Bifidobacterial surface-exopolysaccharide facilitates commensal-host interaction through immune modulation and pathogen protection

Typeset version

 

TY  - JOUR
  - Fanning, S,Hall, LJ,Cronin, M,Zomer, A,MacSharry, J,Goulding, D,Motherway, MO,Shanahan, F,Nally, K,Dougan, G,van Sinderen, D
  - 2012
  - January
  - Proceedings of The National Academy of Sciences of The United States of America
  - Bifidobacterial surface-exopolysaccharide facilitates commensal-host interaction through immune modulation and pathogen protection
  - Validated
  - ()
  - NK CELLS COLONIZATION POLYSACCHARIDE BACTERIUM RESPONSES MILK GENE
  - 109
  - 2108
  - 2113
  - Bifidobacteria comprise a significant proportion of the human gut microbiota. Several bifidobacterial strains are currently used as therapeutic interventions, claiming various health benefits by acting as probiotics. However, the precise mechanisms by which they maintain habitation within their host and consequently provide these benefits are not fully understood. Here we show that Bifidobacterium breve UCC2003 produces a cell surface-associated exopolysaccharide (EPS), the biosynthesis of which is directed by either half of a bidirectional gene cluster, thus leading to production of one of two possible EPSs. Alternate transcription of the two opposing halves of this cluster appears to be the result of promoter reorientation. Surface EPS provided stress tolerance and promoted in vivo persistence, but not initial colonization. Marked differences were observed in host immune response: strains producing surface EPS (EPS+) failed to elicit a strong immune response compared with EPS-deficient variants. Specifically, EPS production was shown to be linked to the evasion of adaptive B-cell responses. Furthermore, presence of EPS+ B. breve reduced colonization levels of the gut pathogen Citrobacter rodentium. Our data thus assigns a pivotal and beneficial role for EPS in modulating various aspects of bifidobacterial-host interaction, including the ability of commensal bacteria to remain immunologically silent and in turn provide pathogen protection. This finding enforces the probiotic concept and provides mechanistic insights into health-promoting benefits for both animal and human hosts.
  - DOI 10.1073/pnas.1115621109
DA  - 2012/01
ER  - 
@article{V160747664,
   = {Fanning,  S and Hall,  LJ and Cronin,  M and Zomer,  A and MacSharry,  J and Goulding,  D and Motherway,  MO and Shanahan,  F and Nally,  K and Dougan,  G and van Sinderen,  D },
   = {2012},
   = {January},
   = {Proceedings of The National Academy of Sciences of The United States of America},
   = {Bifidobacterial surface-exopolysaccharide facilitates commensal-host interaction through immune modulation and pathogen protection},
   = {Validated},
   = {()},
   = {NK CELLS COLONIZATION POLYSACCHARIDE BACTERIUM RESPONSES MILK GENE},
   = {109},
  pages = {2108--2113},
   = {{Bifidobacteria comprise a significant proportion of the human gut microbiota. Several bifidobacterial strains are currently used as therapeutic interventions, claiming various health benefits by acting as probiotics. However, the precise mechanisms by which they maintain habitation within their host and consequently provide these benefits are not fully understood. Here we show that Bifidobacterium breve UCC2003 produces a cell surface-associated exopolysaccharide (EPS), the biosynthesis of which is directed by either half of a bidirectional gene cluster, thus leading to production of one of two possible EPSs. Alternate transcription of the two opposing halves of this cluster appears to be the result of promoter reorientation. Surface EPS provided stress tolerance and promoted in vivo persistence, but not initial colonization. Marked differences were observed in host immune response: strains producing surface EPS (EPS+) failed to elicit a strong immune response compared with EPS-deficient variants. Specifically, EPS production was shown to be linked to the evasion of adaptive B-cell responses. Furthermore, presence of EPS+ B. breve reduced colonization levels of the gut pathogen Citrobacter rodentium. Our data thus assigns a pivotal and beneficial role for EPS in modulating various aspects of bifidobacterial-host interaction, including the ability of commensal bacteria to remain immunologically silent and in turn provide pathogen protection. This finding enforces the probiotic concept and provides mechanistic insights into health-promoting benefits for both animal and human hosts.}},
   = {DOI 10.1073/pnas.1115621109},
  source = {IRIS}
}
AUTHORSFanning, S,Hall, LJ,Cronin, M,Zomer, A,MacSharry, J,Goulding, D,Motherway, MO,Shanahan, F,Nally, K,Dougan, G,van Sinderen, D
YEAR2012
MONTHJanuary
JOURNAL_CODEProceedings of The National Academy of Sciences of The United States of America
TITLEBifidobacterial surface-exopolysaccharide facilitates commensal-host interaction through immune modulation and pathogen protection
STATUSValidated
TIMES_CITED()
SEARCH_KEYWORDNK CELLS COLONIZATION POLYSACCHARIDE BACTERIUM RESPONSES MILK GENE
VOLUME109
ISSUE
START_PAGE2108
END_PAGE2113
ABSTRACTBifidobacteria comprise a significant proportion of the human gut microbiota. Several bifidobacterial strains are currently used as therapeutic interventions, claiming various health benefits by acting as probiotics. However, the precise mechanisms by which they maintain habitation within their host and consequently provide these benefits are not fully understood. Here we show that Bifidobacterium breve UCC2003 produces a cell surface-associated exopolysaccharide (EPS), the biosynthesis of which is directed by either half of a bidirectional gene cluster, thus leading to production of one of two possible EPSs. Alternate transcription of the two opposing halves of this cluster appears to be the result of promoter reorientation. Surface EPS provided stress tolerance and promoted in vivo persistence, but not initial colonization. Marked differences were observed in host immune response: strains producing surface EPS (EPS+) failed to elicit a strong immune response compared with EPS-deficient variants. Specifically, EPS production was shown to be linked to the evasion of adaptive B-cell responses. Furthermore, presence of EPS+ B. breve reduced colonization levels of the gut pathogen Citrobacter rodentium. Our data thus assigns a pivotal and beneficial role for EPS in modulating various aspects of bifidobacterial-host interaction, including the ability of commensal bacteria to remain immunologically silent and in turn provide pathogen protection. This finding enforces the probiotic concept and provides mechanistic insights into health-promoting benefits for both animal and human hosts.
PUBLISHER_LOCATION
ISBN_ISSN
EDITION
URL
DOI_LINKDOI 10.1073/pnas.1115621109
FUNDING_BODY
GRANT_DETAILS