Biology Reference
In-Depth Information
sufficient to abrogate NleC proteolytic activity ( Petty et al., 2010 ; Yen et al.,
2010 ; Baruch et al., 2011 ; Muhlen et al., 2011 ; Pearson et al., 2011 ). Further-
more, NleC cleaves p65 within its conserved DNA-binding domain, suggesting
that other NF-κB family members containing the same sequence could be sub-
strates of NleC ( Baruch et al., 2011 ). The acetyltransferase p300 was recently
identified as an additional target of NleC ( Shames et al., 2011 ). p300 functions
as a co-activator in the transcription of many genes by acetylating p65 (among
other transcription factors), thereby enhancing expression of genes downstream
of κB-containing enhancers ( Chen et al., 2001 ) such as IL-8. Overexpression
of p300 can indeed antagonize repression of IL-8 secretion induced by EPEC
infection ( Shames et al., 2011 ). While infection with EPEC nleC mutant does
not result in increased IL-8 production, presumably due to functional redun-
dancy with NleE and NleB, infection with an EPEC nleEC double mutant or
an EPEC nleBEC triple mutant results in significantly higher IL-8 production
by host cells compared with infection with EPEC nleE or EPEC nleBE alone
( Baruch et al., 2011 ; Pearson et al., 2011 ). Thus, NleC appears to complement
the activities of NleE and NleB by eliminating activated p65/p50 and/or its acet-
yltransferase p300 that would positively influence transcription and expression
of pro-inflammatory cytokines.
Together with NleC, the effector NleD was identified as zinc-dependent
metalloprotease as it also harbors the consensus sequence ( 142 HELLH 146 )
( Marches et al., 2005 ). NleD specifically targets MAP kinases JNK and p38
but not ERK, thereby blocking nuclear translocation of the AP-1 transcription
factor, which regulates multiple cell processes including inflammation, differ-
entiation, proliferation, and apoptosis ( Baruch et al., 2011 ). Infection with an
EPEC nleBECD mutant results in an increase of expression and secretion of
IL-8 compared to the nleBEC mutant, indicating an anti-inflammatory role for
NleD ( Baruch et al., 2011 ).
The NleH family of proteins is also conserved amongst EPEC and EHEC
strains and the C-termini of NleH1 and NleH2 proteins display significant amino
acid sequence similarity with the S. flexneri T3SS anti-inflammatory effector
OspG ( Kim et al., 2005 ). NleH1 and NleH2 have critical roles in inhibiting host
cell apoptosis; however, these effectors have also been implicated in the sup-
pression of NF-κB activation. NleH1 and NleH2 decrease IκKβ-induced NF-κB
activity by decreasing TNFα-induced degradation by preventing ubiquitination
of phospho-IκBα. Although the mechanism is not clear yet, stabilization of IκBα
is dependent on conserved lysine residues K159 in NleH1 and K169 in NleH2
from EPEC E2348/69 respectively ( Royan et al., 2010 ). It was recently shown
that the N-terminus of both NleH1 and NleH2 could bind the ribosomal protein
S3 (RPS3) ( Gao et al., 2009 ). RPS3 is a non-Rel subunit of NF-κB complexes
( Wan et al., 2007 ). RPS3 is considered a 'specifier' subunit of NF-κB, because it
facilitates high-affinity binding of DNA and determines the specificity of NF-κB
for selected target genes ( Wan et al., 2007 ). NleH1 of EHEC selectively blocks
the transcription of NF-κB target genes by attenuating nuclear translocation of
Search WWH ::




Custom Search