• No results found

RAGE and the innate immune response in infection and inflammation - Chapter 4: Receptor for advanced glycation end products protects against Klebsiella pneumoniae induced pneumonia in mice

N/A
N/A
Protected

Academic year: 2021

Share "RAGE and the innate immune response in infection and inflammation - Chapter 4: Receptor for advanced glycation end products protects against Klebsiella pneumoniae induced pneumonia in mice"

Copied!
18
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

UvA-DARE is a service provided by the library of the University of Amsterdam (https://dare.uva.nl)

RAGE and the innate immune response in infection and inflammation

van Zoelen, M.A.D.

Publication date 2009

Link to publication

Citation for published version (APA):

van Zoelen, M. A. D. (2009). RAGE and the innate immune response in infection and inflammation.

General rights

It is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), other than for strictly personal, individual use, unless the work is under an open content license (like Creative Commons).

Disclaimer/Complaints regulations

If you believe that digital publication of certain material infringes any of your rights or (privacy) interests, please let the Library know, stating your reasons. In case of a legitimate complaint, the Library will make the material inaccessible and/or remove it from the website. Please Ask the Library: https://uba.uva.nl/en/contact, or a letter to: Library of the University of Amsterdam, Secretariat, Singel 425, 1012 WP Amsterdam, The Netherlands. You will be contacted as soon as possible.

(2)

4

C

ha

p

te

r

Receptor for advanced glycation end

products protects against

Klebsiella

pneumoniae induced pneumonia in

mice

Marieke A.D. van Zoelen,1,2 Alex F. de Vos,1,2 Sandrine Florquin,3 Angelika Bierhaus,4

Regina de Beer,1,2 Peter P. Nawroth4 and Tom van der Poll1,2

Submitted

1Center for Infection and Immunity Amsterdam (CINIMA), 2Center for Experimental and Molecular Medicine

(CEMM), 3Department of Pathology; Academic Medical Center, University of Amsterdam, Amsterdam, The

Netherlands

(3)

Abstract

Klebsiella species is the second most commonly isolated gram-negative organism in

sepsis and a frequent causative pathogen in pneumonia. The receptor for advanced glycation end products (RAGE) is expressed on different cell types and plays a key role in diverse inflammatory responses. We here investigated the role of RAGE in the host response to Klebsiella (K.) pneumoniae pneumonia and intranasally inoculated RAGE

deficient (RAGE-/-) and normal wild-type (Wt) mice with K. pneumoniae. K. pneumoniae

pneumonia was associated with an increased pulmonary expression of RAGE compared to healthy, uninfected mice. RAGE deficiency impaired host defense as reflected by a worsened survival, increased bacterial outgrowth and dissemination in RAGE deficient mice. RAGE deficient mice infected with K. pneumoniae showed similar lung inflammation,

and slightly elevated - if any - cytokine and chemokine levels and unchanged hepatocellular injury. In addition, RAGE deficient mice displayed an unaltered response to intranasally instilled Klebsiella lipopolysaccharide with respect to pulmonary cell recruitment and

local release of cytokines and chemokines. These data suggest that RAGE contributes to an effective antibacterial host response during K. pneumoniae pneumonia. Furthermore,

we established that RAGE plays an insignificant part in the lung inflammatory response to either intact Klebsiella or Klebsiella LPS.

(4)

Introduction

Gram-negative pneumonia is a common and serious illness that is a major cause of morbidity and mortality in humans. Klebsiella (K.) pneumoniae is a frequently isolated

causative pathogen in lower respiratory tract infection (1-3). The increasing microbial resistance to antibiotics resulting in therapy failure and higher mortality rates is an issue of major concern (1). Therefore it is important to gain more insight into the pathogenesis of pneumonia.

Triggering of receptor for advanced glycation end products (RAGE) results in sustained receptor-dependent signaling and activation of nuclear factor-κB and mitogen-activated protein kinase pathways. Its known ligands are amongst others advanced glycation end products (4), amyloid (5), β-sheet fibrils (6), high mobility group box 1 (7, 8) and some members of the S100 family, including S100A12 (9), S100B (10) and S100P (11). Inhibition of RAGE signaling has been found to reduce inflammatory responses in animal models of hepatic injury (12-14), diabetic atherosclerosis (15, 16), delayed type hypersensitivity (17, 18), type II collagen-induced arthritis (18) and sepsis (17).

RAGE is expressed in normal, healthy lungs (19-23) and pulmonary RAGE expression is enhanced in patients with pneumonia (21). Earlier, we found that RAGE deficiency protects against pneumonia casued by the gram-positive bacterium Streptococcus pneumoniae (S. pneumoniae) as reflected by an enhanced survival, diminished outgrowth

at the primary site of infection and a decreased spreading of bacteria to other body compartments together with reduced lung damage (24) (chapter 3). Whereas S. pneumoniaeis the most commonly isolated pathogen in patients with community-acquiredpneumonia, K. pneumoniae is a causative organism inboth community-acquired and nosocomial pneumonia (25, 26). We here sought to determine the role of RAGE in pneumonia caused by K. pneumoniae. For this, we first investigated RAGE expression in

the lungs during K. pneumoniae pneumonia and next, we intranasally infected RAGE

deficient mice (RAGE-/- mice) with K. pneumoniae and compared the course of the

(5)

Materials and Methods

Animals

Ten week old male RAGE deficient (RAGE-/-) mice were generated as previously described

(17) and backcrossed ten times to a C57Bl/6 background. Wt type C57Bl/6 mice were obtained from Harlan Sprague Dawley Inc. (Horst, The Netherlands). The Institutional Animal Care and Use Committee of the Academic Medical Center, University of Amsterdam, approved all experiments.

Induction of pneumonia and LPS induced lung inflammation

Pneumonia was induced as described earlier (27-29). K. pneumoniae serotype 2 (ATCC

43816; American Type Culture Collecion, Manassas, VA) was cultured for 16 h at 37 ˚C in 5% CO2 in tryptic soy broth (TSH, Difco, Detroit, MI). This suspension was diluted 1:100 in fresh medium and grown for 3 h to midlogarithmic phase. Bacteria were harvested by centrifugation at 1,500 x g for 15 min., washed twice in sterile 0.9% saline and resuspended in saline. The number of colony forming units (CFUs) was determined by plating 10-fold dilutions of the suspensions on blood agar plates. After preparation of the bacterial inoculum, mice were slightly anesthetized by inhalation of isoflurane (Abott, Queensborough, Kent, UK) and 50 µl of bacterial suspension (1 x 104 CFUs) was inoculated intranasally. In survival studies, mice were monitored for 2

weeks (1 x 104 CFUs). In a separate experiment, 50 µl of lipopolysaccharide (LPS) from

K. pneumoniae (Sigma, St. Louis, MO) (100 µg) was inoculated intranasally. Uninfected

(control) mice and mice inoculated with K. pneumoniae or with LPS from K. pneumoniae

were anesthetized with ketamine (Eurovet Animal Health BV, Bladel, The Netherlands) and medetomidine (Pfizer Animal Health BV, Capelle aan de IJssel, The Netherlands) at indicated time points for sample harvesting.

Preparation of blood samples and organ homogenates

Blood was collected from the vena cava inferior in heparin containing tubes and centrifuged

at 1500 x g for 10 minutes, after which plasma was collected and frozen at -20 ºC until assayed. The lungs were removed and processed as described previously (27, 30, 31). Lungs, liver and spleen were harvested and homogenized at 4 ºC in 4 volumes of sterile isotonic saline with a tissue homogenizer (Biospect Products, Bartlesville, OK) which was carefully cleaned and desinfected with 70% ethanol after each homogenization. Serial 10-fold dilutions in sterile saline were made from these homogenates and blood and 50 μl volumes were plated onto sheep-blood agar plates and incubated at 37 ºC and 5% CO2. CFUs were counted after 24 h. For cytokine, chemokine and myeloperoxidase (MPO) measurements, lung homogenates were lysed in 1 volume of lysis buffer (300 mM NaCl, 15 mM Tris [tris(hydroxymethyl)aminomethane], 2 mM MgCl2, 2 mM Triton X-100,

(6)

pepstatin A, leupeptin and aprotinine [20 ng/ml], pH 7.4) on ice for 30 minutes and centrifuged at 1500 x g at 4 ºC for 10 minutes. The supernatants were frozen at -20 ºC until assayed.

Bronchoalveolar lavage

The trachea was exposed through a midline incision and cannulated with a sterile 22-gauge Abbocath-T catheter (Abbott Laboratories, Sligo, Ireland). Bilateral bronchoaolveolar lavage (BAL) was performed by instilling two 0.5-ml aliquots of sterile isotonic saline. 0.9-1 ml of lavage fluid was retrieved per mouse.

Cell counts and differentials

Cell counts were determined in BALF using an automated counter (Beckham Coulter, Coulter ZF, Mijdrecht, The Netherlands). Subsequently, BALF was centrifuged and supernatant was stored at -20 ºC until further assayed; the pellet was suspended in phosphate-buffered saline until a final concentration of 105 cells/ml and differential

cell counts were performed on cytospin Giemsa stain (Diff-Quick; Dade Behring AG, Düdingen, Switzerland).

Histological examination

Lungs for histologic examination were harvested after 24 and 48 h, fixed in 4% formaldehyde and embedded in paraffin. 4-µm thick sections were stained with hematoxylin-eosin and analyzed by a pathologist who whad no knowledge of the genotype of the mice. To score lung inflammation and damage, the lung samples were screened for the following parameters: interstitial inflammation, vaculitis, bronchitis, edema and pleuritis. Each parameter was graded on a scale of 0 to 5 (0, absent; 1, very mild; 2, mild; 3, moderate; 4, severe and 5, very severe). The total histology score was expressed as the (mean) sum of the score for all parameters. Immunostaining for RAGE was performed on paraffin slides after deparaffinization and rehydration using standard procedures. Endogenous peroxidase activity was quenched using 1.5% H2O2 in PBS. Primary antibodies used were goat anti-mouse RAGE polyclonal antibodies (Neuromics, Edina, MN) and secondary antibodies were biotinlylated rabbit anti-goat antibodies (DakoCytomation, Glostrup, Denmark). ABC solution (DakoCytomation, Glostrup, Denmark) was used as the detection enyzme. DAB peroxidase (Sigma, St. Louis, MO) was used as substrate for visualization. Counterstaining was performed with methylgreen (Sigma, St. Louis, MO).

Assays

Tumor necrosis factor (TNF)-α, interleukin (IL)-6, monocyte chemoattractant protein (MCP)-1 and IL-10 levels were determined using a cytometric beads array (CBA)

(7)

multiplex assay (BD Biosciences, San Jose, CA) in accordance with the manufacturer’s recommendations. Keratinocyte-derived chemokine (KC) and macrophage inflammatory

protein 2 (MIP-2) levels were measured by ELISA (R&D Sytstems, Abingdon, United Kingdom) according to the manufacturer’s instructions. Myeloperoxidase (MPO) was measured by ELISA(Hycult Biotechnology BV, Uden, The Netherlands).Aspartate aminotransferase (AST) and alanine aminotransferase (ALT) were determined with commercially available kits (Sigma-Aldrich, St. Louis, MO), using a Hitachi analyzer (Boehringer Mannheim, Mannheim, Germany).

Statistical analysis

Alldata are expressed asmeans ± SEM. Differences between groups were analyzed by Mann-Whitney U test. Values of p < 0.05 were considered to represent a statistically

significant difference.

Results

RAGE expression in the lungs

To obtain constitutive and K. pneumoniae-induced RAGE expression, we performed

immunhistochemical stainings of RAGE of lung tissue from healthy, uninfected Wt mice and from Wt mice after inoculation with K. pneumoniae. In accordance with the

literature (19-23), the normal, healthy mice showed extensive RAGE staining in their lungs (Fig. 1A), being mainly present in the interalveolar septae in an endothelial pattern, while bronchial epithelial cells were negative for RAGE staining (Fig. 1A, arrow). The specificity of the RAGE staining was confirmed by immunohistochemical analysis of lungs obtained from RAGE-/- mice, used as negative controls (Fig. 1B). Lungs from K.

pneumoniae infected mice displayed the same pattern of RAGE positivity as lungs from

Figure 1. Expression of RAGE in lungs during K. pneumoniae pneumonia. Representative view of

a lung from a normal, uninfected Wt mouse (A) displaying ubiquitous expression of RAGE on the surface of endothelium. (B) Absence of RAGE positivity in the lung of a RAGE-/- mouse. (C) Lungs

from a Wt mouse 48 h after the inoculation of K. pneumoniae. Arrow indicates bronchial epithelium in healthy lungs (A), being negative for RAGE staining. RAGE staining; original magnification x10.

(8)

healthy Wt mice, i.e. the interalveolar septae stained positive for RAGE staining with

an endothelial pattern; however, RAGE expression was enhanced following pulmonary infection with K. pneumoniae as reflected by more intense staining (Fig. 1C).

RAGE deficiency enhances lethality due to

K. pneumoniae pneumonia

To study the contribution of RAGE to the outcome of Klebsiella pneumonia, Wt and

RAGE-/- mice were intranasally inoculated with K. pneumoniae and observed for 14

days (Fig. 2). Although the first deaths occurred after 2 days in both strains, all RAGE -/- mice had died after 10 days, while only 50% of the Wt mice had died at the end of the

observation period (p < 0.05). Thus, RAGE deficiency rendered mice more susceptible to K. pneumoniae induced death.

Figure 2. RAGE-/- mice demonstrate an increased

mortality during K. pneumoniae pneumonia. Survival or Wt and RAGE-/- mice after intranasal

inoculation with 1 x 104 CFUs K. pneumoniae.

Mortality was assessed for 14 days (n = 13-14 mice per genotype in each experiment).

RAGE deficiency facilitates early bacterial outgrowth and dissemination

To obtain insight in the mechanism underlying the higher mortaility of RAGE-/- mice,

we repeated this experiment and sacrificed mice 24 and 48 h after infection (i.e. directly

before the first mice started dying) to enumerate bacterial counts in lungs, blood, liver and spleen. At 24 h after infection, bacterial outgrowth in the lungs were similar in Wt and RAGE-/- mice. However, after 48 h, the number of Klebsiella CFUs was higher in

the lungs of RAGE-/- mice when compared to the Wt mice (p < 0.05; Fig. 3A). At 24 h,

significantly increased CFU counts were recovered from blood, liver and spleen harvested from the RAGE-/- mice compared to Wt mice. Liver homogenates from RAGE-/- mice

showed increased bacterial loads at 48 h as well (all p < 0.05; Fig. 3B-D). Thus, RAGE

serves to limit the outgrowth of K. pneumoniae in the lungs and the ensuing dissemination

(9)

Figure 3. RAGE-/- mice

demonstrate an enhanced local bacterial outgrowth and dissemination during K.

pneumoniae pneumonia. Bacterial

loads in lung homogenate (A), blood (B), liver (C) and spleen (D) were determined in Wt and RAGE-/- mice 24 and 48 h after

intranasal inoculation 1 x 104 CFUs

K. pneumoniae. Data are means ±

SEM of 8-10 mice per genotype at each time point. *, p < 0.05 vs Wt mice.

Figure 4. Unchanged lung inflammation

during Klebsiella pneumonia. Wt and RAGE-/- mice were inoculated

intranasally with 1 x 104 CFUs K.

pneumoniae. Representative HE

stainings of lung tissue at 24 (A and B) and 48 (C and D) h post inoculation in Wt (A and C) and RAGE-/- (B and

D) mice. Original magnification x20. Graphical representation of the degree of lung inflammation at 24 and 48 h (E) determined according to the scoring system described in the Materials and Methods section. Myeloperoxidase (MPO) levels in lung tissues (F). Data are means ± SEM of 8-10 mice per genotype at each time point.

(10)

RAGE deficiency does not impact on lung inflammation during

K.

pneumoniae pneumonia

Considering that RAGE signaling results in sustained cellular activation we were interested to study the role of RAGE in lung inflammation during Klebsiella pneumonia. Thus, we

analyzed lung tissue slides obtained from Wt and RAGE-/- mice 24 and 48 h after infection.

At both time points, both mouse strains displayed interstitial inflammation together with vasculitis, peri-bronchitis, edema and pleuritis (Fig. 4A-D). Importantly, in contrast to our expectation, the extent of lung inflammation, as determined by the semi-quantitative scoring system described in the Materials and Methods section, analyzing the severity of vasculitis, bronchitis, edema and pleuritis, was not different between Wt and RAGE

-/-mice (Fig. 4E). In addition, MPO concentrations in lung homogenates of Wt and RAGE -/- mice were similar at both time points (Fig. 4F), indicating that RAGE deficiency did

not influence neutrophil recruitment. Together these data suggest that RAGE does not play a significant role in the lung inflammation that accompanies Klebsiella pneumonia.

Cytokine and chemokine levels

In pulmonary infection, cytokines and chemokines production is an important factor in the host immune response (32, 33). We determined the influence of RAGE deficiency on pulmonary and systemic cytokine and chemokine concentrations during Klebsiella

pneumonia. Levels of the cytokines tumor necrosis factor (TNF)-α, IL-6, MCP-1 and IL-10 and of chemokines KC and MIP-2 did not differ between the two mouse strains at 24 h. At 48 h, MCP-1 and KC concentrations were increased in the lungs (p < 0.05;

Table I). In plasma, TNF-α, IL-6 nd MCP-1 levels were similar between the two mouse strains at both time points, while IL-10 was elevated in the RAGE-/- mice at 48 h (p <

0.05; Table I).

Wt and RAGE

-/-

mice display similar hepatocellular injury

This model of Klebsiella pneumonia is associated with hepatocellular injury (29).

Considering the enhanced lethality and sustained elevated bacterial loads in liver homogenates in RAGE-/- mice, we were interested to examine the extent of hepatocellular

injury in both mouse strains (Fig. 5). At 24 h after infection, neither RAGE-/- nor Wt mice

demonstrated elevated plasma concentrations of AST or ALT. At 48 h post infection, both mouse strains had strongly elevated plasma transaminase levels; although these levels tended to be higher in RAGE-/- mice, the differences with Wt mice did not reach

(11)

Table I. Cytokine and chemokine levels in Wt and RAGE-/- micea 24 h 48 h Wt RAGE-/- Wt RAGE Lung homogenate (pg/ml) TNF-α 724 ± 187 446 ± 66 1,679 ± 932 5,151 ± 1617 IL-6 1348 ± 252 723 ± 240 706 ± 301 844 ± 223 MCP-1 6172 ± 381 6006 ± 444 2,861 ± 282 4,838 ± 623 ** IL-10 545 ± 99 482 ± 50 26 ± 3 32 ± 6 KC 13991 ± 1847 13026 ± 1851 11,257 ± 2,251 20,478 ± 3,372 * MIP-2 8082 ± 1818 11298 ± 2300 12,085 ± 4,367 7,548 ± 1,467 Plasma (pg/ml) TNF-α 87 ± 30 141 ± 47 62 ± 18 56 ± 26 IL-6 211 ± 49 211 ± 58 277 ± 159 478 ± 157 MCP-1 827 ± 188 1,777 ± 444 519 ± 139 526 ± 231 IL-10 14 ± 3 21 ± 3 1 ± 1 8 ± 4 *

a Data are means ± SEM at 24 or 48 h post inoculation of 1 x 104 CFUs K. pneumoniae.

n = 8-10 mice per group.

*, p < 0.05 vs Wt mice; **, p < 0.01 vs Wt mice.

Figure 5. Hepatocellular injury during K. pneumoniae pneumonia. Wt and RAGE-/- mice were

inoculated intranasally with 1 x 104 CFUs K. pneumoniae and sacrificed after 24 and 48 h.

Aspartate aminotransferase (AST, A) and alanine aminotransferase (ALT, B) in plasma of Wt and RAGE-/- mice. Data are means ± SEM of 8-10 mice per genotype at each time point.

(12)

RAGE in Klebsiella pneumonia

Table II. Cell counts, cytokine and chemokine levels in bronchoalveolar lavage fluid after Klebsiella LPSa

Wt RAGE-/- Cells (x 105/ml) Total cells 5.8 ± 1.0 8.2 ± 1.4 Neutrophils 5.5 ± 1.0 7.7 ± 1.3 Cytokines (pg/ml) TNF-α 5,561 ± 819 6,212 ± 568 IL-6 2,542 ± 973 3,989 ± 583 KC 1327 ± 209 870 ± 81 MIP-2 661 ± 79 516 ± 34

a Data are means ± SEM at 6 h post inoculation of 100 μg

LPS from K. pneumoniae. n = 8-10 mice per group.

RAGE

-/-

mice demonstrate an unchanged inflammatory response to

K.

pneumoniae LPS

In light of the strong expression of RAGE in the lung and its reported role as a receptor mediating pro-inflammatory effects, we were surprised to find unaltered lung inflammation and (if anything) higher cytokine levels in RAGE-/- mice during Klebsiella

pneumonia. To obtain further evidence for a modest role of RAGE in the induction of lung inflammation in response to a gram-negative bacterium, we compared the inflammatory response to Klebsiella lipopolysaccharide (LPS), administered via the airways, in Wt and

RAGE-/- mice. In these studies we harvested bronchoalveolar lavage fluid (BALF) 6 h

after LPS administration considering that this time point is representative for examining LPS responses in the pulmonary compartment (27, 34, 35). In line with the observations during respiratory tract infection with live K. pneumoniae, RAGE-/- mice displayed an

unaltered response to intranasally instilled Klebsiella LPS with respect to pulmonary cell

recruitment and local release of cytokines and chemokines (Table II).

Discussion

Klebsiella species is the second most commonly isolated gram-negative organism in sepsis

(36, 37) and a frequent causative pathogen in pneumonia (1). RAGE is a multiligand receptor of the immunoglobulin superfamily that is expressed in all tissues by a wide range of cell types, including cells involved in the innate immune system, e.g. neutrophils,

(13)

signaling pathways, resulting in pro-inflammatory gene expression upon interaction with several distinct endogenous proinflammatory ligands. Therefore, RAGE may function as a sensor of danger signals leading to a certain amount of inflammation and hence play a beneficial role in bacterial eradication during infection. However, interaction of RAGE with its ligands and the subsequently induced inflammation can also worsen tissue damage, thereby exerting detrimental effects. Earlier, we found that RAGE impairs host defense in gram-positive pneumonia. We here established that RAGE contributes to an effective antibacterial host response during K. pneumoniae pneumonia. Indeed, RAGE

-/-mice displayed higher bacterial counts at the primary source of infection and increased spreading of bacteria to blood and distant organs, which was associated with a decreased survival.

We confirmed previous studies showing that normal healthy lungs constitutively express RAGE, especially in the alveolar septae in an endothelial pattern (19-23). Earlier, pulmonary RAGE expression was shown to be enhanced in patients with postobstructive pneumonia (21). Our present finding that K. pneumoniae pneumonia is associated with

an upregulation of interalveolar RAGE expression in the lungs together with similar observations in pneumococcal pneumonia strongly suggests that enhanced pulmonary RAGE expression is a common response to bacterial pneumonia.

The current data should be considered in the context of several earlier studies on the role of RAGE during bacterial infections. In a model of polymicrobial abdominal sepsis induced by cecal ligation and puncture, RAGE-/- mice had an improved survival together

with a reduced NF-κB activation in the peritoneum; bacterial growth and dissemination were not determined in this study (17). We previously investigated the role of RAGE during abdominal sepsis induced by the gram-negative bacterium Escherichia coli (E. coli),

showing that RAGE deficiency was associated with an enhanced outgrowth of E. coli

locally and in distant organs together with more severe liver injury (chapter 2). In contrast, we found that in a model of pneumonia by the gram-positive bacterium S. pneumoniae,

RAGE deficiency was associated with a reduced bacterial outgrowth and dissemination and less severe lung damage (24) (chapter 3). RAGE deficiency or inhibition did not impact on the growth of Listeria monocytogenes after intraperitoneal injection (39).

These data suggest that RAGE plays differential roles in antibacterial defense during experimental infections with different pathogens.

The recruitment of neutrophils is an important part of host defense against pneumonia (40, 41). RAGE has been implicated as a mediator of cell trafficking. Indeed, RAGE

(14)

ligation and puncture (17) and a reduction in neutrophil influx in the peritoneal cavity during thioglycollate peritonitis (42). Furthermore, in vivo studies have suggested that

RAGE is an endothelial counter receptor for the β2 integrin Mac-1 (42, 43). During pneumococcal pneumonia RAGE-/- mice showed an attenuated influx of neutrophils

into the lungs (24) (chapter 3). In contrast, we did not find an effect of RAGE deficiency on cell influx during K. pneumoniae pneumonia, as indicated by histopathology and

pulmonary MPO concentrations. Moreover, leukocyte counts and differentials in BALF harvested after intrapulmonary delivery of Klebsiella LPS were similar in Wt and RAGE-/- mice.

Together these data suggest that RAGE does not play a role of importance in leukocyte recruitment to the lungs during gram-negative infection, and that the impact of RAGE on cell trafficking may depend on the inflammatory stimulus and the organ involved. Of note, the model of (sterile) lung inflammation induced by Klebsiella LPS was also

used to study the role of RAGE while avoiding potential bias due to differences in the pro-inflammatory stimulus caused by differences in pulmonary bacterial loads between Wt and RAGE-/- mice. These experiments confirmed the results obtained after infection

with viable Klebsiella, i.e. that RAGE plays an insignificant part in the lung inflammatory

response to either intact Klebsiella or Klebsiella LPS.

Pneumonia represents a major cause of morbidity and mortality. We here show that RAGE plays a protective role during respiratory tract infection by a common gram-negative causative pathogen, K. pneumoniae, by improving antibacterial defense in lungs

and reducing bacterial dissemination.

Acknowledgements

We thank Regina de Beer, Joost Daalhuisen and Marieke S. ten Brink for expert technical assistance.

(15)

References

1. Burwen, D. R., S. N. Banerjee, and R. P. Gaynes. 1994. Ceftazidime resistance among selected nosocomial gram-negative bacilli in the United States. National Nosocomial Infections Surveillance System. J. Infect. Dis. 170:1622-1625.

2. Podschun, R. and U. Ullmann. 1998. Klebsiella spp. as nosocomial pathogens: epidemiology, taxonomy, typing methods, and pathogenicity factors. Clin. Microbiol. Rev. 11:589-603.

3. Yinnon, A. M., A. Butnaru, D. Raveh, Z. Jerassy, and B. Rudensky. 1996. Klebsiella bacteraemia: community versus nosocomial infection. QJM. 89:933-941.

4. Kislinger, T., C. Fu, B. Huber, W. Qu, A. Taguchi, Y. S. DU, M. Hofmann, S. F. Yan, M. Pischetsrieder, D. Stern, and A. M. Schmidt. 1999. N(epsilon)-(carboxymethyl)lysine adducts of proteins are ligands for receptor for advanced glycation end products that activate cell signaling pathways and modulate gene expression. J. Biol. Chem. 274:31740-31749.

5. Yan, S. D., H. Zhu, A. Zhu, A. Golabek, H. Du, A. Roher, J. Yu, C. Soto, A. M. Schmidt, D. Stern, and M. Kindy. 2000. Receptor-dependent cell stress and amyloid accumulation in systemic amyloidosis. Nat. Med. 6:643-651.

6. Yan, S. D., X. Chen, J. Fu, M. Chen, H. Zhu, A. Roher, T. Slattery, L. Zhao, M. Nagashima, J. Morser, A. Migheli, P. Nawroth, D. Stern, and A. M. Schmidt. 1996. RAGE and amyloid-beta peptide neurotoxicity in Alzheimer’s disease. Nature 382:685-691.

7. Sorci, G., F. Riuzzi, C. Arcuri, I. Giambanco, and R. Donato. 2004. Amphoterin stimulates myogenesis and counteracts the antimyogenic factors basic fibroblast growth factor and S100B via RAGE binding. Mol. Cell Biol. 24:4880-4894.

8. Hori, O., J. Brett, T. Slattery, R. Cao, J. Zhang, J. X. Chen, M. Nagashima, E. R. Lundh, S. Vijay, D. Nitecki, and . 1995. The receptor for advanced glycation end products (RAGE) is a cellular binding site for amphoterin. Mediation of neurite outgrowth and co-expression of rage and amphoterin in the developing nervous system. J. Biol. Chem. 270:25752-25761.

9. Moroz, O. V., A. A. Antson, E. J. Dodson, H. J. Burrell, S. J. Grist, R. M. Lloyd, N. J. Maitland, G. G. Dodson, K. S. Wilson, E. Lukanidin, and I. B. Bronstein. 2002. The structure of S100A12 in a hexameric form and its proposed role in receptor signalling.

Acta Crystallogr. D. Biol. Crystallogr. 58:407-413.

10. Valencia, J. V., M. Mone, J. Zhang, M. Weetall, F. P. Buxton, and T. E. Hughes. 2004. Divergent pathways of gene expression are activated by the RAGE ligands S100b and AGE-BSA. Diabetes 53:743-751.

11. Arumugam, T., D. M. Simeone, A. M. Schmidt, and C. D. Logsdon. 2004. S100P stimulates cell proliferation and survival via receptor for activated glycation end products (RAGE). J. Biol. Chem. 279:5059-5065.

12. Cataldegirmen, G., S. Zeng, N. Feirt, N. Ippagunta, H. Dun, W. Qu, Y. Lu, L. L. Rong, M. A. Hofmann, T. Kislinger, S. I. Pachydaki, D. G. Jenkins, A. Weinberg, J. Lefkowitch, X.

(16)

Rogiers, S. F. Yan, A. M. Schmidt, and J. C. Emond. 2005. RAGE limits regeneration after massive liver injury by coordinated suppression of TNF-alpha and NF-kappaB. J. Exp. Med. 201:473-484.

13. Zeng, S., N. Feirt, M. Goldstein, J. Guarrera, N. Ippagunta, U. Ekong, H. Dun, Y. Lu, W. Qu, A. M. Schmidt, and J. C. Emond. 2004. Blockade of receptor for advanced glycation end product (RAGE) attenuates ischemia and reperfusion injury to the liver in mice.

Hepatology 39:422-432.

14. Ekong, U., S. Zeng, H. Dun, N. Feirt, J. Guo, N. Ippagunta, J. V. Guarrera, Y. Lu, A. Weinberg, W. Qu, R. Ramasamy, A. M. Schmidt, and J. C. Emond. 2006. Blockade of the receptor for advanced glycation end products attenuates acetaminophen-induced hepatotoxicity in mice. J. Gastroenterol. Hepatol. 21:682-688.

15. Park, L., K. G. Raman, K. J. Lee, Y. Lu, L. J. Ferran, Jr., W. S. Chow, D. Stern, and A. M. Schmidt. 1998. Suppression of accelerated diabetic atherosclerosis by the soluble receptor for advanced glycation endproducts. Nat. Med. 4:1025-1031.

16. Goova, M. T., J. Li, T. Kislinger, W. Qu, Y. Lu, L. G. Bucciarelli, S. Nowygrod, B. M. Wolf, X. Caliste, S. F. Yan, D. M. Stern, and A. M. Schmidt. 2001. Blockade of receptor for advanced glycation end-products restores effective wound healing in diabetic mice. Am. J. Pathol. 159:513-525.

17. Liliensiek, B., M. A. Weigand, A. Bierhaus, W. Nicklas, M. Kasper, S. Hofer, J. Plachky, H. J. Grone, F. C. Kurschus, A. M. Schmidt, S. D. Yan, E. Martin, E. Schleicher, D. M. Stern, G. G. Hammerling, P. P. Nawroth, and B. Arnold. 2004. Receptor for advanced glycation end products (RAGE) regulates sepsis but not the adaptive immune response. J. Clin. Invest 113:1641-1650.

18. Hofmann, M. A., S. Drury, B. I. Hudson, M. R. Gleason, W. Qu, Y. Lu, E. Lalla, S. Chitnis, J. Monteiro, M. H. Stickland, L. G. Bucciarelli, B. Moser, G. Moxley, S. Itescu, P. J. Grant, P. K. Gregersen, D. M. Stern, and A. M. Schmidt. 2002. RAGE and arthritis: the G82S polymorphism amplifies the inflammatory response. Genes Immun. 3:123-135.

19. Cheng, C., K. Tsuneyama, R. Kominami, H. Shinohara, S. Sakurai, H. Yonekura, T. Watanabe, Y. Takano, H. Yamamoto, and Y. Yamamoto. 2005. Expression profiling of endogenous secretory receptor for advanced glycation end products in human organs.

Mod. Pathol. 18:1385-1396.

20. Englert, J. M., L. E. Hanford, N. Kaminski, J. M. Tobolewski, R. J. Tan, C. L. Fattman, L. Ramsgaard, T. J. Richards, I. Loutaev, P. P. Nawroth, M. Kasper, A. Bierhaus, and T. D. Oury. 2008. A role for the receptor for advanced glycation end products in idiopathic pulmonary fibrosis. Am. J. Pathol. 172:583-591.

21. Morbini, P., C. Villa, I. Campo, M. Zorzetto, S. Inghilleri, and M. Luisetti. 2006. The receptor for advanced glycation end products and its ligands: a new inflammatory pathway in lung disease? Mod. Pathol. 19:1437-1445.

(17)

22. Uchida, T., M. Shirasawa, L. B. Ware, K. Kojima, Y. Hata, K. Makita, G. Mednick, Z. A. Matthay, and M. A. Matthay. 2006. Receptor for advanced glycation end-products is a marker of type I cell injury in acute lung injury. Am. J. Respir. Crit Care Med.

173:1008-1015.

23. Wittkowski, H., A. Sturrock, M. A. van Zoelen, D. Viemann, T. van der Poll, J. R. Hoidal, J. Roth, and D. Foell. 2007. Neutrophil-derived S100A12 in acute lung injury and respiratory distress syndrome. Crit Care Med. 35:1369-1375.

24. van Zoelen, M. A., M. Schouten, A. F. de Vos, S. Florquin, J. C. Meijers, P. P. Nawroth, A. Bierhaus, and T. van der Poll. 2009. The receptor for advanced glycation end products impairs host defense in pneumococcal pneumonia. J. Immunol. 182:4349-4356.

25. Bartlett, J. G. and L. M. Mundy. 1995. Community-acquired pneumonia. N. Engl. J. Med. 333:1618-1624.

26. Brown, P. D. and S. A. Lerner. 1998. Community-acquired pneumonia. Lancet

352:1295-1302.

27. Rijneveld, A. W., S. Weijer, S. Florquin, C. T. Esmon, J. C. Meijers, P. Speelman, P. H. Reitsma, H. Ten Cate, and T. van der Poll. 2004. Thrombomodulin mutant mice with a strongly reduced capacity to generate activated protein C have an unaltered pulmonary immune response to respiratory pathogens and lipopolysaccharide. Blood

103:1702-1709.

28. van Westerloo, D. J., S. Knapp, C. van’t Veer, W. A. Buurman, A. F. de Vos, S. Florquin, and T. van der Poll. 2005. Aspiration pneumonitis primes the host for an exaggerated inflammatory response during pneumonia. Crit Care Med. 33:1770-1778.

29. Renckens, R., J. J. Roelofs, P. I. Bonta, S. Florquin, C. J. de Vries, M. Levi, P. Carmeliet, C. van’t Veer, and T. van der Poll. 2007. Plasminogen activator inhibitor type 1 is protective during severe gram-negative pneumonia. Blood 109:1593-1601.

30. Knapp, S., C. W. Wieland, C. van ‘t Veer, O. Takeuchi, S. Akira, S. Florquin, and T. van der Poll. 2004. Toll-like receptor 2 plays a role in the early inflammatory response to murine pneumococcal pneumonia but does not contribute to antibacterial defense. J Immunol

172:3132-8.

31. Wieland, C. W., S. Florquin, N. A. Maris, K. Hoebe, B. Beutler, K. Takeda, S. Akira, and T. van der Poll. 2005. The MyD88-dependent, but not the MyD88-independent, pathway of TLR4 signaling is important in clearing nontypeable haemophilus influenzae from the mouse lung. J. Immunol. 175:6042-6049.

32. Knapp, S., M. J. Schultz, and T. van der Poll. 2005. Pneumonia models and innate immunity to respiratory bacterial pathogens. Shock 24 Suppl 1:12-18.

33. Strieter, R. M., J. A. Belperio, and M. P. Keane. 2003. Host innate defenses in the lung: the role of cytokines. Curr. Opin. Infect. Dis. 16:193-198.

(18)

2002. Differential role of interleukin-6 in lung inflammation induced by lipoteichoic acid and peptidoglycan from Staphylococcus aureus. Am. J. Respir. Crit Care Med.

165:1445-1450.

35. Knapp, S., A. S. von, M. Leendertse, I. Haslinger, C. Draing, D. T. Golenbock, and T. van der Poll. 2008. Lipoteichoic acid-induced lung inflammation depends on TLR2 and the concerted action of TLR4 and the platelet-activating factor receptor. J. Immunol.

180:3478-3484.

36. Bernard, G. R., J. L. Vincent, P. F. Laterre, S. P. LaRosa, J. F. Dhainaut, A. Lopez-Rodriguez, J. S. Steingrub, G. E. Garber, J. D. Helterbrand, E. W. Ely, and C. J. Fisher, Jr. 2001. Efficacy and safety of recombinant human activated protein C for severe sepsis. N Engl J Med

344:699-709.

37. Fisher, C. J., Jr., J. M. Agosti, S. M. Opal, S. F. Lowry, R. A. Balk, J. C. Sadoff, E. Abraham, R. M. Schein, and E. Benjamin. 1996. Treatment of septic shock with the tumor necrosis factor receptor:Fc fusion protein. The Soluble TNF Receptor Sepsis Study Group. N. Engl. J. Med. 334:1697-1702.

38. Bierhaus, A., D. M. Stern, and P. P. Nawroth. 2006. RAGE in inflammation: a new therapeutic target? Curr. Opin. Investig. Drugs 7:985-991.

39. Lutterloh, E. C., S. M. Opal, D. D. Pittman, J. C. Keith, Jr., X. Y. Tan, B. M. Clancy, H. Palmer, K. Milarski, Y. Sun, J. E. Palardy, N. A. Parejo, and N. Kessimian. 2007. Inhibition of the RAGE products increases survival in experimental models of severe sepsis and systemic infection. Crit Care 11:R122.

40. Zhang, P., W. R. Summer, G. J. Bagby, and S. Nelson. 2000. Innate immunity and pulmonary host defense. Immunol. Rev. 173:39-51.

41. Strieter, R. M., J. A. Belperio, and M. P. Keane. 2002. Cytokines in innate host defense in the lung. J. Clin. Invest 109:699-705.

42. Chavakis, T., A. Bierhaus, N. Al Fakhri, D. Schneider, S. Witte, T. Linn, M. Nagashima, J. Morser, B. Arnold, K. T. Preissner, and P. P. Nawroth. 2003. The pattern recognition receptor (RAGE) is a counterreceptor for leukocyte integrins: a novel pathway for inflammatory cell recruitment. J. Exp. Med. 198:1507-1515.

43. Zen, K., C. X. Chen, Y. T. Chen, R. Wilton, and Y. Liu. 2007. Receptor for advanced glycation endproducts mediates neutrophil migration across intestinal epithelium. J. Immunol. 178:2483-2490.

Referenties

GERELATEERDE DOCUMENTEN

The findings obtained in this dissertation shed a first light on factors promoting parochial and universal cooperation. This dissertation thus provides useful

Het gebouw, ook wel de Tabakgarage genoemd, verrees hier tussen 1966 en 1971 naar ontwerp van het architectenbureau Zanstra, Gmelig Meyling, De Clercq, Zubli in opdracht

69 geopolitical units have been thus considered (see Table 1 ). Other ecological or biogeographical subdivision systems of the European mainland could have been envisaged, but are

However, no e¡ect of annexin V binding was observed on the product desorption rates from PS/PE/PC membranes containing 10 mole % of lysoPC and oleic acid (result not shown). It has

Post hoc analysis revealed that the region in blue (LIPL) is smaller in subjects with dyslexia and that the regions in red (LOFG and ROFG) are larger in subjects with

A Resource-based perspective of collective action in the distribution of agricultural input and primary health services in the Couffo region, Benin..

8.3.18.3.1 Strategy of collective action in the distribution of primary health services 213 8.3.28.3.2 Structure of collective action in the distribution of primary health services

reformss in five sub-prefectures of the Couffo region (1996-97) 121 Tablee 5.11 Breakdown of a sample of villages according to quarales of respondents for. theirr perceived