UvA-DARE (Digital Academic Repository)

TREM receptors in infection and inflammation

Hommes, T.J.

Publication date 2015 Document Version Final published version

Link to publication

Citation for published version (APA): Hommes, T. J. (2015). TREM receptors 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 . You will be contacted as soon as possible.

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

Download date:06 Oct 2021

TREM receptors in infection and inflammation

Tijmen Hommes COLOFON

TREM receptors in infection and inflammation Academic Thesis, University of Amsterdam, The Netherlands

ISBN: 978-94-6169-766-0

Copyright © 2015 by T. J. Hommes, Amsterdam, The Netherlands All rights reserved. No part of this thesis may be reproduced, stored, or transmitted in any form or by any means, without prior permission of the author.

Author: Tijmen J. Hommes Cover design: Stefan Glerum Lay-out: Optima Grafische Communicatie, Rotterdam, The Netherlands Printed by: Optima Grafische Communicatie, Rotterdam, The Netherlands

Printing of this thesis was financially supported by: BD Bioscience, Chipsoft, Dr. Falk Pharma Benelux B.V., Erbe, HyCult, Olympus, Pfizer, Stichting Wetenschap en Onder- zoek Interne Geneeskunde OLVG, University of Amsterdam and Zambon TREM receptors in infection and inflammation

ACADEMISCH PROEFSCHRIFT

ter verkrijging van de graad van doctor

aan de Universiteit van Amsterdam

op gezag van de Rector Magnificus

prof. dr. D.C. van den Boom ten overstaan van een door het College voor Promoties ingestelde commissie,

in het openbaar te verdedigen in de Agnietenkapel

op vrijdag 13 november 2015, te 14:00 uur

door Tijmen Johannes Hommes

geboren te Hilversum Promotiecommissie

Promotor: Prof. Dr. T. van der Poll Universiteit van Amsterdam

Copromotor: Dr. A. F. de Vos Universiteit van Amsterdam

Overige leden: Prof. Dr. G. R. van den Brink Universiteit van Amsterdam Prof. Dr. T. B. H. Geijtenbeek Universiteit van Amsterdam Prof. Dr. S. Knapp Universiteit van Wenen Prof. Dr. M. J. Schultz Universiteit van Amsterdam Prof. Dr. P. J. Sterk Universiteit van Amsterdam Dr. W. J. Wiersinga Universiteit van Amsterdam

Faculteit der Geneeskunde “Omtoch”

Voor papa

Table of contents

Chapter 1 General introduction and thesis outline 9

Chapter 2 Triggering Receptor Expressed on Myeloid Cells (TREM)-1 19 improves host defence in pneumococcal pneumonia

Chapter 3 DNAX-activating protein of 12 kDa (DAP12) impairs host defense 45 in pneumococcal pneumonia

Chapter 4 Triggering Receptor Expressed on Myeloid Cells (TREM)-1 65 improves host defense during Staphylococcus aureus pneumonia

Chapter 5 TREM-1/3 contributes to protective immunity in Klebsiella 81 derived pneumosepsis whereas TREM-2 does not

Chapter 6 Triggering Receptor Expressed on Myeloid Cells (TREM)-2 impairs 105 host defense in Gram-negative sepsis (melioidosis)

Chapter 7 Role of Nucleotide-binding Oligomerization Domain-containing 129 protein (NOD) 2 in host defense during pneumococcal pneumonia

Chapter 8 TREM-1/3 contributes to house dust mite induced allergic lung 145 inflammation

Chapter 9 Summary and general discussion 159

Addendum Nederlandse samenvatting 171 PhD portfolio 175 List of publications 177 About the author 179 Acknowledgements (Dankwoord) 181

1 General introduction and thesis outline

General introduction and thesis outline

This thesis describes research conducted to obtain more insight in activation of the innate immune system during lung infection and inflammation, focusing on bacterial pneumonia and asthma, and the role of Triggering Receptor Expressed by Myeloid cells (TREM)-1 and -2 herein. In this introductory chapter we briefly describe the diseases and 1 innate immune receptors studied in this thesis, and we provide an outline of its contents.

Sepsis & pneumonia Sepsis is a clinical syndrome characterized by an uncontrolled and unbalanced host re- sponse to infection ultimately leading to tissue injury, organ dysfunction and oftentimes death1. Sepsis is a major cause of morbidity and mortality worldwide2,3. Although this heterogeneous syndrome has been recognized for more than a thousand years, spe- cific therapies are not available. Thus far, the best treatment remains antibiotic therapy started as soon as possible, where needed combined with source control together with supportive care, including intravenous fluid resuscitation, mechanical ventilation and vasoactive medication4. Despite years of research effective therapies targeting specific derailed host response pathways have not been developed. Numerous clinical trials investigating the efficacy of immune modulatory and anticoagulant therapies have not been successful5. Therefore, a clear understanding of the immunopathological processes underlying sepsis is pivotal in our way to new therapies3. The most important cause of sepsis is pneumonia. Streptococcus pneumoniae is a Gram-positive bacterium and the most frequently isolated causative agent of commu- nity-acquired pneumonia6 while Klebsiella pneumoniae (Gram-negative) is a common pathogen in hospital-acquired pneumonia7. Another Gram-negative bacterium causing community-acquired pneumonia and sepsis is Burkholderia pseudomallei8. This patho- gen is commonly found in Southeast-Asia and Northern Australia and known to cause a life-threatening disease called melioidosis8. Nowadays the emergence of resistant bacteria is a major concern for both physicians and patients. Methicillin-resistant Staphy- lococcus aureus (MRSA) used to be a frequently isolated pathogen in hospital-acquired pneumonia9 but in recent years virulent strains of community-associated MRSA have also emerged, causing infections in individuals without recognized risk factors10.

Host defense during pneumonia The lung with its large surface area is continuously exposed to the outside world and under nonstop attack of invading microbes. During evolution different defense mecha- nisms have evolved to defend the host. These vary from simple physical mechanisms such as coughing and sneezing to more subtle instruments such as alveolar macro- phages and airway epithelial cells. These latter cell types reside constitutively in the lung and can respond to pathogens that reach the lower airways by mounting a protective innate immune response. Central in the detection of microbes by innate immune cells

11 Chapter 1 are pattern recognition receptors (PRRs). Four different families of PRRs are currently recognized: Toll-like receptors (TLRs), Nucleotide-binding Oligomerization Domain (NOD)-like receptors (NLRs), RIG-I-like receptors and C-type lectin receptors.

Toll-like receptors and NOD-like receptors TLRs are evolutionarily conserved receptors expressed by a wide array of cells able to recognize highly conserved microbial motifs known as pathogen-associated microbial patterns (PAMPs)11. In addition, TLRs can recognize host derived molecules released upon cell injury, so-called alarmins or damage-associated molecular patterns (DAMPs). Stimulation of TLRs by their corresponding ligands initiates signaling cascades ultimately giving rise to a proinflammatory response aimed at eradicating pathogens. NLRs are a family of PRRs located within the cytosol12. Different members are recog- nized of which NOD1 and NOD2 are the most prominent ones. Both NOD1 and NOD2 recognize different parts of peptidoglycan, a PAMP expressed by Gram-positive and Gram-negative bacteria13. Activation of both receptors leads to a pro-inflammatory re- sponse through activation of p38 mitogen-activated protein (MAP) kinase and Nuclear factor –kappa B (NF-κB)13.

TREM-receptors: Fine tuning the innate immune response Although the innate immune response is crucial for host survival during infection, it must be kept in check in order to prevent excessive inflammation and subsequent tissue damage. Different proteins exist to help set the threshold for cellular responses to spe- cific stimuli and to modulate the magnitude of the response. TREM proteins are a family of cell surface receptors primarily expressed by leukocytes which are able to amplify or dampen innate immune responses14 thereby fine-tuning innate immune responses. TREM-1 was the first TREM family member characterized15. TREM-1 is a member of the immunoglobulin super family and is expressed mainly by neutrophils, monocytes and macrophages and is able to amplify TLR and NLR-signaling15,16 thereby lowering the threshold for pathogen detection. TREM-1 is upregulated in response to lipopolysaccha- ride and other microbial products. In vivo TREM-1 is highly expressed in inflammatory lesions caused by bacteria, more so than in lesions from non-infectious origin17. TREM-1 associates with adaptor molecule DNAX-activating protein of 12kDa (DAP12), which contains immunoreceptor tyrosine-based activation motifs (ITAMs)15. In response to receptor ligation the tyrosine residues in the ITAMs of DAP12 become phosphorylated which results in downstream signal transduction through extracellular signal-related kinase (ERK) 1/2 and NF-kB translocation to the nucleus causing proinflammatory cytokine secretion15 (Figure 1). How exactly TREM-1 is activated remains elusive. Differ- ent endogenous ligands have been described, including High-mobility group protein B1 (HMGB1), heat shock protein 70 (HSP70)18, a ligand expressed by human platelets19

12 General introduction and thesis outline

1

Figure 1. Thesis outline and (simplified) overview of TREM- and TLR signaling. The different research questions are depicted in red. DAP12: DNAX-activating protein of 12kDa; HDM: House dust mite HMGB1: High Mobility Group Box 1; HSP70: heat shock protein 70; MRSA: methicillin resistant Staphylococcus au- reus; NOD2: nucleotide-oligomerization domain-containing protein 2; NFkB: nuclear factor kappa B; PG- LYRP1: peptidoglycan recognition protein 1; Rip2: receptor-interacting protein 2; TLR: Toll-like receptor; TREM: Triggering Receptor Expressed on Myeloid cells. and more recently peptidoglycan recognition protein 1 (PGLYRP1)20. Additionally to a membrane bound form of TREM-1, a soluble form exists. It is thought that TREM-1 is cleaved from the cell membrane by metalloproteinases to give rise to a soluble form21. Soluble TREM-1 has been proposed as a diagnostic marker for different diseases, espe- cially infections22. TREM-2 is closely related to TREM-1 although its expression profile and function are different. TREM-2 is mainly expressed by macrophages, dendritic cells, microglia and osteoclasts. In humans TREM-2 mutations have been linked to a rare syndrome called Nasu-Hakola, which is associated with pre-senile dementia and bone cysts23. Opposite to TREM-1, TREM-2 is able to inhibit TLR responses in leukocytes and serves as a phago- cytic receptor for different bacterial species24-26. Like TREM-1, TREM-2 signals through DAP12 although the downstream signaling pathways have not been elucidated quite as extensively as for TREM-1. Quite interestingly, since both TREM-1 and TREM-2 signal through DAP12 their functions seem opposite. A model has been proposed where

13 Chapter 1 ligand-receptor avidity mediates the net effect they have on either cellular activation or inhibition27. Both TREM-1 and TREM-2 serve roles in host defense against bacterial infections. Blocking TREM-1 during severe sepsis lead to increased host survival in a murine model of abdominal sepsis17. In contrast, TREM-2 was found to have a protective role in this model28, exemplifying their opposite functions. However in pneumonia derived sepsis their roles remain ill-defined.

Allergic lung inflammation Asthma is a disease characterized by chronic airway inflammation. Typically patients experience recurrent periods of wheezing, shortness of breath and coughing29. Asthma puts a great burden on healthcare expenditure and is a major cause of morbidity world- wide30. Immunologically asthma represents a Th2 driven response with a central role for lymphocytes, eosinophils and the subsequent production of IgE31. Relatively new is the finding of a role for the innate immune system in the development of the disease31. Recently it was found TLR4 has a role in the pathogenesis of allergic airway inflammation in mice32,33. House dust mite is a major allergen in human asthma34.

Thesis outline

The general aim of this thesis is to increase our insight in the immunopathogenesis of pulmonary inflammatory responses during bacterial infection and allergic lung re- sponses with a focus on fine-tuning of innate immune responses by TREM-1 and TREM-2. The different research topics covered in this thesis are described below and are sche- matically depicted in Figure 1.

In Chapter 2 we investigate the role of TREM-1 during Gram-positive pneumonia by intranasally infecting TREM-1/3 deficient and wild-type mice with serotype 2Streptococ - cus pneumoniae. Subsequently, the early and late host responses were evaluated as well as in vitro leukocyte function. To increase our insight in TREM-1 signaling, in Chapter 3 we investigate the role of DAP12, the signaling adaptor molecule for TREM-receptors, in pneumococcal pneumonia caused by serotype 3 pneumococci. In Chapter 4 another Gram-positive pathogen is used to investigate the role of TREM-1 in pulmonary infec- tion by infecting TREM-1/3 deficient and wild-type mice with MRSA. In Chapter 5 and 6 we set out to dissect the presumptive opposite roles of TREM-1 and TREM-2 in Gram- negative pneumonia derived sepsis caused by Klebsiella pneumoniae or Burkholderia pseudomallei respectively. Since TREM-1 can modulate NOD2 signaling, in Chapter 7 the role of NOD2 is investigated in lower respiratory tract infection caused by differ-

14 General introduction and thesis outline ent strains of S. pneumoniae. In addition to the different infectious models, Chapter 8 describes the role of TREM-1 in a murine model of allergic airway disease triggered by repeated administration of house dust mite. Finally, in Chapter 9, we briefly summarize the results and conclusions presented in this work. 1

15 Chapter 1

References

1. Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med 2013;​369:​840‑51. 2. Adhikari NK, Fowler RA, Bhagwanjee S, Rubenfeld GD. Critical care and the global burden of criti- cal illness in adults. Lancet 2010;​376:​1339‑46. 3. Cohen J, Vincent JL, Adhikari NK, et al. Sepsis: a roadmap for future research. Lancet Infect Dis 2015;​15:​581‑614. 4. Dellinger RP, Levy MM, Rhodes A, et al. Surviving sepsis campaign: international guidelines for management of severe sepsis and septic shock: 2012. Crit Care Med 2013;​41:​580‑637. 5. Opal SM, Dellinger RP, Vincent JL, Masur H, Angus DC. The next generation of sepsis clinical trial designs: what is next after the demise of recombinant human activated protein C?. Crit Care Med 2014;​42:​1714‑21. 6. van der Poll T, Opal SM. Pathogenesis, treatment, and prevention of pneumococcal pneumonia. Lancet 2009;​374:​1543‑56. 7. Podschun R, Ullmann U. Klebsiella spp. as nosocomial pathogens: epidemiology, taxonomy, typ- ing methods, and pathogenicity factors. Clin Microbiol Rev 1998;​11:​589‑603. 8. Wiersinga WJ, Currie BJ, Peacock SJ. Melioidosis. N Engl J Med 2012;​367:​1035‑44. 9. Chambers HF, Deleo FR. Waves of resistance: Staphylococcus aureus in the antibiotic era. Nat Rev Microbiol 2009;​7:​629‑41. 10. David MZ, Daum RS. Community-associated methicillin-resistant Staphylococcus aureus: epide- miology and clinical consequences of an emerging epidemic. Clin Microbiol Rev 2010;​23:​616‑87. 11. Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011;​34:​637‑50. 12. Franchi L, Warner N, Viani K, Nunez G. Function of Nod-like receptors in microbial recognition and host defense. Immunol Rev 2009;​227:​106‑28. 13. Caruso R, Warner N, Inohara N, Nunez G. NOD1 and NOD2: Signaling, Host Defense, and Inflam- matory Disease. Immunity 2014;​41:​898‑908. 14. Klesney-Tait J, Turnbull IR, Colonna M. The TREM receptor family and signal integration. Nat Im- munol 2006;​7:​1266‑73. 15. Bouchon A, Dietrich J, Colonna M. Cutting edge: inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J Immunol 2000;​164:​4991‑5. 16. Netea MG, Azam T, Ferwerda G, Girardin SE, Kim SH, Dinarello CA. Triggering receptor expressed on myeloid cells-1 (TREM-1) amplifies the signals induced by the NACHT-LRR (NLR) pattern recog- nition receptors. J Leukoc Biol 2006;​80:​1454‑61. 17. Bouchon A, Facchetti F, Weigand MA, Colonna M. TREM-1 amplifies inflammation and is a crucial mediator of septic shock. Nature 2001;​410:​1103‑7. 18. El Mezayen R, El Gazzar M, Seeds MC, McCall CE, Dreskin SC, Nicolls MR. Endogenous signals released from necrotic cells augment inflammatory responses to bacterial endotoxin. Immunol Lett 2007;​111:​36‑44. 19. Haselmayer P, Grosse-Hovest L, von Landenberg P, Schild H, Radsak MP. TREM-1 ligand expression on platelets enhances neutrophil activation. Blood 2007;​110:​1029‑35. 20. Read CB, Kuijper JL, Hjorth SA, et al. Cutting Edge: identification of neutrophil PGLYRP1 as a ligand for TREM-1. J Immunol 2015;​194:​1417‑21. 21. Gibot S, Kolopp-Sarda MN, Bene MC, et al. A soluble form of the triggering receptor expressed on myeloid cells-1 modulates the inflammatory response in murine sepsis. J Exp Med 2004;​200:​ 1419‑26.

16 General introduction and thesis outline

22. Gibot S, Cravoisy A, Levy B, Bene MC, Faure G, Bollaert PE. Soluble triggering receptor expressed on myeloid cells and the diagnosis of pneumonia. N Engl J Med 2004;​350:​451‑8. 23. Paloneva J, Manninen T, Christman G, et al. Mutations in two genes encoding different subunits of a receptor signaling complex result in an identical disease phenotype. Am J Hum Genet 2002;​ 1 71:​656‑62. 24. Hamerman JA, Jarjoura JR, Humphrey MB, Nakamura MC, Seaman WE, Lanier LL. Cutting edge: inhibition of TLR and FcR responses in macrophages by triggering receptor expressed on myeloid cells (TREM)-2 and DAP12. J Immunol 2006;​177:​2051‑5. 25. Turnbull IR, Gilfillan S, Cella M, et al. Cutting Edge: TREM-2 Attenuates Macrophage Activation. The Journal of Immunology 2006;​177:​3520‑4. 26. N’Diaye EN, Branda CS, Branda SS, et al. TREM-2 (triggering receptor expressed on myeloid cells 2) is a phagocytic receptor for bacteria. J Cell Biol 2009;​184:​215‑23. 27. Turnbull IR, Colonna M. Activating and inhibitory functions of DAP12. Nat Rev Immunol 2007;​7:​ 155‑61. 28. Chen Q, Zhang K, Jin Y, et al. Triggering Receptor Expressed on Myeloid Cells-2 Protects against Polymicrobial Sepsis by Enhancing Bacterial Clearance. Am J Respir Crit Care Med 2013. 29. Martinez FD, Vercelli D. Asthma. Lancet 2013;​382:​1360‑72. 30. Eder W, Ege MJ, von Mutius E. The asthma epidemic. N Engl J Med 2006;​355:​2226‑35. 31. Lambrecht BN, Hammad H. The immunology of asthma. Nat Immunol 2014;​16:​45‑56. 32. Eisenbarth SC, Piggott DA, Huleatt JW, Visintin I, Herrick CA, Bottomly K. - enhanced, toll-like receptor 4-dependent T helper cell type 2 responses to inhaled antigen. J Exp Med 2002;​196:​1645‑51. 33. Hammad H, Chieppa M, Perros F, Willart MA, Germain RN, Lambrecht BN. House dust mite al- lergen induces asthma via Toll-like receptor 4 triggering of airway structural cells. Nat Med 2009;​ 15:​410‑6. 34. Thomas WR, Hales BJ, Smith WA. House dust mite allergens in asthma and allergy. Trends Mol Med 2010;​16:​321‑8.

17

Triggering Receptor Expressed on Myeloid Cells (TREM)-1 improves host defence in pneumococcal pneumonia

Tijmen J. Hommes1,2, Arie J. Hoogendijk1,2, Mark C. Dessing3, Cornelis van ’t Veer1,2, Sandrine Florquin3, Marco Colonna4, Alex F. de Vos1,2, Tom van der Poll1,2,5

2 Academic Medical Center, University of Amsterdam, the Netherlands: 1Center for Experimental and Molecular Medicine 2Center for Infection and Immunity 3Department of Pathology 5Division of Infectious Diseases 4Washington University in St. Louis, St. Louis, Missouri, USA: Department of Pathology

Journal of Pathology. 2014 Aug;233(4):357-67 Chapter 2

Abstract

Streptococcus (S.) pneumoniae is a common Gram-positive pathogen in community-ac- quired pneumonia and sepsis. Triggering Receptor Expressed on Myeloid cells-1 (TREM- 1) is a receptor on phagocytes known to amplify inflammatory responses. Previous studies showed that TREM-1 inhibition protects against lethality during experimental Gram-negative sepsis. We here aimed to investigate the role of TREM-1 in an experi- mental model of pneumococcal pneumonia using TREM-1/3 deficient (Trem-1/3−/−) and wild type (Wt) mice. Additionally ex vivo responsiveness of Trem-1/3−/− neutrophils and macrophages was examined. S. pneumoniae infection resulted in a rapid recruitment of TREM-1 positive neutrophils into the bronchoalveolar space while high constitutive TREM-1 expression on alveolar macrophages remained unchanged. TREM-1/3 defi- ciency led to increased lethality accompanied by enhanced growth of S. pneumoniae at the primary site of infection and increased dissemination to distant organs. Within the first 3-6 hours of infection Trem-1/3−/− mice demonstrated a strongly impaired in- nate immune response in the airways, as reflected by reduced local release of cytokines and chemokines and a delayed influx of neutrophils. Trem-1/3−/− alveolar macrophages produced less cytokines upon exposure to S. pneumoniae in vitro and were less capable of phagocytosing this pathogen. TREM-1/3 deficiency did not influence neutrophil re- sponsiveness to S. pneumoniae. These results identify TREM-1 as key player in protective innate immunity during pneumococcal pneumonia most likely by enhancing the early immune response of alveolar macrophages.

20 TREM-1 improves host defence in pneumococcal pneumonia

Introduction

Infections with Streptococcus (S.) pneumoniae are a major burden for public health in the world1. The majority of cases of community-acquired pneumonia (CAP) is caused by this Gram-positive pathogen2. CAP is a common cause of sepsis, leading to 10 mil- lion deaths annually3. With the growing incidence of antibiotic resistance knowledge of mechanisms underlying host-pathogen interactions in pneumococcal pneumonia is increasingly important4. 2 The host response to pathogens is initiated by the recognition of pathogen associated molecular patterns (PAMPs) via pattern recognition receptors such as Toll-like receptors (TLRs) and NOD-like Receptors (NLRs)5. Triggering Receptor Expressed on Myeloid cells- 1 (TREM-1) is a membrane bound receptor expressed on neutrophils, monocytes and macrophages, that also exists as a soluble form (sTREM-1) when cleaved from the cell membrane6,7. Cell-associated TREM-1 is upregulated in the presence of bacteria and microbial ligands6. By binding of a currently unknown ligand TREM-1 is able to amplify TLR- and NLR-signaling6,8. TREM-1 associates with the adaptor molecule DNAX activating protein of 12kDa (DAP12) leading to downstream activation of MAP kinases and NFκB translocation, resulting in pro-inflammatory cytokine- and chemokine production6. In fulminant sepsis exaggerated inflammation may contribute to collateral damage of tissues9. Since TREM-1 is an amplifier of inflammation, this receptor has been considered a therapeutic target for severe sepsis10. Indeed, in models of lethal sepsis associated with acute severe inflammation different strategies to block TREM-1 have been shown to improve outcome. Inhibition of TREM-1 signaling increased survival in several models of abdominal sepsis11-13. Likewise, in acute pneumonia caused by Pseudomonas (P.) ae- ruginosa antagonizing TREM-1 signaling attenuated lung inflammation and injury and reduced lethality14. In contrast, however, a very recent study has suggested a role for TREM-1 in protective immunity during P. aeruginosa pneumonia in mice15. Thus far, the role of TREM-1 in Gram-positive infection is largely unknown. Here, we used the recently generated TREM-1/3 deficient (Trem-1/3−/−) mice15 to determine the contribution of TREM-1 to the host response during respiratory tract infection caused by S. pneumoniae.

Methods

Detailed methods are provided in the supplement.

21 Chapter 2

Mice Pathogen-free 8- to 10-week old male wild type (Wt) C57BL/6 mice were purchased from Charles River (Maastricht, The Netherlands). Trem-1/3−/− mice15 were backcrossed > 97% to a C57BL/6 genetic background. All experiments were approved by the Animal Care and Use Committee of the Academic Medical Center, Amsterdam, the Netherlands.

Experimental design Pneumonia was induced by intranasal inoculation with 2 × 107 colony forming units (CFU) serotype 2 S. pneumoniae (strain D39). Mice were euthanized at predefined time points (n = 7-8 per group) or followed for 12 days after infection (n = 15 per group). Col- lection and handling of samples were done as described16-18. In separate experiments BALF was collected (3 and 6 hours time points only).

TREM-1 expression TREM-1 expression on lung and blood leukocytes was evaluated using flow cytometry in essence as described19. Wild type mice were infected with S. pneumoniae and terminated before or after 6, 24 or 48 hours after infection. Blood and broncheoalveolar lavage fluid (BALF) were collected and leukocyte TREM-1 expression was determined using FACS.

Assays Tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, Chemokine (C-X-C motif) ligand 1, 2 and 5, TREM-1 (all R&D systems, Minneapolis, MN) and myeloperoxidase (MPO; Hycult Biotechnology BV, Uden, the Netherlands) were measured using specific ELISAs according to manufacturers’ recommendations.

Histology After embedding, lungs were stained with haematoxylin and eosin to score inflamma- tion or stained and quantified for Ly-6G using immunohistochemical procedures as described16,18.

Neutrophil activation Bone marrow neutrophils from Trem-1/3−/− and Wt mice were isolated as described20. Neutrophils (1 × 105/well) were incubated with medium, heat killed S. pneumoniae (1 × 107/ml), or phorbol myristate acetate (PMA, 10ng/ml) for 1 hour, washed, stained for Ly- 6G, CD11b and CD62L (all BD Pharming) and analyzed by FACS in essence as described elsewhere20.

22 TREM-1 improves host defence in pneumococcal pneumonia

Phagocytosis Phagocytosis of S. pneumoniae by blood neutrophils, bone marrow derived macrophages (BMDMs; prepared as described21) and alveolar macrophages (obtained as described22) from Trem-1/3−/− and Wt mice was done in essence as previously described18,23. Briefly, whole blood, BMDMs and alveolar macrophages were incubated with FITC or CFSE labeled S. pneumoniae and internalization was measured by FACS.

Macrophage stimulation 2 BMDMs and alveolar macrophages from Trem-1/3−/− and Wt mice were stimulated for 6 or 24 hours with or without the indicated multiplicity of infection (MOI) of heat-killed S. pneumoniae and supernatants were taken and stored at −20°C until assayed for TNF-α and CXCL1.

Statistical analysis Values are expressed as scatter plots with medians or as mean ± SEM. Serial changes in TREM-1 expression in time were analyzed by Kruskal-Wallis test with the Dunn post-hoc test. Differences between groups were analyzed by Mann-Whitney U test. For survival analysis, Kaplan-Meier analysis followed by log-rank test was performed. Analyses were performed using GraphPad Prism version 5.0, GraphPad Software (San Diego, CA). Values of P less than 0.05 were considered statistically significant.

Results

TREM-1 levels increase during pneumococcal pneumonia To obtain insight into TREM-1 expression Wt mice were intranasally inoculated with S. pneumoniae and sTREM-1 levels were measured in BALF and lung homogenates before and after infection. sTREM-1 was detectable in BALF of uninfected mice; following in- fection BALF sTREM-1 concentrations increased modestly but significantly, peaking at 24 hours (Figure 1A). Total lung sTREM-1 levels were significantly elevated compared with baseline values at 6 and 24 hours after infection (Figure 1B; P < 0.001 and P < 0.01 respectively). Using FACS-analysis, we observed a gradual increase in TREM-1 expression on neu- trophils recovered from BALF reaching peak values at 48 hours (Figure 1C; P < 0.001). Alveolar macrophages from uninfected mice showed high constitutive TREM-1 expres- sion which did not significantly change after infection (Figure 1D). In blood, TREM-1 expression increased on both neutrophils (Figure 1E; P < 0.01) and monocytes (Figure 1F; P < 0.01) following infection with S. pneumoniae.

23 Chapter 2

A B BALF 1000 25 Lung homogenates F g L *** * n A 800

u 20 l B

l l m m / ** / 600 15 g g n p

1 1 - - 400 10 M M E E R R 200 5 T s T s 0 0 0h 6h 24h 48h 0h 6h 24h 48h

C D Alveolar macrophages 150 BALF granulocytes 300 ** I I F 100 F 200 M M

1 1 - - M M E E R 50 ** R 100 T ** T ND 0 0 0h 6h 24h 48h 0h 6h 24h 48h

E F Blood granulocytes 200 150 Blood monocytes ** I 150 I F F 100 M M

** 1 1 - 100 - M M E E R R 50 T 50 T

0 0 0h 6h 24h 48h 0h 6h 24h 48h

Figure 1: Increased expression of TREM-1 pneumococcal pneumonia. (s)TREM-1 levels in bronchoal- veolar lavage fluid (BALF) (A), whole lung homogenates (B) and cell-surface TREM-1 expression on relevant lung and blood cells was determined in naive mice and 6, 24 and 48 hours after intranasal inoculation with 2 × 107 colony forming units (CFU) Streptococcus (S.) pneumoniae. TREM-1 cell-surface expression is depict- ed over time on neutrophils and macrophages/monocytes in BALF and whole blood. Data are mean ± SEM values from 6-8 mice per group at each time point (one experiment per time point). Asterisks represent sta- tistical significance for the difference with t = 0 (by Dunn post-hoc test) after testing for the overall response (Kruskal-Wallis test). MFI, mean fluorescence intensity; ND, not determined (not enough garnulocytes for adequate flow cytometry). *P < 0.05, **P < 0.01, and ***P < 0.001 versus t = 0 h.

24 TREM-1 improves host defence in pneumococcal pneumonia

TREM-1/3 deficiency impairs host defense during pneumococcal pneumonia To investigate the functional role of TREM-1 during pneumococcal pneumonia, Trem- 1/3−/− and Wt mice were infected intranasally with S. pneumoniae and observed during 12 days. While only 1 of 15 Wt mice died, 8 of 15 Trem-1/3−/− mice deceased (Figure 2A; P < 0.001), indicating an essential protective role for TREM-1. To substantiate this finding, in a separate experiment we determined bacterial outgrowth in lungs, blood, spleen and liver 24 and 48 hours after infection. Bacterial loads were markedly higher in lungs of Trem-1/3−/− mice at these time points (Figure 2B; 2 P < 0.001 and P < 0.01 respectively). Remarkably, Trem-1/3−/− mice displayed significantly enhanced dissemination of the infection reflected by the fact that S. pneumoniae could be cultured from the blood of 6 of 8 Trem-1/3−/− mice, compared with 0 of 8 Wt mice after 24 hours, and 5 of 8 Trem-1/3−/− mice compared with 2 of 8 Wt mice after 48 hours (Figure 2C; P < 0.01 and P = 0.12 respectively). Increased dissemination in Trem-1/3−/− mice was

A B 100 8

g 7

l * n

a 80 u v l 6 i

l v r

*** m 5 *** / u 60 s U

t F 4 n C

e 40 3 g c r o l e Wt 2 P 20 0 Trem-1/3 -/- 1 1 0 0 0 2 4 6 8 10 12 14 24h 48h Days C D 8 8 n d 7 7 e o e l o l

6 p 6 b s

l ** 5 l 5 m m /

** / U

4 U 4

F ** F C C

3 3 g g o o l l

2 2

0 0 1 1 1 1 0 0 24h 48h 24h 48h Figure 2: TREM-1/3 deficiency increases mortality and bacterial growth and dissemination during pneumococcal pneumonia. Wildtype (Wt) and Trem-1/3−/− mice (n = 15 per group) were intranasally in- fected with 2 × 107 colony forming units (CFU) Streptococcus (S.) pneumoniae and survival was monitored for 12 days (A). In separate experiments Wt and Trem-1/3−/− mice (n = 7-8 per group) were intranasally in- fected with 2 × 107 CFU S. pneumoniae and sacrificed 24 and 48 hours later. Bacterial loads were determined in lung homogenates (B), blood (C) and spleen (D). Horizontal lines represent medians; data are from one experiment for each time point and for survival; * P < 0.05, ** P < 0.01, *** P < 0.001 versus Wt mice.

25 Chapter 2 also reflected by higher bacterial loads in spleen after 24 and 48 hours (Figure 2D; both P < 0.01) and liver after 48 hours (data not shown; P < 0.01). Together these data suggest that TREM-1/3 protect against lethality during pneumo- coccal pneumonia by facilitating bacterial clearance and limiting dissemination of the infection.

Impact of TREM-1/3 deficiency on lung inflammation during late stage pneumococcal pneumonia Since TREM-1 is an amplifier of inflammation and promotes neutrophil migration15, we assessed pulmonary histopathology and determined the extent of neutrophil influx into

A B C 20 Wt Trem-1/3 -/- e r 15 o c s

y

g 10 o l o h t

a 5 P -/- Wt (24h) Trem-1/3 (24h) 0 DD E F 24h 20 * e r 15 o c s

y

g 10 o l o h t

a 5 P

-/- Wt (48h) Trem-1/3 (48h) 0 G H 48h 3 50 ) % ( 40 a l e r

2 m a /

g 30 g n n

u O l

P 20 +

1 M G 6

- 10 y L 0 0 24h 48h 24h 48h Figure 3: TREM-1/3 deficiency aggravates lung pathology during late stage pneumococcal pneumo- nia. Wildtype (Wt) and Trem-1/3−/− mice (7-8 per group) were intranasally inoculated with 2 × 107 colony forming units (CFU) Streptococcus (S.) pneumoniae and sacrificed 24 and 48 hours later. Representative he- matoxylin and eosin (HE) stainings of lung tissue of Wt (A, D) and Trem-1/3−/− (B, E) 24 and 48 hours after inoculation with S. pneumoniae (original magnification ×200). Panels C and F show quantitative lung pa- thology score (means ± SEM) 24 and 48 hours after infection as determined by the scoring system listed in the online methods section. Neutrophil influx, as measured by Ly-6G positivity of histopathological slides (G) and pulmonary MPO levels (H) did not differ between Wt and Trem-1/3−/− mice at either 24 or 48 hours after infection; data are from one experiment for each time point. *P < 0.05 versus Wt mice.

26 TREM-1 improves host defence in pneumococcal pneumonia the lungs of Trem-1/3−/− and Wt mice. Notably, after 48 hours Trem-1/3−/− mice showed exaggerated lung pathology with enhanced interstitial inflammation, endothelialitis and larger surfaces of confluent inflammation infiltrates (Figure 3A-F; P < 0.05). TREM-1/3 deficiency did not impact on neutrophil recruitment as reflected by similar numbers of Ly6G positive cells in lung tissue (Figure 3G and Supplementary Figure S1) and similar MPO concentrations in whole lung homogenates (Figure 3H) prepared from Trem-1/3−/− and Wt mice. Next we measured cytokine (TNF-α, IL-1β and IL-6) and CXCL1 levels in whole lung 2 homogenates after induction of pneumonia. Only CXLC1 levels differed between strains, being lower in Trem-1/3−/− mice at 24 hours (Table 1; P < 0.01).

Table 1: Cytokine and chemokine concentrations in lung homogenates of Wildtype and Trem-1/3−/− mice during pneumococcal pneumonia. 24h 48h Wild type Trem-1/3−/− Wild type Trem-1/3−/− TNF-α [pg/mL] 2510±96 2301±126 325±28 375±38 IL-1β [pg/mL] 7046±623 7092±731 988±40 1511±150 IL-6 [pg/mL] 8324±1430 5289±457 862±52 1031±58 CXCL1 [ng/mL] 10.6±0.7 6.8±0.7** 6.3±0.4 8.0±1.1 Data are expressed as means ± SEM of n = 7-8 mice per group per time point, CXCL Chemokine (C-X-C motif) ligand, IL interleukin, TNF-α tumor necrosis factor-α, Trem-1/3−/− Triggering Receptor Expressed on Myeloid Cells-1/3 deficient, **P < 0.01 versus wild type mice (Mann-Whitney U test).

TREM-1/3 impacts on early host response during pneumococcal pneumonia In order to elucidate the mechanism underlying the impaired host defense in the ab- sence of TREM-1/3, we set out to investigate the early immune response (3 and 6 hours after infection) to S. pneumoniae. To this end we inoculated Trem-1/3−/− and Wt mice with S. pneumoniae and collected BALF, lungs and blood. No differences in bacterial loads between Trem-1/3−/− and Wt mice were found at either time point (Figure 4A-C). While at 3 hours after infection total leukocyte numbers were similar in BALF harvested from Trem-1/3−/− and Wt mice, at 6 hours BALF leukocyte counts were lower in the for- mer group (Figure 4D; P < 0.01). Already at 3 hours post infection Trem-1/3−/− mice had reduced neutrophil numbers in BALF when compared with Wt mice and this attenuated neutrophil influx in the absence of TREM-1/3 was still present at 6 hours (Figure 4E; both P < 0.05). Accordingly, MPO concentrations in whole lung homogenates (Figure 4F) and the number of Ly6G positive cells in lung tissue were reduced in Trem-1/3−/− relative to Wt mice (Figure 4G-I; both P < 0.05). The number of alveolar macrophages in BALF did not differ between strains at these time points (supplemental Figure S2). Together these data suggest that TREM-1/3 deficiency impairs neutrophil recruitment to lung

27 Chapter 2

Figure 4: TREM-1/3 deficiency leads to an attenuated early host response during pneumococcal pneumonia. Wildtype (Wt) and Trem-1/3−/− mice (n = 7-8 per group) were intranasally infected with 2 × 107 colony forming units (CFU) Streptococcus (S.) pneumoniae and sacrificed 3 and 6 hours later. Bacterial loads were determined in lung homogenates (A), bronchoalveolar lavage fluid (BALF; B) and blood (C). Total number of leukocytes (D) and neutrophils (E) in BALF of Wt and Trem-1/3−/− mice. Panels G (Wt) and H (Trem-1/3−/−) show Ly6G staining of histopathological slides (original magnification ×200) at 6 hours after infection. Neutrophil influx, as measured by Ly-6G positivity (I), and pulmonary MPO levels (J). Levels of Tumor Necrosis Factor (TNF)-α (J), Chemokine (C-X-C motif) ligand 1 (CXCL1) (K), CXCL2 (L) and CXCL5 (M). Data are representive for two experiments and mean ± SEM are shown; *P < 0.05; **P < 0.01; ***P < 0.001. tissue and the bronchoalveolar space early after infection with S. pneumoniae via the airways. We next measured levels of TNF-α, CXCL1, CXCL2 and CXCL5 in BALF, and TNF-α, CXCL1 and 2 in whole lung homogenates. TNF-α levels were impressively lower in both BALF and lung homogenates collected from Trem-1/3−/− mice 3 and 6 hours after infection (Figure 4J, and supplementary Figure S3A; P < 0.001). Similarly, BALF CXCL1 and CXCL5 concentrations were lower 6 hours after infection (Figure 4K and M; P < 0.01 and P < 0.05 respectively) and CXCL2 levels 3 and 6 hours after infection (Figure 4L; both P < 0.05) in Trem-1/3−/− mice. Whole lung CXCL1 was lower in Trem-1/3−/− mice at 3 and 6 hours

28 TREM-1 improves host defence in pneumococcal pneumonia

(both P < 0.01); no differences were found in whole lung CXCL2 early during infection (supplementary Figure S3B). These data demonstrate the importance of TREM-1/3 for local cytokine and chemokine production in response to pulmonary infection with S. pneumoniae.

TREM-1/3 deficiency does not influence neutrophil activation or phagocytosis Having established that TREM-1/3 plays an important role in early neutrophil recruit- ment, we next tested the capacity of Trem-1/3−/− neutrophils to respond to S. pneu- 2 moniae. First, we exposed purified Trem-1/3−/− and Wt neutrophils to S. pneumoniae or (as an aspecific stimulus) PMA in vitro for one hour and determined the expression of CD11b (expected to become upregulated upon activation) and CD62L (downregulated

A medium S.pneu PMA

80% 2% 80% 2% 21% 12% 0% 1%

Wt

L 12% 6% 5% 62% 1% 98%

78% 2% 20% 5% 0% 1% CD62

-/- Trem-1/3

12% 8% 13% 62% 4% 95%

CD11b B C 2000 Wt 150 Trem-1/3 -/- 1500 L b

2 100 1 6 1 D D C

C 1000

I I F F M

M 50 500

0 0 M S.pneu PMA M S.pneu PMA D 30000 x e d n i 20000 s i s o t y c

o 10000 g a h P

0 4°C 37°C Figure 5: TREM-1/3 deficiency does not impact on neutrophil function. Bone marrow derived neutro- phils from wildtype (Wt) and Trem-1/3−/− mice were incubated with medium, heat killed Streptococcus (S.) pneumoniae or phorbol 12-myristate 13-acetate (PMA) for 1 hour and CD11b and CD62L expression was analysed by flow cytometry to evaluate cellular activation (A). Mean fluorescence index was calculated for both CD11b and CD62L (B-C). Whole blood from Wt and Trem-1/3−/− mice was incubated with carboxyfluo- rescein succinimidyl ester (CFSE) labeled viable S. pneumoniae for 1 hour and the degree of phagocytosis of blood neutrophils was assessed by flow cytometry (D). Data are representative for 2 independent experi- ments and are expressed as mean ± SEM.

29 Chapter 2 upon activation) (Figure 5A-C). Both S. pneumoniae and PMA enhanced CD11b and re- duced CD62L expression on Trem-1/3−/− and Wt neutrophils, and responses were similar in both genotypes. Second, we tested another major effector function of neutrophils during infection, phagocytosis. We found no differences between Trem-1/3−/− and Wt neutrophils with regard to their capacity to internalize S. pneumoniae (Figure 5D). In our hands mouse neutrophils are unable to kill S. pneumoniae D39 in vitro. Therefore, to obtain insight in the potential role of TREM-1/3 in neutrophil mediated killing of S. pneu- moniae, we sorted TREM-1 high and TREM-1 low neutrophils from human blood and determined their capacity to kill S. pneumoniae. No differences were observed between human TREM-1 high and TREM-1 low neutrophils with regard to their capacity to kill S. pneumoniae (supplemental Figure S4).

Trem-1/3−/− macrophages show a reduced responsiveness to pneumococci and are less capable of phagocytosing S. pneumoniae Since we found lower levels of cytokines and chemokines in BALF from Trem-1/3−/− mice early after infection we hypothesized that macrophages could be hampered in their ability to secrete these mediators upon stimulation with S. pneumoniae. To test this we generated BMDMs from both Trem-1/3−/− and Wt mice, incubated these with S. pneumoniae for 6 or 24 hours and measured TNF-α and CXCL1 in supernatants by ELISA. Trem-1/3−/− BMDMs produced significantly less TNF-α and CXCL1 after 6 and 24 hours of incubation with S. pneumoniae (Figure 6A,C; both P < 0.01). We confirmed these findings using purified alveolar macrophages: similar to BMDMs, alveolar macrophages from Trem-1/3−/− mice produced lower levels of TNF-α and CXCL1 when exposed to S. pneumoniae for 6 or 24 hours (Figure 6B,D; P < 0.01). Phagocytosis represents an essential task of macrophages in host defense against in- vading pathogens24. To examine a possible role for TREM-1/3 herein we incubated Trem- 1/3−/− and Wt BMDMs or alveolar macrophages with viable S. pneumoniae and analysed phagocytosis by FACS. Trem-1/3−/− BMDMs and alveolar macrophages were reduced in their capacity to internalize S. pneumoniae (Figure 6E,F; P < 0.01).

Discussion

TREM-1 has been implicated as an amplifier of the innate immune response by virtue of its capacity to enhance TLR and NLR signaling upon recognition of microbial com- ponents. Previous studies examined the role of TREM-1 in host defense against severe Gram-negative infection11-13,15. To the best of our knowledge, we here for the first time report on the role of TREM-1 in Gram-positive pneumonia. We found that S. pneumoniae lung infection resulted in local sTREM-1 release and recruitment of TREM-1 express-

30 TREM-1 improves host defence in pneumococcal pneumonia

A B BMDM AM 3000 Wt 4000 Trem-1/3 -/- l

l ** 3000 2000 m m 1000 ** pg/ pg/ 800 2000 - - F F 600 N N T T 400 * 1000 200 * * 0 0 2 M 1:10 1:100 M 1:10 1:100 M 1:50 M 1:50 1:250 6h 24h 6h 24h C D 8000 800

6000 l l 3000 600 m m pg/ pg/ 1 1 2000 400 L L * C C X X C C 1000 * 200 ** ** ** 0 0 M 1:10 1:100 M 1:10 1:100 M 1:50 M 1:50 1:250 6h 24h 6h 24h E F 6000 2000 x x e 5000 e * 1500 nd nd i 4000 i * s s i i s s o o t 3000 t 1000 cy cy 2000 go go a a 500 h h P 1000 P

0 0 4°C 37°C 4°C 37°C Figure 6: TREM-1/3 deficiency results in lower cytokine and chemokine production and phagocytosis by macrophages in vitro. Bone marrow derived macrophages (BMDMs) and alveolar macrophages (AMs) from wildtype (Wt) and Trem-1/3−/− mice were incubated with medium (M) or heat killed Streptococcus (S). pneumoniae (MOI 1:10 and 1:100) for 6 and 24 hours and levels of Tumor Necrosis Factor (TNF)-α (A-B), Chemokine (C-X-C motif) ligand 1 (C-D) in supernatants were measured. Representative figures of three independent experiments are shown (n = 4-8). BMDMs (E) and AMs (F) from Wt and Trem-1/3−/− mice were incubated with heat killed fluorescein isothiocyanate (FITC) labeledS. pneumoniae (MOI 1:100) for 1 hour at 4°C and 37°C and phagocytosis was assessed by flow cytometry after quenching. Data are representative for 2-3 independent experiments and are expressed as mean ± SEM *P < 0.05; **P < 0.01. ing neutrophils, while high constitutive TREM-1 expression by alveolar macrophages remained unaltered. Importantly, TREM-1/3 deficiency led to increased mortality, most likely as a consequence of enhanced bacterial growth and dissemination caused by a strongly impaired early innate immune response in the airways. Our results suggest that

31 Chapter 2 the beneficial role of TREM-1/3 results from its expression by alveolar macrophages and its involvement in phagocytosis of S. pneumoniae and cytokine/chemokine production by these resident lung cells, thereby facilitating the recruitment of neutrophils to the site of infection and the clearance of bacteria. Increased levels of sTREM-1 have been detected in plasma of patients with pneumo- coccal pneumonia25. In addition, mice with respiratory tract infection caused by S. pneu- moniae demonstrated increased levels of TREM-1 protein in whole lung homogenates25. Our current data expand these earlier results: we demonstrate constitutive TREM-1 expression on alveolar macrophages and enhanced expression of TREM-1 on recruited neutrophils after infection with S. pneumoniae. Our laboratory previously reported similar findings with regard to cell-associated TREM-1 expression in the bronchoal- veolar space of mice during severe Gram-negative pneumonia derived sepsis caused by Burkholderia pseudomallei19. Other studies have confirmed the constitutive expression of TREM-1 on human alveolar macrophages and the increased TREM-1 expression on human blood neutrophils during severe infection6,11,19,26. As a model for TREM-1 deficiency we used mice deficient for both TREM-1 and TREM-3. In mice, TREM-3 is highly homologous to TREM-1 and both receptors have a similar cel- lular distribution. Like TREM-1, TREM-3 is an activating receptor on mouse macrophages signaling through DAP1227. In contrast, in humans Trem3 is a pseudogene. Thus to mimic the human situation, we used Trem-1/3−/− mice to obtain insight into the role of TREM-1 in pneumococcal pneumonia. The same approach was used in a recent study investigat- ing the contribution of TREM-1 to the host response during P. aeruginosa pneumonia15. While this earlier investigation also reported an impaired host defense of Trem-1/3−/− mice after infection with P. aeruginosa via the airways, as reflected by increased bacterial loads and mortality when compared with Wt mice15, the underlying mechanisms clearly differed from our current findings during pneumococcal pneumonia. Indeed, during Pseudomonas pneumonia Trem-1/3−/− mice showed an attenuated influx of neutrophils in BALF in spite of increased neutrophil numbers in lung tissue, suggestive for a role of TREM-1 in transepithelial migration of neutrophils15. In contrast, we here found a reduced influx of neutrophils in both lung tissue and BALF of Trem-1/3−/− mice early after infection with S. pneumoniae. During Pseudomonas pneumonia Trem-1/3−/− mice displayed higher cytokine and chemokine concentrations in BALF and lungs than Wt mice; however, measurements were done 14 hours after infection, when Trem-1/3−/− mice had much higher bacterial loads in the pulmonary compartment providing a much more potent stimulus for inflammatory mediator production15. We focused on early time points after infection (3 and 6 hours), when bacterial burdens were still similar in both mouse strains and showed strongly reduced levels of TNF-α, CXCL1, CXCL2 and CXCL5. In accordance, Trem-1/3−/− BMDMs and alveolar macrophages released less TNF-α and CXCL1 upon exposure to S. pneumoniae in vitro, which is in agreement with earlier re-

32 TREM-1 improves host defence in pneumococcal pneumonia ports showing enhanced cytokine production by leukocytes in response to TLR and NLR ligands when pre-incubated with an agonistic TREM-1 antibody6,8,25. To gain more insight into TREM-1 signaling during pneumococcal pneumonia it would be of great value to assess which downstream pathways are implicated upon ligation of this receptor. It is know both Erk1/2 and NFB pathways are involved in human and murine leukocytes6,11. Further studies, in particular in primary alveolar macrophages, are required to unravel the precise mechanism by which TREM-1 influences cytokine production in the lower respiratory tract infected with S. pneumoniae. Nonetheless, our results suggests that 2 during pneumococcal pneumonia the reduced responsiveness of Trem-1/3−/− alveolar macrophages played a pivotal role in the diminished release of cytokines and chemo- kines in the bronchoalveolar space early after infection, subsequently resulting in an attenuated neutrophil recruitment to the site of the infection and an impaired clearance of pneumococci. Notably, especially TNF-α levels were dramatically lower in BALF and lung homogenates from Trem-1/3−/− mice. Since TNF-α is a key proinflammatory cytokine in protective immunity during airway infections with S. pneumoniae28,29, reduced TNF-α release likely contributed to the enhanced susceptibility of Trem-1/3−/− mice. Host de- fense in Trem-1/3−/− mice was further impaired as a consequence of the reduced capacity of Trem-1/3−/− macrophages to internalize S. pneumoniae. Ligation of TREM-1 by agonistic antibodies has been shown to affect degranulation and phagocytosis of neutrophils6,11,30. We here followed the opposite approach by ex- amining the impact of TREM-1/3 deficiency on neutrophil activation by S. pneumoniae and the aspecific stimulus PMA. Our data revealed unaltered neutrophil responsiveness and phagocytosis in the absence of TREM-1/3. In accordance, Trem-1/3−/− neutrophils showed intact chemotaxis and an unaltered capacity to phagocytose P. aeruginosa15. Together these data argue against a role for TREM-1/3 in activation of neutrophil func- tions important for antibacterial defense. An earlier investigation reported that administration of an agonistic TREM-1 antibody accelerated bacterial clearance and improved survival during murine pneumococcal pneumonia25. Notably, while this previous study suggests that exogenous stimulation of TREM-1 may be beneficial for host defense duringS. pneumoniae pneumonia, it does not provide insight into the role of endogenous TREM-1 in this infection25. Indeed, ligands for TREM-1 have not been identified and thus far it is unclear how TREM-1 signaling is triggered during infection. Pneumolysin is a pneumococcal derived pore-forming toxin responsible for much of the inflammatory response to infection with S. pneumoniae31. Pneumolysin may acti- vate immune cells via either TLR432 or NOD233, both receptors of which signaling can be amplified by TREM-16,8. As such, one could speculate that pneumolysin has a role in the TREM-1 dependent cytokine response and thereby in TREM-1 mediated protection during pneumococcal pneumonia. It would be interesting to address this issue in future

33 Chapter 2 research by using a genetically modified strain of the serotype 2 S. pneumoniae (D39) lacking pneumolysin. TREM-1 has been implicated as a therapeutic target in severe bacterial infection and sepsis in light of strong protective effects exerted by TREM-1 inhibition in various models of fulminant Gram-negative sepsis10. In contrast, our current data suggest a protective role for TREM-1 in a model of CAP induced by the common Gram-positive respiratory pathogen S. pneumoniae. These results illustrate the complex nature of innate immunity, which in the initial phase of localized infection is essential for an adequate response to invading bacteria but during exaggerated inflammation elicited by high bacterial loads can contribute to collateral damage and tissue injury.

Acknowledgements

The authors thank Marieke ten Brink and Joost Daalhuisen for their expert technical assistance during the animal experiments, Daan de Boer and Ahmed Achouiti for their assistance in processing the experimental samples and Regina de Beer for performing histopathological and immunohistochemical stainings. This work was supported by a PhD Scholarship from the AMC Graduate School, Uni- versity of Amsterdam, the Netherlands. The funder had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript

34 TREM-1 improves host defence in pneumococcal pneumonia

References

1. van der Poll T, Opal SM. Pathogenesis, treatment, and prevention of pneumococcal pneumonia. Lancet 2009;​374:​1543‑56. 2. Mandell LA, Wunderink RG, Anzueto A, et al. Infectious Diseases Society of America/American Thoracic Society consensus guidelines on the management of community-acquired pneumonia in adults. Clin Infect Dis 2007;​44 Suppl 2:​S27‑72. 3. Bartlett JG, Dowell SF, Mandell LA, File Jr TM, Musher DM, Fine MJ. Practice guidelines for the man- agement of community-acquired pneumonia in adults. Infectious Diseases Society of America. 2 Clin Infect Dis 2000;​31:​347‑82. 4. Feldman C. Clinical relevance of in the management of pneumococcal community-acquired pneumonia. J Lab Clin Med 2004;​143:​269‑83. 5. Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011;​34:​637‑50. 6. Bouchon A, Dietrich J, Colonna M. Cutting edge: inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J Immunol 2000;​164:​4991‑5. 7. Gibot S, Kolopp-Sarda MN, Bene MC, et al. A soluble form of the triggering receptor expressed on myeloid cells-1 modulates the inflammatory response in murine sepsis. J Exp Med 2004;​200:​ 1419‑26. 8. Netea MG, Azam T, Ferwerda G, Girardin SE, Kim SH, Dinarello CA. Triggering receptor expressed on myeloid cells-1 (TREM-1) amplifies the signals induced by the NACHT-LRR (NLR) pattern recog- nition receptors. J Leukoc Biol 2006;​80:​1454‑61. 9. Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med 2013;​369:​840‑51. 10. Derive M, Massin F, Gibot S. Triggering receptor expressed on myeloid cells-1 as a new therapeutic target during inflammatory diseases. Self Nonself 2010;​1:​225‑30. 11. Bouchon A, Facchetti F, Weigand MA, Colonna M. TREM-1 amplifies inflammation and is a crucial mediator of septic shock. Nature 2001;​410:​1103‑7. 12. Gibot S, Buonsanti C, Massin F, et al. Modulation of the triggering receptor expressed on the myeloid cell type 1 pathway in murine septic shock. Infect Immun 2006;​74:​2823‑30. 13. Gibot S, Massin F, Marcou M, et al. TREM-1 promotes survival during septic shock in mice. Eur J Immunol 2007;​37:​456‑66. 14. Gibot S, Alauzet C, Massin F, et al. Modulation of the triggering receptor expressed on myeloid cells-1 pathway during pneumonia in rats. J Infect Dis 2006;​194:​975‑83. 15. Klesney-Tait J, Keck K, Li X, et al. Transepithelial migration of neutrophils into the lung requires TREM-1. J Clin Invest 2013;​123:​138‑49. 16. Rijneveld AW, Weijer S, Florquin S, et al. 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 2004;​103:​1702‑9. 17. Dessing MC, Florquin S, Paton JC, van der Poll T. Toll-like receptor 2 contributes to antibacterial defence against pneumolysin-deficient pneumococci. Cell Microbiol 2008;​10:​237‑46. 18. Achouiti A, Vogl T, Urban CF, et al. Myeloid-related protein-14 contributes to protective immunity in gram-negative pneumonia derived sepsis. PLoS Pathog 2012;​8:​e1002987. 19. Wiersinga WJ, Veer Cvt, Wieland CW, et al. Expression Profile and Function of Triggering Recep- tor Expressed on Myeloid Cells-1 during Melioidosis. Journal of Infectious Diseases 2007;​196:​ 1707‑16.

35 Chapter 2

20. Van Ziffle JA, Lowell CA. Neutrophil-specific deletion of Syk kinase results in reduced host defense to bacterial infection. Blood 2009;​114:​4871‑82. 21. Eske K, Breitbach K, Kohler J, Wongprompitak P, Steinmetz I. Generation of murine bone marrow derived macrophages in a standardised serum-free cell culture system. J Immunol Methods 2009;​ 342:​13‑9. 22. Knapp S, Leemans JC, Florquin S, et al. Alveolar Macrophages Have a Protective Antiinflammatory Role during Murine Pneumococcal Pneumonia. Am J Respir Crit Care Med 2003;​167:​171‑9. 23. Weijer S, Sewnath ME, de Vos AF, et al. Interleukin-18 facilitates the early antimicrobial host response to Escherichia coli peritonitis. Infect Immun 2003;​71:​5488‑97. 24. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Im- munol 2011;​11:​723‑37. 25. Lagler H, Sharif O, Haslinger I, et al. TREM-1 activation alters the dynamics of pulmonary IRAK-M expression in vivo and improves host defense during pneumococcal pneumonia. J Immunol 2009;​183:​2027‑36. 26. Hoogerwerf JJ, de Vos AF, van’t Veer C, et al. Priming of alveolar macrophages upon instillation of lipopolysaccharide in the human lung. Am J Respir Cell Mol Biol 2010;​42:​349‑56. 27. Chung DH, Seaman WE, Daws MR. Characterization of TREM-3, an activating receptor on mouse macrophages: definition of a family of single Ig domain receptors on mouse chromosome 17. Eur J Immunol 2002;​32:​59‑66. 28. van der Poll T, Keogh CV, Buurman WA, Lowry SF. Passive immunization against tumor necrosis factor-alpha impairs host defense during pneumococcal pneumonia in mice. Am J Respir Crit Care Med 1997;​155:​603‑8. 29. Rijneveld AW, Florquin S, Branger J, Speelman P, Van Deventer SJ, van der Poll T. TNF-alpha com- pensates for the impaired host defense of IL-1 type I receptor-deficient mice during pneumococ- cal pneumonia. J Immunol 2001;​167:​5240‑6. 30. Radsak MP, Salih HR, Rammensee HG, Schild H. Triggering receptor expressed on myeloid cells-1 in neutrophil inflammatory responses: differential regulation of activation and survival. J Immu- nol 2004;​172:​4956‑63. 31. Hirst RA, Kadioglu A, O’Callaghan C, Andrew PW. The role of pneumolysin in pneumococcal pneumonia and meningitis. Clin Exp Immunol 2004;​138:​195‑201. 32. Malley R, Henneke P, Morse SC, et al. Recognition of pneumolysin by Toll-like receptor 4 confers resistance to pneumococcal infection. Proc Natl Acad Sci U S A 2003;​100:​1966‑71. 33. Davis KM, Nakamura S, Weiser JN. Nod2 sensing of lysozyme-digested peptidoglycan promotes macrophage recruitment and clearance of S. pneumoniae colonization in mice. J Clin Invest 2011;​ 121:​3666‑76.

36 TREM-1 improves host defence in pneumococcal pneumonia

Supplement

Material and Methods

Induction of pneumonia Mice were intranasally inoculated with 2 × 107 Streptococcus (S.) pneumoniae serotype 2 (strain D39) in a 50 µl saline solution and sacrificed 3, 6, 24 or 48 hours thereafter1,2. Male Trem-1/3−/− mice were compared with a sex and age matched wild type (Wt) control 2 group that was infected at the same time with the exact same inoculum (n = 7-8 per strain for each time point). Collection and handling of samples were done as previ- ously described1,2. In brief, blood was drawn into heparinized tubes and organs were removed aseptically and homogenised in 4 volumes of sterile isotonic saline using a tissue homogenizer (Biospec Products, Bartlesville, UK). To determine bacterial loads, ten-fold dilutions were plated on blood agar plates and incubated at 37°C for 16 hours. Bronchoalveolar lavage (BAL) fluid was obtained from separate groups of infected Trem-1/3−/− and Wt mice (n = 8 per group) at 3 and 6 hours of infection. The trachea was exposed through a midline incision and cannulated with a sterile 22-gauge Abbocath-T catheter (Abbott Laboratories, Sligo, Ireland). BAL was performed by instilling two 0.5 ml aliquots of sterile phosphate buffered saline (PBS) as described earlier3. 0.5–1 ml of BAL fluid was retrieved per mouse. Cell-free BAL supernatants were stored at –20°C for cytokine measurement. Cell numbers were determined using a hemocytometer, and Giemsa-stained cytospin preparations were used for differential cell counts.

TREM-1 expression TREM-1 expression was assessed in essence as previously described4. Wt mice were ter- minated before or 6, 24 or 48 hours after infection (n = 6 at each time point) and blood and bronchoalveolar lavage fluid (BALF) were obtained for FACS analysis as described5. The following antibodies were used (all in concentrations recommended by the manu- facturers): rat anti-mouse TREM-1-phycoerythrin [PE] (clone 174031; R&D systems, Min- neapolis, MN), rat anti-mouse Ly-6G-fluorescein isothiocyanate [FITC] (Pharmingen, San Diego, CA) and rat anti-mouse F4/80- allophycocyanin [APC] (Serotec, Oxford, UK). After staining, cells were fixed in 2% paraformaldehyde. Flow cytometry was done directly after sample collection using a FACS-Calibur instrument (BD Biosciences, San Jose, CA). TREM-1 mean fluorescence intensity (MFI) was measured in the Ly-6G-high gate (granulocytes), in the side scatter-low and F4/80-positive gate (monocytes) and in the side scatter-high and F4/80-positive gate (macrophages). Data are presented as the difference between MFI of specifically stained cells and nonspecifically stained cells.

37 Chapter 2

Histology Lung pathology scores were determined as described elsewhere1. In brief, lungs were harvested at the indicated time points, fixed in 10% buffered formalin, and embedded in paraffin. 4 µm sections were stained with haematoxylin and eosin (HE) and analyzed by a pathologist blinded for groups. To score lung inflammation and damage, the entire lung surface was analyzed with respect to the following parameters: bronchitis, edema, interstitial inflammation, intra-alveolar inflammation, pleuritis, endothelialitis and percentage of the lung surface demonstrating confluent inflammatory infiltrate. Each parameter was graded 0–4, with 0 being ‘absent’ and 4 being ‘severe’. The total pathol- ogy score was expressed as the sum of the score for all parameters. Granulocyte staining was done using FITC-labeled rat anti-mouse Ly-6G mAb (clone RB6-8C5, Pharmingen, San Diego, CA, USA) as described earlier3. Ly-6G expression in the lung tissue sections was quantified by digital image analysis6. In short, lung sections were scanned using the Olympus Slide system (Olympus, Tokyo, Japan) and TIF images, spanning the full tissue section were generated. In these images Ly-6G positivity and total surface area were measured using Image J (U.S. National Institutes of Health, Bethesda, MD, http://rsb.info. nih.gov/ij); the amount of Ly-6G positivity was expressed as a percentage of the total surface area.

Neutrophil activation Bone marrow derived neutrophils from Wt and Trem-1/3−/− mice were isolated from femurs and tibias as previously described7. Bones were flushed with HBSS (without Ca2+/ Mg2+; Gibco Life Technologies, Rockville, MD), washed twice and then resuspended in 3 ml of a 45% Percoll (GE Healthcare) solution in Ca2+/Mg2+-free HBSS. Leukocytes were then loaded on top of a Percoll density gradient by layering successively 2 ml each of 62%, 55%, and 50% Percoll solutions on top of 3 ml of a 81% Percoll solution. Cells were then centrifuged at 1,200 g for 30 minutes at room temperature. The cell band formed between the 81% and 62% layer was harvested using a Pasteur pipette and diluted into fresh Ca2+/Mg2+ free HBSS, washed twice, resuspended in HBSS containing Ca2+/ Mg2+ and seeded at a concentration of 1 × 105/ 100 μl per well in a 96-well U-bottom microtiter plate. Medium, 1 × 107 heat-killed S. pneumoniae/ml or phorbol myristate acetate (PMA 10ng/ml; Sigma Aldrich, St. Louis, MO) was added and incubated to the wells with neutrophils for 1 hour at 37°C in 5% CO2. Subsequently cells were washed and stained with Ly-6G FITC (BD, Pharmingen), CD11b PE (BD, Pharmingen) and CD62L APC (BD, Pharmingen) and analyzed by FACS. The MFI for CD11b and CD62L were calculated in the Ly-6G–high gate (neutrophils).

38 TREM-1 improves host defence in pneumococcal pneumonia

Phagocytosis Phagocytosis of S. pneumoniae was determined in essence as described before2,8. In brief, S. pneumoniae were cultured and washed with pyrogen-free sterile saline and resuspended in sterile PBS to a concentration of 2×109 bacteria/ml. The concentrated S. pneumoniae preparation was treated for 1 h at 37°C with 50 µg/ml Mitomycin C (Sigma- Aldrich; Zwijndrecht, the Netherlands) to prepare alive but growth-arrested bacteria. Subsequently, the growth-arrested S. pneumoniae preparation was washed twice in ice-cold sterile PBS by centrifugation at 4°C, and the final pellet was dispersed in ice-cold 2 PBS in the initial volume and transferred to sterile tubes. Undiluted samples of these preparations failed to generate any bacterial colonies when plated on BA plates, indicat- ing successful growth arrest. Subsequently the bacteria were labeled with carboxyfluo- rescein succinimidyl ester (CFSE, Invitrogen, Breda, the Netherlands) for whole blood incubations. Heat-killed and fluorescein isothiocyanate (FITC, Invitrogen, Breda, the Netherlands) labeled bacteria were used for macrophage experiments. 100 µl heparin- ized whole blood from Trem-1/3−/− and Wt mice was incubated with 10 µl CFSE labeled bacteria in RPMI (Gibco, Life Technologies, Rockville, MD) (end concentration of 1 × 107 CFU/ml) at 37°C (n = 8 per group) or 4°C (n = 4 per group). After 60 minutes, samples were put on ice to stop phagocytosis. Afterwards, red blood cells were lysed using isotonic

NH4Cl solution (155 mM NH4Cl, 10 mM KHCO3, 100 mM EDTA, pH 7.4). Neutrophils were labeled using anti-Ly-6G-PE (clone 1A8, BD Pharmingen, San Diego, CA) and washed twice in FACS-buffer (0.5% BSA, 0.01% NaN3, 0.35 mM EDTA in PBS) for analysis. Bone marrow derived macrophages (BDMDs) were generated using recombinant mouse granulocyte-macrophage colony stimulating factor (GM-CSF, Invitrogen) as previously described9 and were pooled (derived from 4 mice), washed twice and resuspended in RPMI containing 2 mM L-glutamine, and 5% Panexin (cat#: P04-951SA2, PAN biotech, Aidenbach, Germany). Alveolar macrophages were obtained from 8 mice per strain as previously described5 and resuspended in RPMI containing 2 mM L-glutamine and 10% fetal calf serum (FCS). 1 × 105 cells per well were seeded in a 96-well flat-bottom plate in 100 µL to adhere overnight at 37°C, 5% CO2. The following day, macrophages were washed with pre-warmed medium to wash away non-adherent cells. Heat-killed FITC labeled bacteria and macrophages were spun at 1000 RPM for 5 minutes and incubated at 37°C (n = 8 wells per strain) or 4°C (n = 4 wells per strain). After 1 hour, samples were washed with ice-cold PBS, then thoroughly scraped from the bottom, quenched by the addition of 50μl of Trypan Blue (0.4%) and left for one minute on ice after which samples were washed twice in FACS-buffer. The degree of phagocytosis was determined using FACS-Calibur (BD Biosciences, San Jose, CA). The phagocytosis index of each sample was calculated as follows: geomean fluorescence × % positive cells.

39 Chapter 2

Opsonophagocytic killing assay Sorted neutrophils were obtained in essence as described earlier4. Briefly, leukocytes derived from 2 healthy male volunteers (mean age, 28 years; age range, 27–29 years) were sorted into TREM-1 high and TREM-1 low expressing granulocytes by FACS. Un- stimulated whole-blood leukocytes were labeled with TREM-1–PE (R&D systems, Min- neapolis, MN) and CD66b-FITC (BD, Biosciences, San Jose, CA). Subsequently, TREM-1 high and TREM-1 low granulocytes were separated by use of a FACSAria instrument (BD, Biosciences, San Jose, CA). All sorted subsets were >95% pure. Subsequently we assessed opsonophagocytic killing by neutrophils with an ex vivo assay, as described earlier10. A 200-μl assay in Hank’s buffer (with calcium and magnesium) plus 0.1% gela- tin was composed of 100 PBS-washed bacteria, 105 neutrophils in Hank’s buffer (with calcium and magnesium) (Gibco, Life Technologies, Rockville, MD) plus 0.1% gelatin and 20 μl of fresh human serum as a source of complement. The assays were incubated for 45 min at 37°C with rotation. After stopping the reaction by incubation at 4°C, viable counts were determined in serial dilutions. Bacterial killing was determined relative to the same experimental condition without the addition of neutrophils and expressed as percentage bacterial viability.

Macrophage stimulation BMDMs (1 × 105/well) and alveolar macrophages (5 × 104/well) from Wt and Trem-1/3−/− mice were resuspended in RPMI containing 2 mM l-glutamine, penicillin, streptomycin and 5% Panexin (BMDMs) or 10% FCS (alveolar macrophages) and were plated in a

96-well flat-bottom plate and allowed to adhere for 16 hours at 37°C in 5% CO2. Subse- quently the cells were washed with PBS to remove nonadherent cells. Next, the adherent cells were stimulated for 6 or 24 hours with or without the indicated MOI of heat-killed

S. pneumoniae at 37°C in 5% CO2. Supernatants were taken and stored at −20°C until assayed.

40 TREM-1 improves host defence in pneumococcal pneumonia

References

1. Rijneveld AW, Weijer S, Florquin S, et al. 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 2004;​103:​1702‑9. 2. Achouiti A, Vogl T, Urban CF, et al. Myeloid-related protein-14 contributes to protective immunity in gram-negative pneumonia derived sepsis. PLoS Pathog 2012;​8:​e1002987. 3. Knapp S, Wieland CW, van ‘t Veer C, et al. Toll-like receptor 2 plays a role in the early inflammatory response to murine pneumococcal pneumonia but does not contribute to antibacterial defense. 2 J Immunol 2004;​172:​3132‑8. 4. Wiersinga WJ, Veer Cvt, Wieland CW, et al. Expression Profile and Function of Triggering Recep- tor Expressed on Myeloid Cells-1 during Melioidosis. Journal of Infectious Diseases 2007;​196:​ 1707‑16. 5. Knapp S, Leemans JC, Florquin S, et al. Alveolar Macrophages Have a Protective Antiinflammatory Role during Murine Pneumococcal Pneumonia. Am J Respir Crit Care Med 2003;​167:​171‑9. 6. Lammers AJ, de Porto AP, Florquin S, et al. Enhanced vulnerability for Streptococcus pneumoniae sepsis during asplenia is determined by the bacterial capsule. Immunobiology 2011;​216:​863‑70. 7. Van Ziffle JA, Lowell CA. Neutrophil-specific deletion of Syk kinase results in reduced host defense to bacterial infection. Blood 2009;​114:​4871‑82. 8. Wiersinga WJ, Kager LM, Hovius JW, et al. Urokinase receptor is necessary for bacterial defense against pneumonia-derived septic melioidosis by facilitating phagocytosis. J Immunol 2010;​184:​ 3079‑86. 9. Eske K, Breitbach K, Kohler J, Wongprompitak P, Steinmetz I. Generation of murine bone marrow derived macrophages in a standardised serum-free cell culture system. J Immunol Methods 2009;​ 342:​13‑9. 10. Lysenko ES, Clarke TB, Shchepetov M, et al. Nod1 signaling overcomes resistance of S. pneu- moniae to opsonophagocytic killing. PLoS Pathog 2007;​3:​e118.

41 Chapter 2

A B

C D

Figure S1: No difference between Wildtype and Trem-1/3−/− mice in neutrophil influx into the lung during pneumococcal pneumonia. Wildtype (Wt) and Trem-1/3−/− mice (7-8 per group) were intranasally inoculated with 2 × 107 colony forming units (CFU) Streptococcus (S.) pneumoniae and sacrificed 24 and 48 hours later. Representative Ly-6G stained histopathological slides of Wt (A, C) and Trem-1/3−/− (B, D) 24 and 48 hours after inoculation with S. pneumoniae (original magnification ×200).

5 2.0 10 Wt Trem-1/3 -/- s e

g 1.5 105 a oph 1.0 105 macr l l

a 4 t 5.0 10 To

0.0 3h 6h Figure S2: No difference in number of alveolar macrophages between Wildtype and Trem-1/3−/− mice during pneumococcal pneumonia. Wildtype (Wt) and Trem-1/3−/− mice (7-8 per group) were intranasally inoculated with 2 × 107 colony forming units (CFU) Streptococcus (S.) pneumoniae and sacrificed 3 and 6 hours later and the number of macrophages were determined in bronchoalveolar lavage fluid (BALF).

42 TREM-1 improves host defence in pneumococcal pneumonia

A B C 10000 Wt 50000 60000 Trem-1/3 -/- 8000 40000 l l l * m m

m 40000 6000 30000 pg/ pg/ pg/ 2 1 L L - F C 4000 C 20000 X X N 20000 T C *** *** C ** 2000 10000

0 0 0 3h 6h 3h 6h 3h 6h Figure S3: Cytokine and chemokine concentrations in lung homogenates of Wildtype and Trem-1/3−/− mice during pneumococcal pneumonia. Wildtype (Wt) and Trem-1/3−/− mice (7-8 per group) were intrana- 2 sally inoculated with 2 × 107 colony forming units (CFU) Streptococcus (S.) pneumoniae and sacrificed 3 and 6 hours later. Levels of Tumor Necrosis Factor (TNF)-α (A), Chemokine (C-X-C motif) ligand 1 (CXCL1) (B) and CXCL2 (C) were measured by ELISA. Data are representative for 2 repeated experiments and mean ± SEM are shown; *P < 0.05; **P < 0.01; ***P < 0.001.

Figure S4: No difference in killing capacity by TREM-1 high and low expressing human neutrophils. Neutrophils from healthy volunteers were sorted in TREM-1 high expressing cells and cells expressing low levels of TREM-1 on their surface (Figure S4A). Killing of S. pneumoniae was determined in vitro. No differ- ence was observed between TREM-1 high and low expressing neutrophils (Figure S4B). Data are represen- tative for 2 repeated experiments and mean ± SEM are shown.

43

DNAX-activating protein of 12 kDa (DAP12) impairs host defense in pneumococcal pneumonia

Tijmen J. Hommes, MD1,2; Arie J. Hoogendijk, PhD1,2; Mark C. Dessing, PhD3; Cornelis van ’t Veer, PhD1,2; Sandrine Florquin, MD, PhD3; Alex F. de Vos, PhD1,2; Tom van der Poll, 3 MD, PhD1,2,4 1Center for Experimental and Molecular Medicine and 2Center for Infection and Immunity, Academic Medical Center, University of Amsterdam, the Netherlands; 3Department of Pathology; Academic Medical Center, University of Amsterdam, The Netherlands; 4Division of Infectious Diseases, Academic Medical Center, University of Amsterdam, the Netherlands

Critical Care Medicine 2014 Dec;42(12):e783-90 Chapter 3

Abstract

Objective: Streptococcus (S.) pneumoniae is the most common causative organism in community-acquired pneumonia responsible for millions of deaths every year. DNAX – activating protein of 12 kDa (DAP12) is an adapter molecule for different myeloid expressed receptors involved in innate immunity. Design: Animal study Setting: University research laboratory Subjects: DAP12 deficient (dap12−/−) and wild type (Wt) mice Interventions: Mice were intranasally infected with S. pneumoniae. In addition, ex vivo responsiveness of alveolar macrophages was examined. Measurements and main results: dap12−/− alveolar macrophages released more tumor necrosis factor-α upon stimulation with S. pneumoniae and displayed increased phago- cytosis of this pathogen as compared with Wt cells. After infection with S. pneumoniae via the airways dap12−/− mice demonstrated reduced bacterial outgrowth in the lungs together with delayed dissemination to distant body sites relative to Wt mice. This favor- able response in dap12−/− mice was accompanied by reduced lung inflammation and an improved survival. Conclusion: These data suggest that DAP12 impairs host defense during pneumococcal pneumonia at the primary site of infection at least in part by inhibiting phagocytosis by alveolar macrophages.

46 DAP12 impairs host defense in pneumococcal pneumonia

Introduction

The immune system uses a wide array of receptors to sense and fight off invading pathogens. A well-known family of pattern recognition receptors, the Toll-like recep- tors (TLRs), recognizes specific molecular patterns present in microbes, an interaction that leads to activation of the innate immune system1. Receptors that signal through immunoreceptor tyrosine-based activation motifs (ITAMs) represent another important family of receptors that are able to cross-regulate heterologous receptor signaling and thereby “fine-tune” the immune response2,3. Many of these receptors signal through the adaptor molecule DNAX-activating protein of 12kDa (DAP12), also known as TYROBP or KARAP4,5. DAP12 is a homodimeric ITAM-bearing transmembrane adaptor protein 3 that shows homology with the FcRγ signaling molecule and was originally identified as a mediator of natural killer (NK) cell activation6. Since then, DAP12 was found to be expressed in variety of myeloid cells including dendritic cells, macrophages, neutrophils, microglia, osteoclasts and, more recently, B-cells7-10. DAP12 pairs with numerous cell- surface receptors and can either have stimulatory or inhibiting effects on cells depend- ing on the avidity of the interaction between the DAP12-associated receptor and its ligand11. DAP12-associated receptors have been shown to recognize both host derived and microbial ligands, suggesting that they play an important role in innate immune responses5. Upon ligation of a DAP12-coupled receptor, Src family kinases become activated and the tyrosine residues within the ITAM of DAP12 are phosphorylated. Subsequently this leads to the recruitment and phosphorylation of Syk, which results in downstream signaling through phospholipase-Cγ, ERK and PI3-kinase pathways12,13. In macrophages this ultimately results in the secretion of cytokines and chemokines14. Early research established a protective role for DAP12 in host defense against viral infections, at least in part due to its involvement in NK cell activation6,8. Knowledge of the contribution of DAP12 to the pathogenesis of bacterial infections is limited. During endotoxemia and septic peritonitis DAP12 deficient (dap12−/−) mice showed a reduced lethality when compared to wild type (Wt) mice13. Additionally, dap12−/− mice had lower bacterial burdens after intravenous infection with the intracellular pathogen Listeria (L.) monocytogenes12. In contrast, during infection with Salmonella enterica serovar Ty- phimurium, dap12−/− mice displayed an impaired host defense as reflected by higher bacterial burdens and increased inflammation compared to Wt mice15. Streptococcus (S.) pneumoniae is the leading cause of community-acquired pneumo- nia and is associated with high morbidity and mortality16. In the United States alone, this Gram-positive diplococcus is responsible for more than half a million pneumonia cases and 50,000 episodes of bacteremia each year, with case fatality rates of 7 and 20% respectively17; similar estimates have been reported for Europe18. Worldwide S. pneu- moniae is accountable for an estimated 2 million deaths every year19. As such, infections

47 Chapter 3 caused by S. pneumoniae represent a major health burden. Here we set out to investigate the involvement of DAP12 in host defense against pneumococcal pneumonia.

Material & Methods

Detailed methods are provided in the supplement.

Mice dap12−/− mice were generated as previously described and backcrossed to a C57BL/6 genetic background20. Age and sex matched Wt C57BL/6 mice were purchased from Charles River (Maastricht, The Netherlands). The Institutional Animal Care and Use Com- mittee approved all animal experiments.

Cell culture and whole blood stimulation Alveolar macrophages from 8 individual dap12−/− and Wt mice were obtained as de- scribed elsewhere21. Heparinized whole blood was obtained from 8 individual dap12−/− and Wt mice in a separate experiment. Growth-arrested bacteria were prepared as described22. Alveolar macrophages and whole blood were incubated for 24 hours with or without viable, growth-arrested S. pneumoniae at the indicated multiplicity of and supernatants were taken and stored at −20°C until assayed for tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6, Il-10 and keratinocyte-derived chemokine (KC).

Phagocytosis Phagocytosis of S. pneumoniae by alveolar macrophages (obtained as described23) and blood neutrophils was done in essence as previously described24,25. Briefly alveolar macrophages and whole blood were incubated with CFSE labeled S. pneumoniae and internalization was measured by FACS.

Experimental design Pneumonia was induced by intranasal inoculation with S. pneumoniae serotype 3 (American Type Culture Collection, ATCC 6303, Rockville, MD; 5 × 104 CFU) as described previously26,27. Mice were sacrificed 6, 24 or 48 hours thereafter (n = 7-8 per strain at each time point). Collection and handling of samples were done as previously described25-27. In survival studies mice (n = 15 per strain) were intranasally inoculated with 5 × 104 or 5 × 103 CFU S. pneumoniae and monitored for up to 14 days after infection.

48 DAP12 impairs host defense in pneumococcal pneumonia mRNA expression analysis DAP12, TREM-1 and TREM-2 expression in whole lung homogenates after intranasal infection with S. pneumoniae was analyzed using quantitative RT-PCR.

Assays Lung homogenates were prepared for immune assays as described before25-27. TNF-α, IL- 1β, IL-6, KC, macrophage inflammatory protein 2 (MIP-2) (all R&D systems, Minneapolis, MN) and myeloperoxidase (MPO; Hycult Biotechnology BV, Uden, the Netherlands) were measured using specific ELISAs according to manufacturers’ recommendations. In cell culture supernatants IL-1β, IL-6, KC and IL-10 were measured by cytometric bead array flex set assay (BD Biosciences, San Jose, CA). 3 Histology After embedding, lungs were stained with haematoxylin and eosin to score inflamma- tion or stained and quantified for Ly-6G using immunohistochemical procedures as described25-27.

Statistical analysis Data are expressed as scatter plots with median or as medians with interquartile ranges. Comparisons between groups were conducted using overall nonparametric analysis of variance with modeled effects for strain, time, and their interaction (if appropriate), followed by post-hoc Mann-Whitney U tests at the individual time points. For survival studies Kaplan-Meier analyses followed by log rank test were performed. All analyses were done using GraphPad Prism version 5.01 (GraphPad Software, San Diego, CA). P- values less than 0.05 were considered statistically significant.

Results

DAP12 deficiency results in increased TNF-α production by alveolar macrophages and blood leukocytes in response to S. pneumoniae Previous reports have shown enhanced cytokine responses of dap12−/− bone marrow derived macrophages when stimulated with TLR ligands2,12. To obtain a first insight in the role of DAP12 in the host defense against S. pneumoniae, we set out to investigate the responsiveness of dap12−/− and Wt alveolar macrophages and whole blood leuko- cytes to S. pneumoniae in vitro. To this end we stimulated alveolar macrophages and whole blood prepared from both genotypes for 24 hours with growth arrested S. pneu- moniae and measured TNF-α, IL-1β, IL-6, KC and IL-10 in supernatants. dap12−/− alveolar macrophages produced more TNF-α than Wt cells (Figure 1A P < 0.05), whereas other

49 Chapter 3

A Alveolar macrophages B Whole blood 1000 2500 Wt ** dap12 -/- 800 2000 * l l m m 600 1500 pg/ pg/ F 400 F 1000 N N T T 200 500

0 0 5 6 7 medium S. pneumoniae medium 10 10 10 S. pneumoniae Figure 1: Increased TNF-α release by DAP12 deficient alveolar macrophages and blood leukocytes in response to S. pneumoniae. Alveolar macrophages (A) and whole blood (B) prepared from DAP12 defi- cient (dap12−/−) and wild type (Wt) mice were incubated with viable, growth-arrested S. pneumoniae for 24 hours (105, 106 and 107/ml for whole blood and MOI 100:1 (bacteria:cell ratio) for macrophages), after which TNF-α was measured in supernatants. Data are expressed as median with interquartile ranges; *P < 0.05 **P < 0.01 (both versus Wt cells). cytokines remained undetectable. Blood leukocytes from dap12−/− mice produced more TNF-α, IL-β, IL-6 and IL-10 than Wt cells (Figure 1B and Supplemental figure 1 A, B and D respectively; P < 0.05 and P < 0.01). These data demonstrate an inhibiting role for DAP12 in leukocyte responses to S. pneumoniae.

DAP12 deficiency enhances phagocytic capacity of alveolar macrophages Different DAP12 associated receptors have been implicated in phagocytosis by macro- phages28,29. To examine a possible influence of DAP12 on phagocytosis ofS. pneumoniae, we incubated alveolar macrophages and whole blood from dap12−/− and Wt mice with

A Alveolar macrophages B Neutrophils 1500 Wt 15000 x x dap12 -/- e e nd nd i i 1000 ** 10000 s s i i s s o o t t cy cy 500 5000 go go a a h h P P

0 0 4°C 37°C 4°C 37°C Figure 2: Enhanced phagocytosis of S. pneumoniae by DAP12 deficient alveolar macrophages. Al- veolar macrophages (A) or whole blood (B) from DAP12 deficient (dap12−/−) and wild type (Wt) mice was incubated at 4°C (n = 4) or 37°C (n = 8) with viable, growth-arrested carboxyfluorescein succinimidyl ester (CFSE) labeled viable Streptococcus (S.) pneumoniae (107/ml) or 1 hour and the degree of phagocytosis was assessed by flow cytometry. The phagocytosis index of each sample was calculated as geo mean fluores- cence × % positive cells. Data are expressed as median with interquartile ranges; **P < 0.01 versus Wt cells.

50 DAP12 impairs host defense in pneumococcal pneumonia viable, growth arrested pneumococci for 1 hour and determined internalization by flow cytometry. Alveolar macrophages from dap12−/− mice displayed an enhanced capacity to internalize S. pneumoniae (Figure 2A, P < 0.01), while phagocytosis of S. pneumoniae by dap12−/− and Wt neutrophils was similar (Figure 2B).

Pneumonia results in enhanced expression of DAP12, TREM-1 and TREM-2 mRNAs in lungs Inoculation with S. pneumoniae via the airways resulted in increases in DAP12, TREM-1 and TREM-2 mRNA levels in whole lung homogenates 24 and 48 hours post infection (Supplemental figure 2; P < 0.01 or P < 0.001).

DAP12 deficient mice demonstrate reduced bacterial outgrowth during 3 pneumococcal pneumonia To investigate the functional role of DAP12 in host defense during Gram-positive pneumonia, we infected dap12−/− and Wt mice with viable S. pneumoniae and harvested lungs, blood and spleens at predefined time points for quantitative cultures (Figure 3). At 6 hours after infection bacterial loads were similar in lungs of dap12−/− and Wt mice, while blood and distant organs showed no growth of pneumococci, indicating that the infection was still localized at this early time point. Remarkably, however, after 24 and 48 hours of infection dap12−/− mice had 44 and 6 fold lower bacterial counts in their lungs respectively compared to Wt mice (Figure 3A; P < 0.01 for both time points). At these later time points, S. pneumoniae could also be recovered from extrapulmonary body sites. Notably, dap12−/− mice displayed a significantly delayed dissemination of the infection: at 24 hours after infection S. pneumoniae could be cultured from the blood of only 3 of 8 dap12−/− mice, compared with 8 of 8 Wt mice (Figure 3B; P < 0.01). Likewise only 3 of 8 dap12−/− mice, compared with 8 of 8 Wt mice had positive spleen cultures 24 hours after infection (Figure 3C; P < 0.01). After 48 hours of infection bacterial burdens

A Lung B Blood C Spleen 109 109 109 8 ** 8 8 10 * 10 10 107 107 107 g 106 g 106 g 106 l lo l 105 lo l 105 lo l 105 m m m / 4 / 4 / 4 U 10 U 10 ** U 10 F 3 F 3 F 3 ** C 10 C 10 C 10 102 102 102 101 101 101 100 100 100 6h 24h 48h 24h 48h 24h 48h

Figure 3: DAP12 deficiency results in reduced outgrowth of S. pneumoniae in lungs and delayed dis- semination to distant body sites. Wild type (Wt) and DAP12 deficient (dap12−/−) mice (n = 7-8 per group) were intranasally infected with 5 × 104 CFU S. pneumoniae and sacrificed 6, 24 and 48 hours later. Bacterial loads were determined in lung homogenates (A), blood (B) and spleen (C). Horizontal lines represent medi- ans; * P < 0.05 ** P < 0.01 versus Wt mice.

51 Chapter 3 were similar in blood and spleen, although 6 of 8 dap12−/− mice and 7 of 7 Wt mice had positive blood cultures at this time point (Figure 4B; P = 0.13). These data indicate that the presence of DAP12 facilitates pneumococcal growth at the primary site of infection and subsequent (early) dissemination.

A Pathology score B % confluent infiltrates 25 Wt 50 dap12 -/- 20 * 40

15 30

% * 10 * 20 5 10 ND 0 0 * 6h 24h 48h 6h 24h 48h C D

-/- Wt (24h) dap12 (24h)

E F

-/- Wt (48h) dap12 (48h)

Figure 4: Reduced pulmonary pathology in DAP12 deficient mice during pneumococcal pneumonia. Total pathology score at indicated time points post infection in Wt and dap12−/− mice was determined ac- cording to the scoring system described in the Methods section (A). Area of confluent inflammation -ex pressed as percentage of total lung surface (B). Representative slides of lung haematoxylin and eosin (HE) staining of wild type (Wt) (C) and DAP12 deficient (dap12−/−) mice (D) 24 hours, Wt (E); dap12−/− mice (F) 48 hours after intranasal infection with 5 × 104 CFU S. pneumoniae. Magnification 200×. Data are expressed as median with interquartile ranges (7-8 mice per group at each time point); * P < 0.05 versus Wt mice at the same time point.

52 DAP12 impairs host defense in pneumococcal pneumonia

DAP12 deficiency results in decreased lung pathology during pneumococcal pneumonia To obtain further insight in the role of DAP12 in the regulation of lung inflammation induced by S. pneumoniae, we semi-quantitatively analyzed lung tissue slides from infected dap12−/− and Wt mice using the scoring system outlined in the Methods section (Figure 4). Already at 6 hours after infection there was mild pulmonary inflammation in the lungs of all mice, gradually increasing thereafter. dap12−/− mice showed reduced lung pathology at both 24 and 48 hours after infection (Figure 4A; P < 0.05 versus Wt mice). In addition, dap12−/− mice had a smaller surface area of confluent inflammatory infiltrate compared to Wt mice at these time points (Figure 4B; P < 0.05).

Impact of DAP12 deficiency on neutrophil influx into the lungs during 3 pneumococcal pneumonia A hallmark of host defense against pneumococcal pneumonia is the strong influx of neutrophils into the lungs30,31. Therefore we determined the extent of neutrophil influx in dap12−/− and Wt mice at 6, 24 and 48 hours after intranasal infection with S. pneumoniae by assessing the area of Ly-6G positivity in lung tissue slides by digital image analysis and by measuring MPO concentrations in whole lung homogenates. Consistent with their reduced lung pathology score (Figure 4), dap12−/− mice displayed a lower number of Ly6+ cells in lung tissue 24 hours after inoculation (Figure 5A; P < 0.05 versus Wt mice), whereas MPO levels in lung homogenates were not significantly different (Figure 5B; P = 0.14). At 48 hours, differences between genotypes had subsided.

Table 1: Lower cytokine and chemokine concentrations in lung homogenates of dap12−/− mice dur- ing pneumococcal pneumonia. Lung cytokines 6 hours 24 hours 48 hours Wt dap12−/− Wt dap12−/− Wt dap12−/− TNF-α (pg/ml) 679 680 270 326 954 1207 (582-749) (651-751) (199-357) (247-403) (741-1482) (972-1858) IL-1β (pg/ml) 283 320 1049 391 3830 4815 (189-450) (259-643) (678-3243) (178-1178)* (2885-5632) (2999-7919) IL-6 (pg/ml) 266 311 2179 364 4306 3491 (256-342) (283-358) (1648-6433) (342-1249)** (3548-6814) (2611-4638) KC (ng/ml) 986 341 4814 722 11682 6007 (545-1715) (284-465)** (2723-7324) (334-2723)** (10640-26728) (4111-7621) Mip-2 (ng/ml) 772 814 2123 1926 52419 25510 (691-884) (736-1006) (1951-3646) (1481-2407) (45311-88352) (14604-32485)* Pro-inflammatory cytokine (TNF-α, IL-1β, IL-6) and chemokine (KC and MIP-2) levels in lung homogenates at 6, 24 and 48 hours after intranasal Streptococcus (S.) pneumoniae infection in wild type (Wt) and DAP12 deficient (dap12−/−) mice. Data are medians with interquartile ranges; n = 7-8 mice per group per time point; * P < 0.05, **P < 0.01 (both versus Wt mice).

53 Chapter 3

A B

60 30

Wt ) -/- % dap12 (

l

40 m 20 area O ng/ O lung lung P +

20 M 10 G G

6 * - y L 0 0 6h 24h 48h 6h 24h 48h C D

-/- Wt (24h) dap12 (24h)

Figure 5: Temporarily reduced neutrophil influx into lung tissue of DAP12 deficient mice during pneumococcal pneumonia. Quantification of pulmonary Ly-6G positivity (A) and MPO levels in whole lung homogenates (B) 6, 24 and 48 hours after intranasal infection with 5 × 104 CFU S. pneumoniae of wild type (Wt) and DAP12 deficient (dap12−/−) mice. Representative neutrophil stainings (brown) of Wt (C) and dap12−/− mice (D) 24 hours after induction of pneumococcal pneumonia are shown. Data are expressed as median with interquartile ranges (7-8 mice per group at each time point); * P < 0.05. Magnification 200×.

A 50,000 CFU B 5000 CFU 100 100 Wt -/- l l 80 dap12 80 va va i i rv rv u u 60 60 s s

t t n n 40 40 erce erce P P 20 20 P=0.17 P<0.05 0 0 0 2 4 6 8 10 12 14 0 2 4 6 8 10 12 14 Days Days

Figure 6: DAP12 deficiency protects against lethality in pneumococcal pneumonia. Wild type (Wt) and DAP12 deficient (dap12−/−) mice (n = 15 per strain in each experiment) were intranasally infected with with 5 × 104 CFU (A) or with 5 × 103 CFU S. pneumoniae (B) and survival was monitored for 14 days. P values indicate differences between groups.

54 DAP12 impairs host defense in pneumococcal pneumonia

DAP12 deficiency reduces the lung levels of some cytokines and chemokines during S. pneumoniae pneumonia To study the impact of DAP12 deficiency on cytokine and chemokine productionin vivo, we measured the levels of cytokines (TNF-α, IL-1β, IL-6) and chemokines (KC and MIP-2) in lungs harvested from dap12−/− and Wt mice after infection with S. pneumoniae (Table 1). The concentrations of these mediators were relatively low at 6 hours after inoculation, increasing thereafter. dap12−/− mice showed reduced pulmonary levels of IL-6 (24 hours; P < 0.01 versus Wt mice), KC (6 and 24 hours, both P < 0.01) and MIP-2 (48 hours; P < 0.05).

DAP12 negatively affects mortality during pneumococcal pneumonia Finally, to determine the impact of DAP12 deficiency on survival we performed an ob- 3 servational study using the same infectious dose as used in the experiments described above (5 × 104 S. pneumoniae). In this study dap12−/− mice displayed a non-significant survival advantage: 5 of 15 (33%) dap12−/− mice survived compared to 1 of 15 (7%) Wt mice (Figure 6A; P = 0.17). Arguing that the infectious dose might have been too high to reveal a detrimental effect of DAP12 on survival, we repeated the experiment with a 10-fold lower inoculum (5 × 103 S. pneumoniae). This infectious challenge still resulted in 60% lethality in Wt mice 6 days after infection, while mortality was significantly lower amongst dap12−/− mice (27%; Figure 6B; P < 0.05). These data demonstrate a detrimental role of DAP12 in protective immunity that appears to be dose dependent.

Discussion

DAP12 is a transmembrane signaling adaptor associated with different activating or inhibiting cell surface receptors in humans and mice5. Limited data exist on the role of DAP12 in bacterial infection. To the best of our knowledge, we here for the first time evaluated the role of DAP12 in bacterial respiratory tract infection, using a clinically relevant human pathogen, the pneumococcus, the main causative agent in community- acquired pneumonia. We here show that DAP12 deficiency improved host defense dur- ing pulmonary infection with S. pneumoniae, as reflected by reduced bacterial growth at the primary site of infection, delayed dissemination of pneumococci to distant body sites, attenuated lung inflammation and an improved survival. Our results suggest that the deleterious role of DAP12 results at least in part from its inhibiting effect on phago- cytosis of S. pneumoniae by alveolar macrophages. In a model of polymicrobial abdominal sepsis induced by cecal ligation and puncture dap12−/− mice demonstrated a reduced lethality accompanied by an attenuated inflam- matory response13. Similarly, dap12−/− mice showed diminished mortality together with mitigated proinflammatory cytokine release during endotoxic shock13. Together these

55 Chapter 3 findings suggest that during severe systemic Gram-negative infection DAP12 may serve as a lethal amplifier of inflammation. In a colitis model induced by streptomycin ad- ministration and oral infection with Salmonella Typhimurium, dap12−/− mice had higher bacterial burdens in their ceca than Wt mice, which was associated with increased lo- cal inflammation15. Bacterial loads did not differ between dap12−/− and Wt mice in organs distant from the gut and neither did survival. While this study15 hinted to a role for DAP12 in antibacterial defense, at least in the intestine, dap12−/− mice displayed reduced bacterial loads after intravenous infection with the intracellular bacterium L. monocytogenes12. Our current study clearly differs from these earlier investigations: we used a model of community-acquired pneumonia characterized by a gradually growing bacterial load at the primary site of infection followed by bacterial spreading, allowing to study a potential role of DAP12 in both the initial immune response as well as the subsequent harmful inflammation phase. In this respect it should be noted that the lung is of particular interest for studies on DAP12 and antibacterial defense considering its constitutive expression in this organ32,33. DAP12 has been shown to inhibit TLR responses of macrophages in vitro12. Since al- veolar macrophages are important for host defense during pneumococcal pneumonia34, we first sought to obtain insight into the function of DAP12 in responsiveness of these cells to S. pneumoniae in vitro. In accordance with earlier findings12, we found enhanced TNF-α release by dap12−/− alveolar macrophages upon stimulation with growth arrested S. pneumoniae. Similarly, dap12−/− blood leukocytes produced higher TNF-α levels than Wt cells when exposed to pneumococci. Interestingly, like dap12−/− macrophages, macrophages lacking the DAP12 coupled receptor Triggering Receptor Expressed by Myeloid Cells (TREM)-2 also display increased cytokine responses to stimulation with TLR ligands35. Whether TREM-2 is responsible for the inhibiting effect of DAP12 in response to S. pneumoniae awaits further research. Although a brisk induction of proinflammatory cytokine release, and particularly of TNF-α, plays an eminent role in protective immunity during pneumococcal pneumonia36, it seems less likely that the enhanced responsiveness of dap12−/− alveolar macrophages significantly contributed to the improved host defense of dap12−/− mice in vivo, considering that we did not find evidence for increased cytokine release (or lung inflammation) in these animals after infection with S. pneumoniae via the airways. If anything, dap12−/− mice had lower lung cytokine levels after infection, which was likely caused by the lower bacterial burdens in these mice. Alternatively, cell types other than alveolar macrophages probably contribute to the production of cytokines in lungs during pneumonia. Interestingly, alveolar macrophages from dap12−/− mice displayed enhanced phagocytosis of viable pneumococci. Although one report showed defective phagocytosis of Escherichia coli by dap12−/− bone marrow derived macrophages28, another report found dap12−/− mac- rophages to phagocytose zymosan more efficiently than Wt cells4. These discrepancies

56 DAP12 impairs host defense in pneumococcal pneumonia likely relate to the variety of receptors with which DAP12 is linked. Further research is necessary to determine how DAP12 interferes with phagocytosis of S. pneumoniae by alveolar macrophages, and whether this is connected to its inhibitory effect on release of proinflammatory mediators. DAP12 deficiency did not influence pulmonary inflammation during the early phase of pneumococcal pneumonia, while at later time points less inflammation was present in the lungs of dap12−/− mice. The attenuated inflammatory response in dap12−/− mice was also reflected by a reduced number of neutrophils in lungs of these mice at 24 hours after infection. It was shown recently that the DAP12 coupled receptor TREM-1 is crucially involved in the transepithelial migration of neutrophils in murine lungs during bacterial infection37. Hence the absence of DAP12 and thus of TREM-1 signaling could have resulted 3 in lower influx of neutrophils into to the lungs. Furthermore, lower levels of pulmonary KC and IL-1β could have contributed to reduced neutrophil recruitment in dap12−/− mice. Since DAP12 is a promiscuous signaling adaptor for many receptors involved in innate immune responses, it is difficult to pinpoint which receptor is responsible for the observed phenotype. Likely, DAP12 is concurrently activated by several receptors during severe infections, either by the causative pathogen or components thereof, or by molecules released by or from host cells. Studies examining the role of DAP12 in infection in vivo are further complicated by the fact that activation of this adaptor can lead to either decreased or enhanced inflammatory or immunological consequences, in which the net effect of DAP12 activation likely depends on the pathogen and the sever- ity and primary source of the infection, with differential engagement of DAP12-coupled receptors and differential roles of distinct DAP12 expressing cell types in host defense against the causative organism of the infection.

Conclusions The present study expands current knowledge of the involvement of DAP12 in host defense against intracellular microorganisms12,15,38 and influenza virus33,38, showing for the first time that DAP12 impairs host defense during respiratory tract infection caused by S. pneumoniae. Our studies were limited to one pneumococcal strain. Extrapolation to humans should be done with caution and further studies are warranted to investigate the role of DAP12 and its potential as a drug target in human infections. This would also require additional animal investigations in which DAP12 is inhibited in a delayed manner after induction of pneumonia in the context of antibiotic therapy.

Acknowledgements: We thank Marieke ten Brink, Joost Daalhuisen and Regina de Beer for their expert techni- cal assistance.

57 Chapter 3

References:

1. Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011;​34:​637‑50. 2. Wang L, Gordon RA, Huynh L, et al. Indirect inhibition of Toll-like receptor and type I interferon responses by ITAM-coupled receptors and integrins. Immunity 2010;​32:​518‑30. 3. Nathan C, Ding A. Nonresolving inflammation. Cell 2010;​140:​871‑82. 4. Hamerman JA, Ni M, Killebrew JR, Chu CL, Lowell CA. The expanding roles of ITAM adapters FcRgamma and DAP12 in myeloid cells. Immunol Rev 2009;​232:​42‑58. 5. Lanier LL. DAP10- and DAP12-associated receptors in innate immunity. Immunol Rev 2009;​227:​ 150‑60. 6. Lanier LL, Corliss BC, Wu J, Leong C, Phillips JH. Immunoreceptor DAP12 bearing a tyrosine-based activation motif is involved in activating NK cells. Nature 1998;​391:​703‑7. 7. Tomasello E, Olcese L, Vely F, et al. Gene structure, expression pattern, and biological activity of mouse killer cell activating receptor-associated protein (KARAP)/DAP-12. J Biol Chem 1998;​273:​ 34115‑9. 8. Lanier LL, Corliss B, Wu J, Phillips JH. Association of DAP12 with activating CD94/NKG2C NK cell receptors. Immunity 1998;​8:​693‑701. 9. Bouchon A, Dietrich J, Colonna M. Cutting edge: inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J Immunol 2000;​164:​4991‑5. 10. Nakano-Yokomizo T, Tahara-Hanaoka S, Nakahashi-Oda C, et al. The immunoreceptor adapter protein DAP12 suppresses B lymphocyte-driven adaptive immune responses. J Exp Med 2011;​ 208:​1661‑71. 11. Turnbull IR, Colonna M. Activating and inhibitory functions of DAP12. Nat Rev Immunol 2007;​7:​ 155‑61. 12. Hamerman JA, Tchao NK, Lowell CA, Lanier LL. Enhanced Toll-like receptor responses in the absence of signaling adaptor DAP12. Nat Immunol 2005;​6:​579‑86. 13. Turnbull IR, McDunn JE, Takai T, Townsend RR, Cobb JP, Colonna M. DAP12 (KARAP) amplifies inflammation and increases mortality from endotoxemia and septic peritonitis. J Exp Med 2005;​ 202:​363‑9. 14. Bouchon A, Facchetti F, Weigand MA, Colonna M. TREM-1 amplifies inflammation and is a crucial mediator of septic shock. Nature 2001;​410:​1103‑7. 15. Charles JF, Humphrey MB, Zhao X, et al. The innate immune response to Salmonella enterica serovar Typhimurium by macrophages is dependent on TREM2-DAP12. Infect Immun 2008;​76:​ 2439‑47. 16. Mandell LA, Wunderink RG, Anzueto A, et al. Infectious Diseases Society of America/American Thoracic Society consensus guidelines on the management of community-acquired pneumonia in adults. Clin Infect Dis 2007;​44 Suppl 2:​S27‑72. 17. Prevention of pneumococcal disease: recommendations of the Advisory Committee on Immuni- zation Practices (ACIP). MMWR Recomm Rep 1997;​46:​1‑24. 18. Welte T, Torres A, Nathwani D. Clinical and economic burden of community-acquired pneumonia among adults in Europe. Thorax 2012;​67:​71‑9. 19. Bartlett JG, Dowell SF, Mandell LA, File Jr TM, Musher DM, Fine MJ. Practice guidelines for the man- agement of community-acquired pneumonia in adults. Infectious Diseases Society of America. Clin Infect Dis 2000;​31:​347‑82.

58 DAP12 impairs host defense in pneumococcal pneumonia

20. Kaifu T, Nakahara J, Inui M, et al. Osteopetrosis and thalamic hypomyelinosis with synaptic degen- eration in DAP12-deficient mice. J Clin Invest 2003;​111:​323‑32. 21. Wiersinga WJ, Wieland CW, Dessing MC, et al. Toll-like receptor 2 impairs host defense in gram- negative sepsis caused by Burkholderia pseudomallei (Melioidosis). PLoS Med 2007;​4:​e248. 22. Wiersinga WJ, Wieland CW, Roelofs JJ, van der Poll T. MyD88 dependent signaling contributes to protective host defense against Burkholderia pseudomallei. PLoS One 2008;​3:​e3494. 23. Knapp S, Leemans JC, Florquin S, et al. Alveolar Macrophages Have a Protective Antiinflammatory Role during Murine Pneumococcal Pneumonia. Am J Respir Crit Care Med 2003;​167:​171‑9. 24. Wiersinga WJ, Kager LM, Hovius JW, et al. Urokinase receptor is necessary for bacterial defense against pneumonia-derived septic melioidosis by facilitating phagocytosis. J Immunol 2010;​184:​ 3079‑86. 25. Achouiti A, Vogl T, Urban CF, et al. Myeloid-related protein-14 contributes to protective immunity in gram-negative pneumonia derived sepsis. PLoS Pathog 2012;​8:​e1002987. 3 26. Rijneveld AW, Weijer S, Florquin S, et al. 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 2004;​103:​1702‑9. 27. van der Windt GJ, Blok DC, Hoogerwerf JJ, et al. Interleukin 1 receptor-associated kinase m im- pairs host defense during pneumococcal pneumonia. J Infect Dis 2012;​205:​1849‑57. 28. N’Diaye EN, Branda CS, Branda SS, et al. TREM-2 (triggering receptor expressed on myeloid cells 2) is a phagocytic receptor for bacteria. J Cell Biol 2009;​184:​215‑23. 29. Radsak MP, Salih HR, Rammensee HG, Schild H. Triggering receptor expressed on myeloid cells-1 in neutrophil inflammatory responses: differential regulation of activation and survival. J Immu- nol 2004;​172:​4956‑63. 30. Paterson GK, Mitchell TJ. Innate immunity and the pneumococcus. Microbiology 2006;​152:​ 285‑93. 31. van der Poll T, Opal SM. Pathogenesis, treatment, and prevention of pneumococcal pneumonia. Lancet 2009;​374:​1543‑56. 32. Aoki N, Zganiacz A, Margetts P, Xing Z. Differential regulation of DAP12 and molecules associated with DAP12 during host responses to mycobacterial infection. Infect Immun 2004;​72:​2477‑83. 33. McCormick S, Shaler CR, Small CL, et al. Control of pathogenic CD4 T cells and lethal immuno- pathology by signaling immunoadaptor DAP12 during influenza infection. J Immunol 2011;​187:​ 4280‑92. 34. Dockrell DH, Whyte MK, Mitchell TJ. Pneumococcal pneumonia: mechanisms of infection and resolution. Chest 2012;​142:​482‑91. 35. Hamerman JA, Jarjoura JR, Humphrey MB, Nakamura MC, Seaman WE, Lanier LL. Cutting edge: inhibition of TLR and FcR responses in macrophages by triggering receptor expressed on myeloid cells (TREM)-2 and DAP12. J Immunol 2006;​177:​2051‑5. 36. Rijneveld AW, Florquin S, Branger J, Speelman P, Van Deventer SJ, van der Poll T. TNF-alpha com- pensates for the impaired host defense of IL-1 type I receptor-deficient mice during pneumococ- cal pneumonia. J Immunol 2001;​167:​5240‑6. 37. Klesney-Tait J, Keck K, Li X, et al. Transepithelial migration of neutrophils into the lung requires TREM-1. J Clin Invest 2013;​123:​138‑49. 38. Divangahi M, Yang T, Kugathasan K, et al. Critical negative regulation of type 1 T cell immunity and immunopathology by signaling adaptor DAP12 during intracellular infection. J Immunol 2007;​179:​4015‑26.

59 Chapter 3

Supplement

Material and Methods

Mice dap12−/− mice were generated as previously described and backcrossed to a C57BL/6 ge- netic background1. Age and sex matched Wt C57BL/6 mice were purchased from Charles River (Maastricht, The Netherlands) and maintained at the animal care facility of the Academic Medical Centre (University of Amsterdam), according to national guidelines with free access to food and water. The Institutional Animal Care and Use Committee approved all animal experiments.

Cell culture and whole blood stimulation Alveolar macrophages from 8 individual dap12−/− and Wt mice were obtained as described elsewhere2. Cells were resuspended in RPMI containing 2 mM l-glutamine, penicillin-streptomycin and 10% fetal calf serum (FCS; Gibco). 5 × 104 cells per well were seeded in a 96-well flat-bottom plate in 100 µL to adhere overnight at 37°C, 5% CO2. Sub- sequently, cells were washed twice with phosphate buffered saline (PBS) to remove non- adherent cells. Heparinized whole blood was obtained from 8 individual dap12−/− and Wt mice in a separate experiment. Growth-arrested bacteria were prepared as described3. In brief, S. pneumoniae (American Type Culture Collection, ATCC 6303, Rockville, MD) were cultured and washed with pyrogen-free sterile saline and resuspended in sterile PBS to a concentration of 2 × 109 bacteria/ml. This S. pneumoniae preparation was treated for 1 hour at 37°C with 50 µg/ml Mitomycin C (Sigma-Aldrich, Zwijndrecht, the Netherlands) to prepare alive but growth-arrested bacteria. Subsequently, the growth-arrested S. pneumoniae preparation was washed twice in ice-cold sterile PBS by centrifugation at 4°C, and the final pellet was dispersed in ice-cold PBS in the initial volume and trans- ferred to sterile tubes. Undiluted samples of these preparations failed to generate any bacterial colonies when plated on blood agar plates, indicating successful growth arrest. Alveolar macrophages and whole blood were incubated for 24 hours with or without viable, growth-arrested S. pneumoniae at the indicated multiplicity of infection in RPMI containing 2mM l-glutamine, penicillin-streptomycin and 10% FCS (macrophages). Supernatants were taken and stored at −20°C until assayed for tumor necrosis factor (TNF)-α, interleukin (IL)-1-β, IL-6, Il-10 and keratinocyte-derived chemokine (KC).

Phagocytosis Phagocytosis of S. pneumoniae was determined in essence as described before4,5. Growth-arrested bacteria were labeled with carboxyfluorescein succinimidyl ester (CFSE, Invitrogen, Breda, the Netherlands). 100 µl heparinized whole blood was incubated with

60 DAP12 impairs host defense in pneumococcal pneumonia

10 µl bacteria in RPMI (end concentration of 1 × 107 bacteria/ml) at 37°C (n = 8 per group) or 4°C (n = 4 per group). After 60 minutes, samples were put on ice to stop phagocytosis.

Afterwards, red blood cells were lysed using isotonic NH4Cl solution (155 mM NH4Cl, 10 mM KHCO3, 100 mM EDTA, pH 7.4). Neutrophils were labeled using anti-Ly-6G-PE (clone 1A8, BD Pharmingen, San Diego, CA) and washed twice in FACS-buffer (0.5% BSA, 0.01%

NaN3, 0.35 mM EDTA in PBS) for analysis. Alveolar macrophages were pooled (derived from 8 mice per strain), washed twice and resuspended in RPMI containing 2 mM L- glutamine, penicillin-streptomycin and 10% FCS. 1 × 105 cells per well were seeded in a

96-well flat-bottom plate in 100 µL to adhere overnight at 37°C, 5% CO2. The following day, macrophages were washed with pre-warmed medium to wash away non-adherent cells. Growth-arrested bacteria were opsonized for 30 minutes at 37°C in 10% normal 3 mouse serum and washed twice in PBS before they were added to the cells at a mul- tiplicity of infection of 100 in a volume of 10 µL. Bacteria and macrophages were spun at 1000 RPM for 5 minutes and incubated at 37°C (n = 8 wells per strain) or 4°C (n = 4 wells per strain). After 1 hour, samples were washed with ice-cold PBS, then thoroughly scraped from the bottom and washed again in FACS-buffer. The degree of phagocytosis was determined using FACSCalibur (Becton Dickinson Immunocytometry, San Jose, CA) The phagocytosis index of each sample was calculated as follows: geometric mean fluorescence × % positive cells.

Experimental design Pneumonia was induced by intranasal inoculation with S. pneumoniae serotype 3 (American Type Culture Collection, ATCC 6303, Rockville, MD; 5 × 104 CFU) as described previously6,7. Mice were sacrificed 6, 24 or 48 hours thereafter (n = 7-8 per strain at each time point). Collection and handling of samples were done as previously described5-7. In brief, blood was drawn into heparinized tubes and organs were removed aseptically and homogenized in 4 volumes of sterile isotonic saline using a tissue homogenizer (Biospec Products, Bartlesville, UK). To determine bacterial loads, ten-fold dilutions were plated on blood agar plates and incubated at 37°C for 16 hours. In survival studies mice (n = 15 per strain) were intranasally inoculated with 5 × 104 or 5 × 103 CFU S. pneumoniae and monitored for up to 14 days after infection. mRNA expression analysis RNA was isolated from lung homogenates that were immediately dissolved in TRIzol (Invitrogen, Breda, the Netherlands), as described by the manufacturer and reverse transcribed using oligo dT (Invitrogen) and Moloney murine leukemia virus reverse transcriptase (Invitrogen). Reverse-transcription polymerase chain reactions were per- formed using LightCycler SYBR Green I Master Mix (Roche, Mijdrecht, the Netherlands) and measured in a LightCycler 480 (Roche) apparatus using the following conditions:

61 Chapter 3

5-minute 95°C hot-start, followed by 40 cycles of amplification (95°C for 10 seconds, 60°C for 5 seconds, 72°C for 15 seconds). For quantification, standard curves were con- structed by PCR on serial dilutions of a concentrated cDNA, and data were analyzed using LightCycler software. Gene expression is presented as a ratio of the expression of the housekeeping gene β2-microglobulin (B2M). Primers were as follows: B2M F: 5′-TG- GTCTTTCTGGTGCTTGTCT and R: 5′-ATTTTTTTCCCGTTCTTCAGC; dap12 F: 5′-CGAAAA- CAACACATTGCTGA and R: 5′-GGGCATAGAGTGGGCTCAT Trem1 F: 5′-GCGTCCCATCCT- TATTACCA and R: 5′-AAACCAGGCTCTTGCTGAGA; Trem2 F: 5′-ACCCACCTCCATTCTTCTCC and R: 5′-GGGTCCAGTGAGGATCTGAA.

Assays Lung homogenates were prepared for immune assays as described before5-7. In brief, lung homogenates were lysed in lysis buffer (300 mM NaCl, 15 mM Tris, 2 mM MgCl2, 1% Triton X-100, pepstatin A, leupeptin, and aprotinin (20 ng/mL); pH 7.4) for 30 minutes on ice and spun at 1500g at 4°C for 10 minutes; the supernatant was frozen at -20°C for cytokine measurement. TNF-α, interleukin (IL)-1-β, IL-6, Keratinocyte-derived chemo- kine (KC), macrophage inflammatory protein 2 (MIP-2) (all R&D systems, Minneapolis, MN) and myeloperoxidase (MPO; Hycult Biotechnology BV, Uden, the Netherlands) were measured using specific ELISAs according to manufacturers’ recommendations. IL-1β, IL-6, KC and IL-10 in supernatants were measured by cytometric bead array flex set assay (BD Biosciences, San Jose, CA) in accordance to the manufacturer’s instructions.

Histology Lung pathology scores were determined as described elsewhere5-7. In brief, lungs were harvested at the indicated time points, fixed in 10% buffered formalin, and embedded in paraffin. 4 µm sections were stained with haematoxylin and eosin (HE) and analyzed by a pathologist blinded for groups. To score lung inflammation and damage, the entire lung surface was analyzed with respect to the following parameters: bronchitis, edema, inter- stitial inflammation, intra-alveolar inflammation, pleuritis, endothelialitis and percentage of the lung surface demonstrating confluent inflammatory infiltrate (>10 fields). Each parameter was graded 0–4, with 0 being ‘absent’ and 4 being ‘severe’. The total pathol- ogy score was expressed as the sum of the score for all parameters (maximum score 24). Granulocyte staining was done using FITC-labeled rat anti-mouse Ly-6 mAb (Pharmingen, San Diego, CA) as described earlier8. Ly-6G expression in the lung tissue sections was quantified by digital image analysis9. In short, lung sections were scanned using the Olym- pus Slide system (Olympus, Tokyo, Japan) and TIF images, spanning the full tissue section were generated. In these images Ly-6G positivity and total surface area were measured using Image J (U.S. National Institutes of Health, Bethesda, MD, http://rsb.info.nih.gov/ij); the amount of Ly-6G positivity was expressed as a percentage of the total surface area.

62 DAP12 impairs host defense in pneumococcal pneumonia

References

1. Kaifu T, Nakahara J, Inui M, et al. Osteopetrosis and thalamic hypomyelinosis with synaptic degen- eration in DAP12-deficient mice. J Clin Invest 2003;​111:​323‑32. 2. Wiersinga WJ, Wieland CW, Dessing MC, et al. Toll-like receptor 2 impairs host defense in gram- negative sepsis caused by Burkholderia pseudomallei (Melioidosis). PLoS Med 2007;​4:​e248. 3. Wiersinga WJ, Wieland CW, Roelofs JJ, van der Poll T. MyD88 dependent signaling contributes to protective host defense against Burkholderia pseudomallei. PLoS One 2008;​3:​e3494. 4. Wiersinga WJ, Kager LM, Hovius JW, et al. Urokinase receptor is necessary for bacterial defense against pneumonia-derived septic melioidosis by facilitating phagocytosis. J Immunol 2010;​184:​ 3079‑86. 5. Achouiti A, Vogl T, Urban CF, et al. Myeloid-related protein-14 contributes to protective immunity in gram-negative pneumonia derived sepsis. PLoS Pathog 2012;​8:​e1002987. 6. Rijneveld AW, Weijer S, Florquin S, et al. Thrombomodulin mutant mice with a strongly reduced 3 capacity to generate activated protein C have an unaltered pulmonary immune response to respiratory pathogens and lipopolysaccharide. Blood 2004;​103:​1702‑9. 7. van der Windt GJ, Blok DC, Hoogerwerf JJ, et al. Interleukin 1 receptor-associated kinase m im- pairs host defense during pneumococcal pneumonia. J Infect Dis 2012;​205:​1849‑57. 8. Knapp S, Wieland CW, van ‘t Veer C, et al. 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 2004;​172:​3132‑8. 9. Lammers AJ, de Porto AP, Florquin S, et al. Enhanced vulnerability for Streptococcus pneumoniae sepsis during asplenia is determined by the bacterial capsule. Immunobiology 2011;​216:​863‑70.

63 Chapter 3

A IL-1 B IL-6 150 400 ** 300

l ** 100 l m m pg/

pg/ 200 6 - 1 IL IL 50 100

0 0 medium 105 106 107 medium 105 106 107 S. pneumoniae S. pneumoniae

C KC D IL-10 80 50

40 ** 60 l l m m 30 pg/

pg/ 40 10

- 20 KC KC IL 20 10

0 0 medium 105 106 107 medium 105 106 107 S. pneumoniae S. pneumoniae Supplemental figure 1: Increased cytokine and chemokine release by DAP12 deficient blood leu- kocytes in response to S. pneumoniae. Whole blood prepared from DAP12 deficient (dap12−/−) and wild type (Wt) mice was incubated with viable, growth-arrested S. pneumoniae for 24 hours (107/ml), after which IL-1b (A), IL-6 (B), KC (C) and IL-10 (D) were measured in supernatants. Data are expressed as median with interquartile ranges; **P<0.01 (versus Wt cells).

A DAP12 B TREM-1 C TREM-2 15 0.8 4 *** ** 0.6 3 10 *** o o o i i i t t 0.4 t 2 ra ra ra 5 *** 0.2 1

0 0.0 0 t=0 t=3 t=24 t=48 t=0 t=3 t=24 t=48 t=0 t=3 t=24 t=48 Supplemental figure2: Increased DAP12, TREM-1 and TREM-2 mRNA expression during pneumococ- cal pneumonia in lungs in vivo. Wild type mice (8 mice per group) were intranasally infected with viable S. pneumoniae and sacrificed 3, 24 and 48 hours later to measure mRNA transcripts in whole lung homog- enates. mRNA expression of DAP12 (A), TREM-1 (B) and TREM-2 (C) compared to uninfected mice (t=0) is presented as a ratio of the expression of the housekeeping gene β2-microglobulin. Data are expressed as median with interquartile ranges; **P < 0.01 ***P < 0.001.

64 Triggering Receptor Expressed on Myeloid Cells (TREM) -1 improves host defense during Staphylococcus aureus pneumonia

Tijmen J. Hommes1,2, Ahmed Achouiti1,2, Mark C. Dessing3, Sandrine Florquin3, Marco Colonna4, Cornelis van ’t Veer1,2, 4 Alex F. de Vos1,2, Tom van der Poll1,2,5 Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands: 1Center for Experimental and Molecular Medicine, 2Center for Infection and Immunity, 3Department of Pathology, 5Division of Infectious Diseases 4Washington University in St. Louis, St. Louis, Missouri, USA: Department of Pathology

submitted Chapter 4

Abstract

Background: Pneumonia caused by Staphyloccocus (S.) aureus is an upcoming clinical problem. Triggering Receptor Expressed on Myeloid cells (TREM)-1 is an activating receptor expressed by leukocytes known to potentiate innate immune responses. We here aimed to analyse the role of TREM-1 in the host response during pneumonia caused by S. aureus. Methods: Wild type (Wt) and TREM-1/3 deficient mice were intranasally inoculated with 107 S. aureus USA300. Mice were sacrificed at 6, 24, 48 or 72 hours after infection to harvest bronchoalveolar lavage (BAL) fluid, blood and organs to evaluate the host response. Responsiveness of macrophages isolated from TREM-1/3 deficient mice to S. aureus was tested in vitro. Results: Macrophages deficient for TREM-1/3 were less responsive to S. aureus in vitro. During staphylococcal pneumonia soluble TREM-1 levels increased in BAL fluid. TREM- 1/3 deficient mice demonstrated an impaired bacterial clearance from their lungs when compared to Wt mice. This was accompanied by reduced lung inflammation, as reflected by histopathology and cytokine levels, while neutrophil recruitment was not affected. Conclusion: These data suggest that TREM-1 contributes to protective immunity during S. aureus pneumonia.

66 TREM-1 improves host defense during Staphylococcus aureus pneumonia

Introduction

Staphylococcus (S.) aureus is a frequent colonizer of the human body that is able to cause a wide array of clinical syndromes ranging from minor skin infections to severe pneumo- nia and sepsis1-3. Infection of the lower respiratory tract by this Gram-positive pathogen is classically confined to health-care settings4. In recent years, however, the prevalence of staphylococcal pneumonia in the general population has increased, in particular due to the emergence of community associated methicillin-resistant S. aureus5. These highly virulent strains are able to cause life-threatening disease in otherwise healthy individu- als5,6. The increasing incidence of antibiotic resistance of this pathogen imposes a high burden on health care7. Therefore, more insight into the host defense mechanisms that influence the outcome of S. aureus infections will help the development of new – im- munomodulating – therapies. The family of TREM (Triggering Receptor Expressed on Myeloid cells) receptors has gained much attention as regulators of the immune response8-10. TREM receptors are 4 able to shape and fine-tune the host response to pathogens initiated by pathogen associated molecular patterns (PAMPs) via pattern recognition receptors (PRRs)8-11. In particular TREM-1, a membrane bound receptor expressed on neutrophils, monocytes and macrophages, is able to amplify signaling through Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD)-like Receptors (NLRs)12-14. Activation of TREM-1 results in the production and secretion of proinflammatory mediators via an intracellular pathway triggered by DNAX-activating protein of 12 kDa (DAP12)12,13. The inflammatory response to staphylococci is mainly initiated via the recognition of lipoteichoic acid (LTA) by TLR2, and of muramyl dipeptide by NOD23,15. Given that TREM-1 enhances signalling of both TLR2 and NOD214, we considered it plausible that TREM-1 might have a role in the host response to S. aureus. Indeed, TREM-1 expression is enhanced on human neutrophils and monocytes incubated with S. aureus13. Similarly, TREM-1 is upregulated on murine macrophages in response to S. aureus LTA in a TLR2 dependent manner16. In vivo, TREM-1 is highly expressed on neutrophils associated with skin lesions caused by S. aureus, such as folliculitis and impetigo13. Here we set out to study the role of TREM-1 in lower airway infection caused by S. aureus by making use of recently generated TREM-1/3 deficient (Trem-1/3−/−) mice17.

67 Chapter 4

Methods

Ethics statement Experiments were carried out in accordance with the Dutch Experiment on Animals Act and approved by the Animal Care and Use Committee of the University of Amsterdam (Permit number: DIX100121).

Mice Pathogen-free 8- to 10-week old male wild type (Wt) C57BL/6 mice were purchased from Charles River (Maastricht, The Netherlands). Trem-1/3−/− mice17 were backcrossed > 97% to a C57BL/6 genetic background and bred in the animal facility of the Academic Medi- cal Center (Amsterdam, the Netherlands).

Macrophage stimulation Bone marrow-derived macrophages (BMDMs) were generated using recombinant mouse granulocyte-macrophage colony stimulating factor (GM-CSF, Invitrogen, Breda, the Netherlands) as previously described18,19. Macrophages were pooled (derived from 3-4 mice), washed twice and resuspended in RPMI containing 2 mM L-glutamine, and 5% Panexin (cat#: P04-951SA2, PAN biotech, Aidenbach, Germany). BMDMs (1 × 105/well) from Trem-1/3−/− and Wt mice were resuspended in RPMI containing 2 mM l-glutamine, penicillin, streptomycin and 5% Panexin and were plated in a 96-well flat-bottom plate and allowed to adhere for 16 hours at 37°C in 5% CO2. Subsequently, the cells were washed with PBS to remove nonadherent cells. Next, the adherent cells were stimulated for 24 hours with or without the indicated multiplicity of infection (MOI) of heat-killed

S. aureus at 37°C in 5% CO2. Supernatants were harvested and stored at −20°C until as- sayed for cytokines and chemokines.

Experimental design This model of S. aureus pneumonia was described in detail previously20,21. Mice were lightly anesthesized by inhalation of isoflurane (Abbot Laboratories, Queensborough, Kent, UK) and intranasally inoculated with a sub-lethal dose of 1 × 107 S. aureus USA 300 (BK 11540) in a 50 μl saline solution (n = 7-8 per strain). Mice were sacrificed 6, 24, 48 or 72 hours thereafter20.

Bacterial cultures Collection and handling of samples were done as previously described19,20. In brief, blood was drawn into heparinized tubes, bronchoalveolar lavage fluid (BAL fluid, see below) was obtained and livers were removed aseptically and homogenised in 4 volumes of sterile isotonic saline using a tissue homogenizer (Biospec Products, Bartlesville, UK). To

68 TREM-1 improves host defense during Staphylococcus aureus pneumonia determine bacterial loads, ten-fold dilutions were plated on blood agar (BA) plates and incubated at 37°C for 16 h.

Bronchoalveolar lavage The trachea and bronchi were exposed through a midline incision. The left main bron- chus was ligated and the trachea was cannulated with a sterile 22-gauge Abbocath-T catheter (Abbott Laboratories, Sligo, Ireland). Unilateral, right-sided BAL was performed by instilling three 0.3 ml aliquots of sterile phosphate buffered saline (PBS). 0.7-0.9 ml of BAL fluid was retrieved per mouse. Total cell numbers in BAL fluid were counted using a Z2 Coulter particle count and size analyzer (Beckman-Coulter, Inc., Miami, FL). BAL fluid differential cell counts were carried out on cytospin preparations stained with modified Giemsa stain (Diff-Quick; Baxter, McGraw Park, IL).

Assays Soluble (s)TREM-1, tumor necrosis factor (TNF)-α, interleukin (IL)-1β, IL-6 and macro- 4 phage inflammatory protein 2 (MIP-2)(all R&D systems, Minneapolis, MN) were measured using specific ELISAs according to manufacturer’s recommendations.

Histology Lung and pathology scores were determined as described before19,20. In brief, lungs were harvested at the indicated time points, fixed in 10% buffered formalin, and embedded in paraffin. Four µm sections were stained with haematoxylin and eosin (HE) and analyzed by a pathologist blinded for groups. To score lung inflammation and damage, the entire lung surface was analyzed with respect to the following parameters: bronchitis, edema, interstitial inflammation, intra-alveolar inflammation, pleuritis, endothelialitis and percentage of the lung surface demonstrating confluent inflammatory infiltrate. Each parameter was graded 0–4, with 0 being ‘absent’ and 4 being ‘severe’. The total pathol- ogy score was expressed as the sum of the score for all parameters.

Statistical analysis Data are expressed as scatter plots with medians or as mean ± SEM. Differences between Trem-1/3−/− and Wt mice were analyzed by Mann-Whitney U test. Analyses were done using GraphPad Prism version 5.0, Graphpad Software (San Diego, CA). Values of P < 0.05 were considered statistically significant different.

69 Chapter 4

Results

TREM-1/3 deficiency results in reduced responsiveness of macrophages to S. aureus in vitro TREM-1 has been shown to amplify TLR and NLR signaling in leukocytes12-14. To obtain a first insight in the role of TREM-1 in host defense against S. aureus, we tested the responsiveness of macrophages derived from Trem-1/3−/− and Wt mice to S. aureus in vitro. To this end we generated BMDMs from both genotypes and incubated these with staphylococci for 24 hours and measured TNF-α, IL-1β, IL-6 and MIP-2 in supernatants by ELISA. Trem-1/3−/− BMDMs produced significantly less TNF-α, IL-6 and MIP-2 after 24 hours of incubation with S. aureus (Figure 1; P < 0.05 or P < 0.01); IL-1β remained unde- tectable in all supernatants.

A B C 15000 TNF-α 5000 IL-6 10000 MIP-2 Wt -/- Trem-1/3 4000 8000 10000 l l 3000 l 6000 m m m * pg/ pg/ pg/ 2000 * 4000 5000 ** 1000 2000 * 0 0 0 medium 1:10 1:100 medium 1:10 1:100 medium 1:10 1:100 S. aureus S. aureus S. aureus Figure 1: TREM-1/3 deficiency results in lower cytokine and chemokine production by macrophages in vitro. Bone marrow-derived macrophages (BMDMs) from wild type (Wt) and Trem-1/3−/− mice were incu- bated with medium (M) or heat-killed S. aureus (MOI 1:10 and 1:100) for 24 h and levels of TNFα (A), IL-6 (B) and MIP-2 (C) were measured in supernatants. Representative figures of two independent experiments are shown (n = 4–8) and are expressed as mean ± SEM; *P < 0.05; **P < 0.01 versus Wt cells. sTREM-1 levels increase in BAL fluid during staphylococcal pneumonia sTREM-1 concentrations in BAL fluid have been used to identify patients with bacterial pneumonia22. To obtain insight in sTREM-1 release in the airways during staphylococcal pneumonia, Wt mice were intranasally inoculated with a sub-lethal dose of 107 S. aureus and sTREM-1 levels were measured in BAL fluid before and 6, 24, 48 and 72 hours after infection. sTREM-1 was detectable in BAL fluid; following infection BAL fluid sTREM-1 concentrations increased strongly, peaking at 6 hours post-infection (Figure 2; P < 0.01 for 6 and 24 hours compared to t = 0).

TREM-1/3 deficiency impairs host defense during staphylococcal pneumonia To investigate the functional role of TREM-1 in host defense during staphylococcal pneumonia, Trem-1/3−/− and Wt mice were infected intranasally with 107 viable S. aureus and BAL fluid and lungs were harvested after 6, 24, 48 and 72 hours for quantitative cultures. Bacterial loads were markedly higher in BAL fluid of Trem-1/3−/− mice 6 and 48 hours after infection (Figure 3A; P < 0.001 6h and P < 0.01 48h). Whole lungs of Trem-

70 TREM-1 improves host defense during Staphylococcus aureus pneumonia

1 500 **

l BALF l 1 000 m pg/ 1

M- 500 ** E R T s 0 0h 6h 24h 48h 72h

Figure 2: sTREM-1 levels increase in BAL fluid during infection with S. aureus. Soluble TREM-1 levels in bronchoalveolar lavage fluid (BALF) were determined in naive mice and 6, 24, 48 and 72 hours after intra- nasal inoculation with 107 CFU of S. aureus. Data are mean ± SEM values from n = 8 mice group at each time point; **P < 0.01 versus t = 0 h.

A B BALF Lung Wt 9 8 -/- Trem-1/3 7 *** 8 ** **

l l 4 6 7 m m / / 6 U U 5 F F ** 5 ** C C 4 4 log log log log 3 3 ** 10 10 2 2 1 1 0 0 6h 24h 48h 72h 6h 24h 48h 72h Figure 3: TREM-1/3 deficiency results in enhanced bacterial outgrowth locally during staphylococ- cal pneumonia. Wild type (Wt) and Trem-1/3−/− mice (n = 8 per group) were intranasally infected with 107 CFU S. aureus and sacrificed 6, 24, 48 or 72 hours later. Bacterial loads were determined in bronchoalveolar lavage fluid (BALF; A) and lung homogenates (B). Horizontal lines represent medians; *P < 0.05; **P < 0.01; ***p < 0.01 versus Wt mice.

1/3−/− mice contained significantly higher numbers of S. aureus at all time points (Figure 3B; P < 0.01). In accordance with our previous studies20,21, virtually no dissemination of bacteria to distant body sites was observed; no differences in bacterial loads in blood or livers were seen between experimental groups (data not shown). These data indicate that TREM-1/3 contributes to bacterial control locally in the lung during pulmonary infection with S. aureus.

TREM-1/3 deficiency reduces pulmonary inflammation during staphylococcal pneumonia Pneumonia caused by S. aureus results in local inflammation and inflammatory cell recruitment, which is an integral part of the host immune response23. Considering the role of TREM-1 as an amplifier of inflammation, we assessed pulmonary histopathology

71 Chapter 4 of Trem-1/3−/− and Wt mice during staphylococcal pneumonia using a semi-quantitative scoring system described in the Methods section. Already at 6 hours, all mice displayed signs of severe pulmonary inflammation, although without differences between mouse strains. The highest pathology scores were found at 24 hours in both Trem-1/3−/− and Wt mice (Figure 4). Thereafter, the extent of lung pathology decreased in Trem-1/3−/− but not in Wt mice; indeed, at 48 and 72 hours of infection, Trem-1/3−/− mice displayed reduced

A Pathology score B % Confluent infiltrates

25 Wt 50 Trem-1/3 -/- 20 40 15 * * 30 % 10 20 * *

5 10 ND 0 0 6h 24h 48h 72h 6h 24h 48h 72h C D -/- Wt (48h) Trem-1/3 (48h)

E F

-/- Wt (72h) Trem-1/3 (72h)

Figure 4: TREM-1/3 deficiency results in reduced pulmonary pathology during staphylococcal pneu- monia. Wild type (Wt) and Trem-1/3−/− mice (n = 8 per group) were intranasally infected with 107 CFU S. aureus and sacrificed 6, 24, 48 or 72 hours later. Quantitative lung pathology score (A) was determined by the scoring system described in the method section. Area of confluent inflammation expressed as percent- age of total lung surface (B). Representative haematoxylin and eosin (HE) stainings of lung tissue of Wt (C,E) and Trem-1/3−/− (D,F) lungs 48h (C,D) and 72h (E,F)after inoculation with S. aureus (original magnification= 200×). Data are expressed as mean ± SEM; *P < 0.05 versus Wt mice.

72 TREM-1 improves host defense during Staphylococcus aureus pneumonia endothelialitis, bronchitis, oedema and pleuritis (Figure 4C-D for 48h and E-F for 72h; P < 0.05 for both 48 and 72 hours). Similarly, Trem-1/3−/− mice had smaller surface areas of confluent inflammation at these time points (Figure 4B; P < 0.05 for both 48 and 72 hours).

TREM-1/3 deficiency reduces pulmonary cytokine and chemokine levels during staphylococcal pneumonia TREM-1 amplifies the release of proinflammatory mediators12,13 (and Figure 1). To study the impact of TREM-1 deficiency on cytokine and chemokine release in the airways during lower respiratory tract infection caused by S. aureus, we measured TNF-α, IL-1β, IL-6 and MIP-2 in BAL fluid recovered from Trem-1/3−/− and Wt mice after induction of pneumonia. All cytokines and chemokines reached peak concentrations 6 hours after inoculation. At this time point we found strikingly lower levels of TNF-α and IL-6 in BAL recovered from Trem-1/3−/− mice (Figure 5A and C; P < 0.01). Similarly, after 24 hours we found lower levels of TNF-α, IL-1β, and MIP-2 in BAL recovered from Trem-1/3−/− mice 4 (Figure 5A, B and D; P < 0.01). Cytokine and chemokine decreased gradually during

A B 100 TNF-α 1.5 IL-1β Wt 80 Trem-1/3 -/- 1.0 l 60 l m m ng/ 40 ng/ ** 0.5 20 ** ** 0 0.0 6h 24h 48h 72h 6h 24h 48h 72h C D 40 IL-6 10 MIP-2

8 30 l l 6 m 20 m ng/ ng/ 4 10 ** 2

0 0 ** 6h 24h 48h 72h 6h 24h 48h 72h Figure 5: Reduced local cytokine and chemokine levels during staphylococcal pneumonia in TREM- 1/3 deficient mice. Wild type (Wt) and Trem-1/3−/− mice (n = 8 per group) were intranasally infected with 107 CFU S. aureus and sacrificed 6, 24, 48 or 72 hours later. Pulmonary cytokine and chemokine levels were determined in bronchoalveolar lavage fluid (BALF) by ELISA. Data are expressed as mean ± SEM; *P < 0.05; **P < 0.01 versus Wt mice.

73 Chapter 4 pulmonary infection with S. aureus and were hardly detectable beyond 48 hours after infection.

TREM-1/3 deficiency does not impact on neutrophil influx into lungs during staphylococcal pneumonia A hallmark of innate host defense during bacterial pneumonia is the influx of neutro- phils into the lung24. Previous studies have described an important role for neutrophils in innate defense during staphylococcal airway infection23. To study the impact of TREM- 1/3 deficiency in our model of staphylococcal pneumonia we determined the extent of leukocyte influx and the cellular composition thereof in BAL fluid from Trem-1/3−/− and Wt mice at different time points during S. aureus pneumonia. No difference in total BAL cells at any of the time points after induction of staphylococcal pneumonia between Trem-1/3−/− and Wt mice was detected (Figure 6A). Neither did we find a difference in alveolar recruited neutrophils between both strains during infection with S. aureus (Figure 6B).

A B Total cells Neutrophils 10 Wt 10 Trem-1/3 -/- l 8 l 8 m m / / s s ll 6 ll 6 ce ce

5 5 E 4 E 4 10 10

x 2 x 2

0 0 6h 24h 48h 72h 6h 24h 48h 72h

Figure 6: Alveolar recruitment of neutrophils is not influenced by TREM-1/3 deficiency during staphy- lococcal pneumonia. Wild type (Wt) and Trem-1/3−/− mice (n = 8 per group) were intranasally infected with 107 CFU S. aureus and sacrificed 6, 24, 48 or 72 hours later. Total number of leukocytes (A) and neutrophils (B) in bronchoalveolar lavage fluid (BALF) of Wt and Trem-1/3−/− mice. Data are expressed as mean ± SEM.

Discussion

The emergence of virulent methicillin-resistant S. aureus strains like USA300 in the general population is a cause for major concern5,6,25. Although mainly causing soft tissue and skin infections these strains are able to cause fatal diseases such as necro- tizing pneumonia5,26,27. Pulmonary inflammation due to S. aureus USA300 is initially caused by a number of virulence factors but may be exaggerated by the host immune response6. A clear understanding of host pathogen interactions is critical to secure the development of new strategies to control S. aureus infections in the future. In this light

74 TREM-1 improves host defense during Staphylococcus aureus pneumonia we were interested in the contribution of TREM-1, a receptor previously implicated in inflammation in different types of infection12-14. While earlier investigations documented involvement of TREM-1 in cellular activation by S. aureus in vitro13,16, we here for the first time studied the role of this receptor in the host response during pneumonia caused by S. aureus in vivo. Our main finding is that TREM-1/3 deficiency impaired host defense, as reflected by increased bacterial loads in the pulmonary department during staphylococ- cal pneumonia, most likely due to attenuated early proinflammatory cytokine release in the airways. Similar to previous studies assessing the role of TREM-1 in infectious disease17,19, we used mice deficient for both TREM-1 and TREM-3 to investigate the role of TREM-1 in S. aureus-induced pneumonia. In mice, TREM-3 is highly homologous to TREM-1 and both receptors have a similar cellular distribution. TREM-3, like TREM-1, is an activat- ing receptor on mouse macrophages also signaling through DAP1228. In contrast, in humans Trem3 is a pseudogene. In order to approach the human situation, we used a TREM-1/3 double knockout mouse to obtain insight into the role of TREM-1 in staphy- 4 lococcal pneumonia. We have recently shown that macrophages lacking TREM-1/3 are less responsive towards S. pneumoniae in vitro19. In line with this, TREM-1/3 deficient macrophages were less responsive towards S. aureus compared to Wt cells, confirming and extending earlier reports12,13,19. TREM-1 is constitutively expressed on local and peripheral myeloid cells and is up- regulated upon exposure to microbes or microbial components12,13,16,19,29. In addition, myeloid cells are able to release a soluble form of TREM-1 into the (local) environment, which is highly correlated to active infection22,30. Indeed, sTREM-1 concentrations in BAL fluid may serve as a biomarker that indicates the presence of pneumonia in patients22,30. In accordance, we found strongly increased sTREM-1 levels in BAL fluid of mice with staphylococcal pneumonia. Clearly, this response is not limited to S. aureus infection, considering previous findings of elevated sTREM-1 levels in BAL fluid of mice with respi- ratory tract infection caused by S. pneumoniae19,31 or Burkholderia pseudomallei29. The current results, showing increased bacterial loads in the lungs of Trem-1/3−/− mice, are in agreement with previous studies assessing the role of TREM-1 during pneumonia caused by S. pneumoniae or P. aeruginosa17,19,31. The underlying mechanism responsible for the beneficial role of TREM-1 likely depends on the pathogen. For example, during pneumonia caused by P. aeruginosa the observed phenotype was explained by impaired migration of TREM-1/3 deficient neutrophils across the pulmonary epithelial barrier17. However, in the current study the number of neutrophils recruited to the alveoli did not differ between Trem-1/3−/− and Wt mice. Our previous work demonstrated impaired early local immune response in Trem-1/3−/− mice allowing for increased bacterial replica- tion and dissemination during pneumococcal pneumonia19. Similarly, we here found decreased pulmonary inflammation reflected by lower pathology scores and reduced

75 Chapter 4 levels of pulmonary inflammatory mediators during staphylococcal pneumonia. Accord- ingly, a recent study reported that TREM-1 deficiency led to attenuated inflammation in parasitic, viral and bacterial infection, although pathogen clearance was not affected; similar to our current study no differences in neutrophil influx were observed in lungs32. One might speculate that differences in pathogens, infectious dose, route and kinetics of the infection, and the variable release of PAMPs/DAMPs allow for differential roles of TREM-1 in the different infection models reported to date. TREM-1 has been shown to amplify signalling through different PRRs including TLR2, TLR4, TLR9 and NOD212,14. In the context of the host defense against S. aureus, TLR2 is of particular importance33. Indeed Tlr2−/− macrophages are less responsive to LTA and heat killed S. aureus33,34, corresponding to reduced cytokine release and enhanced lethality during S. aureus sepsis induced by intravenous infection33,35. Tlr2−/− mice also displayed an impaired defense after cutaneous infection with this pathogen36. While a recent study described increased lethality of Tlr2−/− mice after infection with S. aureus via the airways, data on bacterial clearance or induction of inflammation were not reported37. Notably, deficiency of the common TLR adaptor MyD88 did not influence bacterial clearance dur- ing S. aureus pneumonia despite blunted local inflammation in the lungs38. In contrast, TLR9 impaired rather than facilitated host defense during staphylococcal pneumonia39, while TLR4 did not contribute to a significant extent20. NOD2 contributed to lung inflammation caused by S. aureus pneumonia, as reflected by less severe lung pathol- ogy, diminished recruitment of neutrophils, and reduced cytokine levels in BAL fluid in Nod2−/− mice40. Bacterial loads in lung tissue were not affected by NOD2 deficiency during staphylococcal pneumonia, while bacterial numbers in BAL fluid were modestly reduced in Nod2−/− mice at a single time point (day 2 after infection)40. Together these data suggest that TREM-1 modifies the host response to S. aureus in the airways in part by amplification of TLR2 and NOD2 signalling. TREM-1 has emerged as an important player in innate immunity by virtue of its capac- ity to amplify proinflammatory signalling by various PRRs. While earlier investigations have suggested that TREM-1 may be a therapeutic target in severe bacterial infection and sepsis11, the current study adds to recent reports that TREM-1 assists in mounting a beneficial host response during respiratory tract infection17,19. These results exemplify the double-edged sword character of innate immunity, which on the one hand is essen- tial for an adequate response to invading bacteria at the primary site of infection, and on the other hand can cause collateral tissue damage during exaggerated inflammation elicited by ongoing bacterial multiplication. The data presented provide new insight in the pathophysiology of staphylococcal pneumonia and contribute to the understand- ing of the complex role of TREM-1 in innate immunity.

76 TREM-1 improves host defense during Staphylococcus aureus pneumonia

Acknowledgements

The authors thank Marieke ten Brink and Joost Daalhuisen for their expert technical assistance during the animal experiments, Anne-Jan van der Meer for his assistance in processing the experimental samples and Regina de Beer for performing histopatho- logical stainings.

4

77 Chapter 4

References

1. Lowy FD. Staphylococcus aureus infections. N Engl J Med 1998;​339:​520‑32. 2. Clements MO, Foster SJ. Stress resistance in Staphylococcus aureus. Trends Microbiol 1999;​7:​ 458‑62. 3. Fournier B, Philpott DJ. Recognition of Staphylococcus aureus by the innate immune system. Clin Microbiol Rev 2005;​18:​521‑40. 4. American Thoracic S, Infectious Diseases Society of A. Guidelines for the management of adults with hospital-acquired, ventilator-associated, and healthcare-associated pneumonia. Am J Respir Crit Care Med 2005;​171:​388‑416. 5. Francis JS, Doherty MC, Lopatin U, et al. Severe community-onset pneumonia in healthy adults caused by methicillin-resistant Staphylococcus aureus carrying the Panton-Valentine leukocidin genes. Clin Infect Dis 2005;​40:​100‑7. 6. Parker D, Prince A. Immunopathogenesis of Staphylococcus aureus pulmonary infection. Semin Immunopathol 2012;​34:​281‑97. 7. Boucher HW, Corey GR. Epidemiology of methicillin-resistant Staphylococcus aureus. Clin Infect Dis 2008;​46 Suppl 5:​S344‑9. 8. Klesney-Tait J, Turnbull IR, Colonna M. The TREM receptor family and signal integration. Nat Im- munol 2006;​7:​1266‑73. 9. Sharif O, Knapp S. From expression to signaling: roles of TREM-1 and TREM-2 in innate immunity and bacterial infection. Immunobiology 2008;​213:​701‑13. 10. Ford JW, McVicar DW. TREM and TREM-like receptors in inflammation and disease. Curr Opin Im- munol 2009;​21:​38‑46. 11. Derive M, Massin F, Gibot S. Triggering receptor expressed on myeloid cells-1 as a new therapeutic target during inflammatory diseases. Self Nonself 2010;​1:​225‑30. 12. Bouchon A, Dietrich J, Colonna M. Cutting edge: inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J Immunol 2000;​164:​4991‑5. 13. Bouchon A, Facchetti F, Weigand MA, Colonna M. TREM-1 amplifies inflammation and is a crucial mediator of septic shock. Nature 2001;​410:​1103‑7. 14. Netea MG, Azam T, Ferwerda G, Girardin SE, Kim SH, Dinarello CA. Triggering receptor expressed on myeloid cells-1 (TREM-1) amplifies the signals induced by the NACHT-LRR (NLR) pattern recog- nition receptors. J Leukoc Biol 2006;​80:​1454‑61. 15. Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011;​34:​637‑50. 16. Zheng H, Heiderscheidt CA, Joo M, et al. MYD88-dependent and -independent activation of TREM-1 via specific TLR ligands. Eur J Immunol 2010;​40:​162‑71. 17. Klesney-Tait J, Keck K, Li X, et al. Transepithelial migration of neutrophils into the lung requires TREM-1. J Clin Invest 2013;​123:​138‑49. 18. Eske K, Breitbach K, Kohler J, Wongprompitak P, Steinmetz I. Generation of murine bone marrow derived macrophages in a standardised serum-free cell culture system. J Immunol Methods 2009;​ 342:​13‑9. 19. Hommes TJ, Hoogendijk AJ, Dessing MC, et al. Triggering receptor expressed on myeloid cells-1 (TREM-1) improves host defence in pneumococcal pneumonia. J Pathol 2014;​233:​357‑67. 20. Achouiti A, van der Meer AJ, Florquin S, et al. High-mobility group box 1 and the receptor for advanced glycation end products contribute to lung injury during Staphylococcus aureus pneu- monia. Crit Care 2013;​17:​R296.

78 TREM-1 improves host defense during Staphylococcus aureus pneumonia

21. Achouiti A, Vogl T, van der Meer AJ, et al. Myeloid-related protein-14 deficiency promotes pulmo- nary inflammation in Staphylococcus aureus pneumonia. European Respiratory Journal. 22. Gibot S, Cravoisy A, Levy B, Bene MC, Faure G, Bollaert PE. Soluble triggering receptor expressed on myeloid cells and the diagnosis of pneumonia. N Engl J Med 2004;​350:​451‑8. 23. Bartlett AH, Foster TJ, Hayashida A, Park PW. Alpha-toxin facilitates the generation of CXC chemo- kine gradients and stimulates neutrophil homing in Staphylococcus aureus pneumonia. J Infect Dis 2008;​198:​1529‑35. 24. Mizgerd JP. Acute lower respiratory tract infection. N Engl J Med 2008;​358:​716‑27. 25. Chambers HF, Deleo FR. Waves of resistance: Staphylococcus aureus in the antibiotic era. Nat Rev Microbiol 2009;​7:​629‑41. 26. Gillet Y, Issartel B, Vanhems P, et al. Association between Staphylococcus aureus strains carrying gene for Panton-Valentine leukocidin and highly lethal necrotising pneumonia in young immu- nocompetent patients. Lancet 2002;​359:​753‑9. 27. Centers for Disease C, Prevention. Severe coinfection with seasonal influenza A (H3N2) virus and Staphylococcus aureus--Maryland, February-March 2012. MMWR Morb Mortal Wkly Rep 2012;​61:​ 289‑91. 28. Chung DH, Seaman WE, Daws MR. Characterization of TREM-3, an activating receptor on mouse macrophages: definition of a family of single Ig domain receptors on mouse chromosome 17. Eur 4 J Immunol 2002;​32:​59‑66. 29. Wiersinga WJ, Veer Cvt, Wieland CW, et al. Expression Profile and Function of Triggering Receptor Expressed on Myeloid Cells-1 during Melioidosis. Journal of Infectious Diseases 2007;​196:​1707‑16. 30. Determann RM, Millo JL, Gibot S, et al. Serial changes in soluble triggering receptor expressed on myeloid cells in the lung during development of ventilator-associated pneumonia. Intensive Care Med 2005;​31:​1495‑500. 31. Lagler H, Sharif O, Haslinger I, et al. TREM-1 activation alters the dynamics of pulmonary IRAK-M expression in vivo and improves host defense during pneumococcal pneumonia. J Immunol 2009;​183:​2027‑36. 32. Weber B, Schuster S, Zysset D, et al. TREM-1 deficiency can attenuate disease severity without affecting pathogen clearance. PLoS Pathog 2014;​10:​e1003900. 33. Takeuchi O, Hoshino K, Akira S. Cutting edge: TLR2-deficient and MyD88-deficient mice are highly susceptible to Staphylococcus aureus infection. J Immunol 2000;​165:​5392‑6. 34. Takeuchi O, Hoshino K, Kawai T, et al. Differential roles of TLR2 and TLR4 in recognition of gram- negative and gram-positive bacterial cell wall components. Immunity 1999;​11:​443‑51. 35. Yimin, Kohanawa M, Zhao S, et al. Contribution of Toll-Like Receptor 2 to the Innate Response against Staphylococcus aureus Infection in Mice. PLoS One 2013;​8:​e74287. 36. Hoebe K, Georgel P, Rutschmann S, et al. CD36 is a sensor of diacylglycerides. Nature 2005;​433:​523‑7. 37. Blanchet C, Jouvion G, Fitting C, Cavaillon JM, Adib-Conquy M. Protective or deleterious role of scavenger receptors SR-A and CD36 on host resistance to Staphylococcus aureus depends on the site of infection. PLoS One 2014;​9:​e87927. 38. Skerrett SJ, Liggitt HD, Hajjar AM, Wilson CB. Cutting edge: myeloid differentiation factor 88 is essential for pulmonary host defense against Pseudomonas aeruginosa but not Staphylococcus aureus. J Immunol 2004;​172:​3377‑81. 39. Parker D, Prince A. Staphylococcus aureus induces type I IFN signaling in dendritic cells via TLR9. J Immunol 2012;​189:​4040‑6. 40. Kapetanovic R, Jouvion G, Fitting C, et al. Contribution of NOD2 to lung inflammation during Staphylococcus aureus-induced pneumonia. Microbes Infect 2010;​12:​759‑67.

79

TREM-1/3 contributes to protective immunity in Klebsiella derived pneumosepsis whereas TREM-2 does not

Tijmen J. Hommes1,2, Mark C. Dessing3, Cornelis van ’t Veer1,2, Sandrine Florquin3, Marco Colonna4, Alex F. de Vos1,2, 5 Tom van der Poll1,2,5 Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands: 1Center for Experimental and Molecular Medicine, 2Center for Infection and Immunity, 3Department of Pathology, 5Division of Infectious Diseases 4Washington University in St. Louis, St. Louis, Missouri, USA: Department of Pathology

Am J Respir Cell Mol Biol. 2015 Apr 10. [Epub ahead of print] Chapter 5

Abstract

Triggering Receptor Expressed on Myeloid cells (TREM)-1 and -2 can affect Toll-like receptor mediated activation of immune cells. Klebsiella (K.) pneumoniae is a common cause of pneumonia-derived sepsis. Here we studied the role of TREM-1/3 and TREM-2 in the host response during Klebsiella pneumonia. Macrophages lacking either TREM- 1/3 or TREM-2 were tested for their responsiveness towards K. pneumoniae as well as their capacity to internalize this pathogen in vitro. TREM-1/3 and TREM-2 deficient mice were infected with K. pneumoniae via the airways and their responses were compared with those in wild type mice. TREM-1/3 deficient macrophages produced lower cytokine levels upon exposure to K. pneumoniae, while on the opposite TREM-2 deficient mac- rophages released higher cytokine concentrations. TREM-2 deficient, but not TREM-1/3 deficient, macrophages showed a reduced capacity to phagocytose K. pneumoniae. TREM-1/3 deficient mice showed an impaired host defense duringKlebsiella pneumonia, as reflected by worsened survival and increased bacterial growth and dissemination. In contrast, TREM-2 deficiency did not impact on disease outcome. Although TREM-1/3 and TREM-2 influence macrophage responsiveness to K. pneumoniae in vitro, only TREM-1/3 contribute to the host response during Klebsiella pneumonia in vivo, serving a protective role.

82 TREM-1/3 contributes to protective immunity in Klebsiella derived pneumosepsis whereas TREM-2 does not

Introduction

The immune system detects invading pathogens through the recognition of structur- ally conserved microbial molecules by Toll-like receptors (TLRs) and NOD-like receptors (NLRs), which activate innate immune cells to clear the infection and shape the adaptive immune response1. Fine-tuning of this response is critical to prevent excessive inflam- mation and tissue damage2,3. TREM-receptors (‘Triggering Receptor Expressed on My- eloid cells’) are a structurally related family of cell surface receptors expressed broadly on a variety of innate cells of the myeloid lineage including neutrophils, monocytes, macrophages and dendritic cells, and are able to modulate cellular responses4,5. TREM-1 was the first TREM protein identified and has been shown to strongly potenti- ate the activation of leukocytes by enhancing TLR and NLR signaling6-8. TREM-1 pairs with signaling adaptor molecule DNAX activating Protein of 12 kDa (DAP12), which con- tains multiple immunoreceptor tyrosine-based activation motifs (ITAMs)6. Engagement of TREM-1 results in the tyrosine phosphorylation of the ITAMs within DAP12 and the subsequent release of pro-inflammatory cytokines through phosphorylation of MAP ki- nases and NFκB translocation6. TREM-2 is primarily expressed on macrophages, dendritic cells, osteoclasts and microglia9-11. Although TREM-2 also activates DAP12, this receptor negatively regulates TLR signaling in macrophages and dendritic cells via downstream 5 signaling pathways less well defined as for TREM-112,13. Alongside of its inhibiting effect on leukocytes, TREM-2 can bind directly to bacteria and serve as a phagocytic recep- tor14,15. Clinically, TREM-2 mutations have been associated with Nasu-Hakola syndrome11 and more recently polymorphisms in the Trem2 gene have been linked with Alzheimer’s disease16,17. Several studies have examined the role of TREM-1 in severe infection. Inhibition of TREM-1 signaling through the administration of soluble TREM-17, a synthetic TREM-1 antagonist (LP17)18 or small interference RNA duplexes19 improved survival in experi- mental models of abdominal sepsis in mice at least in part by attenuating exaggerated inflammatory responses. Similarly, administration of LP17 diminished lung inflammation and reduced mortality in acute pneumonia caused by Pseudomonas (P.) aeruginosa20. However, we and others recently reported that TREM-1 contributes to protective im- munity during pneumonia induced by either P. aeruginosa21 or Streptococcus (S.) pneu- moniae in mice22, suggesting that the inflammation amplifying function of this receptor can either harm or protect the host during infection depending on the type and severity of the bacterial challenge. Knowledge of the function of TREM-2 in host defense against infection is more limited. A recent study reported a protective role for TREM-2 in poly- microbial sepsis induced by cecal ligation and puncture by a mechanism that involved improved bacterial clearance23. In contrast, TREM-2 was found to impair host defense during pneumonia caused by S. pneumoniae24.

83 Chapter 5

Klebsiella (K.) pneumoniae is a frequent cause of pneumonia25 and the second most common cause of Gram-negative sepsis26,27. This pathogen clearly differs from previously studied bacteria in the context of TREMs. Indeed, while Pseudomonas causes an hyper- acute inflammation of the airways,Klebsiella gradually grows in the lungs, subsequently disseminating to distant organs, and different TLRs contribute to host defense; similarly, TLRs involved in the recognition of S. pneumoniae and K. pneumoniae in the airways are different28. Therefore, we here aimed to study the roles of TREM-1 and TREM-2 in the host response during severe pneumonia derived sepsis caused by K. pneumoniae. To study the role of TREM-1 we used a recently described TREM-1/3 deficient mouse strain21. Like TREM-1, TREM-3 is an activating receptor in murine macrophages signaling through DAP12 and exists as a pseudo-gene in humans29. The Trem3 gene likely arose from a duplication event during evolution and is located adjacent to the Trem1 gene29. Hence, to model the human situation, a mouse deficient for both TREM-1 and TREM-3 was used.

Material & Methods

Mice TREM-1/3 (Trem1/3−/−) and TREM-2 deficient (Trem2−/−) mice were generated as previ- ously described and backcrossed at least six times to a C57BL/6 genetic background21,30. Age and sex matched wild type (Wt) C57BL/6 mice were purchased from Charles River (Maastricht, The Netherlands) and maintained at the animal care facility of the Academic Medical Centre (University of Amsterdam), according to national guidelines with free access to food and water. The response of C57BL/6 mice from different commercial suppliers was tested when the current model of Klebsiella pneumonia (see below) was set up. No differences were found between the different Wt mice tested with regard to host response. The Institutional Animal Care and Use Committee approved all animal experiments.

Cell culture and stimulation Bone marrow derived macrophages (BMDMs) were prepared from Trem1/3−/−, Trem2−/− and Wt mice as described previously31 and details can be found in the Online methods. BMDMs were stimulated for 24 hours in medium containing 2mM L-glutamine, penicil- lin-streptomycin and 5% Panexin with or without heat-killed K. pneumoniae (30 minutes,

65°C; multiplicity of infection (MOI) 1:1, 10:1 or 100:1) at 37°C in 5% CO2. Supernatants were taken and stored at −20°C until assayed.

84 TREM-1/3 contributes to protective immunity in Klebsiella derived pneumosepsis whereas TREM-2 does not

Phagocytosis Phagocytosis of K. pneumoniae by Trem1/3−/− and Trem2−/− BMDMs was determined in essence as described before32,33. See Online supplement for details.

Induction of pneumonia Mice were intranasally inoculated with 104 K. pneumoniae serotype 2 (ATCC 43816, Rockville, MD) in a 50 µl saline solution and sacrificed 6, 24 or 48 hours thereafter as previously described33,34. Trem1/3−/− and Trem2−/− mice were studied in separate experi- ments, on each occasion compared with a sex and age matched Wt control group that was infected at the same time with the exact same inoculum (n = 8 per strain for each time point). Collection and handling of samples were done as previously described33,34 and details can be found in the online supplement. In survival studies mice (n = 16 per strain) were intranasally inoculated with 104 K. pneumoniae and monitored for up to 8 days after infection.

Assays Lung homogenates were prepared for immune assays as described before33,34. TNF-α, interleukin (IL)-1-β, IL-10, Keratinocyte-derived chemokine (KC), macrophage inflamma- tory protein 2 (MIP-2) (all R&D systems, Minneapolis, MN) and myeloperoxidase (MPO; 5 Hycult Biotechnology BV, Uden, the Netherlands) were measured using specific ELISAs according to manufacturers’ recommendations.

Histology After embedding, lungs were stained with haematoxylin and eosin to score inflam- mation or stained and quantified for Ly6G using immunohistochemical procedures as described33,34.

Statistical analysis Data are expressed as box-and-whisker diagrams depicting the smallest observa- tion, lower quartile, median, upper quartile and largest observation unless indicated otherwise. Comparisons between groups were conducted using the Mann-Whitney U or Chi square test. For survival studies Kaplan-Meier analyses followed by log rank test were performed. All analyses were done using GraphPad Prism version 5.01 (GraphPad Software, San Diego, CA). P-values less than 0.05 were considered statistically significant.

85 Chapter 5

Results

K. pneumoniae induced cytokine response is impaired in Trem1/3−/− and enhanced in Trem2−/− macrophages To obtain a first insight in the role of TREM-1 and TREM-2 in host defense toK. pneumoni- ae, we set out to investigate the responsiveness of Trem1/3−/−, Trem2−/− and Wt BMDMs to K. pneumoniae in vitro. To this end we stimulated BMDMs of different genotypes for 6 (Figure 1A-B) and 24 hours (Figure 1C-D) with heat killed K. pneumoniae and measured TNF-α and KC in culture supernatants. We found Trem1/3−/− BMDMs to produce less TNF-α and KC compared to Wt BMDMs when incubated with heat killed K. pneumoniae. BMDMs from Trem2−/− mice on the other hand showed higher levels of TNF-α and KC compared to Wt cells in supernatants.

A A B B 600 600 Wt Wt 12501250 TremTr1e/3m -1/-/3 -/- * * ** ** ****** 10001000 -/- -/- l l TremTr2e m2

l l ** ** m 40m 0 400 m 75m 0 750 pg/ pg/ pg/ pg/

F F 500 500 N N * * KC KC ****** T 20T 0 200 ****** 250 250 ** **

0 0 0 0 medmiumedium 1:1 1:1 1:101:10 1:1010:100 medmiumedium 1:1 1:1 1:101:10 1:1010:100 KlebKslebiellsaie (Mlla OI(M)OI) KlebKslebiellsaie (Mlla OI(M)OI) C C D D 12001200 300030000 0 * * ** ** l l l l m 80m 0 800 200020000 0 m m pg/ pg/

* * * * pg/ pg/ F F N N * * KC KC T 40T 0 400 100010000 0 ** ** ** ** * * 0 0 0 0 ****** medmiumedium 1:1 1:1 1:101:10 1:1010:100 medmiumedium 1:1 1:1 1:101:10 1:1010:100 KlebKslebiellsaie (Mlla OI(M)OI) KlebKslebiellsaie (Mlla OI(M)OI) Figure 1: Impaired cytokine responses of Trem1/3−/− cells and enhanced responses of Trem2−/− cells to K. pneumoniae. Bone marrow derived macrophages from wild type (Wt), Trem1/3−/− and Trem2−/− were stimulated for 6 hours (Figure 1A & B) and 24 hours (Figure 1C & D) with the indicated multiplicity of infec- tion (MOI) heat killed K. pneumoniae and TNF-α (A & C) and KC (B & D) were measured in supernatants. Data are expressed as box-and-whisker diagrams depicting the smallest observation, lower quartile, median, up- per quartile and largest observation. Representative graphs are shown from 3 independent experiments; *P < 0.05 **P < 0.01 ***P < 0.001 versus Wt cells.

Trem2−/− but not Trem1/3−/− macrophages show impaired phagocytosis of K. pneumoniae The early control of invading microbes by the innate immune system primarily depends on professional phagocytes – neutrophils and macrophages. In vitro studies have sug- gested that both TREM-1 and TREM-2 may enhance phagocytosis14,35. To investigate the role of TREM-1/3 and TREM-2 in phagocytosis of K. pneumoniae, we harvested

86 TREM-1/3 contributes to protective immunity in Klebsiella derived pneumosepsis whereas TREM-2 does not

A A NeutrophilsNeutrophils B B MacrophagesMacrophages 4000 4000 Wt Wt 1000010000 x x x x Trem1T/3re -m/- 1/3 -/- e e e e 3000 3000 -/- -/- 7500 7500 nd nd nd nd Trem2T rem2 i i i i * * s s s s i i i i s s s s o o o o t t t 200t 0 2000 5000 5000 cy cy cy cy go go go go a a a 100a 0 1000 2500 2500 h h h h P P P P

0 0 0 0 4°C 4°C 37°C37°C 4°C 4°C 37°C37°C Figure 2: Phagocytosis is impaired in Trem2−/− deficient macrophages. Growth arrested, CFSE-labelled K. pneumoniae were incubated with peripheral blood neutrophils (A) or FITC labeled K. pneumoniae with bone marrow derived macrophages (B) from wild type (Wt), Trem1/3−/− and Trem2−/− mice at 4°C (n = 3–4 per mouse strain) or 37°C (n = 6–8 per mouse strain) for 1 hour after which phagocytosis was quantified. Data are expressed as box-and-whisker diagrams depicting the smallest observation, lower quartile, median, upper quartile and largest observation; *P < 0.05 versus Wt cells. whole blood and BMDMs from naïve Trem1/3−/−, Trem2−/− and Wt mice and compared the capacity of neutrophils and macrophages to internalize growth-arrested K. pneu- moniae by FACS. Neither TREM-1/3 nor TREM-2 influenced neutrophil phagocytosis of K. pneumoniae (Figure 2A). In contrast, Trem2−/− BMDMs were hampered in their ability to internalize K. pneumoniae when compared to Wt BMDMs, whereas Trem1/3−/− BMDMs 5 showed no defect (Figure 2B; P < 0.01).

Trem1/3−/− but not Trem2−/− mice show an impaired host defense during Klebsiella pneumonia derived sepsis To investigate the functional role of TREM-1 and TREM-2 in host defense during Gram- negative pneumonia derived sepsis, Trem1/3−/−, Trem2−/− and Wt mice were infected with 104 viable K. pneumoniae and lungs, blood and spleens were harvested at predefined time points for quantitative cultures, seeking to collect data representative for local defense, at the primary site of infection, and subsequent dissemination. Trem1/3−/− mice demonstrated higher bacterial burdens in their lungs 48 hours after infection (Figure 3A); at earlier time points pulmonary bacterial loads did not differ between Trem1/3−/− and Wt mice. In addition, Trem1/3−/− mice showed enhanced early dissemination of the infection, as reflected by higher bacterial loads in blood and spleen 24 hours after infection (Figure 3C,E). In contrast, Trem2−/− and Wt mice had similar bacterial loads in all body sites at all time points (Figure 3B,D and F). To determine whether the enhanced bacterial growth and dissemination in Trem1/3−/− mice impacted on outcome, we next infected Trem1/3−/− and Wt mice via the airways with the same infectious dose of K. pneumoniae and followed them 8 days (Figure 4). Although death occurred after 2 days in all mouse strains, Trem1/3−/− mice displayed accelerated lethality. The median survival time of Trem1/3−/− mice was 65 hours (2.7 days) versus 106 hours (4.5 days) for

87 Chapter 5

A Lung C Blood E Spleen 9 Wt 9 9 8 -/- * 8 8 Trem1/3 l l 7 7 l 7 m m / / m

6 / U 6 U 6 U F F ** 5 F 5 C C 5 ** C 4 4 log log 4 log 3 log 3 10 10 3 2 10 2 2 1 1 1 0 0 6h 24h 48h 24h 48h 24h 48h B D F 9 Wt 9 9 8 8 8 Trem2 -/- l 7 l 7 l 7 m m m / / 6 / 6 U 6 U U F F 5 F 5 C 5 C C 4 4 log log 4 log log 3 3 10 3 10 2 10 2 2 1 1 1 0 0 6h 24h 48h 24h 48h 24h 48h

Figure 3: Trem1/3−/− mice show enhanced bacterial dissemination during pneumonia derived sepsis caused by K. pneumoniae. Wild type (Wt), Trem1/3−/− (A,C,E) and Trem2−/− mice (B,D,F) were intranasally infected with 104 colony forming units (CFU) K. pneumoniae and sacrificed 6, 24 or 48 hours later. Bacterial loads were determined in the lung (A,B), blood (C,D) and spleen (E,F). Data are scatter plots with medians (8 mice per group at each time point). *P < 0.05 and **P < 0.01 versus Wt mice at the same time point.

Wt mice. Eventually all Trem1/3−/−mice died versus 75% of the Wt mice (P < 0.001). These data establish a protective role for TREM1/3 in Klebsiella pneumonia derived sepsis as reflected by an enhanced lethality and increased bacterial growth and dissemination in Trem1/3−/− mice, whereas the role of TREM-2 seems limited.

100 Wt -/-

l Trem1/3 80 va i rv

u 60 s

t n 40 erce

P 20 *** 0 0 2 4 6 8 days Figure 4: Trem1/3−/− mice show increased mortality during pneumonia derived sepsis caused by K. pneumoniae. Wild type (Wt) and Trem1/3−/− mice (n = 16 per group) were intranasally infected with 104 colony forming units (CFU) K. pneumoniae and survival was monitored for 12 days). *** P < 0.001 versus Wt mice

88 TREM-1/3 contributes to protective immunity in Klebsiella derived pneumosepsis whereas TREM-2 does not

Trem1/3−/− but not Trem2−/− mice show increased lung pathology during Klebsiella pneumonia This model of Klebsiella pneumonia is associated with profound lung pathology33,34,36. To study the role of TREM-1/3 and TREM-2 herein we analyzed HE-stained lung tissue slides obtained from infected Trem1/3−/−, Trem2−/− and Wt mice using the semi-quantitative scoring system described in the Materials and Methods section (Figure 5 and Supple-

5

Figure 5: Trem1/3−/− mice show enhanced lung pathology during K. pneumoniae pneumonia. Representative slides of lung haematoxylin and eosin (HE) staining (magnification ×4; insets show magni- fication ×10) of wild type (Wt, A) and Trem1/3−/− mice (B) 6 hours, Wt (C) and Trem1/3−/− mice (D) 24 hours and Wt (E) and Trem1/3−/− mice (F) 48 hours after intranasal K. pneumoniae infection. Total pathology score at indicated time points post infection in Wt and Trem1/3−/− mice was determined according to the scor- ing system described in the Online methods section (G). Data are expressed as box-and-whisker diagrams depicting the smallest observation, lower quartile, median, upper quartile and largest observation (8 mice per group at each time point). * P < 0.05 versus Wt mice at the same time point.

89 Chapter 5

Table 1: Cytokine and chemokine concentrations in lung homogenates of Wt and Trem1/3−/− mice during Klebsiella pneumonia. Lung cytokines 6 hours 24 hours 48 hours Wt Trem1/3−/− Wt Trem1/3−/− Wt Trem1/3−/− TNF-α (pg/ml) 549 610 1225 644 1539 2229 (349-643) (448-688) (828-1676) (671-2044) (780-6779) (1040-2799) IL-1β (pg/ml) 827 861 3512 2315 3995 6271 (585-997) (573-1544) (1523-6269) (1602-3005) (2580-6632) (4946-7880) IL-6 (pg/ml) 336 436 1231 855 1483 3435 (293-465) (323-570) (521-2074) (739-1228) (720-2853) (2148-3954) IL-10 (pg/ml) 581 619 205 207 527 550 (450-664) (571-864) (112-275) (134-250) (488-602) (539-570) KC (ng/ml) 4.0 3.9 17.2 16.9 26.9 30.9 (3.0-4.8) (2.7-5.6) (11.9-55.6) (7.6-20.5) (8.8-43.2) (22.4-63.0) MIP-2 (ng/ml) 1.5 1.8 4.7 3.6 6.4 10.5 (1.2-1.9) (1.4-2.0) (1.4-7.4) (3.2-4.6) (1.9-9.2) (9.9-10.7) Proinflammatory cytokine (TNF-α, IL-1β, IL-6), anti-inflammatory (IL-10) and chemokine (KC and MIP-2) lev- els in lung homogenates at 6, 24 and 48 hours after intranasal K. pneumoniae infection in wild type (Wt) and Trem1/3−/− mice. Data are medians (interquartile ranges); n = 8 mice per group per time point.

mentary Figure E1). Already at 6 hours after infection mild interstitial inflammation and pleuritis were found in all mice; at later stages endothelialitis, bronchitis and edema became apparent. Interestingly, Trem1/3−/− mice showed exaggerated lung pathology at both 6 and 24 hours after infection when compared with Wt mice (both P < 0.05), with enhanced interstitial inflammation and pleuritis after 6 hours of infection and enhanced interstitial inflammation, endothelialitis, oedema and larger surfaces of confluent in- flammatory infiltrate 24 hours after infection. After 48 hours of infection no differences were observed anymore between Trem1/3−/− and Wt mice. Lung pathology scores did not differ between Trem2−/− and Wt mice after 6 and 24 hours of infection (Supplemen- tary Figure E1). At 48 hours, Trem2−/− mice showed modestly attenuated lung pathology when compared with Wt mice (P < 0.05). To study the impact of TREM-1/3 and TREM-2 deficiency on the induction of cytokines and chemokines in lungs, we measured the levels of TNF-α, IL-1β, IL-6, IL-10, KC and MIP-2 in whole lung homogenates obtained from Trem1/3−/−, Trem2−/− and Wt mice 6, 24 or 48 hours after airway infection with K. pneumoniae; no differences between mouse strains were found (shown for Trem1/3−/− versus Wt mice in Table 1).

Neither TREM-1/3 nor TREM-2 deficiency influences neutrophil recruitment into the lungs Shortly after the onset of bacterial pneumonia neutrophils become recruited to the site of infection reflecting an important part of the host response to infection37,38. Previous

90 TREM-1/3 contributes to protective immunity in Klebsiella derived pneumosepsis whereas TREM-2 does not studies have documented an important protective role for neutrophils in the innate response to Klebsiella airway infection39,40. Recently it was reported that Trem1/3−/− neu- trophils are hampered in their ability to cross the epithelial barrier during pulmonary infection with P. aeruginosa21. Therefore, we determined neutrophil influx in Trem1/3−/−, Trem2−/− and Wt mice during Klebsiella pneumonia after 6, 24, and 48 hours of infection by quantifying the number of Ly6G positive cells in lung tissue sections and by measur- ing MPO concentrations in whole lung homogenates. No differences in Ly6G positive

5

Figure 6: Neutrophil influx in Klebsiella pneumonia is not influenced by TREM-1/3 deficiency. MPO levels in whole lung homogenates (A) and quantification of pulmonary Ly-6G positivity (B) and 6, 24 and 48 hours after infection in Wt and Trem1/3−/− mice. Representative neutrophil staining (brown) of Wt (C) and Trem1/3−/− mice (D); original magnification 100×. Neutrophil influx into BALF (E) 24 hours after induction of Klebsiella pneumonia. Data are expressed as box-and-whisker diagrams depicting the smallest observa- tion, lower quartile, median, upper quartile and largest observation (8 mice per group at each time point).

91 Chapter 5 cells or MPO concentrations were observed at any time point between Trem1/3−/− and Wt mice (Figure 6) or between Trem2−/− and Wt mice (data not shown). Considering the reported role of TREM-1 in neutrophil migration across the epithelium during Pseudomo- nas pneumonia21, we also determined neutrophil recruitment into the bronchoalveolar space by counting the number of neutrophils in BALF harvested from Trem1/3−/− and Wt mice 24 hours after infection. The total cell influx did not differ at this time point neither did the number of neutrophils (Figure 6).

Discussion

TREM-family proteins are potent immunomodulatory receptors expressed on innate im- mune cells and are therefore considered potential therapeutic targets for the treatment of inflammatory conditions including pneumonia and sepsis41. TREM-1 and TREM-2 are prominent members of this receptor family that in vitro seem to mediate opposite ef- fects: whereas TREM-1 acts as an amplifier of inflammation, TREM-2 can function as an inhibitor. Interfering with TREM-signaling in vivo either dictates survival or lethality dur- ing severe abdominal sepsis depending on the receptor antagonized: blocking TREM-1 improved survival7,18,19, while blockade of TREM-2 had the opposite effect23. In contrast, during pneumonia caused by P. aeruginosa or S. pneumoniae pneumonia TREM-1/3 deficiency resulted in impaired host defense21,22, while TREM-2 deficiency improved sur- vival during pneumococcal pneumonia24. To further dissect the roles of these receptors during severe infection we evaluated host responses of mice deficient for TREM-1 and TREM-3 or TREM-2 during Klebsiella pneumonia. We here show that TREM-1/3 enhanced cytokine release by macrophages upon exposure to Klebsiella, while TREM-2 inhibited this. In addition, TREM-2 but not TREM-1/3, facilitated macrophage phagocytosis of Klebsiella. Importantly, the presence of TREM-1 and TREM-3 improved host defense dur- ing Klebsiella pneumonia derived sepsis, while TREM-2 did not contribute. These data in part contrast with previous studies on this topic7,18,19,23,24 and exemplify the complex functions of TREM receptors in the innate immune response during sepsis. Both TREM-1 and TREM-2 have been shown to influence TLR signaling. TREM-1 syner- gizes with different TLRs. For example, human monocytes co-incubated with an agonistic TREM-1 antibody and TLR ligands, produced higher cytokine levels than after incubation with either stimulus alone6,7. Accordingly, TREM-1 silenced RAW macrophages displayed reduced IL-1β and IL-6 mRNA levels when stimulated with LPS42. In agreement, we report that Trem1/3−/− macrophages produce less TNF-α and KC when incubated with Klebsiella LPS or heat killed K. pneumoniae. In addition, we showed Trem2−/− BMDMs to produce higher levels of cytokines in response to these stimuli. This is in line with earlier reports documenting increased levels of TNF-α and IL-6 secreted by TREM-2 deficient BMDMs

92 TREM-1/3 contributes to protective immunity in Klebsiella derived pneumosepsis whereas TREM-2 does not when incubated with LPS, zymosan, CpG or S. pneumoniae12,24. Correspondingly, TREM-2 silenced murine alveolar macrophages showed increased TNF-α levels when incubated with LPS43 and bone marrow myeloid cells transduced with TREM-2 showed an anti- inflammatory phenotype44. In addition to cytokine production by macrophages we assessed the capacity of Trem1/3−/− and Trem2−/− leukocytes to internalize K. pneumoniae. TREM-1/3 deficiency did not affect the degree of phagocytosis of this pathogen by macrophages or neu- trophils. In agreement, Trem1/3−/− neutrophils had an unaltered ability to internalize P. aeruginosa in vitro21 and a TREM-1 blocking fusion protein did not affect phagocytosis by macrophages of this pathogen45. In contrast, TREM-2 deficiency resulted in reduced phagocytosis by macrophages of Escherichia (E.) coli, Staphylococcus aureus and S. pneu- moniae14,24. Similarly, transducing myeloid cells with TREM-2 enhanced their phagocytic capacity44. In accordance, we found around 30% less phagocytosis of K. pneumoniae by Trem2−/− macrophages. Notably, TREM-2 deficiency did not affect neutrophil phagocyto- sis of this pathogen, which corresponds with the lack of evidence for TREM-2 expression by neutrophils. To investigate the role of TREM-1 in Klebsiella pneumonia, we used a mouse lacking both TREM-1 and TREM-3, an approach also used by Klesney-Tait et al to study the role of TREM-1 in Pseudomonas pneumonia21. The Trem3 gene is highly homologues to the 5 Trem1 gene suggesting redundant functions of these two proteins21. Indeed, similar to TREM-1, TREM-3 was found to be an activating receptor on murine macrophages29. Therefore to simulate the effect of antagonizing TREM-1 in humans, a mouse deficient for both receptors was generated. We found Trem1/3−/− mice to be more susceptible to Klebsiella pneumonia, as reflected by increased lethality accompanied by enhanced bacterial growth and dissemination. Although at a first glance these data are similar to those reported after infection with Pseudomonas via the airways21, important differ- ences exist. First, although both Gram-negative respiratory pathogens, Pseudomonas is cleared from the lungs of previously healthy mice, even after high dose infection, while Klebsiella gradually grows at the primary site of infection subsequently leading to systemic infection28. As a consequence thereof, lung inflammation is more acute in the Pseudomonas model, resembling a pulmonary LPS challenge. The differential char- acteristics of experimental murine Pseudomonas and Klebsiella pneumonia are further illustrated by earlier reports revealing opposite roles for the prototypic proinflammatory cytokines TNF-α and IL-1 in host defense during pneumonia caused by Pseudomonas (harmful)46,47 or Klebsiella (protective)48. Second, TREM-1 was reported to strongly impact on neutrophil migration through lung epithelium after infection with Pseudomonas21. Considering that these cells play an important role in the host defense against patho- gens in the lung37, we determined the extent of neutrophil recruitment into lung tissue (by Ly6 staining of lung slides and by measurement of MPO in whole lung homogenates)

93 Chapter 5 and the bronchoalveolar space (by determining neutrophil counts in BALF) in Trem1/3−/− and Wt mice. Strikingly, we did not find any differences, strongly arguing against a role for TREM-1 in this classic inflammatory response after infection with Klebsiella. Finally, Trem1/3−/− mice displayed higher pro-inflammatory cytokine levels in their lungs after inoculation with Pseudomonas, which most likely reflected the increased pulmonary bac- terial loads in these animals21. We here did not detect differences in lung cytokine levels between Trem1/3−/− and Wt mice. Although the reduced responsiveness of Trem1/3−/− macrophages as shown in vitro strongly suggests a role for TREM-1/3 in the induction of early inflammation, TREM-1/3 deficiency did not impact on cytokine production in whole lungs in vivo, suggesting that the measurement of cytokine levels between 6 and 48 hours after infection is not sensitive enough to expose more subtle impairments in the TLR-initiated innate immune response. In contrast, the enhanced susceptibility of Trem1/3−/− mice to lower respiratory tract infection caused by S. pneumoniae was linked to an attenuated early inflammatory response in the lungs22. Together these data sug- gest a protective role for TREM-1 in host defense during pneumonia caused by different bacterial pathogens relevant for human disease, albeit likely via at least partially distinct mechanisms. Of note, a role for TREM-3 cannot be completely ruled out and more research is necessary to address this matter perhaps by making use of a TREM-1 and/ or TREM-3 single knockout mouse. Another limitation of this study is that no detailed insight is revealed with regard to the underlying mechanisms that mediate the function of TREM-1/3 and TREM2 during Klebsiella pneumonia at the cellular level. Interestingly, Trem2−/− mice did not have a noticeable phenotype during Klebsiella pneumonia despite the reduced capacity of TREM-2 deficient macrophages to internal- ize K. pneumoniae in vitro. Apparently the in vitro defect in phagocytosis of these cells is not relevant in vivo and is compensated for by other cells and/or mechanisms. This finding is in accordance with a recent paper showing defective phagocytosis of E. coli by TREM-2 knockout macrophages in vitro without a subsequent in vivo phenotype during peritonitis caused by this pathogen49. Another recent paper reported a protec- tive effect of TREM-2 during severe sepsis induced by cecal ligation and puncture, as indicated by a reduced survival in mice treated with a blocking TREM-2 fusion protein and an increased survival in mice administered with TREM-2 overexpressing bone mar- row derived myeloid cells23. The protective effect mediated by TREM-2 was attributed to the phagocytic function of TREM-2 since administration of the blocking TREM-2 fusion protein increased bacterial loads, whereas TREM-2 overexpressing bone marrow my- eloid cells reduced these; furthermore TREM-2 overexpressing cells displayed improved phagocytosis of beads and whole bacteria23. By contrast, in another investigation TREM- 2 was reported to weaken defense against pneumococci in the airways most likely by impairing phagocytosis by alveolar macrophages24. Together these data suggest that the immune modifying properties of TREM-2 as demonstrated in vitro translate into a

94 TREM-1/3 contributes to protective immunity in Klebsiella derived pneumosepsis whereas TREM-2 does not variable impact on host defense during severe infection in vivo at least in part depend- ing on the source and the type of the bacterial challenge. K. pneumoniae is an important cause of lower respiratory tract infection and sepsis25-27. We here used our established model of pneumonia derived sepsis caused by this patho- gen, characterized by a gradually growing bacterial burden at the primary site of infec- tion followed by bacterial spreading to blood and distant organs. This model not only resembles a likely clinical scenario of human disease but also allows for evaluation of the potential protective and collateral damaging effects of innate immune activation. Our results reveal that while both TREM-1/3 and TREM-2 impact on macrophage responsive- ness toward K. pneumoniae, only TREM-1/3 contributes to host defense during Klebsi- ella pneumonia. Preclinical research on potential therapeutic targets for sepsis requires studies in multiple clinically relevant experimental models. In this respect, studies of pneumonia and peritonitis are of importance considering that these are the two most common sources of sepsis in humans50. While interpretation of this and earlier studies may be influenced by the use of different interventions and mouse strains (targeting only a single TREM or TREM1/3), our current results on pneumonia taken together with previous abdomen-derived sepsis studies on TREM-17,18,19,21,22 and TREM-223,24 illustrate that the roles of these innate immune receptors in the host response to severe infection is not uniform in different sepsis models (for overview see Supplemental Table 1). 5

Acknowledgements

We thank Marieke ten Brink, Joost Daalhuisen and Regina de Beer for their expert techni- cal assistance.

95 Chapter 5

References

1. Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011;​34:​637‑50. 2. Ivashkiv LB. Cross-regulation of signaling by ITAM-associated receptors. Nat Immunol 2009;​10:​ 340‑7. 3. Nathan C, Ding A. Nonresolving inflammation. Cell 2010;​140:​871‑82. 4. Klesney-Tait J, Turnbull IR, Colonna M. The TREM receptor family and signal integration. Nat Im- munol 2006;​7:​1266‑73. 5. Sharif O, Knapp S. From expression to signaling: roles of TREM-1 and TREM-2 in innate immunity and bacterial infection. Immunobiology 2008;​213:​701‑13. 6. Bouchon A, Dietrich J, Colonna M. Cutting edge: inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J Immunol 2000;​164:​4991‑5. 7. Bouchon A, Facchetti F, Weigand MA, Colonna M. TREM-1 amplifies inflammation and is a crucial mediator of septic shock. Nature 2001;​410:​1103‑7. 8. Netea MG, Azam T, Ferwerda G, Girardin SE, Kim SH, Dinarello CA. Triggering receptor expressed on myeloid cells-1 (TREM-1) amplifies the signals induced by the NACHT-LRR (NLR) pattern recog- nition receptors. J Leukoc Biol 2006;​80:​1454‑61. 9. Daws MR, Lanier LL, Seaman WE, Ryan JC. Cloning and characterization of a novel mouse myeloid DAP12-associated receptor family. Eur J Immunol 2001;​31:​783‑91. 10. Schmid CD, Sautkulis LN, Danielson PE, et al. Heterogeneous expression of the triggering recep- tor expressed on myeloid cells-2 on adult murine microglia. J Neurochem 2002;​83:​1309‑20. 11. Cella M, Buonsanti C, Strader C, Kondo T, Salmaggi A, Colonna M. Impaired differentiation of osteoclasts in TREM-2-deficient individuals. J Exp Med 2003;​198:​645‑51. 12. Turnbull IR, Gilfillan S, Cella M, et al. Cutting Edge: TREM-2 Attenuates Macrophage Activation. The Journal of Immunology 2006;​177:​3520‑4. 13. Hamerman JA, Jarjoura JR, Humphrey MB, Nakamura MC, Seaman WE, Lanier LL. Cutting edge: inhibition of TLR and FcR responses in macrophages by triggering receptor expressed on myeloid cells (TREM)-2 and DAP12. J Immunol 2006;​177:​2051‑5. 14. N’Diaye EN, Branda CS, Branda SS, et al. TREM-2 (triggering receptor expressed on myeloid cells 2) is a phagocytic receptor for bacteria. J Cell Biol 2009;​184:​215‑23. 15. Daws MR, Sullam PM, Niemi EC, Chen TT, Tchao NK, Seaman WE. Pattern recognition by TREM-2: binding of anionic ligands. J Immunol 2003;​171:​594‑9. 16. Guerreiro R, Wojtas A, Bras J, et al. TREM2 variants in Alzheimer’s disease. N Engl J Med 2013;​368:​ 117‑27. 17. Jonsson T, Stefansson H, Steinberg S, et al. Variant of TREM2 associated with the risk of Alzheim- er’s disease. N Engl J Med 2013;​368:​107‑16. 18. Gibot S, Buonsanti C, Massin F, et al. Modulation of the triggering receptor expressed on the myeloid cell type 1 pathway in murine septic shock. Infect Immun 2006;​74:​2823‑30. 19. Gibot S, Massin F, Marcou M, et al. TREM-1 promotes survival during septic shock in mice. Eur J Immunol 2007;​37:​456‑66. 20. Gibot S, Alauzet C, Massin F, et al. Modulation of the triggering receptor expressed on myeloid cells-1 pathway during pneumonia in rats. J Infect Dis 2006;​194:​975‑83. 21. Klesney-Tait J, Keck K, Li X, et al. Transepithelial migration of neutrophils into the lung requires TREM-1. J Clin Invest 2013;​123:​138‑49.

96 TREM-1/3 contributes to protective immunity in Klebsiella derived pneumosepsis whereas TREM-2 does not

22. Hommes TJ, Hoogendijk AJ, Dessing MC, et al. Triggering receptor expressed on myeloid cells-1 (TREM-1) improves host defence in pneumococcal pneumonia. J Pathol 2014;​233:​357‑67. 23. Chen Q, Zhang K, Jin Y, et al. Triggering Receptor Expressed on Myeloid Cells-2 Protects against Polymicrobial Sepsis by Enhancing Bacterial Clearance. Am J Respir Crit Care Med 2013. 24. Sharif O, Gawish R, Warszawska JM, et al. The triggering receptor expressed on myeloid cells 2 inhibits complement component 1q effector mechanisms and exerts detrimental effects during pneumococcal pneumonia. PLoS Pathog 2014;​10:​e1004167. 25. Podschun R, Ullmann U. Klebsiella spp. as nosocomial pathogens: epidemiology, taxonomy, typ- ing methods, and pathogenicity factors. Clin Microbiol Rev 1998;​11:​589‑603. 26. Kang CI, Song JH, Chung DR, et al. Risk factors and pathogenic significance of severe sepsis and septic shock in 2286 patients with gram-negative bacteremia. J Infect 2011;​62:​26‑33. 27. Shorr AF, Tabak YP, Killian AD, Gupta V, Liu LZ, Kollef MH. Healthcare-associated bloodstream infection: A distinct entity? Insights from a large U.S. database. Crit Care Med 2006;​34:​2588‑95. 28. Knapp S, Schultz MJ, van der Poll T. Pneumonia models and innate immunity to respiratory bacte- rial pathogens. Shock 2005;​24 Suppl 1:​12‑8. 29. Chung DH, Seaman WE, Daws MR. Characterization of TREM-3, an activating receptor on mouse macrophages: definition of a family of single Ig domain receptors on mouse chromosome 17. Eur J Immunol 2002;​32:​59‑66. 30. Turnbull IR, Gilfillan S, Cella M, et al. Cutting edge: TREM-2 attenuates macrophage activation. J Immunol 2006;​177:​3520‑4. 31. Eske K, Breitbach K, Kohler J, Wongprompitak P, Steinmetz I. Generation of murine bone marrow derived macrophages in a standardised serum-free cell culture system. J Immunol Methods 2009;​ 342:​13‑9. 5 32. Wiersinga WJ, Kager LM, Hovius JW, et al. Urokinase receptor is necessary for bacterial defense against pneumonia-derived septic melioidosis by facilitating phagocytosis. J Immunol 2010;​184:​ 3079‑86. 33. Achouiti A, Vogl T, Urban CF, et al. Myeloid-related protein-14 contributes to protective immunity in gram-negative pneumonia derived sepsis. PLoS Pathog 2012;​8:​e1002987. 34. Rijneveld AW, Weijer S, Florquin S, et al. 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 2004;​103:​1702‑9. 35. Radsak MP, Salih HR, Rammensee HG, Schild H. Triggering receptor expressed on myeloid cells-1 in neutrophil inflammatory responses: differential regulation of activation and survival. J Immu- nol 2004;​172:​4956‑63. 36. Renckens R, Roelofs JJ, Bonta PI, et al. Plasminogen activator inhibitor type 1 is protective during severe Gram-negative pneumonia. Blood 2007;​109:​1593‑601. 37. Mizgerd JP. Acute lower respiratory tract infection. N Engl J Med 2008;​358:​716‑27. 38. Opitz B, van Laak V, Eitel J, Suttorp N. Innate immune recognition in infectious and noninfectious diseases of the lung. Am J Respir Crit Care Med 2010;​181:​1294‑309. 39. Tsai WC, Strieter RM, Wilkowski JM, et al. Lung-specific transgenic expression of KC enhances resistance to Klebsiella pneumoniae in mice. J Immunol 1998;​161:​2435‑40. 40. Ye P, Rodriguez FH, Kanaly S, et al. Requirement of interleukin 17 receptor signaling for lung CXC chemokine and granulocyte colony-stimulating factor expression, neutrophil recruitment, and host defense. J Exp Med 2001;​194:​519‑27. 41. Derive M, Massin F, Gibot S. Triggering receptor expressed on myeloid cells-1 as a new therapeutic target during inflammatory diseases. Self Nonself 2010;​1:​225‑30.

97 Chapter 5

42. Ornatowska M, Azim AC, Wang X, et al. Functional genomics of silencing TREM-1 on TLR4 signal- ing in macrophages. Am J Physiol Lung Cell Mol Physiol 2007;​293:​L1377‑84. 43. Gao X, Dong Y, Liu Z, Niu B. Silencing of triggering receptor expressed on myeloid cells-2 en- hances the inflammatory responses of alveolar macrophages to lipopolysaccharide. Mol Med Report 2013. 44. Takahashi K, Prinz M, Stagi M, Chechneva O, Neumann H. TREM2-transduced myeloid precursors mediate nervous tissue debris clearance and facilitate recovery in an animal model of multiple sclerosis. PLoS Med 2007;​4:​e124. 45. Wang F, Liu S, Wu S, et al. Blocking TREM-1 signaling prolongs survival of mice with Pseudomonas aeruginosa induced sepsis. Cell Immunol 2012;​272:​251‑8. 46. Skerrett SJ, Martin TR, Chi EY, Peschon JJ, Mohler KM, Wilson CB. Role of the type 1 TNF receptor in lung inflammation after inhalation of endotoxin or Pseudomonas aeruginosa. Am J Physiol 1999;​ 276:​L715‑27. 47. Schultz MJ, Rijneveld AW, Florquin S, Edwards CK, Dinarello CA, van der Poll T. Role of interleukin-1 in the pulmonary immune response during Pseudomonas aeruginosa pneumonia. Am J Physiol Lung Cell Mol Physiol 2002;​282:​L285‑90. 48. Cai S, Batra S, Wakamatsu N, Pacher P, Jeyaseelan S. NLRC4 inflammasome-mediated production of IL-1beta modulates mucosal immunity in the lung against gram-negative bacterial infection. J Immunol 2012;​188:​5623‑35. 49. Gawish R, Martins R, Bohm B, et al. Triggering receptor expressed on myeloid cells-2 fine-tunes inflammatory responses in murine Gram-negative sepsis. FASEB J 2014. 50. Angus DC, Linde-Zwirble WT, Lidicker J, Clermont G, Carcillo J, Pinsky MR. Epidemiology of severe sepsis in the United States: analysis of incidence, outcome, and associated costs of care. Crit Care Med 2001;​29:​1303‑10.

98 TREM-1/3 contributes to protective immunity in Klebsiella derived pneumosepsis whereas TREM-2 does not

Supplement

Methods

Cell culture Bone marrow derived macrophages (BDMDs) were generated using recombinant mouse granulocyte-macrophage colony stimulating factor (GM-CSF, Invitrogen, Breda, the Netherlands) as previously described1 and were pooled (derived from 3-4 mice). In brief, femurs and tibias were removed aseptically and flushed with sterile phosphate buffered saline (PBS) and washed twice. Next the bone marrow cells were resuspended in RPMI containing 2 mM l-glutamine, penicillin-streptomycin, 5% Panexin BMM (PAN-Biotech, Aidenbach, Germany), 50 µM β-mercaptoethanol (Sigma-Aldrich, Zwijndrecht, the

Netherlands) and 2 ng/ml GM-CSF. Cells were incubated at 37°C, 5% CO2 and medium was refreshed at day 5 and 7. Subsequently the cells were harvested at day 9 or 10 after washing them with PBS to remove non-adherent cells. Next cells were plated at the indi- cated concentration per well in a 96-well flat-bottom plate in 100 µL to adhere overnight at 37°C, 5% CO2. Subsequently the cells were washed with PBS to remove non-adherent cells and stimulated with heat-killed K. pneumoniae for 24 hours as indicated (5 × 104 5 cells per well) or used for phagocytosis (1 × 105cells per well).

Phagocytosis Growth-arrested bacteria were prepared as described before2. In brief, Klebsiella (K.) pneumoniae were cultured and washed with pyrogen-free sterile saline and resus- pended in sterile PBS to a concentration of 2 × 109 bacteria/ml. The concentrated K. pneumoniae preparation was treated for 1 hour at 37°C with 50 µg/ml Mitomycin C (Sigma-Aldrich; Zwijndrecht, the Netherlands) to prepare alive but growth-arrested bacteria. Subsequently, the growth-arrested K. pneumoniae preparation was washed twice in ice-cold sterile PBS by centrifugation at 4°C, and the final pellet was dispersed in ice-cold PBS in the initial volume and transferred to sterile tubes and subsequently labeled with carboxyfluorescein succinimidyl ester (CFSE, Invitrogen, Breda, the Nether- lands) for whole blood incubations or fluorescein isothiocyanate (FITC) for macrophage incubations. 100 µl heparinized whole blood was incubated with 10 µl bacteria in RPMI (Gibco) (end concentration of 1 × 107 CFU/ml) at 37°C (n = 8 per group) or 4°C (n = 4 per group). After 20 and 60 minutes, samples were put on ice to stop phagocytosis.

Afterwards, red blood cells were lysed using isotonic NH4Cl solution (155 mM NH4Cl, 10 mM KHCO3, 100 mM EDTA, pH 7.4). Neutrophils were labeled using using anti-Ly-6G-PE (clone 1A8, BD Pharmingen, San Diego, CA) and washed twice in FACS-buffer (0.5% BSA,

0.01% NaN3, 0.35 mM EDTA in PBS) for analysis. Growth-arrested, FITC labelled bacteria

99 Chapter 5 were opsonised for 30 minutes at 37°C in 10% normal mouse serum and washed twice in PBS before they were added to the macrophages at a multiplicity of infection of 100 in a volume of 10 µL. Bacteria and macrophages were spun at 1000 RPM for 5 minutes and incubated at 37°C (n = 8 wells per strain) or 4°C (n = 4 wells per strain). After 1 hour, samples were washed with ice-cold PBS, then thoroughly scraped from the bottom and quenched by addition of 50 µL 4% Trypan blue (Sigma-Aldrich; Zwijndrecht, the Netherlands) for 1 minute. Next cells were washed twice in FACS-buffer. The degree of phagocytosis was determined using FACSCalibur (Becton Dickinson Immunocytometry, San Jose, CA.) The phagocytosis index of each sample was calculated as follows: geo- metric mean fluorescence × % positive cells.

Sample collection and handeling Blood was drawn into heparinized tubes and organs were removed aseptically and homogenised in 4 volumes of sterile isotonic saline using a tissue homogenizer (Biospec Products, Bartlesville, UK). To determine bacterial loads, ten-fold dilutions were plated on blood agar plates and incubated at 37°C for 16 hours. Bronchoalveolar lavage (BAL) fluid was obtained from a separate group of infected Trem1/3−/− and Wt mice (n = 8 per group) at 24 hours of infection. The trachea was exposed through a midline incision and cannulated with a sterile 22-gauge Abbocath-T catheter (Abbott Laboratories, Sligo, Ireland). BAL was performed by instilling two 0.5 ml aliquots of sterile PBS as described earlier3. 0.5–1 ml of BAL fluid was retrieved per mouse. Cell-free BAL supernatants were stored at –20°C for cytokine measurement. Cell numbers were determined using a hemocytometer, and Giemsa-stained cytospin preparations were used for differential cell counts.

Histology Lungs were harvested at the indicated time points, fixed in 10% buffered formalin, and embedded in paraffin. 4 µm sections were stained with haematoxylin and eosin (HE) and analyzed by a pathologist blinded for groups. To score lung inflammation and damage, the entire lung surface was analyzed with respect to the following parameters: bronchitis, edema, interstitial inflammation, intra-alveolar inflammation, pleuritis, en- dothelialitis and percentage of the lung surface demonstrating confluent inflammatory infiltrate. Each parameter was graded 0–4, with 0 being ‘absent’ and 4 being ‘severe’. The total pathology score was expressed as the sum of the score for all parameters. Granu- locyte staining was done using FITC-labeled rat anti-mouse Ly-6G mAb (Pharmingen, San Diego, CA) as described earlier3. Ly-6G expression in the lung tissue sections was quantified by digital image analysis4. In short, lung sections were scanned using the Olympus Slide system (Olympus, Tokyo, Japan) and TIF images, spanning the full tissue section were generated. In these images Ly-6G positivity and total surface area were

100 TREM-1/3 contributes to protective immunity in Klebsiella derived pneumosepsis whereas TREM-2 does not measured using Image J (U.S. National Institutes of Health, Bethesda, MD, http://rsb.info. nih.gov/ij); the amount of Ly-6G positivity was expressed as a percentage of the total surface area.

5

101 Chapter 5

References

1. Eske K, Breitbach K, Kohler J, Wongprompitak P, Steinmetz I. Generation of murine bone marrow derived macrophages in a standardised serum-free cell culture system. J Immunol Methods 2009;​ 342:​13‑9. 2. Achouiti A, Vogl T, Urban CF, et al. Myeloid-related protein-14 contributes to protective immunity in gram-negative pneumonia derived sepsis. PLoS Pathog 2012;​8:​e1002987. 3. Knapp S, Wieland CW, van ‘t Veer C, et al. 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 2004;​172:​3132‑8. 4. Lammers AJ, de Porto AP, Florquin S, et al. Enhanced vulnerability for Streptococcus pneumoniae sepsis during asplenia is determined by the bacterial capsule. Immunobiology 2011;​216:​863‑70.

102 TREM-1/3 contributes to protective immunity in Klebsiella derived pneumosepsis whereas TREM-2 does not

Supplemental Table 1 Mouse Model TREM-1 TREM-1/3 KO TREM-2 SEPSIS MODELS: Endotoxemia (LPS Inhibition via TREM-1/Fc fusion - KO: improved survival4 shock) protein1, siRNA2 or LP173 improves survival Cecal ligation and Inhibition via TREM-1/Fc fusion Improved Blocking AB: worsens survival, puncture protein improves survival1; siRNA survival (online higher CFUs6; administration of dose dependent effect: high patent)5 TREM-2 overexpressing myeloid dose worsens survival, low dose cells protects6 improves survival2 Escherichia coli Inhibition via TREM-1/Fc fusion - KO: no phenotype4 peritonitis protein improves survival1 Burkholderia LP17 improves survival, lowers - - pseudomallei CFUs7 (melioidosis) PNEUMONIA MODELS: Streptococcus Agonistic AB: improves survival, Worsened KO: improved survival, lower pneumoniae lowers CFUs8 survival, higher CFUs10 CFUs9 Pseudomonas Worsened aeruginosa survival, higher CFUs11 Legionella KO: no phenotype with respect to - - 5 pneumophila bacterial load12 Klebsiella - Worsened KO: no phenotype* pneumoniae survival, higher CFUs* 1Bouchon, Nature, 2001; 2Gibot, Eur J Immunol, 2007; 3Gibot, I&I, 2006; 4Gawisch, FASEB, 2014; 5Colonna, online patent #US20100306863, 2010; 6Chen, AJRCCM, 2013; 7Wiersinga, JID, 2007; 8Lagler, JI, 2009; 9Hom- mes, J Pathol, 2014; 10Sharif, PLOS Pathogens, 2014; 11Klesney-Tait, JCI, 2013; 12Weber, PLOS Pathogens, 2014; *current manuscript. AB = antibody; CFU = colony forming unit; KO = knockout; LP17 = synthetic TREM-1 inhibitor; LPS = lipo- polysaccharide; TREM = Triggering Receptor Expressed on Myeloid Cells

103 Chapter 5

Supplemental Figure E1: Lung pathology in Trem2−/− mice after infection with K. pneumoniae. Rep- resentative slides of lung haematoxylin and eosin (HE) staining of wild type (Wt, A) and Trem2−/− mice (B) 6 hours, Wt (C) and Trem2−/− mice (D) 24 hours and Wt (E) and Trem2−/− mice (F) 48 hours after intranasal K. pneumoniae infection. Total pathology score at indicated time points post infection in Wt and Trem2−/− mice was determined according to the scoring system described in the Online methods section (G). Data are expressed as box-and-whisker diagrams depicting the smallest observation, lower quartile, median, upper quartile and largest observation (8 mice per group at each time point). * P < 0.05 versus Wt mice at the same time point.

104 Triggering Receptor Expressed on Myeloid Cells (TREM) -2 impairs host defense in Gram-negative sepsis (melioidosis)

Tassili A. F. Weehuizen1,2, Tijmen J. Hommes1,2, Jacqueline M. Lankelma1,2, Hanna K. de Jong1,2, Joris. J.T.H. Roelofs3, Alex F. de Vos1,2, Marco Colonna4, Tom van der Poll MD1,2,5, W. Joost 6 Wiersinga1,2,5 Academic Medical Center, University of Amsterdam, the Netherlands: 1Center for Experimental and Molecular Medicine 2Center for Infection and Immunity 3Department of Pathology 5Division of Infectious Diseases 4Washington University in St. Louis, St. Louis, Missouri, USA: Department of Pathology

submitted Chapter 6

Abstract

Objective: Triggering receptor on myeloid cells (TREM) -1 and TREM-2 are key regulators of the inflammatory response that are involved in the clearance of invading pathogens. Melioidosis, caused by the “Tier 1” biothreat agent Burkholderia (B.) pseudomallei, is a common form of community-acquired sepsis in Southeast-Asia. TREM-1 has been sug- gested as a biomarker for sepsis and melioidosis. We aimed to characterize the expres- sion and function of TREM-1 and TREM-2 in melioidosis. Design: Animal study. Setting: University research laboratory. Subjects: Wild-type, TREM-1/3 and TREM-2 deficient mice. Interventions: Mice were intranasally infected with live B. pseudomallei and killed after 24, and/or 72 h for harvesting of lungs, liver, spleen, and blood. Additionally, survival studies were performed. Measurements and Main results: TREM-1 and TREM-2 expression was increased both in the lung and liver of B. pseudomallei-infected mice. Strikingly, Trem-2−/−, but not Trem-1/3−/−, mice displayed a markedly improved host defense as reflected by a strong survival advantage together with decreased bacterial loads, less inflammation and re- duced organ injury. Cellular responsiveness of TREM-2, but not TREM-1, deficient blood and bone-marrow derived macrophages (BMDM) was diminished upon exposure to B. pseudomallei. Phagocytosis and intracellular killing of B. pseudomallei by BMDM and alveolar macrophages were TREM-1 and TREM-2-independent. Conclusion: TREM-2, and to a lesser extent TREM-1, plays a remarkable detrimental role in the host defense against a clinically relevant Gram-negative pathogen: TREM-2 deficiency restricts the inflammatory response, thereby decreasing organ damage and mortality.

106 TREM-2 impairs host defense in Gram-negative sepsis (melioidosis)

Introduction

In sepsis, defined as a deregulated host response to a life-threatening infection, a careful balance between inflammatory and anti-inflammatory responses is vital1-3. Pathogen- or danger-associated molecular patterns are recognized by intracellular sensory complexes and cell surface receptors expressed on innate immune cells that can initiate the inflam- matory and anti-microbial response. Well-known examples of these pattern recognition receptors (PRRs) are the Toll-like receptor (TLR), nucleotide-oligomerization domain-like receptor (NLR) and C-type lectin receptor (CLR) families4. A more recently discovered group of innate immune receptors are the membrane-bound triggering receptors on myeloid cells (TREMs), which act as key modulators, rather than as initiators, of the inflammatory response5-7. TREM-1 and TREM-2 are the most studied members of the TREM-family, however their exact role in the pathogenesis of sepsis remains ill-defined. Upon recognition of partially still unspecified ligands, TREM-1 and TREM-2 recruit and phosphorylate the adaptor mol- ecule DNAX adaptor protein 12 (DAP12) after which the cellular response is initiated8,9. Only recently, binding of TREM-1 to a complex of peptidoglycan recognition protein 1 (PGLYRP1) and bacterially derived peptidoglycan has been demonstrated10. TREM-1 is expressed on neutrophils and monocyte subsets11 and amplifies pro-inflammatory responses in vitro, enhancing the TLR-response to specific TLR ligands12. There are con- flicting reports on the role of TREM-1 in in vivo infection models. TREM-1 deficiency impaired bacterial clearance in a model of Klebsiella pneumonia-induced liver abscess formation13, pneumococcal14 and Pseudomonas (P.) aeruginosa pneumonia15. However, 6 blocking TREM-1 with an analogue synthetic peptide derived from the extracellular moiety of TREM-1 (LP17) actually improved survival during P. aeruginosa pneumonia16 and endotoxaemia in rodent models17. Interestingly, in a murine pneumonia model of Legionella pneumonia no impact of TREM-1 deficiency was found on bacterial clearance or neutrophil influx towards the primary site of infection18. TREM-2 is primarily expressed on macrophages, dendritic cells, microglia and osteo- clasts19-22. In contrast to TREM-1, TREM-2 acts as a negative regulator of inflammatory responses in macrophages and dendritic cells19,21. In addition, TREM-2 is involved in phagocytosis23,24 and killing of bacteria by macrophages25. Blocking TREM-2 in vivo by a recombinant protein in a polymicrobial sepsis model revealed that TREM-2 is required for bacterial clearance and improves survival26. In contrast, TREM-2 plays a detrimental role during pneumococcal pneumonia24. Melioidosis, considered to be an excellent model for Gram-negative sepsis, is caused by the Tier 1 biological treat agent Burkholderia pseudomallei27,28. Melioidosis is charac- terized by pneumonia and abscess formation and an important cause of community- acquired sepsis in Southeast Asia and Northern Australia27. The high mortality rate,

107 Chapter 6 that can approach 40%, and the emerging antibiotic resistance of B.pseudomallei29 emphasize the need to better understand the pathogenesis of melioidosis, which could ultimately lead to novel treatment strategies. We previously found increased soluble (s) TREM-1 plasma levels and TREM-1 surface expression on monocytes of patients with melioidosis30, suggesting an important role for TREM-1 in the host defense against B. pseudomallei. Treatment with a peptide mimicking a conserved-domain of sTREM-1 partially protected mice from B. pseudomallei induced lethality30. In this study we now examine the role of TREM-1 and TREM-2 during experimental melioidosis, utilizing recently generated Trem-1/3-deficient (Trem-1/3−/−)15 and Trem-2- deficient (Trem-2−/−) mice19 to determine their contribution to the host response against B. pseudomallei. We hypothesized that TREM-1 deficiency would decrease inflammation and improve survival during experimental melioidosis while TREM-2 deficiency would instead lead towards increased inflammation and a worsened survival. Unexpectedly however, we found that TREM-2, but not TREM-1, plays an important detrimental role during melioidosis. TREM-2 deficiency improves survival of B. pseudomallei infected mice, by limiting inflammation and organ damage. These data identify TREM-2 as a potential treatment target for sepsis caused by B. pseudomallei.

Materials and methods

Detailed methods are provided in the supplement.

Mice Pathogen-free 8- to 10-week-old male wild-type (WT) C57BL/6 mice were purchased from Charles River (Leiden, The Netherlands). Trem-1/3−/− 6,14 and Trem-2−/− 19 mice were backcrossed >97% to a C57BL/6 genetic background. TREM-1 and -2 deficiency was con- firmed by polymerase chain reaction. All mice were kept at the animal care facility of the Academic Medical Center (University of Amsterdam) with unlimited access to food and water. The Animal Care and Use of Committee of the University of Amsterdam approved all experiments.

Experimental infection Experimental melioidosis was induced by intranasal inoculation with 5 × 102 colony forming units (CFU) of B. pseudomallei strain 1026b (a clinical isolate) as described31-33. For survival experiments mice were observed 4-6 times daily, up to 14 days post-infection. Sample harvesting, processing, and determina­tion of bacterial growth were performed as described.32,33.

108 TREM-2 impairs host defense in Gram-negative sepsis (melioidosis)

TREM-1 and TREM-2 expression Total RNA was isolated using the Isolate II RNA mini kit (Bioline, Taunton, MA, USA), treated with DNase (Bioline) and reverse transcribed using an oligo(dT) primer and Moloney murine leukemia virus RT (Promega, Madison, WI, USA). Primers and RT-PCR conditions can be found in the supplemental data (Supplemental Digital Content 1) Data were analyzed using the comparative Ct method.

Assays Plasma mouse tumour necrosis factor-α (TNF-α), interleukin (IL)-6, IL-10, IL-12p70, monocyte chemoattractant protein-1 (MCP-1) and interferon (IFN)-γ were measured by cytometric bead array multiplex assay (BD Biosciences, San Jose, CA, USA). Keratinocyte chemoattractant (KC), TNF-α, IL-1ß and IL-6 levels in lung and BALF were measured by ELISA (R&D systems, Minneapolis, MN). Alanine aminotransaminase34 and aspartate aminotransferase (AST), blood urea nitrogen (BUN) and lactate dehydrogenase (LDH) levels were determined with the Cobas 8000 module c702 (Roche Diagnostics).

(Immuno)histology Paraffin-embedded 4-μm lung, liver and spleen sections were stained with haematoxylin and eosin and analyzed for inflammation and tissue damage, as described previously14,33. Granulocyte (Ly6G) staining was done exactly as described previously35.

Whole blood and macrophage stimulation Whole blood, alveolar macrophages (AM) and bone-marrow derived macrophages 6 (BMDM) were harvested from naïve WT and Trem1/3−/− and Trem-2−/− mice as de- scribed33,36,37 and stimulated overnight with either medium, ultrapure LPS (Invivogen, San Diego, CA, USA) or B. pseudomallei (107 CFU/ml or MOI of 50), after which superna- tant was harvested and stored at -20°C until assayed for TNF-α.

Phagocytosis and bacterial killing Phagocytosis was determined as described previously38. In brief, AM and BMDM (5 × 104 cells/well) were incubated with or without heat-inactivated FITC-labelled B. pseudomal- lei (MOI 50) for 60 and/or 120 minutes at 37°C and 5% CO2 air and internalization was measured directly after collection by flow cytometry. Bacterial killing was evaluated as described36,39. In short, BMDM were incubated with to log-phase grown B. pseudomallei (MOI 30) for 20 minutes at 37°C in 5% CO2 air, after which they were washed and incubated with kanamycin 250 µg/ml for 30 minutes at 36 37°C in 5% CO2 air (this point was taken as time zero) . At designated time points the BMDM were washed and lysed and appropriate dilutions of these lysates were plated onto blood-agar plates and incubated at 37°C for 24–48 h before CFU were counted.

109 Chapter 6

Statistical analysis Values are expressed as mean ± standard error of the mean (SEM). Differences between groups were analyzed by Mann-Whitney U test. For survival analysis, Kaplan-Meier analysis followed by log-rank test was performed. These analyses were performed using GraphPad Prism version 5.01. Values of P < 0.05 were considered statistically significant.

Results

Increased TREM-1 and TREM-2 expression in the lung and liver during experimental melioidosis Septic melioidosis patients present with pneumonia and bacterial dissemination to distant body sites27,40. Since it is not feasible to study TREM-1 and TREM-2 mRNA ex- pression at tissue level in these patients, we used our well-established murine model of pneumonia-derived melioidosis in which mice are intranasally infected with a lethal dose of B. pseudomallei32,33. Mice were killed at 0, 24, and 72h after infection (i.e., directly before the first predicted death), and TREM-1/-2 mRNA expression was determined in lungs and livers. At baseline, TREM-1 and TREM-2 expression was low, corresponding with our previous data on sTREM-1 levels in melioidosis patients30, TREM-1 was strongly up-regulated in lung and liver tissue (P < 0.05 lung at 24h, P < 0.01 liver at 72h; Fig. 1, A and B). TREM-2 mRNA expression was increased in experimental melioidosis as well (P < 0.05 for both lung and liver; Fig. 1, C and D). The increase in both TREM-1 and TREM-2 expression was much more pronounced at the primary site of infection, the pulmonary compartment, when compared to the hepatic compartment.

Figure 1: Increased TREM-1 and TREM-2 expression in experimental melioidosis. TREM-1 and TREM-2 mRNA expression was determined in wild type (WT) mice priorto infection or at 24 or 72h post-infection with 5 × 102 CFU B.pseudomallei intranasally. TREM-1 mRNA expression in lung (A) and liver (B) was deter- mined. Likewise, TREM-2 mRNA expression was measured in lung (C) and liver (D) tissue. Data arepresented as fold induction compared to the mRNA expression in uninfected mice (all RNA data are normalized to GAPDH). Data are mean ± SEM, n = 4-5 mice/group. *P < 0.05, **P < 0.01, compared to gene-expression at t = 0h (Mann-Whitney U test).

110 TREM-2 impairs host defense in Gram-negative sepsis (melioidosis)

Trem-2−/− mice, but not Trem-1/3−/− mice, are protected from B. pseudomallei- induced mortality Having established that both TREM-1 and TREM-2 are highly up-regulated during meli- oidosis, we further investigated the involvement of these receptors in the outcome of melioidosis. Therefore, we infected WT, Trem-1/3−/− and Trem-2−/− mice intranasally with a lethal dose of B. pseudomallei and observed them for 14 days (Fig. 2). There was no significant difference in survival between Trem-1/3−/− and WT mice following a lethal B. pseudomallei challenge: 95% of Trem-1/3−/− and WT mice died within 6 days after inocu- lation (Fig. 2A). Strikingly however, Trem-2−/− mice were significantly protected: 70% of Trem-2−/− survived until the end of the 14-day observation period while all WT mice died within 6 days (P < 0.001; Fig. 2B).

Figure 2: Survival of Trem-2−/− mice, but not of Trem-1/3−/− mice, is enhanced in experimental melioidosis. Survival was observed for every 4-6h, up to a maximum of 14 days after intra- nasal inoculation with 5 × 102 CFU B. pseudomallei in wild-type (WT; closed circles) and Trem-1/3−/− mice (open circles; A). Similarly, survival of WT (closed circles) and Trem-2−/− mice (open circles) was determined (B) (n = 20 per group). P values indicate significance of the difference in survival between Trem-2−/− and WT mice (Kaplan-Meier analysis, followed by a log-rank test). ns = not significant. 6

Enhanced bacterial clearance in Trem-2−/− mice To substantiate the finding that Trem-2−/− mice are protected during melioidosis, we determined bacterial loads in lung and BALF as well as in blood, liver and spleen 72h

Figure 3: TREM-2 deficiency reduces bacterial loads and dissemination. WT and Trem-2−/− mice were infected with 5 × 102 colony forming units (CFU) of B. pseudomallei intranasally (n = 5-6 mice per group). To determine bacterial loads, WT (closed circles) and Trem-2−/− (open circles) mice were sacrificed 72 h post- infection and CFUs were determined in lung homogenates (A), broncho-alveolar lavage fluid (BALF) B( ), whole blood (C), liver (D) and spleen (E). Data are expressed as mean ± SEM, n = 5-6/group. ** P < 0.01. BC+ denotes positive blood cultures (Mann-Whitney U test).

111 Chapter 6 post-infection. Relative to WT mice, Trem2−/− mice displayed strongly reduced bacterial loads both at the primary site of infection (P < 0.01 for lung and BALF; Fig. 3, A and B) as well as in distant organs and the systemic compartment (P < 0.01 for blood and spleen; Fig. 3, C-E). 72h post-infection 100% of WT but only 20% of Trem2−/− mice had become bacteraemic. These findings indicate that TREM-2 plays a key deleterious role during experimental melioidosis by antagonizing bacterial clearance leading to increased dis- semination of infection.

Trem-2−/− mice demonstrate reduced lung inflammation Since TREM-2 has been described as a negative regulator of inflammation19,20, we next assessed the inflammatory response in the pulmonary compartment. Therefore we stud- ied the extent of inflammation in lung homogenates and BALF. We observed markedly decreased levels of pro-inflammatory cytokines TNF-α, IL-6, IL-1β and the chemokine KC in both lung homogenates and BALF of TREM-2 deficient mice compared to controls

Table 1. Cytokine response in lung homogenates, BALF and plasma of WT and Trem-2−/− mice during experimental melioidosis. T = 72h WT Trem-2−/− pg/ml Lung homogenate TNF-α 1680 ± 222 512 ± 156 ** IL-6 7025 ±1408 450 ± 66* KC 59588 ± 9304 10580 ± 2233** IL-1β 31292 ± 4975 1860 ± 516** BALF TNF-α 7054 ± 1578 1689 ± 171* IL-6 18478 ± 4471 406 ± 204 ** KC 30702 ± 6626 1055 ± 454** IL-1β 10469 ± 2424 217 ± 81** Plasma TNF-α 2324 ± 909 91 ± 19** IL-6 2641 ± 526 31 ± 5** IL-10 11 ± 3 5 ± 0** MCP-1 1675 ± 453 11 ± 1** IFN-γ 158 ± 62 17 ± 3** IL-1β 1123 ± 303 125 ± 25* Cytokine levels in lung homogenate, broncho-alveolar fluid (BALF) and plasma were measured after intra- nasal infection with 5 × 102 CFU wild-type B. pseudomallei. Wild-type (WT) and Trem-2−/− mice were sacri- ficed 72 h after infection. Data are represented as means ± SEM (n = 5-6/group). TNF-α = Tumor necrosis factor-α; IL = Interleukin; MCP-1 = Monocyte Chemoattractant Protein-1; KC = Keratinocyte Chemoattrac- tant; IFN-γ = Interferon-γ; * P < 0.05; ** P < 0.01.

112 TREM-2 impairs host defense in Gram-negative sepsis (melioidosis)

(P < 0.01-0.05; Table 1). To further obtain insight into the involvement of TREM-2 in the in- flammatory response followingB. pseudomallei infection, we semi-quantitatively scored lung histology slides generated from Trem-2−/− and WT mice. However, all mice displayed severe pulmonary inflammation and no differences were observed between the mouse strains (Fig. 4, A-C). Neutrophil recruitment to the lung is an essential part of the inflam- matory host response to melioidosis31,41,42. Therefore, we investigated the granulocyte influx into the pulmonary compartment as analyzed by Ly6G-immunostaining in WT and Trem-2−/− mice 72h post-infection with B. pseudomallei (Fig. 4, D-F). Corresponding to the diminished bacterial loads and decreased levels of cyto- and chemokines in lung tissue, a reduced influx of granulocytes in lungs of Trem-2−/− mice was found (P < 0.05, Fig. 4D).

6

Figure 4: Reduced neutrophil influx in lungs of Trem-2−/− mice, without affecting lungpathology. Lung pathology was determined in WT (black bars) and Trem-2−/− mice (white bars) infected with 5 × 102 CFU B. pseudomallei at 72h post-infection as described in the Methods section (A). Representative lung slides of WT (B) and Trem-2−/− mice (C) (original magnification 10×). Neutrophil influx was defined by Ly6G positivity (expressed as % of total lung surface; D). Representative photographs of Ly6G-immunostaining for granu- locytes on lung slides of WT (E) and Trem-2−/− mice (F) (original magnification 10×). Data are expressed as mean ± SEM, n = 5-6 mice per group per time point. *P < 0.05. (Mann-Whitney U test).

113 Chapter 6

Trem-2 deficiency leads to decreased distant organ injury during experimental melioidosis To evaluate the role of TREM-2 in the systemic inflammatory response, we determined plasma cytokine levels 72h post-infection with B. pseudomallei. Consistent with the lower pulmonary cytokine levels and bacterial loads, we found that the plasma levels of TNF-α, IL-6, IL-1β, MCP-1, IL-10, IFN-γ and KC were all significantly reduced in Trem- 2−/− mice compared to WTs (P < 0.01-0.05, Table 1). Furthermore, we obtained spleen pathology scores and performed routine clinical chemistry tests to evaluate hepatic, renal and systemic injury. In line with the observed decreased splenic bacterial loads, Trem-2−/− mice showed less inflammation compared to WT mice 72h after inoculation with B. pseudomallei (P < 0.05; Fig. 5A). Plasma AST levels of Trem-2−/− mice were de- creased when compared to controls 72h post-infection, reflecting decreased hepatocel- lular injury in these animals (P < 0.05; Fig. 5B). Consistently, we observed a trend towards lower ALT, BUN and LDH levels in Trem-2−/− mice compared to controls suggesting better renal function and less organ damage respectively (Fig. 5, C-E).

Figure 5: Reduced distant organ damage in Trem-2−/− mice. At 72h post-infection with 5 × 102 CFU B. pseudomallei intranasally splenic injury (A) in WT (black bars) and Trem-2−/− mice (white bars) was quantified as described in the Methods section. Plasma levels of aspartate transaminase (AST; B), alanine transaminase (ALT; C), Lactate dehydrogenase (LDH; D) and blood urea nitrogen (BUN; E) in WT and Trem-2−/− mice were determined. Data are expressed as mean ± SEM. n = 5-6 mice per group per time point. *P < 0.05 (Mann- Whitney U test).

114 TREM-2 impairs host defense in Gram-negative sepsis (melioidosis)

Lack of TREM-2 leads to a reduced inflammatory response ex vivo, but does not impact on phagocytosis of B. pseudomallei by macrophages Having established that TREM-2 plays an important deleterious role during experi- mental melioidosis and is involved in the inflammatory response, we next assessed what cells are responsible for these effects. It is known that blood monocytes, alveolar macrophages (AM) and BMDM express TREM-224, therefore we harvested these cells and first stimulated them overnight with the TLR4-ligand LPS and B. pseudomallei. We found a clear trend towards lower TNF-α levels when whole blood, AM or BMDM of Trem-2−/− mice were stimulated with LPS (Fig. 6, A-C). This effect was even more pronounced after stimulation with B. pseudomallei: the TNF-α response of whole blood and BMDM derived from TREM-2 deficient mice was significantly reduced compared to controls (P < 0.05; Fig. 6, A and B). Considering TREM-2’s known phagocytic properties23,24 and the observed

6

Figure 6: TREM-2 deficiency reduces cellular responsiveness ex vivo. Whole blood (A), bone marrow derived macrophages (BMDM; B) and alveolar macrophages (AM; C) of WT (black bars) and Trem-2−/− mice (white bars) were stimulated with medium, E.coli LPS (100 ng/ml) or heat-inactivated B. pseudomallei (107 CFU/ml or MOI of 50). Supernatant was collected after 20 h of stimulation and assayed for TNF-α. In addi- tion, WT and Trem-2−/− BMDM (D) and AM (E) were incubated at 37°C with FITC labeled heat-inactivated B. pseudomallei after which time-dependent phagocytosis was determined. Data are presented as mean ± SEM and are representative of two or three independent experiments. n = 4 or 8 per group. * P < 0.05 (Mann-Whitney U test).

115 Chapter 6 lower local and systemic bacterial loads in TREM-2-deficient mice, we determined the phagocytic capacity of AM and BMDM harvested from WT and Trem-2−/− mice. Despite a trend towards enhanced phagocytosis of FITC-labelled B. pseudomallei by TREM-2 defi- cient macrophages, no significant differences were found (Fig. 6, D and E). In line, TREM-2 did not impact on the intracellular killing of B. pseudomallei by BMDM (data not shown).

Limited role of TREM-1/3 in the host defense during experimental melioidosis In a final set of experiments we studied the role of TREM-1 in the host defense against B. pseudomallei using Trem-1/3−/− mice. In contrast to the data derived from Trem-2−/− mice, no differences in bacterial counts in lung or BALF were observed betweenB. pseudomallei- challenged Trem-1/3−/− and WT mice (Fig. 7, A and B). In line, TREM-1 deficiency did not impact on lung pathology and cytokine levels, except for decreased KC levels, which did not influence pulmonary neutrophilic content as determined by Ly6-stainings (Fig. 7, E and F and Table 2). However, TREM-1 did influence bacterial dissemination as bacterial loads in blood and liver were significantly decreased inTrem-1/3 −/− mice compared to WTs 72h after infection (P < 0.01; Fig. 7, C and D). We next evaluated TREM-1’s role in systemic inflammation and end organ damage. At 72h post-infection, the levels of key regulatory cytokines in the systemic compartment (TNF-α, IL-6, IL-10, MCP-1 and IFN-γ) did not differ between Trem-1/3−/− mice and WT (Table 2). Induced pathology of the spleen (Fig.7G) and

Figure 7: Effect of TREM-1 deficiency on bacterial clearance, pulmonary neutrophil influx and or- gan damage during experimental melioidosis. WT (closed circles/black bars) and Trem-1/3−/− mice (open circles/ white bars) were intranasally infected with 5 × 102 CFU of B. pseudomallei and sacrificed 24 and 72 h post-infection, followed by determination of bacterial loads in lung homogenate (A), BALF (B), blood (C) and liver (D). Neutrophil influx as determined by % Ly6G positive surface of lung slides was calculated for WT and Trem-1/3−/− mice (E). Lung (F) and spleen (G) pathology was scored as described in the Methods section. Aspartate transaminase (AST; H), alanine transaminase (ALT; I), lactate dehydrogenase (LDH; J) and blood urea nitrogen (BUN; K) were measured as a marker for end organ damage. Data are expressed as mean ± SEM. n = 7-8 mice per group. *P < 0.05; **P < 0.01 (Mann-Whitney U test).

116 TREM-2 impairs host defense in Gram-negative sepsis (melioidosis)

Table 2. Cytokine responses in lung homogenates, BALF and plasma of WT and Trem-1/3−/− mice dur- ing experimental melioidosis. T = 24h T = 72h WT Trem-1/3−/− WT Trem-1/3−/− pg/ml Lung homogenate TNF-α 1406 ± 220 1227 ± 215 2135 ± 312 2312 ± 386 IL-6 2380 ± 293 2913 ± 353 17694 ± 3121 29910 ± 4668 KC 18817 ± 5116 14277 ± 1981 71606 ± 6071 69866 ± 8092 pg/ml BALF TNF-α 883 ± 100 808 ±95 2472 ± 359 3624 ± 831 IL-6 1675 ± 285 654 ± 97** 6041 ± 942 9449 ± 1914 KC 992 ± 183 711 ± 113 19039 ± 1614 12251 ± 1678* pg/ml Plasma TNF-α 12 ± 1 9 ±1 616 ± 160 307 ± 77 IL-6 143 ± 36 133 ± 19 2868 ± 818 2695 ± 313 IL-10 8 ± 2 20 ± 6 199 ± 77 81 ± 36 MCP-1 263 ± 63 92 ± 25* 2335 ± 94 2975 ± 226 IFN-γ 28 ± 4 26 ± 4 1606 ± 445 1275 ± 242 Cytokine levels in plasma, lung homogenate and broncho-alveolar fluid (BALF) were measured after in- tranasal infection with 5 × 102 CFU wild-type B. pseudomallei. Wild-type (WT) and Trem-1/3−/− mice were sacrificed 24 and 72 h after infection. Data are represented as means ± SEM (n = 7 or 8/group per time point). TNF-α = Tumor necrosis factor-α; IL = Interleukin; MCP-1 = Monocyte Chemoattractant Protein-1; KC = Keratinocyte Chemoattractant; IFN-γ = Interferon-γ; *P < 0.05; **P < 0.01. liver (data not shown) was similar in Trem-1/3−/− and WT mice. In correspondence with the 6 lower hepatic bacterial counts at 72h, we found lower levels of the hepatocellular injury markers AST and ALT levels in Trem-1/3−/− mice compared to WT mice. LDH levels, reflecting general organ injury, were elevated in Trem-1/3−/− mice at 24 h, while they were reduced compared to their WT counterparts at 72h post-infection (P < 0.05; Fig. 7J). No difference in plasma BUN levels was observed between mice strains (Fig. 7K).

TREM-1 deficiency does not impact on ex vivo cytokine responsiveness and phagocytosis nor intracellular killing of B. pseudomallei TREM-1 is abundantly expressed on monocytes and macrophages following exposure to B. pseudomallei30. In line with previous findings11, Trem-1/3−/− BMDM produced less TNF-α in response to LPS stimulation (P < 0.05; Fig. 8D). Surprisingly, no differences in cellular responsiveness were found between AM and whole blood derived from WT and Trem-1/3−/−mice (Fig.8 A-C). Lastly, we wished to determine whether TREM-1 contributes to phagocytosis and/or killing of B. pseudomallei. No differences in phagocytic and kill- ing capacities between WT and TREM deficient BMDM or AM were observed (Fig. 8, data not shown).

117 Chapter 6

Figure 8: No effect of TREM-1 deficiency on the cellular responsiveness and phagocytosis or intra- cellular killing of B. pseudomallei. Whole blood (A), bone marrow derived macrophages (BMDM; B) and alveolar macrophages (AM; C) of WT and Trem-1/3−/− mice were stimulated with medium, E.coli LPS(100 ng/ ml) or heat-inactivated wild type B. pseudomallei (107 CFU/ml at a MOI of 50). TNF-α levels were measured in the supernatant obtained after 20 h of stimulation. BMDM (D) and AM (E) of WT and Trem-1/3−/− mice were incubated at 37°C with FITC labeled heat-inactivated B. pseudomallei after which time-dependent phagocytosis was determined. Data are expressed as mean ± SEM and are representative of two or three independent experiments. n = 4 or 8 (for the whole blood assay) per group. *P < 0.05 (Mann-Whitney U test).

Discussion

TREM-1 and TREM-2 are innate immune receptors that have demonstrated to either amplify or regulate TLR and NLR signaling after recognition of pathogen-associated molecular patterns. Our study is the first to examine the role of both TREM-1 and TREM-2 during experimental melioidosis. We observed increased TREM-1 and TREM-2 expres- sion during experimental melioidosis, both at the local site of infection and systemically. Subsequently, we found that TREM-2 impairs the host defense against B. pseudomallei- induced sepsis, as demonstrated by an improved survival of infected Trem-2−/− mice as a direct result of diminished bacterial dissemination, decreased inflammation and less or- gan damage. Our ex vivo studies suggest that the protective effect of TREM-2 deficiency in part results from the diminished capacity of TREM-2-deficient macrophages to elicit a pro-inflammatory response which is an important contributor to organ injury in the event of sepsis. TREM-1 was also found to play a detrimental role during B. pseudomallei

118 TREM-2 impairs host defense in Gram-negative sepsis (melioidosis) infection, which is in line with our earlier finding that blocking TREM-1 could improve survival during melioidosis30. However when compared to TREM-2 the role of TREM-1 in the host response against B. pseudomallei seems to be limited Previous studies have demonstrated that soluble TREM-1 levels are up-regulated in plasma of patients with sepsis, pneumonia and melioidosis30,43,44. In addition, it is known that surface TREM-1 expression is increased on monocytes of melioidosis pa- tients30. However, soluble TREM-1 levels in septic patients do not always correlate to the expression of membrane-bound TREM-1 on different myeloid cell types30,45. Less is known about the kinetics of TREM-2 expression during infection. A recent study demon- strated that during sepsis TREM-2 expression on ascites-retrieved cells of patients with abdominal sepsis was increased26. Correspondingly, TREM-2 was up-regulated on AM of mice infected with S. pneumoniae24. In line with these earlier studies, we now show that both TREM-1 and TREM-2 mRNA expression is elevated in lung and liver tissue of mice infected with B. pseudomallei. The in vivo role of TREM-2 in infectious diseases remains ill defined. In a model studying P. aeruginosa keratitis TREM-2 deficiency increased corneal bacterial loads46. More recently, Chen et al. demonstrated that TREM-2 is required for efficient bacterial clearance in a murine polymicrobial sepsis model using a TREM-2 blocking recombinant protein26. In the same study it was shown that administration of TREM-2 overexpressing bone marrow derived myeloid cells improved survival during polymicrobial sepsis, but not endotoxaemia26. In sharp contrast, Sharif et al. demonstrated a beneficial effect of TREM-2 deficiency during S. pneumoniae pneumonia24. To evaluate how TREM-2 defi- ciency led to increased clearance of B. pseudomallei, we assessed the functional roles 6 of macrophages that express TREM-219,24. TREM-2 is known to be involved in direct kill- ing26,46 and phagocytosis of bacteria by macrophages23,24. Interestingly, we did not find impaired bacterial killing or phagocytosis of B. pseudomallei by BMDM or AM of Trem-2−/− mice. Several characteristics of this facultative intracellular bacterium when compared to other bacteria might in part explain these discrepancies; B. pseudomallei is capable of invading both phagocytic and non-phagocytic cells47 and circumvents intracellular defense mechanisms efficiently in order to replicate and spread to adjacent cells48,49. TREM-2 is traditionally regarded as a negative regulator of the in vitro inflammatory response in response to TLR-ligands19,21. In contrast, our study now demonstrates that TREM-2 deficiency leads to a reduced inflammatory response to B. pseudomallei both ex vivo and in vivo suggesting that TREM-2’s role during inflammation may not be that upfront. This is in line with recent studies investigating the role of TREM-2 in models of pneumococcal pneumonia24, post-stroke inflammation50 and DSS-induced colitis51. Different elements, can explain these inconsistencies: differences in mice strains used (BALB/C versus C57Bl/6), different experimental murine models (e.g. caecal ligation and puncture (CLP)- model versus a intranasal inhalation model for sepsis), differences in

119 Chapter 6

TREM-2 blockade (e.g. by using TREM-2 deficient mice or TREM-2 antibodies) and lastly the difference of an in vitro approach in contrast to our ex vivo cellular challenge model. Of importance, neutrophil recruitment to the lung, an important defense mechanism during melioidosis31,42, was reduced in Trem-2−/− mice during experimental melioidosis as a potential result of the decreased inflammatory response and production of che- mokines following infection. In this respect, it is noteworthy, that IL-1β – which we and others have shown to be involved in excessive deleterious neutrophil influx during experimental melioidosis37,52 – was also significantly reduced inTrem-2 −/− mice. Excessive inflammation and neutrophil activation can lead towards multi-organ failure, which is almost universally seen in lethal cases of melioidosis. Distant organ injury was signifi- cantly reduced in Trem-2−/− mice, potentially as a result of a reduced influx of inflamma- tory cells. Trem-2−/− mice displayed an evidently reduced inflammatory response, which resulted in a strong survival benefit. In addition, it is well known thatB. pseudomallei can replicate intracellularly27, and neutrophils may act as its permissive host cell52. We could therefore hypothesize that the reduced bacterial loads seen in TREM-2 deficient mice are a result of decreased intracellular bacterial replication at the infection site, due to reduced neutrophilic influx. Taken together, during melioidosis, TREM-2 deficiency resulted in a restricted inflam- matory response, thereby decreasing organ damage and mortality. Future research should focus on the potential of anti-TREM-2 treatment of B.pseudomallei-infected mice. TREM-1 amplifies TLR-responses and therefore might dangerously enhance the inflammatory response to bacterial infection18. Controversial results have been found on the role of TREM-1 during bacterial infection. TREM-1 deficiency has shown to be detrimental during endotoxaemia17 and polymicrobial sepsis12,53, while in contrast, mod- erate levels of TREM-1 can improve survival during polymicrobial sepsis, but not endo- toxaemia54. Blockade of TREM-1 with a peptide called LP17 could partially protect mice from B. pseudomallei induced lethality30. In this study however, we observed, using the same infection model, that survival of B. pseudomallei-infected TREM-1-deficient mice was similar to WTs. This might be explained by the fact that these mice were completely TREM-1-deficient and in addition lacked TREM-3, a DAP12-coupled activating receptor on murine macrophages55. In contrast, in humans TREM-3 is a pseudogene55. However, since DAP12 is known to both potentiate and attenuate TLR-signaling, it is perhaps not surprising that the net-effect on bacterial clearance of B. pseudomallei is not affected. TREM-1 has other functions next to TLR-signaling enhancement, such as phagocytosis and the production of reactive oxygen species56. Furthermore, TREM-1 has been recently linked to trans-epithelial migration of neutrophils after infection with P. aeruginosa15. Blocking TREM-1 completely could therefore interfere with these important antibac- terial mechanisms. We did not find a role for TREM-1 in the killing or phagocytosis of B. pseudomallei, which is in line with the fact that TREM-1/3 deficiency in neutrophils

120 TREM-2 impairs host defense in Gram-negative sepsis (melioidosis) neither impacts on bacterial killing, phagocytosis and chemotaxis of P. aeruginosa15. This suggests that other phagocytic receptors on leukocytes are more important for the efficient eradication of B. pseudomallei38,41,57. In conclusion, we here demonstrate that human and murine melioidosis are associ- ated with increased TREM-1 and -2 expression. TREM-2 deficiency is beneficial during experimental Gram-negative sepsis caused by a clinical relevant pathogen, resulting in lower bacterial loads, reduced organ damage, decreased inflammation and improved survival. When compared to TREM-2, TREM-1 plays a limited detrimental role during experimental melioidosis. These results provide new information on the expression and function of TREM-2 during melioidosis and demonstrate its potential therapeutic usefulness.

Acknowledgements

We thank Marieke ten Brink and Joost Daalhuisen for expert technical assistance. We are grateful for the help of Regina de Beer who performed histopathological and immunohistochemical stainings, and Onno de Boer for his assistance in analyzing im- munohistologically stained slides.

6

121 Chapter 6

References

1. Hotchkiss RS, Monneret G, Payen D. Sepsis-induced immunosuppression: from cellular dysfunc- tions to immunotherapy. Nat Rev Immunol 2013;​13:​862‑74. 2. Angus DC, van der Poll T. Severe sepsis and septic shock. N Engl J Med 2013;​369:​2063. 3. Wiersinga WJ. Current insights in sepsis: from pathogenesis to new treatment targets. Curr Opin Crit Care 2011;​17:​480‑6. 4. Broz P, Monack DM. Newly described pattern recognition receptors team up against intracellular pathogens. Nat Rev Immunol 2013;​13:​551‑65. 5. Arts RJ, Joosten LA, van der Meer JW, Netea MG. TREM-1: intracellular signaling pathways and interaction with pattern recognition receptors. J Leukoc Biol 2013;​93:​209‑15. 6. Klesney-Tait J, Turnbull IR, Colonna M. The TREM receptor family and signal integration. Nat Im- munol 2006;​7:​1266‑73. 7. Sharif O, Knapp S. From expression to signaling: roles of TREM-1 and TREM-2 in innate immunity and bacterial infection. Immunobiology 2008;​213:​701‑13. 8. Aoki N, Kimura S, Xing Z. Role of DAP12 in innate and adaptive immune responses. Curr Pharm Des 2003;​9:​7‑10. 9. Tessarz AS, Cerwenka A. The TREM-1/DAP12 pathway. Immunol Lett 2008;​116:​111‑6. 10. Read CB, Kuijper JL, Hjorth SA, et al. Cutting Edge: Identification of Neutrophil PGLYRP1 as a Ligand for TREM-1. J Immunol 2015. 11. Bouchon A, Dietrich J, Colonna M. Cutting edge: inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J Immunol 2000;​164:​4991‑5. 12. Bouchon A, Facchetti F, Weigand MA, Colonna M. TREM-1 amplifies inflammation and is a crucial mediator of septic shock. Nature 2001;​410:​1103‑7. 13. Lin YT, Tseng KY, Yeh YC, Yang FC, Fung CP, Chen NJ. Triggering Receptor Expressed on Myeloid Cells-1 promotes survival during Klebsiella pneumoniae liver abscess in mice. Infect Immun 2014. 14. Hommes TJ, Hoogendijk AJ, Dessing MC, et al. Triggering receptor expressed on myeloid cells-1 (TREM-1) improves host defence in pneumococcal pneumonia. J Pathol 2014;​233:​357‑67. 15. Klesney-Tait J, Keck K, Li X, et al. Transepithelial migration of neutrophils into the lung requires TREM-1. J Clin Invest 2013;​123:​138‑49. 16. Gibot S, Alauzet C, Massin F, et al. Modulation of the triggering receptor expressed on myeloid cells-1 pathway during pneumonia in rats. J Infect Dis 2006;​194:​975‑83. 17. Gibot S, Buonsanti C, Massin F, et al. Modulation of the triggering receptor expressed on the myeloid cell type 1 pathway in murine septic shock. Infect Immun 2006;​74:​2823‑30. 18. Weber B, Schuster S, Zysset D, et al. TREM-1 deficiency can attenuate disease severity without affecting pathogen clearance. PLoS Pathog 2014;​10:​e1003900. 19. Turnbull IR, Gilfillan S, Cella M, et al. Cutting edge: TREM-2 attenuates macrophage activation. J Immunol 2006;​177:​3520‑4. 20. Hamerman JA, Jarjoura JR, Humphrey MB, Nakamura MC, Seaman WE, Lanier LL. Cutting edge: inhibition of TLR and FcR responses in macrophages by triggering receptor expressed on myeloid cells (TREM)-2 and DAP12. J Immunol 2006;​177:​2051‑5. 21. Ito H, Hamerman JA. TREM-2, triggering receptor expressed on myeloid cell-2, negatively regu- lates TLR responses in dendritic cells. Eur J Immunol 2012;​42:​176‑85. 22. Takahashi K, Rochford CD, Neumann H. Clearance of apoptotic neurons without inflammation by microglial triggering receptor expressed on myeloid cells-2. J Exp Med 2005;​201:​647‑57.

122 TREM-2 impairs host defense in Gram-negative sepsis (melioidosis)

23. N’Diaye EN, Branda CS, Branda SS, et al. TREM-2 (triggering receptor expressed on myeloid cells 2) is a phagocytic receptor for bacteria. J Cell Biol 2009;​184:​215‑23. 24. Sharif O, Gawish R, Warszawska JM, et al. The triggering receptor expressed on myeloid cells 2 inhibits complement component 1q effector mechanisms and exerts detrimental effects during pneumococcal pneumonia. PLoS Pathog 2014;​10:​e1004167. 25. Zhu M, Li D, Wu Y, Huang X, Wu M. TREM-2 promotes macrophage-mediated eradication of Pseu- domonas aeruginosa via a PI3K/Akt pathway. Scand J Immunol 2014. 26. Chen Q, Zhang K, Jin Y, et al. Triggering receptor expressed on myeloid cells-2 protects against polymicrobial sepsis by enhancing bacterial clearance. Am J Respir Crit Care Med 2013;​188:​ 201‑12. 27. Wiersinga WJ, Currie BJ, Peacock SJ. Melioidosis. N Engl J Med 2012;​367:​1035‑44. 28. Simpson AJ. Melioidosis: a clinical model for gram-negative sepsis. J Med Microbiol 2001;​50:​ 657‑8. 29. Schweizer HP. Mechanisms of antibiotic resistance in Burkholderia pseudomallei: implications for treatment of melioidosis. Future Microbiol 2012;​7:​1389‑99. 30. Wiersinga WJ, Veer C, Wieland CW, et al. Expression profile and function of triggering receptor expressed on myeloid cells-1 during melioidosis. J Infect Dis 2007;​196:​1707‑16. 31. Wiersinga WJ, de Vos AF, de Beer R, et al. Inflammation patterns induced by different Burkholderia species in mice. Cell Microbiol 2008;​10:​81‑7. 32. Weehuizen TA, Wieland CW, van der Windt GJ, et al. Expression and function of transforming growth factor beta in melioidosis. Infect Immun 2012;​80:​1853‑7. 33. Wiersinga WJ, Wieland CW, Dessing MC, et al. Toll-like receptor 2 impairs host defense in gram- negative sepsis caused by Burkholderia pseudomallei (Melioidosis). PLoS Med 2007;​4:​e248. 34. Centers for Disease C, Prevention DoH, Human S. Possession, use, and transfer of select agents and toxins; biennial review. Final rule. Fed Regist 2012;​77:​61083‑115. 35. Kager LM, Wiersinga WJ, Roelofs JJ, et al. Endogenous protein C has a protective role during Gram-negative pneumosepsis (melioidosis). J Thromb Haemost 2013;​11:​282‑92. 6 36. Eske K, Breitbach K, Kohler J, Wongprompitak P, Steinmetz I. Generation of murine bone marrow derived macrophages in a standardised serum-free cell culture system. J Immunol Methods 2009;​ 342:​13‑9. 37. Koh GC, Weehuizen TA, Breitbach K, et al. Glyburide reduces bacterial dissemination in a mouse model of melioidosis. PLoS Negl Trop Dis 2013;​7:​e2500. 38. Wiersinga WJ, Kager LM, Hovius JW, et al. Urokinase receptor is necessary for bacterial defense against pneumonia-derived septic melioidosis by facilitating phagocytosis. J Immunol 2010;​184:​ 3079‑86. 39. Breitbach K, Sun GW, Kohler J, et al. Caspase-1 mediates resistance in murine melioidosis. Infect Immun 2009;​77:​1589‑95. 40. Currie BJ, Ward L, Cheng AC. The epidemiology and clinical spectrum of melioidosis: 540 cases from the 20 year Darwin prospective study. PLoS Negl Trop Dis 2010;​4:​e900. 41. Wiersinga WJ, van der Poll T, White NJ, Day NP, Peacock SJ. Melioidosis: insights into the pathoge- nicity of Burkholderia pseudomallei. Nat Rev Microbiol 2006;​4:​272‑82. 42. Easton A, Haque A, Chu K, Lukaszewski R, Bancroft GJ. A critical role for neutrophils in resistance to experimental infection with Burkholderia pseudomallei. J Infect Dis 2007;​195:​99‑107. 43. Gibot S, Kolopp-Sarda MN, Bene MC, et al. Plasma level of a triggering receptor expressed on myeloid cells-1: its diagnostic accuracy in patients with suspected sepsis. Ann Intern Med 2004;​ 141:​9‑15.

123 Chapter 6

44. Gibot S, Cravoisy A, Levy B, Bene MC, Faure G, Bollaert PE. Soluble triggering receptor expressed on myeloid cells and the diagnosis of pneumonia. N Engl J Med 2004;​350:​451‑8. 45. Oku R, Oda S, Nakada TA, et al. Differential pattern of cell-surface and soluble TREM-1 between sepsis and SIRS. Cytokine;​61:​112‑7. 46. Sun M, Zhu M, Chen K, et al. TREM-2 promotes host resistance against Pseudomonas aeruginosa infection by suppressing corneal inflammation via a PI3K/Akt signaling pathway. Invest Ophthal- mol Vis Sci;​54:​3451‑62. 47. Jones AL, Beveridge TJ, Woods DE. Intracellular survival of Burkholderia pseudomallei. Infect Im- mun 1996;​64:​782‑90. 48. Kespichayawattana W, Rattanachetkul S, Wanun T, Utaisincharoen P, Sirisinha S. Burkholderia pseudomallei induces cell fusion and actin-associated membrane protrusion: a possible mecha- nism for cell-to-cell spreading. Infect Immun 2000;​68:​5377‑84. 49. Allwood EM, Devenish RJ, Prescott M, Adler B, Boyce JD. Strategies for Intracellular Survival of Burkholderia pseudomallei. Front Microbiol 2011;​2:​170. 50. Sieber MW, Jaenisch N, Brehm M, et al. Attenuated inflammatory response in triggering receptor expressed on myeloid cells 2 (TREM2) knock-out mice following stroke. PLoS One;​8:​e52982. 51. Correale C, Genua M, Vetrano S, et al. Bacterial sensor triggering receptor expressed on myeloid cells-2 regulates the mucosal inflammatory response. Gastroenterology 2013;​144:​346-56 e3. 52. Ceballos-Olvera I, Sahoo M, Miller MA, Del Barrio L, Re F. Inflammasome-dependent pyroptosis and IL-18 protect against Burkholderia pseudomallei lung infection while IL-1beta is deleterious. PLoS Pathog 2011;​7:​e1002452. 53. Gibot S, Kolopp-Sarda MN, Bene MC, et al. A soluble form of the triggering receptor expressed on myeloid cells-1 modulates the inflammatory response in murine sepsis. J Exp Med 2004;​200:​ 1419‑26. 54. Gibot S, Massin F, Marcou M, et al. TREM-1 promotes survival during septic shock in mice. Eur J Immunol 2007;​37:​456‑66. 55. Chung DH, Seaman WE, Daws MR. Characterization of TREM-3, an activating receptor on mouse macrophages: definition of a family of single Ig domain receptors on mouse chromosome 17. Eur J Immunol 2002;​32:​59‑66. 56. Radsak MP, Salih HR, Rammensee HG, Schild H. Triggering receptor expressed on myeloid cells-1 in neutrophil inflammatory responses: differential regulation of activation and survival. J Immu- nol 2004;​172:​4956‑63. 57. Stevens MP, Galyov EE. Exploitation of host cells by Burkholderia pseudomallei. Int J Med Micro- biol 2004;​293:​549‑55.

124 TREM-2 impairs host defense in Gram-negative sepsis (melioidosis)

Supplement

Materials and Methods

Experimental infection 24 and 72 hours (h) after infection, mice were sacrificed under intraperitoneal anesthesia containing ketamin (Eurovet Animal Health, Bladel, The Netherlands) and medetomidin (Pfizer Animal Health Care, Capelle aan den IJssel, The Netherlands). Lungs, liver and spleen were harvested and homogenized at 4°C in 4 volumes of sterile saline using a tissue homogenizer (Biospec Products, Bartlesville, OK). Broncho-alveolar lavage (BAL) was performed by instilling two 0.5 ml aliquots of sterile saline into the airway through cannulation of the trachea with a sterile 22-gauge Abbocath-T catheter (Abbott, Sligo, Ireland). Serial dilutions of organ homogenates, BAL fluid (BALF) and blood were plated on blood agar plates and incubated at 37°C at 5% CO2 overnight. Lung homogenates were diluted 1:1 in Greenberger lysis buffer containing 300 mM NaCl, 30 mM Tris, 2 mM

MgCl2, 2 mMCaCl2, 1% Triton X-100 and protease inhibitors (Roche, Indianapolis, IN), incubated at 4°C for 30 min and centrifuged at 1730×g at 4°C for 10 min. Supernatants of lung and BALF and plasma were sterilized using 0.22 μm pore-size filters (Millipore, Billerica, MA) and stored at -20°C until further analysis.

Trem-1 and Trem-2 expression For RT-PCR on murine lung and liver tissues the following conditions were used: 6 min 6 at 95°C, followed by 45 cycles of amplification (95°C for 10 s, 60°C for 5 s, and 72°C for 15 s). Data were analysed by the comparative Ct method using GAPDH as the house- keeping gene and presented as a fold induction compared to gene-expression in naïve mice. The forward primer used for mTREM-1 was (5’- 3’) TGTTGTGCTCTTCCATCCTG and the reverse was (5’- 3’) GGGATCGGGTTGTAGTTGTG. For mTREM-2 these were (5’- 3’) AGATGACCAAGATGCTGGAGA and (5’- 3’) ATGCTGGCTGCAAGAAACTT respectively. For mGAPDH the forward primer (5’- 3’) ATGGCCTTCCGTGTTCCTAC and reverse primer (5’- 3’) AGGAGACAACCTGGTCCTCA were used. Oligonucleotides were purchased from Eurogentec (Maastricht, The Netherlands).

(Immuno)histology Lungs, livers and spleens for histology were harvested after infection, fixed in 100% formalin and embedded in paraffin. Sections of 4 μm were stained with haematoxy- lin–eosin (HE), and analysed by a pathologist who was blinded to the groups1-3. Lung pathology was scored based on affected surface, necrosis and/or abscess formation, interstitial inflammation, endothelialitis, bronchitis, edema, thrombus formation, and

125 Chapter 6 pleuritis. Each parameter was graded on a scale of 0 to 4. The total “mean histological score” was expressed as the sum of the scores for each parameter, (maximum of 32). Spleen and liver sections were scored on inflammation, necrosis/abscess formation, and thrombus formation using the scale given above (maximum score of 12). Granulocyte (Ly6G) staining was performed as previously described4 The Ly6G anti- body detects Ly6G, formerly known as myeloid differentiation antigen Gr-1, a 21–25- kDa GPI-anchored protein that can be found on granulocytes, including neutrophils and eosinophils. Slides were counterstained with methylgreen (Sigma-Aldrich, St. Louis, MO, USA). The total tissue area of the Ly-6-stained slides was scanned with a slide scanner (Olympus dotSlide, Tokyo, Japan) and the obtained scans were exported in TIFF format for digital image analysis. The digital images were analyzed with IMAGEJ (version 1.47v, National Institutes of Health, Bethesda, MD, USA) and the immunopositive (Ly6 +) area was expressed as the percentage of the total lung surface area.

Whole blood and macrophage stimulation Whole blood, alveolar macrophages (AM) and bone-marrow derived macrophages (BMDM) were harvested from naïve WT and Trem1/3−/− and Trem-2−/− mice as de- scribed2,5,6. 50 µl of whole blood was stimulated with 50 µl of stimulus in a 96-well V-bottom plate (Greiner Bio-one, Alphen a/d Rijn, the Netherlands), either plain RPMI 1640 (Life Technologies, Bleiswijk, The Netherlands), B. pseudomallei 2 × 107 CFU/ml or lipopolysaccharide (LPS) of E.coli O111:B4 (end concentration of 100 ng/ml) (Invitrogen,

San Diego, CA, USA) overnight at 37°C and 5% CO2 air after which supernatant was harvested and stored at -20°C until assayed. AM (5 × 104/well) and BMDM (1 × 105/well) were stimulated in a 96-well flat bottom plate (Greiner) either with RPMI + FCS 10% (Life technologies) B. pseudomallei (MOI 50) or LPS of E.coli (end concentration 100 ng/ml) at

37°C and 5% CO2 air overnight after which the supernatants were stored at -20°C until assayed.

126 TREM-2 impairs host defense in Gram-negative sepsis (melioidosis)

References

1. Hommes TJ, Hoogendijk AJ, Dessing MC, et al. Triggering receptor expressed on myeloid cells-1 (TREM-1) improves host defence in pneumococcal pneumonia. J Pathol 2014;​233:​357‑67. 2. Wiersinga WJ, Wieland CW, Dessing MC, et al. Toll-like receptor 2 impairs host defense in gram- negative sepsis caused by Burkholderia pseudomallei (Melioidosis). PLoS Med 2007;​4:​e248. 3. Wiersinga WJ, de Vos AF, de Beer R, et al. Inflammation patterns induced by different Burkholderia species in mice. Cell Microbiol 2008;​10:​81‑7. 4. Kager LM, Wiersinga WJ, Roelofs JJ, et al. Endogenous protein C has a protective role during Gram-negative pneumosepsis (melioidosis). J Thromb Haemost 2013;​11:​282‑92. 5. Eske K, Breitbach K, Kohler J, Wongprompitak P, Steinmetz I. Generation of murine bone marrow derived macrophages in a standardised serum-free cell culture system. J Immunol Methods 2009;​ 342:​13‑9. 6. Koh GC, Weehuizen TA, Breitbach K, et al. Glyburide reduces bacterial dissemination in a mouse model of melioidosis. PLoS Negl Trop Dis 2013;​7:​e2500.

6

127

Role of Nucleotide-binding Oligomerization Domain-containing (NOD) 2 in host defense during pneumococcal pneumonia

Tijmen J. Hommes1,2, Miriam H. van Lieshout1,2, Cornelis van ‘t Veer1,2, Sandrine Florquin3, Hester J. Bootsma4, Peter W. Hermans4, Alex F. de Vos1,2, Tom van der Poll1,2,5

7 Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands: 1Center for Experimental and Molecular Medicine, 2Center for Infection and Immunity, 3Department of Pathology, 5Division of Infectious Diseases. Radboud University Medical Center, Nijmegen, the Netherlands: 4Laboratory of Pediatric Infectious Diseases.

submitted Chapter 7

Abstract

Streptococcus (S.) pneumoniae is the most common causative pathogen in community- acquired pneumonia. Nucleotide-binding oligomerization domain-containing (NOD) 2 is a pattern recognition receptor located in the cytosol of myeloid cells that is able to detect peptidoglycan fragments of S. pneumoniae. We here aimed to investigate the role of NOD2 in the host response during pneumococcal pneumonia. Phagocytosis of S. pneumoniae was studied in NOD2 deficient (Nod2−/−) and wild-type (Wt) alveolar macrophages and neutrophils in vitro. In subsequent in vivo experiments Nod2−/− and Wt mice were inoculated with serotype 2 S. pneumoniae (D39), an isogenic capsule locus deletion mutant (D39∆cps) or serotype 3 S. pneumoniae (6303) via the airways, and bacterial growth and dissemination and the lung inflammatory response were evalu- ated. Nod2−/− alveolar macrophages and blood neutrophils displayed a reduced capacity to internalize pneumococci in vitro. During pneumonia caused by S. pneumoniae D39 Nod2−/− mice were indistinguishable from Wt mice with regard to bacterial loads in lungs and distant organs, lung pathology and neutrophil recruitment. While Nod2−/− and Wt mice also had similar bacterial loads after infection with the more virulent S. pneumoniae 6303 strain, Nod2−/− mice displayed a reduced bacterial clearance of the normally aviru- lent unencapsulated D39∆cps strain. These results suggest that NOD2 does not contribute to host defense during pneumococcal pneumonia and that the pneumococcal capsule impairs recognition of S. pneumoniae by NOD2.

130 Role of NOD2 in host defense during pneumococcal pneumonia

Introduction

Invading pathogens are sensed by a wide array of pattern recognition receptors (PRRs) that initiate the innate immune response and shape adaptive immunity1. Nucleotide- binding oligomerization domain (NOD)-like receptors (NLRs) are a family of intracellular PRRs that recognize specific microbial components2. NOD1 and NOD2 are prominent members of this family. Unlike NOD1, which is expressed by all cell types, NOD2 is mainly expressed by macrophages, monocytes and Paneth cells in the gut2. NOD2 recognizes bacterial molecules that are produced during the synthesis and/or degradation of pep- tidoglycan and is considered to act as a general sensor for most bacteria3. NOD2 signals through the adaptor protein receptor interacting protein (RIP)2, ultimately resulting in nuclear factor (NF)-κB translocation to the nucleus and the subsequent production and release of proinflammatory mediators, thereby triggering an immune response aimed at restricting bacterial growth. Streptococcus (S.) pneumoniae is the most common causative micro-organism in community-acquired pneumonia and an important cause of mortality world-wide4,5. The mortality rate associated with pneumococcal pneumonia ranges from 6 to >40%, largely depending on age and health care settings. Several studies have implicated NOD2 in the recognition of S. pneumoniae by innate immune cells6-8. NOD2 senses internalized pneumococci6 by a mechanism that depends on lysozyme-dependent digestion of S. pneumoniae and subsequent delivery of pneumococcal peptidoglycan fragments into the host cell cytosol mediated by pneumolysin, an important virulence factor of this pathogen8. During colonization of the upper airways, S. pneumoniae recognition by NOD2 induces the production of CC-chemokine ligand 2, leading to the recruitment of inflammatory macrophages necessary for bacterial clearance8. Thus far the role of NOD2 in lower respiratory tract infection by S. pneumoniae has not been studied. Here we set out to determine the contribution of NOD2 to the host 7 response during pneumococcal pneumonia by infecting NOD2 deficient (Nod2−/−) mice with a variety of wild-type (Wt) and genetically modified S. pneumoniae strains via the airways.

Materials and Methods

Ethics statement Experiments were carried out in accordance with the Dutch Experiment on Animals Act and approved by the Animal Care and Use Committee of the University of Amsterdam (Permit number DIX100121).

131 Chapter 7

Mice Nod2−/− C57BL/6 mice were purchased from Jackson Laboratories (Bar Harbor, ME). Age and sex matched Wt C57BL/6 mice were purchased from Charles River (Maastricht, the Netherlands) and maintained at the animal care facility of the Academic Medical Centre (University of Amsterdam), according to national guidelines with free access to food and water.

Phagocytosis Phagocytosis of S. pneumoniae by alveolar macrophages (obtained as described9) and blood neutrophils from Nod2−/− and Wt mice was done in essence as previously de- scribed10. Briefly, alveolar macrophages and whole blood were incubated with growth- inhibited, carboxyfluorescein succinimidyl ester (CFSE)- or fluorescein isothiocyanate (FITC)-labeled serotype 2 S. pneumoniae (D39) and internalization was measured by flow cytometry using FACS-Calibur (BD Biosciences, San Jose, CA).

Induction of pneumonia Pneumococcal pneumonia was induced by either serotype 2 S. pneumoniae (D39), a non-encapsulated mutant strain (isogenic capsule locus(cps) deletion mutant D39Δcps) of D3911,12 or a serotype 3 pneumococcal strain (ATCC 6303, American Type Culture Col- lection, Manassas, VA). All bacterial strains were grown for 3-6 hours to midlogarithmic phase at 37ºC in Todd-Hewitt broth (Difco, Detroit, MI), supplemented with yeast extract (0.5%). Bacteria were harvested by centrifugation at 4000 rpm, and washed twice in sterile isotonic saline. Next, mice (n = 8 per strain for each time point), were inoculated with 107 colony forming units (CFU) D39, 108 CFU D39Δcps or 5 × 104 CFU serotype 3 S. pneumoniae per mouse in a 50 µl saline solution and sacrificed 6, 24 or 48 hours there- after as described10,12. Collection and handling of samples were done as described10,12. In brief, blood was drawn into heparinized tubes and organs were removed aseptically and homogenised in 4 volumes of sterile isotonic saline using a tissue homogenizer (Biospec Products, Bartlesville, UK). To determine bacterial loads, ten-fold dilutions were plated on blood agar plates and incubated at 37°C for 16 hours.

Assays Lung homogenates were prepared for immune assays as described10. Tumor necrosis fac- tor (TNF)-α, interleukin (IL)-1-β, IL-6, Keratinocyte-derived chemokine (KC), macrophage inflammatory protein 2 (MIP-2) (all R&D systems, Minneapolis, MN) and myeloperoxidase (MPO; Hycult Biotechnology BV, Uden, the Netherlands) were measured using specific ELISAs according to manufacturers’ recommendations.

132 Role of NOD2 in host defense during pneumococcal pneumonia

Histology After embedding, lungs were stained with haematoxylin and eosin. To score lung inflam- mation and damage, the entire lung surface was analyzed with respect to the following parameters: bronchitis, edema, interstitial inflammation, intra-alveolar inflammation, pleuritis, endothelialitis and percentage of the lung surface demonstrating confluent inflammatory infiltrate. Each parameter was graded 0–4, with 0 being ‘absent’ and 4 being ‘severe’. The total pathology score was expressed as the sum of the score for all parameters. Granulocyte staining was done using FITC-labeled rat anti-mouse Ly-6G mAb (Pharmingen, San Diego, CA, USA) as described earlier10. Ly-6G expression in the lung tissue sections was quantified by digital image analysis13. In short, lung sections were scanned using the Olympus Slide system (Olympus, Tokyo, Japan) and TIF images, spanning the full tissue section were generated. In these images Ly-6G positivity and total surface area were measured using Image J (U.S. National Institutes of Health, Bethesda, MD, http://rsb.info.nih.gov/ij); the amount of Ly-6G positivity was expressed as a percentage of the total surface area.

Statistical analysis Data are expressed as box-and-whisker diagrams depicting the smallest observation, lower quartile, median, upper quartile and largest observation, or as medians with inter- quartile ranges as indicated. Comparisons between groups were conducted using the Mann-Whitney U. All analyses were done using GraphPad Prism version 5.01 (GraphPad Software, San Diego, CA). P-values less than 0.05 were considered statistically significant.

Results

NOD2 deficiency reduces the capacity of alveolar macrophages and neutrophils 7 to phagocytose S. pneumoniae in vitro Since NOD2 has been shown to promote phagocytosis of Staphylococcus aureus14, we first tested the capacity of Nod2−/− and Wt leukocytes to internalize S. pneumoniae (D39) in vitro. To this end we harvested alveolar macrophages and whole blood from naïve Nod2−/− and Wt mice and quantified fluorochrome labelled internalized S. pneumoniae by FACS. Interestingly, both Nod2−/− alveolar macrophages and blood neutrophils showed impaired internalization of S. pneumoniae in vitro (Figure 1 A and B; P < 0.05 and P < 0.001 respectively).

133 Chapter 7

A Alveolar macrophages B Neutrophils

400 Wt 50000 -/- x Nod2 x e e 40000 300 nd * nd i i s s i i 30000 s s o o t 200 t cy cy 20000 go go a 100 a *** h h 10000 P P

0 0 4°C 37°C 4°C 37°C Figure 1: NOD2 deficiency reduces the capacity of alveolar macrophages and neutrophils to internal- ize S. pneumoniae in vitro. Growth arrested, FITC labeled S. pneumoniae D39 were incubated with alveolar macrophages (A) or CFSE-labeled S. pneumoniae D39 with peripheral blood neutrophils (B) from wild-type (Wt) and Nod2−/− mice at 4°C (n = 3–4 per mouse strain) or 37°C (n = 6–8 per mouse strain) for 1 hour af- ter which phagocytosis was quantified. Data are expressed as box-and-whisker diagrams depicting the smallest observation, lower quartile, median, upper quartile and largest observation; *P < 0.05, ***P < 0.001 versus Wt cells.

NOD2 deficiency does not impact on bacterial growth or dissemination during pneumonia caused by serotype 2 S. pneumoniae (D39) To test whether the reduced phagocytic capacity of leukocytes in vitro results in im- paired host defense during pneumonia in vivo, we infected Nod2−/− and Wt mice with S. pneumoniae D39 via the airways and harvested blood and organs 6, 24 and 48 hours later to determine bacterial burdens. We observed no differences in bacterial loads in lungs, blood or spleens between Nod2−/− and Wt mice (Figure 2).

A Lung B Blood C Spleen 10 Wt 10 10 9 9 9 Nod2 -/-

l 8 l 8 l 8 m m m

/ 7 / 7 / 7

U 6 U 6 U 6 F F F

C 5 C 5 C 5 4 4 4 log log log log 3 3 3 10 10 10 2 2 2 1 1 1 0 0 0 6h 24h 48h 6h 24h 48h 6h 24h 48h Figure 2: NOD2 deficiency does not impact on bacterial growth or dissemination during pneumonia caused by serotype 2 S. pneumoniae (D39). Wild-type (Wt) and Nod2−/− mice were intranasally infected with 107 CFU of S. pneumoniae and sacrificed 6, 24 or 48 hours later. Bacterial loads were determined in lung homogenates (A), blood (B) and spleen (C). Data are expressed as box-and-whisker diagrams depicting the smallest observation, lower quartile, median, upper quartile and largest observation (8 mice per group at each time point). Differences between groups were not significant.

134 Role of NOD2 in host defense during pneumococcal pneumonia

NOD2 deficiency does not impact on inflammation during pneumonia caused by serotype 2 S. pneumoniae (D39) Considering that NOD2 has been found to contribute to S. pneumoniae induced in- flammatory responses in macrophages6, we studied the extent of lung inflammation in Nod2−/− and Wt mice during pneumonia. To this end we measured cytokines and chemokines in whole lung homogenates obtained from Nod2−/−and Wt mice at several time points after infection. We found elevated IL-6 levels in lung homogenates taken from Nod2−/− mice at 24 and 48 hours post-infection (Table 1; P < 0.05 and P < 0.01). Similarly at 24 and 48 hours post-infection Nod2−/− displayed increased levels of MIP-2 in their lungs compared to Wt mice (Table 1; P < 0.01 and P < 0.05). Lung pathology, semi- quantitatively scored by methods previously described10 (Figure 3A-C), and the extent of neutrophil recruitment into the lungs, measured by the number of Ly6G-positive cells in lung tissue (Figure 3D-F) and MPO concentrations in whole lung homogenates (Figure 3G), were not different betweenNod2 −/− and Wt mice. These data argue against a role for NOD2 in lung inflammation during pneumonia caused by S. pneumoniae D39.

Table 1: Cytokine and chemokine concentrations in lung homogenates of wild-type and Nod2−/− mice during pneumococcal pneumonia caused by serotype 2 S. pneumoniae (D39). Lung cytokines 6 hours 24 hours 48 hours Wt Nod2−/− Wt Nod2−/− Wt Nod2−/− TNF-α (pg/ml) 3394 (2597- 2531 (2035- 2282 (2163- 2239 (2036- 1144 (635- 1490 (891- 4733) 3817) 2419) 2453) 1892) 1865) IL-1β (pg/ml) 1913 (1865- 2201 (1857- 307 (215-399) 344 (220-405) 757 (709-768) 980 (740- 2407) 2602) 1129) IL-6 (pg/ml) 1874 (1392- 2256 (2064- 462 (441-509) 901 (705- 426 (400-498) 585 (500- 2266) 3038) 947)** 1114)* KC (ng/ml) 68.9 (53.2- 51.0 (47.9- 17.6 (13.8- 22.2 (16.0- 11.5 (9.6-23.5) 5.6 (5.1- 8.7) 92.1) 65.2) 21.6) 26.5) MIP-2 (ng/ml) 19.3 (16.1- 22.2 (19.0- 9.6 (7.6-10.9) 22.2 (20.7- 6.0 (5.2-7.1) 14.1 (8.1- 7 24.5) 25.7) 34.2)** 21.4)* Proinflammatory cytokine (TNF-α, IL-1β, IL-6) and chemokine (KC and MIP-2) levels in lung homogenates at 6, 24 and 48 hours after intranasal S. pneumoniae D39 infection in wild-type (Wt) and Nod2−/− mice. Data are medians (interquartile ranges); n = 8 mice per group per time point; * P < 0.05, ** P < 0.01.

135 Chapter 7

A B C

20 Wt Nod2 -/- 15 e r o

sc 10

A P 5

-/- Wt (24h) Nod2 (24h) 0 6h 24h 48h D E F

8

) % ( 6 area 4 lung lung + G G

6 2 - y

-/- L Wt (24h) Nod2 (24h) 0 6h 24h 48h G 100000

80000 l

m 60000 O ng/ O

P 40000 M 20000

0 6h 24h 48h Figure 3: NOD2 deficiency does not influence lung pathology and neutrophil recruitment during pneumonia caused by serotype 2 S. pneumoniae (D39). Wild-type (Wt) and Nod2−/− mice were intra- nasally infected with 107 CFU of S. pneumoniae D39 and sacrificed 6, 24 or 48 hours later. Representative hematoxylin and eosin (HE) stainings of lung tissue of Wt (A) and Nod2−/− mice (B) 24 hours after inoculation with S. pneumoniae D39 (original magnification ×200). Quantification of pulmonary Ly-6G positivity (F) and MPO levels in whole lung homogenates (G) 6, 24 or 48 hours after intranasal infection with S. pneumoniae D39 of wild-type (Wt) and Nod2−/− mice. Representative neutrophil stainings (brown) of Wt (D) and Nod2−/− mice (E) 24 hours after induction of pneumococcal pneumonia are shown (original magnification ×200). Data are expressed as box-and-whisker diagrams depicting the smallest observation, lower quartile, me- dian, upper quartile and largest observation (8 mice per group at each time point). Differences between groups were not significant.

NOD2 deficiency results in defective pulmonary clearance of non-encapsulated serotype 2 S. pneumoniae (D39Δcps) We have recently shown that the thick polysaccharide capsule impairs recognition of Toll-like receptor (TLR) ligands expressed by S. pneumoniae12. To test whether this also holds true for NOD2, we next infected Nod2−/−and Wt mice with the non-encapsulated mutant serotype 2 S. pneumonia D39Δcps. Interestingly, we observed increased bac- terial loads in lungs of Nod2−/− mice compared to Wt mice after 24 hours of infection (Figure 4; P < 0.05). No dissemination into blood or distant organs was found in either Wt or Nod2−/− mice. Cytokine and chemokine levels in whole lung homogenates were not different between Nod2−/− and Wt mice with the exception of IL-6 concentrations, which were higher in lungs of Nod2−/− mice 24 hours after infection (Table 2; P < 0.05).

136 Role of NOD2 in host defense during pneumococcal pneumonia Lung Lung 10 Wt 190 WNod2t -/- 9 l 8 Nod2 -/- m

l 8 / 7 U m / 67 F U C 6 F 5 C 45 log log 34 * log log 10 3 *

10 2 12 01 0 6h 24h Figure 4: NOD2 deficiency results in defective6h pulmonary clearance24h of non-encapsulated mutant S. pneumoniae D39Δcps. Wild-type (Wt) and Nod2−/− mice were intranasally infected with 108 CFU of S. pneu- moniae D39Δcps and sacrificed 6 or 24 hours later. Bacterial loads were determined in lung homogenates. Data are expressed as box-and-whisker diagrams depicting the smallest observation, lower quartile, me- dian, upper quartile and largest observation (8 mice per group at each time point); * P < 0.05.

Likewise, histopathology scores (Figure 5A-C), the number of Ly6 positive cells in lung tissue slides (Figure 5D-F) and whole lung MPO concentrations (Figure 5G) were similar in Nod2−/− and Wt mice 6 and 24 hours after inoculation with S. pneumoniae D39Δcps.

Table 2: Cytokine and chemokine concentrations in lung homogenates of wild-type (Wt) and Nod2−/− mice during pneumococcal pneumonia caused by an unencapsulated mutant strain serotype 2 S. pneumoniae D39Δcps. Lung cytokines 6 hours 24 hours Wt Nod2−/− Wt Nod2−/− TNF-α (pg/ml) 1720 (1430-2617) 993 (581-2855) 1009 (773-1676) 1493 (1052-1887) IL-1β (pg/ml) 461 (304-585) 383 (186-942) 1299 (888-1330) 1430 (1174-2021) IL-6 (pg/ml) 457 (441-518) 362 (348-519) 530 (501-588) 709 (644-835)* KC (ng/ml) 5.1 (4.3-6.8) 4.6 (3.6-6.9) 11.8 (10.1-14.0) 9.7 (8.9-13.1) MIP-2 (ng/ml) 8.7 (7.7-9.3) 7.4 (6.5-8.6) 15.4 (11.7-18.1) 18.1 (14.2-22.1) 7 Proinflammatory cytokine (TNF-α, IL-1β, IL-6) and chemokine (KC and MIP-2) levels in lung homogenates at 6 and 24 hours after intranasal S. pneumoniae D39Δcps infection in wild-type (Wt) and Nod2−/− mice. Data are medians (interquartile ranges); n = 8 mice per group per time point; * P < 0.05.

NOD2 does not contribute to the host response during pneumonia caused by serotype 3 S. pneumoniae In patients serotype 3 pneumococci are associated with severe disease15-18. Thus, to validate our findings of the limited role of NOD2 in host defense during pneumococ- cal pneumonia, we performed additional studies with the highly virulent serotype 3 S. pneumoniae 6303 strain. Similarly to experiments with the serotype 2 S. pneumoniae D39 strain, Nod2−/− and Wt mice had similar bacterial loads in lungs, blood and spleen at 6, 24 and 48 hours after infection (Figure 6A-C).

137 Chapter 7

A B C

15

e 10 r o sc

A

P 5

-/- Wt (24h) Nod2 (24h) 0 6h 24h D E F

4

) % ( 3 area 2 lung lung + G G

6 1 - y

-/- L Wt (24h) Nod2 (24h) 0 6h 24h G 60000 l

m 40000 O ng/ O P

M 20000

0 6h 24h Figure 5: NOD2 deficiency does not influence lung pathology and neutrophil recruitment during pneumonia caused by an unencapsulated mutant S. pneumoniae D39Δcps. Wild-type (Wt) and Nod2−/− mice were intranasally infected with 108 CFU of S. pneumoniae D39Δcps and sacrificed 6 or 24 hours later. Representative hematoxylin and eosin (HE) stainings of lung tissue of Wt (A) and Nod2−/− mice (B) 24 hours after inoculation with S. pneumoniae (original magnification ×200). Quantification of pulmonary Ly-6G positivity (F) and MPO levels in whole lung homogenates (G) 6 or 24 hours after intranasal infection with S. pneumoniae D39Δcps of wild-type (Wt) and Nod2−/− mice. Representative neutrophil stainings (brown) of Wt (D) and Nod2−/− mice (E) 24 hours after induction of pneumococcal pneumonia are shown (original magnification ×200). Data are expressed as box-and-whisker diagrams depicting the smallest observation, lower quartile, median, upper quartile and largest observation (8 mice per group at each time point). Dif- ferences between groups were not significant.

A Lung B Blood C Spleen 10 Wt 10 10 9 9 9 Nod2 -/-

l 8

l 8

l 8 m m m / / 7 / 7 7 U U U 6

6 F 6 F F C C C 5 5 5 4 4 4 log log log log log log 3 3 3 10 10 10 2 2 2 1 1 1 0 0 0 6h 24h 48h 6h 24h 48h 6h 24h 48h Figure 6: NOD2 deficiency does not impact on bacterial growth or dissemination during pneumonia caused by serotype 3 S. pneumoniae. Wild-type (Wt) and Nod2−/− mice were intranasally infected with 104 CFU of S. pneumoniae (6303) and sacrificed 6, 24 or 48 hours later. Bacterial loads were determined in lung homogenates (A), blood (B) and spleen (C). Data are expressed as box-and-whisker diagrams depicting the smallest observation, lower quartile, median, upper quartile and largest observation (8 mice per group at each time point). Differences between groups were not significant.

138 Role of NOD2 in host defense during pneumococcal pneumonia

Discussion

NOD2 is a prominent member of the NLR family able to recognize microbial derived ligands in the cytosol2. While previous studies found a role for NOD2 in host defense against Gram-negative pneumonia19,20, limited data exists on the involvement of NOD2 in host defense in Gram-positive pneumonia. In the present study we aimed to charac- terize the in vivo relevance of NOD2 in pneumococcal pneumonia. Since S. pneumoniae is sensed by NOD26, we hypothesized that NOD2 deficiency would result in impaired innate immunity during lower airway infection by this common respiratory pathogen. However, we here found no role for NOD2 in host defense during pneumonia caused by two S. pneumoniae strains (serotype 2 D39 and the more virulent serotype 3 6303), as reflected by similar bacterial loads at the primary site of infection and distant organs in Nod2−/− and Wt mice at multiple time points after infection. Interestingly, when we used a mutant strain of S. pneumoniae lacking the thick polysaccharide capsule, we observed a modestly impaired pneumococcal clearance locally in lungs of Nod2−/− mice. Together these data suggest that NOD2, while contributing to antibacterial defense against normally avirulent pneumococci, does not play a significant role in the innate immune response during pneumococcal pneumonia in vivo. To evaluate a functional role for NOD2 in pneumococcal pneumonia, we first assessed the capacity of neutrophils and alveolar macrophages, the main cell types involved in pulmonary clearance of S. pneumoniae4,21, to internalize this pathogen in vitro. In ac- cordance with an earlier report showing decreased uptake of S. aureus by bone mar- row derived neutrophils14, neutrophils and alveolar macrophages displayed impaired phagocytosis of S. pneumoniae. The mechanism by which NOD2 contributes to phago- cytosis of S. pneumoniae might involve induction of matrix metalloproteinase (MMP)-9, considering that the pneumococcus induces MMP9 in a NOD2 dependent manner7 and that MMP9 is important for phagocytosis of this bacterium by neutrophils22. A role for 7 NOD2 in phagocytosis is not uniform: NOD2 does not contribute to internalization of Mycobacterium (M.) bovis BCG or M. tuberculosis by alveolar macrophages23. Similarly, bone marrow derived macrophages deficient for RIP2 – the adapter molecule for NOD2 – showed no defect in internalization of Chlamydia pneumophila20. In a previous study Nod2−/− mice showed reduced lung inflammation and neutrophil recruitment during S. aureus pneumonia24. Since S. aureus is rapidly cleared from the lungs of normal immune competent mice, this model used very high infectious doses (109 CFU)24, lessening its clinical relevance. As a consequence, these earlier results are difficult to compare with the current investigation, which not only involved another pathogen, but also much lower infectious doses of more virulent bacterial strains. Of the two pneumococcal strains tested, S. pneumoniae 6303 clearly is the most virulent. Indeed, while S. pneumoniae D39 is slowly cleared from mouse lungs, low dose infection

139 Chapter 7 with S. pneumoniae 6303 results in a high mortality in immune competent mice caused by a gradual growth and subsequent dissemination of bacteria12,25,26. NOD2 did not con- tribute to either clearance of S. pneumoniae D39 or limiting the growth of S. pneumoniae 6303, strengthening the evidence that this cytosolic PRR is not involved in host defense during pneumococcal pneumonia. Our results are in line with a recent paper showing that NOD2 deficiency does not impact on the clearance ofS. pneumoniae from the upper airways in a model of nasopharyngeal colonization8. Lung inflammation was not altered inNod2 −/− mice during S. pneumoniae pneumonia, except for elevated levels of IL-6 and MIP-2 in whole lung homogenates. These results are in line with a previous report of elevated cytokine levels in lung homogenates of Nod2−/− mice during pneumonia caused by Legionella despite unaltered pulmonary bac- terial loads19. These data suggest NOD2 might reduce the proinflammatory response to S. pneumoniae. Since NOD2 is able to inhibit TLR2 mediated cytokine responses through NFκB27, and TLR2 is implicated in the early inflammatory response to S. pneumoniae26, one could speculate that the elevated IL-6 and MIP-2 levels in Nod2−/− mice are caused by absence of this inhibiting effect of NOD2 on TLR2 signalling. On the other hand, bone marrow derived macrophages lacking NOD2 produced less IL-6 in vitro compared to Wt cells when incubated with serotype 4 S. pneumoniae8. Considering the unaltered bacterial loads and lung pathology in Nod2−/− mice the modest effect of NOD2 on S. pneumoniae induced cytokine production is of little biological significance. Interestingly, when we used an unencapsulated pneumococcal strain we found Nod2−/− mice to have increased pulmonary bacterial loads 24 hours after infection. We12,13 and others28,29 have previously shown a role for the pneumococcal capsule in the capacity of S. pneumoniae to multiply in the lower airways and to induce severe pneu- monia. The polysaccharide capsule is a crucial virulence factor protecting S. pneumoniae from various harmful cellular processes including phagocytosis30,31. Since phagocytosis of S. pneumoniae is important for its degradation by lysozyme, releasing various ligands in the cytosol where it can be sensed by NOD28, we hypothesize that in the absence of the capsule S. pneumoniae is delivered more easily to the cytosol where it can be sensed by NOD2. Recently, our group demonstrated that part of the virulence of encapsulated pneumococci relies on the capacity of the capsule to impair recognition of TLR ligands expressed by S. pneumoniae12. The present data suggest that the same mechanism may be at play for recognition of the pneumococcus by NOD2. In conclusion, we here found no evidence for an important role for NOD2 in host defense during pneumonia caused by two different S. pneumoniae strains. Possibly, the innate immune response to S. pneumoniae is triggered by a simultaneous action of different PRRs. Indeed, during a pneumococcal colonization model, Nod2−/− mice were not hampered in clearance of S. pneumoniae from the upper airways; however, when mice lacking both NOD2 and TLR2 were used, impaired clearance of pneumococci was

140 Role of NOD2 in host defense during pneumococcal pneumonia observed8. These data, together with findings that the bacterial capsule can shield S. pneumoniae from recognition by certain PRRs, exemplify the complex nature of immune defense against this common and clinically relevant pathogen.

Acknowledgements

We thank Marieke ten Brink, Joost Daalhuisen and Regina de Beer for their expert techni- cal assistance.

7

141 Chapter 7

References

1. Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011;​34:​637‑50. 2. Caruso R, Warner N, Inohara N, Nunez G. NOD1 and NOD2: Signaling, Host Defense, and Inflam- matory Disease. Immunity 2014;​41:​898‑908. 3. Sorbara MT, Philpott DJ. Peptidoglycan: a critical activator of the mammalian immune system during infection and homeostasis. Immunol Rev 2011;​243:​40‑60. 4. van der Poll T, Opal SM. Pathogenesis, treatment, and prevention of pneumococcal pneumonia. Lancet 2009;​374:​1543‑56. 5. Welte T, Torres A, Nathwani D. Clinical and economic burden of community-acquired pneumonia among adults in Europe. Thorax 2012;​67:​71‑9. 6. Opitz B, Puschel A, Schmeck B, et al. Nucleotide-binding oligomerization domain proteins are innate immune receptors for internalized Streptococcus pneumoniae. J Biol Chem 2004;​279:​ 36426‑32. 7. Vissers M, Hartman Y, Groh L, de Jong DJ, de Jonge MI, Ferwerda G. Recognition of Streptococcus pneumoniae and muramyl dipeptide by NOD2 results in potent induction of MMP-9, which can be controlled by lipopolysaccharide stimulation. Infect Immun 2014;​82:​4952‑8. 8. Davis KM, Nakamura S, Weiser JN. Nod2 sensing of lysozyme-digested peptidoglycan promotes macrophage recruitment and clearance of S. pneumoniae colonization in mice. J Clin Invest 2011;​ 121:​3666‑76. 9. Knapp S, Leemans JC, Florquin S, et al. Alveolar Macrophages Have a Protective Antiinflammatory Role during Murine Pneumococcal Pneumonia. Am J Respir Crit Care Med 2003;​167:​171‑9. 10. Hommes TJ, Hoogendijk AJ, Dessing MC, et al. Triggering receptor expressed on myeloid cells-1 (TREM-1) improves host defence in pneumococcal pneumonia. J Pathol 2014;​233:​357‑67. 11. Bootsma HJ, Egmont-Petersen M, Hermans PW. Analysis of the in vitro transcriptional response of human pharyngeal epithelial cells to adherent Streptococcus pneumoniae: evidence for a distinct response to encapsulated strains. Infect Immun 2007;​75:​5489‑99. 12. de Vos AF, Dessing MC, Lammers AJ, et al. The Polysaccharide Capsule of Streptococcus pneumo- nia Partially Impedes MyD88-Mediated Immunity during Pneumonia in Mice. PLoS One 2015;​10:​ e0118181. 13. Lammers AJ, de Porto AP, Florquin S, et al. Enhanced vulnerability for Streptococcus pneumoniae sepsis during asplenia is determined by the bacterial capsule. Immunobiology 2011;​216:​863‑70. 14. Deshmukh HS, Hamburger JB, Ahn SH, McCafferty DG, Yang SR, Fowler VG, Jr. Critical role of NOD2 in regulating the immune response to Staphylococcus aureus. Infect Immun 2009;​77:​1376‑82. 15. Bender JM, Ampofo K, Byington CL, et al. Epidemiology of Streptococcus pneumoniae-induced hemolytic uremic syndrome in Utah children. Pediatr Infect Dis J 2010;​29:​712‑6. 16. Burgos J, Lujan M, Falco V, et al. The spectrum of pneumococcal empyema in adults in the early 21st century. Clin Infect Dis 2011;​53:​254‑61. 17. Weinberger DM, Harboe ZB, Sanders EA, et al. Association of serotype with risk of death due to pneumococcal pneumonia: a meta-analysis. Clin Infect Dis 2010;​51:​692‑9. 18. Yu J, Salamon D, Marcon M, Nahm MH. Pneumococcal serotypes causing pneumonia with pleural effusion in pediatric patients. J Clin Microbiol 2011;​49:​534‑8. 19. Berrington WR, Iyer R, Wells RD, Smith KD, Skerrett SJ, Hawn TR. NOD1 and NOD2 regulation of pulmonary innate immunity to Legionella pneumophila. Eur J Immunol 2010;​40:​3519‑27.

142 Role of NOD2 in host defense during pneumococcal pneumonia

20. Shimada K, Chen S, Dempsey PW, et al. The NOD/RIP2 pathway is essential for host defenses against Chlamydophila pneumoniae lung infection. PLoS Pathog 2009;​5:​e1000379. 21. Koppe U, Suttorp N, Opitz B. Recognition of Streptococcus pneumoniae by the innate immune system. Cell Microbiol 2012;​14:​460‑6. 22. Hong JS, Greenlee KJ, Pitchumani R, et al. Dual protective mechanisms of matrix metallopro- teinases 2 and 9 in immune defense against Streptococcus pneumoniae. J Immunol 2011;​186:​ 6427‑36. 23. Divangahi M, Mostowy S, Coulombe F, et al. NOD2-deficient mice have impaired resistance to Mycobacterium tuberculosis infection through defective innate and adaptive immunity. J Im- munol 2008;​181:​7157‑65. 24. Kapetanovic R, Jouvion G, Fitting C, et al. Contribution of NOD2 to lung inflammation during Staphylococcus aureus-induced pneumonia. Microbes Infect 2010;​12:​759‑67. 25. Dessing MC, Knapp S, Florquin S, de Vos AF, van der Poll T. CD14 facilitates invasive respiratory tract infection by Streptococcus pneumoniae. Am J Respir Crit Care Med 2007;​175:​604‑11. 26. Knapp S, Wieland CW, van ‘t Veer C, et al. 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 2004;​172:​3132‑8. 27. Watanabe T, Kitani A, Murray PJ, Strober W. NOD2 is a negative regulator of Toll-like receptor 2-mediated T helper type 1 responses. Nat Immunol 2004;​5:​800‑8. 28. Magee AD, Yother J. Requirement for capsule in colonization by Streptococcus pneumoniae. Infect Immun 2001;​69:​3755‑61. 29. Morona JK, Miller DC, Morona R, Paton JC. The effect that mutations in the conserved capsular polysaccharide biosynthesis genes cpsA, cpsB, and cpsD have on virulence of Streptococcus pneumoniae. J Infect Dis 2004;​189:​1905‑13. 30. Kadioglu A, Andrew PW. The innate immune response to pneumococcal lung infection: the untold story. Trends Immunol 2004;​25:​143‑9. 31. Hyams C, Camberlein E, Cohen JM, Bax K, Brown JS. The Streptococcus pneumoniae capsule in- hibits complement activity and neutrophil phagocytosis by multiple mechanisms. Infect Immun 2010;​78:​704‑15. 7

143

TREM-1/3 contributes to house dust mite induced allergic lung inflammation

Tijmen J. Hommes1,2, J. Daan de Boer1,2, Adam A. Anas1,2, Joris J. T. H. Roelofs3, Marco Colonna4, Cornelis van ’t Veer1,2, Alex F. de Vos1,2, Tom van der Poll1,2,5

8 Academic Medical Center, University of Amsterdam, Amsterdam, the Netherlands: 1Center for Experimental and Molecular Medicine; 2Center for Infection and Immunity; 3Department of Pathology; 5Division of Infectious Diseases. 4Washington University in St. Louis, St. Louis, Missouri, USA: Department of Pathology.

submitted Chapter 8

Abstract

Triggering Receptor Expressed on Myeloid cells (TREM)-1 and 2 are innate immune receptors able to fine-tune inflammatory responses initiated by pattern recognition receptors. Recent research has implicated Toll-like receptor (TLR)4 in the pathogenesis of house dust mite (HDM) induced experimental asthma. Since TREM-1 and TREM-2 respectively can enhance and inhibit TLR4 signaling, we here set out to investigate the role of TREM-1 and TREM-2 in HDM-induced allergic airway inflammation. Mice deficient for TREM-1/3 or TREM-2 and wild type mice were sensitized and chal- lenged with HDM extract via the airways, and cell influx, histopathology, mucus produc- tion and serum IgE levels were measured. TREM-1/3 deficiency was associated with attenuated eosinophil and neutrophil influx into bronchoalveolar lavage fluid, and reduced eosinophil accumulation in lung tissue; the extent of lung pathology and HDM-induced mucus production in the airways were not affected. TREM-1/3 deficiency also led to a diminished plasma IgE response. TREM-2 deficiency did not modify any response to HDM exposure. TREM-1/3 augments components of the allergic inflammatory airway response to HDM, suggesting this immune enhancing receptor may be a therapeutic target in al- lergic asthma.

146 TREM-1/3 contributes to house dust mite induced allergic lung inflammation

Introduction

“Triggering Receptor Expressed on Myeloid cells” (TREM) proteins are a family of cell surface receptors able to modulate cellular responses1. Best known family members are the innate cell surface receptors TREM-1 and TREM-2, which are both expressed by leukocytes and can augment or inhibit cellular responses respectively1. While most re- search has focused on their role in infectious diseases such as pneumonia and sepsis2-5, recent evidence has implicated TREM receptors in the pathogenesis of non-infectious conditions. For example deletion of TREM-1 reduced liver tumorigenesis in mice6 and blocking TREM-1 in a murine model of inflammatory bowel disease attenuated disease severity7. Likewise, TREM-2 may be involved in osteoclastogenesis8 and Alzheimer’s disease9. Recent studies have suggested that TREM-1 and -2 might contribute to the allergic lung inflammation accompanying asthma10-12. Asthma is a chronic disease of the airways characterized by bronchial hyper-reactivity, mucus overproduction and airway remodeling, affecting up to 300 million people world- wide13,14. Clinically, asthma is a disease characterized by episodes of reversible airway obstruction, dyspnea and wheezing. The allergic airway inflammation accompanying most forms of asthma are caused by a type 2 immune response driven by T helper (Th)2 lymphocytes, eosinophils and enhanced production of IgE15. Of note, however, it has become increasingly clear that the innate immune system plays a crucial role in the pathophysiology asthma16, wherein innate immune cells together with the respiratory epithelium help to shape a Th2 driven adaptive immune response15,17. Toll-like receptors (TLRs) comprise a family of pattern recognition receptors that are of pivotal importance for the initiation of innate immune responses18. Several lines of evidence have impli- cated TLRs in the pathogenesis of allergic asthma. Polymorphisms in genes encoding TLRs have been linked to the occurrence and manifestations of human asthma and allergies19. The main house dust mite (HDM) allergen, Dermatophagoides pteronyssinus (Der p) 2, has structural and functional homology with MD-2, the lipopolysaccharide (LPS)-binding component of the TLR4 signaling complex, and can facilitate LPS-driven TLR4 signaling in the absence of MD-220. In accordance, mice deficient for either TLR421-23 or the common TLR adaptor myeloid differentiation primary 88 (MyD88)24 showed a 8 markedly attenuated allergic lung inflammation upon exposure to HDM via the airways. TREM-1 and TREM-2 have opposite effects on TLR signaling: whereas TREM-1 enhances signaling upon ligation of several TLRs2,25, TREM-2 inhibits TLR-dependent cellular ef- fects26-28. Of most relevance for allergic HDM-driven asthma, TREM-1 and TREM-2 clearly influence LPS-induced activation of TLR425-28. Thus, considering the emerging role of TLR signaling in the pathogenesis of asthma, we here hypothesized that TREM-1 and TREM-2 might contribute to allergic airway inflammation induced by HDM. Specifically, we postulated that TREM-1 might enhance and TREM-2 might attenuate HDM-induced

147 Chapter 8 inflammation. We here tested this hypothesis by studying TREM-1/34 and TREM-228 deficient mice in a recently established mouse model of asthma using repeated HDM instillation via the airways29.

Materials and Methods

Animals TREM-1/3 (Trem-1/3−/−) and TREM-2 deficient (Trem-2−/−) mice were generated as previ- ously described and backcrossed at least six times to a C57BL/6 genetic background4,28. Age and sex matched wild type (Wt) C57BL/6 mice were purchased from Charles River (Maastricht, The Netherlands) and maintained at the animal care facility of the Academic Medical Centre (University of Amsterdam), according to national guidelines with free access to food and water. The Institutional Animal Care and Use Committee approved all animal experiments.

HDM-induced mouse asthma model HDM allergen whole body extract (Greer Laboratories, Lenoir, NC), derived from the common European HDM species Der p, was used to induce allergic lung inflammation as described29. Briefly, mice were inoculated intranasally on day 0, 1 and 2 with 25 μg HDM (sensitization phase) and on day 14, 15, 18 and 19 with 6.25 μg HDM (challenge phase). Controls received isotonic sterile saline intranasally on each occasion. Inoculum volume was 20 μl for every HDM and saline exposure and inoculation procedures were performed during isoflurane inhalation anesthesia. The experiment was ended at day 21 by euthanizing the mice and the subsequent collection and processing of samples: in one experiment BALF and blood were collected, in a separate experiment one lung was obtained for pathology using procedures as described in 29 and below.

Bronchoalveolar lavage and tissue handling Bronchoalveolar lavage fluid (BALF) was harvested after exposing the trachea through a midline incision and instilling and retrieving 1 mL of sterile saline 0.9% (in 500 μl aliquots). Cell counts were determined for each BALF sample in a hemocytometer (Beck- man Coulter, Fullerton, CA) and differential cell counts by cytospin preparations stained with Giemsa stain (Diff-Quick; Dade Behring AG, Düdingen, Switzerland). In independent experiments non-lavaged lungs were fixed in 10% formalin.

Pathology Lungs were embedded in paraffin after fixation in 10% formalin; 4 μm thick paraffin sections were stained with hematoxylin-eosin. Parameters of allergic lung inflammation

148 TREM-1/3 contributes to house dust mite induced allergic lung inflammation were scored in a blinded fashion by an experienced pathologist: interstitial inflammation, peribronchial inflammation, perivascular inflammation, edema and endothelialitis, each graded on a scale of 0 to 4 (0: absent, 1: mild, 2: moderate, 3: severe, 4: very severe). The total pathology score was expressed as the sum of the score for all parameters29. Periodic Acid Schiff (PAS)-D staining for carbohydrates in mucus was performed to quantify the amount of mucus. The amount of mucus per lung section was scored in a semiquantita- tive fashion on a scale from 0-8 (0-4 for presence of intrabronchial mucus plugs, 0-4 for the extent of goblet cells within the bronchial tree)29. Eosinophil staining was performed using a monoclonal antibody against major basic protein (MBP). Entire sections were digitized with a slide scanner using the 10× objective (Olympus dotSlide, Tokyo, Japan). Images were exported in the TIFF format for quantification. Influx of eosinophils was de- termined by measuring the MBP immunopositive area by digital image analysis (ImageJ 1.46, National Institute of Health, Bethesda, Maryland), and subsequently expressed as a percentage of the total lung area. The average of ten images was used for analysis of eosinophilic pulmonary influx29.

IgE assay Total IgE in plasma was measured using rat anti-mouse IgE as a capture antibody and biotin rat-anti-mouse IgE as a detection antibody; purified mouse IgE was used as stan- dard (all from BD Biosciences Pharmingen, Breda, the Netherlands).

Statistical analysis Values are expressed as mean ± SE. Differences between groups were tested by Mann- Whitney U test. GraphPad Prism version 5.0, GraphPad Software (San Diego, CA) was used for all analyses. Values of P < 0.05 were considered statistically significant.

Results

TREM-1/3 deficiency results in decreased cell influx in BALF after HDM exposure 8 To study the impact of TREM-1/3 deficiency on allergic airway inflammation we repeat- edly inoculated Trem-1/3−/− and Wt mice intranasally with HDM. As expected29,30, HDM induced recruitment of predominantly eosinophils and neutrophils into BALF of Wt mice. Trem-1/3−/− mice displayed a strongly reduced influx of leukocytes into BALF com- pared to Wt mice (Figure 1A, P < 0.001). This difference was mainly caused by a (mean) 65% reduction in eosinophil numbers (Figure 1B; P < 0.01). Neutrophil, lymphocyte and macrophage numbers (Figure 1C, D and Supplemental Figure 1, P < 0.05 or P < 0.01) were also decreased in BALF recovered from Trem-1/3−/− mice. To further analyse pulmonary

149 Chapter 8

A B C D

Total cells Eosinophils Neutrophils Lymphocytes 2.5 20 0.8 2.5 Wt Trem-1/3 -/- 6 5 5 2.0 5 2.0 0 0 0 0 15 0.6 1

x1 x x1 x1

s s 1.5 s 1.5 s ll ll ll ll 10 0.4 ce ce ce ce l l l l l l 1.0 l 1.0 a a a *** a t t t t ** To To To To 5 0.2 ** 0.5 * 0.5

0.0 0 0.0 0.0

Figure 1: TREM-1/3 deficiency is associated with reduced leukocyte recruitment upon HDM chal- lenge. Total cell counts (A), eosinophils (B), neutrophils (C) and lymphocytes (D) in BALF recovered from Trem-1/3−/− and wild type (Wt) mice after HDM exposure (dotted line indicates mean values in saline group not challenged with HDM). Data are means with SE of 8 mice per group; * P < 0.05, **P < 0.01 and ***P < 0.001.

A B C D 1.5 e ac

f 1.0 r u s * ung

l 0.5 %

-/- Wt saline Wt HDM Trem-1/3 HDM 0.0 Wt Trem-1/3 -/- Figure 2: TREM-1/3 deficiency is associated with reduced eosinophil recruitment upon HDM chal- lenge. Percentage of lung surface stained positive for eosinophils quantified by digitally imaging of MBP staining (see Methods). Representative MBP staining of lung tissue-slides of Wt mice exposed to saline (B), Wt mice exposed to HDM (C) and Trem-1/3−/− mice exposed to HDM (D). Scale bar indicates 200µm. Data are means with SE of 8 mice per group; * P < 0.05, **P < 0.01 and ***P < 0.001. eosinophil accumulation we stained lung tissue with an eosinophil specific anti-MBP antibody. Consistent with the profound influx of eosinophils observed in BALF, lung tissue showed increased MBP staining after HDM exposure (Figure 2 A-C). The percent- age of MBP stained eosinophils was lower in lungs of HDM-challenged Trem-1/3−/− mice compared to lungs of Wt mice (Figure 2A and 2D; P < 0.05). Together, these data show TREM-1/3 deficiency is associated with a reduced cell influx into BALF after HDM expo- sure.

TREM-1/3 deficiency does not impact on the severity of lung pathology during HDM induced airway inflammation To study the impact of TREM-1/3 deficiency on lung pathology during allergic airway inflammation we made use of a semi-quantitative scoring system described in the Ma- terials and Methods section. Intranasal HDM installation provoked marked lung inflam- mation in Trem-1/3−/− and Wt mice compared to saline controls. Surprisingly TREM1/3 deficiency did not impact on the severity of lung inflammation compared to Wt mice

150 TREM-1/3 contributes to house dust mite induced allergic lung inflammation

A B C D 5

e 4 r o sc 3 y og l 2 ho t a

P 1 -/- Wt saline Wt HDM Trem-1/3 HDM 0 Wt Trem-1/3 -/- Figure 3: TREM-1/3 deficiency does not impact lung pathology after HDM exposure. Pathology scores (A) as determined by the semi-quantitative score described in the Methods section (dotted line indicates mean score in saline group not challenged with HDM). Representative slides of lung HE staining of Wt mice exposed to saline (B), HDM (C) and Trem-1/3−/− mice exposed to HDM (D). Scale bar indicates 200µm. Data are means with SE of 8 mice per group.

(Figure 3A-D). Moreover, distinct histological features such as peribronchial inflamma- tion, endothelialitis, edema or pleuritis were not different between Trem-1/3−/− and Wt mice to (data not shown). Since human asthma is characterized by increased airway mucus production31, we next set out to investigate whether TREM-1/3 deficiency impacted on this feature by PAS-D staining of lung tissue. HDM exposure caused a clear increase in PAS-D staining compared to saline treated animals; however, no differences were found betweenTrem- 1/3−/− and Wt mice (Figure 4).

A B C D 3 e r

o 2 sc

s u c

u 1 M

-/- Wt saline Wt HDM Trem-1/3 HDM 0 Wt Trem-1/3 -/-

Figure 4: TREM-1/3 deficiency does not influence airway mucus production upon HDM exposure. (A) Mean (with SE) mucus production scores as determined by the semi-quantitative score described in the Methods section (dotted line indicates mean score in saline group not challenged with HDM). Representa- tive slides of lung PAS-D staining of Wt mice exposed to saline (B), HDM (C) Trem-1/3−/− mice exposed to HDM (D). Scale bar indicates 200µm. Data are means with SE of 8 mice per group. 8

TREM-1/3 deficiency attenuates IgE responses after HDM challenge. Since the presence of IgE in the circulation is the prototypic hallmark of Th2 driven immunity in allergic asthma15, we next measured IgE levels in plasma collected from Trem-1/3−/− and Wt mice after repeated intranasal instillation with HDM. Compared to saline controls, HDM exposed mice had elevated plasma levels of total IgE. Trem-1/3−/− mice had lower IgE plasma levels compared to Wt mice after HDM challenge (Figure 5; P < 0.01).

151 Chapter 8

IgE plasma

800 Wt Trem-1/3 -/- 600 l

m 400 ng/

200 **

0 saline HDM

Figure 5: TREM-1/3 deficiency results in a reduced systemic IgE response after HDM exposure. Mean (with SE) IgE levels. Data are means with SE of 8 mice per group; **P < 0.01.

TREM-2 deficiency does not impact HDM-induced airway inflammation To study the impact of TREM-2 deficiency on allergic airway inflammation, we repeated the experiments with Trem-2−/− mice. We found no differences in total cell influx or in influx of cell subsets into BALF (Figure 6A-D), and no difference in the numbers of eosin- ophils in lung tissue between Trem-2−/− and Wt mice (Figure 7A-D). Similarly, the extent of lung inflammation (Supplemental Figure 2) and mucus accumulation (Supplemental Figure 3) did not differ between Trem-2−/− and WT mice.

A B C D

Total cells Eosinophils Neutrophils Lymphocytes 2.5 25 0.8 1.5 Wt -/- Trem-2 5 6 5 2.0 5 20 0 0 0 0 0.6 1 1 1

x1 1.0 x x

x

1.5 s s s 15 s ll ll ll ll 0.4 ce ce ce ce l l l l l l 1.0 l 10 a a a a t t t

t 0.5 To To To 0.2 0.5 To 5

0.0 0 0.0 0.0

Figure 6: TREM-2 deficiency does not influence HDM-induced leukocyte influx into the lungs. Total cell counts (A), eosinophils (B), neutrophils (C) and lymphocytes (D) in BALF recovered from Trem-2−/− and wild type (Wt) mice after HDM exposure (dotted line indicates mean values in saline group not challenged with HDM). Data are means with SE of 8 mice per group.

152 TREM-1/3 contributes to house dust mite induced allergic lung inflammation

A B C D 1.5 e ac

f 1.0 r u s

ung

l 0.5 %

-/- Wt saline Wt HDM Trem-2 HDM 0.0 Wt Trem-2 -/-

Figure 7: TREM-2 deficiency does not impact lung pathology after HDM exposure. Percentage of lung surface stained positive for eosinophils quantified by digitally imaging of MBP staining (see Methods). Rep- resentative MBP staining of lung tissue-slides of Wt mice exposed to saline (B), Wt mice exposed to HDM (C) and Trem-2−/− mice exposed to HDM (D). Scale bar indicates 200µm. Data are means with SE of 8 mice per group.

Discussion

Asthma is a highly prevalent chronic disease of the airways associated with considerable morbidity and a tremendous burden on healthcare expenditure, stressing the need for a better understanding of underlying pathophysiological mechanisms and the develop- ment of new therapies13,32. Recent studies have implicated TLRs, and especially TLR4, in allergic asthma19-24. Since TLR signalling can be influenced by TREM-1 (enhancing TLR effects2,25) and TREM-2 (attenuating TLR effects26-28), we here tested the hypothesis that TREM-1 and TREM-2 might have opposite roles in allergic lung inflammation elicited by repeated exposure to HDM via the airways. We indeed show that TREM-1/3 deficiency is associated with attenuated leukocyte recruitment into the pulmonary compartment of mice inoculated with HDM, with strong reductions in eosinophil and neutrophil num- bers in BALF, and diminished eosinophil accumulation in lung tissue, accompanied by reduced systemic IgE levels. In contrast with our hypothesis, TREM-2 deficiency did not impact HDM-induced lung inflammation. Our model makes use of HDM as inflammation inducing stimulus29,30. HDM allergens are an important cause for allergic lung inflammation in asthma patients33. The allergic potential of HDM allergens partially is the result of the ability to activate the innate im- 8 mune system34. Der p 2, a major HDM allergen, can mimic the function of MD-2, the LPS binding component of the TLR4 receptor complex, thereby facilitating TLR4 signaling by LPS20. This effect of HDM on LPS-TLR4 signaling is relevant in the context of asthma, since exposure to LPS, as an environmental pollutant, is associated with asthma severity and the occurrence of asthma exacerbations35,36. Hence, the use of HDM to induce allergic inflammation in mice is not only clinically relevant, but also suitable to investigate the potential role of TREMs in asthma. Thus far, knowledge of the possible role of TREMs in allergy and/or asthma is highly limited. Asthma patients were reported to have elevated

153 Chapter 8 plasma levels of soluble TREM-111,12. In addition, mice with fungal asthma had increased lung soluble TREM-1 concentrations10, while TREM-1 expression was reduced on lung neutrophils of mice exposed to HDM, suggesting that TREM-1 might be shed during allergic airway inflammation37. In contrast, alveolar macrophages displayed transient increase in TREM-2 expression during HDM-induced pulmonary inflammation in mice37. Similar to previous studies3,4, we used a mouse deficient for both TREM-1 and TREM-3 to study the role of TREM-1 in HDM-induced responses. TREM-3 is a pseudogene in humans but an activating receptor in mice displaying high homology to TREM-138. For this reason a TREM-1 and 3 double knockout mouse was used in order to more accurately approach the human situation. Nonetheless, whereas in humans TREM-3 is not functional, we cannot exclude that in mice TREM-3 contributes to the phenotype of Trem-1/3−/− mice. Our data in Trem-1/3−/− mice are consistent with earlier investigations that implicated TLR signaling in HDM-induced allergic lung inflammation. Mice deficient in MyD88 were protected from the principal features of allergic asthma elicited by repeated HDM ad- ministration, including pulmonary accumulation of eosinophils and neutrophils24. TLR4 deficiency reproduced most of the phenotype associated with MyD88 deficiency after HDM exposure, including attenuated eosinophil recruitment into the lungs, but did not affect neutrophil influx; in contrast, TLR2 deficient mice were indistinguishable from Wt mice24. The importance of TLR4 in HDM-induced inflammation in the lower airways was confirmed in another investigation23. In accordance with the reduced IgE response in Trem-1/3−/− mice, Myd88−/− mice also showed a strongly reduced Th2 associated im- munoglobulin response24. Likewise, our current finding that TREM-1/3 deficiency does not impact on mucus production in HDM exposed airways is corroborated by similar results in Myd88−/− and Tlr4−/− mice24. Together these data suggest distinct partially TLR independent pathways in the induction of different components of the HDM response in the lungs. Our study does not reveal which TREM-1/3 expressing cell type drives the phenotype of Trem-1/3−/− mice in this model. TREM-1 is especially expressed by monocytes/mac- rophages and neutrophils1, whereas expression of TREM-3 seems to be limited to mac- rophages38. Notably, however, TREM-1 expression could be induced by active vitamin D in human airway epithelial cells39, and gastric epithelium expresses TREM-1 during infection with Helicobacter pylori 40. Experiments using TLR4 bone marrow chimeras have shown that absent TLR4 expression on radioresistant lung structural cells but not on hematopoietic cells abolished HDM-driven allergic airway inflammation, suggesting an essential role for airway epithelial TLR4 herein21. A more recent study making use of the Cre-lox system to generate airway epithelial cell and myeloid TLR4 deficient mice indicated that TLR4 expression by hematopoietic cells is critical for neutrophilic airway inflammation following HDM exposure, whereas TLR4 expression by airway epithelial cells drives eosinophilic airway inflammation22. Together these data suggest that TLR4

154 TREM-1/3 contributes to house dust mite induced allergic lung inflammation expression by hematopoietic and airway epithelial cells may control distinct arms of the immune response to inhaled HDM. Although we here show that TREM-1/3 deficiency affects both neutrophil and eosinophil influx, future investigations are warranted to es- tablish whether TREM-1/3 modifies HDM-TLR4 effects in hematopoietic and/or epithelial cells. Like TREM-1, TREM-2 is able to modulate TLR signaling26-28. TREM-2 deficient macro- phages produce increased levels of proinflammatory cytokines in response to various TLR ligands27. In contrast to the clear role of TREM-1/3, we found no role for TREM-2 in HDM-induced allergic airway inflammation, as reflected by unaltered cell influx, lung inflammation and mucus formation in Trem-2−/− mice when compared with Wt mice. Possible explanations include differential cellular expression of TREM-1/3 and TREM-2 and/or differential impact on HDM-TLR signaling by these receptors. In conclusion, we here for the first time show that TREM-1/3 contributes to airway inflammation induced by a clinically relevant human allergen. TREM-1 has been pro- posed as therapeutic target for sepsis1,2. The current data suggest that TREM-1 inhibitory strategies could be a potentially valuable adjunctive therapy for allergic asthma.

Acknowledgements

The authors thank Marieke ten Brink and Joost Daalhuizen (Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands) for their expert technical assistance during the animal experiments, Regina de Beer (Center for Experimental and Molecular Medicine, Academic Medical Center, Amsterdam, the Netherlands) for performing histopathological stainings, Onno de Boer (Department op Pathology, Academic Medical Center, Amsterdam, the Netherlands) for assistance with digital analysis of MBP immunostaining, and Dr. Nancy Lee and Prof. James Lee (Mayo Clinic Arizona, Scottsdale, USA) for providing the anti-MBP antibody. 8

155 Chapter 8

References

1. Sharif O, Knapp S. From expression to signaling: roles of TREM-1 and TREM-2 in innate immunity and bacterial infection. Immunobiology 2008;​213:​701‑13. 2. Bouchon A, Facchetti F, Weigand MA, Colonna M. TREM-1 amplifies inflammation and is a crucial mediator of septic shock. Nature 2001;​410:​1103‑7. 3. Hommes TJ, Hoogendijk AJ, Dessing MC, et al. Triggering receptor expressed on myeloid cells-1 (TREM-1) improves host defence in pneumococcal pneumonia. J Pathol 2014;​233:​357‑67. 4. Klesney-Tait J, Keck K, Li X, et al. Transepithelial migration of neutrophils into the lung requires TREM-1. J Clin Invest 2013;​123:​138‑49. 5. Sharif O, Gawish R, Warszawska JM, et al. The triggering receptor expressed on myeloid cells 2 inhibits complement component 1q effector mechanisms and exerts detrimental effects during pneumococcal pneumonia. PLoS Pathog 2014;​10:​e1004167. 6. Wu J, Li J, Salcedo R, Mivechi NF, Trinchieri G, Horuzsko A. The proinflammatory myeloid cell receptor TREM-1 controls Kupffer cell activation and development of hepatocellular carcinoma. Cancer Res 2012;​72:​3977‑86. 7. Schenk M, Bouchon A, Seibold F, Mueller C. TREM-1--expressing intestinal macrophages crucially amplify chronic inflammation in experimental colitis and inflammatory bowel diseases. J Clin Invest 2007;​117:​3097‑106. 8. Paloneva J, Manninen T, Christman G, et al. Mutations in two genes encoding different subunits of a receptor signaling complex result in an identical disease phenotype. Am J Hum Genet 2002;​ 71:​656‑62. 9. Jonsson T, Stefansson H, Steinberg S, et al. Variant of TREM2 associated with the risk of Alzheim- er’s disease. N Engl J Med 2013;​368:​107‑16. 10. Buckland KF, Ramaprakash H, Murray LA, et al. Triggering receptor expressed on myeloid cells-1 (TREM-1) modulates immune responses to Aspergillus fumigatus during fungal asthma in mice. Immunol Invest 2011;​40:​692‑722. 11. Bucova M, Suchankova M, Dzurilla M, et al. Inflammatory marker sTREM-1 reflects the clinical stage and respiratory tract obstruction in allergic asthma bronchiale patients and correlates with number of neutrophils. Mediators Inflamm 2012;​2012:​628754. 12. Phua J, Koay ES, Zhang D, et al. Soluble triggering receptor expressed on myeloid cells-1 in acute respiratory infections. Eur Respir J 2006;​28:​695‑702. 13. Braman SS. The global burden of asthma. Chest 2006;​130:​4S-12S. 14. Martinez FD, Vercelli D. Asthma. Lancet 2013;​382:​1360‑72. 15. Lambrecht BN, Hammad H. The immunology of asthma. Nat Immunol 2014;​16:​45‑56. 16. Holgate ST. Innate and adaptive immune responses in asthma. Nat Med 2012;​18:​673‑83. 17. Lambrecht BN, Hammad H. The airway epithelium in asthma. Nat Med 2012;​18:​684‑92. 18. Kawai T, Akira S. Toll-like receptors and their crosstalk with other innate receptors in infection and immunity. Immunity 2011;​34:​637‑50. 19. Tesse R, Pandey RC, Kabesch M. Genetic variations in toll-like receptor pathway genes influence asthma and atopy. Allergy 2011;​66:​307‑16. 20. Trompette A, Divanovic S, Visintin A, et al. Allergenicity resulting from functional mimicry of a Toll-like receptor complex protein. Nature 2009;​457:​585‑8. 21. Hammad H, Chieppa M, Perros F, Willart MA, Germain RN, Lambrecht BN. House dust mite al- lergen induces asthma via Toll-like receptor 4 triggering of airway structural cells. Nat Med 2009;​ 15:​410‑6.

156 TREM-1/3 contributes to house dust mite induced allergic lung inflammation

22. McAlees JW, Whitehead GS, Harley IT, et al. Distinct Tlr4-expressing cell compartments control neutrophilic and eosinophilic airway inflammation. Mucosal Immunol 2014. 23. Ryu JH, Yoo JY, Kim MJ, et al. Distinct TLR-mediated pathways regulate house dust mite-induced allergic disease in the upper and lower airways. J Allergy Clin Immunol 2013;​131:​549‑61. 24. Phipps S, Lam CE, Kaiko GE, et al. Toll/IL-1 signaling is critical for house dust mite-specific helper T cell type 2 and type 17 [corrected] responses. Am J Respir Crit Care Med 2009;​179:​883‑93. 25. Bouchon A, Dietrich J, Colonna M. Cutting edge: inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J Immunol 2000;​164:​4991‑5. 26. Gawish R, Martins R, Bohm B, et al. Triggering receptor expressed on myeloid cells-2 fine-tunes inflammatory responses in murine Gram-negative sepsis. FASEB J 2014. 27. Hamerman JA, Jarjoura JR, Humphrey MB, Nakamura MC, Seaman WE, Lanier LL. Cutting edge: inhibition of TLR and FcR responses in macrophages by triggering receptor expressed on myeloid cells (TREM)-2 and DAP12. J Immunol 2006;​177:​2051‑5. 28. Turnbull IR, Gilfillan S, Cella M, et al. Cutting edge: TREM-2 attenuates macrophage activation. J Immunol 2006;​177:​3520‑4. 29. Daan de Boer J, Roelofs JJ, de Vos AF, et al. Lipopolysaccharide inhibits Th2 lung inflammation induced by house dust mite allergens in mice. Am J Respir Cell Mol Biol 2013;​48:​382‑9. 30. de Boer JD, Yang J, van den Boogaard FE, et al. Mast cell-deficient kit mice develop house dust mite-induced lung inflammation despite impaired eosinophil recruitment. J Innate Immun 2014;​ 6:​219‑26. 31. Thomson NC, Chaudhuri R, Messow CM, et al. Chronic cough and sputum production are associ- ated with worse clinical outcomes in stable asthma. Respir Med 2013;​107:​1501‑8. 32. Barnett SB, Nurmagambetov TA. Costs of asthma in the United States: 2002-2007. J Allergy Clin Immunol 2011;​127:​145‑52. 33. Platts-Mills TA, Erwin EA, Heymann PW, Woodfolk JA. Pro: The evidence for a causal role of dust mites in asthma. Am J Respir Crit Care Med 2009;​180:​109-13; discussion 20‑1. 34. Gregory LG, Lloyd CM. Orchestrating house dust mite-associated allergy in the lung. Trends Im- munol 2011;​32:​402‑11. 35. Celedon JC, Milton DK, Ramsey CD, et al. Exposure to dust mite allergen and endotoxin in early life and asthma and atopy in childhood. J Allergy Clin Immunol 2007;​120:​144‑9. 36. Michel O, Kips J, Duchateau J, et al. Severity of asthma is related to endotoxin in house dust. Am J Respir Crit Care Med 1996;​154:​1641‑6. 37. Habibzay M, Saldana JI, Goulding J, Lloyd CM, Hussell T. Altered regulation of Toll-like receptor responses impairs antibacterial immunity in the allergic lung. Mucosal Immunol 2012;​5:​524‑34. 38. Chung DH, Seaman WE, Daws MR. Characterization of TREM-3, an activating receptor on mouse macrophages: definition of a family of single Ig domain receptors on mouse chromosome 17. Eur 8 J Immunol 2002;​32:​59‑66. 39. Rigo I, McMahon L, Dhawan P, et al. Induction of triggering receptor expressed on myeloid cells (TREM-1) in airway epithelial cells by 1,25(OH)(2) vitamin D(3). Innate Immun 2012;​18:​250‑7. 40. Schmausser B, Endrich S, Beier D, et al. Triggering receptor expressed on myeloid cells-1 (TREM-1) expression on gastric epithelium: implication for a role of TREM-1 in Helicobacter pylori infection. Clin Exp Immunol 2008;​152:​88‑94.

157 Chapter 8

Supplemental

Macrophages

6 Wt Trem-1/3 -/- 5 0

x1 4

s ll ce

l l ** a

t 2 To

0

Supplemental Figure 1: TREM-1/3 deficiency is associated with reduced macrophage recruitment upon HDM challenge. Macrophage counts in BALF recovered from Trem-1/3−/− and wild type (Wt) mice after HDM exposure (dotted line indicates mean values in saline group not challenged with HDM). Data are means with SE of 8 mice per group; **P < 0.01.

A B C D 8 e r 6 o sc

y 4 og l ho t

a 2 P Wt saline Wt HDM Trem-2 - /- HDM 0 Wt Trem-2 -/- Supplemental Figure 2: TREM-2 deficiency does not impact lung pathology after HDM exposure. Pa- thology scores (A) as determined by the semi-quantitative score described in the Methods section (dotted line indicates mean score in saline group not challenged with HDM). Representative slides of lung HE stain- ing of Wt mice exposed to saline (B), HDM (C) and Trem-2−/− mice exposed to HDM (D). Scale bar indicates 200µm. Data are means with SE of 8 mice per group. a) b) c) d) 3 e r

o 2 sc

s u c

u 1 M

Wt saline Wt HDM Trem-2 - /- HDM 0 Wt Trem-2 -/- Supplemental Figure 3: TREM-2 deficiency does not influence airway mucus production upon HDM exposure. (A) Mean (with SE) mucus production scores as determined by the semi-quantitative score de- scribed in the Methods section (dotted line indicates mean score in saline group not challenged with HDM). Representative slides of lung PAS-D staining of Wt mice exposed to saline (B), HDM (C) Trem-2−/− mice ex- posed to HDM (D). Scale bar indicates 200µm. Data are means with SE of 8 mice per group.

158 9 Summary and general discussion

Summary and general discussion

Summary

Lung diseases like pneumonia and asthma are major causes of morbidity and mortal- ity worldwide. The innate immune system is the first line of defense against invading pathogens. It acts rapidly upon recognition of microbial motifs by pattern recognition receptors (PRRs), which initiates an inflammatory response aimed at pathogenic clear- ance. The Triggering Receptor Expressed on Myeloid cells (TREM) protein family consists of innate immune receptors that are able to amplify or dampen signaling through PRRs. This thesis focuses on the role of TREM receptors in infectious diseases and allergic airway inflammation.

Chapter 1 provides a short overview of the different disease models used in this re- search and briefly discusses pulmonary innate immunity. We particularly discuss the role of Toll-like and NOD like receptors and introduce TREM receptors. Especially the role and function of TREM-1 and TREM-2 are discussed on more detail. In Chapter 2 we investigate the role of TREM-1 in a murine model of pneumococcal pneumonia by instill- ing live S. pneumoniae intranasally in both wild-type and TREM-1 deficient animals. We found that TREM-1 is crucially involved in early host defense against S. pneumoniae by potentiating local cytokine and chemokine production and neutrophil recruitment to the site of infection. TREM-1 deficient mice displayed enhanced lethality associated with increased bacterial growth and dissemination. We found that alveolar macrophages from TREM-1 deficient mice are hampered in their ability to produce cytokines and chemokines in response to pneumococci. Additionally, we found that these cells were less capable of internalizing S. pneumoniae in vitro. TREM-1 deficient neutrophils on the other hand, did not show defective phagocytosis or activation upon incubation with the same pathogen. Interestingly, TREM-2 was shown to ameliorate host defense during the same disease model1. Since both TREM-1 and TREM-2 signal through adaptor mol- ecule DNAX-activation protein of 12kDa (DAP12) in Chapter 3 we investigate the role of DAP12 during pneumococcal pneumonia. We show that DAP12 deficiency improved host defense during pulmonary infection S. pneumoniae. DAP12 deficient mice displayed reduced lethality associated with decreased bacterial growth and dissemination, and attenuated lung inflammation. We foundin vitro that alveolar macrophages from DAP12 deficient mice produced more TNF-α. Additionally, we found that these cells were more capable of internalizing S. pneumoniae in vitro. To obtain more insight into the role of TREM-1 during Gram-positive pneumonia we evaluated host responses of TREM-1 9 deficient mice during pneumonia caused by another clinically relevant pathogen: in Chapter 4 we studied the role of TREM-1 in pneumonia caused by methicillin resistant Staphylococcus aureus (MRSA). Our main finding is that also in this model TREM-1 defi- ciency impaired host defense, as reflected by increased bacterial loads in the pulmonary

161 Chapter 9 department during staphylococcal pneumonia. This was accompanied by reduced lung inflammation, as reflected by histopathology and cytokine levels, while neutrophil recruitment was not affected. Macrophages deficient for TREM-1 were less responsive to S. aureus in vitro. After exploring the role of TREM-1 in Gram-positive pneumonia, in Chapter 5 and Chapter 6 we further dissect the role of TREM-1 in pneumonia by using two Gram-negative pathogens. Since no data of TREM-2 in Gram-negative pneumonia exist, we also investigated the role of this receptor in these models. In Chapter 5 we describe a crucial role for TREM-1 in host defense against Klebsiella (K.) pneumoniae. In contrast, we did not find a role for TREM-2 in this model. TREM-1 deficient mice displayed enhanced lethality associated with increased bacterial growth and dissemination. We found that macrophages from TREM-1 deficient mice were hampered in their ability to produce cytokines and chemokines in response to Klebsiella but showed no defect in the phagocytosis of this pathogen. In contrast, we found that TREM-2 deficient cells showed increased cytokine responses and were less capable of internalizing K. pneumoniae in vitro. Chapter 6 describes the role of both TREM-1 and TREM-2 in melioidosis (Gram- negative pneumosepsis caused by Burkholderia (B.) pseudomallei). Surprisingly we only found a modest role for TREM-1 during B. pseudomallei infection reflected by unaltered survival of TREM-1 deficient mice. Inflammation and bacterial loads were not affected with the exception of mildly reduced systemic outgrowth in TREM-1 deficient animals. Interestingly, we found TREM-2 to impair host defense against B. pseudomallei-induced sepsis, reflected by improved survival, diminished bacterial dissemination, decreased inflammation and less organ damage of TREM-2 deficient mice. Our ex vivo studies sug- gest that the protective effect of TREM-2 deficiency in part results from the diminished capacity of TREM-2-deficient macrophages to elicit a proinflammatory response, which is an important contributor to organ injury in the event of sepsis. In Chapter 7 we ex- plore the role of NOD2, a cytosolic PRR which recognizes specific pneumococcal derived motifs. We here found no role for NOD2 in host defense during pneumonia caused by two S. pneumoniae strains (serotype 2 D39 and the more virulent serotype 3 6303), as reflected by similar bacterial loads at the primary site of infection and distant organs in NOD2 deficient and wild-type mice at multiple time points after infection. Interest- ingly, when we used a mutant strain of S. pneumoniae lacking the thick polysaccharide capsule, we observed a modestly impaired pneumococcal clearance locally in lungs of NOD2 deficient mice. Having established an important role for TREM-1 in Gram-positive pneumonia caused by S. pneumoniae and S. aureus and a detrimental role for TREM-2 in Gram-negative pneumonia caused by B. pseudomallei, in Chapter 8 we set out to evalu- ate the role of both receptors in sterile allergic airway inflammation caused by repeated house dust mite (HDM) instillation. Interestingly, we found that TREM-1 deficient mice are protected from allergic airway inflammation: TREM-1 deficiency resulted in reduced influx of total inflammatory cells into bronchoalveolar lavage fluid, marked reductions

162 Summary and general discussion in eosinophil and neutrophil numbers and diminished eosinophil accumulation in lung tissue, accompanied by reduced systemic IgE levels. Unexpectedly, TREM-2 deficiency did not impact HDM-induced lung inflammation.

Discussion

With the studies presented in this thesis we aimed to obtain further insight into the role of TREM-receptors during pneumonia, sepsis and allergic lung inflammation. TREM- receptors are a novel family of proteins implicated in innate immune responses and are considered potential targets for the treatment of infectious diseases. TREM-1 is an activating receptor on macrophages and neutrophils that can potentiate signaling by PRRs. Previous work that studied the contribution of TREM-1 to sepsis pathogenesis has indicated that TREM-1 adversely affects sepsis outcome by causing excessive inflamma- tion2. These findings have raised interest in developing TREM-1 inhibiting strategies for the treatment of sepsis. Indeed interfering with TREM-1 signaling improves outcome in different sepsis models2-4. However in the work presented in this thesis we find TREM-1 to actually improve host defense during different models of pneumonia/pneumosepsis. During Gram-positive pneumonia caused by S. pneumoniae and S. aureus, TREM-1 de- ficiency leads to impaired host defense. These results are in line with an earlier report showing that TREM-1 is important for protective immunity during pneumonia caused by Pseudomonas aeruginosa5. It is known that an inflammatory response is necessary to overcome invading pathogens. Indeed in these two pneumonia models we found TREM-1 deficiency to result in reduced levels of pulmonary proinflammatory cytokines. In the case of pneumococcal pneumonia this resulted in a reduced influx of neutro- phils, while this was not the case in staphylococcal pneumonia, indicating different underlying mechanisms. These results add to the notion that host defense mechanisms differ between pathogens. Similar in both these studies however, are the in vitro results obtained from macrophage stimulation experiments. TREM-1 deficient cells displayed impaired cytokine production when incubated with S. pneumoniae and S. aureus. This may be explained by the amplificatory role of TREM-1 on different TLR and NLR signaling cascades6,7. How this is regulated at the molecular level, however, is not known. Liga- tion of TREM-1 results in mitogen-actived protein (MAP) kinases phosphorylation and nuclear factor kappa B (NFkB) translocation to the nucleus resulting in proinflammaroty cytokine release, partially explaining our results. Comparable results were obtained 9 when using K. pneumoniae although in vivo, during pneumosepsis caused by this patho- gen, we did not find differences in cytokine levels in lungs of TREM-1 deficient mice. Nonetheless, these animals displayed worsened survival compared to their wild-type counterparts also indicating a protective effect of TREM-1. How exactly this is explained

163 Chapter 9 merits further research. One might speculate that perhaps killing of Klebsiella by TREM-1 deficient cells is impaired although no data exists on a potential role of TREM-1 in bacte- rial killing. In contrast with earlier work demonstrating a beneficial effect of blocking TREM-1 during Gram-negative pneumonia caused by B. pseudomallei (melioidosis)8 we only found a very modest phenotype of mice lacking TREM-1 during this disease. Obvi- ously comparing both studies is difficult since the first study used a TREM-1 mimicking peptide to inhibit TREM-1 signaling in wild-type mice while the latter investigation used a genetically modified mouse strain totally devoid of TREM-1.

Unlike TREM-1, which is an activating receptor, TREM-2 is an inhibiting receptor on leukocytes suggesting opposite roles of both receptors. Since sepsis is characterized by overwhelming inflammation, the inhibiting effect of TREM-2 is thought to be beneficial in this syndrome. Indeed blocking TREM-2 after cecal ligation and puncture, a severe ab- dominal sepsis model, survival of treated animals is worsened9. In correspondence with an earlier report showing a deleterious role for TREM-2 during pneumonia caused by S. pneumoniae1, we also found a clear harmful role for TREM-2 during melioidosis. However, in contrast to our hypothesis, we did not find a role for TREM-1 during K. pneumoniae pneumonia derived sepsis. Of note, in vitro we did find increased cytokine production and impaired phagocytosis of this bacterium by TREM-2 deficient macrophages which is in line with the literature suggesting TREM-2 can inhibit TLR signaling and is a phago- cytic receptor for bacteria10-12. On the other hand, internalization of B. pseudomallei was not affected by TREM-2 deficiency. Several characteristics of this facultative intracellular bacterium might in part explain these discrepancies; B. pseudomallei is capable of invad- ing both phagocytic and non-phagocytic cells and circumvents intracellular defense mechanisms efficiently in order to replicate and spread to adjacent cells13.

Since TREM-1 and TREM-2 both signal through the adaptor molecule DAP12, we inves- tigated the role of this protein during pneumococcal pneumonia caused by serotype 3 pneumococci. Interestingly we found DAP12 deficient mice to be protected. Because DAP12 is the adaptor molecule for many other receptors than TREM proteins, it is not possible to pinpoint its effect to a single receptor. We found an opposite phenotype for TREM-1 deficient mice during pneumococcal pneumonia although we used a different strain of S. pneumoniae in these experiments (serotype 2). Interestingly, the protected phenotype of mice lacking DAP12 is similar to that of TREM-2 deficient mice in the same infectious model1. The net effect of DAP12 activation likely depends on the pathogen and the severity and primary source of the infection, with differential engagement of DAP12-coupled receptors and differential roles of distinct DAP12 expressing cell types in host defense against the causative organism of the infection.

164 Summary and general discussion

A previous report has shown TREM-1 is able to amplify NOD2 signaling7. Since NOD2 recognizes S. pneumoniae in vitro14 and since TREM-1 has a role in the host defense against this bacterium in vitro and in vivo, we set out to assess the role of NOD2 in a model of pneumonia caused by this pathogen. We only found a modest role for this NLR. Although NOD2 deficient neutrophils and macrophages displayed a reduced capacity to phagocytose S. pneumoniae, NOD2 deficient mice had an unremarkable host defense upon infection with either a serotype 2 or serotype 3 pneumococcal strain via the air- ways in vivo. Notably, NOD2 deficient mice did show increased pneumococcal burdens in their lungs after infection with an unencapsulated pneumococcal strain. The polysac- charide capsule is a key virulence factor expressed by S. pneumoniae15, which amongst other protects the pneumococcus of being phagocytosed by immune cells. Possibly, in the absence of the capsule components of S. pneumoniae, including the NOD2 li- gands peptidoglycan fragments, are delivered more easily to the cytosol where they can be sensed by NOD2. Recently, part of the virulence of encapsulated pneumococci was shown to rely on the capacity of the capsule to impair recognition of TLR ligands expressed by S. pneumoniae16. The present data indicate that the same mechanism may apply for recognition of the pneumococcus by NOD2.

While absence of TREM-1 signaling was harmful in pneumonia models leading to worsened disease outcome, we found TREM-1 deficiency to be beneficial during allergic inflammation of the airways. This makes TREM-1 a potential therapeutic target for the treatment of this disease. Indeed the innate immune system and TLR signaling in par- ticular, is a new promising field of research in asthma17. Future research should point out whether interfering with TREM-1 signaling - perhaps by using small molecule inhibitors/ peptides - is beneficial in experimental asthma.

In many chapters of this thesis we used mice deficient for both TREM-1 and TREM-3 as a model for TREM-1 deficiency. In mice, TREM-3 is highly homologous to TREM-1 and both receptors have a similar cellular distribution18. While in humans Trem3 is a pseudogene, in mice TREM-3 (like TREM-1) is an activating receptor on macrophages signaling through DAP1218. Hence, to mimic the human situation, we used TREM-1/TREM-3 deficient mice to investigate the role of TREM-1 in infection and inflammation. Nonetheless, caution is warranted with regard to extrapolation of our results to human disease. While this is ac- curate for extrapolation of mouse models to human disease in general, this is especially true for studies on TREM-1. 9

Conclusion Taken together the work presented in this thesis has contributed to our knowledge on TREM receptors during infectious diseases and allergic lung inflammation. In sharp con-

165 Chapter 9 trast to what is known about the role of TREM-1 in systemic inflammation, we here show that TREM-1 serves a protective role during both Gram-positive and Gram-negative pneumonia, while worsening disease outcome during allergic lung inflammation. TREM- 2 on the other hand impairs host defense during Gram-negative pneumonia and has no role in allergic airway inflammation. These results illustrate the complex nature of innate immunity, which in the initial phase of localized infection is essential for an adequate response to invading bacteria but during exaggerated inflammation elicited by high bacterial loads or during sustained allergic inflammation can contribute to collateral damage and tissue injury. While TREM receptors might serve as potential targets for the treatment of inflammatory diseases one must keep in mind there is a fine line between mitigating excessive inflammation and making the immune response ineffective.

166 Summary and general discussion

References

1. Sharif O, Gawish R, Warszawska JM, et al. The triggering receptor expressed on myeloid cells 2 inhibits complement component 1q effector mechanisms and exerts detrimental effects during pneumococcal pneumonia. PLoS Pathog 2014;​10:​e1004167. 2. Bouchon A, Facchetti F, Weigand MA, Colonna M. TREM-1 amplifies inflammation and is a crucial mediator of septic shock. Nature 2001;​410:​1103‑7. 3. Gibot S, Buonsanti C, Massin F, et al. Modulation of the triggering receptor expressed on the myeloid cell type 1 pathway in murine septic shock. Infect Immun 2006;​74:​2823‑30. 4. Gibot S, Kolopp-Sarda MN, Bene MC, et al. A soluble form of the triggering receptor expressed on myeloid cells-1 modulates the inflammatory response in murine sepsis. J Exp Med 2004;​200:​ 1419‑26. 5. Liao R, Liu Z, Wei S, Xu F, Chen Z, Gong J. Triggering receptor in myeloid cells (TREM-1) specific expression in peripheral blood mononuclear cells of sepsis patients with acute cholangitis. In- flammation 2009;​32:​182‑90. 6. Bouchon A, Dietrich J, Colonna M. Cutting edge: inflammatory responses can be triggered by TREM-1, a novel receptor expressed on neutrophils and monocytes. J Immunol 2000;​164:​4991‑5. 7. Netea MG, Azam T, Ferwerda G, Girardin SE, Kim SH, Dinarello CA. Triggering receptor expressed on myeloid cells-1 (TREM-1) amplifies the signals induced by the NACHT-LRR (NLR) pattern recog- nition receptors. J Leukoc Biol 2006;​80:​1454‑61. 8. Wiersinga WJ, Veer Cvt, Wieland CW, et al. Expression Profile and Function of Triggering Receptor Expressed on Myeloid Cells-1 during Melioidosis. Journal of Infectious Diseases 2007;196:​ ​1707‑16. 9. Chen Q, Zhang K, Jin Y, et al. Triggering Receptor Expressed on Myeloid Cells-2 Protects against Polymicrobial Sepsis by Enhancing Bacterial Clearance. Am J Respir Crit Care Med 2013. 10. Hamerman JA, Jarjoura JR, Humphrey MB, Nakamura MC, Seaman WE, Lanier LL. Cutting edge: inhibition of TLR and FcR responses in macrophages by triggering receptor expressed on myeloid cells (TREM)-2 and DAP12. J Immunol 2006;​177:​2051‑5. 11. N’Diaye EN, Branda CS, Branda SS, et al. TREM-2 (triggering receptor expressed on myeloid cells 2) is a phagocytic receptor for bacteria. J Cell Biol 2009;​184:​215‑23. 12. Turnbull IR, Gilfillan S, Cella M, et al. Cutting edge: TREM-2 attenuates macrophage activation. J Immunol 2006;​177:​3520‑4. 13. Allwood EM, Devenish RJ, Prescott M, Adler B, Boyce JD. Strategies for Intracellular Survival of Burkholderia pseudomallei. Front Microbiol 2011;​2:​170. 14. Opitz B, Puschel A, Schmeck B, et al. Nucleotide-binding oligomerization domain proteins are innate immune receptors for internalized Streptococcus pneumoniae. J Biol Chem 2004;​279:​ 36426‑32. 15. Hyams C, Camberlein E, Cohen JM, Bax K, Brown JS. The Streptococcus pneumoniae capsule in- hibits complement activity and neutrophil phagocytosis by multiple mechanisms. Infect Immun 2010;​78:​704‑15. 16. de Vos AF, Dessing MC, Lammers AJ, et al. The Polysaccharide Capsule of Streptococcus pneumo- nia Partially Impedes MyD88-Mediated Immunity during Pneumonia in Mice. PLoS One 2015;​10:​ 9 e0118181. 17. Lambrecht BN, Hammad H. The immunology of asthma. Nat Immunol 2014;​16:​45‑56. 18. Chung DH, Seaman WE, Daws MR. Characterization of TREM-3, an activating receptor on mouse macrophages: definition of a family of single Ig domain receptors on mouse chromosome 17. Eur J Immunol 2002;​32:​59‑66.

167

A Addendum

Nederlandse samenvatting

Nederlandse samenvatting

Dit proefschrift heeft als doel de rol van TREM-receptoren te onderzoeken in pneumonie (longontsteking) en astma. Beide ziektebeelden zijn verantwoordelijk voor veel mor- biditeit in de westerse wereld. In Hoofdstuk 1 geven we een kort overzicht van deze ziektebeelden, de afweerreactie zoals deze optreedt in de long en met name de rol van TREM-receptoren in de aangeboren afweer. Longontsteking is een infectie van de lagere luchtwegen gekenmerkt door symptomen bestaande uit koorts, benauwdheid en hoesten. Jaarlijks krijgen in Nederland 172.000 mensen een pneumonie. Astma is een allergische ontsteking van de luchtwegen en wordt gekenmerkt door episoden van benauwdheid, piepende ademhaling en hoesten. Gezien de opkomst van resistente bacteriën doemt er een scenario op waarin onze zogenaamde “last-resort” antibiotica niet meer werkzaam zullen zijn. Derhalve is een goed begrip van ons afweersysteem tegen bacteriële infecties van groot belang in het kader van de behandeling van pneu- monie. Onze aangeboren afweer (“innate immunity”) behelst een snelle, niet specifieke reactie opgestart door verschillende soorten afweercellen. In de long, de focus van dit proefschrift, zijn dat met name macrofagen. Deze witte bloedcellen zijn uitgerust met eiwitten (receptoren) die kleine stukjes van bacteriën kunnen herkennen (de zoge- naamde “pattern recognition receptors” zoals Toll-like receptoren). Herkenning van een bacterie door een dergelijke receptor initieert een afweerreactie waarbij er verschillende ontstekingsmediatoren vrij komen (lokaal in de long en in het bloed), die er voor zorgen dat er witte bloedcellen vanuit het beenmerg naar de long migreren om daar de infectie te lijf te gaan. Naast een robuuste en adequate afweerreactie is regulatie van dit proces erg belangrijk daar een te veel aan ontsteking weefselschade met zich mee kan bren- gen. Derhalve zijn er verschillende mechanismen in het lichaam aanwezig om dit proces te reguleren. Dit proefschrift gaat over Triggering Receptor Expressed on Myeloid cells (TREM)-receptoren. Deze eiwitten komen met name voor op afweercellen en kunnen de afweerreactie versterken (TREM-1) of afremmen (TREM-2). Deze receptoren kunnen dus als het ware de aangeboren afweer “fine-tunen” en zijn veelbelovende targets in de ontwikkeling van medicijnen tegen onder andere infectieziekten. Eerder onderzoek liet een gunstig effect zien van het remmen van TREM-1 in sepsis (bloedvergiftiging). Sepsis is een klinisch syndroom, vaak ontstaan vanuit een longontsteking, waarbij er sprake is van een niet gebalanceerde reactie van het lichaam op fragmenten van de bacteriële celwand in het bloed (systemische infectie). De gedachte is dat het afremmen van een overmatige ontstekingsreactie een positief effect heeft op de uitkomst van deze ziekte. Inderdaad bleek een TREM-1 remmer erg succesvol als therapie in muizen met ernstige sepsis. De rol van TREM-1 in lokale infectieziekten zoals longontsteking is echter nog nauwelijks onderzocht. In alle hieronder beschreven studies maken we gebruik van zogenoemde “knockout” muizen. Dit zijn genetisch gemodificeerde muizen A

171 Nederlandse samenvatting die het gen/eiwit van interesse niet hebben. In Hoofdstuk 2 onderzochten we de rol van TREM-1 in pneumonie veroorzaakt door een vaak voorkomende Gram-positieve verwekker van dit ziektebeeld, Streptococcus pneumoniae (de “pneumokok”). Hiertoe infecteerden we wild-type muizen (de controle groep) en TREM-1 deficiënte (knockout) muizen met pneumokokken en onderzochten we de afweerreactie door het bepalen van de bacteriële uitgroei en overleving, als mede de werking van macrofagen zonder TREM-1. We vonden dat muizen zonder TREM-1 een slechtere overleving hadden, gekenmerkt door meer bacteriële uitgroei in longen en bloed. Opvallend was dat de vroege afweerrespons in deze muizen ernstig afgezwakt was. Macrofagen zonder TREM-1 bleken minder goed in staat te zijn ontstekingseiwitten te produceren en ook minder goed pneumokokken op te kunnen nemen (fagocyteren). We concludeerden dat TREM-1 op macrofagen de vroege afweerreactie versterkt en noodzakelijk is voor de afweer tegen pneumokokken in de long. TREM-1 en TREM-2 alsmede een aantal andere receptoren die betrokken zijn bij verschillende cellulaire processen, signaleren via een adaptor molecuul genaamd DNAX-protein of 12kDa (DAP12). In Hoofdstuk 3 deden we onderzoek naar de rol van DAP12 in pneumokokken pneumonie. We vonden dat DAP12 deficiënte muizen een verbeterde overleving hadden met minder pneumokokken in de longen en in lever, milt en bloed. Ook hier onderzochten we de werking van witte bloedcellen zonder DAP12. We vonden dat deze cellen beter in staat waren pneumokok- ken op te nemen en heftiger reageerden door meer ontstekingseiwitten te produceren vergeleken met controle cellen. Mogelijk verklaren we hiermee de verbeterde afweer van DAP12 deficiënte muizen in dit type longontsteking. Een andere veel voorkomende verwekker van pneumonie binnen het ziekenhuis maar ook steeds vaker buiten het ziekenhuis is Staphylococcus aureus. Tevens komen er van deze bacterie steeds meer resistente stammen voor (MRSA). Derhalve onderzochten we in Hoofdstuk 4 de rol van TREM-1 in de aangeboren afweerreactie tegen deze bacterie. We vonden dat, net als in het geval van pneumokokken pneumonie, TREM-1 een gunstig effect heeft op de afweer tegen pneumonie veroorzaakt door deze bacterie. Muizen zonder TREM-1 hadden na- melijk meer bacteriën in hun longen en tegelijkertijd minder ontstekingsmediatoren en minder ernstige longpathologie. Na in voorgaande hoofdstukken naar Gram-positieve verwekkers van longontsteking gekeken te hebben, onderzochten we in Hoofdstuk 5 & 6 de rol van TREM-1 en TREM-2 in Gram-negatieve longontsteking. In Hoofdstuk 5 keken we naar zowel de rol van TREM-1 en TREM-2 in pneumonie veroorzaakt door Klebsiella pneumoniae, een vaak voorkomende verwekker van pneumonie binnen het ziekenhuis. We vonden dat muizen zonder TREM-1 sneller stierven en meer bacteriële uitgroei hadden. Tevens beschrijven we dat macrofagen zonder TREM-1 minder ontstekingsme- diatoren konden produceren. Tot onze verbazing vonden we geen rol voor TREM-2 in dit ziektebeeld ook al bleken macrofagen zonder TREM-2 meer cytokines te produceren en minder goed Klebsiella op te kunnen nemen. In Hoofdstuk 6 onderzochten we de

172 Nederlandse samenvatting rol van beide TREM-receptoren in een tropische Gram-negatieve sepsis (melioidose). In tegenstelling tot onze bevindingen met Klebsiella bleek het verhaal hier andersom en vonden we een bescheiden rol voor TREM-1 maar een belangrijke negatieve rol voor TREM-2. TREM-2 deficiënte muizen lieten een sterk verbeterde overleving zien met veel minder bacteriën in longen, bloed, lever en milt. TREM-1 deficiënte muizen daar en tegen waren qua overleving en bacterie aantallen nauwelijks te onderscheiden van con- trole muizen. In Hoofdstuk 7 deden we onderzoek naar een ander soort receptor met een belangrijke rol binnen de aangeboren afweer. Nucleotide-binding Oligomerization Domain-containing (NOD) 2 behoort tot een groep receptoren die fragmenten van de bacteriecelwand binnenin de cel kan herkennen en daardoor bijdraagt aan de aangebo- ren afweer. NOD2 is gebleken in staat stukjes van pneumokokken te herkennen. Gezien het feit dat TREM-1 de afweerreactie die via NOD2 verloopt, versterkt, onderzochten we de rol van NOD2 in pneumokokken pneumonie. In tegenstelling tot onze hypothese, vonden we geen rol voor NOD2 in deze ziekte. We gebruikten verschillende soorten pneumokokken maar vonden slechts verschillen tussen controle muizen en NOD2 deficiënte muizen (met meer uitgroei in longen van laatst genoemde muizen) wanneer we een pneumokok gebruikten die zodanig genetisch gemodificeerd was dat deze geen dik kapsel meer had. Door de afwezigheid van dit kapsel wordt de bacterie vermoedelijk makkelijker opgenomen en herkent door NOD2 in witte bloedcellen. Na onderzoek naar TREM receptoren gedaan te hebben in infecties (pneumonie en sepsis), keken we in Hoofdstuk 8 naar de rol van deze receptoren in allergische longont- steking. Hier toe stelden we TREM-1 deficiëntie, TREM-2 deficiënte en controle muizen herhaaldelijk bloot aan huisstofmijt om zodoende een allergische ontsteking van de luchtwegen te bewerkstelligen als model voor humaan astma. Wanneer we TREM-1 de- ficiënte muizen astma gaven, vonden we dat deze muizen een sterk afgezwakte allergi- sche respons lieten zien. Dit bleek onder andere uit veel lagere ontstekingscel aantallen in de longen en verlaagd IgE in het bloed van TREM-1 deficiënte muizen. Tegen onze verwachtingen in vonden we geen rol voor TREM-2 in deze ziekte.

Conclusie De studies die in dit proefschrift zijn opgenomen hebben sterk bijgedragen aan onze kennis over de rol van TREM-receptoren in infectieziekten en allergische longontste- king. In tegenstelling tot wat tot dusver bekend is over de rol van TREM-1 in sepsis, laten we hier zien dat TREM-1 een beschermende rol heeft in zowel Gram-positieve als Gram-negatieve pneumonie, terwijl het juist allergische luchtwegontsteking ver- ergert. TREM-2 daarentegen, verslechtert juist de afweer tijdens sommige vormen van Gram-negatieve pneumonie en heeft geen rol in allergische luchtwegontsteking. Deze resultaten illustreren de complexheid van de aangeboren afweer, die in de beginfase van gelokaliseerde infectie essentieel is voor een adequate reactie op binnendringende A

173 Nederlandse samenvatting bacteriën maar tijdens overdreven ontsteking veroorzaakt door hoge bacterie aantallen of tijdens langdurige allergische ontsteking kan bijdragen tot nevenschade en weef- selbeschadiging. TREM receptoren zouden mogelijk kunnen dienen als doelwit voor de behandeling van ontstekingsziekten echter door de complexe biologie van deze eiwit- ten zal verder onderzoek noodzakelijk zijn alvorens deze dierexperimentele resultaten naar de kliniek geëxtrapoleerd kunnen worden.

174 PhD portfolio

PhD portfolio

Year Workload (ECTS) Courses Practical Biostatistics 2010 1.1 Laboratory Animals Course (article 9) 2009 3.9 Basic Laboratory Safety 2009 0.4 The AMC world of science 2009 1 Seminars, workshops and master classes Masterclass by Professor Jos van der Meer 2012 0.2 Masterclass by Professor Charles A. Dinarello 2011 0.2 Ruysch Lectures 2010-2013 0.3 Poster presentations TREM-1 improves host defense in pneumococcal pneumonia. 2013 0.5 15th International Congress of Immunology (ICI), Milan, Italy DAP12 impairs host defense during pneumococcal pneumonia. 2013 0.5 15th International Congress of Immunology (ICI), Milan, Italy TREM-1 improves host defense in pneumococcal pneumonia. 2012 0.5 7th International Shock Congress, Miami, USA TREM-1 improves host defense in pneumococcal pneumonia. 2011 0.5 Toll 2011 Meeting, Garda, Italy Congresses and symposia 15th International Congress of Immunology (ICI), Milan, Italy 2013 0.75 7th International Shock Congress, Miami Beach, USA. 2012 1.25 Training the immunity, Nijmegen, The Netherlands 2012 0.3 Toll 2011: Decoding Innate Immunity, Riva del Garda, Italy 2011 1 8th World Congress on Trauma, Shock, Inflammation and Sepsis (TSIS), München, 2010 0.75 Germany Lecturing Invited speaker Center for Inflammatory Bowel Diseases, UCLA, Los Angeles, USA. 2012 0.3 Lecture on TLR signaling in infectious diseases. Tutoring Research project (student): Role of TREM-1 and TREM-2 in klebsiella pneumonia 2012 0.3 Grants Travel Award, International Federation of Shock Societies. 2012 Spinoza grant, Universiteitsbeurs, University of Amsterdam 2012 AMC PhD scholarship, Graduate School, Academic Medical Center 2009

A

175

List of publications

List of publications

Scientific journals: Hommes TJ, Dessing MC, van ‘t Veer C, Florquin S, Colonna M, de Vos AF, Poll TV. TREM- 1/3 Contribute to Protective Immunity in Klebsiella Derived Pneumosepsis Whereas TREM-2 Does Not. Am J Respir Cell Mol Biol. 2015 Apr 10. [Epub ahead of print]

Hommes TJ, Hoogendijk AJ, Dessing MC, Van’t Veer C, Florquin S, de Vos AF, van der Poll T. DNAX-activating protein of 12 kDa impairs host defense in pneumococcal pneumonia. Crit Care Med. 2014 Dec;42(12):e783-90

Hommes TJ, Hoogendijk AJ, Dessing MC, Van’t Veer C, Florquin S, Colonna M, de Vos AF, van der Poll T. Triggering receptor expressed on myeloid cells-1 (TREM-1) improves host defence in pneumococcal pneumonia. J Pathol. 2014 Aug;233(4):357-67. de Boer JD, Roelofs JJ, de Vos AF, de Beer R, Schouten M, Hommes TJ, Hoogendijk AJ, de Boer OJ, Stroo I, van der Zee JS, Veer CV, van der Poll T. Lipopolysaccharide inhibits Th2 lung inflammation induced by house dust mite allergens in mice. Am J Respir Cell Mol Biol. 2013 Mar;48(3):382-9.

Achouiti A, Vogl T, Urban CF, Hommes TJ, van Zoelen MA, Florquin S, Roth J, van ‘t Veer C, de Vos AF, van der Poll T. Myeloid-related protein-14 contributes to protective immunity in Gram-negative pneumonia derived sepsis. PLoS Pathog. 2012; 8(10):e1002987 van der Windt GJ, Hoogendijk AJ, Schouten M, Hommes TJ, de Vos AF, Florquin S, van der Poll T. Osteopontin impairs host defense during pneumococcal pneumonia. J Infect Dis. 2011 Jun 15;203(12):1850-8.

Braat H, Stokkers P, Hommes TJ, Cohn D, Vogels E, Pronk I, Spek A, van Kampen A, van De- venter S, Peppelenbosch M, Hommes D. Consequence of functional Nod2 and Tlr4 muta- tions on gene transcription in Crohn’s disease patients. J Mol Med. 2005 Aug;83(8):601-9.

Book chapters: Hommes TJ, Wiersinga WJ, van der Poll T. The host response to sepsis. In: Vincent J.L. (ed.). Yearbook of Intensive Care and Emergency Medicine 2009. Springer-Verlag. 2009: 39-50. A

177

About the author

About the author

Tijmen Johannes Hommes was born in Hilversum, the Netherlands in 1981. After obtaining his VWO exam at the RSG Magister Alvinus te Sneek in 1999, he moved to Amsterdam to study biomedical sciences (University of Amsterdam). In 2000 he enrolled in a Bachelor of Science program at Indiana University, Bloomington, Indiana. In 2002 het started studying medicine at the University of Amsterdam. During his medical study he worked as a student under the supervision of Professor Sander van Deventer in the lab for experimental internal medicine in the AMC studying host pathogen interactions in inflammatory bowel disease. In 2006 Tijmen gained additional life and lab experience in Barcelona, Spain. Finally he obtained his medical degree in 2009 after doing an elec- tive internship in internal medicine at Tygerberg University, Cape Town, South Africa. He started his PhD project in September 2009 under the supervision of Professor Tom van der Poll at the Center for Molecular and Experimental Medicine at the AMC, studying the innate immune system during sepsis and pneumonia. In January 2014 he started his internal medicine residency at the Onze Lieve Vrouwe Gasthuis in Amsterdam under the supervison of dr. Y. Smets. He will finish his gastroenterology career at Leiden University Medical Center under the supervision of dr. R. Veenendaal.

A

179

Dankwoord

Dankwoord

Dit boekje is tot stand gekomen door de hulp, adviezen en mentale support van heel veel mensen. Ik wil graag een aantal mensen persoonlijk noemen. Te beginnen met mijn promotor Tom van der Poll. Tom, tijdens mijn coschappen wandelde ik op aanraden van Joost Wiersinga jouw kantoor binnen en begonnen we te praten over een promotietra- ject. Vrij snel sleepten we een beurs binnen en was de deal rond. Erg knap hoe je zoveel slimme, talentvolle maar ook gezellige mensen bij elkaar weet te krijgen. Een groep met een unieke open, niet-competitieve sfeer. Daarnaast bewonder ik je schrijftalent; waar ik veel van geleerd heb. Verder is je begeleidende en adviserende rol erg prettig. Je houdt de teugels vrij losjes maar weet op de juiste momenten aan te sporen. Heel veel dank voor het vertrouwen en de mogelijkheid vier jaar lang onderzoek te doen in jouw groep. Dan mijn co-promotor Alex de Vos aan wie ik zeer veel dank verschuldigd ben. Alex, ontzettend bedankt voor je al rake adviezen en praktische begeleiding de afgelopen ja- ren. Zonder jouw hulp had ik het niet kunnen doen. Kees ook jou wil ik bedanken voor je begeleidende rol. Prachtig om te zien hoe enthousiast jij van moleculaire biologie kunt worden. Jouw kennis en inzicht hebben vele stenen bijgedragen aan mijn onderzoek. Mark Dessing, zonder jou geen TREM knockouts! Dank ook voor het meedenken en het sparren over onze TREM-hobby. A special thanks to Sylvia Knapp. It was always very inspiring to have our TREM-talk. Gijs van de Brink, jou wil ik ook bedanken voor de diverse keren dat we hebben gespro- ken over mijn project. Joost Wiersinga, ook met jou kon ik altijd van gedachten wisselen over mijn favoriet eiwitten. Dank voor je bruisende enthousiasme en support. Graag wil ook de overige leden van mijn promotiecommissie danken voor het voor het kritisch beoordelen van mijn manuscript. Verder wil ik de volgende mensen van de afdeling pathologie bedanken. Allereerst Sandrine: merci beaucoup voor het scoren van de pathologie van de pneumonie proeven. Joris bedankt voor het astma deel. En zonder Onno zou er niets te scoren zijn geweest, dank. Vervolgens mijn collega’s, de Tommies. Dit boekje zou net zo dik zijn als jullie boeken als ik op papier zou zetten hoe dankbaar ik jullie stuk voor stuk ben voor de mooie tijd en voor alle steun tijdens de zware momenten. Het waren vier waanzinnige jaren met zeer veel hoogte punten met daar doorheen verweven ook moeilijke momenten. Veel dank voor alle geweldige herinneringen op het lab tijdens de proeven in de MLII, op congres, bij Kelly, in het gasthuis, tijdens de colaatjes drinken, etc. Jullie zijn de zonder twijfel de reden dat ik deze weg tot de laatste meters heb kunnen bewandelen. Daan & Katja, mijn paranimfen. Wat was ik geweest zonder jullie?! Geweldig dat ik jul- lie paranimf mocht zijn en wat vereerd ben ik dat jullie nu de mijne willen zijn. Oneindig veel dank voor jullie luisterende oor in de moeilijke tijden zowel in als buiten het lab. A

181 Dankwoord

En fijn dat we geen collega’s meer zijn maar vrienden! Katja, wat een mooie jaren heb- ben we gehad zowel in als buiten het lab. Samen proeven doen, tikken, gitaar spelen, vlees-kaas-wijn etentjes met Sacha. Prachtige herinneringen voor altijd. Daan, ik weet het nog als de dag van gisteren. Jij was de eerste die mij in de wondere wereld van het promovendus zijn de weg wees. Dit ben je blijven doen tot de laatste letter getypt was. Ontzettend bedankt voor alle adviezen, verhitte discussies, flauwe grappen, roze mix drankjes en het introduceren van een huisstofmijt geïnduceerd allergie muismodel… Veel dank aan de harde kern van het CEMM lab: Danielle, Regina, Sanne, Miranda, Hakima en Monique. Zonder jullie hulp zijn wij dokters met twee linker pipetteerhanden niets! Joost & Marieke, dank voor alle geduld en hulp bij de dierproeven. (Marieke ik hoop dat er geen microfoons hingen in het IWO… Wat een lol hebben wij gehad!) En natuurlijk Heleen en Monique dank voor alle ondersteunende hulp! Ook wil ik mijn collega’s uit het OLVG danken voor de support tijdens het zware vorige jaar en bij het afronden van mijn proefschrift dit jaar. Vrienden, het zit erop. Onderzoek doen, gaat niet over rozen. Dank voor jullie luiste- rende oor, begrip en gin-tonics tijdens de moeilijke momenten. Jullie zijn mij allemaal erg dierbaar en een heerlijke diverse groep mensen om altijd op terug te vallen! ’t Dak gaat eraf vrijdag de 13de! RIFF: Nies, Wiets, Peet en Bo wat een heerlijke uitlaatklep vind ik altijd bij jullie in het repetitiehok. Lieve ouders, dank voor jullie onvoorwaardelijke steun en vertrouwen. Jullie hebben mij van jongs af aan altijd in alles gesteund en alle mogelijkheden geboden om mezelf op wat voor manier dan ook te ontwikkelen. Lieve mama, dank voor het altijd voor me klaar staan en altijd naar me luisteren en je fijne relativeringsvermogen. Lieve papa, wat jammer dat je er de 13de niet bij bent en het uiteindelijke resultaat niet hebt gezien. Maar toch weet ik zeker dat je wist dat het goed ging komen en dat je apetrots bent. En zoals beloofd: ik ga het groots vieren! Daan, lieve broer, speciaal zou ik jou ook willen bedanken voor alle adviezen, inspiratie en motivatie die jij mij altijd geeft. Je energie en enthousiasme werken aanstekelijk en hebben mij mijn gehele promotietraject enorm geholpen. Tot slot, allerliefste Annabel, wat heerlijk dat jij de zon in mijn leven terug hebt ge- bracht. Tijdens die zwaar wegende laatste loodjes stond je me bij en gaf me de energie om dit proefschrift af te maken. Wat heb ik veel zin met jou verder op avontuur te gaan! “Podemos irnos juntos lejos de este mundo tú y yo.”

182