Skip to main content
Erschienen in: Molecular and Cellular Pediatrics 1/2016

Open Access 01.12.2016 | Mini review

Diverse roles of endoplasmic reticulum stress sensors in bacterial infection

verfasst von: Helena Pillich, Maria Loose, Klaus-Peter Zimmer, Trinad Chakraborty

Erschienen in: Molecular and Cellular Pediatrics | Ausgabe 1/2016

Abstract

Bacterial infection often leads to cellular damage, primarily marked by loss of cellular integrity and cell death. However, in recent years, it is being increasingly recognized that, in individual cells, there are graded responses collectively termed cell-autonomous defense mechanisms that induce cellular processes designed to limit cell damage, enable repair, and eliminate bacteria. Many of these responses are triggered not by detection of a particular bacterial effector or ligand but rather by their effects on key cellular processes and changes in homeostasis induced by microbial effectors when recognized. These in turn lead to a decrease in essential cellular functions such as protein translation or mitochondrial respiration and the induction of innate immune responses that may be specific to the cellular deficit induced. These processes are often associated with specific cell compartments, e.g., the endoplasmic reticulum (ER). Under non-infection conditions, these systems are generally involved in sensing cellular stress and in inducing and orchestrating the subsequent cellular response. Thus, perturbations of ER homeostasis result in accumulation of unfolded proteins which are detected by ER stress sensors in order to restore the normal condition. The ER is also important during bacterial infection, and bacterial effectors that activate the ER stress sensors have been discovered. Increasing evidence now indicate that bacteria have evolved strategies to differentially activate different arms of ER stress sensors resulting in specific host cell response. In this review, we will describe the mechanisms used by bacteria to activate the ER stress sensors and discuss their role during infection.
Hinweise

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

HP and TC conceived and wrote the manuscript. ML and KPZ critically revised the manuscript. All authors read and approved the final manuscript.
Abkürzungen
ATF3/4/6
activating transcription factor 3/4/6
BiP
immunoglobulin heavy chain-binding protein
BMM
bone marrow-derived macrophages
CHOP
DNA-damage-inducible transcript 3
CT
cholera toxin
DAPK1
death-associated protein kinase 1
eIF2α
eukaryotic translation initiation factor 2α
ER
endoplasmic reticulum
ERAD
ER-associated degradation
ESAT-6
early secretory antigenic target 6
Grp94
heat shock protein 90 kDa beta member 1
IκB
inhibitor of kappa B
IL-6
interleukin 6
IRE1
inositol-requiring enzyme 1
LLO
listeriolysin O
LPS
lipopolysaccharide
MAPK
mitogen-activated protein kinase
mRNA
messenger RNA
PERK
protein kinase RNA (PKR)-like ER kinase
PFT
pore-forming toxin
RIDD
regulated IRE1-dependent decay
RIG-I
retinoic acid inducible gene I
ROS
reactive oxygen species
STEC
Shiga toxigenic Escherichia coli
Stx
Shiga toxin
SubAB
subtilase cytotoxin
TLR2/4/9
toll-like receptor 2/4/9
UPR
unfolded protein response
XBP1
X-box-binding protein 1

Introduction

Newly synthesized transmembrane and secretory proteins are folded and post-translationally modified within the endoplasmic reticulum (ER). Certain conditions such as hypoxia, Ca2+ perturbation, and reactive oxygen species (ROS) cause continued accumulation of unfolded proteins within the ER, a condition termed ER stress. To counteract ER stress and maintain ER function, cells activate the unfolded protein response (UPR), a signaling cascade composed of three axes which enables the reduction of protein amount entering the ER by translational inhibition, enhancement of protein folding by transcriptional upregulation of ER chaperones, and degradation of misfolded proteins through ER-associated degradation (ERAD). If ER stress is prolonged and severe, UPR induces apoptosis.
Several studies have now implicated UPR in bacterial infections. Surprisingly, it becomes clear that some bacteria have evolved strategies to activate all three UPR-signaling pathways. Recent studies have revealed that the execution of a particular UPR-signaling pathway does not occur randomly and new functions of the ER stress sensors have been described. In this review, we summarize the mechanisms used by bacteria to induce UPR and discuss the importance of certain UPR-signaling pathway activation.

UPR-signaling pathways

In higher eukaryotes, UPR signaling is mediated by the ER-transmembrane proteins detecting ER stress: inositol-requiring enzyme 1 (IRE1), protein kinase RNA (PKR)-like ER kinase (PERK), and activating transcription factor 6 (ATF6). The ER-resident chaperone immunoglobulin heavy chain-binding protein (BiP) binds to the luminal domain of the ER stress sensors and keeps them in an inactive state. Accumulation of unfolded proteins leads to release of BiP from the ER stress sensors and subsequent binding to the unfolded proteins [1].
Unbound IRE1 oligomerizes, autophosphorylates, and activates its endoribonuclease which mediates unconventional splicing of an intron from X-box-binding protein 1 (xbp1) messenger RNA (mRNA) producing a potent transcription factor (spliced-XBP1) [1] (Fig. 1).
Disruption of the PERK-BiP complex results in dimerization, autophosphorylation and activation of the kinase domain of PERK. Active PERK phosphorylates serine 51 of eukaryotic translation initiation factor 2α (eIF2α) consequently leading to translational inhibition. Thus, the protein load that enters the ER decreases. However, under this condition, the translation of some mRNAs, such as that of the transcription factor atf4, is increased [1] (Fig. 1).
Release of BiP from ATF6 permits the translocation of ATF6 to the Golgi apparatus where it is cleaved by two proteases. The cytosolic fragment migrates to the nucleus and regulates the transcription of UPR target genes [1] (Fig. 1).

Bacterial mechanisms to induce UPR

Bacteria have evolved different virulence factors that trigger the activation of UPR. One well-known example is lipopolysaccharide (LPS), an endotoxin located in the outer membrane of Gram-negative bacteria. LPS is detected by toll-like receptor 4 (TLR4) that is delivered by the ER chaperone heat shock protein 90 kDa beta member 1 (Grp94) from the ER to the plasma membrane. Expression of TLR4 and Grp94 is increased after LPS treatment. However, folding and plasma membrane translocation of TLR4 is not sufficient because the expression level of Grp94 is lower than that of TLR4 [2] (Fig. 2 (A)).
Another major group of toxins that induce UPR are pore-forming toxins (PFT) including aerolysin, Cry5B, listeriolysin O (LLO), and the small protein early secretory antigenic target 6 (ESAT-6) [35]. PFT-mediated pore formation on the plasma membrane which leads to perturbance of Ca2+ levels, activation of mitogen-activated protein kinase (MAPK), and induction of ROS production triggers UPR activation [3, 4] (Fig. 2 (B)). These circumstances are also important for UPR-activation mechanisms utilized by other bacteria, for instance Pseudomonas aeruginosa and H2O2-positive Streptococcus pneumoniae [6, 7] (Fig. 2 (C)).
Other bacteria have evolved strategies to activate UPR by release of factors that are able to enter the lumen of the ER. This includes tunicamycin, an inhibitor of protein N-linked glycosylation, which is very often used as a positive control for UPR induction [5, 7, 8] (Fig. 2 (D)). Also, AB5 toxins (Shiga toxin (Stx), subtilase cytotoxin (SubAB), cholera toxin (CT)) that comprise of a cell surface receptor-binding pentameric B-subunit are retrogradely trafficked via the Golgi apparatus to the ER where they trigger ER stress [911] (Fig. 2 (E)). The subunits of Stx1 are sensed as misfolded proteins within the ER and lead to UPR initiation [9, 12]. SubAB-triggered UPR activation is induced by the cleavage of BiP by this proteolytic toxin, resulting in subsequent loss of the inactivation process of the ER stress sensors [10]. CT, on the other hand, interacts directly with IRE1 leading to its activation [11].

The role of individual ER stress marker activation

A closer look on the activation of the ER stress sensors illustrates that some bacterial factors induce all three axes of ER stress while others are more specific (Table 1). Examples of bacterial factors which trigger the activation of all three pathways include LPS, LLO, and SubAB [2, 5, 10], whereas Chlamydia pneumoniae, S. pneumoniae, Legionella pneumophila, and CT induce only one of the ER stress sensors [6, 11, 13, 14]. Considering that S. pneumoniae executes ER stress through a general response, that is through an increase in ROS, the activation of only the PERK-pathway observed might be due to suppression of IRE1- and ATF6-signaling pathway. Indeed, L. pneumophila, an intracellular pathogen which replicates within ER-like vacuoles, cleaves ATF6 but does not activate the PERK-signaling pathway, and it even blocks thapsigargin-induced xbp1 splicing by expression of the two effectors Lgt1 and Lgt2 that glycosylate serine 53 in the elongation factor eEF1A causing protein synthesis inhibition [14]. Furthermore, ER stress sensor activation is cell-type specific as demonstrated in studies with Stx1 [9, 12]. Because a particular cell type is involved in a certain response within the host, this suggests that the activation of an individual ER stress marker may be cell-type specific and related to cell function.
Table 1
Bacteria and bacterial products that activate the ER stress sensors
Bacterium
Virulence factor
Cell type
UPR-specific host response
Mechanism
Reference
IRE1
PERK
ATF6
Aeromonas hydrophila
Aerolysin
HeLa
XBP1-s
n.d.
n.d.
 
[3]
Bacillus thuringiensis
Cry5B
C. elegans
xbp1-s
n.d.
n.d.
p38
[3]
Brucella abortus
 
BMM
xbp1-s
n.d.
n.d.
 
[16]
Chlamydia pneumoniae
 
HEp-2
eIF2α-p
Persistent infection
[13]
Francisella tularensis
 
BMM
xbp1-s
n.d.
n.d.
TLR2
[8]
Gram-negative bacteria
LPS
Monocytic THP-1
XBP1-s
PERK-p, eIF2α-p
ATF6 cleavage
TLR4
[2]
Helicobacter pylori
HP0175
AGS
n.d.
PERK-p, CHOP, ATF4, eIF2α-p
n.d.
 
[23]
Legionella pneumophila
 
BMM, HEK-293 FCγ, RAW264.7
Block of xbp1-s
Block of CHOP translation
ATF6 cleavage
 
[14]
Listeria monocytogenes
LLO
P388D1, HeLa
xbp1-s
eIF2α-p
ATF6 cleavage
 
[5]
Mycobacterium tuberculosis
ESAT-6
A549
xbp1-s
eIF2α-p, ATF4, chop
n.d.
ER Ca2+ release, ROS
[4]
Pseudomonas aeruginosa
 
C. elegans
xbp1-s
n.d.
n.d.
PMK-1 (p38 orthologue)
[7]
Shigella dysenteriae, STEC
Stx1
Monocytic THP-1
IRE1, xbp1-s
PERK-p, chop
ATF6 cleavage
Unfolded Stx (not for IRE1)
[9]
Macrophage-like THP-1
IRE1-p, xbp1-s
PERK-p, CHOP
Unfolded Stx
[12]
Staphylococcus aureus
 
BMM, RAW264.7
xbp1-s
n.d.
n.d.
TLR2/4/9
[17]
STEC
SubAB
Vero, MEF
xbp1-s
chop, eIF2α-p, atf4
ATF6 cleavage
BiP cleavage
[10]
Streptococcus pneumoniae
H2O2
H441
decrease of xbp1-s
PERK-p, atf4, eIF2α-p, atf3, chop
ROS
[6]
Streptomyces sp.
Tunicamycin
P388D1, HeLa, C. elegans, J774
xbp1-s, IRE1-p
PERK-p, chop
ATF6 cleavage
Inhibition of N-linked glycosylation
[5, 7, 8]
Vibrio cholerae
CT
T84
IRE1-p,
xbp1-s
Interaction with IRE1
[11]
Yersinia pseudotuberculosis
 
MEF, RAW264.7
n.d.
eIF2α-p, atf3
n.d.
 
[21]

The role of IRE1 activation

Recent studies now show that, in addition to the transcriptional regulation of UPR targets, the IRE1-XBP1 arm also affects other host cell responses. Firstly, both IRE1 and XBP1 are implicated in cell-autonomous defense mechanisms against PFTs as ire-1- and xbp-1-negative Caenorhabditis elegans are more sensitive to Cry5B. In contrast, pek-1 (a PERK homolog) did not contribute to resistance against Cry5B [3]. In addition, xbp-1 but not atf-6 or pek-1 is required for the development and survival of C. elegans against P. aeruginosa [7]. Moreover, xbp1 −/− mice exhibit abnormalities within the intestine that include disseminated cellular ER stress, apoptosis of Paneth cells, and reduced expression of bactericidal molecules as well as mucin 2, thus permitting the dissemination of orally administered Listeria monocytogenes [15]. In addition, loss of xbp1 increases the Francisella tularensis burden in the liver, spleen, and lung following aerosolic infection of mice. IRE1-triggered splicing of xbp1 augments also the production of pro-inflammatory cytokines (interleukin 6 (IL-6)) in a TLR-dependent manner, as exemplified during F. tularensis infection [8]. Moreover, CT-activated IRE1 degrades mRNA associated with the ER membranes, a mechanism termed regulated IRE1-dependent decay (RIDD). The resulting mRNA fragments are sensed by retinoic acid inducible gene I (RIG-I) leading to production of cytokines like IL-6. IRE1α- and RIG-I-dependent expression of IL-6 is also observed after treatment of mouse embryonic fibroblasts with enzymatically inactive Stx suggesting a general mechanism of immune activation by AB5 toxins [11]. Recently, it was demonstrated that IRE1-extended ROS production stimulated caspase-2-Bid-mediated mitochondrial damage which activated the inflammasome [16]. Finally, Staphylococcus aureus induced IRE1-activation results in sustained generation of ROS which enables bacterial killing [17].
Secondly, in Drosophila S2 as well as in mammalian cells ire1 but not perk and atf6 is required for the replication of Brucella melitensis, a bacterium which grows in an ER-like compartment [18].

The role of PERK activation

Translational attenuation allows a rapid spatial response to infection and contributes to host defenses through several mechanisms. Recently, it was described that PERK is involved in induction of the innate immune response which is triggered by the activation of the pro-inflammatory transcription factor NF-κB upon release of inhibitor of kappa B (IκB) [1]. Due to the short half-life of IκB, translational attenuation results in loss of NF-κB binding and subsequent expression of pro-inflammatory cytokines, as demonstrated during L. pneumophila infection [1, 19]. Moreover, PERK regulates the expression of the pro-apoptotic factor DNA-damage-inducible transcript 3 (ddit3, also known as chop) which contributes to host defense as shown during Mycobacterium tuberculosis infection where RNAi-mediated chop depletion results in an increased number of intracellular bacteria [20]. In addition, infection of cells defective in eIF2α phosphorylation results in a higher intracellular L. monocytogenes, Yersinia pseudotuberculosis, and Chlamydia trachomatis number [21]. On the other hand, phosphorylation of eIF2α is accompanied by increased expression of BiP which rescues host cells from stress contributing to host survival as observed during interferon gamma-induced persistent C. pneumoniae infection [13]. These studies suggest that translational attenuation serves as a mechanism of the host to detect bacteria and to activate an appropriate anti-bacterial defense.
A recent study showed that SubAB induces stress granules in various cells in a PERK-dependent manner [22]. Furthermore, Halder et al. reported that the Helicobacter pylori produced HP0175, a peptidyl-prolyl cis-trans isomerase, activates the PERK arm consequently leading to production of ATF4 and CHOP which both induce the expression of autophagy-related genes [23]. In contrast, SubAB-triggered PERK activation suppresses autophagy [24]. Thus, PERK-dependent autophagy induction seems to be insult dependent.

The role of ATF6 activation

The role of bacterial-conditioned ATF6 activation is much less studied than that of IRE1 or PERK. Nonetheless, a recent study showed that atf6 −/− mice are highly susceptible to Bacillus anthracis infection and exhibit increased bacterial load in the spleens and livers. These effects are associated with reduced autophagic bacterial degradation as ATF6 was shown to be required for the expression of death-associated protein kinase 1 (dapk1) which promotes autophagy [25].

Summary and future perspectives

It is now clear that host cells detect and respond to impairments of key cellular processes induced by microbial effectors rather than directly detecting the microbes themselves. These responses are generally accompanied by selective shutdown of essential cellular process such as protein biosynthesis. Indeed, it is remarkable that a large number of bacterial effector proteins, including toxins such as Stx, SubAB, or PFTs, target the eukaryotic translation machinery. UPR was described as a response of a cell to counteract the accumulation of the unfolded proteins within the ER. Surprisingly, during a bacterial infection, different ER stress sensors are activated indicating that bacteria have evolved strategies to induce a particular UPR pathway. Indeed, recent studies have revealed that the ER stress sensors modulate also other cell-autonomous processes such as autophagy, RIDD, the inflammasome, or stress granule formation. Chemical and pharmacological modulation of ER stress was shown to be essential for bacterial elimination. Interestingly, these compounds were not only used in cellular in vitro conditions but also in in vivo mouse experiments. Thus, these studies indicate that ER stress functions as a target in many pathologies, including acute infections and chronic neurodegenerative diseases. Therefore, targeting ER stress might be effective as an adjunct therapy in threatening bacterial infections.

Acknowledgements

We thank Dr. Petra Burghaus and Dr. Mobarak Abu Mraheil for the critical reading of the manuscript. This work was supported by the Bundesministerium für Bildung und Forschung (ERA-NET PathoGenoMics LISTRESS and Infect-ERA PROANTILIS to T. C.) and by the Deutsche Forschungsgemeinschaft (SFB-TR84, project A04 to T. C.).
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

HP and TC conceived and wrote the manuscript. ML and KPZ critically revised the manuscript. All authors read and approved the final manuscript.
Literatur
1.
Zurück zum Zitat Chaudhari N, Talwar P, Parimisetty A, Lefebvre d‘Hellencourt C, Ravanan P (2014) A molecular web: endoplasmic reticulum stress, inflammation, and oxidative stress. Front Cell Neurosci 8:213PubMedCentralCrossRefPubMed Chaudhari N, Talwar P, Parimisetty A, Lefebvre d‘Hellencourt C, Ravanan P (2014) A molecular web: endoplasmic reticulum stress, inflammation, and oxidative stress. Front Cell Neurosci 8:213PubMedCentralCrossRefPubMed
2.
Zurück zum Zitat Coope A, Milanski M, Arruda AP, Ignacio-Souza LM, Saad MJ, Anhê GF, Velloso LA (2012) Chaperone insufficiency links TLR4 protein signaling to endoplasmic reticulum stress. J Biol Chem 287:15580–15589PubMedCentralCrossRefPubMed Coope A, Milanski M, Arruda AP, Ignacio-Souza LM, Saad MJ, Anhê GF, Velloso LA (2012) Chaperone insufficiency links TLR4 protein signaling to endoplasmic reticulum stress. J Biol Chem 287:15580–15589PubMedCentralCrossRefPubMed
3.
Zurück zum Zitat Bischof LJ, Kao CY, Los FC, Gonzalez MR, Shen Z, Briggs SP, van der Goot FG, Aroian RV (2008) Activation of the unfolded protein response is required for defenses against bacterial pore-forming toxin in vivo. PLoS Pathog 4:e1000176PubMedCentralCrossRefPubMed Bischof LJ, Kao CY, Los FC, Gonzalez MR, Shen Z, Briggs SP, van der Goot FG, Aroian RV (2008) Activation of the unfolded protein response is required for defenses against bacterial pore-forming toxin in vivo. PLoS Pathog 4:e1000176PubMedCentralCrossRefPubMed
4.
Zurück zum Zitat Choi HH, Shin DM, Kang G, Kim KH, Park JB, Hur GM, Lee HM, Lim YJ, Park JK, Jo EK, Song CH (2010) Endoplasmic reticulum stress response is involved in Mycobacterium tuberculosis protein ESAT-6-mediated apoptosis. FEBS Lett 584:2445–2454CrossRef Choi HH, Shin DM, Kang G, Kim KH, Park JB, Hur GM, Lee HM, Lim YJ, Park JK, Jo EK, Song CH (2010) Endoplasmic reticulum stress response is involved in Mycobacterium tuberculosis protein ESAT-6-mediated apoptosis. FEBS Lett 584:2445–2454CrossRef
5.
Zurück zum Zitat Pillich H, Loose M, Zimmer KP, Chakraborty T (2012) Activation of the unfolded protein response by Listeria monocytogenes. Cell Microbiol 14:949–964CrossRefPubMed Pillich H, Loose M, Zimmer KP, Chakraborty T (2012) Activation of the unfolded protein response by Listeria monocytogenes. Cell Microbiol 14:949–964CrossRefPubMed
6.
Zurück zum Zitat Loose M, Hudel M, Zimmer KP, Garcia E, Hammerschmidt S, Lucas R, Chakraborty T, Pillich H (2015) Pneumococcal hydrogen peroxide-induced stress signaling regulates inflammatory genes. J Infect Dis 211:306–316PubMedCentralCrossRefPubMed Loose M, Hudel M, Zimmer KP, Garcia E, Hammerschmidt S, Lucas R, Chakraborty T, Pillich H (2015) Pneumococcal hydrogen peroxide-induced stress signaling regulates inflammatory genes. J Infect Dis 211:306–316PubMedCentralCrossRefPubMed
7.
Zurück zum Zitat Richardson CE, Kooistra T, Kim DH (2010) An essential role for XBP-1 in host protection against immune activation in C. elegans. Nature 463:1092–1095PubMedCentralCrossRefPubMed Richardson CE, Kooistra T, Kim DH (2010) An essential role for XBP-1 in host protection against immune activation in C. elegans. Nature 463:1092–1095PubMedCentralCrossRefPubMed
8.
Zurück zum Zitat Martinon F, Chen X, Lee AH, Glimcher LH (2010) TLR activation of the transcription factor XBP1 regulates innate immune responses in macrophages. Nat Immunol 11:411–418PubMedCentralCrossRefPubMed Martinon F, Chen X, Lee AH, Glimcher LH (2010) TLR activation of the transcription factor XBP1 regulates innate immune responses in macrophages. Nat Immunol 11:411–418PubMedCentralCrossRefPubMed
9.
Zurück zum Zitat Lee SY, Lee MS, Cherla RP, Tesh VL (2008) Shiga toxin 1 induces apoptosis through the endoplasmic reticulum stress response in human monocytic cells. Cell Microbiol 10:770–780CrossRefPubMed Lee SY, Lee MS, Cherla RP, Tesh VL (2008) Shiga toxin 1 induces apoptosis through the endoplasmic reticulum stress response in human monocytic cells. Cell Microbiol 10:770–780CrossRefPubMed
10.
Zurück zum Zitat Wolfson JJ, May KL, Thorpe CM, Jandhyala DM, Paton JC, Paton AW (2008) Subtilase cytotoxin activates PERK, IRE1 and ATF6 endoplasmic reticulum stress-signalling pathways. Cell Microbiol 10:1775–1786PubMedCentralCrossRefPubMed Wolfson JJ, May KL, Thorpe CM, Jandhyala DM, Paton JC, Paton AW (2008) Subtilase cytotoxin activates PERK, IRE1 and ATF6 endoplasmic reticulum stress-signalling pathways. Cell Microbiol 10:1775–1786PubMedCentralCrossRefPubMed
11.
Zurück zum Zitat Cho JA, Lee AH, Platzer B, Cross BC, Gardner BM, De Luca H, Luong P, Harding HP, Glimcher LH, Walter P, Fiebiger E, Ron D, Kagan JC, Lencer WI (2013) The unfolded protein response element IRE1α senses bacterial proteins invading the ER to activate RIG-I and innate immune signaling. Cell Host Microbe 13:558–569PubMedCentralCrossRefPubMed Cho JA, Lee AH, Platzer B, Cross BC, Gardner BM, De Luca H, Luong P, Harding HP, Glimcher LH, Walter P, Fiebiger E, Ron D, Kagan JC, Lencer WI (2013) The unfolded protein response element IRE1α senses bacterial proteins invading the ER to activate RIG-I and innate immune signaling. Cell Host Microbe 13:558–569PubMedCentralCrossRefPubMed
12.
Zurück zum Zitat Lee MS, Cherla RP, Leyva-Illades D, Tesh VL (2009) Bcl-2 regulates the onset of shiga toxin 1-induced apoptosis in THP-1 cells. Infect Immun 77:5233–5244PubMedCentralCrossRefPubMed Lee MS, Cherla RP, Leyva-Illades D, Tesh VL (2009) Bcl-2 regulates the onset of shiga toxin 1-induced apoptosis in THP-1 cells. Infect Immun 77:5233–5244PubMedCentralCrossRefPubMed
13.
Zurück zum Zitat Shima K, Klinger M, Schütze S, Kaufhold I, Solbach W, Reiling N, Rupp J (2015) The role of endoplasmic reticulum-related BiP/GRP78 in interferon gamma-induced persistent Chlamydia pneumoniae infection. Cell Microbiol 17:923–934CrossRefPubMed Shima K, Klinger M, Schütze S, Kaufhold I, Solbach W, Reiling N, Rupp J (2015) The role of endoplasmic reticulum-related BiP/GRP78 in interferon gamma-induced persistent Chlamydia pneumoniae infection. Cell Microbiol 17:923–934CrossRefPubMed
14.
15.
Zurück zum Zitat Kaser A, Lee AH, Franke A, Glickman JN, Zeissig S, Tilg H, Nieuwenhuis EE, Higgins DE, Schreiber S, Glimcher LH, Blumberg RS (2008) XBP1 links ER stress to intestinal inflammation and confers genetic risk for human inflammatory bowel disease. Cell 134:743–756PubMedCentralCrossRefPubMed Kaser A, Lee AH, Franke A, Glickman JN, Zeissig S, Tilg H, Nieuwenhuis EE, Higgins DE, Schreiber S, Glimcher LH, Blumberg RS (2008) XBP1 links ER stress to intestinal inflammation and confers genetic risk for human inflammatory bowel disease. Cell 134:743–756PubMedCentralCrossRefPubMed
16.
Zurück zum Zitat Bronner DN, Abuaita BH, Chen X, Fitzgerald KA, Nunez G, He Y, Yin XM, O‘Riordan MX (2015) Endoplasmic reticulum stress activates the inflammasome via NLRP3- and caspase-2-driven mitochondrial damage. Immunity 43:451–462CrossRefPubMed Bronner DN, Abuaita BH, Chen X, Fitzgerald KA, Nunez G, He Y, Yin XM, O‘Riordan MX (2015) Endoplasmic reticulum stress activates the inflammasome via NLRP3- and caspase-2-driven mitochondrial damage. Immunity 43:451–462CrossRefPubMed
17.
Zurück zum Zitat Abuaita BH, Burkholder KM, Boles BR, O‘Riordan MX (2015) The endoplasmic reticulum stress sensor inositol-requiring enzyme 1α augments bacterial killing through sustained oxidant production. MBio 6:e00705PubMedCentralCrossRefPubMed Abuaita BH, Burkholder KM, Boles BR, O‘Riordan MX (2015) The endoplasmic reticulum stress sensor inositol-requiring enzyme 1α augments bacterial killing through sustained oxidant production. MBio 6:e00705PubMedCentralCrossRefPubMed
18.
Zurück zum Zitat Qin QM, Pei J, Ancona V, Shaw BD, Ficht TA, de Figueiredo P (2008) RNAi screen of endoplasmic reticulum-associated host factors reveals a role for IRE1alpha in supporting Brucella replication. PLoS Pathog 4:e1000110PubMedCentralCrossRefPubMed Qin QM, Pei J, Ancona V, Shaw BD, Ficht TA, de Figueiredo P (2008) RNAi screen of endoplasmic reticulum-associated host factors reveals a role for IRE1alpha in supporting Brucella replication. PLoS Pathog 4:e1000110PubMedCentralCrossRefPubMed
19.
Zurück zum Zitat Fontana MF, Banga S, Barry KC, Shen X, Tan Y, Luo ZQ, Vance RE (2011) Secreted bacterial effectors that inhibit host protein synthesis are critical for induction of the innate immune response to virulent Legionella pneumophila. PLoS Pathog 7:e1001289PubMedCentralCrossRefPubMed Fontana MF, Banga S, Barry KC, Shen X, Tan Y, Luo ZQ, Vance RE (2011) Secreted bacterial effectors that inhibit host protein synthesis are critical for induction of the innate immune response to virulent Legionella pneumophila. PLoS Pathog 7:e1001289PubMedCentralCrossRefPubMed
20.
Zurück zum Zitat Lim YJ, Choi JA, Choi HH, Cho SN, Kim HJ, Jo EK, Park JK, Song CH (2011) Endoplasmic reticulum stress pathway-mediated apoptosis in macrophages contributes to the survival of Mycobacterium tuberculosis. PLoS One 6:e28531PubMedCentralCrossRefPubMed Lim YJ, Choi JA, Choi HH, Cho SN, Kim HJ, Jo EK, Park JK, Song CH (2011) Endoplasmic reticulum stress pathway-mediated apoptosis in macrophages contributes to the survival of Mycobacterium tuberculosis. PLoS One 6:e28531PubMedCentralCrossRefPubMed
21.
Zurück zum Zitat Shrestha N, Bahnan W, Wiley DJ, Barber G, Fields KA, Schesser K (2012) Eukaryotic initiation factor 2 (eIF2) signaling regulates proinflammatory cytokine expression and bacterial invasion. J Biol Chem 287:28738–28744PubMedCentralCrossRefPubMed Shrestha N, Bahnan W, Wiley DJ, Barber G, Fields KA, Schesser K (2012) Eukaryotic initiation factor 2 (eIF2) signaling regulates proinflammatory cytokine expression and bacterial invasion. J Biol Chem 287:28738–28744PubMedCentralCrossRefPubMed
22.
Zurück zum Zitat Tsutsuki H, Yahiro K, Ogura K, Ichimura K, Iyoda S, Ohnishi M, Nagasawa S, Seto K, Moss J, Noda M (2016) Subtilase cytotoxin produced by locus of enterocyte effacement-negative shiga-toxigenic Escherichia coli induces stress granule formation. Cell Microbiol. doi:10.1111/cmi.12565 PubMed Tsutsuki H, Yahiro K, Ogura K, Ichimura K, Iyoda S, Ohnishi M, Nagasawa S, Seto K, Moss J, Noda M (2016) Subtilase cytotoxin produced by locus of enterocyte effacement-negative shiga-toxigenic Escherichia coli induces stress granule formation. Cell Microbiol. doi:10.​1111/​cmi.​12565 PubMed
23.
Zurück zum Zitat Halder P, Datta C, Kumar R, Sharma AK, Basu J, Kundu M (2015) The secreted antigen, HP0175, of Helicobacter pylori links the unfolded protein response (UPR) to autophagy in gastric epithelial cells. Cell Microbiol 17:714–729CrossRefPubMed Halder P, Datta C, Kumar R, Sharma AK, Basu J, Kundu M (2015) The secreted antigen, HP0175, of Helicobacter pylori links the unfolded protein response (UPR) to autophagy in gastric epithelial cells. Cell Microbiol 17:714–729CrossRefPubMed
24.
Zurück zum Zitat Yahiro K, Tsutsuki H, Ogura K, Nagasawa S, Moss J, Noda M (2014) DAP1, a negative regulator of autophagy, controls SubAB-mediated apoptosis and autophagy. Infect Immun 82:4899–4908PubMedCentralCrossRefPubMed Yahiro K, Tsutsuki H, Ogura K, Nagasawa S, Moss J, Noda M (2014) DAP1, a negative regulator of autophagy, controls SubAB-mediated apoptosis and autophagy. Infect Immun 82:4899–4908PubMedCentralCrossRefPubMed
25.
Zurück zum Zitat Gade P, Ramachandran G, Maachani UB, Rizzo MA, Okada T, Prywes R, Cross AS, Mori K, Kalvakolanu DV (2012) An IFN-γ-stimulated ATF6-C/EBP-β-signaling pathway critical for the expression of death associated protein kinase 1 and induction of autophagy. Proc Natl Acad Sci USA 109:10316–10321PubMedCentralCrossRefPubMed Gade P, Ramachandran G, Maachani UB, Rizzo MA, Okada T, Prywes R, Cross AS, Mori K, Kalvakolanu DV (2012) An IFN-γ-stimulated ATF6-C/EBP-β-signaling pathway critical for the expression of death associated protein kinase 1 and induction of autophagy. Proc Natl Acad Sci USA 109:10316–10321PubMedCentralCrossRefPubMed
Metadaten
Titel
Diverse roles of endoplasmic reticulum stress sensors in bacterial infection
verfasst von
Helena Pillich
Maria Loose
Klaus-Peter Zimmer
Trinad Chakraborty
Publikationsdatum
01.12.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
Molecular and Cellular Pediatrics / Ausgabe 1/2016
Elektronische ISSN: 2194-7791
DOI
https://doi.org/10.1186/s40348-016-0037-7

Weitere Artikel der Ausgabe 1/2016

Molecular and Cellular Pediatrics 1/2016 Zur Ausgabe

ADHS-Medikation erhöht das kardiovaskuläre Risiko

16.05.2024 Herzinsuffizienz Nachrichten

Erwachsene, die Medikamente gegen das Aufmerksamkeitsdefizit-Hyperaktivitätssyndrom einnehmen, laufen offenbar erhöhte Gefahr, an Herzschwäche zu erkranken oder einen Schlaganfall zu erleiden. Es scheint eine Dosis-Wirkungs-Beziehung zu bestehen.

Erstmanifestation eines Diabetes-Typ-1 bei Kindern: Ein Notfall!

16.05.2024 DDG-Jahrestagung 2024 Kongressbericht

Manifestiert sich ein Typ-1-Diabetes bei Kindern, ist das ein Notfall – ebenso wie eine diabetische Ketoazidose. Die Grundsäulen der Therapie bestehen aus Rehydratation, Insulin und Kaliumgabe. Insulin ist das Medikament der Wahl zur Behandlung der Ketoazidose.

Frühe Hypertonie erhöht späteres kardiovaskuläres Risiko

Wie wichtig es ist, pädiatrische Patienten auf Bluthochdruck zu screenen, zeigt eine kanadische Studie: Hypertone Druckwerte in Kindheit und Jugend steigern das Risiko für spätere kardiovaskuläre Komplikationen.

Betalaktam-Allergie: praxisnahes Vorgehen beim Delabeling

16.05.2024 Pädiatrische Allergologie Nachrichten

Die große Mehrheit der vermeintlichen Penicillinallergien sind keine. Da das „Etikett“ Betalaktam-Allergie oft schon in der Kindheit erworben wird, kann ein frühzeitiges Delabeling lebenslange Vorteile bringen. Ein Team von Pädiaterinnen und Pädiatern aus Kanada stellt vor, wie sie dabei vorgehen.

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.