Your browser doesn't support javascript.
loading
A consortia of clinical E. coli strains with distinct in vitro adherent/invasive properties establish their own co-colonization niche and shape the intestinal microbiota in inflammation-susceptible mice.
Bleich, Rachel M; Li, Chuang; Sun, Shan; Ahn, Ju-Hyun; Dogan, Belgin; Barlogio, Cassandra J; Broberg, Christopher A; Franks, Adrienne R; Bulik-Sullivan, Emily; Carroll, Ian M; Simpson, Kenneth W; Fodor, Anthony A; Arthur, Janelle C.
Afiliación
  • Bleich RM; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
  • Li C; Department of Biology, Appalachian State University, Boone, NC, USA.
  • Sun S; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
  • Ahn JH; College of Computing and Informatics, University of North Carolina at Charlotte, Charlotte, NC, USA.
  • Dogan B; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
  • Barlogio CJ; Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY, USA.
  • Broberg CA; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
  • Franks AR; Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
  • Bulik-Sullivan E; Center for Gastrointestinal Biology & Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
  • Carroll IM; Center for Gastrointestinal Biology & Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
  • Simpson KW; Center for Gastrointestinal Biology & Disease, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
  • Fodor AA; Department of Clinical Sciences, Cornell University College of Veterinary Medicine, Ithaca, NY, USA.
  • Arthur JC; College of Computing and Informatics, University of North Carolina at Charlotte, Charlotte, NC, USA.
Microbiome ; 11(1): 277, 2023 Dec 20.
Article en En | MEDLINE | ID: mdl-38124090
ABSTRACT

BACKGROUND:

Inflammatory bowel disease (IBD) patients experience recurrent episodes of intestinal inflammation and often follow an unpredictable disease course. Mucosal colonization with adherent-invasive Escherichia coli (AIEC) are believed to perpetuate intestinal inflammation. However, it remains unclear if the 24-year-old AIEC in vitro definition fully predicts mucosal colonization in vivo. To fill this gap, we have developed a novel molecular barcoding approach to distinguish strain variants in the gut and have integrated this approach to explore mucosal colonization of distinct patient-derived E. coli isolates in gnotobiotic mouse models of colitis.

RESULTS:

Germ-free inflammation-susceptible interleukin-10-deficient (Il10-/-) and inflammation-resistant WT mice were colonized with a consortium of AIEC and non-AIEC strains, then given a murine fecal transplant to provide niche competition. E. coli strains isolated from human intestinal tissue were each marked with a unique molecular barcode that permits identification and quantification by barcode-targeted sequencing. 16S rRNA sequencing was used to evaluate the microbiome response to E. coli colonization. Our data reveal that specific AIEC and non-AIEC strains reproducibly colonize the intestinal mucosa of WT and Il10-/- mice. These E. coli expand in Il10-/- mice during inflammation and induce compositional dysbiosis to the microbiome in an inflammation-dependent manner. In turn, specific microbes co-evolve in inflamed mice, potentially diversifying E. coli colonization patterns. We observed no selectivity in E. coli colonization patterns in the fecal contents, indicating minimal selective pressure in this niche from host-microbe and interbacterial interactions. Because select AIEC and non-AIEC strains colonize the mucosa, this suggests the in vitro AIEC definition may not fully predict in vivo colonization potential. Further comparison of seven E. coli genomes pinpointed unique genomic features contained only in highly colonizing strains (two AIEC and two non-AIEC). Those colonization-associated features may convey metabolic advantages (e.g., iron acquisition and carbohydrate consumption) to promote efficient mucosal colonization.

CONCLUSIONS:

Our findings establish the in vivo mucosal colonizer, not necessarily AIEC, as a principal dysbiosis driver through crosstalk with host and associated microbes. Furthermore, we highlight the utility of high-throughput screens to decode the in vivo colonization dynamics of patient-derived bacteria in murine models. Video Abstract.
Asunto(s)
Palabras clave

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Infecciones por Escherichia coli / Microbioma Gastrointestinal Límite: Adult / Animals / Humans Idioma: En Revista: Microbiome Año: 2023 Tipo del documento: Article País de afiliación: Estados Unidos

Texto completo: 1 Banco de datos: MEDLINE Asunto principal: Infecciones por Escherichia coli / Microbioma Gastrointestinal Límite: Adult / Animals / Humans Idioma: En Revista: Microbiome Año: 2023 Tipo del documento: Article País de afiliación: Estados Unidos