogura Y, Fontalba A et al. Host recognition of bacterial muramyl dipeptide mediated through NOD2. Implications for Crohn’s disease. J. Biol. Chem.278(8), 5509–5512 (2003).
  • Maeda S, Hsu LC, Liu H et al. Nod2 mutation in Crohn’s disease potentiates NF-κB activity and IL-1β processing. Science307(5710), 734–738 (2005).
  • Ahmad T, Armuzzi A, Bunce M et al. The molecular classification of the clinical manifestations of Crohn’s disease. Gastroenterology122(4), 854–866 (2002).
  • Wehkamp J, Stange EF. A new look at Crohn’s disease: breakdown of the mucosal antibacterial defense. Ann. NY Acad. Sci.1072, 321–331 (2006).
  • Eckburg PB, Bik EM, Bernstein CN et al. Diversity of the human intestinal microbial flora. Science308(5728), 1635–1638 (2005).
  • Hayashi H, Sakamoto M, Benno Y. Phylogenetic analysis of the human gut microbiota using 16S rDNA clone libraries and strictly anaerobic culture-based methods. Microbiol. Immunol.46(8), 535–548 (2002).
  • Lepage P, Seksik P, Sutren M et al. Biodiversity of the mucosa-associated microbiota is stable along the distal digestive tract in healthy individuals and patients with IBD. Inflamm. Bowel. Dis.11(5), 473–480 (2005).
  • Baumgart M, Dogan B, Rishniw M et al. Culture independent analysis of ileal mucosa reveals a selective increase in invasive Escherichia coli of novel phylogeny relative to depletion of Clostridiales in Crohn’s disease involving the ileum. ISME J.1(5), 403–418 (2007).
  • Frank DN, St Amand AL, Feldman RA, Boedeker EC, Harpaz N, Pace NR. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl Acad. Sci. USA104(34), 13780–13785 (2007).
  • Gophna U, Sommerfeld K, Gophna S, Doolittle WF, Veldhuyzen van Zanten SJ. Differences between tissue-associated intestinal microfloras of patients with Crohn’s disease and ulcerative colitis. J. Clin. Microbiol.44(11), 4136–4141 (2006).
  • Manichanh C, Rigottier-Gois L, Bonnaud E et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut55(2), 205–211 (2006).
  • Martinez-Medina M, Aldeguer X, Gonzalez-Huix F, Acero D, Garcia-Gil LJ. Abnormal microbiota composition in the ileocolonic mucosa of Crohn’s disease patients as revealed by polymerase chain reaction-denaturing gradient gel electrophoresis. Inflamm. Bowel Dis.12(12), 1136–1145 (2006).
  • Kleessen B, Kroesen AJ, Buhr HJ, Blaut M. Mucosal and invading bacteria in patients with inflammatory bowel disease compared with controls. Scand. J. Gastroenterol.37(9), 1034–1041 (2002).
  • Darfeuille-Michaud A, Boudeau J, Bulois P et al. High prevalence of adherent-invasive Escherichia coli associated with ileal mucosa in Crohn’s disease. Gastroenterology127(2), 412–421 (2004).
  • Darfeuille-Michaud A, Neut C, Barnich N et al. Presence of adherent Escherichia coli strains in ileal mucosa of patients with Crohn’s disease. Gastroenterology,115(6), 1405–1413 (1998).
  • Ryan P, Kelly RG, Lee G et al. Bacterial DNA within granulomas of patients with Crohn’s disease – detection by laser capture microdissection and PCR. Am. J. Gastroenterol.99(8), 1539–1543 (2004).
  • Targan SR, Landers CJ, Yang H et al. Antibodies to CBir1 flagellin define a unique response that is associated independently with complicated Crohn’s disease. Gastroenterology128(7), 2020–2028 (2005).
  • Darfeuille-Michaud A. Adherent-invasive Escherichia coli: a putative new E. coli pathotype associated with Crohn’s disease. Int. J. Med. Microbiol.292(3–4), 185–193 (2002).
  • Martin HM, Campbell BJ, Hart CA et al. Enhanced Escherichia coli adherence and invasion in Crohn’s disease and colon cancer. Gastroenterology127(1), 80–93 (2004).
  • Sasaki M, Sitaraman SV, Babbin BA et al. Invasive Escherichia coli are a feature of Crohn’s disease. Lab. Invest.87(10), 1042–1054 (2007).
  • Boudeau J, Glasser AL, Masseret E, Joly B, Darfeuille-Michaud A. Invasive ability of an Escherichia coli strain isolated from the ileal mucosa of a patient with Crohn’s disease. Infect. Immun.67(9), 4499–4509 (1999).
  • Glasser AL, Boudeau J, Barnich N, Perruchot MH, Colombel JF, Darfeuille-Michaud A. Adherent invasive Escherichia coli strains from patients with Crohn’s disease survive and replicate within macrophages without inducing host cell death. Infect. Immun.69(9), 5529–5537 (2001).
  • Meconi S, Vercellone A, Levillain F et al. Adherent-invasive Escherichia coli isolated from Crohn’s disease patients induce granulomas in vitro. Cell. Microbiol.9(5), 1252–1261 (2007).
  • Barnich N, Carvalho FA, Glasser AL et al. CEACAM6 acts as a receptor for adherent–invasive E. coli, supporting ileal mucosa colonization in Crohn disease. J. Clin. Invest.117(6), 1566–1574 (2007).
  • Kotlowski R, Bernstein CN, Sepehri S, Krause DO. High prevalence of Escherichia coli belonging to the B2+D phylogenetic group in inflammatory bowel disease. Gut56(5), 669–675 (2007).
  • Swidsinski A, Weber J, Loening-Baucke V, Hale LP, Lochs H. Spatial organization and composition of the mucosal flora in patients with inflammatory bowel disease. J. Clin. Microbiol.43(7), 3380–3389 (2005).
  • Simpson KW, Dogan B, Rishniw M et al. Adherent and invasive Escherichia coli is associated with granulomatous colitis in boxer dogs. Infect. Immun.74(8), 4778–4792 (2006).
  • Dutly F, Altwegg M. Whipple’s disease and ‘Tropheryma whippelii’. Clin. Microbiol. Rev.14(3), 561–583 (2001).
  • German AJ, Hall EJ, Kelly DF, Watson AD, Day MJ. An immunohistochemical study of histiocytic ulcerative colitis in boxer dogs. J. Comp. Pathol.122(2–3), 163–175 (2000).
  • Van Kruiningen HJ. Granulomatous colitis of boxer dogs: comparative aspects. Gastroenterology53(1), 114–122 (1967).
  • Van Kruiningen HJ, Montali RJ, Strandberg JD, Kirk RW. A granulomatous colitis of dogs with histologic resemblance to Whipple’s disease. Pathol. Vet.2(6), 521–544 (1965).
  • Masseret E, Boudeau J, Colombel JF et al. Genetically related Escherichia coli strains associated with Crohn’s disease. Gut48(3), 320–325 (2001).
  • Kingsley RA, Humphries AD, Weening EH et al. Molecular and phenotypic analysis of the CS54 island of Salmonella enterica serotype Typhimurium: identification of intestinal colonization and persistence determinants. Infect. Immun.71(2), 629–640 (2003).
  • Pukatzki S, Ma AT, Sturtevant D et al. Identification of a conserved bacterial protein secretion system in Vibrio cholerae using the Dictyostelium host model system. Proc. Natl Acad. Sci. USA103(5), 1528–1533 (2006).
  • Boudeau J, Barnich N, Darfeuille-Michaud A. Type 1 pili-mediated adherence of Escherichia coli strain LF82 isolated from Crohn’s disease is involved in bacterial invasion of intestinal epithelial cells. Mol. Microbiol.39(5), 1272–1284 (2001).
  • Brzuszkiewicz E, Bruggemann H, Liesegang H et al. How to become a uropathogen: comparative genomic analysis of extraintestinal pathogenic Escherichia coli strains. Proc. Natl Acad. Sci. USA103(34), 12879–12884 (2006).
  • Bartholome AL, Albin DM, Baker DH, Holst JJ, Tappenden KA. Supplementation of total parenteral nutrition with butyrate acutely increases structural aspects of intestinal adaptation after an 80% jejunoileal resection in neonatal piglets. J. Parenter. Enteral. Nutr.28(4), 210–222 (2004).
  • Pryde SE, Duncan SH, Hold GL, Stewart CS, Flint HJ. The microbiology of butyrate formation in the human colon. FEMS Microbiol. Lett.217(2), 133–139 (2002).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.