Characterization of antigen-presenting cell function in...

100
Doctoral thesis from the Department of Immunology, Wenner-Gren Institute, Stockholm University, Sweden Characterization of antigen-presenting cell function in vitro and ex vivo Pablo Giusti Stockholm 2011

Transcript of Characterization of antigen-presenting cell function in...

Page 1: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

Doctoral thesis from the Department of Immunology, Wenner-Gren Institute,

Stockholm University, Sweden

Characterization of antigen-presenting cell

function in vitro and ex vivo

Pablo Giusti

Stockholm 2011

Page 2: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

ii

Imagination is more important than knowledge. For knowledge is limited to all we now know and understand, while imagination

embraces the entire world, and all there ever will be to know and understand.

Albert Einstein

Page 3: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

iii

ABSTRACT Long-term protective immune responses depend on proper initiation of adaptive immunity by

professional antigen-presenting cells (APCs). Autoimmune disorders and certain infections

can cause disease through manipulating the immune system and modulation of APCs may be

important for the outcome of these conditions. This work aimed to investigate the behaviour

of different APC subsets during conditions known to cause improper immune responses.

In Paper I, the effects of an anti-inflammatory compound called Rabeximod, intended for

treatment of rheumatoid arthritis (RA) were investigated. The results showed that generation

of monocyte-derived DCs (MoDCs) and inflammatory macrophages (MΦs) was impaired

upon exposure to Rabeximod while differentiation of anti-inflammatory (Ai)-MΦs was

unaffected. When MoDCs and inflammatory MΦs were exposed to Rabeximod they exhibited

a reduced capacity to stimulate allogeneic T cells while Ai- MΦs remained unaffected. These

findings suggest that Rabeximod acts by inhibiting the functionality of inflammatory subsets

of APCs.

In Paper II, the effects of different Plasmodium falciparum-derived stimuli such as hemozoin

(Hz) and infected red blood cells (iRBCs) on MoDCs were investigated. Both stimuli

triggered increased expression of certain co-stimulatory molecules and chemokine receptors,

indicating increased activation and migration of MoDCs. These MoDCs secreted TNF-α, IL-6

and IL-10, but no IL-12. MoDCs exposed to iRBCs induced allogeneic T-cell proliferation

while those exposed to Hz did not. These results indicate that DCs may be improperly

activated during malaria and may therefore be unable to efficiently activate T cells.

In Paper III, innate aspects of immunity in children from the Fulani, that displays better

protection against malaria, were investigated and compared to the sympatric ethnic group the

Dogon. We observed that distinct subsets of APCs from Fulani children expressed increased

levels of activation markers and their toll-like receptor (TLR) responses were unaffected

while undergoing P.falciparum infection. Conversely, the Dogon exhibited decreased

activation of APCs and severely suppressed TLR responses when undergoing infection. These

differences in innate responses, including APC behaviour may indicate an important role for

TLR and APC activation in malarial immunity.

In conclusion, detailed knowledge of precise mechanisms of APC activation can be very

helpful in understanding the proper course of specific effector immune responses. Moreover,

this knowledge would allow for manipulation of APCs that could be used for treatment of

inflammatory disorders and in the generation of efficient vaccines against infectious diseases.

Page 4: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

iv

LIST OF PAPERS

The thesis is based on the following papers which will be referred to by their roman numerals:

I. Giusti P, Frascaroli G, Tammik C, Gredmark-Russ S, Söderberg-Nauclér C,

Varani S. “The novel anti-rheumatic compound Rabeximod impairs

differentiation and function of human pro-inflammatory dendritic cells and

macrophages.” Immunobiology. 2011 Jan-Feb;216(1-2):243-50.

II. Giusti P, Urban B, Frascaroli G, Tinti A, Troye-Blomberg M, Varani S.

“Plasmodium falciparum-infected erythrocytes and β-hematin induce partial

maturation of human dendritic cells and increase their migratory ability in

response to lymphoid chemokines.” Infection & Immunity 2011 Jul;79(7):2727-36

III. Arama C, * Giusti P, * Boström S, Dara V, Traore B, Dolo A, Doumbo O, Varani

S, Troye-Blomberg M, "Interethnic Differences in Antigen-Presenting Cell

Activation and TLR Responses in Malian Children during Plasmodium

falciparum Malaria.” PLoS One. 2011 Mar 31;6(3):e18319 * These authors

contributed equally to this work.

Page 5: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

v

ABBREVIATIONS APC Antigen-presenting cell

Ai-MΦ Anti-inflammatory MΦ

Allo-MΦ Allostimulated MΦ

BCR B-cell receptor

BDCA Blood-dendritic cell antigen

CARD Caspase recruitment domain

CC Chemotactic cytokines

CCR Chemokine receptor

CD Cluster of differentiation

CLIP Class II-associated invariant chain

peptides

CLR C-type lectin receptor

CR Complement receptor

DAMP Danger associated molecular

pattern

DC Dendritic cell

Flt3L FMS-like tyrosine kinase 3 ligand

GPI Glycosylphosphatidylinositol

anchor

GM-CSF Granulocyte macrophage-colony

stimulating factor

HLA Human-leukocyte antigen

HMGB1 High mobility group box 1

HSP Heat shock protein

Hz Hemozoin

Ig Immunoglobulin

IFN Interferon

iRBC Infected red blood cell

IRAK IL-1 receptor-associated kinase

IRF IFN regulatory factor

IL Interleukin

ISG IFN-stimulated genes

ITAM Immunoreceptor tyrosine-based

activation motif

LPS Lipopolysaccharide

M-CSF Monocyte-colony stimulating

factor

MΦ Macrophage

MCP Monocyte-chemoattractant protein

MDA Melanoma-differentiation

associated gene

mDC Myeloid DC

MIP Macrophage-inflammatory protein

MLR Mixed leukocyte reaction

MoDC Monocyte-derived dendritic cell

MMP Matrix metalloprotease

MyD88 Myeloid differentiation primary

response gene 88

NK cell Natural-killer cell

NF-kb Nuclear factor kappa beta

NLR Nod-like receptor

PAMP Pathogen-associated molecular

pattern

PBMC Peripheral blood mononuclear cell

pDC Plasmacytoid DC

PRR Pathogen-recognition receptor

RA Reumathoid arthritis

RANK Receptor activator of NFκB

RANTES Regulated on Activation Normal T

Expressed and Secreted Protein

RIG Retinoic acid-inducible gene

Tc T-cytotoxic cell

Th T-helper cell

TIR Toll/IL-1 receptor domain

TIRAP TIR- adaptor protein

TRAF TNF receptor associated factor

TRIF TIR-domain-containing adapter-

inducing interferon-β

Treg Regulatory-T cell

TCR T-cell receptor

TLR Toll-like receptor

TNF Tumour Necrosis factor

Page 6: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

TABLE OF CONTENTS ABSTRACT ........................................................................................................................................................ iii LIST OF PAPERS .............................................................................................................................................. iv ABBREVIATIONS ............................................................................................................................................. v TABLE OF CONTENTS ................................................................................................................................... vi INTRODUCTION ............................................................................................................................................... 7

THE IMMUNE SYSTEM ......................................................................................................... 7 T cells ....................................................................................................................................................... 10 Antigen-presenting cells of innate immunity........................................................................... 12 Recognition strategies of antigen-presenting cells ................................................................ 19

ANTIGEN PRESENTATION ................................................................................................. 24 Signal 1 .................................................................................................................................................... 24 Signal 2 .................................................................................................................................................... 29 Signal 3 .................................................................................................................................................... 30

INFLAMMATION ................................................................................................................. 35 Leukocyte movement ........................................................................................................................ 36

RELATED BACKGROUND .......................................................................................................................... 38 APCS IN INFLAMMATION AND THE LINK TO ADAPTIVE IMMUNITY ............................. 38

Monocytes .............................................................................................................................................. 38 Macrophages ......................................................................................................................................... 39 Dendritic cells ....................................................................................................................................... 40 Endogenous ligands ........................................................................................................................... 42 The manipulation of APCs by cytokines ..................................................................................... 43

MALARIA ............................................................................................................................. 44 REUMATHOID ARTHRITIS ................................................................................................. 49

THE STUDIES ................................................................................................................................................. 53 GENERAL AIM ..................................................................................................................... 53 SPECIFIC AIMS .................................................................................................................... 53

Paper I ..................................................................................................................................................... 53 Paper II .................................................................................................................................................... 53 Paper III .................................................................................................................................................. 53

METHODS ........................................................................................................................................................ 55 Paper I ..................................................................................................................................................... 55 Paper II .................................................................................................................................................... 55 Paper III .................................................................................................................................................. 56

RESULTS AND DISCUSSION ...................................................................................................................... 57 Paper I ..................................................................................................................................................... 57 Paper II .................................................................................................................................................... 59 Paper III .................................................................................................................................................. 63

GENERAL CONCLUSIONS .......................................................................................................................... 68 FUTURE WORK .............................................................................................................................................. 71 ACKNOWLEDGEMENTS ............................................................................................................................. 72 REFERENCES .................................................................................................................................................. 75

Page 7: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

7

INTRODUCTION

THE IMMUNE SYSTEM

Mammals have evolved in co-existence with a wide variety of viruses, parasites and bacteria.

Many of these microbes have adapted to niches in or outside the body and do not normally

cause disease, they are called commensals. However, these and other microbes can, under

certain conditions, sometimes invade tissues where they can cause disease. Therefore, the

body has developed defence mechanisms to keep tissues free from invading microbes and

maintain tissue homeostasis. Those defence mechanisms comprise antimicrobial peptides,

signalling molecules and a complex network of cells that have evolved to find and eliminate

pathogens and even remember the invading microbes. The immune system is commonly

divided into two, although intervening, different branches that we call the innate and the

adaptive immune system.

The innate immune system has evolved to detect danger signals and indiscriminately attack

pathogens. Innate immunity does not, from a short time perspective, seem to adapt to changes

in the surrounding environment and is very similar in all humans. In order to impede

microbial colonization of the human body, obstacles are created at many different levels.

Physical barriers, such as the skin and mucosal surfaces of the body are the first ones.

Everything from mouth, to stomach, to the end of the intestine can be regarded as a tunnel

through the body and is actually to be considered as the body’s exterior surfaces. These

surfaces are colonized by enormous amounts of microorganisms that do not normally cause

disease. The acidic environment in the stomach is an additional physical barrier. Inside the

body, in circulation, there are antibacterial peptides and complement proteins that recognise

pathogens and initiate a cascade of proteolytic activities ultimately leading to lysis of

microbes.

Page 8: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

8

Finally, there are the cells of the innate immune system. They are equipped with different

kinds of receptors to recognize disruptions of the normal homeostasis and infectious stimuli.

These receptors have evolved to recognize danger signals and recurrent patterns that are

essential to, and therefore conserved among, pathogens. These pathogen associated molecular

patterns (PAMPs) and danger associated molecular patterns (DAMPs) are recognized by

pattern recognition receptors (PRRs). The best studied PRRs are the toll-like receptors (TLRs)

but there are many different PRRs and some debate as to what their function really is,

therefore, they will be further discussed in a separate chapter.

However, since living conditions in different social, cultural and geographical environments

can be variable there is a need for adaptation. In order to be able to adapt to these variable

conditions, thus responding as quickly and efficiently as possible, a distinct branch of the

immune system has evolved to specifically recognize and remember pathogens that it has

been exposed to. This is the adaptive branch of the immune system and is, in contrast to the

innate, unique in every human being.

In summary, the innate immune system is fast acting, unspecific and lacks memory while the

adaptive immunity is slower to act but is specific and has a memory component. The

interaction of these two branches results in an immune system that can act swiftly against all

pathogens and learn to respond more efficiently against pathogens that are common to a given

individual´s specific environment. These branches are dependent on each other for optimal

performance. The innate components are necessary for the adaptive branch to function and the

innate immune responses are much more efficient when aided by adaptive components.

All cells of the immune system originate in the bone marrow and, at certain stages of

development, in the liver. They are found throughout the body, particularly in the lymphatic

tissues and in circulation. These cells are collectively called leukocytes and they all stem from

hematopoietic stem cells. The pluripotent stem cells differentiate into common lymphoid

Page 9: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

9

progenitors or common myeloid progenitors. These then further differentiate into their

respective immune cells. B- , T- and natural killer (NK)- cells and possibly some subsets of

dendritic cells (DCs) stem from a lymphoid progenitor, while granulocytes, monocytes and

most DCs stem from a myeloid progenitor. A vast majority of the circulating immune cells

are granulocytes. In round numbers granulocytes comprise about 60% of leukocytes in blood,

about 30% are lymphocytes and the final 10% are monocytes (Figure 1).

Figure 1. Cells of the immune system

Monocytes, macrophages (MΦs), granulocytes, DCs and NK cells belong to the innate branch

of the immune system. With the exception of NK cells these cells are able to constantly

survey their surroundings through endocytosis.

Leukocytes can communicate through the secretion of cytokines that are sometimes called the

hormones of the immune system. Since they are a means of communication between

Page 10: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

10

leukocytes they are called interleukins (IL). An important subgroup of cytokines that governs

cell motility are the chemotactic cytokines (CCs). These substances are shared among all cells

of the immune system and will be discussed in a separate chapter.

T cells

T lymphocytes are defined by expression of a T-cell receptor (TCR), through which they

recognize antigens. These cells are not able to recognize antigens directly, but the pathogen

needs to be degraded into peptide fragments that in association with human leukocyte antigen

(HLA) molecules on antigen-presenting cells (APCs) are presented to T cells. The details of

how this activation is orchestrated, the cells responsible and its consequences will be

discussed in more detail below.

The TCR lacks an intracellular signalling domain instead it is expressed together with a

molecule called cluster of differentiation (CD) 3. The CD3 molecule contains an

immunoreceptor tyrosine-based activation motif (ITAM) that can elicit intracellular

signalling. Intracellular signalling via CD3 is initiated upon binding of the TCR to an antigen-

HLA complex. The genes coding for the TCR are randomly rearranged during T-cell

development thus providing a unique specificity for each TCR. Because this rearrangement is

at random, T cells must be prevented from recognizing and attacking ones healthy tissues. A

selection process occurs in the thymus where less than 5% of the cells survive [1]. This

process is carried out in order to avoid the release of potentially harmful T cells. The final

outcome of this process is the release of T cells expressing a TCR repertoire with unique

specificities and harmless to healthy self. T cells leaving the thymus can be subdivided

according to the expression of their co-receptors, CD4+ cells are called T- helper (Th) cells,

CD8+ cells are called T-cytotoxic (Tc) cells.

Page 11: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

11

T helper cells

Th cells are defined by the expression of the co-receptor CD4. The activation of naïve CD4+ T

cells can lead to skewing of the T cell in different directions thus creating a heterogeneous

population of Th cells. Depending on the cytokines secreted by the activating APC the naïve

T cell will commit to a given lineage (Figure 2).

Figure 2. Th-cell differentiation and their consequences for immunity

Reprinted from Medzhitov, Nature 449, 819-826 (18 October 2007), doi:10.1038/nature06246 with permission

from Nature Publishing Group.

This lineage is established and sustained by promoting the expression of a specific

transcription factor which then suppresses transcription factors that direct the other lineages.

This was first discovered as the first two lineages defined were shown to be reciprocally

inhibitory and defined as Th1 and Th2 cells [2,3]. Later, additional subsets have been defined

that are now known as regulatory T cells (Tregs) and Th17 [4]. Tregs can either be induced in

the periphery and are then called Tr1 or Th3 cells or they can be released from the thymus in

which case they are referred to as natural (n) Tregs [5]. The transcription factors involved in

Page 12: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

12

establishing the distinct Th lineages are T-bet, GATA-3, RORγt and FoxP3 for Th1, Th2,

Th17 and Tregs respectively [6].

The functions of the different subsets of effector T cells are also different. Th1 responses have

traditionally been linked to the elimination of intracellular bacteria or viruses while Th2

confers protection against parasites and Th17 clears extracellular bacteria and fungi. Finally,

Tregs are necessary to avoid excessive tissue damage upon inflammation. This model

explains how adaptive immunity has evolved to produce diverse responses to different kinds

of pathogens. These directed responses can in turn enhance innate immunity.

T cytotoxic cells

Tc cells are defined by the fact that they express CD8 as co-receptor. Upon activation, Tc

cells kill other cells by induction of apoptosis or release of granules containing perforin or

granulosin that can lyse the infected cell [7]. Much like Th cells, some of the Tc effector cells

become memory cells, in order to sustain long term immunity, while the remaining cells

undergo apoptosis as soon as the primary infection is cleared [8].

Antigen-presenting cells of innate immunity

Professional APCs act as sentinels of the immune system. These cells play a pivotal role in

the induction of adaptive immune responses to antigenic stimulation by presenting antigens to

T cells in the context of HLA molecules expressed on their cell surface. Innate APCs

comprise two types of DCs, i.e. plasmacytoid DCs (pDCs) and myeloid DCs (mDCs),

monocytes and tissue-specific macrophages (MΦs). Importantly, B cells are also capable of

presenting antigens but are not considered as part of innate immunity and will not be dealt

with in this thesis.

Page 13: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

13

Monocytes

Monocytes develop from a myeloid progenitor in the bone marrow and are then released into

the bloodstream. Monocytes constitute about 10% of the blood leucocytes in humans and are

characterized as mononuclear leucocytes expressing the lipopolysaccharide (LPS) co-receptor

CD14. These cells play an important role in tissue homeostasis by patrolling vessels and

tissues [9] and constitute a reservoir for tissue-MΦs and DCs [10].

Monocyte subsets

About 20 years ago, a subpopulation of monocytes expressing Fcγ receptor (FcγR) III/CD16

was described [11]. The classical CD14++

CD16- cell expresses high levels of CD14 and no

CD16, while the non-classical CD14+CD16

++ cell type expresses CD16 and lower levels of

CD14. The different patterns of cytokine secretion of the two subsets have led to the terms

“inflammatory” and “classical” monocytes. However, since the function of the more recently

discovered monocyte subset is still not clearly defined the term “inflammatory” can be

unfortunate and the term “non-classical” is preferred. More recently another monocyte

population, that is present at a low frequency but exhibit unique features and expand in

response to cytokine treatment and inflammation, has been found. The immunophenotype of

this subset is somewhere in between classical and non-classical (CD14++

CD16+) which has

led to the definition of “intermediate” monocytes [12]. The role of monocytes when they enter

tissues or become activated in response to inflammatory stimuli will be further discussed

when discussing the role of APCs in inflammation.

Macrophages

Over 100 years ago, in 1908, Ilya Ilyich Mechnikov and Paul Ehrlich were awarded the Nobel

prise in Medicine for their work on phagocytosis. Much of their work was performed in MΦs

that Mechnikov described as mononuclear phagocytes playing an important role in pathogen

Page 14: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

14

elimination and housekeeping [13]. The term macrophage is derived from Greek and means

“big eater” in contrast to the granulocytes that were called “small eaters”. Phagocytosis by

MΦs can be induced by different types of receptors such as FcRs and will be further discussed

in the chapter of antigen presentation. MΦs have since then been described in various organs

such as lung, liver and nervous tissues as tissue-specific subsets. For the identification of MΦs

intracellular macrosialin, also called CD68, has been widely used [14] although it has been

proposed to be a marker for phagocytosis rather than a specific marker of MΦ subsets [15].

These cells also express the monocyte marker CD14 and, as professional APCs, they typically

express HLA class II, henceforth referred to as HLA-II, molecules on their surface.

Macrophage subsets

More than 25 years ago, interferon (IFN) -γ was identified as an outstanding factor that could

induce production of superoxides in MΦs and activate them to become potent immune cells

[16,17]. It was later described that IL-4 and IL-13 activation of MΦs inhibited the respiratory

burst and secretion of proinflammatory cytokines [18,19]. It was then proposed that these

factors could lead to a different kind of activation, which led to the concept of alternatively

activated MΦs [20].

In analogy with the Th1/Th2 activation of T cells, an M1/M2 characterisation of MΦs has

been suggested. Since the classical Th1 cytokine IFN-γ was the first activating factor

described, stimulation of MΦs induced by this mediator has been defined as “classical”

activation or more recently M1. In contrast, “alternative” activation of monocytes induced by

IL-4, IL-10 or IL-13 leads to M2 polarized MΦs, thus promoting a Th2 type of response

[21,22]. A subcategorizing of M2 polarized MΦs, where the M2 MΦs now include all MΦs

that are not M1, has been proposed [23]. This leads to a concept where the M1 is well defined

but the M2 would then comprise a heterogenous group of cells that have different roles in

infection and wound healing. Therefore this concept has been accused of oversimplification

Page 15: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

15

and a more complex picture of MΦ activation has been suggested that separates MΦ subtypes

according to their functions [24]. This classification defines classically activated M1 as MΦs

devoted to host defence, while the M2 are separated into wound healing MΦs and regulatory

MΦs [25]. This definition was suggested to better account for the role of MΦs in homeostasis.

For simplicity, we will refer to the different MΦ subsets as M1 for classically activated MΦs

and M2 for all other subsets.

Macrophage differentiation in vitro

Two important factors for macrophage differentiation are macrophage colony-stimulating

factor (M-CSF) and granulocyte-macrophage colony stimulating factor (GM-CSF). M-CSF is

constitutively produced by many different cell types and is readily detectable under steady

state conditions while GM-CSF is produced upon stimulation with inflammatory mediators.

Both factors induce MΦ differentiation and they have been suggested to represent the

extremes of the full spectrum of MΦ activation [26].

MΦs can be differentiated in vitro from monocytes using M-CSF or GM-CSF. Both GM-CSF

and M-CSF are individually important in the development of MΦs as lack of either lead to

altered MΦ homeostasis or pathology [27]. Cells obtained by these stimulations are

phenotypically similar. Nevertheless M-CSF-derived MΦs express higher levels of CD16 and

CD163 than GM-CSF derived MΦs. Both cell types are capable of antibody-dependent

cytotoxicity although the M2 MΦs seem slightly more efficient in this function, possibly as a

consequence of the increased CD16 expression [28]. Upon exposure to Mycobacterium, in

contrast to monocyte-derived DCs (Mo)DCs, MΦs do not secrete IL-12 but M1 secrete IL-23

which leads to more efficient T-cell priming, while M2 secrete IL-10 and are poor activators

of T cells [29].

Page 16: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

16

Dendritic Cells

DCs were first described in 1973 by Ralph Steinman and Zanvil Cohn [30,31]. Four years

later the authors concluded that DCs are at least 100 times more effective in stimulating

allogeneic T cells than any other major APC subset [32]. We now know that DCs are

specialized professional APCs and that their life cycle is adapted to find, process and present

antigens to naive T cells in lymphoid tissues. DCs are a very heterogeneous group of cells

both when it comes to their distribution as specialized cells in different tissues and to their

origin [33].

Immature DCs (iDCs) express high levels of PRRs to detect PAMPs and a repertoire of

chemokine receptors (CCRs) that are specialized to detect inflammatory mediators while

these cells patrol peripheral tissues. When DCs encounter inflammatory signals and/or

microbial components, they become activated and the expression of their surface molecules is

modified [34]. One of the most important maturation markers on DCs is the surface antigen

CD83, which is crucial for the induction of activated CD4+ T cells [35].

Down-regulation of PRRs and inflammatory CCRs, such as CCR1, CCR2 and CCR5 is also

part of the maturation process of DCs. Simultaneously, DCs up-regulate HLA molecules, co-

stimulatory molecules and CCRs specific for ligands expressed in lymphoid tissues such as

CCR7 and CXCR4 [36]. This change in CCR expression enables migration of DCs to T-cell

rich areas in secondary lymphoid organs where lymphoid chemokines are produced and

where DCs encounter massive amounts of naive T cells [37]. When DCs and T cells find their

perfect match, that is, an antigen-HLA-II complex on the DC and the TCR of a CD4+ T cell,

co-stimulation and cytokine secretion are increased. This T-cell activation triggers its

proliferation and thousands of T-cell clones will be produced in a matter of days.

Page 17: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

17

DC subsets

In humans, two complementary subsets of DCs called mDCs and pDCs have been described

in peripheral blood [38]. More recently, subgroups of mDCs according to specific surface

markers have been suggested (Table 1). Careful characterisation of these subsets has resulted

in surface markers specific for different DC subsets in circulation [39].

The major circulating mDC subpopulation is defined as lineage negative, CD1c+ CD11c

+

CD123- cells and characterised by blood dendritic cell antigen (BDCA)-1/CD1c expression.

Conversely, pDCs are defined as lineage negative, CD123+ and CD11c

- cells and express

BDCA-2 (CD303). BDCA-3 (CD141) is expressed on a subpopulation of mDCs that is also

defined as lineage negative, CD1c+, CD11c

+, CD123

-. These cells lack the expression of

BDCA-1, CD2, CD32 and CD64.

Blood DCs lack dendrites and markers of mature DCs such as CD83 and thus do not have

typical characteristics of DCs in tissue. These immature DCs appear to be in transit when

circulating in the blood, and they mature into functional DCs only after entering the tissue

[12].

Subtype CD1c CD11c CD123 CD303 CD2 CD16 CD32 CD64

BDCA-1 + + - - + - + +

BDCA-2 - - + + -/+ - - -

BDCA-3 - -/+ - - - - - -

CD16+DC - -/+ + - + + Nd Nd

Table 1. Expression of surface molecules on distinct blood DC subsets.

Abbreviation: Nd- no data. References: [39-41].

Dendritic cell differentiation in vitro

In order to obtain in vitro cultures of DCs, several protocols have been established to obtain

purified mDCs [42-44]. DCs derived from CD34+ cord blood cells differentiate along two

independent pathways in response to GM-CSF and TNF-α. After 5-6 days, two subsets

(CD1a+CD14

- and CD1a

-CD14

+ cells) can be observed; after 12 days in culture, all cells are

CD1a+CD14

-. The CD1a

+ precursors differentiate into Langerhans cells that contain Birbeck

Page 18: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

18

granules, whereas the CD14+ precursors lead to CD1a

+ mDCs that do not produce Birbeck

granules and that possess the characteristics of interstitial DCs [45]. MoDCs with the typical

phenotype of CD1a+CD14

- cells can be obtained in vitro by stimulation of purified monocytes

with IL-4 and GM-CSF for 5-6 days [44]. In vitro-generated MoDCs are considered to have

inflammatory properties [46]. These cells can further mature upon incubation with various

stimuli, such as bacterial LPS, inflammatory cytokines (TNF- and IL-1) and T-cell signals

(e.g. CD40L). A protocol for in vitro generation of the BDCA-3+ DC subset has been recently

reported that will be helpful for comparative studies on different DC populations and enable

manipulation of these cells to be used in different therapeutic settings [47].

Origin of DCs

The origin of DCs remains an unresolved issue. There are many subpopulations of DCs in

peripheral blood and different models for the development of DCs have been suggested [48].

It is believed that monocytes represent a pool of circulatory precursor cells that are capable of

differentiating into mDCs that resemble the features of interstitial DCs [34]. In support of this

hypothesis, it has been shown that monocytes can differentiate into DCs and migrate to the

lymph nodes upon ingestion of latex beads [10]. Similarly, Langerhans DCs in the skin can

arise from monocytes that infiltrate tissues [49], but under steady-state conditions they can

renew themselves to maintain the resident population [50]. Recently, it has been shown that

LPS can induce monocyte differentiation to DCs and that these differentiated cells can

migrate to lymph nodes and activate both CD4+ and CD8

+ T cells [51].

The origin of pDCs is particularly intriguing since they bare gene rearrangement previously

thought to be unique for lymphocytes and it seems that they can originate from both lymphoid

and myeloid precursors [52]. Even though pDCs and mDCs have been suggested to stem from

precursors of different origins it seems that the system allows some plasticity since pDCs

from bone marrow were able to differentiate to mDCs upon viral infection [53].

Page 19: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

19

Recognition strategies of antigen-presenting cells

It has now been more than 20 years since Charles Janeway proposed a link between innate

and adaptive immune responses [54]. He hypothesised that there are recurring PAMPs that are

essential to the function of microbes which are recognized by receptors of the host innate

immunity. These receptors are called PRRs. This was in line with the prevailing idea that

these receptors had evolved to distinguish self from non-self.

Five years later, Polly Matzinger suggested an alternative theory called “the danger model”

[55]. The “danger model” suggested that signals sent by stressed or injured tissues are the key

triggers to APCs. We now know that PRRs expressed on APCs have, in addition to

exogenous microbial derived ligands i.e. PAMPs, many endogenous ligands. Most of these

ligands can be related to stress, injury or necrotic cell death i.e. DAMPs [56-59]. The most

investigated family belonging to the PRRs in humans are the TLRs.

Toll-like receptors

Toll was first discovered as a protein involved in the development of drosophila but its role in

immunity was first shown by Bruno Lemaitre and Jules Hoffman as they showed that toll-

deleted mutants of drosophila were susceptible to lethal fungal infections [60]. The tolls were

later shown to be structurally related to the IL-1 receptor, suggesting an important role in

immunity [61].

The ligands and roles for the different receptors were discovered one after another [62-65]. So

far, ten different TLRs have been characterized in humans. They are all expressed by different

subtypes of APCs and have specific localizations and ligands (Table 2). TLR1, TLR2, TLR4,

TLR5 and TLR6 are found in the outer cell membrane and recognize recurring PAMPs in

bacteria, fungi, parasites and some viruses. Conversely, TLR3, TLR7, TLR8, and TLR9 are

located intracellularly and are specialized in recognition of intracellular bacterial and viral

nucleic acids. The motifs that TLRs recognize are not necessarily unique to pathogens. For

Page 20: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

20

example, it has been shown that TLR7 does not distinguish between influenza- and self-

derived single stranded RNA [66]. However, TLR7 is localized in endolysosomes where host

ssRNA is not normally present, thus avoiding any reaction against self-derived nucleic acids

under steady-state condition.

The expression of TLRs is different in monocytes, MΦs and different DC subsets as

previously mentioned. Different subtypes of DCs are devoted to defend against different kinds

of pathogens and therefore express different TLRs (Table 1). The pDCs express TLR1, TLR6,

TLR7, TLR9 and TLR10, while mDCs express TLR1, TLR2, TLR3, TLR4, TLR5, TLR6,

TLR8 and TLR10 [67].

Receptor Ligand and origin Location mDC pDC MoDC MΦ Monocyte

TLR1 Triacyl LpP (bacteria) Surface + + + + +

TLR2 Zymozan, PGN (fungi, G+ bacteria) Surface + - + + +

TLR3 dsRNA (viruses) Internal + - + - -

TLR4 LPS, proteins (G- bacteria, viruses) Surface - - + + +

TLR5 Flagellin (bacteria) Surface + - +/- + +

TLR6 Diacyl LpP, Zymosan (bacteria, fungi) Surface +/- + + +/- +

TLR7 ssRNA (virus) Internal +/- + - - -

TLR8 ssRNA (virus) Internal + - + - +

TLR9 CpG-rich DNA,(virus, protozoa) Internal - + - - +

TLR10 Nd Surface + + Nd - -

Table 2 TLR localization and ligands.

Abbreviations: mDC-myeloid DC, pDC-plasmacytoid DC, MoDC-monocyte-derived DCs, LpP-Lipoprotein,

PGN-Peptidoglycan, LPS-lipopolysaccharide, ds-double stranded, ss-single stranded, Nd-No data. References:

[67,68].

Signalling through TLRs

TLRs are membrane-spanning proteins with one intracellular signalling domain and one

amino terminal extracellular domain containing leucine-rich repeats, which are responsible for

binding the ligand. The signalling domain signals as a dimer and TLRs can form either

heterodimers or homodimers. For example, it has been shown that TLR2 can form

heterodimers with either TLR1 or TLR6 or form a homodimer and that the ligands for each

conformation can vary [69].

Page 21: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

21

The TLR signalling domain is homologous to the IL-1R signalling domain and is called

Toll/IL-1R (TIR) domain. In general TIR-TIR interaction with an adaptor protein initiates

signalling thus recruiting an IL-1-receptor-associated kinase (IRAK) family protein and the

TNF-receptor associated factor 6 (TRAF6). This complex activates Map kinases, which

eventually lead to the activation of transcription factors [70,71].

All TLRs except TLR3 signal through the adaptor protein myeloid-differentiation primary

response gene 88 (MyD88). TLR3 signals through the TIR-domain-containing adapter-

inducing IFN-β (TRIF) and TNF-receptor associated factor 3 (TRAF3) leading to activation

of the transcription factor IFN regulatory factor (IRF) 3 and secretion of type I IFNs [72]. In

addition to MyD88, TLR1, TLR2, and TLR6 can also signal through toll-interleukin-1

receptor domain containing adaptor protein (TIRAP). TLR4 is unique in that it can signal via

MyD88, TIRAP and TRIF.

Interestingly, signalling through the same adaptor protein can have different outcomes. Both

TLR7 and TLR9 signal through MyD88 and engage IRF7 which leads to secretion of type I

IFNs [73]. Conversely, when the MyD88 - dependent pathway is activated through TLR4 it

leads to IL-12p70 secretion [74] while when TLR4 engages the alternative pathway via TRIF,

it results in IRF3 activation and secretion of type I IFNs [72,75].

In addition, the production of type I IFNs can be induced by two different pathways; either

through IRF3 via TRAF3 or through IRF7 via TRAF6 [76]. TRAF3 was shown to be essential

for the induction of type I IFNs and IL-10. In addition, deficiency of TRAF3 augments

production of IL-12 because of defective IL-10 production [77]. Therefore, this may be an

additional way to create diverse responses. The impact of different TLR activation on

adaptive immune responses has been summarised in a review [74] (Figure 3).

Page 22: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

22

Figure 3. TLRs in the regulation of adaptive immunity.

Reprinted from Manicassamy S, Pulendran B. Modulation of adaptive immunity with Toll-like receptors. Semin

Immunol (2009), doi:10.1016/j.smim.2009.05.005 with permission from Elsevier.

Briefly, IL-12p70 is only weakly induced by TLR2 but strongly induced by TLR4, TLR5 and

TLR8. Conversely, TLR2 activation leads to IL-10 production. TLR4 ligation can lead to

either IL-12p70 or the release of type I IFNs [74]. Viral recognition by TLR3, TLR7 and

TLR9 induces the release type I IFNs by different pathways. Thus, activation of different

TLR2 complexes results in T-helper (Th) 2 or Treg skewing of the immune responses, while

activation of TLR3, TLR4, TLR5, TLR7, TLR8, and TLR9 can lead to a Th1-type of

response [74].

The outcome will be different depending on which TLR is engaged, and thereby a “custom-

made” default response mechanism is put in place to deal with different types of antigens.

This activation process is further complicated by the fact that these signalling pathways

Page 23: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

23

interact with the pathways of other PRRs. This interaction can skew the signalling pathways

in different directions as we will see in the next section.

C-type lectin receptors

Another group of PRRs are the C-type-lectin receptors (CLRs), which belong to a superfamily

of proteins recognising carbohydrate structures on glycosylated surface proteins of pathogens.

Some examples of CLRs are the mannose receptors, DC-SIGN, CLEC9A/DNGR1, Dectin-1

and, importantly, a specific marker of the pDC subset, the BDCA-2 molecule. One of the

functions of these receptors is to internalize antigens for processing and presentation to T

cells, as shown for DC-SIGN [78]. It has been proposed that ligation of CLRs alone might

lead to immunosuppression while ligation of the same CLR during a danger situation would

induce activation [79]. Although many CLRs can induce signalling on their own, some CLRs

act by modulating TLR signalling [80].

Nod-like receptors

A more recently discovered family that senses inflammation includes the Nod-like receptors

(NLRs). These receptors assemble upon activation into large cytoplasmic multiprotein

complexes called inflammasomes [81] that recruit inflammatory caspases and trigger their

activation [82]. In contrast to TLRs, NLRs exhibit the ability to activate caspase-1 that is

necessary for IL-1β production [83,84]. Inflammasome activation may also have a role in

initiating adaptive immunity [85]. It has also been suggested that another NLR, the NLRX1

would have a regulatory function [86] thus indicating additional possibilities of regulating

immune responses.

RIG-I-like receptors

A family of cytoplasmic receptors specific for viral nucleic acids is known as the retinoic

acid-inducible gene I (RIG-I)-like helicases. These receptors have been shown to induce type

Page 24: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

24

I IFNs by phosphorylation of IRF3 [87] through the action of a caspase recruitment domain

(CARD) [88]. RIG-I and the melanoma-differentiation-associated gene (MDA)5, a receptor

belonging to the same family, recognize single stranded RNA and double stranded RNA.

RIG-I responds to influenza-, paramyxoviruses and flaviviruses while MDA5 respond to

picorna viruses [89,90].

In summary, there is a wide variety of receptors that are able to recognize both exogenous

PAMPs and endogenous DAMPs. Some of them are specifically expressed on a given cell

type while expression of other receptors is less restricted. Thus, a myriad of possibilities to

fine tune the responses of APCs according to the specific stimulus are presented.

ANTIGEN PRESENTATION

It is well established that APCs need to transmit two signals in order to activate T cells [91]

and it was later suggested that a third signal is needed [92] which has now been widely

accepted. The three signals required to properly activate a CD4+ T-cell are:

1) Binding of the T-cell receptor to a HLA-II-antigen complex

2) Binding of co-stimulatory molecules

3) Cytokine signalling

Signal 1

The immunological definition of an individual’s “self” lies in the HLA molecules. These

molecules are the main determinants for the rejection of allogeneic grafts upon

transplantation. The cells of a transplant have an aberrant HLA expression, as compared to the

host, and the host will not consider it as self but treat it as foreign and attack. T cells can only

recognize an antigen when it is presented as peptide fragments in association with HLA

molecules.

Page 25: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

25

The classical HLA proteins are subdivided into class I and II. The HLA-I molecules are

expressed on all nucleated cells of the body. They have a peptide-binding groove that holds

fragments of about 8-10 amino acids. The distal part of the HLA molecule then binds to the

CD8 molecule present on T cells [93]. The expression of HLA-II molecules is restricted only

to professional APCs. The HLA-II molecules have a larger groove than the HLA-I and can

therefore hold larger fragments. The distal part of the HLA-II molecules bind CD4 present on

T cells [93].

In the steady state condition HLA-antigen complexes are constantly internalized by immature

APCs, but upon maturation the internalization stops and consequently the cell surface

becomes full of HLA-antigen complexes. This process can be regulated by the actions of

ubiquitin ligase that marks the β-chain of HLA molecules with ubiquitin. This mark induces

internalization of the complexes while ubiquitin ligase is down regulated upon activation of

DCs [94-97].

Antigen uptake

Professional APCs activate the adaptive branch of immunity. The first step in antigen

presentation is to internalize the pathogen, and this is performed by endocytosis. Endocytosis

is a process by which the cell takes up extracellular contents by budding of inwards forming

an endosome of the plasma membrane and internalizing it to the cytoplasm. Pinocytosis is a

form of endocytosis that is also referred to as “cell drinking” or internalization of soluble

products. Another form of endocytosis is “cell eating” or phagocytosis. Pinocytosis is

formation of vesicles seemingly at random that serves to sample the environment for soluble

antigen in a non-specific manner. Phagocytosis is a process that is defined as an actin-

dependent receptor-mediated uptake of particles larger than 0,5µm in diameter [98].

The receptors involved are cell-surface receptors, such as Fc receptors (FcR), complement

receptors (CR) or CLRs [99]. FcR and CR are called opsonic receptors because they

Page 26: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

26

recognize the particles to be phagocytosed only when opsonised i.e when the particles are

bound to antibodies or complement proteins. The zipper model suggests that FcγR binding of

the particle leads to receptor clustering that are sequentially engaged to the surface of the

particle, leading to local actin polymerisation. The polymerisation creates outwards extension

of the cytoskeleton called phagocytic cup [100].

FcR ligation leads to activation of tyrosine kinases, belonging to the Src and Syk families

[101]. They in turn lead to activation of small GTPases that are important for remodelling of

the cytoskeleton [102]. CR3-mediated phagocytosis is not dependent on tyrosine kinases and

seem to be mediated by a different family of small GTPases [103]. Phagocytosis mediated by

CLRs is not as well studied as phagocytosis mediated by FcR or CR. Dectin-1 mediated

phagocytosis seems to use different signalling pathways as it does not require Syk kinases

although it is dependent on tyrosine phosphorylation [104].

It is known that DCs lose their ability to take up antigen from the environment while

maturing, however this seems to be preceded by a brief period of migratory arrest and

enhanced uptake [105]. In line with this model, some evidence suggests that the ability to take

up antigen is regulated by activation of proteins downstream TLR signalling [106]. A clear

link between antigen processing and migration is the fact that gene knock outs of the invariant

chain, a crucial component for antigen processing, migrate faster than wild type DCs [107]. It

has also been shown that APCs become transiently paralyzed by microbial stimuli in order to

increase antigen sampling locally [108,109]. After the brief uptake period, maturation and

migration to lymph nodes will ensue. This enables the APC to carefully sample the

environment, process and select the antigen to be presented before setting of in search of

naïve T cells.

Page 27: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

27

Antigen processing and presentation

Presentation of endogenous peptide fragments of cytosolic proteins on HLA-I molecules is

performed by all nucleated cells of the body. The HLA-I molecules are aided by a chaperone

molecule called β2-microglobulin to increase stability of the complex. Because the HLA-I

processing pathway leads to the transport of cytosolic proteins to the cell surface, this process

is called the cytosolic pathway. The cytosolic proteins are taken up by the proteasome and

degraded to peptide fragments. These fragments are then transported to the endoplasmatic

reticulum through the transporter protein TAP. In the endoplasmatic reticulum the peptide

fragments from the proteasome are then loaded on the HLA-I molecule and follow the normal

secretory pathway through the Golgi to reach the cell surface.

In contrast to the cytosolic pathway, professional APCs use the endocytic pathway to process

particles that are taken up from the extracellular environment and present peptide fragments

on HLA-II molecules. In APCs, extracellular components are transported into the cell through

endosomes therefore; this pathway is called the endocytic pathway. When an antigen is taken

up through an endosome the endosome is fused with lysosomes containing digestive enzymes

in an acidic environment. This endolysosome is where the pathogen is degraded by proteases.

Meanwhile, an HLA-II molecule that is contained in the endoplasmic reticulum bound to a

chaperone molecule called the invariant chain travels to the lysosome. In the lysosome the

chaperone is digested by proteases leaving only a fragment called class II-associated invariant

chain peptides (CLIP) in the peptide groove. The CLIP fragment is then replaced by a

selected peptide fragment from the antigen on the HLA-II molecule. The HLA-II-antigen

complex can then be transported to the APC surface where it is ready for encountering T cells

[110,111].

Proteolysis is an important step in antigen processing and it is believed that excessive protease

activity might destroy the antigen while moderate proteolysis may lead to enhanced antigen

Page 28: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

28

presentation. It has been shown that an antigen that is more resistant to proteolysis induces

more potent T-cell responses than a protease-sensitive antigen [112]. Importantly, the levels

of MHC-II complexes on mature DCs are 10-100 fold higher than on monocytes or B cells

emphasising the important role of DCs in antigen presentation [113].

In line with the notion that DCs are the most potent APCs, it has been demonstrated that DCs

exhibit lower levels of protease activity than MΦs [114]. In addition, it has been shown that

DC phagosomes become transiently alkaline thus preserving the antigen from protease

activity and enhancing antigen presentation, particularly in cross presentation (see below)

[115].

Cross-presentation

Some APCs are capable of processing antigen through an alternative pathway that enables

them to present exogenous antigens on HLA-I molecules. This phenomenon is restricted to

particular subsets of professional APCs that induce antigen-specific CD8+ T cells and is

known as cross-presentation or cross-priming [116]. Cross-priming is induced by receptors

that recognize viruses, in particular TLR9 [117,118]. It has been shown that viral infection

can lead to cross-presentation by DCs and activation of CD8+ T cells without T-cell contact

but dependent on type I IFN production [119].

There is no consensus on the precise mechanism of cross-presentation and different pathways

have been proposed. The phagosomes and endosomes of DCs are constituted by the

endoplasmatic reticulum membrane. This may enable exogenous protein transportation into

the cytosol where proteins could go through the proteasome and TAP, back into the

endosomes and then be loaded on HLA-I molecules as any endogenous proteins are

[120,121]. This has been proposed to be TAP-dependent through the same mechanism that

dislocates misfolded proteins from the endoplasmic reticulum to be degraded [122]. However,

TAP-independent pathways have also been suggested to explain cross-priming [123].

Page 29: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

29

Finally, it has been proposed that DCs migrating to lymph nodes to activate CD4+ T cells can

transfer exogenous antigens to resident DC populations in lymph nodes and that these cells

activate CD8+ T cells [124,125]. This hypothesis is strengthened by the fact that splenic DCs

can cross-present antigens more efficiently than other DC subsets [126].

Signal 2

The expression of co-stimulatory molecules on APCs is fundamental to their function since

they are the providers of signal two needed for the activation of T cells. APCs express a

variety of co-stimulatory molecules that constitute what is called the immunological synapse.

Two important co-stimulatory molecules are the members of the B7-family, CD80/B7.1 and

CD86/B7.2 that bind CD152/CTLA4 and CD28 on T cells. Such binding can mediate

activating or inhibitory signals [127]. For example, ligation of CD28 by CD80 and CD86

leads to T-cell activation while ligation of CD152 suppresses activating signalling in T cells

[128]. Furthermore, evidence indicates that priming of T cells without proper co-stimulation

[129,130] or in the absence of activating cytokines will lead to their differentiation into Tregs

or anergic T cells [131].

Two members of the TNFR family are also known to affect T-cell responses, CD134/OX40

and CD137/4-IBB. CD4+ T cells are more prone to secrete Th2 cytokines if the APC that

activates them express high amounts of OX40L that binds OX40 on activated T cells [132]. If

an APC expresses 4-1BBL, they preferentially induce activation of CD8+ cells and secretion

of IFN-γ [133]. In addition, it has been shown that OX40 can antagonise the induction of

peripherally induced Tregs while both OX40 and 4-1BBL can support proliferation of

naturally occurring Tregs [134].

Another important molecule for co-stimulation is the ICOS. Ligand binding to ICOS has been

shown to be important for triggering T cells as potent activators of B cells [135]. In addition,

Page 30: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

30

ICOS seems to be able to mediate suppression since ligand-binding to ICOS is important in

generating Tregs [136].

Signal 3

After it had been established that signals 1 and 2 are necessary for the activation of naïve T

cells, it soon became clear that a third signal was also needed for the triggering of naïve T

cells into potent effector cells [92]. This signal is constituted by the cytokines secreted by

APCs and varies depending on the manner of activation of the APC. It is now known that

each defined Th lineage has specific requirements for their generation. For example, Th1 cells

can only be induced if IL-12 is secreted while IL-4 is essential to establish a Th2 lineage.

Cytokines

Cytokines are soluble mediators of inflammation secreted by leucocytes and epithelial cells.

As they serve to communicate between leucocytes they are called interleukins (ILs).

Traditionally they have been given names according to the function they were discovered for

but as they are sequenced they are renamed to interleukins. Some key cytokines will be

introduced below and their function will be further discussed when discussing inflammation.

IL-1β

IL-1β is a potent pro-inflammatory cytokine and a powerful pyrogenic factor that does not

seem to play a role in homeostasis. IL-1β induces increased expression of adhesion molecules

on the endothelium at a site of inflammation and acts as a bone marrow stimulant increasing

the number of circulating neutrophils [137]. Monocytes, MΦs and DCs produce IL-1β in

response to inflammation. It is produced as pro-IL-1β and needs to be cleaved in order to

become the active from of IL-1β [138]. In this context, it has recently been shown that

Page 31: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

31

inflammasomes play a vital role in the induction of caspase-1 leading to the subsequent

realease of IL-1β [81].

TNF-α

TNF-α is a member of the TNF super family of proteins, which was initially discovered for

killing of tumour cells, and acts as a potent pro-inflammatory cytokine. TNF-α can trigger

apoptosis via the TNF receptor that activates pro-caspases thus inducing cell death. This

cytokine is crucial in infectious diseases as it efficiently promotes inflammation and helps to

combat the pathogen.

TNF-α is a central player in inflamed tissues in the early phase of inflammation, where it is

known to induce the production of many other pro-inflammatory cytokines [139]. However,

high levels of this cytokine are responsible for tissue damage and pathogenicity [140]. In the

late phase of inflammation, TNF-α can induce an IRF1-dependent autocrine loop of type I

IFN secretion, thus sustaining an inflammatory response and lowering the threshold of TLR

responses [141].

Type I IFNs

These cytokines were named interferons over 50 years ago because they were discovered to

interfere with viral replication [142]. They are found in all vertebrates where they have been

investigated and many subgroups have been discovered [143]. Although there are about 20

different type I IFNs, IFN-α and IFN-β are the classical antiviral IFNs. Type I IFNs can be

produced by a wide variety of cells. Viral recognition triggers transcription of type I IFNs

which when bound to their receptors trigger the JAK/STAT pathway leading to transcription

of IFN stimulated genes (ISG). The most potent producers of IFN-α are pDCs and NK cells as

a response to viral infection. IFN-α acts on immune cells to elicit anti-viral responses and also

functions as an analgesic.

Page 32: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

32

The triggering of RIG-I, MDA5, TLR3, TLR7, and TLR9 activates the transcription factors

AP1, NF-kB, IRF3 and IRF7 leading to secretion of type I IFNs [144]. It has been suggested

that DC secretion of type I IFNs can act in an autocrine manner, thus enhancing activation of

DCs. This was supported by the fact that DCs from type I IFN knockout mice exhibited an

impaired phenotype and were less potent activators of T cells [145]. Although it seems clear

that type I IFNs can play an important role in DC maturation they do not seem to be able to

induce potent maturation on their own [146,147].

IL-6

Upon its discovery this cytokine was called B-cell stimulatory factor-2, but it was renamed as

an interleukin as the cDNA was sequenced [148]. Pro-inflammatory cytokines such as TNF-α

and IL-1β induce IL-6, while its secretion is inhibited by the Th2 cytokines IL-4 and IL-13.

Nevertheless, this cytokine can skew CD4+ T cells to become IL-4 secreting cells and it has

therefore been regarded as a Th2 cytokine [149]. This effect was shown to be induced by

disruption of the autocrine IFN-γ loop that leads to Th1 activation through induction of

SOCS1 [150]. It has now become clear that IL-6 is a key factor in the induction of the

transcription factor RORγt and in the differentiation of naïve T cells to the Th17 lineage

effector cells [151]. Recently, it has been suggested that IL6 is an important regulator of the

balance between Th17/Tregs [152,153].

IL-12 family

IL-12 is the classical Th1-inducing cytokine secreted by neutrophils, monocytes, MΦs and

DCs and induces IFN-γ release in NK cells and T cells. IL-12 was the first member of the IL-

12 family of interleukins discovered and initially called NK-cell stimulatory factor [154].

More recently, IL-23 [155] and IL-27 [156] were added to the IL-12 family. All three are

heterodimers and IL-12 and IL-23 share the p40 subunit. IL-12 is composed of the p40 and

Page 33: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

33

the p35 subunits while IL-23 is composed by the p40 and the p19 subunit. Similarly, the

corresponding receptors are composed by two chains and both IL-12 and IL-23 receptors

share IL-12Rβ1 chain, while the second chain is different for each receptor. In this way the

function these cytokines exert by stimulating the cognate receptors is different.

For instance, IL-23 creates autoimmune inflammation in a mouse model for multiple sclerosis

while IL-12 does not [157]. Secretion of the IL-12 family cytokines can be differentially

regulated depending on the nature of the microbial stimuli. This is evidenced by the fact that

LPS can induce both IL-12 and IL-23 release by DCs whereas peptidoglycan induces IL-23

but not IL-12 secretion [158].

IFN-γ

IFN-γ is an important and well-studied cytokine that mediates strong pro-inflammatory

responses. It was originally termed macrophage-activating factor and is the only member of

the type two IFN class. IFN-γ secretion is induced in activated T cells and NK cells by IL-12

and IL-23 and release of this mediator leads to a classical Th1 response while supressing Th2

responses [159]. NK and T cells produce IFN-γ and release generally follows in response to

virus, intracellular bacteria or tumor cells [160].

IL-4 & IL-13

IL-4 is secreted by granulocytes and various T-cell subsets [161] and represents the hallmark

cytokine for Th2 responses. This mediator was originally known as the B-cell differentiation

factor [162]. IL-13 is another Th2 cytokine with very similar effects to IL-4 but seems to be

more important for immunity against certain infections such as different species of

Leishmania [163]. The IL-4 receptor α-chain can bind both IL-4 and IL-13. However different

receptor complexes can be composed by different subunits enabling both cytokines to mediate

different responses [164].

Page 34: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

34

IL-10

Perhaps the most important cytokine that regulates immune responses is IL-10. It was

originally known as “cytokine synthesis inhibitory factor” because it was shown to inhibit

Th1 cells and was therefore regarded as a Th2 cytokine [165]. It is now known that IL-10 can

be secreted by T cells, B cells, DCs and MΦs during an infection. The inhibition of IL-1 and

TNF-α is a very important anti-inflammatory effect of IL-10, while it also inhibits the

production of many other cytokines, even its own production [86].

The importance of IL-10 as a regulator of immune responses was evaluated in IL-10 deficient

mice. These mice remained healthy until their gut was colonized by bacteria. After

colonization, the animals developed chronic enterocolitis [166] indicating that the immune

responses were not properly regulated. The potency of IL-10 is emphasized by the fact that

some viruses, such as Epstein Barr and human cytomegalovirus, encode an IL-10 homologue

that is released in order to escape immune responses [167].

TGF-β

Another soluble mediator that is a key factor for regulating immune responses is the

transforming growth factor (TGF)-β. There are various isoforms of TGF-β and they are all

members of the TGF-β superfamily. They are secreted in an inactive form often called the

latent TGF-β and it has been suggested that the regulation of activation of the latent form is

the most potent form of regulation of TGF-β [168]. TGF-β is a pleiotropic cytokine that is of

outmost importance for preserving homeostasis. This is emphasized by the fact that mice

lacking the TGF-β1 protein die prematurely of excessive inflammatory tissue damage [169].

APC-derived cytokines in T-cell differentiation

IL-12 secretion by DCs and MΦs is tightly linked to the induction of IFN-γ in naïve T cells

and commitment of these cells to the Th1 lineage [170,171]. IL-12 priming of T cells also

Page 35: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

35

impairs their capability of secreting IL-4 [172], thus further favouring Th1-type responses

rather than Th2. Microbial stimuli such as LPS and Toxoplasma gondii are required and

sufficient for the production of IL-12 by DCs and to initiate their migration to T-cell areas of

the spleen [173,174].

In addition, IL-12 and IFN-γ production is known to be important for the protection against

intracellular infections like mycobacteria and genetic defects in the Th1 axis leads to

susceptibility to disease [175]. Conversely, IL-23 does not induce Th1 responses while it

plays an important role in the protection against extracellular bacteria, such as Klebsiella

pneumoniae [176]. It has now been shown that this is an effect of the induction of Th17

responses [177-180].

INFLAMMATION

Upon sterile or pathogen-mediated tissue damage, necrotic and surrounding cells secrete

inflammatory mediators that in turn recruit and activate immune cells. The subsequent release

of inflammatory mediators leads to an increased permeability of the surrounding vessels, thus

increasing the blood flow at the site of inflammation. Simultaneously, circulating immune

cells start to adhere to the vessel walls as a consequence of increased expression of adhesion

molecules such as selectins and integrins on endothelial cells. The selectins induce rolling of

blood cells on the vessel wall and the integrins mediate transendothelial migration.

Granulocytes, that are normally the first immune cells that are recruited at the site of

inflammation, release their granules containing toxic compounds and phagocytosis of

microbes is induced, leading to their own death. The granulocytes also release proteases that

will start to liquefy the foci of inflammation thus inducing the formation of pus. Monocytes

will also phagocytose pathogens and simultaneously process antigens for subsequent antigen

presentation. Similarly, tissue specific APCs such as DCs that are activated by pathogens and

Page 36: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

36

inflammatory mediators, will carry antigens to the lymph nodes in order to activate

appropriate T cells.

In the early phases of inflammation the proinflammatory cytokines secreted such as IL-1, IL-6

and TNF-α serve to increase inflammation and temperature. Later on, the secretion of soluble

factors will be skewed to regulatory cytokines, such as IL-10, in order to decrease

inflammation. In the normal situation the pathogen is removed, the inflammation resolved,

and homeostasis restored. However, chronic inflammation may persist that can lead to severe

tissue damage.

Leukocyte movement

Inflammation implies recruiting leukocytes to the site of injury or infection. In order for cells

to find their final destination, signalling molecules are secreted that guide the movement of

leukocytes. These molecules are small proteins called chemokines. The name derives from

their ability to induce chemotaxis i.e cell movement towards a gradient of a certain substance.

The chemokines are classified according to structural properties and the C symbolises the

location of highly conserved cystein residues in the peptide chain of chemokines. Chemokines

are divided into two major subgroups; the CC and the CXC [181]. More recently, two smaller

subgroups have been added; i.e. the C and the CX3C subgroups [182]. The chemokine

receptors (CCRs) are coupled to a G protein for intracellular signalling [183] that contain a

characteristic seven transmembrane domain.

Inflammatory CCRs

Inflammatory CCRs often have various ligands and many of them are shared (Table 3). This

is the case for CCR1, CCR2 and CCR5 that typically bind inflammatory mediators such as

macrophage inflammatory protein (MIP)-1α and MIP-1β, Monocytes-Chemoattractant

Protein (MCP)1-4 and Regulated on Activation Normal T Expressed and Secreted Protein

Page 37: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

37

(RANTES) [184]. Cells expressing any of these CCRs will sense inflammatory chemokines

and start to move towards the site of inflammation.

Lymphoid CCRs

In contrast to the inflammatory CCRs, the lymphoid CCRs such as CXCR4 and CCR7, do not

have many known ligands. CCR7 is expressed on naive T cells and it has been shown that

lack of this receptor severely impairs primary immune responses [185]. Both T and B

lymphocytes express CXCR4 and this receptor has a dramatic effect on the development of B

cells [186]. A ligand for CXCR4 is stromal derived factor (SDF)1/CXCL12, while MIP-

3β/CCL19 and secondary lymphoid tissue chemokine (SLC)/CCL21 bind to CCR7 and are

mainly expressed in lymphoid tissues. Both CCR7 and CXCR4 are highly expressed on

mature DCs, allowing their migration to secondary lymphoid organs [36].

Receptor Ligand Location iDC mDC

CCR1 CCL3/MIP-1α, CCL5/RANTES Inflammation + -

CCR2 MCP-1-4 Inflammation + -

CCR5 MIP-1α, CCL4/MIP-1β, CCL5/RANTES Inflammation + -

CCR7 CCL19/ELC/MIP-3β, CCL21/SLC Lymphatic tissue - +

CXCR4 CXCL12/SDF-1α Lymphatic tissue + + Table 3. A few examples of CCRs expressed on immature and mature DCs and some of their ligands. Abbreviations: CCL-Chemokine ligand, iDC-immature DC, mDC-mature DC, MCP-monocyte chemoattractant

protein, MIP-Macrophage inflammatory protein, SDF-stromal derived factor . References: [182,184]

Page 38: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

38

RELATED BACKGROUND

APCs IN INFLAMMATION AND THE LINK TO ADAPTIVE IMMUNITY

Inflammation is the trigger that activates APCs through a variety of different receptors. The

ligation of PAMPs or DAMPs to APC receptors triggers intracellular activation signals. The

sum of these signals decides what action the APC will take.

Antigen presentation is the act that links pathogen recognition by innate immune cells to the

initiation of the adaptive immune responses. Professional APCs are all capable of activating

cells of adaptive immunity. However, different APC subsets can be triggered by distinct

signals and have become specialized in activation of specific branches of the adaptive system.

So far, the mechanisms that orchestrate the activation of adaptive responses are only partially

clarified.

Monocytes

As discussed earlier, human monocytes in circulation are commonly divided into the classical

CD14++

CD16- and the non-classical CD14

+CD16

+ monocytes. The CD14

+CD16

++ subset

expresses higher levels of HLA-DR and was shown to produce higher levels of TNF-α [187]

but lower levels of IL-10 [188] as compared to classical monocytes. Their murine

counterparts have been defined with many similarities. Perhaps the major difference is that in

humans the non-classical represent only a small part of the circulating monocytes while in

mice the subsets are equally distributed [189]. The CD16+ subset is more potent in inducing

T-cell responses in a mixed leukocyte reaction (MLR) than the CD16- monocytes [190].

Monocytes are precursors of MΦs and can give rise to inflammatory [191] and anti-

inflammatory MΦs [192] and to inflammatory DCs [46]. The differentiation of monocytes

seems to be differently regulated during homeostasis and under inflammatory conditions.

Monocytes do not seem to replenish MΦ in the lung [193], or in the spleen, DCs [194] under

Page 39: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

39

steady state conditions. Monocytes can, however, be an important source of DC during

inflammation [10,49,195]. In this context, it has also been shown that the non-classical

monocytes are more prone to trans-endothelial cell migration and differentiation into

inflammatory DCs [196].

Macrophages

Classical activation of MΦs, induced by IFN-γ, leads to differentiation of M1-MΦs that

secrete high levels of IL-12, the most potent Th1 inducing cytokine, and low levels of the

anti-inflammatory protein IL-10. In contrast, MΦ activation by IL-4 and IL-13 leads to M2

polarization and secretion of high levels of IL-10 and low IL-12. In addition, upon stimulation

with IFN-γ and CD40 ligand (CD40L), M1-MΦs secrete high levels of pro-inflammatory

mediators including IL-1β and TNF- while M2-MΦs secrete low levels of these cytokines

but high levels of IL-10. M2-MΦs do, however, secrete higher levels of pro-inflammatory

chemokines perhaps indicating a role in recruitment of other cells and in the regulation of

homeostasis [197].

The roles of M1-MΦs in promoting inflammation and Th1 differentiation are emphasized by

the fact that IFN-γ also enhances the expression of TLR4 and downstream signalling proteins

such as MyD88 [198]. In contrast, the Th2 cytokine IL-13 inhibits LPS-induced caspase-1

activity and modulates gene expression of various genes in the IL-1β system [199] indicating

a dampening function on inflammatory responses.

Selective depletion of MΦs at different stages of inflammation in a model of liver injury has

shown that these cells are important both during the injury and the recovery phase [200].

Specifically, it has been suggested that classical MΦs are involved in tissue degradation that

lead to injury [201] while alternative activation of MΦs promote tissue healing [202].

Generally, alternatively activated MΦs are thought to play an important role in the

maintenance of homeostasis. The fact that the receptors that are upregulated during alternative

Page 40: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

40

activation have various endogenous ligands has been used as an argument to strengthen this

view [203].

Dendritic cells

DCs are well known to become potently activated by microbial stimuli which lead to

maturation, antigen processing and activation of naïve T cells, thus initiating adaptive

immune responses [91]. This could lead to activation of distinct T-helper subsets such as the

previously described Th1, Th2 and Th17 [2,3][204]. Finally, it is becoming clear that DCs

also play an important role in the induction and maintenance of tolerance through the

induction and maintenance of regulatory leukocytes [205,206]. These different pathways of

initiation of adaptive immunity are highly dependent on cytokines.

Myeloid DCs

Myeloid DCs express a broad repertoire of PRRs and respond to bacterial, parasitic, fungal

and viral infections. They also have the capacity to tailor inflammatory and T-cell responses

depending on the nature of the antigen [207]. For example, upon LPS stimulation DCs up

regulate maturation markers and secrete high levels of IL-12, which is known to induce Th1

responses [208]. The secretion of IL-12 by DCs not only initiates Th1 responses but is also

critical for generating IgM-secreting plasma cells [209]. In addition, DC-derived IL-12 can

induce class switching to IgG and IgA [210].

Although the specific characteristics of circulating mDC subsets are being investigated their

different roles are not yet completely clarified. BDCA-3+ DCs have been shown to express

lower levels of HLA-DR and co-stimulatory molecules than the BDCA-1+ subset, indicating

lower levels of activation [39]. In fact, a potential immunomodulatory role for the BDCA-3+

subset has been suggested during malaria infection [211].

Page 41: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

41

In addition, BDCA-3+ DCs of sheep, mice and humans express XCR1 and selectively respond

to the ligand XCL1 expressed preferentially in CD8+ memory T cells [212]. These findings

support the view that these cells may be the human counterpart of the murine DCs devoted to

cross-presentation of viral antigens to CD8+ T cells. In line with this hypothesis, it was shown

that XCR1 knock outs have impaired early CD8+ T-cell responses [213].

Plasmacytoid DCs

The pDCs are specialized to handle viral infections and have several features that enable them

to respond quickly and potently to viruses. They were originally called natural IFN-producing

cells since they release vast amounts of type I IFNs upon recognition of nucleic acids [214].

The pDCs seem to play a crucial role in humoral immunity; high levels of type I IFN

secretion by pDCs can induce plasma cell differentiation and Ig production [215,216] and can

also induce class switching of B cells [217]. Furthermore, pDCs possess intracellular

endosomes containing HLA-I molecules that are quickly mobilized, loaded with antigen and

sent to the surface to activate CD8+ T cells upon viral infection, thus contributing to the

development of an efficient effector response [218].

The pDCs are also able to induce both Th1 and Th2 responses in vitro. It was shown that

virus-stimulated pDCs secreted high levels of IFN-α and lead to Th1 responses while IL-3-

stimulated pDCs upregulated OX40L, secreted low levels of IFN-α and led to Th2 phenotype

[219].

Tolerogenic DCs

As the most important initiators of adaptive immunity, DCs must not only be able to activate

potent inflammatory responses, but should also be able to induce tolerance. DCs that travel

the periphery sample the environment and constantly present self-derived and harmless

environmental peptides on their HLA-II molecules. These cells can, as long as they remain in

Page 42: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

42

an immature state, maintain peripheral tolerance [205] indeed antigen presentation in the

absence of co-stimulation can lead to T-cell anergy [220].

Apoptotic cells display phosphatidylserine on their surface, which is recognized by various

receptors on APCs and can lead to uptake of apoptotic bodies by these cells [221]. The uptake

of material from apoptotic cells does lead to antigen presentation but not maturation of DCs

[222,223]. DCs migrate to local lymph nodes during steady state conditions to present self-

antigen derived from apoptotic cells to T cells without inducing their activation [224,225].

The phenotypic characterization of tolerogenic DCs is still not well defined, however ILT3,

ILT4 and IDO may be surface indicators of tolerogenic DCs [226-228].

Endogenous ligands

As we have seen, APCs and other hematopoietic cells interact intimately to decide whether or

not to respond to a specific microbe/antigen and what type of responses to induce. However,

non-hematopoietoc cells also play a role in this context. One example is the mucosal surfaces,

which constitute the interface between the outside world. For instance, epithelial cells of the

intestine express various TLRs and can secrete soluble factors that participate in inflammatory

responses [229]. Furthermore, under homeostatic conditions, intestinal cells [230-232] as well

as liver parenchyma [233] maintains a DC population that is tolerogenic [234].

Two important factors for induction of tissue tolerance are thymic stromal lymphopoietin

(TSLP) and transforming growth factor (TGF)-β [235]. TSLP induces a mature phenotype on

DCs, with up regulation of HLA-II and co-stimulatory molecules but no secretion of

inflammatory cytokines [236].

There is no doubt that non-hematpoietic cells participate in inflammatory reactions but their

role is not entirely understood. On the one hand, epithelial cells have been shown in vitro to

be necessary for potent antiviral Th1 responses [237]. On the other hand, it has been shown

Page 43: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

43

that tissue responses to LPS are capable of inducing inflammation but not DC activation and

therefore these responses are not sufficient to trigger adaptive immunity [238].

In addition, heat-shock proteins (HSPs) are endogenous ligands that can act as trigger for

inflammation and the most studied is a family called molecular chaperones. Chaperones are

involved in folding of proteins but are also released during stress conditions and have

important stimulatory functions on innate immunity [239].

High mobility group box 1 (HMGB1) is also an important mediator of inflammation. This

nuclear DNA-binding protein is released upon necrotic cell death and is known to be involved

in both sepsis and autoimmune diseases. HMGB1 can bind to various receptors, such as

TLR4, involved in inflammatory responses [240].

The manipulation of APCs by cytokines

Different pathways of initiation of adaptive immunity are highly dependent on APC

differentiation and functionality which is in turn affected by various cytokines.

It is well established that DCs are capable of secreting high amounts of pro-inflammatory Th1

cytokines like IL-12 and type I IFNs and that they are essential in orchestrating adaptive T

cell responses. However, the effect of these cytokines on APCs can have profound effects on

the development on immunity.

For instance, if immature DCs are exposed to TNF-α they become phenotypically mature

expressing high levels of HLA and co-stimulatory molecules but they do not secrete IL-12

[241]. In fact, they can even induce IL-10 secreting antigen-specific T cells that protect from

autoimmune disease [242,243]. In the presence of fibroblasts TNF-α favors the differentiation

of monocytes into DCs rather than MΦs as well as induce maturation of pDCs and mDCs

[244,245].

There are other important inflammatory mediators that can affect the differentiation and

functionality of APCs. Fibroblast secretion of IL-6 is capable of skewing monocyte

Page 44: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

44

differentiation from DCs to MΦs. This cytokine also impairs migration of mature MoDCs and

inhibits DC maturation and subsequent T-cell activation [246,247]. IL-6 may therefore play a

role in the specific differentiation and function of APCs [248].

A crucial cytokine for immune regulation is IL-10 and a well-recognized anti-inflammatory

effect of IL-10 is to dampen APC functions. For example, a major role is to contain

inflammation and this is achieved by preventing MΦs from killing and by inhibiting cytokine

secretion by MΦ and DCs [86]. In fact, DCs that are matured in the presence of IL-10 exhibit

reduced expression of HLA-II, co-stimulatory molecules and impaired secretion of

inflammatory cytokines [220,249-251]. In addition, DCs matured in the presence of IL-10

induce suppressive T cells that do not require IL-10 or TGF-β to exert their suppressive

function [252], and are therefore different from the Th3 and Tr1 subsets.

MALARIA

Malaria burden

Malaria is one of the most devastating infectious diseases in the world and it is tightly

connected to poverty [253,254]. According to the WHO malaria report form 2010, there were

225 million cases of malaria reported in 2009 as compared to 233 million cases in the year

2000. Though the number of cases has varied during this time period the number of deaths has

been steadily decreasing to781 thousand from 985 thousand during the same time period. In

Africa, 85% of the deaths occur in children below the age of 5 and about 98% of the cases are

caused by Plasmodium falciparum [255,256]. Pregnancy associated malaria is another aspect

of the infection that defines pregnant women as an important risk group for malaria infection

[257].

Page 45: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

45

Parasite life cycle

The parasite is transmitted from the definitive host, mosquitoes of the genus Anopheles, to

intermediate hosts such as reptiles, birds and mammals. The four “classical” species infecting

humans are Plasmodium falciparum, vivax, malariae, ovale. A fifth species, Plasmodium

knowlesi, was known to infect macaques but has also been reported to infect humans [258-

264]. The sporozoites are transmitted to humans from the salivary glands of the infected

mosquito (Figure 4).

Figure 4. The Malaria Life cycle

Reprinted from. Ménard, Robert, 2005: Knockout malaria vaccine? (Nature 433, 113-114). with permission from

Nature Publishing Group

The sporozoites immediately infect the liver upon entering the blood stream and develop into

merozoites. The merozoites leave the liver and infect-red blood cells (RBCs), where they

undergo replication. This is the blood stage of the infection and in the RBCs the parasites

develop from ring stage, to trophozoites and finally schizonts. Thousands of new merozoites

are released into the blood stream at the rupture of each schizont. Among the merozoites there

Page 46: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

46

is a small portion of gametocytes released that are necessary for sexual stages in the mosquito

gut.

Hemozoin

The free heme groups that are left upon degradation of haemoglobin are toxic to the parasites.

Therefore, they are transformed into malarial pigment or hemozoin (Hz) that are innocuous

crystals [265]. APCs take up the Hz released by the parasites. In order to create in vitro

models of malaria stimulation, hemozoin crystals can be synthetically formed from hemin

[265] and the product is usually referred to as β-hematin or synthetic (s)Hz. It has been shown

that sHz induces recruitment of leukocytes, mainly monocytes and neutrophil populations,

and trigger potent pro inflammatory responses [266-268]. The immunogenicity of Hz in mice

and in humans will be discussed in the next section.

Innate Immune responses to malaria

MyD88, a central adaptor protein in TLR signalling, has been suggested to play an important

role in the pathogenesis of the malaria. Different parasite-derived or modified molecules have

been shown to bind TLRs and elicit innate immune responses [269]. For example

glycosylphosphatidylinositol anchors (GPIs) of protozoan parasites has been shown to bind

TLR2 [270] and TLR4 [271].

It has also been suggested that Hz activates cells through TLR9 [272]. It was then

demonstrated that Hz functions as a carrier of malarial DNA to the endolysosomes and that

malarial DNA attached to Hz is responsible for TLR9 activation [273]. More recently, it was

postulated that it is not Hz, but rather a protein-DNA complex, later discovered to be the

nucleosome, that triggers TLR9 activation [274,275].

Polymorphism in the TLR4 gene predisposes to the severe form of malaria in children [276]

and polymorphisms in TLR4 and TLR9 genes are associated with increased risk of low birth

Page 47: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

47

weight in the offspring of pregnant women infected with malaria [277]. These findings

suggest that TLRs contribute in determining inflammatory responses upon malaria infection.

In this context it has been shown that secretion of pro- and anti- inflammatory cytokines is

increased upon specific TLR stimulation of whole blood that was primed by malaria antigens

[278].

Taken together, these studies indicate that P. falciparum modulates TLR activity and that

innate immunity, in particular TLRs, plays an important role in the responses against malaria.

Antigen-presenting cells in malaria

APCs are crucial initiators of adaptive immunity, express high amounts of TLRs and may be

fundamental in host responses to malaria. In fact, DC-mediated suppression of T-cell

functionality has been suggested to be the cause of the poor T-cell mediated immune

responses seen in malaria [279,280]. Observations in vivo have shown that children with acute

malaria have a lower expression of HLA-DR on peripheral blood DCs as compared to healthy

children [281]. Moreover, a possible immunomodulatory role has been suggested for the

BDCA-3+ subset of mDCs upon malaria infection in Kenyan children [211].

Observations in vitro have shown that APCs exposed to Hz increase both pro- and anti-

inflammatory mediators as reviewed in [282]. In addition, Hz can mediate activation of pDCs

via TLR9, although it is unclear whether the stimulus that activates these cells is Hz itself,

parasite DNA or both in a complex [272,273,283]. As mentioned above, more recent studies

suggest that TLR9 is bound by a histone-malarial DNA complex [274,275].

Whether or not Hz is responsible for activation of TLR9 there are documented immunological

effects of Hz. Hz-loaded monocytes have been shown to secrete higher levels of TNF-α and

IL-1β [284] than unstimulated cells, but also lower levels of IL-12p70 by an IL-10-dependent

mechanism [285]. Monocytes also exhibit inhibited differentiation to DCs [286] as well as

impaired up-regulation of HLA-II molecules when exposed to Hz [287]. LPS-induced

Page 48: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

48

maturation of human MoDCs was shown to be impaired upon prior exposure to high amounts

of P.falciparum-infected red blood cells (iRBCs) [288]. However, when immature DCs were

exposed to parasite-derived Hz, enhanced maturation was observed [289].

Thus, both pro- and anti- inflammatory effects have been attributed to malarial stimuli when

in contact with APCs. Many factors such as the heterogeneity of APCs used, different parasite

strains, different types of Hz employed and the time of exposure of malarial stimuli to APCs

could explain these discrepancies [290,291]. Taken together, existing data suggest that the

function of various APC subsets is influenced by P.falciparum and that immune responses to

malaria may be hampered by the modulation of APC functions.

Cytokines and clinical implications in malaria

For a successful elimination of parasites it is important to have an early innate response

including secretion of pro-inflammatory cytokines like IL-12 and TNF-α. For optimal

clearance of the parasite, that initial response should be followed by an effective adaptive

immune response including high titres of antimalarial antibodies to clear the parasitemia. On

the other hand, pro-inflammatory cytokines may also play an important role in determining

pathology, in particular in the development of severe malaria. In fact, low levels of IL-10 and

high levels of TNF-α have been associated with disease severity and in severe cases in

Gabonese children IL-12 was found to be lower as compared to the mild cases [292]. In

addition, the development of cerebral malaria has been associated with high plasma levels of

IL-1β [293] and higher levels of IL-6 and IL-10 were associated with severe malaria as

compared to uncomplicated malaria [294].

Fulani and Dogon in northern Mali

The study area in Sahel, northern Mali, lies in Dogon country but in this particular area the

Dogon farmers settled down only 50 years ago. The Fulani, a traditionally nomadic pastoral

Page 49: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

49

people, settled in this area at least 200 years ago with their cattle. The Fulani have been shown

to be less susceptible to malaria than their sympatric counterparts i.e. other ethnicities living

under similar geographical, economic and social conditions [295]. In addition to the relative

protection of symptomatic disease, the Fulani have higher proportions of malaria specific IL-4

and IFN-γ producing cells as compared to their sympatric neighbours. They also have

increased spleen rates and exhibit higher malaria-specific antibody titres [295-298]. Recently,

a lower Treg activity has been shown in a population of Fulani in Burkina Faso as compared

to a neighbouring sympatric group [299]. So far, no satisfactory explanation for the relative

protection to malaria infection of the Fulani has yet been found.

REUMATHOID ARTHRITIS

Reumathoid arthritis (RA) is a chronic inflammatory autoimmune disease of the joints.

Approximately 1% of the world’s population is affected and RA is more common in the

female population [300].

Clinically, RA is characterized by joint pain, stiffness and swelling due to synovial

inflammation and effusion. The joint is composed by the two cartilage covered bone ends

separated by synovial fluid and lined by the synovial membrane. In the healthy state there are

synoviocytes sitting on the membrane lining the joints. In RA, for yet unknown reasons, the

joint is infiltrated by leukocytes and the synoviocytes start to proliferate [301] (Figure 5). In

contrast to other forms of arthritis, RA synovitis tends to disobey tissue boundaries,

infiltrating articular bone and cartilage, thus inducing a pannus formation. Eventually the

tissue, cartilage and the underlying bone are destroyed and the disease causes disability,

cardiovascular complications and premature death if insufficiently treated [302].

Page 50: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

50

Figure 5. Healthy (left) joint and joint affected by rheumatoid arthritis (right).

Reprinted from Strand et al. Nature Reviews Drug Discovery 6, 75–92 (January 2007) | doi:10.1038/nrd2196

with permission from Nature Publishing Group

The aetiology of RA is still unknown and therefore there is no curative treatment. In order to

alleviate the symptoms of the disease, anti-inflammatory drugs can be used but they do not

affect disease progression. In the past decade, several new anti-rheumatic drugs have become

available for the treatment of RA. In particular, biological agents that target specific pro-

inflammatory cytokines such as TNF- have proven to be efficient in combination with

methotrexate [303]. Nevertheless, anti-TNF- treatment exhibits notable side effects and is

inconvenient for patients to use because it is administered by injection. In addition, TNF

inhibition is not always effective, and the extent of therapeutic response varies between

different individuals [302].

Page 51: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

51

Role of MΦs and DCs in RA

Although the cause of RA has not yet been found, research over the past decades has lead to a

better insight into the pathogenesis of the disease. Among others, APCs appear as crucial cells

in determining this inflammatory condition. Large amounts of cells that are found in the

inflamed joint are MΦs and their numbers correlate to tissue destruction [304,305]. The main

mechanism for MΦ involvement in tissue destruction is likely by driving inflammation

through secretion of pro-inflammatory cytokines, such as TNF-α and IL1-β, that are known to

be important for disease progression [306].

Joint destruction starts by cleavage of the proteoglycans by aggrecanases, followed by

degradation of the collagen backbone by matrix metalloproteinases (MMPs) [307]. It has been

shown that synovial MΦs, that are present in large numbers in the cartilage-pannus-junction

[305], are important in driving the secretion of aggrecanases and MMPs [308].

Once the cartilage is degraded the bone erosion starts to take place. One central mechanism of

bone destruction is the generation of osteoclasts that origin from cells of myeloid lineage

[309]. M-CSF is produced by synovial endothelial cells and fibroblasts and its production is

increased by inflammatory mediators such as TNF-α and IL1-β [310,311]. The receptor

activator of NFκB (RANK) is essential for the formation of osteoclasts from myeloid

precursors [312]. It has been shown that monocyte/ MΦs of synovial tissues develop into

osteoclasts in the presence of M-CSF and RANK [313]. In support of the role of MΦs in bone

erosion, it has been shown that the radiological progression of joint destruction correlates with

the degree of synovial MΦ infiltration [304]. Thus, MΦs can contribute to RA pathogenesis

by inducing inflammation at early time points and amplifying cartilage destruction and bone

erosion at later time points.

It is known that resting, immature DCs are present in healthy tissues, including the synovial

tissue [314] and it has been shown that DCs can prime autoimmune responses in local lymph

Page 52: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

52

nodes [315] which could lead to development of autoantibodies and chronic inflammation.

Autoantibodies against citrullinated protein antigens are present in about 70% of RA cases

[316] and are associated with enhanced tissue injury [317] and worse outcome [318,319].

In support for a role of DCs in maintaining inflammation during RA, pDCs and mDCs are

reduced in the circulation of RA patients as compared to healthy controls while present in

synovial tissue in higher amount [320,321]. In addition, mDCs from RA patients were highly

responsive to endogenous and exogenous ligands and produced greater quantities of pro-

inflammatory cytokines than those from healthy controls [322,323]. Although MΦs and

mDCs are believed to be the most important cells for triggering inflammation and

cartilage/bone damages in this chronic disorder, recent findings also describe a role for pDCs.

In fact, when investigating the correlation between pDCs, rheumatoid factor and

autoantibodies, the number of pDCs in the synovium was positively correlated to serum levels

of rheumatoid factor and anti-citrullinated autoantibodies. This subset is known to play a

crucial role in the development of humoral immunity [215] and these observations suggest a

role for pDCs in triggering humoral autoimmunity in RA [321].

FMS-like tyrosine kinase 3 ligand (Flt-3L) has been shown to be strongly expressed in

synovial fluid of RA patients and induce erosive arthritis in mice [324]. Flt3-L is known to

induce differentiation of human CD11c+ DCs and increase their numbers in circulation [325]

thus providing a possible link between DCs and disease progression.

Finally, targeting of co-stimulatory molecules with blockers of the B7 family has proven

effective in reducing the clinical signs and symptoms of inflammation in RA patients [326].

This further emphasizes the role of co-stimulation by APC in initiating and perpetuating

inflammation during this autoimmune disorder.

In summary, various studies indicate APCs as crucial mediators of critical events in RA

pathogenesis.

Page 53: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

53

THE STUDIES

GENERAL AIM

As discussed in previous chapters professional APCs are of crucial importance in the

generation of adaptive immune responses. Much recent research is focused on clarifying the

precise mechanisms of these processes and their role in infection and autoimmunity. We have

tried to create in vitro or ex vivo approaches that allowed us to investigate the behaviour of

these cell types in response to specific stimuli or under the influence of anti-inflammatory

drugs.

SPECIFIC AIMS

Paper I

The aim of this study was to investigate whether the effect of an anti-inflammatory

pharmaceutical compound might act trough modulating the properties of APCs of myeloid

lineage. We hypothesized that the anti-inflammatory effects of the drug, previously observed

in animal models, may be through suppression of APCs.

Paper II

The goal of this study was to characterize the behaviour of MoDCs in malaria by challenging

these cells with iRBCs or sHz in vitro and to analyse their phenotype, migratory behaviour

and cytokine secretion. We hypothesised that an impaired activation of DCs by the parasite or

products derived from infection may contribute to the poor development of immunity to

malaria.

Paper III

The aim of this study was to investigate if the relative protection of the Fulani to malaria

infection could be related to the properties of APC. We hypothesised that early innate

Page 54: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

54

responses, for example via TLRs, and the prevalence or activation status of different APC

subsets of the Fulani children could be related to the enhanced protection against malaria seen

among the Fulani.

Page 55: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

55

METHODS A brief description of the methods used in the different projects is outlined below. For more

elaborated descriptions please see the materials & methods of each paper.

Paper I

Monocytes were purified from peripheral blood mononuclear cells (PBMCs) obtained from

healthy blood donors using Ficoll separation. The monocytes were then cultured in the

presence of different growth factors in order to generate different kind of APCs. The APCs

where then allowed to mature after which their phenotype, cytokine secretion and

immunostimulatory ability were assessed. The anti-inflammatory drug rabeximod was added

at different stages of differentiation and both cells and supernatants obtained at different time

points were analysed. The immunophenotype of the cells was analysed by FACS upon

staining for the relevant surface markers. The levels of secreted cytokines were measured in

the supernatant using cytometric bead array (CBA). The phagocytic capacity of the APCs was

assessed by allowing cells to internalize FITC-labelled Dextran or yeast particles. The

immunostimulatory abilities of the cells were assessed in an allogeneic MLR.

Paper II

Monocytes were purified from PBMCs obtained from healthy blood donors using Ficoll

separation. The monocytes were then cultured in the presence of GM-CSF and IL-4 to

produce MoDCs. The MoDCs were then challenged with sHz, crude malaria antigen or

iRBCs. The phenotype of the cells was analyzed using FACS, their migratory properties were

assessed using a Boyden chamber, their culture supernatant was analysed for cytokine

secretion using CBA and their immunostimulatory abilities was assessed through an

allogeneic MLR.

Page 56: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

56

Paper III

A total of 77 Malian children belonging to Dogon and Fulani ethnic groups were recruited

upon the informed consent of their parents. Giemsa stained slides where made and the

children were divided into groups of malaria infected or non-infected. Of the Dogon children

a total of 40 were enrolled of whom 20 were infected and 20 were non-infected. Of the Fulani

14 infected children were recruited and 23 non-infected making a total of 37 enrolled. Venous

blood samples were collected from all enrolled children and plasma was taken apart and

frozen for future analysis of cytokine and antibody levels in circulation. PBMCs were isolated

and stained for APC subtypes, activation markers and their phenotype was analysed by FACS.

In addition PBMCS were stimulated with TLR-specific ligands and the supernatants of the

stimulated cells were analysed for cytokine secretion using CBA and ELISA.

Page 57: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

57

RESULTS AND DISCUSSION

Paper I

The levels of cytokines and growth factors present during the generation of APCs are what

decide what type of APC they will become. APCs can develop into functionally distinct

subsets promoting different kinds of responses during inflammation. In addition, the manner

of activation of the APC decides what type of response the APC promotes. Thus, the manner

of their activation will affect what type of effector cells the T cells become. These T cells are

crucial for generating potent plasma cells capable of secreting large amounts of antibodies.

Therefore, APCs also play an important role in triggering humoral responses.

RA is a chronic inflammatory disease characterized, among other things, by autoreactive

antibodies. There is increasing evidence that TLRs may be involved in Th lineage decisions

and the maintenance of inflammation in RA [327,328]. This, and the fact that blocking of co

stimulation has yielded promising results in patients non responsive to TNF treatment

emphasize the role of antigen presentation in the development and maintenance of RA [329].

We investigated the effects of a newly developed anti-inflammatory drug, Rabeximod, on

APCs in vitro to evaluate its mode of action. In order to investigate the effect of Rabeximod

on DCs we generated MoDCs. An inflammatory subset of MΦs resembling the M1-type

described in the literature was generated by employing allogeneic stimulation of monocytes.

This stimulation lead to the differentiation of monocytes into MΦs that secrete IFN-, TNF-α,

IL-6, and IL-13 and efficiently support antigen presentation to allogeneic T cells. These cells

will be referred to as allostimulated (Allo)-MΦs. Another subset resembling the M2-MΦ was

also generated using the growth factor M-CSF that will be referred to as anti-inflammatory

(AI)-MΦs.

As expected from the literature [29] the phenotypic analysis showed that the MΦs retained

their CD14 expression while the MoDCs expressed low CD14 but high CD1a. When

Page 58: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

58

Rabeximod was added to monocytes at day 0 before addition of the differentiation stimuli the

MoDCs and Allo-MΦ were impaired in their differentiation whereas differentiation of AI-MΦ

was unaffected. This suggests that Rabeximod might impair the differentiation of APC with

pro- but not anti- inflammatory properties.

A key feature of APCs is the ability to take up antigen from the environment. The capacity of

each APC subset to take up antigen was assessed by allowing them to internalize FITC-

labelled Dextran particles. This assay revealed that MoDCs cultured in the presence of

Rabeximod where less efficient at taking up antigen whereas both MΦ subsets remained

unaffected.

The most potent initiators of T-cell proliferation were shown to be DCs already 30 years ago

[32]. The ability of each APC subset to induce proliferation of allogeneic T cells was assessed

in allogeneic MLRs. As expected, the MoDCs exhibited superior capacity to activate T cells

as compared to the MΦ subsets. Between the two MΦ subsets the Allo-MΦ were more potent

than the AI-MΦ at inducing T-cell proliferation. Both MoDCs and Allo-MΦs exhibited

impaired allostimulatory capacity upon exposure to Rabeximod, while AI-MΦs remained

unaffected by the drug. These findings again indicated that pro-inflammatory responses may

be selectively affected by the drug.

Rabeximod on its own however, does not seem to affect cytokine secretion by MΦ other than

decrease IL-6 production of MoDCs. It is possible that Rabeximod also affects IL-10

secretion but the variations in the results suggest that this effect may be donor dependent.

These results are in agreement with a previous study showing that Rabeximod did not affect

cytokine production in the absence of LPS in murine PBMCs [330]. Because of the crucial

importance of APCs early on in inflammation, our findings are in agreement with previous

work in a mouse model where the most potent effect of the drug was observed during the

early phases of inflammation [331].

Page 59: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

59

Taken together, the results presented here show that Rabeximod impairs the generation and

immunostimulatory ability of inflammatory APCs but do not affect immunomodulatory APC

subsets. Therefore, this may have an alleviating effect on the disease course of RA and

deserves further evaluation.

Paper II

Immunity to malaria is only acquired after many years of repeated exposure and this

immunity is rapidly lost. The reasons underlying the slow acquisition of immunity to malaria

is not well understood. Potent and long lasting immune responses are dependent on adaptive

immunity and the generation of memory lymphocytes. For these cells to be generated DCs are

required to potently induce naïve T cells to become effector cells. Depending on the

conditions under which an APC activates a naive T cell it may differentiate into different

kinds of effector cells or even become anergic. Consequently, to brief a TCR-MHC

interaction or lack of co-stimulatory molecules and/or cytokine secretion may lead to anergic

T cells. Therefore the phenotype and functional properties of APCs during an infection are

crucial in defining the ensuing immune response.

The effects of Hz or iRBCs on monocytes and MoDCs have been investigated in various

studies as reviewed in [332,333]. It has been shown that MoDCs that have been exposed to

high doses of iRBCs do not mature properly in response to LPS [288] and a similar effect on

MoDCs has been seen by Hz [286].

This study was designed to analyze the effect of sHz and iRBCs on phenotypic and functional

properties of MoDCs with regards to maturation, migration and antigen presentation. We

choose a combination of TNF-α and prostaglandin E2 (hereafter referred to as TNF-α/PGE2)

as positive control because of the enhanced migratory capacity seen in MoDCs exposed to

these stimuli [334].

Page 60: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

60

A clear dose response pattern was observed upon incubation with sHz with regards to

maturation markers CD80, CD83 and HLA-DR on MoDCs. However, only the surface

markers CD80 and CXCR4 were significantly increased. Although a slight up regulation of

CD83, HLA-DR and CCR7 was also observed. This up regulation of maturation markers

upon sHz stimulation was markedly lower than the one observed after stimulation with TNF-

α/PGE2. MoDCs were also analyzed for kinetics of the expression of surface markers on

MoDCs was assessed after 12, 36 and 60 hours of stimulation with sHz.

The results indicated that up regulation of CD83 by sHz was transient as compared to the up

regulation of the same marker induced by TNF-α/PGE2. The surface marker CD83 is

important for DC maturation and can influence DC-T cell interactions [35]. Therefore, these

findings may indicate that MoDCs exposed to sHz are only partially and transiently activated

and consequently become poor inducers of adaptive immune responses.

A similar phenotype with only marginal up regulation of maturation markers was found when

MoDCs were incubated with iRBCs including significantly increased CXCR4 expression.

These results are in line with other findings reporting of a partial maturation of DCs upon

incubation with malaria-derived products [289,335].

Because of the increased expression of the lymphoid CCRs observed upon stimulation with

sHz we assessed the migratory ability of these cells towards the ligands for CCR7 and

CXCR4, i.e CCL19/MIP-3β and CXCL12/SDF-1α, respectively. The results revealed that

immature MoDCs exposed to 20 µg sHz increased their migration towards both ligands as

compared to the cells that where incubated with medium alone.

The fact that partially mature MoDCs increased their migration towards chemotactic ligands,

important for homing to lymph nodes, in vitro may indicate that partially mature circulating

DCs are induced to travel to the lymph nodes during a natural P. falciparum infection.

However, MoDCs failed to properly up regulate CCR7 when they were exposed to sHz before

Page 61: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

61

TNF-α/PGE2 was added to the culture. Expression of CCR7 not only induces chemotaxis

towards lymphoid organs but has also been reported to enhance maturation, survival and

migratory speed of DCs [336]. Therefore we hypothesize that malaria, or products derived

from infection, could negatively affect DC responses in the presence of other maturation

stimuli.

The cytokines released by MoDC in response to sHz and iRBCs were also analyzed. An

increased release of IL-6, IL-10, TNF-α but not IL-12 was seen when MoDCs were incubated

with 20 µg sHz. Similarly, incubation of DCs with iRBCs increased the secretion levels of the

same cytokines. In addition stimulation of MoDCs with iRBCs lead to secretion of IL-1β that

was not observed upon stimulation with sHz.

It has been shown that IL-10 secreted by monocytes/MΦs upon contact with Hz decreases the

capacities of PBMCs in culture to secrete IL-2, IL-12 and IFN-γ [337], thus leading to

impaired induction of potent Th1 type responses. Consequently, lack of potent Th1 induction

would significantly impair the induction of a potent adaptive immune response. Secretion of

pro-inflammatory cytokines is generally important in the initial phases of malaria infection.

However, when high levels of pro-inflammatory cytokines are maintained they have been

associated with inflammatory induced tissue injury [338]

Finally, we assessed the ability of MoDCs to activate allogeneic T cells in an MLR after

contact with sHz or iRBCs. We found that cells that had been in contact with sHz induced

proliferation to a degree similar to the cells incubated with medium alone or uninfected RBCs.

In contrast, proliferation induced by MoDCs stimulated with iRBCs was similar to that of

cells stimulated with TNF-α/PGE2. The fact that iRBC-stimulated MoDCs induced

considerably higher T-cell activation despite their immature phenotype indicates that some

other mode of action must be involved. It is possible that a hitherto unknown mechanism or

soluble factor related to malaria infection may influence these processes.

Page 62: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

62

Taken together, our results show partially increased expression of maturation markers,

increased migration towards lymphoid chemokines and increased secretion of IL-10 but not

IL-12 by MoDCs upon contact with malaria derived products. However, MoDCs exposed to

sHz did not induce proliferation of allogeneic T cells whereas those exposed to intact infected

erythrocytes did. This suggests different modes of action of these activation patterns.

The fact that iRBCs induce secretion of IL-1β also indicates that differential inflammatory

responses are triggered by iRBCs as compared to sHz. It is known, that the Nalp3

inflammasome is activated by sHZ suggesting that caspase1 may be activated by both

mechanisms. However the production of pro-IL-1β is also required which may or may not be

triggered by sHz.

These in vitro observations suggest that DC responses in malaria are potent enough for DCs to

make contact with naïve T cells in the lymph nodes. However, proper induction of adaptive

immunity requires high expression of co-stimulatory molecules and IL-12 release by DCs.

This experimental model does not appear to yield such potent MoDCs but seem to be more in

line with studies of murine malaria.

In fact, it has been shown that DCs migrate to CD4 T-cell areas of the spleen upon blood

stage infection [339]. However, it was also shown that the P. chaubadi chaubadi parasite

induced only marginal maturation of DC [335,340]. Moreover, although these DCs displayed

an activated phenotype the formation of T-cell clusters and induction of differentiation was

impaired [340].

It is therefore possible that the actions of malarial products on MoDCs, with induction of

partial maturation and increased IL-10 secretion which in turn lead to impaired differentiation

of host T cells. The poor activation of T cells may then lead to impaired adaptive immune

responses and therefore insufficient clearance of the parasites.

Page 63: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

63

We therefore hypothesise that the lack of stable CD83 expression and no secretion of IL-12

are signs of hampered activation of APCs by the parasite or by products derived from

infection and that this may contribute to the poor development of immunity to malaria.

Paper III

It is well known that the Fulani show less clinical symptoms of infection than sympatric

ethnic groups although they are exposed to the same parasite pressure [296]. Different aspects

of the immune responses to malaria of the Fulani have been investigated in different African

countries [295,341]. The Fulani exhibit higher antibody titres, enlarged spleen rates and

higher levels of cytokines in circulation compared to sympatric ethnic groups.

This study was designed to investigate some aspects of the innate immune responses in

children belonging to the Fulani ethnic group in Mali. Children from a neighbouring village

belonging to a different ethnic group, the Dogon, were recruited to the study as a control

population. The aim of the study was to investigate the prevalence and activation status of

APCs as well as responses to specific TLR stimuli of Fulani and Dogon children and if this

could be involved in their different response to malaria infection.

In addition to the data presented in the paper included in this thesis we confirmed that Fulani

children have higher levels of malaria specific antibodies (Figure 6 and Figure 7) and higher

levels of cytokines (Figure 8) in circulation as compared to Dogon. The Fulani had higher

levels of total antimalarial IgG and IgM and of the IgG subclasses IgG1, IgG2 and IgG3 but

not IgG4.

The levels of cytokines involved in inflammation were measured in serum of children

belonging to the two ethnic groups. It was found that Fulani children had higher levels of

IFN-α, IFN-γ, IL-6, IL-8 and IL-12(p70) than the Dogon regardless of infectious status

(Figure 8). These findings are in line with previous findings of our group in the adult

populations of these and other villages [342-344].

Page 64: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

64

Figure 6. Specific antibody responses to P. falciparum are higher in Fulani children than in the Dogon

children

Blood plasma samples from 77 children were analysed for antimalarial IgG (A) and IgM (B). The samples were

subdivided according to ethnicity and slide positivity to malaria; i.e. uninfected Dogon (n=20), infected Dogon

(n=20), uninfected Fulani (n=23) and infected Fulani (n=14). The y-axis depicts the concentration of the

respective antibody/ml of plasma . The boxplots illustrate the medians and the 25th and 75th quartile and the

whiskers represent the 10% and 90% percentiles. Data were analyzed using Mann-Whitney rank sum test. *;

p≤0.05. **;p<0.01. ***; p<0.001.

Figure 7. Malaria specific IgG subclass responses to P. falciparum in Fulani and Dogon children.

Blood plasma samples from 77 children were analysed for antimalarial IgG1 (A), 2 (B), 3 (C) and 4 (D). The

samples were subdivided according to ethnicity and slide positivity to malaria; i.e. uninfected Dogon (n=20),

infected Dogon (n=20), uninfected Fulani (n=23) and infected Fulani (n=14). The y-axis represent µg of the

respective antibody/ml of plasma. The boxplots illustrate the medians and the 25th and 75th quartile and the

whiskers represent the 10% and 90% percentiles.. Data were analysed using Mann-Whitney rank sum test. *;

p≤0.05. **;p<0.01. ***; p<0.001.

Page 65: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

65

Figure 8. Intraethnic and interethnic difference in cytokine plasma levels.

Blood plasma samples from 77 children were analyzed for cytokines. The levels of IL-6 (A), IL-8 (B) and IL-

12p70 (C) were measured using cytometric bead array. The levels of IFN- α (D) and IFN-γ (E) were analyzed

using commercial ELISA kits. The samples were subdivided according to ethnicity and slide positivity to

malaria; i.e. uninfected Dogon (n=20), infected Dogon (n=20), uninfected Fulani (n=23) and infected Fulani

(n=14).The y-axis depicts picograms (pg) of the respective cytokine/ml of plasma. The boxplots illustrate the

medians and the 25th and 75th quartile and the whiskers represent the 10% and 90% percentiles. Data were

analyzed using Mann-Whitney rank sum test. *; p≤0.05.***; p<0.001.

We then analyzed four subtypes of blood DCs as well as their activation status in the children

recruited to the study. A decreased expression of the activation marker HLA-DR was found

on all DC subsets of infected Dogon children as compared to their uninfected peers.

Conversely, the expression of HLA-DR was increased in Fulani children undergoing infection

on BDCA-2+ and BDCA-3

+ subsets when compared to their uninfected peers. The expression

of CD86 followed the same pattern i.e. decreased in the infected Dogon children while it is

increased or unaltered in infected Fulani children.

Page 66: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

66

We then analysed the frequency of the different subsets in circulation. An increase of

circulating BDCA-2+ and BDCA-3

+ DCs was observed in the infected Dogon as compared to

the uninfected Dogon. In contrast, a decrease of circulating BDCA-2+ and BDCA-3

+ DCs was

observed in the infected Fulani as compared to their uninfected peers.

The activation status of the monocytic subsets in circulation was reduced in terms of

decreased expression of HLA-DR and CD86 in infected children regardless of ethnicity.

However, this decrease was more pronounced in the Dogon, consequently Fulani children

undergoing infection maintained higher levels of activation markers on classical monocytes

than the infected Dogon.

The expression of TLRs differs between the different APC subsets. In order to further

characterize the responses of these cells we stimulated their PBMCs with specific ligands for

TLR4, TLR7 and TLR9 that is LPS, Imiquimod and CpG respectively. We then analysed the

supernatants of the PBMCs stimulated with these ligands for cytokine secretion.

We found that secretion of practically all investigated cytokines was supressed in Dogon

children undergoing infection while Fulani children seemed unaffected. Strikingly, the

secretion of IFNγ was completely abolished in Dogon children undergoing infection. Not

surprisingly the ratio of IFNγ/IL-10 was severely decreased in Dogon undergoing infection

but unaffected in the Fulani. This pattern of supressed pro-inflammatory responses of the

Dogon were confirmed when the ratio of TNF-α/IL-10 was calculated. A decreased ratio of

TNF-α/IL-10 upon stimulation of PBMCs with ligands to TLR7, TLR9 but not TLR4 was

observed. This might indicate that the TLR4 signalling pathway is differently affected than

the TLR7 and TLR9 pathways.

In summary, the activation status was increased in DCs of Fulani undergoing malaria

infection but decreased in Dogon undergoing infection while the prevalence of these cells in

circulation was decreased in infected Fulani and increased in infected Dogon. We therefore

Page 67: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

67

hypothesize that the different levels of DCs in the blood of the two ethnic groups undergoing

malaria might be explained by the different activation status of these cells. The increased

activation status of circulating DC subsets in infected Fulani children may indicate that these

cells have migrated to lymphoid organs to initiate adaptive immunity and are therefore no

longer found in circulation. Conversely, in the Dogon ethnic group, malaria infection may

impair activation of circulating DCs thus possibly affecting downstream cellular and humoral

immune responses.

We observed that PBMCs of infected Dogon children are suppressed in their responses to

specific TLR ligands but the cells from the Fulani remain unaffected when undergoing

infection. This is in line with our findings that DCs of Fulani in circulation seem to become

more activated upon infection while DCs of the Dogon are suppressed.

Taken together, these findings suggest that there is a difference in the prevalence and

activation status of blood DCs and cytokine secretion between the Fulani and the Dogon in

response to malaria infection. Because of the crucial importance of APCs in initiation of

adaptive immune responses these differences may very well be important for the relative

protection against malaria seen in the Fulani as compared to the Dogon.

Page 68: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

68

GENERAL CONCLUSIONS It has become evident that APCs play a crucial part in the early stages of an immune response.

It is also increasingly evident that different manners of APC activation can have very different

consequences, and that what we commonly regard as phenotypic activation may not lead to

initiation of adaptive immunity. Microbial stimuli have been shown to induce potent

responses in DCs including secretion of IL-12 and initiation of adaptive immunity [173,174].

In mice it has been shown that CD8α- DCs express TLR7 but CD8α

+ DCs do not. Upon

injection of TLR7 ligand both subsets display a mature phenotype but only CD8α- DCs,

which respond directly to TLR7, secrete IL-12 [345]. In line with these studies it has been

shown that DCs secrete TNF-α and IL-6 in response to microbial antigens but only the TLR4-

mediated responses lead to IL-12 production [346]. These findings suggest that TLR ligation

is necessary and sufficient to mount an effective immune response.

Taking these data into consideration, Joffre et al. suggested that all DCs respond to

inflammatory stimuli by migrating to the lymph nodes but only those cells that have sensed

microbes directly induce effector T cells. The bystander DCs that have not directly sensed the

pathogen become activated but would instead present tissue antigens from the infectious foci

and “rescue” these antigens from the immune response thereby evading excessive and

detrimental self-responses [347]. These bystander cells may therefore display an activated

phenotype but be unable to induce efficient T-cell responses. Supporting this hypothesis is the

fact that un-activated DCs continuously present self-derived antigens from tissues or apoptotic

cells without leading to initiation of immunity [241].

We suggest that rendering APCs to act as bystander cells, i.e. not efficient in inducing proper

T-cell responses may be an effective way of subverting immune responses by pathogens such

as P. Falciparum. Moreover, inappropriate signalling while recognizing and presenting self-

Page 69: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

69

antigens may activate APCs and lead to prolonged and detrimental immune responses, thus

leading to autoimmunity.

Our first study revealed that a drug that has shown anti-inflammatory effects in a murine

model of autoimmune arthritis acts efficiently by inhibiting inflammatory subsets of human

APCs in vitro. This could be an effective mechanism to break the maintenance of

inflammation during RA and contribute to decelerate disease progress. It also suggests a

central role for APCs in determining detrimental inflammation in autoimmunity.

Our second study revealed that products of malaria infection, such as Hz and iRBCs do

induce phenotypic activation and migration of MoDCs. We also observed that iRBCs induce

T-cell proliferation but none of these malaria-stimulated DCs produce IL-12. This might

indicate that malaria infection can somehow induce APCs to act as bystander APCs,

ineffective at activating T cells, even though they have had previous contact with the parasite.

In the third study, we compared the Dogon population of Mali to the Fulani, an ethnic group

that displays a relatively better protection against malaria. We observed that TLR responses of

Dogon children are severely suppressed and their APCs express decreased levels of activation

markers when undergoing malaria infection, whereas the Fulani exhibit unaltered TLR

responses and increased APC activation while undergoing infection. These differences in

TLR responses and APC behaviour may indicate an important role for APCs in malarial

immunity.

In summary, these data support the view that when APCs are improperly activated the ensuing

adaptive responses may become inefficient or excessive leading to pathology (Figure 9). This

suggests a central role for APCs in contributing to autoimmunity and dysregulated responses

to pathogens. Our observations may suggest that the malaria parasite has found a way to limit

APC responses to that of bystander cells and therefore insufficient long-term immunity is

initiated upon contact with malaria derived products. In addition, the Fulani ethnic group may

Page 70: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

70

somehow have overcome this suppression and their APCs respond more potently to malaria

infection. This could lead to increased levels of IFN secretion and augmented adaptive

responses leading to higher titres of protective antibodies.

Figure 9. Consequences of immune activation under different circumstances. Different challenges to the immune system induce different responses in APCs. The responses elicited in APCs

lead to distinct T-helper responses according to the different situations. We suggest that malaria has a distinct

way of manipulating these responses so that efficient T-cell responses are dampened. Moreover, inappropriate

triggering of APC responses may play an important role in the induction of autoimmunity.

Page 71: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

71

FUTURE WORK Our studies, together with previous findings, indicate that APCs are important players in the

orchestration of immune responses both in autoimmune disease and in malaria infection.

Much recent progress has been made in elucidating the mechanisms through which the

malaria parasite activates innate immune receptors, in particular TLR9. Because of the

functional differences observed in TLR responses between Dogon and Fulani, it would be

interesting to investigate known mutations in various TLR genes in order to unravel whether

these differences are genetically determined.

In addition, there are many details about the mechanisms surrounding APC responses to

malarial products that would be interesting topics for further research. For instance, detailed

information about the effects of different strains of parasites on different APC subsets and on

APC precursors. Since increased levels of BDCA-3+ DCs have been associated with severe

disease and an increase of that subset in circulation is seen in the Dogon that are more

susceptible to disease. More detailed investigations into the role of BDCA-3+ DCs in malaria

as compared to other circulating subsets of DCs is needed. More detailed knowledge is also

needed of the mechanisms behind the relatively better protection to malaria seen among the

Fulani, in particular the role of APCs. This knowledge would be of outmost importance for

understanding how a potent response to malaria is achieved and may also be helpful in

vaccine development.

Finally, and perhaps most important, is the underlying question of how different activation

patterns of APCs lead to different effector responses. In general, a better understanding of

APC activation and downstream responses would be invaluable for coming up with efficient

strategies to combat infections and immune disorders.

Page 72: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

72

ACKNOWLEDGEMENTS This work would not have been possible to complete were it not for the invaluable support

and friendship of many wonderful individuals that I have crossed paths with during the years.

Therefore, I would like to thank the whole department of immunology as well as other

departments, institutes and universities that have in any way contributed to my education and

development within the fields of malaria and immunology.

The most important person for the realization of my PhD studies has of course been my

supervisor Marita Troye-Blomberg. I thank you first for giving me the opportunity to work

with you but also for always taking your time to help me in every way and for sharing your

vast experience.

The next key person to the work behind this thesis from whom I learnt most practical aspects

and with whom I spent lots of time during the first years of my studies is my co supervisor

Stefania Varani. I thank you for your patience while working with my stubborn self through

all these years.

I would also like to thank all the other seniors at our or other departments for sharing their

knowledge and experience. In particular Carmen Fernandez, muchísimas gracias por todo tipo

de apoyo en la ciencia y en lo demas, I am also grateful to Klavs Berzins, Eva Sverremark-

Ekström and Eva Severinson and Ann-Kristin Östlund Farrants for great company and many

interesting discussions during the years.

My thanks also goes to the backbone of the department, Gelana Yadeta, Margareta ”Maggan”

Hagstedt, Ann Sjölund och Anna-Leena Jarva tack för all hjälp och för att ni stått ut med mina

galenskaper.

I next want to thank my friends, fellow malariologists and future doctors Charles Arama for

all the long talks, un monde meilleur est nécessaire et possible! Stéphanie Boström om du

hoppar av vetenskapen kan du ju alltid bli bagare ;) with both of whom I have spent good and

bad times all over the planet, I am really glad I have had the pleasure to work closely with

Page 73: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

73

these two wonderful personalities during these years. And also to the latest addition in the

malaria team Ioana “thevampire” Bujila with whom I have also had the pleasure to work

closely together and shared many enjoyable experiences. A special thanks also to Lisa

Israelsson whose good spirits and pleasurable company I had the privilege to enjoy during the

first years.

Then there are those that I haven’t had the chance to work so close to but with whom I have

shared many happy and less happy moments Ebba “toomanynicknamestoremember”

Sohlberg, Maria Johansson, för kryssen, kaffet, kakorna och alla andra äventyr vi delat.

Then there is the long list of past and present students at the department special thanks also to

Petra Amoudruz, Halima Balogun, Jubayer Rahman, Salah Farouk and Amre Nasr for their

outstanding personalities and kind manners. I am also glad to have gotten to know Nancy

Awah, Olga Chuquimia Flores, Manijeh Vafa, Shanie Saghafian Hedengren, Natalija

Gerasimick, Yeneneh Haileselassie, Ylva Sjögren Bolin, Ulrika Homlund, Nora Bachmayer,

Yvonne Sjölund, Nnaemeka Iriemenam, and John Arko-Mensah that have contributed to

many good times in the department.

Then there are the people I have come in contact to for one reason or another that I have had

the privilege to work with or at least get to know like Anna Tjärnlund, Yohannes Assefaw-

Redda, Valentina Mangano, Sabina Dahlström, Samad Ibitokou and Maiga Boubacar.

My fellow malariologists from the Swedish Malaria Network including Pedro and Isabel of

the Portuguese resistance group, the “Chen girls” Sandra Nilsson and Kim Brolin, Anna

Färnert with students Ann Liljander and Klara Lundbom, finally the later additions of Maja

Malmberg and Mia Palmqvist.

I would also like to extend my gratitude to co-authors and all the people senior or students

who I have come in contact with during these years.

Page 74: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

74

Last but not least are the people everywhere outside of the scientific world that I am fortunate

enough to share my life with. You are all too many to mention here but you have been every

bit as important as the people mentioned above.

Para toda mi extendida familia, pasada, presente y porvenir, de sangre o recuparada por los

caminos los llevare por siempre conmigo.

Till mina L(o)uisar, Mamma, Marre och Erre jag hade inte kunnat välja en bättre familj om

jag hade kunnat, har er att tacka för allt jag är och önskar er all lycka i världen!

Page 75: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

75

REFERENCES

1. Starr TK, Jameson SC, Hogquist KA. (2003) Positive and negative selection of T cells.

Annu Rev Immunol 21: 139-176.

2. Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL. (1986) Two types of

murine helper T cell clone. I. definition according to profiles of lymphokine activities

and secreted proteins. J Immunol 136(7): 2348-2357.

3. Mosmann TR, Coffman RL. (1989) TH1 and TH2 cells: Different patterns of lymphokine

secretion lead to different functional properties. Annu Rev Immunol 7: 145-173.

4. Korn T, Bettelli E, Oukka M, Kuchroo VK. (2009) IL-17 and Th17 cells. Annu Rev

Immunol 27: 485-517.

5. Bluestone JA, Abbas AK. (2003) Natural versus adaptive regulatory T cells. Nat Rev

Immunol 3(3): 253-257.

6. Zhu J, Paul WE. (2010) Peripheral CD4+ T-cell differentiation regulated by networks of

cytokines and transcription factors. Immunol Rev 238(1): 247-262.

7. Corazza N, Muller S, Brunner T, Kagi D, Mueller C. (2000) Differential contribution of

fas- and perforin-mediated mechanisms to the cell-mediated cytotoxic activity of naive

and in vivo-primed intestinal intraepithelial lymphocytes. J Immunol 164(1): 398-403.

8. Joshi NS, Kaech SM. (2008) Effector CD8 T cell development: A balancing act between

memory cell potential and terminal differentiation. J Immunol 180(3): 1309-1315.

9. Auffray C, Fogg D, Garfa M, Elain G, Join-Lambert O, et al. (2007) Monitoring of blood

vessels and tissues by a population of monocytes with patrolling behavior. Science

317(5838): 666-670.

10. Randolph GJ, Inaba K, Robbiani DF, Steinman RM, Muller WA. (1999) Differentiation of

phagocytic monocytes into lymph node dendritic cells in vivo. Immunity 11(6): 753-761.

11. Passlick B, Flieger D, Ziegler-Heitbrock HW. (1989) Identification and characterization

of a novel monocyte subpopulation in human peripheral blood. Blood 74(7): 2527-2534.

12. Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, et al. (2010) Nomenclature of

monocytes and dendritic cells in blood. Blood 116(16): e74-80.

13. Mechnikov II. (1988) Immunity in infective diseases. by il'ia il'ich mechnikov, 1905. Rev

Infect Dis 10(1): 223-227.

14. Kunisch E, Fuhrmann R, Roth A, Winter R, Lungershausen W, et al. (2004) Macrophage

specificity of three anti-CD68 monoclonal antibodies (KP1, EBM11, and PGM1) widely

used for immunohistochemistry and flow cytometry. Ann Rheum Dis 63(7): 774-784.

15. Beranek JT. (2005) CD68 is not a macrophage-specific antigen. Ann Rheum Dis 64(2):

342-3; author reply 343-4.

Page 76: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

76

16. Nathan CF, Murray HW, Wiebe ME, Rubin BY. (1983) Identification of interferon-

gamma as the lymphokine that activates human macrophage oxidative metabolism and

antimicrobial activity. J Exp Med 158(3): 670-689.

17. Nathan CF, Prendergast TJ, Wiebe ME, Stanley ER, Platzer E, et al. (1984) Activation of

human macrophages. comparison of other cytokines with interferon-gamma. J Exp Med

160(2): 600-605.

18. Abramson SL, Gallin JI. (1990) IL-4 inhibits superoxide production by human

mononuclear phagocytes. J Immunol 144(2): 625-630.

19. Doherty TM, Kastelein R, Menon S, Andrade S, Coffman RL. (1993) Modulation of

murine macrophage function by IL-13. J Immunol 151(12): 7151-7160.

20. Stein M, Keshav S, Harris N, Gordon S. (1992) Interleukin 4 potently enhances murine

macrophage mannose receptor activity: A marker of alternative immunologic

macrophage activation. J Exp Med 176(1): 287-292.

21. Goerdt S, Politz O, Schledzewski K, Birk R, Gratchev A, et al. (1999) Alternative versus

classical activation of macrophages. Pathobiology 67(5-6): 222-226.

22. Mantovani A, Sozzani S, Locati M, Allavena P, Sica A. (2002) Macrophage polarization:

Tumor-associated macrophages as a paradigm for polarized M2 mononuclear

phagocytes. Trends Immunol 23(11): 549-555.

23. Sica A, Schioppa T, Mantovani A, Allavena P. (2006) Tumour-associated macrophages

are a distinct M2 polarised population promoting tumour progression: Potential targets of

anti-cancer therapy. Eur J Cancer 42(6): 717-727.

24. Gordon S. (2003) Alternative activation of macrophages. Nat Rev Immunol 3(1): 23-35.

25. Mosser DM, Edwards JP. (2008) Exploring the full spectrum of macrophage activation.

Nat Rev Immunol 8(12): 958-969.

26. Hamilton JA. (2008) Colony-stimulating factors in inflammation and autoimmunity. Nat

Rev Immunol 8(7): 533-544.

27. Fleetwood AJ, Lawrence T, Hamilton JA, Cook AD. (2007) Granulocyte-macrophage

colony-stimulating factor (CSF) and macrophage CSF-dependent macrophage

phenotypes display differences in cytokine profiles and transcription factor activities:

Implications for CSF blockade in inflammation. J Immunol 178(8): 5245-5252.

28. Young DA, Lowe LD, Clark SC. (1990) Comparison of the effects of IL-3, granulocyte-

macrophage colony-stimulating factor, and macrophage colony-stimulating factor in

supporting monocyte differentiation in culture. analysis of macrophage antibody-

dependent cellular cytotoxicity. J Immunol 145(2): 607-615.

29. Verreck FA, de Boer T, Langenberg DM, Hoeve MA, Kramer M, et al. (2004) Human IL-

23-producing type 1 macrophages promote but IL-10-producing type 2 macrophages

subvert immunity to (myco)bacteria. Proc Natl Acad Sci U S A 101(13): 4560-4565.

Page 77: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

77

30. Steinman RM, Cohn ZA. (1973) Identification of a novel cell type in peripheral lymphoid

organs of mice. I. morphology, quantitation, tissue distribution. J Exp Med 137(5): 1142-

1162.

31. Steinman RM, Lustig DS, Cohn ZA. (1974) Identification of a novel cell type in

peripheral lymphoid organs of mice. 3. functional properties in vivo. J Exp Med 139(6):

1431-1445.

32. Steinman RM, Witmer MD. (1978) Lymphoid dendritic cells are potent stimulators of the

primary mixed leukocyte reaction in mice. Proc Natl Acad Sci U S A 75(10): 5132-5136.

33. Naik SH. (2008) Demystifying the development of dendritic cell subtypes, a little.

Immunol Cell Biol 86(5): 439-452.

34. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, et al. (2000) Immunobiology of

dendritic cells. Annu Rev Immunol 18: 767-811.

35. Lechmann M, Zinser E, Golka A, Steinkasserer A. (2002) Role of CD83 in the

immunomodulation of dendritic cells. Int Arch Allergy Immunol 129(2): 113-118.

36. Sallusto F, Schaerli P, Loetscher P, Schaniel C, Lenig D, et al. (1998) Rapid and

coordinated switch in chemokine receptor expression during dendritic cell maturation.

Eur J Immunol 28(9): 2760-2769.

37. Bachmann MF, Kopf M, Marsland BJ. (2006) Chemokines: More than just road signs.

Nat Rev Immunol 6(2): 159-164.

38. Jarrossay D, Napolitani G, Colonna M, Sallusto F, Lanzavecchia A. (2001) Specialization

and complementarity in microbial molecule recognition by human myeloid and

plasmacytoid dendritic cells. Eur J Immunol 31(11): 3388-3393.

39. Dzionek A, Fuchs A, Schmidt P, Cremer S, Zysk M, et al. (2000) BDCA-2, BDCA-3, and

BDCA-4: Three markers for distinct subsets of dendritic cells in human peripheral blood.

J Immunol 165(11): 6037-6046.

40. Piccioli D, Tavarini S, Borgogni E, Steri V, Nuti S, et al. (2007) Functional specialization

of human circulating CD16 and CD1c myeloid dendritic-cell subsets. Blood 109(12):

5371-5379.

41. MacDonald KP, Munster DJ, Clark GJ, Dzionek A, Schmitz J, et al. (2002)

Characterization of human blood dendritic cell subsets. Blood 100(13): 4512-4520.

42. Inaba K, Inaba M, Romani N, Aya H, Deguchi M, et al. (1992) Generation of large

numbers of dendritic cells from mouse bone marrow cultures supplemented with

granulocyte/macrophage colony-stimulating factor. J Exp Med 176(6): 1693-1702.

43. Romani N, Gruner S, Brang D, Kampgen E, Lenz A, et al. (1994) Proliferating dendritic

cell progenitors in human blood. J Exp Med 180(1): 83-93.

Page 78: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

78

44. Sallusto F, Lanzavecchia A. (1994) Efficient presentation of soluble antigen by cultured

human dendritic cells is maintained by granulocyte/macrophage colony-stimulating

factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med

179(4): 1109-1118.

45. Caux C, Vanbervliet B, Massacrier C, Dezutter-Dambuyant C, de Saint-Vis B, et al.

(1996) CD34+ hematopoietic progenitors from human cord blood differentiate along two

independent dendritic cell pathways in response to GM-CSF+TNF alpha. J Exp Med

184(2): 695-706.

46. Shortman K, Naik SH. (2007) Steady-state and inflammatory dendritic-cell development.

Nat Rev Immunol 7(1): 19-30.

47. Poulin LF, Salio M, Griessinger E, Anjos-Afonso F, Craciun L, et al. (2010)

Characterization of human DNGR-1+ BDCA3+ leukocytes as putative equivalents of

mouse CD8alpha+ dendritic cells. J Exp Med 207(6): 1261-1271.

48. Shortman K, Liu YJ. (2002) Mouse and human dendritic cell subtypes. Nat Rev Immunol

2(3): 151-161.

49. Ginhoux F, Tacke F, Angeli V, Bogunovic M, Loubeau M, et al. (2006) Langerhans cells

arise from monocytes in vivo. Nat Immunol 7(3): 265-273.

50. Merad M, Manz MG, Karsunky H, Wagers A, Peters W, et al. (2002) Langerhans cells

renew in the skin throughout life under steady-state conditions. Nat Immunol 3(12):

1135-1141.

51. Cheong C, Matos I, Choi JH, Dandamudi DB, Shrestha E, et al. (2010) Microbial

stimulation fully differentiates monocytes to DC-SIGN/CD209(+) dendritic cells for

immune T cell areas. Cell 143(3): 416-429.

52. Shigematsu H, Reizis B, Iwasaki H, Mizuno S, Hu D, et al. (2004) Plasmacytoid dendritic

cells activate lymphoid-specific genetic programs irrespective of their cellular origin.

Immunity 21(1): 43-53.

53. Zuniga EI, McGavern DB, Pruneda-Paz JL, Teng C, Oldstone MB. (2004) Bone marrow

plasmacytoid dendritic cells can differentiate into myeloid dendritic cells upon virus

infection. Nat Immunol 5(12): 1227-1234.

54. Janeway CA,Jr. (1989) Approaching the asymptote? evolution and revolution in

immunology. Cold Spring Harb Symp Quant Biol 54 Pt 1: 1-13.

55. Matzinger P. (1994) Tolerance, danger, and the extended family. Annu Rev Immunol 12:

991-1045.

56. Fearon DT, Locksley RM. (1996) The instructive role of innate immunity in the acquired

immune response. Science 272(5258): 50-53.

57. Janeway CA,Jr, Medzhitov R. (2002) Innate immune recognition. Annu Rev Immunol 20:

197-216.

Page 79: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

79

58. Rifkin IR, Leadbetter EA, Busconi L, Viglianti G, Marshak-Rothstein A. (2005) Toll-like

receptors, endogenous ligands, and systemic autoimmune disease. Immunol Rev 204: 27-

42.

59. Zhang Z, Schluesener HJ. (2006) Mammalian toll-like receptors: From endogenous

ligands to tissue regeneration. Cell Mol Life Sci 63(24): 2901-2907.

60. Lemaitre B, Nicolas E, Michaut L, Reichhart JM, Hoffmann JA. (1996) The dorsoventral

regulatory gene cassette spatzle/Toll/cactus controls the potent antifungal response in

drosophila adults. Cell 86(6): 973-983.

61. Lemaitre B. (2004) The road to toll. Nat Rev Immunol 4(7): 521-527.

62. Poltorak A, He X, Smirnova I, Liu MY, Van Huffel C, et al. (1998) Defective LPS

signaling in C3H/HeJ and C57BL/10ScCr mice: Mutations in Tlr4 gene. Science

282(5396): 2085-2088.

63. Takeuchi O, Hoshino K, Kawai T, Sanjo H, Takada H, et al. (1999) Differential roles of

TLR2 and TLR4 in recognition of gram-negative and gram-positive bacterial cell wall

components. Immunity 11(4): 443-451.

64. Hemmi H, Takeuchi O, Kawai T, Kaisho T, Sato S, et al. (2000) A toll-like receptor

recognizes bacterial DNA. Nature 408(6813): 740-745.

65. Takeuchi O, Kawai T, Muhlradt PF, Morr M, Radolf JD, et al. (2001) Discrimination of

bacterial lipoproteins by toll-like receptor 6. Int Immunol 13(7): 933-940.

66. Diebold SS, Massacrier C, Akira S, Paturel C, Morel Y, et al. (2006) Nucleic acid agonists

for toll-like receptor 7 are defined by the presence of uridine ribonucleotides. Eur J

Immunol 36(12): 3256-3267.

67. Iwasaki A, Medzhitov R. (2004) Toll-like receptor control of the adaptive immune

responses. Nat Immunol 5(10): 987-995.

68. Akira S, Takeda K. (2004) Toll-like receptor signalling. Nat Rev Immunol 4(7): 499-511.

69. Ozinsky A, Underhill DM, Fontenot JD, Hajjar AM, Smith KD, et al. (2000) The

repertoire for pattern recognition of pathogens by the innate immune system is defined by

cooperation between toll-like receptors. Proc Natl Acad Sci U S A 97(25): 13766-13771.

70. Dunne A, O'Neill LA. (2003) The interleukin-1 receptor/Toll-like receptor superfamily:

Signal transduction during inflammation and host defense. Sci STKE 2003(171): re3.

71. Takeda K, Kaisho T, Akira S. (2003) Toll-like receptors. Annu Rev Immunol 21: 335-

376.

72. Doyle S, Vaidya S, O'Connell R, Dadgostar H, Dempsey P, et al. (2002) IRF3 mediates a

TLR3/TLR4-specific antiviral gene program. Immunity 17(3): 251-263.

Page 80: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

80

73. Hemmi H, Kaisho T, Takeda K, Akira S. (2003) The roles of toll-like receptor 9, MyD88,

and DNA-dependent protein kinase catalytic subunit in the effects of two distinct CpG

DNAs on dendritic cell subsets. J Immunol 170(6): 3059-3064.

74. Manicassamy S, Pulendran B. (2009) Modulation of adaptive immunity with toll-like

receptors. Semin Immunol .

75. Kawai T, Adachi O, Ogawa T, Takeda K, Akira S. (1999) Unresponsiveness of MyD88-

deficient mice to endotoxin. Immunity 11(1): 115-122.

76. Hoebe K, Beutler B. (2006) TRAF3: A new component of the TLR-signaling apparatus.

Trends Mol Med 12(5): 187-189.

77. Hacker H, Redecke V, Blagoev B, Kratchmarova I, Hsu LC, et al. (2006) Specificity in

toll-like receptor signalling through distinct effector functions of TRAF3 and TRAF6.

Nature 439(7073): 204-207.

78. Engering A, Geijtenbeek TB, van Vliet SJ, Wijers M, van Liempt E, et al. (2002) The

dendritic cell-specific adhesion receptor DC-SIGN internalizes antigen for presentation

to T cells. J Immunol 168(5): 2118-2126.

79. Geijtenbeek TB, van Vliet SJ, Engering A, 't Hart BA, van Kooyk Y. (2004) Self- and

nonself-recognition by C-type lectins on dendritic cells. Annu Rev Immunol 22: 33-54.

80. Geijtenbeek TB, Gringhuis SI. (2009) Signalling through C-type lectin receptors: Shaping

immune responses. Nat Rev Immunol 9(7): 465-479.

81. Martinon F, Burns K, Tschopp J. (2002) The inflammasome: A molecular platform

triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell

10(2): 417-426.

82. Lamkanfi M, Kanneganti TD, Franchi L, Nunez G. (2007) Caspase-1 inflammasomes in

infection and inflammation. J Leukoc Biol 82(2): 220-225.

83. Lich JD, Arthur JC, Ting JP. (2006) Cryopyrin: In from the cold. Immunity 24(3): 241-

243.

84. Ting JP, Kastner DL, Hoffman HM. (2006) CATERPILLERs, pyrin and hereditary

immunological disorders. Nat Rev Immunol 6(3): 183-195.

85. Eisenbarth SC, Flavell RA. (2009) Innate instruction of adaptive immunity revisited: The

inflammasome. EMBO Mol Med 1(2): 92-98.

86. Moore KW, de Waal Malefyt R, Coffman RL, O'Garra A. (2001) Interleukin-10 and the

interleukin-10 receptor. Annu Rev Immunol 19: 683-765.

87. Yoneyama M, Suhara W, Fukuhara Y, Fukuda M, Nishida E, et al. (1998) Direct

triggering of the type I interferon system by virus infection: Activation of a transcription

factor complex containing IRF-3 and CBP/p300. EMBO J 17(4): 1087-1095.

Page 81: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

81

88. Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T, et al. (2004) The RNA

helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral

responses. Nat Immunol 5(7): 730-737.

89. Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, et al. (2006) Differential roles

of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature 441(7089):

101-105.

90. Gitlin L, Barchet W, Gilfillan S, Cella M, Beutler B, et al. (2006) Essential role of mda-5

in type I IFN responses to polyriboinosinic:Polyribocytidylic acid and

encephalomyocarditis picornavirus. Proc Natl Acad Sci U S A 103(22): 8459-8464.

91. Banchereau J, Steinman RM. (1998) Dendritic cells and the control of immunity. Nature

392(6673): 245-252.

92. Kalinski P, Hilkens CM, Wierenga EA, Kapsenberg ML. (1999) T-cell priming by type-1

and type-2 polarized dendritic cells: The concept of a third signal. Immunol Today

20(12): 561-567.

93. Garcia KC, Degano M, Stanfield RL, Brunmark A, Jackson MR, et al. (1996) An

alphabeta T cell receptor structure at 2.5 A and its orientation in the TCR-MHC complex.

Science 274(5285): 209-219.

94. Shin JS, Ebersold M, Pypaert M, Delamarre L, Hartley A, et al. (2006) Surface expression

of MHC class II in dendritic cells is controlled by regulated ubiquitination. Nature

444(7115): 115-118.

95. Ohmura-Hoshino M, Matsuki Y, Aoki M, Goto E, Mito M, et al. (2006) Inhibition of

MHC class II expression and immune responses by c-MIR. J Immunol 177(1): 341-354.

96. van Niel G, Wubbolts R, Ten Broeke T, Buschow SI, Ossendorp FA, et al. (2006)

Dendritic cells regulate exposure of MHC class II at their plasma membrane by

oligoubiquitination. Immunity 25(6): 885-894.

97. De Gassart A, Camosseto V, Thibodeau J, Ceppi M, Catalan N, et al. (2008) MHC class II

stabilization at the surface of human dendritic cells is the result of maturation-dependent

MARCH I down-regulation. Proc Natl Acad Sci U S A 105(9): 3491-3496.

98. Aderem A, Underhill DM. (1999) Mechanisms of phagocytosis in macrophages. Annu

Rev Immunol 17: 593-623.

99. Kerrigan AM, Brown GD. (2009) C-type lectins and phagocytosis. Immunobiology .

100. Swanson JA. (2008) Shaping cups into phagosomes and macropinosomes. Nat Rev Mol

Cell Biol 9(8): 639-649.

101. Strzelecka-Kiliszek A, Kwiatkowska K, Sobota A. (2002) Lyn and syk kinases are

sequentially engaged in phagocytosis mediated by fc gamma R. J Immunol 169(12):

6787-6794.

Page 82: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

82

102. Niedergang F, Chavrier P. (2005) Regulation of phagocytosis by rho GTPases. Curr Top

Microbiol Immunol 291: 43-60.

103. Hall AB, Gakidis MA, Glogauer M, Wilsbacher JL, Gao S, et al. (2006) Requirements

for vav guanine nucleotide exchange factors and rho GTPases in FcgammaR- and

complement-mediated phagocytosis. Immunity 24(3): 305-316.

104. Herre J, Marshall AS, Caron E, Edwards AD, Williams DL, et al. (2004) Dectin-1 uses

novel mechanisms for yeast phagocytosis in macrophages. Blood 104(13): 4038-4045.

105. West MA, Wallin RP, Matthews SP, Svensson HG, Zaru R, et al. (2004) Enhanced

dendritic cell antigen capture via toll-like receptor-induced actin remodeling. Science

305(5687): 1153-1157.

106. Zaru R, Ronkina N, Gaestel M, Arthur JS, Watts C. (2007) The MAPK-activated kinase

rsk controls an acute toll-like receptor signaling response in dendritic cells and is

activated through two distinct pathways. Nat Immunol 8(11): 1227-1235.

107. Faure-Andre G, Vargas P, Yuseff MI, Heuze M, Diaz J, et al. (2008) Regulation of

dendritic cell migration by CD74, the MHC class II-associated invariant chain. Science

322(5908): 1705-1710.

108. West MA, Prescott AR, Chan KM, Zhou Z, Rose-John S, et al. (2008) TLR ligand-

induced podosome disassembly in dendritic cells is ADAM17 dependent. J Cell Biol

182(5): 993-1005.

109. Ng LG, Hsu A, Mandell MA, Roediger B, Hoeller C, et al. (2008) Migratory dermal

dendritic cells act as rapid sensors of protozoan parasites. PLoS Pathog 4(11): e1000222.

110. Cresswell P. (1996) Invariant chain structure and MHC class II function. Cell 84(4): 505-

507.

111. Watts C. (2004) The exogenous pathway for antigen presentation on major

histocompatibility complex class II and CD1 molecules. Nat Immunol 5(7): 685-692.

112. Delamarre L, Couture R, Mellman I, Trombetta ES. (2006) Enhancing immunogenicity

by limiting susceptibility to lysosomal proteolysis. J Exp Med 203(9): 2049-2055.

113. Inaba K, Pack M, Inaba M, Sakuta H, Isdell F, et al. (1997) High levels of a major

histocompatibility complex II-self peptide complex on dendritic cells from the T cell

areas of lymph nodes. J Exp Med 186(5): 665-672.

114. Delamarre L, Pack M, Chang H, Mellman I, Trombetta ES. (2005) Differential

lysosomal proteolysis in antigen-presenting cells determines antigen fate. Science

307(5715): 1630-1634.

115. Savina A, Jancic C, Hugues S, Guermonprez P, Vargas P, et al. (2006) NOX2 controls

phagosomal pH to regulate antigen processing during crosspresentation by dendritic

cells. Cell 126(1): 205-218.

Page 83: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

83

116. Albert ML, Sauter B, Bhardwaj N. (1998) Dendritic cells acquire antigen from apoptotic

cells and induce class I-restricted CTLs. Nature 392(6671): 86-89.

117. Schwarz K, Storni T, Manolova V, Didierlaurent A, Sirard JC, et al. (2003) Role of toll-

like receptors in costimulating cytotoxic T cell responses. Eur J Immunol 33(6): 1465-

1470.

118. Datta SK, Redecke V, Prilliman KR, Takabayashi K, Corr M, et al. (2003) A subset of

toll-like receptor ligands induces cross-presentation by bone marrow-derived dendritic

cells. J Immunol 170(8): 4102-4110.

119. Le Bon A, Etchart N, Rossmann C, Ashton M, Hou S, et al. (2003) Cross-priming of

CD8+ T cells stimulated by virus-induced type I interferon. Nat Immunol 4(10): 1009-

1015.

120. Guermonprez P, Saveanu L, Kleijmeer M, Davoust J, Van Endert P, et al. (2003) ER-

phagosome fusion defines an MHC class I cross-presentation compartment in dendritic

cells. Nature 425(6956): 397-402.

121. Houde M, Bertholet S, Gagnon E, Brunet S, Goyette G, et al. (2003) Phagosomes are

competent organelles for antigen cross-presentation. Nature 425(6956): 402-406.

122. Ackerman AL, Giodini A, Cresswell P. (2006) A role for the endoplasmic reticulum

protein retrotranslocation machinery during crosspresentation by dendritic cells.

Immunity 25(4): 607-617.

123. Shen L, Sigal LJ, Boes M, Rock KL. (2004) Important role of cathepsin S in generating

peptides for TAP-independent MHC class I crosspresentation in vivo. Immunity 21(2):

155-165.

124. Allan RS, Waithman J, Bedoui S, Jones CM, Villadangos JA, et al. (2006) Migratory

dendritic cells transfer antigen to a lymph node-resident dendritic cell population for

efficient CTL priming. Immunity 25(1): 153-162.

125. Carbone FR, Belz GT, Heath WR. (2004) Transfer of antigen between migrating and

lymph node-resident DCs in peripheral T-cell tolerance and immunity. Trends Immunol

25(12): 655-658.

126. Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C, et al. (2007)

Differential antigen processing by dendritic cell subsets in vivo. Science 315(5808): 107-

111.

127. Chen L. (2004) Co-inhibitory molecules of the B7-CD28 family in the control of T-cell

immunity. Nat Rev Immunol 4(5): 336-347.

128. Krummel MF, Allison JP. (1995) CD28 and CTLA-4 have opposing effects on the

response of T cells to stimulation. J Exp Med 182(2): 459-465.

129. Harlan DM, Hengartner H, Huang ML, Kang YH, Abe R, et al. (1994) Mice expressing

both B7-1 and viral glycoprotein on pancreatic beta cells along with glycoprotein-

Page 84: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

84

specific transgenic T cells develop diabetes due to a breakdown of T-lymphocyte

unresponsiveness. Proc Natl Acad Sci U S A 91(8): 3137-3141.

130. von Herrath MG, Guerder S, Lewicki H, Flavell RA, Oldstone MB. (1995) Coexpression

of B7-1 and viral ("self") transgenes in pancreatic beta cells can break peripheral

ignorance and lead to spontaneous autoimmune diabetes. Immunity 3(6): 727-738.

131. Adler AJ, Marsh DW, Yochum GS, Guzzo JL, Nigam A, et al. (1998) CD4+ T cell

tolerance to parenchymal self-antigens requires presentation by bone marrow-derived

antigen-presenting cells. J Exp Med 187(10): 1555-1564.

132. Lane P. (2000) Role of OX40 signals in coordinating CD4 T cell selection, migration,

and cytokine differentiation in T helper (th)1 and Th2 cells. J Exp Med 191(2): 201-206.

133. Shuford WW, Klussman K, Tritchler DD, Loo DT, Chalupny J, et al. (1997) 4-1BB

costimulatory signals preferentially induce CD8+ T cell proliferation and lead to the

amplification in vivo of cytotoxic T cell responses. J Exp Med 186(1): 47-55.

134. So T, Lee SW, Croft M. (2008) Immune regulation and control of regulatory T cells by

OX40 and 4-1BB. Cytokine Growth Factor Rev 19(3-4): 253-262.

135. Hutloff A, Dittrich AM, Beier KC, Eljaschewitsch B, Kraft R, et al. (1999) ICOS is an

inducible T-cell co-stimulator structurally and functionally related to CD28. Nature

397(6716): 263-266.

136. Ito T, Yang M, Wang YH, Lande R, Gregorio J, et al. (2007) Plasmacytoid dendritic

cells prime IL-10-producing T regulatory cells by inducible costimulator ligand. J Exp

Med 204(1): 105-115.

137. Dinarello CA. (2005) Interleukin-1beta. Crit Care Med 33(12 Suppl): S460-2.

138. Dinarello CA. (1996) Biologic basis for interleukin-1 in disease. Blood 87(6): 2095-

2147.

139. Parameswaran N, Patial S. (2010) Tumor necrosis factor-alpha signaling in

macrophages. Crit Rev Eukaryot Gene Expr 20(2): 87-103.

140. Beutler B, Grau GE. (1993) Tumor necrosis factor in the pathogenesis of infectious

diseases. Crit Care Med 21(10 Suppl): S423-35.

141. Yarilina A, Park-Min KH, Antoniv T, Hu X, Ivashkiv LB. (2008) TNF activates an

IRF1-dependent autocrine loop leading to sustained expression of chemokines and

STAT1-dependent type I interferon-response genes. Nat Immunol 9(4): 378-387.

142. ISAACS A, LINDENMANN J. (1957) Virus interference. I. the interferon. Proc R Soc

Lond B Biol Sci 147(927): 258-267.

143. Pestka S, Krause CD, Walter MR. (2004) Interferons, interferon-like cytokines, and their

receptors. Immunol Rev 202: 8-32.

Page 85: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

85

144. Takeuchi O, Akira S. (2009) Innate immunity to virus infection. Immunol Rev 227(1):

75-86.

145. Montoya M, Schiavoni G, Mattei F, Gresser I, Belardelli F, et al. (2002) Type I

interferons produced by dendritic cells promote their phenotypic and functional

activation. Blood 99(9): 3263-3271.

146. Luft T, Jefford M, Luetjens P, Toy T, Hochrein H, et al. (2002) Functionally distinct

dendritic cell (DC) populations induced by physiologic stimuli: Prostaglandin E(2)

regulates the migratory capacity of specific DC subsets. Blood 100(4): 1362-1372.

147. Honda K, Sakaguchi S, Nakajima C, Watanabe A, Yanai H, et al. (2003) Selective

contribution of IFN-alpha/beta signaling to the maturation of dendritic cells induced by

double-stranded RNA or viral infection. Proc Natl Acad Sci U S A 100(19): 10872-

10877.

148. Hirano T, Yasukawa K, Harada H, Taga T, Watanabe Y, et al. (1986) Complementary

DNA for a novel human interleukin (BSF-2) that induces B lymphocytes to produce

immunoglobulin. Nature 324(6092): 73-76.

149. Rincon M, Anguita J, Nakamura T, Fikrig E, Flavell RA. (1997) Interleukin (IL)-6

directs the differentiation of IL-4-producing CD4+ T cells. J Exp Med 185(3): 461-469.

150. Diehl S, Anguita J, Hoffmeyer A, Zapton T, Ihle JN, et al. (2000) Inhibition of Th1

differentiation by IL-6 is mediated by SOCS1. Immunity 13(6): 805-815.

151. Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, et al. (2006) The orphan

nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-

17+ T helper cells. Cell 126(6): 1121-1133.

152. Dienz O, Rincon M. (2009) The effects of IL-6 on CD4 T cell responses. Clin Immunol

130(1): 27-33.

153. Kimura A, Kishimoto T. (2010) IL-6: Regulator of Treg/Th17 balance. Eur J Immunol

40(7): 1830-1835.

154. Kobayashi M, Fitz L, Ryan M, Hewick RM, Clark SC, et al. (1989) Identification and

purification of natural killer cell stimulatory factor (NKSF), a cytokine with multiple

biologic effects on human lymphocytes. J Exp Med 170(3): 827-845.

155. Oppmann B, Lesley R, Blom B, Timans JC, Xu Y, et al. (2000) Novel p19 protein

engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as

distinct from IL-12. Immunity 13(5): 715-725.

156. Pflanz S, Timans JC, Cheung J, Rosales R, Kanzler H, et al. (2002) IL-27, a

heterodimeric cytokine composed of EBI3 and p28 protein, induces proliferation of naive

CD4(+) T cells. Immunity 16(6): 779-790.

Page 86: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

86

157. Cua DJ, Sherlock J, Chen Y, Murphy CA, Joyce B, et al. (2003) Interleukin-23 rather

than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain.

Nature 421(6924): 744-748.

158. Re F, Strominger JL. (2001) Toll-like receptor 2 (TLR2) and TLR4 differentially

activate human dendritic cells. J Biol Chem 276(40): 37692-37699.

159. Farrar MA, Schreiber RD. (1993) The molecular cell biology of interferon-gamma and

its receptor. Annu Rev Immunol 11: 571-611.

160. Schroder K, Hertzog PJ, Ravasi T, Hume DA. (2004) Interferon-gamma: An overview of

signals, mechanisms and functions. J Leukoc Biol 75(2): 163-189.

161. Tay SS, Plain KM, Bishop GA. (2009) Role of IL-4 and Th2 responses in allograft

rejection and tolerance. Curr Opin Organ Transplant 14(1): 16-22.

162. Coffman RL, Varkila K, Scott P, Chatelain R. (1991) Role of cytokines in the

differentiation of CD4+ T-cell subsets in vivo. Immunol Rev 123: 189-207.

163. Wynn TA. (2003) IL-13 effector functions. Annu Rev Immunol 21: 425-456.

164. LaPorte SL, Juo ZS, Vaclavikova J, Colf LA, Qi X, et al. (2008) Molecular and

structural basis of cytokine receptor pleiotropy in the interleukin-4/13 system. Cell

132(2): 259-272.

165. Fiorentino DF, Bond MW, Mosmann TR. (1989) Two types of mouse T helper cell. IV.

Th2 clones secrete a factor that inhibits cytokine production by Th1 clones. J Exp Med

170(6): 2081-2095.

166. Kuhn R, Lohler J, Rennick D, Rajewsky K, Muller W. (1993) Interleukin-10-deficient

mice develop chronic enterocolitis. Cell 75(2): 263-274.

167. Moore KW, Vieira P, Fiorentino DF, Trounstine ML, Khan TA, et al. (1990) Homology

of cytokine synthesis inhibitory factor (IL-10) to the epstein-barr virus gene BCRFI.

Science 248(4960): 1230-1234.

168. Annes JP, Munger JS, Rifkin DB. (2003) Making sense of latent TGFbeta activation. J

Cell Sci 116(Pt 2): 217-224.

169. Kulkarni AB, Huh CG, Becker D, Geiser A, Lyght M, et al. (1993) Transforming growth

factor beta 1 null mutation in mice causes excessive inflammatory response and early

death. Proc Natl Acad Sci U S A 90(2): 770-774.

170. Hsieh CS, Macatonia SE, Tripp CS, Wolf SF, O'Garra A, et al. (1993) Development of

TH1 CD4+ T cells through IL-12 produced by listeria-induced macrophages. Science

260(5107): 547-549.

171. Macatonia SE, Hosken NA, Litton M, Vieira P, Hsieh CS, et al. (1995) Dendritic cells

produce IL-12 and direct the development of Th1 cells from naive CD4+ T cells. J

Immunol 154(10): 5071-5079.

Page 87: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

87

172. Manetti R, Parronchi P, Giudizi MG, Piccinni MP, Maggi E, et al. (1993) Natural killer

cell stimulatory factor (interleukin 12 [IL-12]) induces T helper type 1 (Th1)-specific

immune responses and inhibits the development of IL-4-producing th cells. J Exp Med

177(4): 1199-1204.

173. Schulz O, Edwards AD, Schito M, Aliberti J, Manickasingham S, et al. (2000) CD40

triggering of heterodimeric IL-12 p70 production by dendritic cells in vivo requires a

microbial priming signal. Immunity 13(4): 453-462.

174. Reis e Sousa C, Hieny S, Scharton-Kersten T, Jankovic D, Charest H, et al. (1997) In

vivo microbial stimulation induces rapid CD40 ligand-independent production of

interleukin 12 by dendritic cells and their redistribution to T cell areas. J Exp Med

186(11): 1819-1829.

175. Dorman SE, Holland SM. (2000) Interferon-gamma and interleukin-12 pathway defects

and human disease. Cytokine Growth Factor Rev 11(4): 321-333.

176. Happel KI, Dubin PJ, Zheng M, Ghilardi N, Lockhart C, et al. (2005) Divergent roles of

IL-23 and IL-12 in host defense against klebsiella pneumoniae. J Exp Med 202(6): 761-

769.

177. Park H, Li Z, Yang XO, Chang SH, Nurieva R, et al. (2005) A distinct lineage of CD4 T

cells regulates tissue inflammation by producing interleukin 17. Nat Immunol 6(11):

1133-1141.

178. Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, et al. (2005) IL-23

drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med

201(2): 233-240.

179. Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. (2006) TGFbeta in

the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-

producing T cells. Immunity 24(2): 179-189.

180. Volpe E, Servant N, Zollinger R, Bogiatzi SI, Hupe P, et al. (2008) A critical function

for transforming growth factor-beta, interleukin 23 and proinflammatory cytokines in

driving and modulating human T(H)-17 responses. Nat Immunol 9(6): 650-657.

181. Murphy PM, Baggiolini M, Charo IF, Hebert CA, Horuk R, et al. (2000) International

union of pharmacology. XXII. nomenclature for chemokine receptors. Pharmacol Rev

52(1): 145-176.

182. Charo IF, Ransohoff RM. (2006) The many roles of chemokines and chemokine

receptors in inflammation. N Engl J Med 354(6): 610-621.

183. Murphy PM. (1994) The molecular biology of leukocyte chemoattractant receptors.

Annu Rev Immunol 12: 593-633.

184. Mantovani A, Bonecchi R, Locati M. (2006) Tuning inflammation and immunity by

chemokine sequestration: Decoys and more. Nat Rev Immunol 6(12): 907-918.

Page 88: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

88

185. Forster R, Schubel A, Breitfeld D, Kremmer E, Renner-Muller I, et al. (1999) CCR7

coordinates the primary immune response by establishing functional microenvironments

in secondary lymphoid organs. Cell 99(1): 23-33.

186. Zou YR, Kottmann AH, Kuroda M, Taniuchi I, Littman DR. (1998) Function of the

chemokine receptor CXCR4 in haematopoiesis and in cerebellar development. Nature

393(6685): 595-599.

187. Belge KU, Dayyani F, Horelt A, Siedlar M, Frankenberger M, et al. (2002) The

proinflammatory CD14+CD16+DR++ monocytes are a major source of TNF. J Immunol

168(7): 3536-3542.

188. Frankenberger M, Sternsdorf T, Pechumer H, Pforte A, Ziegler-Heitbrock HW. (1996)

Differential cytokine expression in human blood monocyte subpopulations: A

polymerase chain reaction analysis. Blood 87(1): 373-377.

189. Randolph GJ, Jakubzick C, Qu C. (2008) Antigen presentation by monocytes and

monocyte-derived cells. Curr Opin Immunol 20(1): 52-60.

190. Ziegler-Heitbrock L. (2007) The CD14+ CD16+ blood monocytes: Their role in

infection and inflammation. J Leukoc Biol 81(3): 584-592.

191. Swirski FK, Pittet MJ, Kircher MF, Aikawa E, Jaffer FA, et al. (2006) Monocyte

accumulation in mouse atherogenesis is progressive and proportional to extent of disease.

Proc Natl Acad Sci U S A 103(27): 10340-10345.

192. Arnold L, Henry A, Poron F, Baba-Amer Y, van Rooijen N, et al. (2007) Inflammatory

monocytes recruited after skeletal muscle injury switch into antiinflammatory

macrophages to support myogenesis. J Exp Med 204(5): 1057-1069.

193. Sawyer RT. (1986) The significance of local resident pulmonary alveolar macrophage

proliferation to population renewal. J Leukoc Biol 39(1): 77-87.

194. Naik SH, Metcalf D, van Nieuwenhuijze A, Wicks I, Wu L, et al. (2006) Intrasplenic

steady-state dendritic cell precursors that are distinct from monocytes. Nat Immunol 7(6):

663-671.

195. Leon B, Lopez-Bravo M, Ardavin C. (2007) Monocyte-derived dendritic cells formed at

the infection site control the induction of protective T helper 1 responses against

leishmania. Immunity 26(4): 519-531.

196. Randolph GJ, Sanchez-Schmitz G, Liebman RM, Schakel K. (2002) The CD16(+)

(FcgammaRIII(+)) subset of human monocytes preferentially becomes migratory

dendritic cells in a model tissue setting. J Exp Med 196(4): 517-527.

197. Verreck FA, de Boer T, Langenberg DM, van der Zanden L, Ottenhoff TH. (2006)

Phenotypic and functional profiling of human proinflammatory type-1 and anti-

inflammatory type-2 macrophages in response to microbial antigens and IFN-gamma-

and CD40L-mediated costimulation. J Leukoc Biol 79(2): 285-293.

Page 89: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

89

198. Bosisio D, Polentarutti N, Sironi M, Bernasconi S, Miyake K, et al. (2002) Stimulation

of toll-like receptor 4 expression in human mononuclear phagocytes by interferon-

gamma: A molecular basis for priming and synergism with bacterial lipopolysaccharide.

Blood 99(9): 3427-3431.

199. Scotton CJ, Martinez FO, Smelt MJ, Sironi M, Locati M, et al. (2005) Transcriptional

profiling reveals complex regulation of the monocyte IL-1 beta system by IL-13. J

Immunol 174(2): 834-845.

200. Duffield JS, Forbes SJ, Constandinou CM, Clay S, Partolina M, et al. (2005) Selective

depletion of macrophages reveals distinct, opposing roles during liver injury and repair. J

Clin Invest 115(1): 56-65.

201. Gibbs DF, Warner RL, Weiss SJ, Johnson KJ, Varani J. (1999) Characterization of

matrix metalloproteinases produced by rat alveolar macrophages. Am J Respir Cell Mol

Biol 20(6): 1136-1144.

202. Gratchev A, Guillot P, Hakiy N, Politz O, Orfanos CE, et al. (2001) Alternatively

activated macrophages differentially express fibronectin and its splice variants and the

extracellular matrix protein betaIG-H3. Scand J Immunol 53(4): 386-392.

203. Taylor PR, Martinez-Pomares L, Stacey M, Lin HH, Brown GD, et al. (2005)

Macrophage receptors and immune recognition. Annu Rev Immunol 23: 901-944.

204. Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, et al. (2005) Interleukin

17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type

1 and 2 lineages. Nat Immunol 6(11): 1123-1132.

205. Steinman RM, Hawiger D, Nussenzweig MC. (2003) Tolerogenic dendritic cells. Annu

Rev Immunol 21: 685-711.

206. Maldonado RA, von Andrian UH. (2010) How tolerogenic dendritic cells induce

regulatory T cells. Adv Immunol 108: 111-165.

207. Huang Q, Liu D, Majewski P, Schulte LC, Korn JM, et al. (2001) The plasticity of

dendritic cell responses to pathogens and their components. Science 294(5543): 870-875.

208. Verhasselt V, Buelens C, Willems F, De Groote D, Haeffner-Cavaillon N, et al. (1997)

Bacterial lipopolysaccharide stimulates the production of cytokines and the expression of

costimulatory molecules by human peripheral blood dendritic cells: Evidence for a

soluble CD14-dependent pathway. J Immunol 158(6): 2919-2925.

209. Dubois B, Massacrier C, Vanbervliet B, Fayette J, Briere F, et al. (1998) Critical role of

IL-12 in dendritic cell-induced differentiation of naive B lymphocytes. J Immunol

161(5): 2223-2231.

210. Fayette J, Durand I, Bridon JM, Arpin C, Dubois B, et al. (1998) Dendritic cells enhance

the differentiation of naive B cells into plasma cells in vitro. Scand J Immunol 48(6):

563-570.

Page 90: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

90

211. Urban BC, Cordery D, Shafi MJ, Bull PC, Newbold CI, et al. (2006) The frequency of

BDCA3-positive dendritic cells is increased in the peripheral circulation of kenyan

children with severe malaria. Infect Immun 74(12): 6700-6706.

212. Bachem A, Guttler S, Hartung E, Ebstein F, Schaefer M, et al. (2010) Superior antigen

cross-presentation and XCR1 expression define human CD11c+CD141+ cells as

homologues of mouse CD8+ dendritic cells. J Exp Med 207(6): 1273-1281.

213. Crozat K, Guiton R, Contreras V, Feuillet V, Dutertre CA, et al. (2010) The XC

chemokine receptor 1 is a conserved selective marker of mammalian cells homologous to

mouse CD8alpha+ dendritic cells. J Exp Med 207(6): 1283-1292.

214. Siegal FP, Kadowaki N, Shodell M, Fitzgerald-Bocarsly PA, Shah K, et al. (1999) The

nature of the principal type 1 interferon-producing cells in human blood. Science

284(5421): 1835-1837.

215. Jego G, Palucka AK, Blanck JP, Chalouni C, Pascual V, et al. (2003) Plasmacytoid

dendritic cells induce plasma cell differentiation through type I interferon and interleukin

6. Immunity 19(2): 225-234.

216. Poeck H, Wagner M, Battiany J, Rothenfusser S, Wellisch D, et al. (2004) Plasmacytoid

dendritic cells, antigen, and CpG-C license human B cells for plasma cell differentiation

and immunoglobulin production in the absence of T-cell help. Blood 103(8): 3058-3064.

217. Le Bon A, Schiavoni G, D'Agostino G, Gresser I, Belardelli F, et al. (2001) Type i

interferons potently enhance humoral immunity and can promote isotype switching by

stimulating dendritic cells in vivo. Immunity 14(4): 461-470.

218. Di Pucchio T, Chatterjee B, Smed-Sorensen A, Clayton S, Palazzo A, et al. (2008) Direct

proteasome-independent cross-presentation of viral antigen by plasmacytoid dendritic

cells on major histocompatibility complex class I. Nat Immunol 9(5): 551-557.

219. Ito T, Amakawa R, Inaba M, Hori T, Ota M, et al. (2004) Plasmacytoid dendritic cells

regulate th cell responses through OX40 ligand and type I IFNs. J Immunol 172(7):

4253-4259.

220. Mahnke K, Schmitt E, Bonifaz L, Enk AH, Jonuleit H. (2002) Immature, but not

inactive: The tolerogenic function of immature dendritic cells. Immunol Cell Biol 80(5):

477-483.

221. Ravichandran KS. (2010) Find-me and eat-me signals in apoptotic cell clearance:

Progress and conundrums. J Exp Med 207(9): 1807-1817.

222. Schulz O, Reis e Sousa C. (2002) Cross-presentation of cell-associated antigens by

CD8alpha+ dendritic cells is attributable to their ability to internalize dead cells.

Immunology 107(2): 183-189.

223. Inaba K, Turley S, Yamaide F, Iyoda T, Mahnke K, et al. (1998) Efficient presentation of

phagocytosed cellular fragments on the major histocompatibility complex class II

products of dendritic cells. J Exp Med 188(11): 2163-2173.

Page 91: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

91

224. Huang FP, Platt N, Wykes M, Major JR, Powell TJ, et al. (2000) A discrete

subpopulation of dendritic cells transports apoptotic intestinal epithelial cells to T cell

areas of mesenteric lymph nodes. J Exp Med 191(3): 435-444.

225. Hemmi H, Yoshino M, Yamazaki H, Naito M, Iyoda T, et al. (2001) Skin antigens in the

steady state are trafficked to regional lymph nodes by transforming growth factor-beta1-

dependent cells. Int Immunol 13(5): 695-704.

226. Manavalan JS, Rossi PC, Vlad G, Piazza F, Yarilina A, et al. (2003) High expression of

ILT3 and ILT4 is a general feature of tolerogenic dendritic cells. Transpl Immunol 11(3-

4): 245-258.

227. Munn DH, Sharma MD, Lee JR, Jhaver KG, Johnson TS, et al. (2002) Potential

regulatory function of human dendritic cells expressing indoleamine 2,3-dioxygenase.

Science 297(5588): 1867-1870.

228. Munn DH, Sharma MD, Baban B, Harding HP, Zhang Y, et al. (2005) GCN2 kinase in T

cells mediates proliferative arrest and anergy induction in response to indoleamine 2,3-

dioxygenase. Immunity 22(5): 633-642.

229. Abreu MT. (2010) Toll-like receptor signalling in the intestinal epithelium: How

bacterial recognition shapes intestinal function. Nat Rev Immunol 10(2): 131-144.

230. Rimoldi M, Chieppa M, Salucci V, Avogadri F, Sonzogni A, et al. (2005) Intestinal

immune homeostasis is regulated by the crosstalk between epithelial cells and dendritic

cells. Nat Immunol 6(5): 507-514.

231. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. (2004)

Recognition of commensal microflora by toll-like receptors is required for intestinal

homeostasis. Cell 118(2): 229-241.

232. Iwasaki A. (2007) Division of labor by dendritic cells. Cell 128(3): 435-436.

233. Bamboat ZM, Stableford JA, Plitas G, Burt BM, Nguyen HM, et al. (2009) Human liver

dendritic cells promote T cell hyporesponsiveness. J Immunol 182(4): 1901-1911.

234. Thomson AW, Knolle PA. (2010) Antigen-presenting cell function in the tolerogenic

liver environment. Nat Rev Immunol 10(11): 753-766.

235. Iliev ID, Matteoli G, Rescigno M. (2007) The yin and yang of intestinal epithelial cells

in controlling dendritic cell function. J Exp Med 204(10): 2253-2257.

236. Liu YJ, Soumelis V, Watanabe N, Ito T, Wang YH, et al. (2007) TSLP: An epithelial

cell cytokine that regulates T cell differentiation by conditioning dendritic cell

maturation. Annu Rev Immunol 25: 193-219.

237. Sato A, Iwasaki A. (2004) Induction of antiviral immunity requires toll-like receptor

signaling in both stromal and dendritic cell compartments. Proc Natl Acad Sci U S A

101(46): 16274-16279.

Page 92: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

92

238. Nolte MA, Leibundgut-Landmann S, Joffre O, Reis e Sousa C. (2007) Dendritic cell

quiescence during systemic inflammation driven by LPS stimulation of radioresistant

cells in vivo. J Exp Med 204(6): 1487-1501.

239. Richter K, Haslbeck M, Buchner J. (2010) The heat shock response: Life on the verge of

death. Mol Cell 40(2): 253-266.

240. Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. (2010) HMGB1 and RAGE in

inflammation and cancer. Annu Rev Immunol 28: 367-388.

241. Lutz MB, Schuler G. (2002) Immature, semi-mature and fully mature dendritic cells:

Which signals induce tolerance or immunity? Trends Immunol 23(9): 445-449.

242. Menges M, Rossner S, Voigtlander C, Schindler H, Kukutsch NA, et al. (2002)

Repetitive injections of dendritic cells matured with tumor necrosis factor alpha induce

antigen-specific protection of mice from autoimmunity. J Exp Med 195(1): 15-21.

243. Verginis P, Li HS, Carayanniotis G. (2005) Tolerogenic semimature dendritic cells

suppress experimental autoimmune thyroiditis by activation of thyroglobulin-specific

CD4+CD25+ T cells. J Immunol 174(11): 7433-7439.

244. Chomarat P, Dantin C, Bennett L, Banchereau J, Palucka AK. (2003) TNF skews

monocyte differentiation from macrophages to dendritic cells. J Immunol 171(5): 2262-

2269.

245. Kadowaki N, Antonenko S, Lau JY, Liu YJ. (2000) Natural interferon alpha/beta-

producing cells link innate and adaptive immunity. J Exp Med 192(2): 219-226.

246. Kitamura H, Kamon H, Sawa S, Park SJ, Katunuma N, et al. (2005) IL-6-STAT3

controls intracellular MHC class II alphabeta dimer level through cathepsin S activity in

dendritic cells. Immunity 23(5): 491-502.

247. Hegde S, Pahne J, Smola-Hess S. (2004) Novel immunosuppressive properties of

interleukin-6 in dendritic cells: Inhibition of NF-kappaB binding activity and CCR7

expression. FASEB J 18(12): 1439-1441.

248. Chomarat P, Banchereau J, Davoust J, Palucka AK. (2000) IL-6 switches the

differentiation of monocytes from dendritic cells to macrophages. Nat Immunol 1(6):

510-514.

249. de Waal Malefyt R, Haanen J, Spits H, Roncarolo MG, te Velde A, et al. (1991)

Interleukin 10 (IL-10) and viral IL-10 strongly reduce antigen-specific human T cell

proliferation by diminishing the antigen-presenting capacity of monocytes via

downregulation of class II major histocompatibility complex expression. J Exp Med

174(4): 915-924.

250. Ding L, Linsley PS, Huang LY, Germain RN, Shevach EM. (1993) IL-10 inhibits

macrophage costimulatory activity by selectively inhibiting the up-regulation of B7

expression. J Immunol 151(3): 1224-1234.

Page 93: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

93

251. Knodler A, Schmidt SM, Bringmann A, Weck MM, Brauer KM, et al. (2009) Post-

transcriptional regulation of adapter molecules by IL-10 inhibits TLR-mediated

activation of antigen-presenting cells. Leukemia 23(3): 535-544.

252. Adler HS, Steinbrink K. (2007) Tolerogenic dendritic cells in health and disease: Friend

and foe! Eur J Dermatol 17(6): 476-491.

253. Gallup JL, Sachs JD. (2001) The economic burden of malaria. Am J Trop Med Hyg

64(1-2 Suppl): 85-96.

254. Teklehaimanot A, Mejia P. (2008) Malaria and poverty. Ann N Y Acad Sci 1136: 32-37.

255. World health Organization. (2008) World malaria report 2008. .

256. World health Organization. (2010) World malaria report 2010. .

257. Rogerson SJ, Mwapasa V, Meshnick SR. (2007) Malaria in pregnancy: Linking

immunity and pathogenesis to prevention. Am J Trop Med Hyg 77(6 Suppl): 14-22.

258. Jongwutiwes S, Putaporntip C, Iwasaki T, Sata T, Kanbara H. (2004) Naturally acquired

plasmodium knowlesi malaria in human, thailand. Emerg Infect Dis 10(12): 2211-2213.

259. Singh B, Kim Sung L, Matusop A, Radhakrishnan A, Shamsul SS, et al. (2004) A large

focus of naturally acquired plasmodium knowlesi infections in human beings. Lancet

363(9414): 1017-1024.

260. Zhu HM, Li J, Zheng H. (2006) Human natural infection of plasmodium knowlesi.

Zhongguo Ji Sheng Chong Xue Yu Ji Sheng Chong Bing Za Zhi 24(1): 70-71.

261. Cox-Singh J, Davis TM, Lee KS, Shamsul SS, Matusop A, et al. (2008) Plasmodium

knowlesi malaria in humans is widely distributed and potentially life threatening. Clin

Infect Dis 46(2): 165-171.

262. Luchavez J, Espino F, Curameng P, Espina R, Bell D, et al. (2008) Human infections

with plasmodium knowlesi, the philippines. Emerg Infect Dis 14(5): 811-813.

263. Ng OT, Ooi EE, Lee CC, Lee PJ, Ng LC, et al. (2008) Naturally acquired human

plasmodium knowlesi infection, singapore. Emerg Infect Dis 14(5): 814-816.

264. Vythilingam I, Noorazian YM, Huat TC, Jiram AI, Yusri YM, et al. (2008) Plasmodium

knowlesi in humans, macaques and mosquitoes in peninsular malaysia. Parasit Vectors

1(1): 26.

265. Slater AF, Swiggard WJ, Orton BR, Flitter WD, Goldberg DE, et al. (1991) An iron-

carboxylate bond links the heme units of malaria pigment. Proc Natl Acad Sci U S A

88(2): 325-329.

266. Jaramillo M, Plante I, Ouellet N, Vandal K, Tessier PA, et al. (2004) Hemozoin-

inducible proinflammatory events in vivo: Potential role in malaria infection. J Immunol

172(5): 3101-3110.

Page 94: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

94

267. Jaramillo M, Godbout M, Olivier M. (2005) Hemozoin induces macrophage chemokine

expression through oxidative stress-dependent and -independent mechanisms. J Immunol

174(1): 475-484.

268. Huy NT, Trang DT, Kariu T, Sasai M, Saida K, et al. (2006) Leukocyte activation by

malarial pigment. Parasitol Int 55(1): 75-81.

269. Ropert C, Franklin BS, Gazzinelli RT. (2008) Role of TLRs/MyD88 in host resistance

and pathogenesis during protozoan infection: Lessons from malaria. Semin

Immunopathol 30(1): 41-51.

270. Campos MA, Almeida IC, Takeuchi O, Akira S, Valente EP, et al. (2001) Activation of

toll-like receptor-2 by glycosylphosphatidylinositol anchors from a protozoan parasite. J

Immunol 167(1): 416-423.

271. Oliveira AC, Peixoto JR, de Arruda LB, Campos MA, Gazzinelli RT, et al. (2004)

Expression of functional TLR4 confers proinflammatory responsiveness to trypanosoma

cruzi glycoinositolphospholipids and higher resistance to infection with T. cruzi. J

Immunol 173(9): 5688-5696.

272. Coban C, Ishii KJ, Kawai T, Hemmi H, Sato S, et al. (2005) Toll-like receptor 9

mediates innate immune activation by the malaria pigment hemozoin. J Exp Med 201(1):

19-25.

273. Parroche P, Lauw FN, Goutagny N, Latz E, Monks BG, et al. (2007) Malaria hemozoin

is immunologically inert but radically enhances innate responses by presenting malaria

DNA to toll-like receptor 9. Proc Natl Acad Sci U S A 104(6): 1919-1924.

274. Wu X, Gowda NM, Kumar S, Gowda DC. (2010) Protein-DNA complex is the exclusive

malaria parasite component that activates dendritic cells and triggers innate immune

responses. J Immunol 184(8): 4338-4348.

275. Gowda NM, Wu X, Gowda DC. (2011) The nucleosome (histone-DNA complex) is the

TLR9-specific immunostimulatory component of plasmodium falciparum that activates

DCs. PLoS One 6(6): e20398.

276. Mockenhaupt FP, Cramer JP, Hamann L, Stegemann MS, Eckert J, et al. (2006) Toll-like

receptor (TLR) polymorphisms in african children: Common TLR-4 variants predispose

to severe malaria. Proc Natl Acad Sci U S A 103(1): 177-182.

277. Mockenhaupt FP, Hamann L, von Gaertner C, Bedu-Addo G, von Kleinsorgen C, et al.

(2006) Common polymorphisms of toll-like receptors 4 and 9 are associated with the

clinical manifestation of malaria during pregnancy. J Infect Dis 194(2): 184-188.

278. McCall MB, Netea MG, Hermsen CC, Jansen T, Jacobs L, et al. (2007) Plasmodium

falciparum infection causes proinflammatory priming of human TLR responses. J

Immunol 179(1): 162-171.

279. Ocana-Morgner C, Mota MM, Rodriguez A. (2003) Malaria blood stage suppression of

liver stage immunity by dendritic cells. J Exp Med 197(2): 143-151.

Page 95: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

95

280. Urban BC, Roberts DJ. (2003) Inhibition of T cell function during malaria: Implications

for immunology and vaccinology. J Exp Med 197(2): 137-141.

281. Urban BC, Mwangi T, Ross A, Kinyanjui S, Mosobo M, et al. (2001) Peripheral blood

dendritic cells in children with acute plasmodium falciparum malaria. Blood 98(9): 2859-

2861.

282. Coban C, Ishii KJ, Horii T, Akira S. (2007) Manipulation of host innate immune

responses by the malaria parasite. Trends Microbiol 15(6): 271-278.

283. Pichyangkul S, Yongvanitchit K, Kum-arb U, Hemmi H, Akira S, et al. (2004) Malaria

blood stage parasites activate human plasmacytoid dendritic cells and murine dendritic

cells through a toll-like receptor 9-dependent pathway. J Immunol 172(8): 4926-4933.

284. Pichyangkul S, Saengkrai P, Webster HK. (1994) Plasmodium falciparum pigment

induces monocytes to release high levels of tumor necrosis factor-alpha and interleukin-1

beta. Am J Trop Med Hyg 51(4): 430-435.

285. Keller CC, Yamo O, Ouma C, Ong'echa JM, Ounah D, et al. (2006) Acquisition of

hemozoin by monocytes down-regulates interleukin-12 p40 (IL-12p40) transcripts and

circulating IL-12p70 through an IL-10-dependent mechanism: In vivo and in vitro

findings in severe malarial anemia. Infect Immun 74(9): 5249-5260.

286. Skorokhod OA, Alessio M, Mordmuller B, Arese P, Schwarzer E. (2004) Hemozoin

(malarial pigment) inhibits differentiation and maturation of human monocyte-derived

dendritic cells: A peroxisome proliferator-activated receptor-gamma-mediated effect. J

Immunol 173(6): 4066-4074.

287. Schwarzer E, Alessio M, Ulliers D, Arese P. (1998) Phagocytosis of the malarial

pigment, hemozoin, impairs expression of major histocompatibility complex class II

antigen, CD54, and CD11c in human monocytes. Infect Immun 66(4): 1601-1606.

288. Urban BC, Ferguson DJ, Pain A, Willcox N, Plebanski M, et al. (1999) Plasmodium

falciparum-infected erythrocytes modulate the maturation of dendritic cells. Nature

400(6739): 73-77.

289. Coban C, Ishii KJ, Sullivan DJ, Kumar N. (2002) Purified malaria pigment (hemozoin)

enhances dendritic cell maturation and modulates the isotype of antibodies induced by a

DNA vaccine. Infect Immun 70(7): 3939-3943.

290. Urban BC, Todryk S. (2006) Malaria pigment paralyzes dendritic cells. J Biol 5(2): 4.

291. Bousheri S, Cao H. (2008) New insight into the role of dendritic cells in malaria immune

pathogenesis. Trends Parasitol 24(5): 199-200.

292. Luty AJ, Perkins DJ, Lell B, Schmidt-Ott R, Lehman LG, et al. (2000) Low interleukin-

12 activity in severe plasmodium falciparum malaria. Infect Immun 68(7): 3909-3915.

Page 96: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

96

293. Prakash D, Fesel C, Jain R, Cazenave PA, Mishra GC, et al. (2006) Clusters of cytokines

determine malaria severity in plasmodium falciparum-infected patients from endemic

areas of central india. J Infect Dis 194(2): 198-207.

294. Lyke KE, Burges R, Cissoko Y, Sangare L, Dao M, et al. (2004) Serum levels of the

proinflammatory cytokines interleukin-1 beta (IL-1beta), IL-6, IL-8, IL-10, tumor

necrosis factor alpha, and IL-12(p70) in malian children with severe plasmodium

falciparum malaria and matched uncomplicated malaria or healthy controls. Infect

Immun 72(10): 5630-5637.

295. Dolo A, Modiano D, Maiga B, Daou M, Dolo G, et al. (2005) Difference in susceptibility

to malaria between two sympatric ethnic groups in mali. Am J Trop Med Hyg 72(3):

243-248.

296. Modiano D, Petrarca V, Sirima BS, Nebie I, Diallo D, et al. (1996) Different response to

plasmodium falciparum malaria in west african sympatric ethnic groups. Proc Natl Acad

Sci U S A 93(23): 13206-13211.

297. Paganotti GM, Babiker HA, Modiano D, Sirima BS, Verra F, et al. (2004) Genetic

complexity of plasmodium falciparum in two ethnic groups of burkina faso with marked

differences in susceptibility to malaria. Am J Trop Med Hyg 71(2): 173-178.

298. Farouk SE, Dolo A, Bereczky S, Kouriba B, Maiga B, et al. (2005) Different antibody-

and cytokine-mediated responses to plasmodium falciparum parasite in two sympatric

ethnic tribes living in mali. Microbes Infect 7(1): 110-117.

299. Torcia MG, Santarlasci V, Cosmi L, Clemente A, Maggi L, et al. (2008) Functional

deficit of T regulatory cells in fulani, an ethnic group with low susceptibility to

plasmodium falciparum malaria. Proc Natl Acad Sci U S A 105(2): 646-651.

300. Joseph A, Brasington R, Kahl L, Ranganathan P, Cheng TP, et al. (2010) Immunologic

rheumatic disorders. J Allergy Clin Immunol 125(2 Suppl 2): S204-15.

301. Strand V, Kimberly R, Isaacs JD. (2007) Biologic therapies in rheumatology: Lessons

learned, future directions. Nat Rev Drug Discov 6(1): 75-92.

302. Smolen JS, Aletaha D, Koeller M, Weisman MH, Emery P. (2007) New therapies for

treatment of rheumatoid arthritis. Lancet 370(9602): 1861-1874.

303. Klareskog L, van der Heijde D, de Jager JP, Gough A, Kalden J, et al. (2004)

Therapeutic effect of the combination of etanercept and methotrexate compared with

each treatment alone in patients with rheumatoid arthritis: Double-blind randomised

controlled trial. Lancet 363(9410): 675-681.

304. Mulherin D, Fitzgerald O, Bresnihan B. (1996) Synovial tissue macrophage populations

and articular damage in rheumatoid arthritis. Arthritis Rheum 39(1): 115-124.

305. Kinne RW, Stuhlmuller B, Burmester GR. (2007) Cells of the synovium in rheumatoid

arthritis. macrophages. Arthritis Res Ther 9(6): 224.

Page 97: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

97

306. McInnes IB, Schett G. (2007) Cytokines in the pathogenesis of rheumatoid arthritis. Nat

Rev Immunol 7(6): 429-442.

307. Schett G, Stach C, Zwerina J, Voll R, Manger B. (2008) How antirheumatic drugs

protect joints from damage in rheumatoid arthritis. Arthritis Rheum 58(10): 2936-2948.

308. Bondeson J, Wainwright SD, Lauder S, Amos N, Hughes CE. (2006) The role of

synovial macrophages and macrophage-produced cytokines in driving aggrecanases,

matrix metalloproteinases, and other destructive and inflammatory responses in

osteoarthritis. Arthritis Res Ther 8(6): R187.

309. Gravallese EM, Manning C, Tsay A, Naito A, Pan C, et al. (2000) Synovial tissue in

rheumatoid arthritis is a source of osteoclast differentiation factor. Arthritis Rheum

43(2): 250-258.

310. Hamilton JA, Filonzi EL, Ianches G. (1993) Regulation of macrophage colony-

stimulating factor (M-CSF) production in cultured human synovial fibroblasts. Growth

Factors 9(2): 157-165.

311. Nakano K, Okada Y, Saito K, Tanikawa R, Sawamukai N, et al. (2007) Rheumatoid

synovial endothelial cells produce macrophage colony-stimulating factor leading to

osteoclastogenesis in rheumatoid arthritis. Rheumatology (Oxford) 46(4): 597-603.

312. Dougall WC, Glaccum M, Charrier K, Rohrbach K, Brasel K, et al. (1999) RANK is

essential for osteoclast and lymph node development. Genes Dev 13(18): 2412-2424.

313. Danks L, Sabokbar A, Gundle R, Athanasou NA. (2002) Synovial macrophage-

osteoclast differentiation in inflammatory arthritis. Ann Rheum Dis 61(10): 916-921.

314. Pettit AR, MacDonald KP, O'Sullivan B, Thomas R. (2000) Differentiated dendritic cells

expressing nuclear RelB are predominantly located in rheumatoid synovial tissue

perivascular mononuclear cell aggregates. Arthritis Rheum 43(4): 791-800.

315. Leung BP, Conacher M, Hunter D, McInnes IB, Liew FY, et al. (2002) A novel dendritic

cell-induced model of erosive inflammatory arthritis: Distinct roles for dendritic cells in

T cell activation and induction of local inflammation. J Immunol 169(12): 7071-7077.

316. Nishimura K, Sugiyama D, Kogata Y, Tsuji G, Nakazawa T, et al. (2007) Meta-analysis:

Diagnostic accuracy of anti-cyclic citrullinated peptide antibody and rheumatoid factor

for rheumatoid arthritis. Ann Intern Med 146(11): 797-808.

317. Kuhn KA, Kulik L, Tomooka B, Braschler KJ, Arend WP, et al. (2006) Antibodies

against citrullinated proteins enhance tissue injury in experimental autoimmune arthritis.

J Clin Invest 116(4): 961-973.

318. Schellekens GA, Visser H, de Jong BA, van den Hoogen FH, Hazes JM, et al. (2000)

The diagnostic properties of rheumatoid arthritis antibodies recognizing a cyclic

citrullinated peptide. Arthritis Rheum 43(1): 155-163.

Page 98: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

98

319. van Gaalen FA, Linn-Rasker SP, van Venrooij WJ, de Jong BA, Breedveld FC, et al.

(2004) Autoantibodies to cyclic citrullinated peptides predict progression to rheumatoid

arthritis in patients with undifferentiated arthritis: A prospective cohort study. Arthritis

Rheum 50(3): 709-715.

320. Jongbloed SL, Lebre MC, Fraser AR, Gracie JA, Sturrock RD, et al. (2006) Enumeration

and phenotypical analysis of distinct dendritic cell subsets in psoriatic arthritis and

rheumatoid arthritis. Arthritis Res Ther 8(1): R15.

321. Lebre MC, Jongbloed SL, Tas SW, Smeets TJ, McInnes IB, et al. (2008) Rheumatoid

arthritis synovium contains two subsets of CD83-DC-LAMP- dendritic cells with distinct

cytokine profiles. Am J Pathol 172(4): 940-950.

322. Roelofs MF, Joosten LA, Abdollahi-Roodsaz S, van Lieshout AW, Sprong T, et al.

(2005) The expression of toll-like receptors 3 and 7 in rheumatoid arthritis synovium is

increased and costimulation of toll-like receptors 3, 4, and 7/8 results in synergistic

cytokine production by dendritic cells. Arthritis Rheum 52(8): 2313-2322.

323. Radstake TR, van Lieshout AW, van Riel PL, van den Berg WB, Adema GJ. (2005)

Dendritic cells, fc{gamma} receptors, and toll-like receptors: Potential allies in the battle

against rheumatoid arthritis. Ann Rheum Dis 64(11): 1532-1538.

324. Dehlin M, Bokarewa M, Rottapel R, Foster SJ, Magnusson M, et al. (2008) Intra-

articular fms-like tyrosine kinase 3 ligand expression is a driving force in induction and

progression of arthritis. PLoS One 3(11): e3633.

325. Maraskovsky E, Daro E, Roux E, Teepe M, Maliszewski CR, et al. (2000) In vivo

generation of human dendritic cell subsets by Flt3 ligand. Blood 96(3): 878-884.

326. Kremer JM, Westhovens R, Leon M, Di Giorgio E, Alten R, et al. (2003) Treatment of

rheumatoid arthritis by selective inhibition of T-cell activation with fusion protein

CTLA4Ig. N Engl J Med 349(20): 1907-1915.

327. Abdollahi-Roodsaz S, Joosten LA, Roelofs MF, Radstake TR, Matera G, et al. (2007)

Inhibition of toll-like receptor 4 breaks the inflammatory loop in autoimmune destructive

arthritis. Arthritis Rheum 56(9): 2957-2967.

328. Abdollahi-Roodsaz S, Joosten LA, Koenders MI, Devesa I, Roelofs MF, et al. (2008)

Stimulation of TLR2 and TLR4 differentially skews the balance of T cells in a mouse

model of arthritis. J Clin Invest 118(1): 205-216.

329. Genovese MC, Becker JC, Schiff M, Luggen M, Sherrer Y, et al. (2005) Abatacept for

rheumatoid arthritis refractory to tumor necrosis factor alpha inhibition. N Engl J Med

353(11): 1114-1123.

330. Hultqvist M, Nandakumar KS, Bjorklund U, Holmdahl R. (2009) The novel small

molecule drug rabeximod is effective in reducing disease severity of mouse models of

autoimmune disorders. Ann Rheum Dis 68(1): 130-135.

Page 99: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

99

331. Hultqvist M, Nandakumar KS, Bjorklund U, Holmdahl R. (2010) Rabeximod reduces

arthritis severity in mice by decreasing activation of inflammatory cells. Ann Rheum Dis

69(8): 1527-1532.

332. Wykes MN, Good MF. (2008) What really happens to dendritic cells during malaria?

Nat Rev Microbiol 6(11): 864-870.

333. Schwarzer E, Skorokhod OA, Barrera V, Arese P. (2008) Hemozoin and the human

monocyte--a brief review of their interactions. Parassitologia 50(1-2): 143-145.

334. van Helden SF, Krooshoop DJ, Broers KC, Raymakers RA, Figdor CG, et al. (2006) A

critical role for prostaglandin E2 in podosome dissolution and induction of high-speed

migration during dendritic cell maturation. J Immunol 177(3): 1567-1574.

335. Mukherjee P, Chauhan VS. (2008) Plasmodium falciparum-free merozoites and infected

RBCs distinctly affect soluble CD40 ligand-mediated maturation of immature monocyte-

derived dendritic cells. J Leukoc Biol 84(1): 244-254.

336. Sanchez-Sanchez N, Riol-Blanco L, Rodriguez-Fernandez JL. (2006) The multiple

personalities of the chemokine receptor CCR7 in dendritic cells. J Immunol 176(9):

5153-5159.

337. Deshpande P, Shastry P. (2004) Modulation of cytokine profiles by malaria pigment--

hemozoin: Role of IL-10 in suppression of proliferative responses of mitogen stimulated

human PBMC. Cytokine 28(6): 205-213.

338. McCormack WJ, Parker AE, O'Neill LA. (2009) Toll-like receptors and NOD-like

receptors in rheumatic diseases. Arthritis Res Ther 11(5): 243.

339. Sponaas AM, Cadman ET, Voisine C, Harrison V, Boonstra A, et al. (2006) Malaria

infection changes the ability of splenic dendritic cell populations to stimulate antigen-

specific T cells. J Exp Med 203(6): 1427-1433.

340. Millington OR, Gibson VB, Rush CM, Zinselmeyer BH, Phillips RS, et al. (2007)

Malaria impairs T cell clustering and immune priming despite normal signal 1 from

dendritic cells. PLoS Pathog 3(10): 1380-1387.

341. Modiano D, Petrarca V, Sirima BS, Bosman A, Nebie I, et al. (1995) Plasmodium

falciparum malaria in sympatric ethnic groups of burkina faso, west africa. Parassitologia

37(2-3): 255-259.

342. Bolad A, Farouk SE, Israelsson E, Dolo A, Doumbo OK, et al. (2005) Distinct

interethnic differences in immunoglobulin G class/subclass and immunoglobulin M

antibody responses to malaria antigens but not in immunoglobulin G responses to

nonmalarial antigens in sympatric tribes living in west africa. Scand J Immunol 61(4):

380-386.

343. Israelsson E, Vafa M, Maiga B, Lysen A, Iriemenam NC, et al. (2008) Differences in

fcgamma receptor IIa genotypes and IgG subclass pattern of anti-malarial antibodies

between sympatric ethnic groups in mali. Malar J 7: 175.

Page 100: Characterization of antigen-presenting cell function in ...su.diva-portal.org/smash/get/diva2:435144/FULLTEXT01.pdf · Doctoral thesis from the Department of Immunology, Wenner-Gren

100

344. Vafa M, Israelsson E, Maiga B, Dolo A, Doumbo OK, et al. (2009) Relationship

between immunoglobulin isotype response to plasmodium falciparum blood stage

antigens and parasitological indexes as well as splenomegaly in sympatric ethnic groups

living in mali. Acta Trop 109(1): 12-16.

345. Doxsee CL, Riter TR, Reiter MJ, Gibson SJ, Vasilakos JP, et al. (2003) The immune

response modifier and toll-like receptor 7 agonist S-27609 selectively induces IL-12 and

TNF-alpha production in CD11c+CD11b+CD8- dendritic cells. J Immunol 171(3): 1156-

1163.

346. Pulendran B, Kumar P, Cutler CW, Mohamadzadeh M, Van Dyke T, et al. (2001)

Lipopolysaccharides from distinct pathogens induce different classes of immune

responses in vivo. J Immunol 167(9): 5067-5076.

347. Joffre O, Nolte MA, Sporri R, Reis e Sousa C. (2009) Inflammatory signals in dendritic

cell activation and the induction of adaptive immunity. Immunol Rev 227(1): 234-247.