71
University of Veterinary Medicine Hannover Immunology Unit & Research Center for Emerging Infections and Zoonoses Role of C-type lectin receptors in bacterial recognition THESIS Submitted in partial fulfilment of the requirements for the degree Doctor rerum naturalium (Dr. rer. nat.) awarded by the University of Veterinary Medicine Hannover by Sabine Mayer-Lambertz from Prüm Hannover, Germany 2019

Role of C-type lectin receptors in bacterial recognition

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

University of Veterinary Medicine Hannover

Immunology Unit &

Research Center for Emerging Infections and Zoonoses

Role of C-type lectin receptors in bacterial

recognition

THESIS

Submitted in partial fulfilment of the requirements for the degree

Doctor rerum naturalium

(Dr. rer. nat.)

awarded by the University of Veterinary Medicine Hannover

by

Sabine Mayer-Lambertz

from Prüm

Hannover, Germany 2019

Supervisor: Prof. Dr. Bernd Lepenies

Supervision Group: Prof. Dr. Bernd Lepenies

Prof. Dr. Maren von Köckritz-Blickwede

Prof. Dr. Bastian Opitz

1rst Evaluation: Prof. Dr. Bernd Lepenies

Immunology Unit & Research Center for Emerging Infections and

Zoonoses

University of Veterinary Medicine Hannover, Foundation

Hannover, Germany

Prof. Dr. Maren von Köckritz-Blickwede

Institute for Biochemistry, Department of Physiological Chemistry

University of Veterinary Medicine Hannover, Foundation

Hannover, Germany

Prof. Dr. Bastian Opitz

Medical Department, Division of Infectious Diseases and

Pulmonary Medicine

Charité - Universitätsmedizin Berlin

Berlin, Germany

2nd Evaluation: Prof. Dr. Thomas A. Kufer

Department of Immunology

University Hohenheim

Stuttgart, Germany

Date of final exam: 03.04.2019

„Imagination is more important than knowledge“

-Albert Einstein-

Parts of the thesis have been published previously:

Mayer S*, Raulf M-K*, Lepenies B (2017) C-type lectins: their network and roles in

pathogen recognition and immunity. Histochemistry and Cell Biology 147 (2):223-237.

doi:10.1007/s00418-016-1523-7

* S. Mayer und M-K. Raulf contributed equally as joined first authors to this work.

Mayer S, Moeller R, Monteiro JT, Ellrott K, Josenhans C, Lepenies B (2018) C-Type Lectin

Receptor (CLR)–Fc Fusion Proteins As Tools to Screen for Novel CLR/Bacteria Interactions:

An Exemplary Study on Preselected Campylobacter jejuni Isolates. Frontiers in Immunology

9:213. doi:10.3389/fimmu.2018.00213

Imai T, Matsumura T*, Mayer-Lambertz S*, Wells CA, Ishikawa E, Butcher SK, Barnett TC,

Walker MJ, Imamura A, Ishida H, Ikebe T, Miyamoto T, Ato M, Ohga S, Lepenies B, van

Sorge NM, Yamasaki S (2018) Lipoteichoic acid anchor triggers Mincle to drive protective

immunity against invasive group A Streptococcus infection. Proceedings of the National

Academy of Sciences 115(45):E10662-E10671. doi: 10.1073/pnas.1809100115

* T. Matsumura and S. Mayer-Lambertz contributed equally as joined second authors to this work.

Further publications:

Revised manuscript submitted to Cell Reports, 22.11.2018:

Raulf M-K, Johannssen T, Matthiesen S, Neumann K, Hachenberg S, Mayer-Lambertz S,

Steinbeis F, Seeberger P-H, Baumgärtner W, Strube C, Ruland J, Lepenies B The C-type

Lectin Receptor CLEC12A Recognizes Plasmodial Hemozoin and Contributes to Cerebral

Malaria Development.

Manuscript in preparation:

Achilli S, Monteiro J, Serna S, Mayer-Lambertz S, Thepaut M, Leroy A, Ebel C, Reichardt

N, Lepenies B, Fieschi F, Vivès C TETRALEC, Artificial Tetrameric Lectins: a tool to

screen ligand and pathogen interaction.

Table of content

List of abbreviations ................................................................................................................... I

Abstract .................................................................................................................................. - 1 -

Zusammenfassung.................................................................................................................. - 3 -

Chapter 1: General introduction............................................................................................. - 6 -

1. The immune system - essential for survival ................................................................... - 6 -

1.1 Innate Immunity .................................................................................................. - 7 -

1.2 Adaptive Immunity ............................................................................................. - 8 -

2. Of PAMPS, DAMPS and PRRs ................................................................................... - 11 -

2.1 TLRs: their discovery changed the understanding of pathogen sensing ........... - 13 -

2.2 NLRs and RLRs: soluble cytoplasmic sensors of PAMPs and DAMPs ........... - 14 -

2.3 CLRs: A diverse group that recognises sweet PAMPs ..................................... - 16 -

2.3.1 Role of CLRs in anti-bacterial immunity ....................................................... - 18 -

3. Aims of the study ......................................................................................................... - 22 -

Chapter 2: Paper I ................................................................................................................ - 24 -

C-type lectins: their network and roles in pathogen recognition and immunity .............. - 24 -

Chapter 3: Paper II ............................................................................................................... - 26 -

C-type lectin receptor (CLR)-Fc fusion proteins as tools to screen for novel CLR/bacteria

interactions: an exemplary study on preselected Campylobacter jejuni isolates ............. - 26 -

Chaper 4: Paper III ............................................................................................................... - 29 -

Lipoteichoic acid anchor triggers Mincle to drive protective immunity against invasive

group A Streptococcus infection ...................................................................................... - 29 -

Chapter 5: Discussion .......................................................................................................... - 32 -

Chapter 6: Future outlook .................................................................................................... - 41 -

Chapter 7: Concluding remarks ........................................................................................... - 43 -

References ............................................................................................................................ - 44 -

Affidavit ............................................................................................................................... - 61 -

Acknowledgement ............................................................................................................... - 62 -

List of abbreviations

I

List of abbreviations

ABTS 2,2'-Azinobis [3-ethylbenzothiazoline-6-sulfonic acid]-diammonium salt

AIM 2 absent in melanoma 2

APC antigen-presenting cell

AP 1 activator protein 1

BMDC bone marrow-derived dendritic cell

BMM bone marrow-derived macrophage

CARD9 caspase recruitment domain family member 9

CD cluster of differentiation

CHO chinese hamster ovary

Clec C-type lectin

CLR C-type lectin receptor

CpG DNA CpG oligodeoxynucleotides

CRD carbohydrate-recognition domain

CTL cytotoxic T lymphocyte

CTLD C-type lectin-like domain

DC dendritic cell

DCAR dendritic cell immunoactivating receptor

DCIR dendritic cell immunoreceptor

DC-SIGN dendritic cell-specific ICAM-grabbing non-integrin

Dectin DC-associated C-type lectin

DGDG diglucosyldiacylglycerol

ds double-stranded

ECM extracellular matrix

ELISA enzyme-linked immunosorbent assay

ER endoplasmic reticulum

FC fragment crystallizable

FcR Fc receptor

FcRγ Fc receptor common gamma-chain

FITC fluorescein isothiocyanate

GAS group A Streptococcus

GalNAc N-acetylgalactosamine

List of abbreviations

II

GBS group B Streptococcus

G-CSF granulocyte-colony stimulating factor

GFP green fluorescence protein

GI gastrointestinal

Glu glutamate

HIF-1 hypoxia-inducible factor-1

HRP horseradish peroxidase

IFN interferon

Ig immunoglobulin

IL interleukin

IPS-1 interferon-beta promoter stimulator 1

ITAM immunoreceptor tyrosine-based activation motif

ITIM immunoreceptor tyrosine-based inhibitory motif

LBP lipopolysaccharide-binding protein

LOS lipooligosaccharide

LPS lipopolysaccharide

LRR leucine-rich repeat

LTA lipoteichoic acid anchor

ManLAM mannose-capped lipoarabinomannan

MAPK mitogen-activated protein kinases

MCL macrophage C-type lectin

MDA5 melanoma differentiation-associated gene 5

MDP muramyl dipeptide

MGDG monoglucosyldiacylglycerol

MGL macrophage galactose-type C-type lectin

MHC major histocompatibility complex

Mincle macrophage-inducible C-type lectin

MIP macrophage inflammatory protein

MR mannose receptor

MyD88 myeloid differentiation factor 88

NFAT nuclear factor of activated T cells

NF-κB nuclear factor kappa-light-chain-enhancer of activated B cells

List of abbreviations

III

NK cell natural killer cell

NLR NOD-like receptor

NO nitric oxide

NOD nucleotide-binding oligomerization domain

PAMP pathogen-associated molecular pattern

PE phycoerythrin

PHDs prolyl hydroxylase domain enzymes

Pro proline

Raf rapidly accelerated fibrosarcoma

RANTES regulated on activation normal T cell expressed and secreted

RIG-I retinoic acid inducible gene I

RLR RIG-I-like receptor

ROS reactive oxygen species

PRRs pattern recognition receptors

SAP130 Sin3A associated protein 130

SIGNR DC-SIGN-related protein

ss single-stranded

STING stimulator of interferon genes

Syk spleen tyrosine kinase

TAP transporter associated with antigen processing

TCR T cell receptor

TDB trehalose-6,6-dibehenate

TDM trehalose-6,6-dimycolate

Th T helper cell

TIR Toll-Interleukin 1 Receptor

TLR Toll-like receptor

TNF tumor necrosis factor

TRAF TNF receptor associated factor

Treg regulatory T cell

TRIF TIR-domain-containing adapter-inducing interferon-β

Abstract

- 1 -

Abstract

Sabine Mayer-Lambertz: Role of C-type lectin receptors in bacterial recognition

The most important function of the innate immune system is the recognition of pathogens that

may cause damage to the host. To sense foreign structures, host immune cells express so-

called pattern-recognition receptors (PRRs) on their surface to detect highly conserved

pathogen-associated molecular patterns (PAMPs) present in all classes of pathogens like

bacteria, viruses, parasites, and fungi. PRRs comprise soluble and transmembrane receptors,

including intracellular receptors as nucleotide-binding oligomerization domain-like receptors,

retinoic acid inducible gene I receptors and Toll-like receptors, but also the extracellularly

located C-type lectin receptors (CLRs). CLRs were originally described as calcium-

dependent carbohydrate-binding proteins. Nowadays, it is known that CLRs additionally

detect other ligands than carbohydrates such as lipids, proteins, ice crystals and inorganic

molecules. Furthermore, also calcium-independent ligand binding of CLRs has been

described. Macrophage-inducible C-type lectin (Mincle) is a type II transmembrane protein

expressed by monocytes, neutrophils, macrophages and dendritic cells. Activation of Mincle

by ligand recognition leads to phosphorylation of spleen-tyrosine kinase that in turn induces

the formation of the ternary CARD9/Bcl10/Malt1 complex and the activation of the

transcription factor NFκ-B. Finally, NFκ-B triggers effector functions such as the production

of cyto- and chemokines, cell degranulation and phagocytosis. Bacteria are known to express

various carbohydrate structures (glycans) on their surface that play important roles in host

cell adhesion, invasion and immune evasion. Since these structures are crucial for bacterial

colonization, survival, growth and initiation of pathogenesis, there is rising interest in

detecting novel CLR/bacteria interactions to prevent infections.

Chapter 3 of this thesis deals with the detailed description of three different methodologies to

detect yet unknown CLR/bacteria interactions using the very frequent human pathogen

Campylobacter jejuni as model organism. All methods presented can be easily applied to

other Gram-negative and Gram-positive bacteria and build on a comprehensive CLR-hFc

fusion protein library. CLR-hFc fusion proteins consist of the Fc part of a human IgG1 fused

to the carbohydrate-recognition domain (CRD) of the respective CLR. First, an ELISA-based

high-throughput assay is described that allows for an initial pre-screening for CLR/bacteria

interactions. This method relies on plate-coated bacteria that are incubated with the respective

Abstract

- 2 -

fusion proteins. Finally, binding is detected by reaction of the colorimetric substrate ABTS

with a secondary anti-human IgG1 Fc antibody coupled to horseradish peroxidase. Among

others, Dectin-1-hFc was found to bind to C. jejuni isolates MHH19 and MHH24. Since

false-positive CLR candidates may occur due to protein aggregation, a following flow

cytometry-based assay was established to confirm potential binding partners. Here, bacteria

were kept in solution, incubated with the CLR-hFc fusion proteins and binding was detected

by the usage of an anti-human IgG1-Fc antibody in a subsequent flow cytometric analysis.

Applying this protocol, binding to Dectin-1-hFc was confirmed for both C. jejuni isolates.

Finally, a confocal microscopy approach is presented that enables direct visualization of the

CLR binding to bacteria. Also, in this assay, Dectin-1-hFc was found to recognise both

C. jejuni isolates. In summary, the described methods based on CLR-hFc fusion proteins

offer the possibility to screen for various novel CLR/bacteria interactions. The detection of

these candidates presents the first step towards bacterial ligand identification and the analysis

of the biological function of the respective CLRs.

The identification of Mincle as CLR that recognises parts of the lipoteichoic acid anchor

(LTA) present in the cell wall of the human pathogen group A Streptococcus (GAS) and

thereby promoting antibacterial immunity is part of chapter 4. Gene expression analysis and

cell stimulation assays revealed that the Mincle adaptor protein CARD9 is involved in the

immune responses against GAS infection. Stimulation of bone marrow-derived dendritic cells

(BMDCs) with purified monoglucosyldiacylglycerol (MGDG) of LTA led to a Mincle-

dependent production of inflammatory cytokines, reactive oxygen species (ROS) and the

activation of inducible nitric oxide synthase (iNOS). In addition, the purified

diglucosyldiacylglycerol (DGDG) of the LTA is recognised by Mincle but does not initiate

signaling. Furthermore, it was found that DGDG serves as suppressor of Mincle whereas

MGDG mediates agonistic Mincle activation. In vivo, Mincle-deficient mice infected with

GAS strains others than the 5448 exhibited high mortality in line with severe bacteraemia and

an increased expression of proinflammatory cytokines. In summary, these results suggest a

protective role for Mincle in anti-GAS immunity.

This work presents comprehensive methods to detect novel CLR/bacteria interactions and

contributed to the identification of GAS-derived MGDG and DGDG as new Mincle ligands.

Zusammenfassung

- 3 -

Zusammenfassung

Sabine Mayer-Lambertz: Die Rolle von C-Typ Lektin Rezeptoren in der

Erkennung von Bakterien

Eine der wichtigsten Aufgabe des angeborenen Immunsystems ist die Erkennung von

Erregern, die in den Wirt eindringen und ihn schädigen. Um diese körperfremden Strukturen

zu detektieren, tragen die Immunzellen des Wirtes sogenannte Mustererkennungsrezeptoren

(engl.: „Pattern-recognition receptors“, PRRs) auf ihrer Oberfläche, die konservierte

Pathogen-assoziierte molekulare Muster (engl.: „Pathogen-associated molecular patterns“,

PAMPs) in allen Klassen von Erregern wie Bakterien, Viren, Parasiten und Pilzen erkennen.

PRRs umfassen lösliche und membrangebundene Rezeptoren sowie intrazelluläre Rezeptoren

wie nucelotide-binding oligomerization domain-like Rezeptoren, retinoic acid inducible gene

I Rezeptoren und Toll-like Rezeptoren. Zusätzlich zählen zu den PRRs auch die extrazellulär

lokalisierten C-Typ Lektin Rezeptoren (CLRs). Ursprünglich wurden CLRs als

kalziumabhängige Kohlenhydrat-bindende Proteine beschrieben. Dieser Kenntnisstand

änderte sich jedoch mit der Entdeckung, dass CLRs auch Lipide, Proteine, Kristalle und

anorganische Moleküle erkennen und dass diese Bindung nicht unbedingt durch Kalzium

vermittelt werden muss. Macrophage-inducible C-type lectin (Mincle) ist ein Typ II

Transmembranprotein, welches von Makrophagen, Dendritischen Zellen, Neutrophilen und

Monozyten exprimiert wird. Die Aktivierung von Mincle durch die Bindung von Liganden an

die Kohlenhydrat-erkennende Domäne (engl.: „Carbohydrate-recognition domain“, CRD)

führt zur Phosphorylierung der „Spleen-tyrosine kinase“ (Syk), die die Bildung des

CARD9/Bcl10/Malt1 Komplexes und die Aktivierung des Transkriptionsfaktors NFκ-B

initiiert. Letztlich werden NFκ-B-vermittelte Effektorfunktionen wie die Produktion von

Zytokinen und Chemokinen, Phagozytose und Degranulation ausgelöst. Bakterien besitzen

eine Vielzahl von Kohlenhydratstrukturen (Glykane) auf ihrer Oberfläche. Diese Strukturen

spielen eine wichtige Rolle in der Adhäsion und dem Eindringen in die Wirtszelle sowie bei

der Immunevasion. Da diese Strukturen essentiell für eine erfolgreiche bakterielle Besiedlung

und Pathogenese im Wirt sind, stellt die Entdeckung neuer CLR/Bakterien-Interaktionen

einen wichtigen ersten Ansatzpunkt dar, um eine bakterielle Infektion zu verhindern.

Das Kapitel 3 beschäftigt sich mit drei unterschiedlichen Methoden, um bislang unbekannte

CLR/Bakterien-Interaktionen aufzudecken. Dabei werden die Methoden anhand des

Zusammenfassung

- 4 -

humanpathogenen Bakteriums Campylobacter jejuni beschrieben. Allerdings können die

Methoden auf weitere Gram-negative sowie Gram-positive Bakterienstämme ausgedehnt

werden. Alle Techniken basieren auf einer CLR-hFc Fusionsprotein-Bibliothek. Die CLR-

hFc Fusionsproteine bestehen aus dem Fc-Teil eines humanen IgG1, das mit der CRD des

entsprechenden Rezeptors fusioniert wurde. Ein ELISA-basierter Hochdurchsatz-Assay wird

als erstes durchgeführt, der initiale Ergebnisse zu möglichen CLR/Bakterien-Interaktionen

liefert. Diese Methode basiert auf der Immobilisierung der Bakterien, die anschließend mit

den CLR-hFc Fusionsproteinen inkubiert werden. Die Umsetzung des Substrats ABTS durch

einen sekundären anti-human IgG1-Fc Antikörper, konjugiert an das Enzym

Meerrettichperoxidase, ermöglicht die Detektion von CLR/Bakterien-Interaktionen. Auf

diese Weise wurde Dectin-1 als Rezeptor für die C. jejuni Isolate MHH19 und MHH24

entdeckt. Da diese Methode durch die Bildung von Proteinaggregaten die Möglichkeit von

falsch-positiven Ergebnissen birgt, wurde ein nachfolgender durchflusszytometrischer Ansatz

zur Verifizierung der Kandidaten durchgeführt. Dabei wurden die Bakterien in Lösung mit

den Fusionsproteinen und einem sekundären anti-human IgG1 Fc Antikörper inkubiert, bevor

die Proben durchflusszytometrisch analysiert wurden. Hierbei konnte die Bindung von

Dectin-1-hFc an beide C. jejuni-Isolate bestätigt werden. Zum Schluss wurde die Dectin-1-

hFc/C. jejuni-Interaktion durch Konfokalmikroskopie visualisiert. Zusammenfassend

ermöglichen die drei auf CLR-Fusionsproteinen basierenden Methoden das Screening von

einer Vielzahl an Bakterienstämmen auf neue CLR-Interaktionen. Dies ist ein erster Schritt,

der zur Identifikation eines distinkten CLR-Liganden und zum besseren Verständnis von

dessen biologischer Funktion führt.

Das Kapitel 4 dieser These beschreibt die Identifikation von Strukturen des

Lipoteichonsäureankers (LTA) in Gruppe A Streptokokken (GAS) als Mincle-Liganden, die

in der Lage sind, eine Immunantwort gegen GAS auszulösen. Genexpressionsstudien und

Zellstimulationsexperimente zeigten, dass CARD9, ein Adaptermolekül von Mincle, an der

Immunantwort gegen GAS beteiligt ist. Die Stimulation von Makrophagen, die aus dem

Knochenmark differenziert wurden, mit aufgereinigtem Monoglucosyldiacylglycerol

(MGDG) aus dem LTA führte zu einer Mincle-abhängigen Produktion von

proinflammatorischen Zytokinen, reaktiven Sauerstoffspezies (ROS) und der Aktivierung der

Stickstoffmonoxid-Synthase (iNOS). Zusätzlich wurde gezeigt, dass von dem LTA

aufgereinigtes Diglucosyldiacylglycerol (DGDG) von Mincle erkannt wird, jedoch zu keiner

Zusammenfassung

- 5 -

Signalweiterleitung innerhalb der Zelle führt. Außerdem konnte gezeigt werden, dass DGDG

Mincle inhibiert, während MGDG als Mincle-Agonist fungiert. Die Infektion von Mäusen

mit einigen GAS-Stämmen zeigte, dass Mincle-Defizienz zu einer gesteigerten Anfälligkeit

auf Grund von erhöhter Bakterienlast und Expression von proinflammatorischen Zytokinen

führte. Alles in allem zeigen diese Ergebnisse, dass Mincle eine schützende Rolle in der

Immunabwehr gegen GAS besitzt.

Zusammenfassend präsentiert diese Arbeit umfangreiche Methoden, um neue Interaktionen

von CLRs und Bakterien zu detektieren. Außerdem trägt diese Arbeit zu der Entdeckung bei,

dass die beiden LTA Bestandteile MGDG und DGDG aus GAS als neue Mincle Liganden

fungieren.

Introduction

- 6 -

Chapter 1: General introduction

1. The immune system - essential for survival

“I find it astonishing that the immune system embodies a degree of complexity which suggests

some more or less superficial though striking analogies with human language, and that this

cognitive system has evolved and functions without assistance of the brain” -Niels K. Jerne-

The human body is challenged every day by multifaceted microorganisms, like fungi, viruses,

parasites and bacteria that attempt to invade and colonize the host. In addition to these

environmental pathogens, the body is naturally colonized by approximately 1014

microorganisms that harbor due to dysbiosis an increased risk for causing disease to the host

(1, 2). Nevertheless, it is the result of the immune system as a powerful defense system that

the host is usually able to limit the infection and eliminate pathogens fast. The immune

system (lat.: immunis; free from, devoid of) is one of the most complex systems in the human

body, forming a remarkable network of different tissues, cells and proteins that cooperate to

protect the body from foreign structures such as microbes, viruses, cancer cells and toxins

(3). The immune system is composed of two potent branches, the innate and the adaptive

immune system (figure 1). Whereas for fast and effective protection of the host both parts

have to collaborate using humoral and cellular components, each part clearly mediates

distinct functions. The innate immune system initiates the first responses after the host is

exposed to pathogens. To allow a fast detection of and response to invading pathogens, the

innate immune system provides various genetically predefined receptors, so-called pattern-

recognition receptors (PRRs), that recognize evolutionarily highly conserved microbial

structures on a broad spectrum of pathogens (Pathogen-associated molecular patterns,

PAMPs) or frequent signs of infections (Danger-associated molecular patterns, DAMPs).

However, this only induces a general immune response that includes the induction of

inflammatory responses, the process of tissue repair, the recruitment of immune cells and the

release of cytokines and chemokines. These processes are activated directly after the

infection occurred and they are not influenced in strength by repeated exposure to the same

antigen (3-5). In contrast, dependent on the activation by the innate immune system, the

adaptive immune system relies on a defined repertoire of specific receptors that enables

response to any antigen (6). Since the development of these highly specific components

requests time, the adaptive immune system is only activated after several days of infection.

The adaptive immune system displays memory, thereby providing an augmented immune

Introduction

- 7 -

reaction towards antigens that had already been encountered previously by the host (4, 7).

However, any disturbed immune function in parts of the immune system increases the

susceptibility of the host to infections, highlighting the importance of an efficient interaction

of the innate and adaptive immune system (3).

1.1 Innate Immunity

The innate immune system is the crucial first line of defence against infections and includes

the epithelium as the initial and main barrier against pathogens. The epithelium is located in

the respiratory tract, skin and gastrointestinal (GI) tract; hence, it is the part that is challenged

most frequently with invading pathogens. Thus, it offers multifaceted strategies to prevent

microbial entry and spread. Tight junctions connect cells with each other and provide an

important physical barrier (8) whereas mucus produced by the respiratory and GI tract

enables rapid entrapment of microbes and protects the epithelial surface. In addition, physical

and chemical barriers such as temperature, low pH or the secretion of antimicrobial

substances kill or neutralize pathogens (4, 9, 10). Importantly, tissue cells that encountered

pathogens produce cytokines that further activate innate immune cells and guide them to the

site of infection (11). The innate response also includes a soluble fraction that comprises

acute-phase proteins, interferons (IFNs), antimicrobial peptides and the complement system

(figure 1). Most notably, the complement system consists of more than 30 plasma proteins

that are cascade-like activated to identify, opsonize and kill pathogens or infected cells either

directly or by phagocytosis. Furthermore, it activates and mobilizes side by side with released

cytokines the cellular component of the innate immune system that comprises monocytes,

granulocytes, macrophages, mast cells, dendritic cells (DCs) and natural killer (NK) cells

(figure 1) (3, 12).

Of these cell subsets, DCs and macrophages are termed professional antigen-presenting cells

(APCs) since they are able to present antigens to T cells in the presence of co-stimulatory

molecules (figure 1), hence linking innate with adaptive immunity. Besides that,

macrophages are characterised by their pronounced phagocytic ability. If the epithelial barrier

is breached by physical damage and pathogens can invade the host, macrophages incorporate

intruders into large intracellular organelles, so-called phagosomes. This compartment fuses

with a lysosome containing enzymes, peptides and proteins to a phagolysosome and

subsequently leads to the degradation of the internal content (10, 13). In addition to the pure

Introduction

- 8 -

elimination of pathogens, peptide fragments can be presented by macrophages on major

histocompatibility complex (MHC) II molecules to T cells, leading to their activation. In

addition, macrophages initiate a potent response towards intracellular pathogens via the

IFNγ-mediated activation by Th1-cells (3, 4).

Whereas macrophages are defined by their phagocytic activity, the primary function of DCs

is the activation of the adaptive immune system by the presentation of antigens to T cells.

Immature DCs are resident throughout the tissues where they capture antigens with surface

located receptors, e.g. CLRs, but also via Fc receptors (FcRs) (14, 15). The processing of

respective antigens into proteolytic peptides is mediated in the next step via phagocytosis,

macropinocytosis or receptor-mediated endocytosis and leads to the maturation of DCs (16).

DCs take up larger organisms such as parasites or fungi and they are able to ingest various

Gram-positive and Gram-negative bacteria as well as mycobacteria (17). The uptake of

Salmonella by the gut epithelium is directly mediated by DCs that open tight junctions to

expose their dendrites to directly capture bacteria (18). In addition, DCs are often the target

of virus infections. Surface molecules on DCs enable viruses to enter the cells and start the

production of virus-associated particles in the cytoplasm. To ensure an effective immune

response towards these various extra- and intracellular stimuli, DCs express two different

classes of MHC molecules that trigger different immune response pathways. Whereas

endogenous (intracellular) peptides presented on MHC I molecules activate CD8+ cytotoxic T

lymphocytes (CTLs), MHC II molecules present exogenous peptides to CD4+ T cells (6, 19).

In addition to this classical antigen-presentation pathway, the mechanism of cross-

presentation enables presentation of extracellular peptides to naïve CD8+ T cells to become

activated CD8+ CTLs. This pathway is especially important for the initiation of immune

responses towards peripheral tissue cells others than antigen-presenting cells infected with

viruses or even against tumor cells (20).

1.2 Adaptive Immunity

Whereas more primitive organisms like invertebrates and plants exclusively rely on the innate

immune system, higher animals further developed the adaptive immune system: a clonal- and

antibody-based second branch that enables immunological memory, hence increasing the

reaction towards reoccurring intruders (12, 21). In parallel to the innate immune system, the

adaptive immune system is activated. The adaptive immune system is characterised by the

Introduction

- 9 -

formation of pathogen-specific pathways. Hallmark cells of this branch are antigen-specific T

cells and antibody-producing B cells that represent clonal lymphocyte populations (figure 1).

Whereas all lymphocyte progenitor cells develop from hematopoietic stem cells in the bone

marrow, where B cells persist to undergo maturation, immature T cells are subsequently

recruited to the thymus to reach their final maturation state (7).

T cells are characterised by specific surface-expressed T cell receptors (TCRs). These TCRs

recognize antigen fragments presented by APCs on MHC I or MHC II molecules. In addition

to the TCR/peptide/MHC interaction, a binding of the costimulatory molecules CD80 or

CD86 located on mature APCs to CD28 expressed on T cells is required to fully activate T

cells (22). Upon recognition of the antigen bound to MHC-I, naïve CD8+ CTLs differentiate

into mature CTLs. Once activated, mature CD8+ T cells release effector molecules, such as

perforin and granzyme B, that induce the apoptosis of infected and abnormal cells (6). After

successful elimination of foreign agents, only few cells survive as memory T cells that

protect the host against reinfection due to their rapid response (23). The presentation of

antigens bound to MHC-II molecules stimulates CD4+ T cells. These cells regulate in

particular the type of immune response by the release of cytokines that enable an enhanced

reaction to eliminate pathogens. Therefore, CD4+ T cells differentiate into various types of T

helper (Th) cells (figure 1) (24). Most notably, Type 1 helper (Th1) cells mediate a cellular

immune response by the release of IFNγ and interleukin (IL)-2, whereas a reaction initiated

by Th2 cells is characterized by IL-4, IL-5, IL-10 and IL-13 production, that in turn

efficiently activates naïve B cells to produce antibodies (humoral immune response) (6).

Th17 cells are characterized by their ability to produce IL-17. IL-17A and IL-17F are known

to be important proinflammatory cytokines that induce the production of IL6 and tumor

necrosis factor α (TNFα) as well as the recruitment of granulocytes (6). Th9 cells play an

important role in allergy and autoimmunity as well as in parasitic helminth infection (25) and

tumor eradication (26). Additionally, regulatory T cells (Tregs), another CD4+ T cell subset,

suppress or downregulate immune responses, thereby playing an important role in

maintaining immune homeostasis (27).

Introduction

- 10 -

Figure 1: Components of the innate and adaptive immune system. Both branches of the immune system

contain cellular and humoral components. The innate immune system resembles the first line of host defense

and comprises phagocytic immune cells like macrophages as well as classical antigen-presenting cells (APCs)

such as dendritic cells (DCs). The humoral part includes host defense peptides like antimicrobial peptides or the

LPS Binding Protein. If the innate immune response is unable to clear the pathogens, the adaptive immune

system is initiated after several hours. Crucial for the activation is the recognition of pathogen-associated

molecular patterns (PAMPs) or damage-associated molecular patterns (DAMPs) by cellular receptors located on

APCs, e.g. C-type lectin receptors and the subsequent presentation of peptide fragments loaded on major

histocompatibility complexes (MHC) to T cells via T cell receptor (TCR)/MHC-interaction. Antigen

presentation stimulates immature T cells to differentiate into cytotoxic T cells (CTL) or T helper (Th) cells.

Activation of Th cells drives down one of several differentiation pathways. One subset represents the regulatory

T cells (Tregs) which dampen immune activation. In contrast, Th1 response activates killing of intruders, Th2

cells are involved in antibody production and clearance of helminths whereas Th17 cells induce inflammatory

responses. Th9 cells play a role in anti-tumor immunity, worm infections as well as in autoimmunity and

allergy. (4)

B cells mediate an important role in the humoral part of the adaptive immune system. Each B

cell expresses approximately 1.5x105 molecules of an unique antigen-binding receptor (BCR)

on its surface that allows a direct recognition and highly specific binding of an antigen (2).

Once activated by foreign antigens, naïve B cells divide rapidly by clonal expansion into

plasma cells or into memory B cells. Plasma cells produce a large amount of respective

Introduction

- 11 -

antibodies that in turn are able to bind to specific antigens on pathogens, hence flagging them

for uptake by phagocytes. Whereas these plasma cells die after one to two weeks, memory B

cells survive and continue the production of antibodies (28). Long-living memory T and B

cells present important survivors of an overcome infection and B cells produce antibodies

that allow immunological memory, enabling a faster and effective immune reaction towards

previously encountered antigens.

2. Of PAMPS, DAMPS and PRRs

For a long time the adaptive immune system was seen as the more powerful arm of

immunity. With the introduction of a model of microbial pattern recognition by Charles

Janeway Jr., the innate immune system got more into the focus of many research groups.

Janeway`s prediction relies on a limited number of germ line-encoded PRRs that have

genetically determined specificities for various microbes and other foreign molecules (5) and

are mainly present on APCs and other immune but also non-immune cells (29). PRRs

recognize highly conserved structures called PAMPs that are abundantly expressed by all

classes of pathogens including viruses, protozoa, fungi as well as Gram-positive and -

negative bacteria (30). Since PAMPs such as LPS or bacterial CpG DNA are exclusively

present in microbes and essential for pathogen survival and integrity, they represent suitable

molecules for microbial sensing by host cells (5, 29). However, besides the recognition of

external-derived PAMPs, PRRs also sense endogenous DAMPs. DAMPs represent host

factors that are either released by the extracellular matrix (ECM) or from cells upon infection,

inflammation or other types of cellular stress (31). Upon disruption of the cell, DAMPs that

for example are under healthy conditions attached to the cytoskeleton or the ECM, such as

actin (32) or fibrinogen (33), are proteolytically released into the extracellular space and are

involved in inflammation and cell regeneration processes in a PRR-dependent manner (34).

In addition to ECM-derived DAMPs, various soluble molecules are intracellularly generated

or accessible during cellular necrosis or apoptosis. According to their origin within the cell,

these molecules can be classified into different groups: mitochondria-derived DAMPs such as

mitochondrial DNA (35); DAMPs like ATP (36, 37) and IL-1β (38) that are produced during

the process of autophagy as well as nuclear (e.g. DNA (39), histones (40)) and cytosolic (e.g.

uric acid (41)) DAMPs (34).

Introduction

- 12 -

Up to now, the receptor system consists of several large families of PRRs (figure 2) including

Toll-like receptors (TLRs) and C-type lectin receptors (CLRs) that are membrane-bound,

hence they detect pathogens present in endocytic compartments or in the extracellular space.

Additionally, unbound intracellular receptors such as NOD-like receptors (NLRs), RIG-I-like

receptors (RLRs) and AIM2-like receptors (ALRs) are located in the cytoplasm, hence they

are responsible for the detection of intracellular agents e.g via the endoplasmic reticulum

membrane-resident adaptor stimulator of interferon genes (STING) (42). Remarkably, this

compartmentalization of the receptors provides important information on the class of detected

pathogens as well as their location, thus indirectly indicating their potential damage to the

host cell (43, 44). For example, it is known that the extracellular presence of Salmonella is

detected in a TLR-dependent manner leading to proinflammatory cytokine production and

phagocytosis whereas the intracellular detection of Salmonella leads to death of the

respective cell, initiated by NLRs (45). Along that line, the recognition of self-derived nucleic

acids is connected to multiple disorders (46, 47). Therefore, compartmentalization of TLRs

and their ligands provides a mechanism that enables the receptor to distinguish between

foreign and self-derived nucleic acids (48). For example, the expression of TLR9 in

intracellular compartments prevents the recognition of self-derived nucleic acids but enables

sensing of virus-derived DNA (49).

Stimulation of PRRs by ligand recognition leads to the activation of distinct adaptor

molecules that in turn trigger specific intracellular signaling pathways. Adaptor proteins play

a crucial role in signal integration from several receptors and enable the detection of various

ligands. Finally, the initiation of an enzymatic signal activates immune responses including

inflammatory pathways such as the stimulation of APCs by the production of

proinflammatory cytokines and IFNs (43, 50). This further elicits the adaptive immune

responses. In addition, the recruitment of PRRs also leads to direct events like autophagy

(51), cell death (52) and phagocytosis (53). These mechanisms finally orchestrate to allow for

a fast and efficient response towards the intruders (43).

Introduction

- 13 -

Figure 2: Pattern-recognition receptors (PRRs) with individual family members and their ligands. Pathogen-associated molecular patterns (PAMPs) are detected by several PRRs located in the cytoplasm of the

immune cells such as NOD-like receptors (NLRs) or RIG-I-like receptors (RLRs). Furthermore, members of the

Toll-like receptor (TLRs) and C-type lectin receptor (CLRs) families are expressed as transmembrane receptors.

This figure shows the main classes of PRRs including some members with their described ligands as examples.

(54, 55)

2.1 TLRs: their discovery changed the understanding of pathogen sensing

TLRs are probably the most intensively investigated class of PRRs. The first receptor

discovered was named Toll and mediates dorsoventral development during embryogenesis in

the fruit fly Drosophila melanogaster (56). Later, it was detected that Toll additionally plays

an important role in the protection of flies against fungal infections (57). Up to now, 11

human (TLR1-10) and 12 murine (TLR1-9 and TLR11-13) homologues of Toll are described

(58). TLRs are type I transmembrane glycoproteins and are mostly expressed by APCs like

macrophages, DCs, and B lymphocytes (48). TLRs are characterized by an extracellular

leucine-rich domain (LRR) that mediates ligand binding and a Toll/IL-1 receptor domain

(TIR) for signal transduction, hence located in the intracellular or cytoplasmic space (59). So

far, TLR ligands that are recognized by the LRR domain are described for most of the TLRs

as well as the role of TLRs in host response to various microbes such as viruses, bacteria,

Introduction

- 14 -

fungi and parasites (50, 60-64). PAMP binding to the respective TLR induces receptor

oligomerization followed by the induction of intracellular signaling that generates a potent

antimicrobial proinflammatory response (65). For most of the TLRs, except for TLR3, the

recruitment of the adaptor protein myeloid differentiation primary response gene 88 (MyD88)

is essential for induction of downstream signaling cascades (66). TLR3 in contrast interacts

with Toll/IL-1R-domain-containing adaptor-inducing IFN-β (TRIF) that together with a

bridging molecule is also used by TLR4. The engagement of various downstream adaptors

involved in signal transduction finally leads to the translocation of the nuclear factor kappa B

(NFκ-B) to the nucleus where it induces subsequently the expression of target genes involved

in inflammatory responses towards intruders (2, 54).

TLRs are classified according to the nature of their PAMP ligand into two subclasses.

Whereas TLR1, TLR2, TLR4, TLR5 and TLR6 recognise lipoproteins and lipoglycans,

TLR3, TLR7, TLR8 and TLR9 are involved in the detection of nucleic acids (50). Focusing

on bacterial TLR ligands, the interaction of TLR4 and bacterial lipopolyssacharide (LPS) that

is ubiquitously present in the cell wall of Gram-negative bacteria has been described most

intensively (67, 68). Since a TLR4/LPS interaction alone only leads to insufficient cell

signaling, the accessory proteins LPS-Binding Protein (LBP) and MD2 assist in LPS binding

and recognition by TLR4 (69, 70). In addition to many bacterial cell wall components that

serve as PAMPs for TLRs including lipoarabinomannan (TLR2 (71)), flagellin (TLR5 (72))

and lipoteichoic acid anchor (LTA, TLR2 (73-75), TLR2/TLR6 (76)), also bacteria-derived

DNA that contains hypomethylated CpG domains is recognized (TLR9 (77)). Furthermore,

TLRs are divided according to their localization within the cell. TLR1, TLR2, TLR4, TLR5

and TLR6 are expressed at the cell surface whereas the nucleic acid sensing TLRs (TLR3,

TLR7, TLR8, TLR9) are located in membranes of the endosomal compartment (48). To

enable sensing of intracellular cytosolic pathogen-derived PAMPs, like viral double-stranded

(ds)RNA or single-stranded (ss)RNA, cells express additionally a large repertoire of cytosolic

PRRs including RLRs and NLRs (65).

2.2 NLRs and RLRs: soluble cytoplasmic sensors of PAMPs and DAMPs

NLRs are a group of PRRs that have gained rising interest in the last years since all described

members are connected to certain human diseases. NLRs are important cytosolic sensors of

mainly bacteria-derived molecules present in the cytoplasm but also of environmental

Introduction

- 15 -

components and host cell molecules (50). Examples of ligands derived from bacteria

are: pore-forming toxins, flagellin proteins, muramyl dipeptide (MDP), anthrax lethal toxin or

microbial RNA and DNA (78). In general, NLRs are characterized by multiple LRRs that

mediate ligand recognition, a nucleotide-binding oligomerization domain (NOD) responsible

for oligomerization upon ligand binding and a protein-protein interaction domain, that is

specific for each NLR (79). Up to now, 22 NLRs are described that, based on their N-

terminal part, are divided into 4 subfamilies including CIITA, NLRB, NLRC and NLRP (80).

Additionally, NLRs can also be classified by their effector functions that they initiate upon

ligand binding (81). Accordingly, some members of NLRs including NLRP1, NLRP3 and

NLRC4 form a signaling complex, the inflammasome, that has a pivotal role in inflammatory

cell death as well as processing and maturation of the proinflammatory cytokines IL-1β and

IL-18 (82). Moreover, activated NLRs like NOD1 and NOD2 affect signal transduction via

downstream adaptor molecules that lead to the activation of NFκ-B or activator protein 1

(AP1), hence enhancing the transcription of various proinflammatory cytokines (83, 84). In

addition to the initiation of signal transduction, NLRs such as CIITA and NLRC5 are

described to induce with the transcription of MHC-II and I genes, respectively, as the lack of

MHC-II expression in patients with bare lymphocyte syndrome can be completely

compensated by CIITA (85-87). Furthermore, NLRs are also involved in the process of

autophagy, a fundamental cellular process of antigen elimination. NOD1 and NOD2 are

known to induce autophagy to remove bacterial components (88).

Whereas NLRs represent soluble receptors that recognise various cytoplasmic pathogens,

RLRs play a pivotal role in the detection and defense against viruses. Up to now, the family

consists of three members: RIG-I, MDA5 and LGP2 which commonly recognize RNA

viruses and subsequently lead to the induction of type I IFN, hence eliciting antiviral

response (54). MDA5 recognizes picornaviruses as encephalomyocarditis virus whereas RIG-

I is described to recognize several ssRNA viruses like Sendai virus, paramyxovirus and

influenza virus (89, 90). The detection of the 5’-triphosphate present in virus ssRNA enables

a selective discrimination of viral RNA from host RNA by RIG-I (91). Ligand recognition by

RIG-I or MDA5 leads to a complex downstream signaling that involves several signaling

adaptor proteins such as IFN-β promotor stimulator 1 (IPS-1) (92). Finally, the activated

signal cascades result in the expression of type I IFNs and proinflammatory factors that are

involved in efficient antiviral defense (54, 93).

Introduction

- 16 -

2.3 CLRs: A diverse group that recognises sweet PAMPs

CLRs represent a highly diverse group of promiscuous receptors that were originally

described to show a strong affinity for mannose, fucose and glucan carbohydrate structures

present in all classes of pathogens, including viruses, parasites, yeast, fungi as well as Gram-

positive and Gram-negative bacteria (94). In general, the group of CLRs comprises both,

soluble and cell membrane-bound proteins (95). CLRs are characterized by the expression of

one or more carbohydrate recognition domains (CRDs) or a C-type lectin-like domain

(CTLD). Classically, CRD/carbohydrate binding is mediated in a Ca2+-dependent manner as

Ca2+-ions are essential for ligand complexation and receptor integrity, hence determining its

activity. CTLDs are also present in CLRs that recognize other PAMPs than carbohydrates

such as proteins, crystals or lipid moieties. In addition, ligand recognition by these CLRs is

mainly mediated in a Ca2+-independent way (96, 97). Based on several criteria like

phylogeny and structure, vertebrate CLRs are divided into 17 groups (98). Commonly, CLRs

of the myeloid group are structured into an extracellular part with the CRD or CTL, a

transmembrane segment and a cytoplasmic domain that in most cases harbors the signaling

motifs of the respective CLRs. In the immune system, CLRs elicit various diverse functions

like cell-cell adhesion, signal transduction and turnover of plasma glycoproteins (99).

Nevertheless, one of the main function of CLRs is the recognition and subsequent uptake of

pathogens. After being internalized, pathogens are degraded in lysosomes and their fragments

are presented on MHC molecules by APCs to induce an adaptive immune response (100).

Due to the large number of members within the CLR superfamily, the following section

focuses on the subfamily of myeloid CLRs that consists of members with a pivotal role in

PAMP detection and subsequent modulation of immune responses.

These CLRs are expressed by myeloid cells, especially APCs like macrophages, DCs and B

cells and are involved in pathogen binding, uptake and degradation, thus playing a role in

activation and regulation of antimicrobial responses (101). Moreover, CLRs are subclassified

by their regulatory effect on the outcome of the initiated immune response. Accordingly,

CLRs such as Dectin-2, macrophage-inducible C-type lectin (Mincle) (figure 3) and DCAR

are associated with adaptor molecules (e.g. Fc Receptor common gamma-chain (FcRγ))

containing immunoreceptor tyrosine-based activation motif (ITAM, YxxL/I) domains that

activate downstream signaling pathways. Alternatively, CLRs like Dectin-1, SIGNR3 and

Introduction

- 17 -

Clec9a carry a single ITAM motif called hemITAM in their intracellular domain (102-104).

Upon ligand detection, ITAM- and also to a certain extent hemITAM bearing CLRs (105,

106) induce cell signaling by recruitment of spleen tyrosine kinase (Syk), that in turn leads to

the phosphorylation of tyrosines and activation of various intermediate molecules, including

the activation of transcriptional pathways such as mitogen-activated protein kinases (MAPK)

(107) and nuclear factor of activated T cells (NFAT) (102, 108). In addition, signaling via

Syk activates the NLRP3 inflammasome that induces the maturation and secretion of IL-1β,

hence playing a role in antifungal host defence (109-112). Essentially, Syk signaling leads to

the formation of an adaptor complex of Caspase recruitment domain-containing 9 (CARD9),

B cell lymphoma 10 (Bcl10) and mucosa-associated lymphoid tissue lymphoma translocation

protein 1 (Malt1) that triggers proinflammatory gene expression via activation of the NFκ-B

pathway (figure 3) (113, 114). CLRs with immunoreceptor tyrosine-based inhibitory motif

(ITIM, S/I/V/LxYxxI/V/L) domains mediate suppression of effector functions by activation

of tyrosine phosphatases (SHP-1, SHP-2, SHIP). This group comprises CLRs such as

Clec12a, DCIR and Clec12b. In addition, CLRs including Mannose-receptor (MR), SIGNR1

and DC-SIGN use complex signaling pathways independent of an ITAM or ITIM motif

(115). However, some CLRs are able to trigger a Syk-independent activation of the NFκ-B

pathway by recruitment of the serine/threonine kinase Raf1 (116). Finally, activation of CLRs

by ligand binding induces various cell responses like endocytosis, phagocytosis, production

of chemokines and cytokines, complement activation and induction of the adaptive immune

system (101).

Since pathogens express multiple PAMPs that can be recognized by several receptors, some

CLRs perform crosstalk with different PRRs, including TLRs to enable a response

concurrently against a broad range of pathogens (117). For example, it is well known that

only the cooperation of DC-SIGN with TLRs triggers a cellular response whereas DC-SIGN

signaling alone does not stimulate cytokine production in general, hence it rather modulates

TLR-mediated responses (118, 119). In addition to crosstalk with other PRRs than CLRs, the

formation of CLR-derived homo- or heterocomplexes, termed multimerization, affects

receptor signaling and effector functions. Especially CLRs with hemITAMs like Clec2 tend

to form homodimers since the successful activation of Syk requires two phosphorylated

tyrosines (120, 121). Moreover, DC-SIGN, a CLR that lacks signaling motifs, forms

tetramers that increases receptor avidity and specificity (122). However, the engagement of

Introduction

- 18 -

CLRs like Mincle (figure 3), Dectin-1 and Dectin-2 induces various immune responses upon

pathogen detection that are independent of other PRRs.

Figure 3: CLR engagement triggers various immune responses. This is displayed in this graphic for Mincle

as example. The Mincle carbohydrate-recognition domain (CRD) senses diverse glycan structures from several

pathogens like fungi and bacteria. Additionally, also danger-associated molecules released by damaged or dead

cells activate Mincle. Upon activation, the spleen-tyrosine kinase (Syk) is recruited via the ITAM-bearing FcRγ

stalk which leads to the formation of the CARD9/Bcl10/Malt1 complex and further to the activation of the

transcription factor NFκB. The subsequent production of cytokines and the activation of other effector functions

initiate important mechanisms involved in host defense, tissue repair mechanisms and autoimmune diseases.

(123)

The following section will provide a comprehensive introduction into the role of CLRs in

anti-bacterial immunity with a focus on the CARD9-dependent CLRs Mincle and Dectin-1.

These CLRs were identified to play an important role in bacterial recognition during the

projects that are included in this thesis.

2.3.1 Role of CLRs in anti-bacterial immunity

Bacteria express various virulence factors such as adhesins, toxins and enzymes that

contribute to pathogenesis in the host. Carbohydrate-based structures such as the M-protein,

lipoteichoic acid anchor (LTA), capsular polysaccharide (CPS) or O-antigen domains derived

from bacterial lipopolyssacharides (LPS) facilitate bacteria/host interactions (124-128).

Introduction

- 19 -

Hence, these structures are essential for adherence and subsequent colonization of host tissue

by bacteria, thus mediating the first and most important step in establishment of an infection.

Since bacterial glycosylation is usually the first “pattern” encountered by cells of the host

immune system, there is growing interest on the role of CLRs in bacterial recognition. The

impact of CLRs is best described for Mycobacterium tuberculosis. Up to now, several CLRs

including MR (129), DC-SIGN (130), Mincle (131, 132) and Dectin-1 (133) have been

described to interact with M. tuberculosis (134). However, whereas several reports on

CLR/M. tuberculosis interactions are described in vitro, the respective receptors showed only

limited to redundant role in clearance of the infection in vivo (130, 135, 136). MR was shown

to recognise mannose-capped lipoarabinomannan (ManLAM) present on the surface of

M. tuberculosis and affected the phagocytosis and assembly of the phagolysosome (129,

137). Additionally, it is reported that the tetrameric transmembrane protein DC-SIGN

recognises mannose moieties in ManLAM structures of mycobacteria and influences the

expression of costimulatory molecules and immune responses in human macrophages (138).

Although several murine homologs of DC-SIGN exist, SIGNR3 has been identified as the

closest functional orthologue of human DC-SIGN based on binding studies (139, 140).

Infection of SIGNR3-deficient mice with M. tuberculosis led to increased bacterial load in

the early phase of infection. However, in the later course of infection, these mice efficiently

exhibited an adaptive immune response, comparable to wild-type mice (130). Besides

M. tuberculosis, DC-SIGN is also described to interact with other bacteria like Helicobacter

pylori and a number of Lactobacillus strains (94). Here, the surface layer A protein of L.

acidophilus was detected as ligand for DC-SIGN (141).

Although contradictory reports on the role of Mincle in murine infection studies with

different bacterial species exist, its adaptor signaling protein CARD9 mediates an essential

role in defense against Mycobacterium species, since CARD9-deficient mice showed

exacerbated pathogenesis and rapid death in an infection study in vivo (142). The

mycobacterial cell wall glycolipid trehalose-6,6-dimycolate (cord factor, TDM) is a well-

known ligand for the CLR Mincle, capable of initiating an inflammatory response including

granuloma formation in mice (131, 132). In addition to TDM, Mincle also recognises the

synthetic analog trehalose-6,6-dibehenate (TDB) (131). The crystal structure of the

recognition sites of bovine and human Mincle binding to trehalose unraveled a canonical C-

type primary monosaccaride-binding site that complexes a Ca2+ ion (143, 144). Moreover, a

Introduction

- 20 -

primary and a proximal secondary binding site complex both glucose moieties in the head

group of the trehalose whereas a hydrophobic groove opposite of the primary binding side

was shown to sense fatty acids with a minimum of 10 carbon atoms (131, 143, 145).

Interestingly, the design of mono- and diacetylated derivatives of trehalose with different

numbers of attached acyl chains resulted in an increased affinity for Mincle binding with

rising number of carbon atoms (146) (for further structural information about Mincle, see

(147)). Mincle is a typ II transmembrane receptor that is mainly expressed by myeloid cells

like macrophages, DCs, monocytes and neutrophils (148-152). However, the receptor was

also found to be present on B cells (153). Activation of Mincle upon ligand binding to the

CRD initiates the phosphorylation of the ITAM motif present in the FcRγ receptor associated

to Mincle. Subsequently, the recruitment of Syk leads to the formation of the

CARD9/Bcl10/Malt1 complex that activates NFκB-signaling pathway resulting in the

expression of chemokines, proinflammatory cytokines such as IL-12 and TNF-α, induction of

phagocytosis and nitric oxide (NO) production (figure 3) (148, 154, 155). Whereas most of

the effector functions are proinflammatory, Mincle also induces the production of IL-10,

hence the activation of anti-inflammatory responses, thereby being involved in both, immune

homeostasis and immune modulation towards microbial pathogens (156). Mincle is described

to interact with a number of diverse bacterial strains, like Streptococcus pneumoniae (157,

158), Klebsiella pneumoniae (159), H. pylori (160) and several Corynebacterium (161) and

Lactobacillus strains (162, 163), but the actual Mincle ligand for most of the interactions in

still unknown. Recently, it was shown that glucosyldiacylglycerol from the important human

pathogens S. pneumoniae and group A Streptococcus (GAS, S. pyogenes) is recognised by

Mincle (157, 164). Glucosyldiacylglycerol is part of the LTA that is the major component of

the cell wall of Gram-positive bacteria. Besides binding of Mincle to the respective bacteria,

it was additionally demonstrated that the lack of Mincle in mice infected with the respective

bacteria resulted in increased mortality and dysregulated anti-bacterial immunity, hence

pinpointing to a protective role of Mincle (157, 164). Aside from sensing bacteria, Mincle

furthermore detects self-molecules such as SAP-130 (155) or β-glucosylceramide that derived

from damaged or necrotic cells (165) and pathogens like parasites (Leishmania major) and

fungi (101).

In fungi, the myeloid cell-derived CLR Dectin-1 is known to be the major receptor for β-1,3-

linked glucan, an ubiquitous cell wall component of fungi (166, 167). Hence, its role in

Introduction

- 21 -

sensing fungal pathogens such as Pneumocystis carinii (168), Candida albicans (168, 169),

Aspergillus fumigatus (170-172) and Cryptococcus neoformans (173) has been intensively

studied (174). Upon ligand detection by the single CTLD, Dectin-1 recruits Syk via

phosphorylation of the hemITAM motif present in the cytoplasmic tail, followed by the

engagement of the CARD9/Bcl10/Malt1 complex and subsequent activation of the NFκ-B

pathway with the production of inflammatory cytokines and induction of other effector

functions (175). Although several studies report a role for Dectin-1 in anti-mycobacterial

defense in vitro, its role in vivo as well as the definite ligand present in mycobacteria still

remains unclear since Dectin-1-deficient mice challenged with M. tuberculosis showed no

significant difference in pathology and induced immune responses compared to wild-type

mice (176). Moreover, Dectin-1 crosstalk with TLR2 is required for efficient response

including phagocytosis, inflammatory cytokine production and induction of the MAPK

pathways towards mycobacterial species in vitro (177).

Aims of the study

- 22 -

3. Aims of the study

CLRs play an important role in pathogen detection and the subsequent initiation and

modulation of various immune responses. Many interactions of CLRs and several pathogens

like viruses, bacteria, parasites and fungi are described. Bacteria present interesting study

targets since they express large numbers of glycoconjugates on their surface. In the field of

microbiology, the role of CLRs is best described for M. tuberculosis, whereas for other

bacteria the impact of CLRs still has to be elucidated more extensively. Importantly, some

bacteria such as S. pneumoniae or H. pylori are shown to interact with CLRs but for most of

the bacteria, distinct ligands of the respective receptors are still unknown. Moreover,

targeting specific CLRs presents an interesting strategy to deliver drugs or vaccine antigens

into specific cells to selectively modulate immune responses. Unraveling new CLR/bacteria

interactions presents the first step towards identification of new CLR ligands and to get better

insights into the biological relevance of the respective receptor.

Aim 1: Establishment of different in vitro methods to detect novel CLR/bacteria

interactions

Based on the usage of a comprehensive CLR-hFc fusion protein library (178) and

Campylobacte jejuni as representative model for various other bacteria, diverse methods have

been established to enable screening for novel CLR/bacteria interactions. First, an ELISA-

based assay allowed for high-throughput screening of plate-bound bacteria. Second, a flow

cytometric assay has been established to verify potential CLR binding candidates and finally,

the binding specificity of the respective CLR has been visualized using a confocal

microscopy-based method. The combination of these three distinct methods should allow for

identification of new CLR interactions with bacterial ligands, hence paving the way for

further functional studies.

Aim 2: Unraveling the role of the CLR Mincle in the recognition of group A

Streptococcus

GAS is an important human pathogen that can cause various diseases, ranging from self-

limiting infections like pharyngitis or impetigo to severe outcomes such as necrotizing

fasciitis or streptococcal toxic shock syndrome (179). Since several studies suggest that next

Aims of the study

- 23 -

to TLRs also other PRRs play an important role in the detection of GAS (180, 181), it was

investigated if CLRs may play a role in sensing GAS. In a collaborative approach,

transcriptome analysis and cell culture-based methods like stimulation of bone marrow-

derived DCs (BMDCs) and bone marrow-derived macrophages (BMMs) were applied to first

identify a novel CLR that interacts with GAS, followed by analysis of the antibacterial

response initiated by this CLR. Finally, the in vivo role of the respective CLR was analysed

by murine infection studies using the respective CLR-deficient mice.

Chapter 2 Paper I

- 24 -

Chapter 2: Paper I

C-type lectins: their network and roles in pathogen recognition and immunity

Histochem Cell Biol., (2017) | https://doi.org/10.1007/s00418-016-1523-7

Title: C-type lectins: their network and roles in pathogen recognition and immunity

Sabine Mayer1°, Marie-Kristin Raulf1,2°, Bernd Lepenies1*

1Immunology Unit & Research Center for Emerging Infections and Zoonoses, University of

Veterinary Medicine, Hannover, Germany

2Institute for Parasitology, Center for Infection Medicine, University of Veterinary Medicine,

Hannover, Germany

°These authors contributed equally to this manuscript.

*Corresponding author:

Prof. Dr. Bernd Lepenies, University of Veterinary Medicine Hannover, Immunology Unit &

Research Center for Emerging Infections and Zoonoses, Bünteweg 17, 30559 Hannover,

Germany

Phone: +49(0)511/953-6135; Email: [email protected]

The extent of Sabine Mayer-Lambertz´s contribution to the article is evaluated according to

the following scale:

A. has contributed to collaboration (0-33%).

B. has contributed significantly (34-66%).

C. has essentially performed this study independently (67-100%).

1. Design of the project including design of individual experiments: /

2. Performing of the experimental part of the study: /

3. Analysis of the experiments: /

4. Presentation and discussion of the study in article form: B

Chapter 2 Paper I

- 25 -

Abstract

C-type lectins (CTLs) represent the most complex family of animal/human lectins that

comprises 17 different groups. During evolution, CTLs have developed by diversification to

cover a broad range of glycan ligands. However, ligand binding by CTLs is not necessarily

restricted to glycans as some CTLs also bind to proteins, lipids, inorganic molecules, or ice

crystals. CTLs share a common fold that harbors a Ca2+ for contact to the sugar and about 18

invariant residues in a phylogenetically conserved pattern. In vertebrates, CTLs have

numerous functions, including serum glycoprotein homeostasis, pathogen sensing, and the

initiation of immune responses. Myeloid CTLs in innate immunity are mainly expressed by

antigen- presenting cells and play a prominent role in the recognition of a variety of

pathogens such as fungi, bacteria, viruses, and parasites. However, myeloid CTLs such as the

macrophage inducible CTL (Mincle) or Clec-9a may also bind to self-antigens and thus

contribute to immune homeostasis. While some CTLs induce pro-inflammatory responses

and thereby lead to activation of adaptive immune responses, other CTLs act as inhibitory

receptors and dampen cellular functions. Since CTLs are key players in pathogen recognition

and innate immunity, targeting CTLs may be a promising strategy for cell-specific delivery of

drugs or vaccine antigens and to modulate immune responses.

Chapter 3 Paper II

- 26 -

Chapter 3: Paper II

C-type lectin receptor (CLR)-Fc fusion proteins as tools to screen for novel

CLR/bacteria interactions: an exemplary study on preselected Campylobacter

jejuni isolates

Front Immunol., (2018) | https://doi.org/10.3389/fimmu.2018.00213

Title: C-type lectin receptor (CLR)-Fc fusion proteins as tools to screen for novel

CLR/bacteria interactions: an exemplary study on preselected Campylobacter jejuni

isolates

Sabine Mayer1, Rebecca Moeller1°, João T. Monteiro1, Kerstin Ellrott2,4, Christine

Josenhans2,3,4,5*, Bernd Lepenies1*

1 Immunology Unit & Research Center for Emerging Infections and Zoonoses, University of

Veterinary Medicine, Hannover, Germany

2 Institute for Medical Microbiology, Medical School Hannover, Hannover, Germany

3 Max von Pettenkofer Institute, Ludwig Maximilian University Munich, Munich, Germany

4 German Center for Infection Research (DZIF), partner site Hannover-Braunschweig

5 German Center for Infection Research (DZIF), partner site Munich

° Present address: Institute for Experimental Virology, TWINCORE, Centre for Experimental

and Clinical Infection Research, a joint venture between the Medical School Hannover and

the Helmholtz Centre for Infection Research, Hannover, Germany

*Corresponding authors:

Prof. Dr. Christine Josenhans, Medical School Hannover, Institute for Medical Microbiology,

Carl-Neuberg-Strasse 1, 30625 Hannover, Germany

Phone: +49511532-4354; Email: [email protected]

Prof. Dr. Bernd Lepenies, University of Veterinary Medicine Hannover, Immunology Unit &

Research Center for Emerging Infections and Zoonoses, Bünteweg 17, 30559 Hannover,

Germany

Phone: +49(0)511/953-6135; Email: [email protected]

Chapter 3 Paper II

- 27 -

The extent of Sabine Mayer-Lambertz´s contribution to the article is evaluated according to

the following scale:

A. has contributed to collaboration (0-33%).

B. has contributed significantly (34-66%).

C. has essentially performed this study independently (67-100%).

1. Design of the project including design of individual experiments: B

2. Performing of the experimental part of the study: C

3. Analysis of the experiments: C

4. Presentation and discussion of the study in article form: B

Chapter 3 Paper II

- 28 -

Abstract

C-type lectin receptors (CLRs) are carbohydrate-binding receptors that recognize their

ligands often in a Ca2+-dependent manner. Upon ligand binding, myeloid CLRs in innate

immunity trigger or inhibit a variety of signaling pathways, thus initiating or modulating

effector functions such as cytokine production, phagocytosis, and antigen presentation. CLRs

bind to various pathogens, including viruses, fungi, parasites, and bacteria. The bacterium

Campylobacter jejuni (C. jejuni) is a very frequent Gram-negative zoonotic pathogen of

humans, causing severe intestinal symptoms. Interestingly, C. jejuni expresses several

glycosylated surface structures, for example, the capsular polysaccharide (CPS),

lipooligosaccharide (LOS), and envelope proteins. This “Methods” paper describes

applications of CLR–Fc fusion proteins to screen for yet unknown CLR/bacteria interactions

using C. jejuni as an example. ELISA-based detection of CLR/bacteria interactions allows a

first prescreening that is further confirmed by flow cytometry-based binding analysis and

visualized using confocal microscopy. By applying these methods, we identified Dectin-1 as

a novel CLR recognizing two selected C. jejuni isolates with different LOS and CPS

genotypes. In conclusion, the here-described applications of CLR–Fc fusion proteins

represent useful methods to screen for and identify novel CLR/bacteria interactions.

Chapter 4 Paper III

- 29 -

Chaper 4: Paper III

Lipoteichoic acid anchor triggers Mincle to drive protective immunity against

invasive group A Streptococcus infection

PNAS, (2018) | https://doi.org/10.1073/pnas.1809100115

Lipoteichoic acid anchor triggers Mincle to drive protective immunity against invasive

group A Streptococcus infection

Takashi Imaia,b,c, Takayuki Matsumurad,1, Sabine Mayer-Lambertze,1, Christine A. Wellsf, Eri

Ishikawab,g, Suzanne K. Butcherf, Timothy C. Barnetth,i, Mark J. Walkerh,i, Akihiro Imamuraj,

Hideharu Ishidak,l, Tadayoshi Ikebem, Tomofumi Miyamoton, Manabu Atod, Shouichi Ohgac,

Bernd Lepeniese, Nina M. van Sorgeo, and Sho Yamasakia,b,g,p,2

aDivision of Molecular Immunology, Medical Institute of Bioregulation, Kyushu University,

Fukuoka 812-8582, Japan

bDepartment of Molecular Immunology, Research Institute for Microbial Diseases,

cDepartment of Pediatrics, Graduate School of Medical Sciences, Kyushu University,

Fukuoka 812-8582, Japan

dDepartment of Immunology, National Institute of Infectious Diseases, Tokyo 162-8640,

Japan

eImmunology Unit and Research Center for Emerging Infections and Zoonoses, University of

Veterinary Medicine, 30559 Hannover, Germany

fThe Centre for Stem Cell Systems, Anatomy and Neuroscience, Faculty of Medicine,

Dentistry and Health Sciences, University of Melbourne, Melbourne, VIC, Australia

gDepartment of Molecular Immunology, Immunology Frontier Research Center, Osaka

University, Osaka 565-0871, Japan

hSchool of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane,

QLD 4072, Australia

iAustralian Infectious Diseases Research Centre, The University of Queensland, Brisbane,

QLD 4072, Australia

jDepartment of Applied Bio-organic Chemistry, Gifu University, Gifu 501-1193, Japan.

kFaculty of Applied Biological Sciences and Center for Highly Advanced Integration of Nano

and Life Sciences (G-CHAIN), Gifu University, Gifu 501-1193, Japan

Chapter 4 Paper III

- 30 -

lCenter for Highly Advanced Integration of Nano and Life Sciences, Gifu University, 501-

1193 Gifu, Japan

mDepartment of Bacteriology I, National Institute of Infectious Diseases, Tokyo 162-8640,

Japan

nDepartment of Natural Products Chemistry, Graduate School of Pharmaceutical Sciences,

Kyushu University, Fukuoka 812-8582, Japan

oMedical Microbiology, University Medical Center Utrecht, Utrecht University, 3584 CX

Utrecht, The Netherlands

pDivision of Molecular Immunology, Medical Mycology Research Center, Chiba University,

Chiba 260-8673, Japan

1These authors contributed equally

2Correspondence to Sho Yamasaki: [email protected]

The extent of Sabine Mayer-Lambertz´s contribution to the article is evaluated according to

the following scale:

A. has contributed to collaboration (0-33%).

B. has contributed significantly (34-66%).

C. has essentially performed this study independently (67-100%).

1. Design of the project including design of individual experiments: A

2. Performing of the experimental part of the study: A

3. Analysis of the experiments: A

4. Presentation and discussion of the study in article form: A

Chapter 4 Paper III

- 31 -

Abstract

Group A Streptococcus (GAS) is a Gram-positive bacterial pathogen that causes a range of

diseases, including fatal invasive infections. However, the mechanisms by which the innate

immune system recognizes GAS are not well understood. We herein report that the C-type

lectin receptor macrophage inducible C-type lectin (Mincle) recognizes GAS and initiates

antibacterial immunity. Gene expression analysis of myeloid cells upon GAS stimulation

revealed the contribution of the caspase recruitment domain-containing protein 9 (CARD9)

pathway to the antibacterial responses. Among receptors signaling through CARD9, Mincle

induced the production of inflammatory cytokines, inducible nitric oxide synthase, and

reactive oxygen species upon recognition of the anchor of lipoteichoic acid,

monoglucosyldiacylglycerol (MGDG), produced by GAS. Upon GAS infection, Mincle-

deficient mice exhibited impaired production of proinflammatory cytokines, severe

bacteremia, and rapid lethality. GAS also possesses another Mincle ligand,

diglucosyldiacylglycerol; however, this glycolipid interfered with MGDG-induced activation.

These results indicate that Mincle plays a central role in protective immunity against acute

GAS infection.

Discussion

- 32 -

Chapter 5: Discussion

This work presents comprehensive methods to detect yet unknown CLR/bacteria interactions

that further enable to investigate the importance of CLRs in the recognition of and the

response to bacterial pathogens, as shown here for the interaction of LTA-derived glycolipids

from GAS and the CLR Mincle.

Sensing pathogens by CLRs expressed on APCs influences the immune responses in several

regards, including antigen presentation, phagocytosis, complement activation and immune

homeostasis (101). Therefore, identification of CLRs and their respective ligands that

enhance or modulate immune responses by inhibition or activation of signaling pathways is

of great interest. Bacteria express a huge repertoire of several carbohydrate-containing

structures on their surface that are essential for bacterial growth and survival (182). Since

CLRs are known to sense carbohydrate structures, bacteria present promising organisms to

study the impact of CLRs in pathogen recognition and induction of signaling pathways. As an

important initial step, the first project (chapter 3, paper II (183)) presents different in vitro

methods that allow for identification of novel CLR/bacteria interactions. It is worth to

mention that all methods can be easily applied to Gram-negative and Gram-positive bacteria,

but each method clearly has its own advantages and disadvantages. The ELISA-based method

relies on the immobilization of the bacteria on a plate which might lead to false-positive

CLR-candidates due to protein aggregation. Although the ELISA offers the possibility for

high-throughput screening, a following flow cytometry-based assay is recommended to

confirm the respective CLR candidates. Application of an adequate gating strategy enables

the identification of bacteria, hence ruling out debris or other contaminants in the sample.

Nevertheless, only bacteria that are detectable by size in the flow cytometer can be analysed.

The detection of small bacteria like mycoplasma may cause problems (184). Finally, a

confocal microscopy-based method allows for co-localisation studies that directly visualize

CLR/bacteria interactions (183).

All methods described are based on soluble fusion proteins, that consist of a Fc portion of a

human IgG1 and the CRD of a CLR (178, 185). In clinical applications, fusion proteins are

frequently used, since they offer several advantages. Favorably, the structure of the Fc part

enables bivalent presentation of the ligand, hence increasing receptor avidity. Additionally,

Discussion

- 33 -

the Fc part impacts important aspects of the fusion proteins like stability and solubility and

folds autonomously (186). Furthermore, purification of the proteins is performed fast, simply

and with low costs via the Fc region using well established protein-G/A affinity

chromatography methods (187). Since the first discovery of a CD4-Fc fusion protein that

prevented infection of T cells with human immune deficiency virus in 1989 (188), the interest

in Fc-fusion molecules has been constantly increasing. Up to now, several Fc-fusions are

already used in therapeutic applications (186). Here, fusion proteins are predominantly used

to target specific receptors and mediate agonistic or antagonistic functions to enhance or

inhibit immune responses, respectively. Among the approved Fc-fusion protein therapeutics,

etanercept is applied as TNF-α-inhibitor that prevents proinflammatory responses in patients

with severe, active rheumatoid arthritis (189). Furthermore, dulaglutide, a recombinant fusion

protein consisting of a glucagon-like peptide-1 analog linked to a modified IgG4 Fc-fragment

was recently approved as therapeutics to control the blood sugar level in patients with type 2

diabetes (190). A current approach is the fusion of peptides that specifically bind cancer cells

to the Fc-domain to induce antibody-dependent cellular cytotoxicity (191).

Apart from their relevance in clinical applications, the use of Fc-fusion proteins in biomedical

research has attracted increasing interest over the last decades. Since the interactions of CLRs

with their carbohydrate ligands are characteristically weak, the expression as Fc-fusion

proteins that form homodimers with bivalent CRD presentation increases the avidity of each

protein (192). This enables even the detection of low affinity receptor/ligand interactions and

renders Fc-fusion proteins interesting tools to analyse these interactions. For example, fusion

proteins are used in microarrays to detect novel receptor ligands. These protein, lectin and

glycan arrays rely on plate-coated biomolecules, like proteins as lectins, or carbohydrates.

The binding of other proteins, cells, carbohydrates, antibodies or Fc-fusion proteins to these

components is commonly evaluated by laser-based methods (193, 194). Recently,

carbohydrate arrays including synthetically produced glycan structures as potential ligands

and CLR-hFc fusion proteins were applied to evaluate the receptor binding preference in

regard to ligand structure and multivalency (195, 196).

Applying a set of recombinant fusion proteins, several CLRs were shown to interact with

bacteria like S. pneumoniae (158) or fungi such as Malassezia pachydermatis (197),

Gnadoderma lucidum (198) and Cordyceps sinensis (198). Although several interactions of

Discussion

- 34 -

CLRs with pathogens are described, the specific ligand involved is still unknown in most

cases. Since bacteria that express glycoconjugates in their surface structures harbor potential

ligands for CLRs, these bacteria are of special interest in the field of CLR research. For

instance, C. jejuni incorporates several different carbohydrates into important structures like

the lipooligosaccharides (LOS), N and O-linked glycoproteins as well as the peptidoglycan

and the polysaccharide capsule (199). One publication showed that N-glycosylated proteins

and LOS containing a terminal N-acetylgalactosamine (GalNAc) derived from C. jejuni were

recognized by the human (h) CLR macrophage galactose-type C-type lectin (MGL) (200).

However, in the study presented in chapter 3, only minor binding of the murine (m)MGL-1 to

the tested C. jejuni isolates was detected. One possible explanation for this might be the

different binding profiles of MGL isoforms that are not only described for mMGL-2, that is

more similar to hMGL, and mMGL-1 (201, 202) but also for other CLRs as the human DC-

SIGN and its eight murine orthologs DC-SIGN-related proteins (SIGNRs) (139, 140).

Despite SIGNR6, a pseudogene, and SIGNR4 all other SIGNR receptors are described to

generally recognize glycan ligands similar to DC-SIGN (139). A similar observation was

made for the human- and mouse-derived Dectin-1. Low-valency β-glucan parts of fucan and

laminarin were able to activate hDectin-1 expressing cells, but not cells expressing the

mDectin-1. Furthermore, stimulation studies with curdlan, a high-valency β-glucan, showed

binding to both human and murine Dectin-1. Further assays including reciprocal mutagenesis

analysis and substitution assays revealed that it is not the ligand-binding site of hDectin-1,

but the intracellular domain of this receptor that mediates its sensitivity to low-valency β-

glucan ligands. Interestingly, the substitution with the hDectin-1-specific amino acids in the

intracellular domain rendered mDectin-1 sensitive to low-valency β-glucans (203).

Dectin-1 is a type II transmembrane protein that is mainly expressed by myeloid cells from

the macrophage/monocyte and neutrophil lineages, but also to a lower extent by DCs and T

cell subpopulations (167, 204). Activation of Dectin-1 includes several cellular functions like

pathogen sensing, uptake and clearance. Since the cytoplasmic tail of Dectin-1 contains a

hemITAM-motif (205), engagement of Dectin-1 leads to the activation of effector functions

including the production of chemokines and cytokines, thus shaping immune responses (105,

174, 206). Applying the methods described in chapter 3 paper II and the CLR-hFc fusion

protein library, Dectin-1 was identified as a receptor for C. jejuni (183). Interestingly,

Dectin -1 is classically described as a major receptor that recognizes specifically β-1,3-

Discussion

- 35 -

glucans present in fungi like C. albicans or S. cerevisae (53). Zymosan, a cell wall polymer

of yeast like S. cerevisae, is recognized by TLR2 and Dectin-1. Whereas Dectin-1 induces

phagocytosis and ROS production that is enhanced by TLR2 signaling, TLR2 triggers

inflammatory cytokine production via NFκ-B that is further augmented by Dectin-1 in return

(207). Moreover, also a cooperation of TLR2 and Dectin-1 was described in the context of

mycobacterial species (133, 208). Interestingly, a study with several virulent, attenuated and

avirulent mycobacterial species showed that, in contrast to an anti-fungal immune response,

Dectin-1 was not able to induce phagocytosis in macrophages upon mycobacterial sensing.

However, the release of immune mediators by macrophages such as the cytokines IL-6,

TNFα, RANTES and G-CSF was impaired in a Dectin-1-dependent manner (208). Strikingly,

up to know there is no data suggesting that mycobacterial cell wall incorporates β-glucan.

Furthermore, Dectin-1 is also known to interact with other bacteria like Haemophilus

influenzae (209) or parasites, such as L. infantum and L. amazonensis (210, 211). These

reports clearly demonstrate that Dectin-1 plays a role in recognition of other pathogens than

fungi, but future work has to be performed to gain more insight into the relevance of Dectin-1

regarding recognition of bacteria, signal transduction and immune modulation. A huge

progress will be the identification of distinct Dectin-1 ligand(s) in these pathogens, which

facilitates research on how Dectin-1 affects immune responses towards pathogens beyond the

focus on fungi (212).

Besides the use of Fc-fusion proteins, also reporter cell line assays are commonly applied to

screen CLR/PAMP interactions, for their ligands and to validate CLR agonists or antagonists

(132, 155, 157, 164, 213-216). These reporter cells express distinct CLRs on their surface

coupled to an intracellular promotor-reporter expression cassette, e.g. NFAT-GFP. Here,

ligand binding to the CRD of the CLR induces the activation of the transcription factor

NFAT. Since subsequent activation of the GFP cassette leads to a fluorescence signal, CLR

activation can be determined by measuring the GFP intensity (155, 164, 216). The application

of reporter cell lines was for example used to extend the finding that S. pneumoniae is

recognized by Mincle (158) by the discovery of S. pneumoniae-derived

glucosyldiacylglycerol present in the LTA as Mincle ligand (157). Along that line, the second

project (chapter 4, paper III (164)) contributed to the finding, that two LTA-derived

components of GAS, namely DGDG and MGDG that differ in one glucose residue are

recognized by Mincle. Moreover, the data suggest that the increased susceptibility of GAS

Discussion

- 36 -

infected Mincle-deficient mice is probably due to an impaired ability to release inflammatory

mediators (164).

GAS is an important human pathogen that expresses several adhesion molecules, like LTA,

to mediate binding to host cells. In contrast, multiple host cells are known to play an

important role in anti-GAS immunity by the release of various inflammatory cytokines in

response to GAS infection to control bacterial growth and dissemination (217-219).

Additionally, the production of NO and ROS increases the clearance of GAS in early stages

of infection (220). Nonetheless, bacteria evolved during evolution efficient and complex

evasion mechanisms to escape the host immune response, such as the expression of nucleases

to prevent entrapment by neutrophil extracellular traps or the blockage of antimicrobial

peptides (221, 222). Modification or masking of critical virulence factors is a common

feature of pathogenic organisms to evade the immune system. For instance, it is assumed that

GAS covers TLR2 agonists like LTA and peptidoglycan by the expression of a thick capsule

to ensure immune evasion (223) until phagocytically active immune cells destroy the capsule

(224). Also the fact that Dectin-1 was not able to detect some pathogenic yeasts like

A. fumigatus or C. neoformans although they express β-glucans (225), argues for the

hypothesis that these strains are able to mask their β-glucan to avoid recognition by PRRs

(226). Moreover, phase variation is a common strategy of several C. jejuni strains to rapidly

adapt to environmental changes and avoid recognition by host cells by controlling the

expression of glycosylated LOS (227), capsule (228, 229) and several other bacterial

determinants (230). Strain-specific genetic differences as well as mechanisms like phase-

variation and antigenic variation that even frequently occur in each individual C. jejuni strain

may additionally explain different study outcomes (183, 200, 231). Recognition of LTA-

derived MGDG by Mincle initiated a protective immune response against GAS whereas

sensing of DGDG suppressed this response without further initiation of signal transduction.

Since it is hypothesize that DGDG serves as suppressor for agonistic PRR-mediated signaling

via Mincle, it is tempting to speculate that bacterial strains might vary their ratio of MGDG

and DGDG in the LTA to escape Mincle-mediated anti-bacterial immunity (164). Whereas it

is shown for group B Streptococcus (GBS) that blockage of the glucosyltransferase, the GBS-

DGDG synthesizing enzyme, led to attenuated virulence of the bacteria in vivo (232), there is

no data existing concerning the precise function of GAS-DGDG.

Discussion

- 37 -

The paper III presented in chapter 4 reported that invasive and noninvasive GAS strains

incorporate different DGDG/MGDG ratios in the LTA (164). Interestingly, amongst the

tested GAS strains, those with increased amounts of DGDG belong to the group that caused

invasive infections (5448, NIH34, NIH44), whereas strains with lower pathogenicity (SF370,

K33, Se235) harbored comparably less DGDG. In correlation with this finding, it was shown

that infection of mice with the different GAS strains influenced the clinical disease course,

which might be partially explained by differences in the DGDG/MGDG ratio (164).

Interestingly, infection of mice with the highly invasive strain 5448 (233, 234) that displayed

the most striking difference in DGDG/MGDG ratio neither significantly initiated Mincle-

dependent immune responses (164) nor impaired the disease progression of the infected mice

(unpublished data), although this GAS strain was shown to be recognized by Mincle. Indeed,

differences between in vitro and in vivo data are also well described in the context of other

CLRs. For instance, infection of mice with S. pneumoniae in a model of invasive

pneumococcal disease did not indicate a role for Mincle in anti-pneumococcal immune

response in vivo although binding of Mincle in a Ca2+-dependent manner to S. pneumoniae

was detected (158). The reasons for the discrepancy between in vitro and in vivo data are

manifold. One explanation might involve the accessibility of the ligand since some ligands

like MGDG and DGDG are incorporated into the LTA present in the bacterial membrane that

is covered by the thick peptidoglycan (235) but can also be shed or exposed to the outer space

of several streptococci (232, 236). Since Mincle did not recognise the whole LTA structure, it

is likely that only portions of shed and degraded moieties are recognized. Most convincingly,

the involvement of other PRRs than CLRs in signaling as well as the observation that other

CLRs can compensate for missing signaling pathways is well known in literature. Thus, we

cannot exclude the contribution of other PRRs in GAS recognition and subsequent initiation

of signal transduction. For example, the CLR MCL is described to be co-expressed and -

regulated with Mincle (237), both being upregulated upon MyD88 activation (238).

Furthermore, whole LTA and the peptidoglycan are well known ligands for TLR2 in Gram-

positive bacteria (239) and TLR2 has been shown to upregulate Mincle expression to induce

downstream signaling upon stimulation with Corynebacterial-derived glycans (161). A clear

role for the TLR adaptor protein MyD88 in initiating immune response towards GAS is

described, since the absence of MyD88 caused a significantly decreased production of TNFα,

type I IFNs, IL6 and IL12 by phagocytic immune cells. This impacted neutrophil recruitment

and led to a higher susceptibility of MyD88-deficient mice challenged with GAS (240-243).

Discussion

- 38 -

Additionally, impaired MyD88 expression in human patients affected TLR-mediated ligand

recognition and may result in severe life-threatening predisposition to several pyogenic

bacterial infections caused by e.g. Staphylococcus aureus and S. pneumoniae (244, 245). In

contrast, whereas TLR9 was shown to play a protective role against GAS in vivo (181) the

impact of TLR2 and TLR13 in anti-GAS immunity was not confirmed in a murine infection

study (180, 217). Moreover, the different pathogenicity profiles of bacterial strains may

explain the partially contradictory results. However, also technical features and the

experimental design like the infection model, infection route and dose might influence the

outcome of in vivo studies in general (246). Indeed, applying a model of focal pneumonia in

infection of mice with a non-invasive S. pneumoniae strain revealed a protective role for

Mincle as indicated by increased bacterial recovery in lungs and pro- and antiinflammatory

cytokine production and larger areas of inflamed tissues in Mincle-deficient mice (157).

Since CLR activation impacts both, the phagocytic activity of APCs and the initiated immune

responses, the knowledge about CLR/pathogen interactions and involved ligands was used in

the last decades to design CLR agonists that specifically target CLR-expressing cells (247).

Indeed, activation of CLRs has been shown to efficiently deliver drugs and genes into

specific cells to initiate immune responses towards vaccines or tumors and also to induce

immune tolerance (248). For example, antibody-mediated targeting of the plasmacytoid DC

(pDC)-specific CLR blood DC antigen 2 (BDCA-2) on BDCA-2-humanized mice impaired

CD4+ and Treg responses (249). Major research on glycan-based CLR targeting has been

performed for the CLR DC-SIGN. Due to its tetrameric CRD presentation, DC-SIGN allows

multivalent targeting (101). Modification of antigens, nanoparticles, liposomes or dendrimers

with DC-SIGN specific glycan ligands has already been successfully introduced to target DC-

SIGN and influence T cell responses (250-256). Furthermore, recognition of glycan-

harboring components as LPS, zymosan or TDB by PRRs is described to initiate expression

of cytokines, enhance internalization of antigens and their presentation on MHC molecules to

T cells, hence influencing the T cell fate (257). This modulatory function renders

carbohydrate ligands interesting candidates for vaccine adjuvants and paves the way for the

field of glycan-based vaccines. Vaccination of mice with liposomes harboring TDB as

Mincle ligand mediated strong anti-GAS immunity after intranasal infection which was

caused by a prominent Th17 immune response (258). GAS infection in vivo characteristically

leads to a Th17 response that plays an important role in the protection against GAS (259,

Discussion

- 39 -

260). Since MGDG was shown to induce a Th17 response (164), this novel Mincle ligand

harbors potential as a future adjuvant candidate. It is known that Mincle recognizes

mycobacterial-derived TDM as well as its synthetic analog TDB and plays an essential role in

enhancing protective immune response via FcRγ/Syk/CARD9 axis in immune cells,

emphasizing its potent adjuvant activity (131, 261). Studies using vaccines with TDB/TDM

as adjuvants showed that both glycolipids mediate immunostimulatory effects towards M.

tuberculosis that involves the FcRγ/Syk/CARD9 pathway and the activation of CD4+ T cells

(262, 263). Up to now, TDB and TDM are applied in liposome-based adjuvants in

experimental vaccines against pathogens like M. tuberculosis, Chlamydia and blood stage

malaria (263-266).

To further investigate the nature of different Mincle ligands, studies were performed to

analyse the crystal structures of human and bovine Mincle. Results showed that Mincle

harbors a CRD that contains a central Ca2+ ion to complex the 3- and 4-OH groups of the

glucose in the trehalose head-group. On the other site of this primary binding site, a

secondary binding site complexes the second glucose residue in the trehalose head-group.

Opposite to this binding pocket, a major hydrophobic groove binds to linear or branched

trehalose glycolipids (143-146). Further studies analysed the influence of the ligand structure

in regard to Mincle binding and signal transduction and showed that branched lipid

backbones (267) with long acyl chains (144) are better complexed by Mincle and induced

cytokine release by macrophages more efficiently (268, 269). Recently, different long-chain,

α-branched trehalose mono- and diesters were synthesized to unravel their capability to be

recognized by Mincle and to induce cell signaling. Interestingly, the prepared diesters of the

trehalose glycolipid mediated an increased agonistic activity of hMincle compared to the

adjuvant TDB (270). The knowledge from these structural data can be used in the future to

develop novel Mincle agonists that mediate efficient adjuvant properties.

In general, CLRs are described to be promiscuous receptors that are often able to detect more

than one ligand (147). For example, Mincle recognizes various nonself-derived glycolipids

and glycoproteins but also self-ligands such as sterols or β-glucosylceramides (213, 271,

272). Obviously, Mincle is also able to bind to the same ligand that derived from different

bacterial species, as shown for the glucosyldiacylglycerol of GAS (164) and S. pneumoniae

(157). Since glucosyldiacylglycerol is a part of the LTA which is a common feature of Gram-

Discussion

- 40 -

positive bacteria, further studies will elucidate if this glycolipid is generally recognized by

Mincle in all Gram-positive bacteria. However, there is insufficient knowledge about

differences in ligand preferences within pathogen species as shown for the recognition of β-

glucan harboring fungi by Dectin-1 (225). The mode of the initiated signal transduction by

CLR engagement is dependent on several factors like receptor density and the nature of the

respective ligands. For example, the affinity and avidity of ligand binding impairs signal

quantity and duration mediated by CLRs. But also the ligand properties like soluble or

particulate as well as the ligand size influence the resulting immune response (273). It was

shown for Mincle that the expression of this receptor in unstimulated cells is only weak,

whereas Mincle is upregulated upon stimulation and translocated to the cell surface via

MyD88 (238). Previous studies have shown that the constitutively expressed CLR MCL

enhances the expression of Mincle upon stimulation with TDM (274). In particular, upon

activation, MCL and Mincle form a complex heterodimer through the binding of the

hydrophobic repeat of MCL to the stalk region of Mincle, hence increasing the cell surface

expression of Mincle (237, 238, 275). These data and the finding that MCL and Mincle both

sense TDM (132, 274, 276) pinpoint to a synergistic relationship between these two

receptors. Furthermore, it is hypothesized that Mincle upon a certain threshold fine-tunes the

signaling that is initiated by MCL ligands to regulate host immune response (277).

In conclusion, the presented work highlights the role of CLRs in anti-bacterial immunity.

Methods were described to detect CLRs that recognize bacteria. This further allows for

functional characterization of CLR/bacteria interactions to unravel host/pathogen

interactions, bacterial pathogenesis and host immune responses. The identification of a

specific CLR ligand, as shown for GAS-derived MGDG and DGDG-binding to Mincle, as

part of a collaborative effort enables additional approaches like glycan-based targeting and

the production or the modification of potent vaccines against GAS.

Future outlook

- 41 -

Chapter 6: Future outlook

Although the previous chapters present examples that CLRs like Dectin-1 and Mincle

mediate an important role in anti-bacterial immunity, still intense work needs to be done to

identify the Dectin-1 ligand in C. jejuni, as well as the underlaying immune mechanisms and

signaling consequences of the MGDG and DGDG/Mincle interaction in GAS.

Bacteria like C. jejuni constantly change their surface structures to adapt to new

environments, including evasion of host immune responses. In addition to inter-strain specific

modifications, also changes of surface structures within one specific strain are described.

Studies are needed that reproducibly screen a broader set of C. jejuni strains that differ in

various characteristics like genetics or lifestyle as generalists or specialists (183). Since this

work only focused on the binding of Dectin-1 to C. jejuni, further studies are needed to show

if this recognition also induces cell signaling or if it is dispensable for the initiation of an

immune response as seen for C. neoformans (173) and other examples of CLR/pathogen

interactions (136, 158, 176). Moreover, the distinct ligand for Dectin-1 present in C. jejuni

has to be determined.

Regarding the detection of LTA-derived MGDG and DGDG as Mincle ligands as part of a

scientific collaboration, it would be helpful if also a larger number of GAS strains isolated

from clinical patients in various stages of infection are analysed concerning their

MGDG/DGDG ratio to enable association between virulence and Mincle-ligand expression.

Since LTA is a ubiquitous component of the Gram-positive cell wall (278), it is interesting to

investigate if Mincle also mediates recognition of other Gram-positive bacteria via LTA-

derived MGDG. The strain 5448 was the only GAS strain investigated with a minor role for

Mincle in vivo despite it was recognized by this receptor (164). The GAS strain 5448 was

isolated from a patient with severe, invasive necrotizing fasciitis (234). Since the infection

and inflammation of the skin is known to cause lower oxygen content as under normal

condition, so-called hypoxia, one should consider the oxygen concentration to obtain

physiologically relevant results. Indeed, publications report that hypoxia-inducible factor-1

(HIF-1), the main transcription factor that regulates the adaptive immune response to hypoxia

has an impact on both, the susceptibility to GAS and the PRR signaling pathways. On the one

hand, GAS 5448 induces HIF-1α (279). A murine infection study showed that HIF-1-

Future outlook

- 42 -

deficient mice are more susceptible to an invasive GAS 5448 infection, probably due to a

decreased phagocytic killing of the bacteria (279). On the other hand, crosstalk of HIF with

PRRs like TLR2 and TLR6 (280) is described in the literature as well as the HIF-induced

upregulation of TLR4 expression in macrophages (281). Interestingly, in response to

activation via TDB, Mincle signaling increased the transcription of the hif1α gene but was not

required for the expression of HIF-1α (282). Furthermore, key regulator molecules like the

adaptor molecule CARD9 and the transcription factor NFκ-B that play an important role in

signaling via Mincle and other CLRs were found to be involved in hypoxia-sensitive

pathways (283-285). Further studies should unravel the role of Mincle in GAS infection

under hypoxic conditions.

Concluding remarks

- 43 -

Chapter 7: Concluding remarks

The presented projects highlight the relevance of CLRs in sensing bacterial pathogens like

C. jejuni or GAS and contributed to unravelling that GAS-derived MGDG and DGDG serve

as novel ligands for Mincle.

CLR-Fc fusion proteins present powerful tools to identify yet unknown CLR/bacteria

interactions. Parts of this thesis describe ELISA-, flow cytometry- and confocal microscopy-

based methods that enable high-throughput screening and subsequent confirmation of CLR

candidates as innate bacterial sensors based on a set of human and murine CLR-Fc fusion

proteins. With the help of these methods, Dectin-1 was detected as a CLR that recognises

different C. jejuni isolates.

Additionally, this thesis contributed to the finding that the LTA-derived glycolipids MGDG

and DGDG from GAS act as novel ligands for Mincle. Whereas MGDG induced activation

of innate immune responses against GAS, DGDG interfered negatively with this response.

Moreover, in vivo data showed that Mincle mediated a protective role in anti-GAS immunity.

The identification of distinct bacterial CLR ligands is important for future vaccination

strategies to induce potent immune responses. Moreover, the analysis of initiated immune

responses by receptor/ligand interactions offers the possibility to therapeutically interfere

with the activated signaling pathway and thus shape immune responses.

References

- 44 -

References

1. Hill DA & Artis D (2010) Intestinal Bacteria and the Regulation of Immune Cell Homeostasis. Annual

Review of Immunology 28(1):623-667.

2. Tao X & Xu A (2016) Chapter 2 Basic Knowledge of Immunology. Amphioxus Immunity), pp 15-42.

3. Marshall JS, Warrington R, Watson W, & Kim HL (2018) An introduction to immunology and

immunopathology. Allergy, Asthma, and Clinical Immunology 14(Suppl 2):49.

4. Turvey SE & Broide DH (2010) Innate immunity. The Journal of allergy and clinical immunology

125(2 Suppl 2):S24-S32.

5. Janeway CA, Jr. (1989) Approaching the asymptote? Evolution and revolution in immunology. Cold

Spring Harbor Symposia on Quantitative Biology 54 Pt 1:1-13.

6. Bonilla FA & Oettgen HC (2010) Adaptive immunity. Journal of Allergy and Clinical Immunology

125(2 Suppl 2):S33-40.

7. Cota AM & Midwinter MJ (2015) The immune system. Anaesthesia & Intensive Care Medicine

16(7):353-355.

8. Ivanov AI, Parkos CA, & Nusrat A (2010) Cytoskeletal regulation of epithelial barrier function during

inflammation. The American journal of pathology 177(2):512-524.

9. Gallo RL & Hooper LV (2012) Epithelial antimicrobial defence of the skin and intestine. Nature

reviews. Immunology 12(7):503-516.

10. Murphy KP, et al. (2008) Janeway´s Immunobiology (New York: Garland Science) 7th Ed.

11. Onyiah JC & Colgan SP (2016) Cytokine responses and epithelial function in the intestinal mucosa.

Cellular and molecular life sciences : CMLS 73(22):4203-4212.

12. Beutler B (2004) Innate immunity: an overview. Molecular Immunology 40(12):845-859.

13. Aderem A & Underhill DM (1999) Mechanisms of phagocytosis in macrophages. Annual Review of

Immunology 17:593-623.

14. Regnault A, et al. (1999) Fcγ receptor-mediated induction of dendritic cell maturation and major

histocompatibility complex class I-restricted antigen presentation after immune complex

internalization. Journal of Experimental Medicine 189(2):371-380.

15. Maurer D, et al. (1998) Fc epsilon receptor I on dendritic cells delivers IgE-bound multivalent antigens

into a cathepsin S-dependent pathway of MHC class II presentation. Journal of Immunology

161(6):2731-2739.

16. Guermonprez P, Valladeau J, Zitvogel L, Thery C, & Amigorena S (2002) Antigen presentation and T

cell stimulation by dendritic cells. Annual Review of Immunology 20:621-667.

17. Bell D, Young JW, & Banchereau J (1999) Dendritic cells. Advances in Immunology 72:255-324.

18. Rescigno M, et al. (2001) Dendritic cells express tight junction proteins and penetrate gut epithelial

monolayers to sample bacteria. Nature Immunology 2(4):361-367.

19. Mellman I & Steinman RM (2001) Dendritic cells: specialized and regulated antigen processing

machines. Cell 106(3):255-258.

References

- 45 -

20. Cruz FM, Colbert JD, Merino E, Kriegsman BA, & Rock KL (2017) The Biology and Underlying

Mechanisms of Cross-Presentation of Exogenous Antigens on MHC-I Molecules. Annual Review of

Immunology 35:149-176.

21. Hoffmann JA, Kafatos FC, Janeway CA, & Ezekowitz RA (1999) Phylogenetic perspectives in innate

immunity. Science 284(5418):1313-1318.

22. Jenkins MK, Taylor PS, Norton SD, & Urdahl KB (1991) CD28 delivers a costimulatory signal

involved in antigen-specific IL-2 production by human T cells. The Journal of Immunology

147(8):2461-2466.

23. Ahmed R & Gray D (1996) Immunological memory and protective immunity: understanding their

relation. Science 272(5258):54-60.

24. Reinhardt RL, Kang SJ, Liang HE, & Locksley RM (2006) T helper cell effector fates--who, how and

where? Current Opinion in Immunology 18(3):271-277.

25. Schmitt E, Klein M, & Bopp T (2014) Th9 cells, new players in adaptive immunity. Trends in

Immunology 35(2):61-68.

26. Lu Y, et al. (2018) Th9 Cells Represent a Unique Subset of CD4+ T Cells Endowed with the Ability to

Eradicate Advanced Tumors. Cancer Cell 33(6):1048-1060.e1047.

27. Chatila TA (2005) Role of regulatory T cells in human diseases. Journal of Allergy and Clinical

Immunology 116(5):949-959.

28. McHeyzer-Williams M, Okitsu S, Wang N, & McHeyzer-Williams L (2011) Molecular programming

of B cell memory. Nature reviews Immunology 12(1):24-34.

29. Liu AH (2008) Innate microbial sensors and their relevance to allergy. Journal of Allergy and Clinical

Immunology 122(5):846-858.

30. Medzhitov R (2009) Approaching the asymptote: 20 years later. Immunity 30(6):766-775.

31. Frevert CW, Felgenhauer J, Wygrecka M, Nastase MV, & Schaefer L (2018) Danger-Associated

Molecular Patterns Derived From the Extracellular Matrix Provide Temporal Control of Innate

Immunity. Journal of Histochemistry and Cytochemistry 66(4):213-227.

32. Srinivasan N, et al. (2016) Actin is an evolutionarily-conserved damage-associated molecular pattern

that signals tissue injury in Drosophila melanogaster. eLife 5:e19662.

33. Smiley ST, King JA, & Hancock WW (2001) Fibrinogen stimulates macrophage chemokine secretion

through toll-like receptor 4. Journal of Immunology 167(5):2887-2894.

34. Schaefer L (2014) Complexity of danger: the diverse nature of damage-associated molecular patterns.

Journal of Biological Chemistry 289(51):35237-35245.

35. Zhang Q, et al. (2010) Circulating mitochondrial DAMPs cause inflammatory responses to injury.

Nature 464(7285):104-107.

36. Krysko DV, et al. (2011) Emerging role of damage-associated molecular patterns derived from

mitochondria in inflammation. Trends in Immunology 32(4):157-164.

37. Fader CM, Aguilera MO, & Colombo MI (2012) ATP is released from autophagic vesicles to the

extracellular space in a VAMP7-dependent manner. Autophagy 8(12):1741-1756.

38. Dupont N, et al. (2011) Autophagy-based unconventional secretory pathway for extracellular delivery

of IL-1beta. EMBO Journal 30(23):4701-4711.

References

- 46 -

39. Tran TT, Groben P, & Pisetsky DS (2008) The release of DNA into the plasma of mice following

hepatic cell death by apoptosis and necrosis. Biomarkers: Biochemical Indicators of Exposure,

Response, and Susceptibility to Chemicals 13(2):184-200.

40. Allam R, et al. (2012) Histones from dying renal cells aggravate kidney injury via TLR2 and TLR4.

Journal of the American Society of Nephrology 23(8):1375-1388.

41. Liu-Bryan R, Scott P, Sydlaske A, Rose DM, & Terkeltaub R (2005) Innate immunity conferred by

Toll-like receptors 2 and 4 and myeloid differentiation factor 88 expression is pivotal to monosodium

urate monohydrate crystal-induced inflammation. Arthritis and Rheumatism 52(9):2936-2946.

42. Burdette DL & Vance RE (2013) STING and the innate immune response to nucleic acids in the

cytosol. Nature Immunology 14(1):19-26.

43. Brubaker SW, Bonham KS, Zanoni I, & Kagan JC (2015) Innate immune pattern recognition: a cell

biological perspective. Annual Review of Immunology 33:257-290.

44. Iwasaki A & Medzhitov R (2015) Control of adaptive immunity by the innate immune system. Nature

Immunology 16(4):343-353.

45. Broz P, Ohlson MB, & Monack DM (2012) Innate immune response to Salmonella typhimurium, a

model enteric pathogen. Gut Microbes 3(2):62-70.

46. Lafyatis R & Marshak-Rothstein A (2007) Toll-like receptors and innate immune responses in systemic

lupus erythematosus. Arthritis Research & Therapy 9(6):222.

47. Mouchess ML, et al. (2011) Transmembrane mutations in Toll-like receptor 9 bypass the requirement

for ectodomain proteolysis and induce fatal inflammation. Immunity 35(5):721-732.

48. Iwasaki A & Medzhitov R (2004) Toll-like receptor control of the adaptive immune responses. Nature

Immunology 5(10):987-995.

49. Barton GM, Kagan JC, & Medzhitov R (2006) Intracellular localization of Toll-like receptor 9 prevents

recognition of self DNA but facilitates access to viral DNA. Nature Immunology 7(1):49-56.

50. Akira S, Uematsu S, & Takeuchi O (2006) Pathogen Recognition and Innate Immunity. Cell

124(4):783-801.

51. Deretic V, Saitoh T, & Akira S (2013) Autophagy in infection, inflammation and immunity. Nature

Reviews: Immunology 13(10):722-737.

52. Lamkanfi M & Dixit Vishva M (2014) Mechanisms and Functions of Inflammasomes. Cell

157(5):1013-1022.

53. Drummond RA & Brown GD (2011) The role of Dectin-1 in the host defence against fungal infections.

Current Opinion in Microbiology 14(4):392-399.

54. Kumagai Y & Akira S (2010) Identification and functions of pattern-recognition receptors. Journal of

Allergy and Clinical Immunology 125(5):985-992.

55. Thomas CJ & Schroder K (2013) Pattern recognition receptor function in neutrophils. Trends in

Immunology 34(7):317-328.

56. Medzhitov R, Preston-Hurlburt P, & Janeway Jr CA (1997) A human homologue of the Drosophila

Toll protein signals activation of adaptive immunity. Nature 388(6640):394–397.

References

- 47 -

57. Lemaitre B, Nicolas E, Michaut L, Reichhart JM, & Hoffmann JA (1996) The dorsoventral regulatory

gene cassette spätzle/Toll/cactus controls the potent antifungal response in Drosophila adults. Cell

86(6):973-983.

58. Rock FL, Hardiman G, Timans JC, Kastelein RA, & Bazan JF (1998) A family of human receptors

structurally related to Drosophila Toll. Proceedings of the National Academy of Sciences of the United

States of America 95(2):588-593.

59. Albiger B, Dahlberg S, Henriques-Normark B, & Normark S (2007) Role of the innate immune system

in host defence against bacterial infections: focus on the Toll-like receptors. Journal of Internal

Medicine 261(6):511-528.

60. Seki E, et al. (2002) Critical roles of myeloid differentiation factor 88-dependent proinflammatory

cytokine release in early phase clearance of Listeria monocytogenes in mice. Journal of Immunology

169(7):3863-3868.

61. Bellocchio S, et al. (2004) The contribution of the Toll-like/IL-1 receptor superfamily to innate and

adaptive immunity to fungal pathogens in vivo. Journal of Immunology 172(5):3059-3069.

62. Hitziger N, Dellacasa I, Albiger B, & Barragan A (2005) Dissemination of Toxoplasma gondii to

immunoprivileged organs and role of Toll/interleukin-1 receptor signalling for host resistance assessed

by in vivo bioluminescence imaging. Cellular Microbiology 7(6):837-848.

63. Adachi K, et al. (2001) Plasmodium berghei infection in mice induces liver injury by an IL-12- and

toll-like receptor/myeloid differentiation factor 88-dependent mechanism. Journal of Immunology

167(10):5928-5934.

64. Abe T, et al. (2007) Hepatitis C virus nonstructural protein 5A modulates the toll-like receptor-

MyD88-dependent signaling pathway in macrophage cell lines. Journal of Virology 81(17):8953-8966.

65. Mogensen TH (2009) Pathogen Recognition and Inflammatory Signaling in Innate Immune Defenses.

Clinical Microbiology Reviews 22(2):240-273.

66. Takeuchi O & Akira S (2010) Pattern Recognition Receptors and Inflammation. Cell 140(6):805-820.

67. Hoshino K, et al. (1999) Cutting edge: Toll-like receptor 4 (TLR4)-deficient mice are hyporesponsive

to lipopolysaccharide: evidence for TLR4 as the LPS gene product. Journal of Immunology

162(7):3749-3752.

68. Park BS & Lee J-O (2013) Recognition of lipopolysaccharide pattern by TLR4 complexes.

Experimental and Molecular Medicine 45(12):e66-e66.

69. Miyake K (2006) Roles for accessory molecules in microbial recognition by Toll-like receptors.

Journal of Endotoxin Research 12(4):195-204.

70. Park BS, et al. (2009) The structural basis of lipopolysaccharide recognition by the TLR4-MD-2

complex. Nature 458(7242):1191-1195.

71. Tapping RI & Tobias PS (2003) Mycobacterial lipoarabinomannan mediates physical interactions

between TLR1 and TLR2 to induce signaling. Journal of Endotoxin Research 9(4):264-268.

72. Hayashi F, et al. (2001) The innate immune response to bacterial flagellin is mediated by Toll-like

receptor 5. Nature 410(6832):1099-1103.

73. Han SH, Kim JH, Martin M, Michalek SM, & Nahm MH (2003) Pneumococcal lipoteichoic acid

(LTA) is not as potent as staphylococcal LTA in stimulating Toll-like receptor 2. Infection and

Immunity 71(10):5541-5548.

References

- 48 -

74. Kim JH, et al. (2005) Monoacyl lipoteichoic acid from pneumococci stimulates human cells but not

mouse cells. Infection and Immunity 73(2):834-840.

75. Schroder NW, et al. (2003) Lipoteichoic acid (LTA) of Streptococcus pneumoniae and Staphylococcus

aureus activates immune cells via Toll-like receptor (TLR)-2, lipopolysaccharide-binding protein

(LBP), and CD14, whereas TLR-4 and MD-2 are not involved. Journal of Biological Chemistry

278(18):15587-15594.

76. Henneke P, et al. (2005) Role of lipoteichoic acid in the phagocyte response to group B streptococcus.

Journal of Immunology 174(10):6449-6455.

77. Bauer S, et al. (2001) Human TLR9 confers responsiveness to bacterial DNA via species-specific CpG

motif recognition. Proceedings of the National Academy of Sciences of the United States of America

98(16):9237-9242.

78. Kawai T & Akira S (2009) The roles of TLRs, RLRs and NLRs in pathogen recognition. International

Immunology 21(4):317-337.

79. Inohara, Chamaillard, McDonald C, & Nunez G (2005) NOD-LRR proteins: role in host-microbial

interactions and inflammatory disease. Annual Review of Biochemistry 74:355-383.

80. Ye Z & Ting JP (2008) NLR, the nucleotide-binding domain leucine-rich repeat containing gene

family. Current Opinion in Immunology 20(1):3-9.

81. Kim YK, Shin JS, & Nahm MH (2016) NOD-Like Receptors in Infection, Immunity, and Diseases.

Yonsei Medical Journal 57(1):5-14.

82. Martinon F, Mayor A, & Tschopp J (2009) The inflammasomes: guardians of the body. Annual Review

of Immunology 27:229-265.

83. Girardin SE, et al. (2003) Nod1 detects a unique muropeptide from gram-negative bacterial

peptidoglycan. Science 300(5625):1584-1587.

84. Girardin SE, et al. (2003) Nod2 is a general sensor of peptidoglycan through muramyl dipeptide

(MDP) detection. Journal of Biological Chemistry 278(11):8869-8872.

85. Steimle V, Otten LA, Zufferey M, & Mach B (1993) Complementation cloning of an MHC class II

transactivator mutated in hereditary MHC class II deficiency (or bare lymphocyte syndrome). Cell

75(1):135-146.

86. Biswas A, Meissner TB, Kawai T, & Kobayashi KS (2012) Cutting edge: impaired MHC class I

expression in mice deficient for Nlrc5/class I transactivator. Journal of Immunology 189(2):516-520.

87. Kobayashi KS & van den Elsen PJ (2012) NLRC5: a key regulator of MHC class I-dependent immune

responses. Nature Reviews: Immunology 12(12):813-820.

88. Travassos LH, et al. (2010) Nod1 and Nod2 direct autophagy by recruiting ATG16L1 to the plasma

membrane at the site of bacterial entry. Nature Immunology 11(1):55-62.

89. Strähle L, et al. (2007) Activation of the β interferon promoter by unnatural Sendai virus infection

requires RIG-I and is inhibited by viral C proteins. Journal of Virology 81(22):12227-12237.

90. Kato H, et al. (2006) Differential roles of MDA5 and RIG-I helicases in the recognition of RNA

viruses. Nature 441(7089):101-105.

91. Takahasi K, et al. (2008) Nonself RNA-sensing mechanism of RIG-I helicase and activation of

antiviral immune responses. Molecular Cell 29(4):428-440.

References

- 49 -

92. Kawai T, et al. (2005) IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon

induction. Nature Immunology 6(10):981-988.

93. Kawai T & Akira S (2006) Innate immune recognition of viral infection. Nature Immunology 7(2):131-

137.

94. Geijtenbeek TB & Gringhuis SI (2009) Signalling through C-type lectin receptors: shaping immune

responses. Nature Reviews: Immunology 9(7):465-479.

95. Cambi A, Koopman M, & Figdor CG (2005) How C-type lectins detect pathogens. Cellular

Microbiology 7(4):481-488.

96. Drickamer K (1999) C-type lectin-like domains. Current Opinion in Structural Biology 9(5):585-590.

97. Kogelberg H & Feizi T (2001) New structural insights into lectin-type proteins of the immune system.

Current Opinion in Structural Biology 11(5):635-643.

98. Zelensky AN & Gready JE (2005) The C-type lectin-like domain superfamily. Febs j 272(24):6179-

6217.

99. Drickamer K & Taylor ME (2015) Recent insights into structures and functions of C-type lectins in the

immune system. Current Opinion in Structural Biology 34:26-34.

100. Figdor CG, van Kooyk Y, & Adema GJ (2002) C-type lectin receptors on dendritic cells and

Langerhans cells. Nature Reviews: Immunology 2(2):77-84.

101. Mayer S, Raulf M-K, & Lepenies B (2017) C-type lectins: their network and roles in pathogen

recognition and immunity. Histochemistry and Cell Biology 147(2):223-237.

102. Kerrigan AM & Brown GD (2011) Syk-coupled C-type lectins in immunity. Trends in immunology

32(4):151-156.

103. Osorio F & Reis e Sousa C (2011) Myeloid C-type lectin receptors in pathogen recognition and host

defense. Immunity 34(5):651-664.

104. Underhill DM & Goodridge HS (2007) The many faces of ITAMs. Trends in Immunology 28(2):66-73.

105. Rogers NC, et al. (2005) Syk-dependent cytokine induction by Dectin-1 reveals a novel pattern

recognition pathway for C type lectins. Immunity 22(4):507-517.

106. Suzuki-Inoue K, et al. (2006) A novel Syk-dependent mechanism of platelet activation by the C-type

lectin receptor CLEC-2. Blood 107(2):542-549.

107. Slack EC, et al. (2007) Syk-dependent ERK activation regulates IL-2 and IL-10 production by DC

stimulated with zymosan. European Journal of Immunology 37(6):1600-1612.

108. Goodridge HS, Simmons RM, & Underhill DM (2007) Dectin-1 stimulation by Candida albicans yeast

or zymosan triggers NFAT activation in macrophages and dendritic cells. Journal of Immunology

178(5):3107-3115.

109. Gross O, et al. (2009) Syk kinase signalling couples to the Nlrp3 inflammasome for anti-fungal host

defence. Nature 459(7245):433-436.

110. Kankkunen P, et al. (2010) (1,3)-β-glucans activate both dectin-1 and NLRP3 inflammasome in human

macrophages. Journal of Immunology 184(11):6335-6342.

References

- 50 -

111. Hise AG, et al. (2009) An essential role for the NLRP3 inflammasome in host defense against the

human fungal pathogen Candida albicans. Cell Host Microbe 5(5):487-497.

112. Said-Sadier N, Padilla E, Langsley G, & Ojcius DM (2010) Aspergillus fumigatus stimulates the

NLRP3 inflammasome through a pathway requiring ROS production and the Syk tyrosine kinase. PloS

One 5(4):e10008.

113. Gross O, et al. (2006) Card9 controls a non-TLR signalling pathway for innate anti-fungal immunity.

Nature 442(7103):651-656.

114. Mócsai A, Ruland J, & Tybulewicz VL (2010) The SYK tyrosine kinase: a crucial player in diverse

biological functions. Nature Reviews: Immunology 10(6):387-402.

115. Barrow AD & Trowsdale J (2006) You say ITAM and I say ITIM, let's call the whole thing off: the

ambiguity of immunoreceptor signalling. European Journal of Immunology 36(7):1646-1653.

116. Gringhuis SI, et al. (2009) Dectin-1 directs T helper cell differentiation by controlling noncanonical

NF-κB activation through Raf-1 and Syk. Nature Immunology 10(2):203-213.

117. Del Fresno C, Iborra S, Saz-Leal P, Martinez-Lopez M, & Sancho D (2018) Flexible Signaling of

Myeloid C-Type Lectin Receptors in Immunity and Inflammation. Frontiers in Immunology 9:804.

118. Hovius JW, et al. (2008) Salp15 binding to DC-SIGN inhibits cytokine expression by impairing both

nucleosome remodeling and mRNA stabilization. PLoS Pathogens 4(2):e31.

119. Rodriguez E, et al. (2017) Fasciola hepatica glycoconjugates immuneregulate dendritic cells through

the Dendritic Cell-Specific Intercellular adhesion molecule-3-Grabbing Non-integrin inducing T cell

anergy. Scientific Reports 7:46748.

120. Watson AA, et al. (2009) The platelet receptor CLEC-2 is active as a dimer. Biochemistry

48(46):10988-10996.

121. Hughes CE, et al. (2010) CLEC-2 activates Syk through dimerization. Blood 115(14):2947-2955.

122. Garcia-Vallejo JJ & van Kooyk Y (2013) The physiological role of DC-SIGN: A tale of mice and men.

Trends in Immunology 34(10):482-486.

123. Lu X, Nagata M, & Yamasaki S (2018) Mincle: 20 years of a versatile sensor of insults. International

Immunology 30(6):233-239.

124. Poole J, Day CJ, von Itzstein M, Paton JC, & Jennings MP (2018) Glycointeractions in bacterial

pathogenesis. Nature Reviews Microbiology 16(7):440-452.

125. Frick IM, Schmidtchen A, & Sjobring U (2003) Interactions between M proteins of Streptococcus

pyogenes and glycosaminoglycans promote bacterial adhesion to host cells. European Journal of

Biochemistry 270(10):2303-2311.

126. Sokurenko EV, et al. (1998) Pathogenic adaptation of Escherichia coli by natural variation of the FimH

adhesin. Proceedings of the National Academy of Sciences of the United States of America

95(15):8922-8926.

127. De Oliveira DM, et al. (2017) Blood Group Antigen Recognition via the Group A Streptococcal M

Protein Mediates Host Colonization. MBio 8(1).

128. Nobbs AH, Lamont RJ, & Jenkinson HF (2009) Streptococcus adherence and colonization.

Microbiology and Molecular Biology Reviews 73(3):407-450, Table of Contents.

References

- 51 -

129. Schlesinger LS, Hull SR, & Kaufman TM (1994) Binding of the terminal mannosyl units of

lipoarabinomannan from a virulent strain of Mycobacterium tuberculosis to human macrophages.

Journal of Immunology 152(8):4070-4079.

130. Tanne A, et al. (2009) A murine DC-SIGN homologue contributes to early host defense against

Mycobacterium tuberculosis. Journal of Experimental Medicine 206(10):2205-2220.

131. Schoenen H, et al. (2010) Cutting edge: Mincle is essential for recognition and adjuvanticity of the

mycobacterial cord factor and its synthetic analog trehalose-dibehenate. Journal of Immunology

184(6):2756-2760.

132. Ishikawa E, et al. (2009) Direct recognition of the mycobacterial glycolipid, trehalose dimycolate, by

C-type lectin Mincle. Journal of Experimental Medicine 206(13):2879-2888.

133. Rothfuchs AG, et al. (2007) Dectin-1 interaction with Mycobacterium tuberculosis leads to enhanced

IL-12p40 production by splenic dendritic cells. Journal of Immunology 179(6):3463-3471.

134. Hoving JC, Wilson GJ, & Brown GD (2014) Signalling C-type lectin receptors, microbial recognition

and immunity. Cellular Microbiology 16(2):185-194.

135. Court N, et al. (2010) Partial redundancy of the pattern recognition receptors, scavenger receptors, and

C-type lectins for the long-term control of Mycobacterium tuberculosis infection. Journal of

Immunology 184(12):7057-7070.

136. Heitmann L, Schoenen H, Ehlers S, Lang R, & Holscher C (2013) Mincle is not essential for

controlling Mycobacterium tuberculosis infection. Immunobiology 218(4):506-516.

137. Kang PB, et al. (2005) The human macrophage mannose receptor directs Mycobacterium tuberculosis

lipoarabinomannan-mediated phagosome biogenesis. Journal of Experimental Medicine 202(7):987-

999.

138. Geijtenbeek TB, et al. (2003) Mycobacteria target DC-SIGN to suppress dendritic cell function.

Journal of Experimental Medicine 197(1):7-17.

139. Powlesland AS, et al. (2006) Widely divergent biochemical properties of the complete set of mouse

DC-SIGN-related proteins. Journal of Biological Chemistry 281(29):20440-20449.

140. Park CG, et al. (2001) Five mouse homologues of the human dendritic cell C-type lectin, DC-SIGN.

International Immunology 13(10):1283-1290.

141. Konstantinov SR, et al. (2008) S layer protein A of Lactobacillus acidophilus NCFM regulates

immature dendritic cell and T cell functions. Proceedings of the National Academy of Sciences of the

United States of America 105(49):19474-19479.

142. Dorhoi A, et al. (2010) The adaptor molecule CARD9 is essential for tuberculosis control. Journal of

Experimental Medicine 207(4):777-792.

143. Feinberg H, et al. (2013) Mechanism for recognition of an unusual mycobacterial glycolipid by the

macrophage receptor mincle. Journal of Biological Chemistry 288(40):28457-28465.

144. Furukawa A, et al. (2013) Structural analysis for glycolipid recognition by the C-type lectins Mincle

and MCL. Proceedings of the National Academy of Sciences of the United States of America

110(43):17438-17443.

145. Feinberg H, et al. (2016) Binding Sites for Acylated Trehalose Analogs of Glycolipid Ligands on an

Extended Carbohydrate Recognition Domain of the Macrophage Receptor Mincle. Journal of

Biological Chemistry 291(40):21222-21233.

References

- 52 -

146. Jegouzo SA, et al. (2014) Defining the conformation of human mincle that interacts with mycobacterial

trehalose dimycolate. Glycobiology 24(12):1291-1300.

147. Williams SJ (2017) Sensing Lipids with Mincle: Structure and Function. Frontiers in Immunology

8:1662-1662.

148. Richardson MB & Williams SJ (2014) MCL and Mincle: C-Type Lectin Receptors That Sense

Damaged Self and Pathogen-Associated Molecular Patterns. Frontiers in Immunology 5:288.

149. Behler F, et al. (2015) Macrophage-inducible C-type lectin Mincle-expressing dendritic cells contribute

to control of splenic Mycobacterium bovis BCG infection in mice. Infection and Immunity 83(1):184-

196.

150. Lee WB, et al. (2012) Neutrophils Promote Mycobacterial Trehalose Dimycolate-Induced Lung

Inflammation via the Mincle Pathway. PLoS Pathogens 8(4):e1002614.

151. Flornes LM, et al. (2004) Identification of lectin-like receptors expressed by antigen presenting cells

and neutrophils and their mapping to a novel gene complex. Immunogenetics 56(7):506-517.

152. Matsumoto M, et al. (1999) A novel LPS-inducible C-type lectin is a transcriptional target of NF-IL6

in macrophages. Journal of Immunology 163(9):5039-5048.

153. Kawata K, et al. (2012) Mincle and human B cell function. Journal of Autoimmunity 39(4):315-322.

154. Kingeter LM & Lin X (2012) C-type lectin receptor-induced NF-κB activation in innate immune and

inflammatory responses. Cellular & Molecular Immunology 9(2):105-112.

155. Yamasaki S, et al. (2008) Mincle is an ITAM-coupled activating receptor that senses damaged cells.

Nature Immunology 9(10):1179-1188.

156. Ostrop J & Lang R (2017) Contact, Collaboration, and Conflict: Signal Integration of Syk-Coupled C-

Type Lectin Receptors. Journal of Immunology 198(4):1403-1414.

157. Behler-Janbeck F, et al. (2016) C-type Lectin Mincle Recognizes Glucosyl-diacylglycerol of

Streptococcus pneumoniae and Plays a Protective Role in Pneumococcal Pneumonia. PLoS Pathogens

12(12):e1006038.

158. Rabes A, et al. (2015) The C-Type Lectin Receptor Mincle Binds to Streptococcus pneumoniae but

Plays a Limited Role in the Anti-Pneumococcal Innate Immune Response. PloS One 10(2):e0117022.

159. Sharma A, Steichen AL, Jondle CN, Mishra BB, & Sharma J (2014) Protective role of Mincle in

bacterial pneumonia by regulation of neutrophil mediated phagocytosis and extracellular trap

formation. Journal of Infectious Diseases 209(11):1837-1846.

160. Devi S, Rajakumara E, & Ahmed N (2015) Induction of Mincle by Helicobacter pylori and consequent

anti-inflammatory signaling denote a bacterial survival strategy. Scientific Reports 5:15049.

161. Schick J, et al. (2017) Toll-Like Receptor 2 and Mincle Cooperatively Sense Corynebacterial Cell Wall

Glycolipids. Infection and Immunity 85(7).

162. Shah S, Nagata M, Yamasaki S, & Williams SJ (2016) Total synthesis of a cyclopropane-fatty acid α-

glucosyl diglyceride from Lactobacillus plantarum and identification of its ability to signal through

Mincle. Chemical Communications (Cambridge, England) 52(72):10902-10905.

163. Sauvageau J, et al. (2012) Isolation and structural characterisation of the major glycolipids from

Lactobacillus plantarum. Carbohydrate Research 357:151-156.

References

- 53 -

164. Imai T, et al. (2018) Lipoteichoic acid anchor triggers Mincle to drive protective immunity against

invasive group A Streptococcus infection. Proceedings of the National Academy of Sciences

115(45):E10662-E10671.

165. Nagata M, et al. (2017) Intracellular metabolite β-glucosylceramide is an endogenous Mincle ligand

possessing immunostimulatory activity. Proceedings of the National Academy of Sciences of the United

States of America 114(16):E3285-e3294.

166. Taylor PR, et al. (2007) Dectin-1 is required for β-glucan recognition and control of fungal infection.

Nature Immunology 8(1):31-38.

167. Brown GD, et al. (2002) Dectin-1 is a major beta-glucan receptor on macrophages. Journal of

Experimental Medicine 196(3):407-412.

168. Saijo S, et al. (2007) Dectin-1 is required for host defense against Pneumocystis carinii but not against

Candida albicans. Nature Immunology 8(1):39-46.

169. Gow NAR, et al. (2007) Immune recognition of Candida albicans β-glucan by dectin-1. The Journal of

infectious diseases 196(10):1565-1571.

170. Steele C, et al. (2005) The β-Glucan Receptor Dectin-1 Recognizes Specific Morphologies of

Aspergillus fumigatus. PLoS Pathogens 1(4):e42.

171. Gersuk GM, Underhill DM, Zhu L, & Marr KA (2006) Dectin-1 and TLRs permit macrophages to

distinguish between different Aspergillus fumigatus cellular states. Journal of Immunology

176(6):3717-3724.

172. Hohl TM, et al. (2005) Aspergillus fumigatus triggers inflammatory responses by stage-specific β-

glucan display. PLoS Pathogens 1(3):e30.

173. Nakamura K, et al. (2007) Dectin-1 is not required for the host defense to Cryptococcus neoformans.

Microbiology and Immunology 51(11):1115-1119.

174. Brown GD (2006) Dectin-1: a signalling non-TLR pattern-recognition receptor. Nature Reviews:

Immunology 6(1):33-43.

175. Plato A, Willment JA, & Brown GD (2013) C-type lectin-like receptors of the dectin-1 cluster: ligands

and signaling pathways. International Reviews of Immunology 32(2):134-156.

176. Marakalala MJ, et al. (2011) The Syk/CARD9-coupled receptor Dectin-1 is not required for host

resistance to Mycobacterium tuberculosis in mice. Microbes and Infection 13(2):198-201.

177. Shin DM, et al. (2008) Mycobacterium abscessus activates the macrophage innate immune response

via a physical and functional interaction between TLR2 and dectin-1. Cellular Microbiology

10(8):1608-1621.

178. Maglinao M, et al. (2014) A platform to screen for C-type lectin receptor-binding carbohydrates and

their potential for cell-specific targeting and immune modulation. Journal of Controlled Release

175:36-42.

179. Walker MJ, et al. (2014) Disease manifestations and pathogenic mechanisms of Group A

Streptococcus. Clinical Microbiology Reviews 27(2):264-301.

180. Fieber C, et al. (2015) Innate Immune Response to Streptococcus pyogenes Depends on the Combined

Activation of TLR13 and TLR2. PloS One 10(3):e0119727.

181. Zinkernagel AS, et al. (2012) Importance of Toll-like receptor 9 in host defense against M1T1 group A

Streptococcus infections. Journal of Innate Immunity 4(2):213-218.

References

- 54 -

182. Flannery A, Gerlach JQ, Joshi L, & Kilcoyne M (2015) Assessing Bacterial Interactions Using

Carbohydrate-Based Microarrays. Microarrays (Basel, Switzerland) 4(4):690-713.

183. Mayer S, et al. (2018) C-Type Lectin Receptor (CLR)–Fc Fusion Proteins As Tools to Screen for

Novel CLR/Bacteria Interactions: An Exemplary Study on Preselected Campylobacter jejuni Isolates.

Frontiers in Immunology 9:213.

184. Razin S (1997) The minimal cellular genome of mycoplasma. Indian Journal of Biochemistry and

Biophysics 34(1-2):124-130.

185. Graham LM, et al. (2006) Soluble Dectin-1 as a tool to detect β-glucans. Journal of Immunological

Methods 314(1):164-169.

186. Czajkowsky DM, Hu J, Shao Z, & Pleass RJ (2012) Fc-fusion proteins: new developments and future

perspectives. EMBO Molecular Medicine 4(10):1015-1028.

187. Carter PJ (2011) Introduction to current and future protein therapeutics: a protein engineering

perspective. Experimental Cell Research 317(9):1261-1269.

188. Capon DJ, et al. (1989) Designing CD4 immunoadhesins for AIDS therapy. Nature 337(6207):525-

531.

189. Goldenberg MM (1999) Etanercept, a novel drug for the treatment of patients with severe, active

rheumatoid arthritis. Clinical Therapeutics 21(1):75-87; discussion 71-72.

190. Scheen AJ (2017) Dulaglutide for the treatment of type 2 diabetes. Expert Opinion on Biological

Therapy 17(4):485-496.

191. Sioud M, Westby P, Olsen JK, & Mobergslien A (2015) Generation of new peptide-Fc fusion proteins

that mediate antibody-dependent cellular cytotoxicity against different types of cancer cells. Mol Ther

Methods Clin Dev 2:15043.

192. Fasting C, et al. (2012) Multivalency as a chemical organization and action principle. Angewandte

Chemie. International Ed. In English 51(42):10472-10498.

193. Kilcoyne M, et al. (2012) Construction of a natural mucin microarray and interrogation for biologically

relevant glyco-epitopes. Analytical Chemistry 84(7):3330-3338.

194. Geissner A, et al. (2019) Microbe-focused glycan array screening platform. Proceedings of the

National Academy of Sciences of the United States of America.

195. Gade M, et al. (2018) Microarray Analysis of Oligosaccharide-Mediated Multivalent Carbohydrate-

Protein Interactions and Their Heterogeneity. Chembiochem: A European Journal of Chemical Biology

19(11):1170-1177.

196. Shanthamurthy CD, et al. (2018) ABO Antigens Active Tri- and Disaccharides Microarray to Evaluate

C-type Lectin Receptor Binding Preferences. Scientific Reports 8(1):6603.

197. Ishikawa T, et al. (2013) Identification of Distinct Ligands for the C-type Lectin Receptors Mincle and

Dectin-2 in the Pathogenic Fungus Malassezia. Cell Host & Microbe 13(4):477-488.

198. Hsu TL, et al. (2009) Profiling carbohydrate-receptor interaction with recombinant innate immunity

receptor-Fc fusion proteins. Journal of Biological Chemistry 284(50):34479-34489.

199. Szymanski CM & Gaynor E (2012) How a sugary bug gets through the day: Recent developments in

understanding fundamental processes impacting Campylobacter jejuni pathogenesis. Gut Microbes

3(2):135-144.

References

- 55 -

200. van Sorge NM, et al. (2009) N-glycosylated proteins and distinct lipooligosaccharide glycoforms of

Campylobacter jejuni target the human C-type lectin receptor MGL. Cellular Microbiology

11(12):1768-1781.

201. Singh SK, et al. (2009) Characterization of murine MGL1 and MGL2 C-type lectins: distinct glycan

specificities and tumor binding properties. Molecular Immunology 46(6):1240-1249.

202. Artigas G, et al. (2017) Glycopeptides as Targets for Dendritic Cells: Exploring MUC1 Glycopeptides

Binding Profile toward Macrophage Galactose-Type Lectin (MGL) Orthologs. Journal of Medicinal

Chemistry 60(21):9012–9021.

203. Takano T, et al. (2017) Dectin-1 intracellular domain determines species-specific ligand spectrum by

modulating receptor sensitivity. Journal of Biological Chemistry 292(41):16933-16941.

204. Taylor PR, et al. (2002) The β-glucan receptor, dectin-1, is predominantly expressed on the surface of

cells of the monocyte/macrophage and neutrophil lineages. Journal of Immunology 169(7):3876-3882.

205. Ariizumi K, et al. (2000) Identification of a novel, dendritic cell-associated molecule, dectin-1, by

subtractive cDNA cloning. Journal of Biological Chemistry 275(26):20157-20167.

206. Brown GD, et al. (2003) Dectin-1 mediates the biological effects of beta-glucans. Journal of

Experimental Medicine 197(9):1119-1124.

207. Gantner BN, Simmons RM, Canavera SJ, Akira S, & Underhill DM (2003) Collaborative induction of

inflammatory responses by dectin-1 and Toll-like receptor 2. Journal of Experimental Medicine

197(9):1107-1117.

208. Yadav M & Schorey JS (2006) The β-glucan receptor dectin-1 functions together with TLR2 to

mediate macrophage activation by mycobacteria. Blood 108(9):3168-3175.

209. Ahren IL, Eriksson E, Egesten A, & Riesbeck K (2003) Nontypeable Haemophilus influenzae activates

human eosinophils through β-glucan receptors. American Journal of Respiratory Cell and Molecular

Biology 29(5):598-605.

210. Lefèvre L, et al. (2013) The C-type Lectin Receptors Dectin-1, MR, and SIGNR3 Contribute Both

Positively and Negatively to the Macrophage Response to Leishmania infantum. Immunity 38(5):1038-

1049.

211. Lima-Junior DS, Mineo TWP, Calich VLG, & Zamboni DS (2017) Dectin-1 Activation during

Leishmania amazonensis Phagocytosis Prompts Syk-Dependent Reactive Oxygen Species Production

To Trigger Inflammasome Assembly and Restriction of Parasite Replication. Journal of Immunology

199(6):2055-2068.

212. Schorey JS & Lawrence C (2008) The pattern recognition receptor Dectin-1: from fungi to

mycobacteria. Current Drug Targets 9(2):123-129.

213. Kiyotake R, et al. (2015) Human Mincle Binds to Cholesterol Crystals and Triggers Innate Immune

Responses. Journal of Biological Chemistry 290(42):25322-25332.

214. Mori D, Shibata K, & Yamasaki S (2017) C-Type Lectin Receptor Dectin-2 Binds to an Endogenous

Protein β-Glucuronidase on Dendritic Cells. PloS One 12(1):e0169562.

215. Yamasaki S, et al. (2009) C-type lectin Mincle is an activating receptor for pathogenic fungus,

Malassezia. Proceedings of the National Academy of Sciences of the United States of America

106(6):1897-1902.

216. Neumann K, et al. (2014) Clec12a is an inhibitory receptor for uric acid crystals that regulates

inflammation in response to cell death. Immunity 40(3):389-399.

References

- 56 -

217. Mishalian I, et al. (2011) Recruited macrophages control dissemination of group A Streptococcus from

infected soft tissues. Journal of Immunology 187(11):6022-6031.

218. Goldmann O, Rohde M, Chhatwal GS, & Medina E (2004) Role of macrophages in host resistance to

group A streptococci. Infection and Immunity 72(5):2956-2963.

219. Loof TG, Rohde M, Chhatwal GS, Jung S, & Medina E (2007) The Contribution of Dendritic Cells to

Host Defenses against Streptococcus pyogenes. The Journal of infectious diseases 196(12):1794-1803.

220. Goldmann O, et al. (2007) Transcriptome analysis of murine macrophages in response to infection with

Streptococcus pyogenes reveals an unusual activation program. Infection and Immunity 75(8):4148-

4157.

221. Finlay BB & McFadden G (2006) Anti-Immunology: Evasion of the Host Immune System by Bacterial

and Viral Pathogens. Cell 124(4):767-782.

222. Laabei M & Ermert D (2019) Catch Me if You Can: Streptococcus pyogenes Complement Evasion

Strategies. Journal of Innate Immunity 11(1):3-12.

223. Stollerman GH & Dale JB (2008) The importance of the group a streptococcus capsule in the

pathogenesis of human infections: a historical perspective. Clinical Infectious Diseases 46(7):1038-

1045.

224. Fieber C & Kovarik P (2014) Responses of innate immune cells to group A Streptococcus. Frontiers in

Cellular and Infection Microbiology 4:140.

225. Herre J, Gordon S, & Brown GD (2004) Dectin-1 and its role in the recognition of β-glucans by

macrophages. Molecular Immunology 40(12):869-876.

226. McGreal EP, Martinez-Pomares L, & Gordon S (2004) Divergent roles for C-type lectins expressed by

cells of the innate immune system. Molecular Immunology 41(11):1109-1121.

227. Guerry P, et al. (2002) Phase variation of Campylobacter jejuni 81-176 lipooligosaccharide affects

ganglioside mimicry and invasiveness in vitro. Infection and Immunity 70(2):787-793.

228. Bacon DJ, et al. (2001) A phase-variable capsule is involved in virulence of Campylobacter jejuni 81-

176. Molecular Microbiology 40(3):769-777.

229. Pequegnat B, et al. (2017) Phase-Variable Changes in the Position of O-Methyl Phosphoramidate

Modifications on the Polysaccharide Capsule of Campylobacter jejuni Modulate Serum Resistance.

Journal of Bacteriology 199(14):e00027-00017.

230. Jerome JP, et al. (2011) Standing genetic variation in contingency loci drives the rapid adaptation of

Campylobacter jejuni to a novel host. PloS One 6(1):e16399.

231. Phongsisay V (2015) Campylobacter jejuni targets immunoglobulin-like receptor LMIR5. Molecular

Immunology 63(2):574-578.

232. Doran KS, et al. (2005) Blood-brain barrier invasion by group B Streptococcus depends upon proper

cell-surface anchoring of lipoteichoic acid. The Journal of Clinical Investigation 115(9):2499-2507.

233. Chatellier S, et al. (2000) Genetic relatedness and superantigen expression in group A streptococcus

serotype M1 isolates from patients with severe and nonsevere invasive diseases. Infection and

Immunity 68(6):3523-3534.

234. Kansal RG, McGeer A, Low DE, Norrby-Teglund A, & Kotb M (2000) Inverse relation between

disease severity and expression of the streptococcal cysteine protease, SpeB, among clonal M1T1

References

- 57 -

isolates recovered from invasive group A streptococcal infection cases. Infection and Immunity

68(11):6362-6369.

235. Percy MG & Gründling A (2014) Lipoteichoic Acid Synthesis and Function in Gram-Positive Bacteria.

Annual Review of Microbiology 68(1):81-100.

236. Ofek I, Simpson WA, & Beachey EH (1982) Formation of Molecular Complexes Between a

Structurally Defined M Protein and Acylated or Deacylated Lipoteichoic Acid of Streptococcus

pyogenes. Journal of Bacteriology 149(2):426-433.

237. Miyake Y, Masatsugu OH, & Yamasaki S (2015) C-Type Lectin Receptor MCL Facilitates Mincle

Expression and Signaling through Complex Formation. Journal of Immunology 194(11):5366-5374.

238. Kerscher B, et al. (2016) Signalling through MyD88 drives surface expression of the mycobacterial

receptors MCL (Clecsf8, Clec4d) and Mincle (Clec4e) following microbial stimulation. Microbes and

Infection 18(7–8):505-509.

239. Oliveira-Nascimento L, Massari P, & Wetzler LM (2012) The Role of TLR2 in Infection and

Immunity. Frontiers in Immunology 18(3):79.

240. Gratz N, et al. (2008) Group A streptococcus activates type I interferon production and MyD88-

dependent signaling without involvement of TLR2, TLR4, and TLR9. Journal of Biological Chemistry

283(29):19879-19887.

241. Gratz N, et al. (2011) Type I interferon production induced by Streptococcus pyogenes-derived nucleic

acids is required for host protection. PLoS Pathogens 7(5):e1001345.

242. Loof TG, Goldmann O, Gessner A, Herwald H, & Medina E (2010) Aberrant inflammatory response to

Streptococcus pyogenes in mice lacking myeloid differentiation factor 88. American Journal of

Pathology 176(2):754-763.

243. Loof TG, Goldmann O, & Medina E (2008) Immune Recognition of Streptococcus pyogenes by

Dendritic Cells. Infection and Immunity 76(6):2785-2792.

244. Picard C, et al. (2003) Pyogenic bacterial infections in humans with IRAK-4 deficiency. Science

299(5615):2076-2079.

245. von Bernuth H, et al. (2008) Pyogenic bacterial infections in humans with MyD88 deficiency. Science

321(5889):691-696.

246. Fiebig A, et al. (2015) Comparative Genomics of Streptococcus pyogenes M1 isolates differing in

virulence and propensity to cause systemic infection in mice. International Journal of Medical

Microbiology 305(6):532-543.

247. Johannssen T & Lepenies B (2017) Glycan-Based Cell Targeting To Modulate Immune Responses.

Trends in Biotechnology 35(4):334-346.

248. Lepenies B, Lee J, & Sonkaria S (2013) Targeting C-type lectin receptors with multivalent

carbohydrate ligands. Advanced Drug Delivery Reviews 65(9):1271-1281.

249. Chappell CP, et al. (2014) Targeting antigens through blood dendritic cell antigen 2 on plasmacytoid

dendritic cells promotes immunologic tolerance. Journal of Immunology 192(12):5789-5801.

250. Unger WWJ, et al. (2012) Glycan-modified liposomes boost CD4+ and CD8+ T-cell responses by

targeting DC-SIGN on dendritic cells. Journal of Controlled Release 160(1):88-95.

References

- 58 -

251. Aarnoudse CA, Bax M, Sanchez-Hernandez M, Garcia-Vallejo JJ, & van Kooyk Y (2008) Glycan

modification of the tumor antigen gp100 targets DC-SIGN to enhance dendritic cell induced antigen

presentation to T cells. International Journal of Cancer 122(4):839-846.

252. Martinez-Avila O, et al. (2009) Gold manno-glyconanoparticles: multivalent systems to block HIV-1

gp120 binding to the lectin DC-SIGN. Chemistry 15(38):9874-9888.

253. Arnaiz B, Martinez-Avila O, Falcon-Perez JM, & Penades S (2012) Cellular uptake of gold

nanoparticles bearing HIV gp120 oligomannosides. Bioconjugate Chemistry 23(4):814-825.

254. Wang SK, et al. (2008) Targeting the carbohydrates on HIV-1: Interaction of oligomannose dendrons

with human monoclonal antibody 2G12 and DC-SIGN. Proceedings of the National Academy of

Sciences of the United States of America 105(10):3690-3695.

255. Chiodo F, et al. (2014) Galactofuranose-coated gold nanoparticles elicit a pro-inflammatory response

in human monocyte-derived dendritic cells and are recognized by DC-SIGN. ACS Chemical Biology

9(2):383-389.

256. Garcia-Vallejo JJ, et al. (2013) Multivalent glycopeptide dendrimers for the targeted delivery of

antigens to dendritic cells. Molecular Immunology 53(4):387-397.

257. Zimmermann S & Lepenies B (2015) Glycans as Vaccine Antigens and Adjuvants: Immunological

Considerations. Methods in Molecular Biology 1331:11-26.

258. Mortensen R, et al. (2017) Local Th17/IgA immunity correlate with protection against intranasal

infection with Streptococcus pyogenes. PloS One 12(4):e0175707-e0175707.

259. Linehan JL, et al. (2015) Generation of Th17 cells in response to intranasal infection requires TGF-β1

from dendritic cells and IL-6 from CD301b+ dendritic cells. Proceedings of the National Academy of

Sciences of the United States of America 112(41):12782-12787.

260. Wang B, et al. (2010) Induction of TGF-β1 and TGF-β1-dependent predominant Th17 differentiation

by group A streptococcal infection. Proceedings of the National Academy of Sciences of the United

States of America 107(13):5937-5942.

261. Ostrop J, et al. (2015) Contribution of MINCLE-SYK Signaling to Activation of Primary Human

APCs by Mycobacterial Cord Factor and the Novel Adjuvant TDB. Journal of Immunology

195(5):2417-2428.

262. Werninghaus K, et al. (2009) Adjuvanticity of a synthetic cord factor analogue for subunit

Mycobacterium tuberculosis vaccination requires FcRγ-Syk-Card9-dependent innate immune

activation. Journal of Experimental Medicine 206(1):89-97.

263. Khader SA, et al. (2007) IL-23 and IL-17 in the establishment of protective pulmonary CD4+ T cell

responses after vaccination and during Mycobacterium tuberculosis challenge. Nature Immunology

8(4):369-377.

264. Agger EM, et al. (2008) Cationic liposomes formulated with synthetic mycobacterial cordfactor

(CAF01): a versatile adjuvant for vaccines with different immunological requirements. PloS One

3(9):e3116.

265. Davidsen J, et al. (2005) Characterization of cationic liposomes based on

dimethyldioctadecylammonium and synthetic cord factor from M. tuberculosis (trehalose 6,6'-

dibehenate)-a novel adjuvant inducing both strong CMI and antibody responses. Biochimica et

Biophysica Acta 1718(1-2):22-31.

266. van Dissel JT, et al. (2014) A novel liposomal adjuvant system, CAF01, promotes long-lived

Mycobacterium tuberculosis-specific T-cell responses in human. Vaccine 32(52):7098-7107.

References

- 59 -

267. Decout A, et al. (2017) Rational design of adjuvants targeting the C-type lectin Mincle. Proceedings of

the National Academy of Sciences 114(10):2675-2680.

268. Stocker BL, Khan AA, Chee SH, Kamena F, & Timmer MS (2014) On one leg: trehalose monoesters

activate macrophages in a Mincle-dependant manner. Chembiochem: A European Journal of Chemical

Biology 15(3):382-388.

269. Khan AA, et al. (2011) Long-chain lipids are required for the innate immune recognition of trehalose

diesters by macrophages. Chembiochem: A European Journal of Chemical Biology 12(17):2572-2576.

270. Bird JH, et al. (2018) Synthesis of Branched Trehalose Glycolipids and Their Mincle Agonist Activity.

Journal of Organic Chemistry 83(15):7593-7605.

271. Kostarnoy AV, et al. (2017) Receptor Mincle promotes skin allergies and is capable of recognizing

cholesterol sulfate. Proceedings of the National Academy of Sciences of the United States of America

114(13):E2758-e2765.

272. Nagata M, et al. (2017) Intracellular metabolite β-glucosylceramide is an endogenous Mincle ligand

possessing immunostimulatory activity. Proceedings of the National Academy of Sciences of the United

States of America 114(16):E3285-E3294.

273. Iborra S & Sancho D (2014) Signalling versatility following self and non-self sensing by myeloid C-

type lectin receptors. Immunobiology 220(2):175-184.

274. Miyake Y, et al. (2013) C-type lectin MCL is an FcRγ-coupled receptor that mediates the adjuvanticity

of mycobacterial cord factor. Immunity 38(5):1050-1062.

275. Lobato-Pascual A, Saether PC, Fossum S, Dissen E, & Daws MR (2013) Mincle, the receptor for

mycobacterial cord factor, forms a functional receptor complex with MCL and FcεRI-γ. European

Journal of Immunology 43(12):3167-3174.

276. Matsunaga I & Moody DB (2009) Mincle is a long sought receptor for mycobacterial cord factor.

Journal of Experimental Medicine 206(13):2865-2868.

277. Patin EC, Orr SJ, & Schaible UE (2017) Macrophage Inducible C-Type Lectin As a Multifunctional

Player in Immunity. Frontiers in Immunology 8:861.

278. Schneewind O & Missiakas D (2014) Lipoteichoic acids, phosphate-containing polymers in the

envelope of gram-positive bacteria. Journal of Bacteriology 196(6):1133-1142.

279. Peyssonnaux C, et al. (2005) HIF-1α expression regulates the bactericidal capacity of phagocytes.

Journal of Clinical Investigation 115(7):1806-1815.

280. Kuhlicke J, Frick JS, Morote-Garcia JC, Rosenberger P, & Eltzschig HK (2007) Hypoxia inducible

factor (HIF)-1 coordinates induction of Toll-like receptors TLR2 and TLR6 during hypoxia. PloS One

2(12):e1364.

281. Kim SY, et al. (2010) Hypoxic stress up-regulates the expression of Toll-like receptor 4 in

macrophages via hypoxia-inducible factor. Immunology 129(4):516-524.

282. Schoenen H, et al. (2014) Differential control of Mincle-dependent cord factor recognition and

macrophage responses by the transcription factors C/EBPβ and HIF1α. Journal of Immunology

193(7):3664-3675.

283. van Uden P, et al. (2011) Evolutionary conserved regulation of HIF-1β by NF-κB. PLoS Genetics

7(1):e1001285.

References

- 60 -

284. van Uden P, Kenneth NS, & Rocha S (2008) Regulation of hypoxia-inducible factor-1α by NF-κB.

Biochemical Journal 412(3):477-484.

285. Yang H, et al. (2007) pVHL acts as an adaptor to promote the inhibitory phosphorylation of the NF-κB

agonist Card9 by CK2. Molecular Cell 28(1):15-27.

Affidavit

- 61 -

Affidavit

I herewith declare that I autonomously carried out the PhD-thesis entitled “Role of C-type

lectin receptors in bacterial recognition”.

No third party assistance has been used.

I did not receive any assistance in return for payment by consulting agencies or any other

person. No one received any kind of payment for direct or indirect assistance in correlation to

the content of the submitted thesis.

I conducted the project at the following institution: Immunology Unit & Research Center for

Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover,

Foundation.

The thesis has not been submitted elsewhere for an exam, as thesis or for evaluation in a

similar context.

I hereby affirm the above statements to be complete and true to the best of my knowledge.

Acknowledgement

- 62 -

Acknowledgement

As we express our gratitude, we must never forget that highest

appreciation is not to utter words, but to live by them. -John F. Kennedy-

First of all, I would like to thank my supervisor Bernd Lepenies for giving me the opportunity

to perform a PhD in his research group. I am grateful for the support, novel ideas and

inspiring discussions throughout the time of the PhD. Thank you for having an open door and

open ears for various problems, small or large. Thank you for your guidance but also for

believing in me, giving me the freedom to do work my own way. Your passion for science

motivated and inspired me throughout the PhD times and I will never forget this.

I also want to thank Maren von Köckritz-Blickwede and Bastian Opitz for participating as

supervisors in my supervision group and their constant support with fruitful discussions and

comments. I am particularly grateful to Maren von Köckritz-Blickwede since you support me

from my time as a Master`s student onwards, hence you hugely contributed to my successful

scientific carrier in a hundred ways and I am immensely honored that you guided me

throughout the times!

Special thanks belongs to the whole immunology group, especially to:

Marie-Kristin Raulf for your practical and mental support during the whole PhD time and

even before that. Tim Ebbecke for the perfect times including a lot of laughing in the lab, the

office and off the work. Mariano Prado Acosta for your support in and off the lab and funny

spanish/german lessons. Dimitri Lindenwald for amazing stories and crucial fun facts of the

day. Guillaume Goyette-Desjardins for help with several computer programs and Silke

Schöneberg for technical support and always a smile on the face.

I am especially grateful to João Monteiro: Thanks so much for your constant encouragement

and your optimism. Your ideas have meant a lot to me and helped in so many ways! I will

never forget our time, especially the starting time. You have been a great colleague to me,

you will be still a great friend to me and for that I will be forever thankful. “In the end,

everything will be fine”

Acknowledgement

- 63 -

A big thanks to present and former PhD and practical students, involved in my projects.

Especially Timo Johannssen, Rebecca Möller, Veronika Breitkopf, Katrin Opitz and Ina Ott.

I also want to thank the Hannover Graduate School for the financial support to attend

conferences.

Additional thanks goes to all collaborators, particularly to Christine Josenhans for providing

the C. jejuni strains and the support in the CLR/C. jejuni project. Furthermore, I would like to

thank Sho Yamasaki and Nina van Sorge for the successful GAS collaboration project. Also a

big thanks to Graham Brogden for the great introduction into the world of lipids.

Furthermore, I would like to thank my friends apart the lab, particularly Melissa Langer for

her support throughout the time and proofreading of this thesis.

Finally, I truely thank my family for their love, support and trust in me. I will be grateful

forever from the deepest of my heart.