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

Document related concepts

Molecular mimicry wikipedia , lookup

Immune system wikipedia , lookup

Phagocyte wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Lymphopoiesis wikipedia , lookup

T cell wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Adaptive immune system wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Immunomics wikipedia , lookup

Innate immune system wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Transcript
Doctoral thesis from the Department of Immunology, the Wenner-Gren Institute,
Stockholm University, Stockholm, Sweden
Phenotypic and functional studies of NK cells
in neonates and during early childhood
Yvonne Sundström
Stockholm 2008
Previously published articles were reproduced with permission from the publisher.
Printed by Universitetsservice AB, Stockholm 2008
Distributor: Stockholm University Library
 Yvonne Sundström, Stockholm 2008
ISBN 978-91-7155-747-6
ii
Till mina föräldrar
iii
iv
SUMMARY
During infancy, before adaptive immunity has matured, innate immunity is thought to be relatively
more important. Human natural killer (NK) cells are innate immune cells involved in the control of
virus-infected cells and can influence adaptive immunity mainly through cytokine production. This
thesis aimed at investigating function and phenotype of NK cells in children from birth and during
early childhood and to see if these features are altered in children that develop early allergy, in
children latently infected by herpes viruses or born by preeclamptic mothers.
In newborn mice, NK-cell function increases and expression of the different NK-cell receptors
changes during the first weeks of life. In humans, similar maturation processes in early life are poorly
studied. We analyzed NK-cell phenotype and function in cord blood (CB) and in the same children at
the age of two and at five years. We demonstrate that the distribution of different NK-cell
subpopulations and expression of NK-cell receptors changed with age. The proportion of NK cells was
higher in CB compared to two years and at five years of age. The proportion of LIR-1+ NK cells was
found to increase with age, while the proportion of CD94+NKG2C- (mainly NKG2A+) NK cells and
the level of expression of NKG2D and NKp30 decreased with age. The induction of IFN-γ was
slightly lower in CB NK cells compared to later time points.
NK-cell cytokines can affect priming of naïve T helper cells and may play a role in development of
allergic reactions. Therefore we evaluated CB NK cells from children that developed early allergy.
Children that became allergic at the age of five years had a smaller proportion of CD56bright NK cells,
which is the NK-cell subpopulation that has a high cytokine producing capacity. CB NK cells from
allergic children had a tendency of an increased expression of the activating receptor NKp30 and
tended to have a decreased proportion of NK-cells expressing CD94/NKG2C compared to the children
that did not become allergic.
Herpesvirus infection associates with a reduced risk of becoming IgE-sensitized, and NK cells are
important in the control of herpes viruses. In an attempt to find a mechanism to the observed
protective effect of herpesvirus latency we studied innate immunity (NK cells and monocytes) in twoyear old children seropositive (SP) or seronegative (SN) for cytomegalovirus (CMV) and Epstein-Barr
virus (EBV). Herpesvirus seropositivity in early life was associated with reduced proportions of IFNγ+ NK cells as well as lower intracellular IFN-γ levels upon in vitro stimulation (IL-15 and
peptidoglycan). IFN-γ measured in cell culture supernatants and in plasma was lower in SP compared
to SN subjects. In addition, SP children also tended to have a decreased proportion of proinflammatory
CD14+CD16+ monocytes and lower levels of IL-6 in culture supernatants.
The pregnancy disorder preeclampsia is associated with increased proinflammatory cytokine
production in the mother and we wanted to study if that had an influence on CB NK cells. In CB, there
was a trend towards a reduced expression of NKG2D on NK cells and a decreased NK-cell function in
children born by preeclamptic mothers. We also demonstrate reduced TNF and increased IL-8 levels
in CB sera from the preeclamptic group. Further, the serum levels of IL-6 in the preeclamptic mothers
and their neonates were positively correlated, whereas no such association was found in the healthy
group.
Taken together, our results suggest that NK-cell populations are dynamic during the first years of
life and start to resemble the phenotype of adults after five years of age. Early alterations in the NKcell populations could lead to insufficient Th1 priming, with an increased risk to develop allergic
disease. Early infection by common herpes viruses can influence NK-cell function and might be one
important factor involved in early maturation processes of adaptive immunity. The altered NK-cell
function and cytokine levels, noticed in CB from pathological pregnancies, suggest that NK cells
could be influenced already in utero. These early alterations of innate immunity may affect the
development of the child’s immune system, sometimes with beneficial outcome but could in some
cases promote pathology.
v
vi
LIST OF PAPERS
This thesis is based on the following original papers, which will be referred to by their
Roman numeral:
I.
Yvonne Sundström, Caroline Nilsson, Gunnar Lilja, Klas Kärre, Marita TroyeBlomberg and Louise Berg. The expression of human NK cell receptors in early
life. Scand J Immunol. 2007 Aug; 66(2-3):335-44.
II.
Yvonne Sundström, Louise Berg, Caroline Nilsson, Gunnar Lilja, Klas Kärre and
Marita Troye-Blomberg. A decreased proportion of CD56bright human natural killer
cells in cord blood from children that develop early allergy. Manuscript.
III.
Shanie Saghafian-Hedengren*, Yvonne Sundström*, Ebba Sohlberg, Caroline
Nilsson, Marita Troye-Blomberg, Louise Berg#, and Eva Sverremark-Ekström#.
Herpesvirus seropositivity in childhood associates with decreased monocyteinduced NK-cell IFN-γ production. Under revision. J Immunol. 2008. *,#Shared
first and last authorship.
vii
TABLE OF CONTENTS
SUMMARY ....................................................................................................................................................................v
LIST OF PAPERS ......................................................................................................................................................vii
TABLE OF CONTENTS ......................................................................................................................................... viii
LIST OF ABBREVIATIONS.....................................................................................................................................xi
INTRODUCTION.......................................................................................................................................................13
THE IMMUNE SYSTEM .......................................................................................................................................13
Innate immunity...................................................................................................................................................13
Cells and functions of innate immunity........................................................................................................................... 13
Pattern recognition receptors ...................................................................................................................................... 14
Granulocytes................................................................................................................................................................ 15
Mast cells ..................................................................................................................................................................... 15
Monocytes/Macrophages ............................................................................................................................................ 16
Dendritic cells.............................................................................................................................................................. 17
NK cells ....................................................................................................................................................................... 18
Adaptive immunity...............................................................................................................................................18
Cells and functions of adaptive immunity....................................................................................................................... 18
Antigen-presenting cells (APCs) and the major histocompatibility complex (MHC)............................................ 18
T cells........................................................................................................................................................................... 20
αβ T cells ............................................................................................................................................................... 20
γδT cells.................................................................................................................................................................. 22
B cells........................................................................................................................................................................... 23
Cytokines..............................................................................................................................................................23
IFN-α/β........................................................................................................................................................................ 24
IFN-γ ............................................................................................................................................................................ 24
TNF .............................................................................................................................................................................. 24
IL-12............................................................................................................................................................................. 25
IL-2 and IL-15 ............................................................................................................................................................. 25
IL-13 and IL-4 ............................................................................................................................................................. 26
IL-10 and TGF-β ......................................................................................................................................................... 26
NK CELLS ...............................................................................................................................................................27
NK-cell definition and distribution ....................................................................................................................27
NK cell receptors.................................................................................................................................................28
The C-type lectin-like receptors....................................................................................................................................... 29
The Ig-like receptors......................................................................................................................................................... 30
The killer Ig-like receptors (KIRs)............................................................................................................................. 30
Leucocyte Ig-like receptor-1 (LIR-1) ........................................................................................................................ 31
The Natural cytotoxicity receptors (NCRs)............................................................................................................... 31
Antibody-dependent cellular cytotoxicity (ADCC) mediated by NK cells.......................................................32
Chemokines and NK-cell homing .......................................................................................................................33
NK-cell generation ..............................................................................................................................................33
Receptor expression during development ..........................................................................................................34
NK-cell education................................................................................................................................................36
NK receptors on T cells ......................................................................................................................................37
NK cells bridge to adaptive immunity................................................................................................................37
NK cells and accessory cells ..............................................................................................................................38
HERPESVIRUSES ..................................................................................................................................................39
ALLERGY................................................................................................................................................................39
The allergic reaction ...........................................................................................................................................40
Type 1 and type 2 cytokines in allergy...............................................................................................................40
The hygiene hypothesis .......................................................................................................................................41
Environmental factors in allergy development ............................................................................................................... 41
NK CELLS IN HEALTH AND DISEASE............................................................................................................42
NK cells in viral infections .................................................................................................................................42
NK cells in allergy...............................................................................................................................................43
THE IMMUNE SYSTEM OF NEONATES AND YOUNG CHILDREN .........................................................44
PREECLAMPSIA....................................................................................................................................................45
viii
AIMS OF THE STUDY .............................................................................................................................................47
SUBJECTS ...................................................................................................................................................................48
STUDY POPULATION (STUDY IV, PRELIMINARY DATA) ..........................................................................................48
MATERIAL AND METHODS.................................................................................................................................49
MATERIAL AND METHODS (STUDY IV, PRELIMINARY DATA).................................................................................49
RESULTS AND DISCUSSION ................................................................................................................................52
PAPER I ......................................................................................................................................................................52
PAPER II.....................................................................................................................................................................53
PAPER III ...................................................................................................................................................................55
STUDY IV ( PRELIMINARY DATA)..............................................................................................................................58
Results ..................................................................................................................................................................58
Discussion............................................................................................................................................................62
CONCLUDING REMARKS AND FUTURE PERSPECTIVES ........................................................................66
ACKNOWLEDGEMENTS .......................................................................................................................................68
REFERENCES ............................................................................................................................................................71
Tables
TABLE 1 ..........................................................................................................................................................................14
TABLE 2 ..........................................................................................................................................................................30
TABLE 3 ..........................................................................................................................................................................32
TABLE 4 ..........................................................................................................................................................................48
Figures
FIGURE 1.........................................................................................................................................................................36
FIGURE 2.........................................................................................................................................................................59
FIGURE 3.........................................................................................................................................................................60
FIGURE 4.........................................................................................................................................................................61
FIGURE 5.........................................................................................................................................................................62
ix
x
LIST OF ABBREVIATIONS
ADCC
AEDS
APC
BCR
BSA
CB
CBA
CBMC
CD
CLMF
CLP
CMP
CMV
Con A
CTL
DC
EBV
ELISA
FcγR
FLK2
Foxp3
GM-CSF
HIV
HLA
HPC
ICAM
iDC
IFN
Ig
IL
ILT
ITAM
ITIM
KIR
Antibody-dependent cellular
cytotoxicity
Atopic eczema/dermatitis
syndrome
Antigen-presenting cell
B-cell receptor
Bovine-serum albumin
Cord blood
Cytometric Bead Array
Cord blood mononuclear cell
Cluster of differentiation
Cytotoxic-lymphocyte
maturation factor
Common lymphoid
progenitor
Common myeloid progenitor
Cytomegalovirus
Concanavalin A
Cytotoxic T-lymphocyte
Dendritic cell
Epstein-Barr virus
Enzyme-linked
immunosorbent assay
Fragment crystallizable
gamma receptor
Fetal liver kinase 2 ligand
Forkhead box P3
Granulocyte-macrophage
colony-stimulating factor
Human immunodeficiency
virus
Human leukocyte antigen
Haematopoietic precursor
cell
Intercellular adhesion
molecule 1
Immature denritic cell
Interferon
Immunoglobulin
Interleukin
Ig-like transcript
Immunoreceptor tyrosinebased activation motif
Immunoreceptor tyrosinebased inhibition motif
Killer immunoglobulin-
LFA-1
LIR
LPS
M1
M2
mDC
MHC
MIC
NCR
NFκB
NK
NKG2D
NKP
NKT
NOD
PAMP
PBMC
pDC
PGN
PHA
PMA
PML
PRR
SCF
SCT
SLE
SPT
TCR
TGF
Th
TLR
TNF
Treg
ULBP
uNK
like receptor
Lymphocyte functionassociated antigen 1
Ig-like receptor
Lipopolysaccharide
Classically activated
macrophage
Alternatively activated
macrophage
Myeloid dendritic cell
Major-histocompatibility
complex
MHC class I C chain-related
protein
Natural-cytotoxicity receptor
Nuclear factor-kappa B
Natural killer
NK group 2, member D
NK-cell precursor
Natural-killer T cell
Nucleotide-binding
oligomerization domain
Pathogen-associated
molecular pattern
Peripheral blood
mononuclear cell
Plasmacytoid dendritic cell
Peptidoglycan
Phytohaemagglutinin
Phorbol 12-myristate 12acetate
Polymorphonuclear
leukocyte
Pattern recognition receptor
Stem cell factor
Syncytiotrophoblast
Systemic lupus erythematosus
Skin prick test
T-cell receptor
Transforming growth factor
T helper
Toll-like receptor
Tumor necrosis factor
Regulatory T cell
UL-16 binding protein
Uterine natural killer cell
xi
xii
INTRODUCTION
THE IMMUNE SYSTEM
The immune system is able to detect the presence of harmful agents in the host, and the
major goal is elimination of the threat. This task is challenging considering the enormous
molecular diversity of pathogens and their high replication and mutation rates. The immune
system has co-evolved with this complexity of pathogens, generating efficient strategies to
fight off invaders. It is composed of several different cell types and soluble factors, which
exert different protective functions against harmful invaders.
The immune system is commonly divided into two parts, the innate and the adaptive.
Although described as separate compartments the innate and adaptive immune systems are
tightly interrelated and the combined responses most often give a highly effective protection
against pathogens/infection.
Innate immunity
The innate or non-specific immune system is the first line of defense against potentially
harmful agents and microorganisms. Preventing entry of pathogens will hold off infections
and the first obstruction for pathogens consists of anatomic barriers such as skin and mucous
membranes. Physiological conditions, such as temperature, pH and different soluble and cellassociated molecules add up the impediment. In addition, the presence of cells specialized in
phagocytosis, cytotoxicity and production of microbicidal agents makes microbial survival
more difficult in the host.
Cells and functions of innate immunity
Cells of the innate immune system recognize and respond to pathogens by non-specific
mechanisms that prevent infection by microorganisms in the host. The mechanisms include
recognition of conserved molecular motifs of microbial origin that are distinguishable from
molecules of the host. These motifs are collectively called pathogen-associated molecular
patterns (PAMPs) and are recognized by pattern recognition receptors (PRRs). By nonspecific we mean that these receptors are conserved between individuals of the same species
and within the individual during ontogeny. However, the interaction between PAMPs and
13
PRRs may be referred to as specific, as not all PAMPs are recognized by all PRRs. The cells
of innate immunity include granulocytes, mast cells, monocytes, macrophages, dendritic cells
(DCs) and natural killer (NK) cells.
Pattern recognition receptors
Among the most evaluated PRRs are the toll-like receptors (TLRs). There are 10 known
functional TLRs in humans [1] (Table 1). TLR2 and TLR4 are expressed on the cell surface
of mainly monocytes/macrophages, DCs and mast cells. TLR2 recognizes peptidoglycan
(PGN), a type of PAMP present in the cell wall of almost all bacteria, and receptor-ligand
interaction induces cellular activation and production of proinflammatory cytokines by the
cell [2].
Ligands for Toll-Like Receptors- What they see on pathogens
TLR
Natural Ligand
TLR1 (partnered with TLR2)
TLR2 (partnered with TLF6)
Bacterial triacyl lipopeptides and certain proteins in parasites
Bacterial diacyl lipopeptides, lipoteichoic acid from Gram-positive
bacteria, and zymosan from the cell wall of yeast, peptidoglycan
Double-stranded RNA from viruses (i.e., West Nile virus)
Endotoxin (lipopolysaccaride) from Gram-negative bacteria
Flagellin from mobile bacteria
Single-stranded RNA from viruses (i.e., HIV)
Single-stranded RNA from viruses (i.e., HIV)
CpG DNA from bacteria or viruses
Unknown
by TLRs. The ligands for each functional human TLR. Adapted from
TLR3
TLR4
TLR5
TLR7
TLR8
TLR9
TLR10
Table 1. Immune recognition
Science 2006; 312 (5771):184–187 [1]. Reprinted with permission from AAAS.
Another type of PRRs are the nucleotide-binding oligomerization domains (NODs) that
mainly are located in the cytosol of monocytes, DCs and epithelial cells [3]. NOD1 and
NOD2 are members of a protein family that are involved in regulation of apoptosis as well as
for innate recognition of specific bacterial components [3,4]. NOD2 recognizes a peptide
derived from PGN that is present in almost all bacteria, while NOD1 mainly recognizes PGNcomponents derived from Gram-negative bacteria and ligand recognition leads to induction of
inflammatory responses through nuclear factor-kappaB (NFκB) signaling pathways [5,6].
14
Granulocytes
Polymorphonuclear leukocytes (PML), or more commonly called, granulocytes are a group
of leukocytes defined by the presence of granules in their cytoplasm and a nucleus that is
often lobed in segments within the cell. There are three different types of granulocytes,
including neutrophils, basophils and eosinophils.
Of the circulating leukocytes in humans, neutrophils are the most numerous (50-70%).
These cells circulate in the blood, but upon infection they are rapidly recruited to the site of
infection and are important in the subsequent progression of immune responses against the
microbe. Neutrophils are very motile phagocytes, but are also involved in the inflammatory
response that is induced through the release of antimicrobial molecules, stored in their
granules, and also through secretion of chemokines that recruit other immune cells [7,8].
Less than 1% of the circulating leukocytes are basophils. Basophils are not phagocytic but
they are suggested to be important in the early regulation of immune responses against
parasitic organisms through the release of IL-4 and IL-13 [9,10]. Basophils have also been
found to play a role in allergic reactions through their release of e.g. histamine [11] and
cytokines [12] that contribute to allergic inflammation.
Eosinophils constitute about 1-6% of all leukocytes. They are found in blood and organs
like thymus, spleen and lymph nodes under normal conditions. Like those of basophils, the
functions of eosinophils include combating parasitic infections [13] and eosinophils are also
associated with the pathogenesis of allergic disease through the release of their granule
proteins [11]. Eosinophils have also been shown to function as professional antigen
presenting cells (APCs) [14].
Mast cells
Like granulocytes, mast cells also have large granules in their cytoplasm and due to their
similarity with basophils it has been suggested that mast cells are the tissue resident form of
basophils. However, the developmental processes, function and morphology differ indicating
that they are different cell types [15].
Mast cells are often mentioned in relation to pathology of allergic disease and therefore
associated with harmful reactions. In a sensitized person allergen-specific immunoglobulin
(Ig) E antibodies bind to the high affinity Fcε receptor I (FcεRI) on tissue-resident mast cells
[16]. The subsequent exposure to the allergen that is recognized by the FcεRI-bound IgE
15
antibodies on the mast cell results in crosslinking of the antibodies, leading to degranulation
of inflammatory mediators such as histamine, heparin and proteases that act on surrounding
tissues. This results in an immediate phase reaction that, within minutes, leads to increased
vascular permeability, mucous secretion and stimulation of sensory nerves that generates
symptoms like nasal congestion, itching and sneezing. Mast cells can also be an important
factor in reactions that are followed, hours later, by recruitment of cells like eosinophils, T
cells and macrophages, to the area through the induction of chemokines and adhesionmolecule expression on endothelial cells. At the site, these cells become activated and the
effector functions of the different cells may result in a prolonged inflammation [17,18].
However, mast cells are of great significance in other immune responses, being one key
effector-cell type that protects against morbidity and pathology of infection. Mast cells are
found in tissues in close proximity to blood vessels and during early responses they are
involved in the induction of inflammation. For instance, upon bacterial infection mast cells
are induced to release tumor necrosis factor (TNF) that is important for the recruitment of
other immune cells, such as neutrophils [19]. Additionally, mast cells exhibit effector
functions such as secretion of antimicrobial peptides that can kill invading bacteria [20]. By
releasing proteases mast cells can disarm endogenous toxins produced to battle bacteria e.g.
endothelin-1 (ET-1), [21]. Mast cells have also been shown to release effector molecules that
degrade other toxins and venoms that are similar to ET-1 [22]. Another interesting feature of
mast cells is the involvement in regulation of immune responses. Mast cell derived IL-10 can
have immunosuppressive functions important in limiting the duration and magnitude of
adaptive immune responses [23]. Taken together, these features of mast cells indicate their
importance in damage control and in regulation of immune responses.
Monocytes/Macrophages
Monocytes originate in the bone marrow and are released into the peripheral circulation. In
human peripheral blood, 5-10% of the leukocytes are monocytes and they are
morphologically and functionally heterogeneous. The initial marker for identification of
monocytes was the expression of CD14. The combination of CD14 and CD16 allows the
identification of two subsets of monocytes: CD14++CD16- cells and CD14+CD16+ cells. In the
blood of healthy individuals the distribution of these populations is about 90% and 10%,
respectively [24]. Functionally, the CD14+CD16+ cells appear to produce more
proinflammatory cytokines and have higher APC activity than the CD14++CD16- monocytes
16
[25]. Monocytes are important effector cells in innate immunity and recognize microbial
components through their expression of PRRs [26]. The CD14+CD16+ subset has been
reported to have higher levels of TLR2 [27] and TLR4 [28]. This indicates an important role
of this subset in microbial responses.
After release from the bone marrow, monocytes circulate in the blood before they enter
tissues were they differentiate into macrophages. Macrophages are phagocytic cells that clear
the body of debris and they are potent APCs. Macrophages can be divided in subpopulations
that reveal different functions depending on the mode of activation. The so-called, classically
activated macrophages (M1) develop in response to IFN-γ and microbial products and are
distinguished through their increased endocytic functions, ability to present antigens and
ability to produce pro-inflammatory cytokines [29]. They also secrete chemokines that induce
recruitment of NK cells and T cells [30]. The alternatively activated macrophages (M2) are
further subdivided into three groups of cells that are activated differently and have separate
functions. M2a macrophages are activated by anti-inflammatory cytokines, like IL-4 and IL13 and the function of M2a cells include secretion of chemokines that recruits eosinophils,
basophils and T cells that promotes anti-inflammatory immune responses [30]. M2b cells are
induced upon stimulation with lipopolysaccharide (LPS) or IL-1β and these cells are
characterized by low IL-12 and high IL-10 production, which promote the subsequent
induction of type 2 adaptive immune responses [30]. Finally, the M2c macrophages are
activated by IL-10, transforming growth factor (TGF)-β or glucocorticoids (a class of steroid
hormones) and their functions include down regulation of pro-inflammatory cytokines and
increased scavenging activity for debris [31].
Dendritic cells
DCs form another heterogeneous family of haematopoietic cells. Two main developmental
pathways from hematopoeitic-progenitor cells in humans generate either myeloid DCs
(mDCs) or plasmacytoid DCs (pDCs). Depending on their location, they display different
morphology and function [32]. Immature DCs have high endocytic activity. Maturation of
DCs is induced by sensing microbial products like PAMPs, tissue damage [33], interaction of
Fc receptors or ligation to CD40L on T cells [34]. Dendritic cells process endocytosed
proteins or cytosolic proteins into proteolytic peptides and load these on to major
histocompatibility complex (MHC) class I and class II molecules. This process induces
17
maturation of DCs and migration to lymph nodes. In the lymph nodes, DCs present antigens
to T cells and thereby initiate antigen-specific adaptive immune responses.
The mDCs are distributed in peripheral tissue, secondary lymphoid organs and in the
blood. Tissue resident DCs called Langerhans cells, are located in the epidermis of the skin
and interstitial DCs are found in interstitial connective (non-lymphoid) tissues. The pDCs are
found in peripheral blood and lymphoid tissues and respond to viruses by producing vast
quantities of type 1 IFNs (IFN-α, IFN-β) [35].
It is generally considered that monocytes are precursors of DCs and human monocytes are
indeed capable of differentiating into some of the DC linages in vitro [36]. It is unclear
whether this occurs under physiological conditions in humans, although experiments using
murine models have reported indications that sustain this hypothesis in vivo [37].
NK cells
NK cells are important cells of innate immunity and are described below (starting page 27).
Adaptive immunity
The adaptive or specific immune system is composed of T- and B-cells that generate
specific responses to antigens. One distinguishing feature of adaptive immunity is the
rearrangement of T-cell receptor (TCR) or B-cell receptor (BCR) genes that results in a vast
diversity in their respective antigen-specific receptors. This results in an enormous variability
of receptors and soluble antibodies that increases the probability for recognition of specific
antigens.
Cells and functions of adaptive immunity
Antigen-presenting cells (APCs) and the major histocompatibility complex (MHC)
Recognition of antigens by T cells requires help/presentation from other cells. These cells
are termed APCs and can be divided into two groups: the professional and the nonprofessional APCs. DCs, monocytes/macrophages and B cells fall into the category of
professional APCs while all nucleated cells are non-professional APCs.
18
Presentation of antigen by cells is achieved through proteins encoded within the major
histocompatibility complex (MHC), called MHC class I and MHC class II molecules. MHC
class I can be further subdivided into MHC class Ia (or classical MHC class I) and MHC class
Ib (or non-classical MHC class I). In humans the MHC complex is termed human leukocyte
antigens (HLA). MHC class Ia includes HLA-A, -B and -C molecules and the MHC class Ib
molecules are named HLA-E, -F and -G. MHC class II molecules include HLA-DP, HLA-DQ
and HLA-DR.
Nearly all nucleated cells (including professional and non-professional APCs) express
MHC class I that presents endogenous antigens. After degradation of cytosolic proteins, e.g.
proteins derived from the cell itself and from viruses or intracellular bacteria, into small
peptides, these are transported to the cell surface as a complex with MHC class I molecules.
The complex is presented to naïve CD3+CD8+ T cells in secondary lymphoid tissues, but for
subsequent generation of CD3+CD8+ effector T cells, sequential signals are required. Ligation
of costimulatory molecules like CD80 and CD86 on the professional APC, engaging CD28 on
the T cells provides one signal that is needed for the differentiation into effector T cells. The
T cell also needs signals derived from cytokines like IL-2. The activated T cells mature and
migrate to sites of inflammation where they find the same antigen-MHC I complex and exert
their effector functions. For CD3+CD8+ T cells these functions include killing of the target
cell and/or production of cytokines that in different ways can enhance the response towards
the pathogen.
Professional APCs endocytose exogenous particles and proteins and process these into
peptides, which are presented by MHC class II on the cell surface. Peptides in association
with MHC class II are recognized by naïve CD3+CD4+ T cells and after the simultaneous
interaction by costimulatory molecules the T cells become activated and are induced to
mature. The functional responses by activated CD3+CD4+ T cells include production of
cytokines, which regulate other parts of the immune system.
The functions of MHC class Ib molecules are in large unknown and their limited
polymorphism restricts the variability of peptides that can be presented. HLA-E bind
sequences derived from MHC class Ia molecules and cell surface expression of HLA-E is
dependent on binding of peptides provided by HLA-A, -B and –C molecules [38,39]. Due to
this dependency, HLA-E is expressed by all cells expressing class Ia molecules, rendering a
broad tissue distribution. Both HLA-E- and HLA-G-peptide complexes, when recognized by
NK cells, either inhibit or trigger NK-cell activity [40,41]. Except for small amounts of HLAG in some human fetal and adult tissue, HLA-G is upregulated on fetal cells (trophoblast
19
cells) in contact with maternal tissue [42] and are suggested to play a role in fetal tolerance by
prevention of NK-cell mediated lysis [40,41]. Four membrane-bound forms (HLA-G1-4) and
three soluble forms (HLA-G5-7) of HLA-G exist [43]. HLA-F transcripts have been detected
in fetal liver, adult skin, placenta, B cells and T cells. The function of HLA-F is still elusive.
The MHC class I chain related (MIC) molecules, MICA and MICB, show homology with
HLA molecules but do not bind peptides needed for antigen presentation. MIC-proteins are
induced on the surface of stressed cells and are recognized by cells expressing the NKG2D
receptor. Some cells, like tumor cells and syncytiotrophoblast cells in the placenta, can shed
soluble MIC molecules. The binding of these soluble molecules to the NKG2D receptor
prevents further NKG2D dependent activation of the T- or NK-cell [44]. CD1 molecules are
also related to the class I MHC molecules and present lipid antigens to T cells and NKT cells.
T cells
Progenitor-T cells emanate from the bone marrow and migrate to the thymus for
maturation. In the thymus, selection of developing T cells (named thymocytes) with
functional TCRs takes place. The selection process authorizes the survival of T cells that
recognize self-MHC molecules (positive selection), but only if the binding to MHC molecules
presenting self-peptides is not too strong (negative selection). Along this maturation/selection
pathway the TCR undergoes genetic rearrangement. In this process, there is recombination of
multiple V, D and J genes that are randomly selected and assembled into two TCR chains,
rendering a TCR with a distinctive specificity for a MHC-peptide complex. The final result of
this process is a T cell with a functional TCR complex including the signaling chains of the
CD3 complex. The TCR is expressed as a heterodimer, made up of a α and a β chain (αβ
TCR), or a γ and a δ chain (γδ TCR).
αβ T cells
T cells with αβ TCRs (αβT cells) can be sub-divided into CD3+CD4+ T cells and
CD3+CD8+ T cells and are generally termed as T helper (Th) cells and cytotoxic Tlymphocytes (CTL), respectively.
Naïve CD3+CD4+ T cells can differentiate into a variety of T-cell subsets depending on the
efficiency of the TCR stimulation, the magnitude of costimulation by APCs and the
20
surrounding cytokine milieu. The involvement of IL-12 induces differentiation of Th1 cells,
IL-4 promotes differentiation of Th2 cells and IL-23 and IL-1β renders Th17 cells [45,46].
The Th1 response, effective against intracellular pathogens, leads to the production of
cytokines like IFN-γ, IL-12 and IL-18 that mainly promotes cellular immunity and gives rise
to inflammation with e.g. activation of macrophages and induction of antigen-presenting
activity [47].
The Th2 response is effective against extracellular pathogens. Th2 cells produce cytokines
like IL-4 that promote humoral immune responses including Ig-class switching to e.g. IgE and
IgG4 by human B cells [48,49].
The characteristic of Th17 cells is the production of IL-17 [50]. All together six different
homodimers exist that form the IL-17 family (IL-17A to IL-17F) of cytokines [51]. Human
blood monocytes cultured in vitro and stimulated with rIL-17A, release TNF and IL-1β
suggesting an important role for IL-17 in inducing and/or maintaining an inflammatory
response [52]. Although the function of this Th subset is not fully clear, present data indicate
a role in the immune responses against extracellular pathogens where Th1 and Th2 type of
immunity are insufficient. This subset of T cells also seems to be involved in driving
autoimmune diseases [53].
Another subset of T cells, called regulatory T cells, (Tregs) is important in controlling the
magnitude and/or duration of immune responses and also to sustain immunological
unresponsiveness to self-antigens, functions that are crucial to prevent detrimental effects to
the host. The main hallmark of Tregs is expression of the transcription factor forkhead box P3
(Foxp3) and CD25, although exceptions exist (see below). Two central Foxp3 expressing
populations described in the literature are referred to as natural CD4+CD25+highFoxp3+ T reg
cells and peripherally induced CD4+CD25+highFoxp3+ T reg cells. The former Treg type
develops from CD4+ T cells within the thymus while the latter is induced by cytokine
stimulation in vitro and is thought to be induced from CD4+CD25- T cells in the periphery in
vivo [54,55]. The immunosuppression mediated by these cells has been suggested to require
cell-cell contact that involves cell-surface bound TGF-β1 [56]. Another type of peripherally
induced Treg cells is termed Tr1 and they are induced from naïve T cells upon IL-10
stimulation. These cells do not express CD25 or Foxp3, but are defined by their production of
IL-10 and TGF-β and are distinguished from Th cells by their minute production of IFN-γ and
no IL-2 or IL-4 [57]. Th3 cells are antigen specific and produce TGF-β upon activation and
some, but not all, appear to express Foxp3 [58].
21
Natural killer T (NKT) cells are a small heterogeneous population of CD4+, CD8+ or CD4CD8- αβT cells that also express NK-cell receptors. Most of the αβ TCRs of human NKT
cells consist of Vα24-Jα18/Vβ11 chains that recognise the MHC-like molecule CD1d. APCs
constitutively express CD1d that binds lipids and glycolipids for presentation to NKT cells.
The activation of NKT cells induces up-regulation of costimulatory molecules on the cell
surface, which augments the antigen presenting activities by DCs and cytokine production of
both Th1 and Th2 types [59].
The CD3+CD8+ T-cell effector function mainly involves induction of apoptosis of target
cells but they are also capable of producing cytokines. Apoptosis can be induced through
release of cytotoxic proteins or through interaction with the membrane-bound Fas ligand
(FasL) on the T cells with Fas on the target cell.
γδ T cells
The majority of mature T cells in peripheral blood are αβT cells, while a minority (1-10%)
expresses an alternative γδ TCR heterodimer in association to CD3 (γδT cells). There are only
a small number of available variable (V) γ and δ genes but the diversity of the γδ TCR
repertoire is still extensive, due to e.g. N nucleotide insertions during gene rearrangement
[60]. In healthy adults, the most prominent type of γδT cells in peripheral blood express the
Vγ9 chain together with Vδ2, while the second most prominent type express various Vγ
chains in association with Vδ1 [61]. However, the lymphocyte distribution in the intestine is
different, where the majority of γδT cells are Vδ1+. The ligands for these subpopulations
differ and the Vγ9/Vδ2 T cells mainly recognize phosphorylated non-peptidic molecules [62],
while e.g. the intestinal Vδ1+ γδT cells recognize stress-induced MHC class I-related MICA
and MICB molecules [63,64].
The recognition of antigens by γδ (Vγ9/Vδ2) T cells occurs independently of class I and
class II MHC molecules or the MHC-related CD1 molecules [62]. Rather, it was recently
suggested that activated γδT cells themselves can have MHC II related antigen presenting
features [65]. The effector functions of γδT cells include killing of target cells through release
of perforin and some γδT cells also express FasL which upon interaction with its receptor
induces apoptosis of the target cell [66]. γδT cells also have the capacity to produce cytokines
and are in particular potent producers of the proinflammatory cytokines IFN-γ and TNF,
indicating an important role in early responses to infections [62].
22
The complexity of γδT-cell biology makes it difficult to designate them to either the innate
or the adaptive part of the immune system. Since γδT cells rearrange TCR genes to produce
receptor diversity and also form memory cells they could be considered as part of the adaptive
immunity. Nevertheless, some subsets respond rapidly to common microbial molecules,
which indicates that specific γδ TCRs may function similarly to PRRs, which are attributes of
innate immunity.
B cells
B cells are essential in the generation of humoral immune responses through the
production of soluble antibodies. The generation of mature immunocompetent B cells takes
place within the bone marrow. During several developmental stages, a random rearrangement
of immunoglobulin genes generates B cells expressing IgM or IgD on the cell surface, as part
of the BCR. Naïve B cells that are not self-reactive exit the bone marrow and migrate to
lymphoid organs. The recognition of an antigen by the BCR on B cells with a simultaneous
interaction with Th cells promotes the development germinal centers in lymph nodes. In the
germinal centers the antigen-specific B cells are expanded, diversified and selected for highaffinity variants of BCRs. These form the compartment of antibody producing plasma cells
and long-lived memory B cells. The quantity and the array of antibody isotypes produced by
the plasma cells is in large determined by the help of cognate T cells through cell-associated
signals obtained from costimulatory molecule interactions, but also through Th cell-derived
cytokines. In human the Ig subclasses include: IgM, IgD, IgG1, IgG2, IgG3, IgG4, IgA1, IgA2
and IgE [67]. In an individual cell the class switch renders immunoglobulins with the same
antigen specificity but the isotype determines the biological function.
Cytokines
Cytokines are proteins involved in cellular communication and are secreted by a variety of
cells, including white blood cells, in response to different stimuli. Chemokines are a group of
cytokines important for the migration and localization of cells to specific sites such as
inflamed tissue. These molecules are very important in modulating both innate and adaptive
immune responses.
Numerous cytokines for cellular communication exist and only a few, of relevance for this
thesis, will be further described.
23
IFN-α/β
IFN-α and β are included in a group of cytokines, type I interferons, which share a
common receptor. An important part of host defense against viral infections is an early
production of type I interferons by the infected cells [68]. The receptor for these cytokines is
expressed on most cells and receptor binding results in transcription of genes encoding other
cytokine and cytokine receptors [69], redistribution of immune cells [70-72], proliferation of
memory CD8 T cells [73] and also help in regulating NK-cell homeostasis, activation and
function [74,75]. The effect of type I interferons on NK cells include intensified NK-cell
mediated cytotoxicity and indirect enhancement of proliferation through induction of IL-15
[76,77]. Type I interferons are also important regulators of NK-cell activity against tumors
[75].
IFN-γ
The type II interferon, IFN-γ, is mainly produced by activated Th1 cells [78], activated
type 1 CD8+ T cells [79] and NK cells after stimulation by cytokines like IL-12 and IL18
[80]. The heterodimeric IFN-γ receptor [81] is expressed on most epithelial cells,
macrophages, B cells and activated T cells [82]. IFN-γ interaction with its receptor induces
transcription of genes, promoting different physiological and cellular responses. Among these
responses are induction of antiviral and anti-tumor effects by e.g. increased NK-cell
cytotoxicity [83], activation of APCs [84,85] and promotion of Th1 differentiation [86]. NK
cells are regulated in an autocrine manner by IFN-γ, resulting in enhanced secretion of the
cytokine [87]. Further, IFN-γ also regulates its own receptor by down-regulating the
expression on the cell surface [85] and in Th2 cells this results in cellular desensitization to
IFN-γ [88]. The effect of IFN-γ on isotype regulation in B cells is inhibition of class switching
to IgE [89,90].
TNF
Macrophages and monocytes are major producers of the pro-inflammatory cytokine TNF,
but many other cell types including NK cells, mast cells, endothelial cells, adipose tissue and
fibroblasts contribute to the production. During early immune reactions against e.g. microbial
24
stimuli large amounts of TNF are released, which induce the production of other proinflammatory mediators that can strengthen the resistance against infection [91]. One
important function of TNF is the induction of apoptotic cell death [92]. The inflammatory
response mediated by TNF can also cause pathological complication associated with
autoimmune disorders including rheumatoid arthritis, Crohn's disease and asthma [91].
IL-12
The pro-inflammatory cytokine IL-12 is a heterodimeric molecule composed of the two
subunits p35 and p40, which together forms the biologically active IL-12p70 molecule.
Macrophages, DCs, monocytes and neutrophils are major sources of IL-12 [93]. IL-12production is induced in these cells upon microbial stimulation or after interaction of CD40
with CD40L on activated T cells in combination with IFN-γ [94]. IL-12 further induces IFN-γ
production in memory Th1 cells that results in a positive feedback loop, with stabilizing
effects on the Th1 polarization.
IL-12 also has a self-regulatory function. In memory Th1 cells IL-12 induces the
expression of IL-10 [95], which inhibits further production of IL-12 [96].
The IL-12 receptor is constitutively expressed, at low levels, on NK cells, which gives
them the ability to respond rapidly to IL-12. IL-12 induces NK cells to produce IFN-γ [97],
increases their cytotoxic activity and have a proliferative effect on pre-activated NK cells (and
T cells) [93].
IL-2 and IL-15
The receptor for IL-2 and IL-15 consists of two shared subunits (β and γ) [98] and a
cytokine-specific α-subunit [99]. Different assembly of the subunits renders different binding
affinity for its ligand. For both IL-2 and IL-15 the β and γ heterodimer has intermediate
affinity, while the high affinity receptor requires assembly of all three subunits (αβγ).
All NK cells express the intermediate affinity IL-2 receptor, while CD56bright NK cells also
constitutively express the high affinity receptor. Since the CD56bright NK cells express the IL2Rαβγ they respond to low doses of IL-2 in vitro by proliferation [100,101]. IL-2 is also part
of induction of cytotoxicity by upregulating the perforin expression in NK cells [102].
IL-15 has been shown to be crucial for NK-cell development and is also important for NKcell proliferation and survival [97,103-105]. NK cell-mediated cytotoxicity requires a
25
conjugate formation where cellular adhesion molecules establish binding between NK cells
and target cells. One such molecule is LFA-1 on NK cells that engage ICAM-1 on target cells.
IL-15 and IL-2 are important in the initiation of NK-cell mediated cytotoxicity by inducing
surface expression of LFA-1 [106].
CD4+ T cells are the major source of IL-2, but also CD8+ T cells and activated DCs can
produce IL-2 [107,108]. IL-15 protein is produced by many different cell types, but primarily
by monocytes and DCs [109,110].
IL-13 and IL-4
Both IL-4 and IL-13 have been implicated in allergic disease. They are each capable of
inducing isotype switching to IgG4 and IgE in human B cells [49,111,112] and can even have
a synergistic effect on IgE synthesis [112].
Both cytokines induce proliferation of activated B cells [48,113] but have different effects
on T cells. IL-4 is produced by Th2 cells and is important for further development and
differentiation of Th2 cells [114] while IL-13 is not capable of inducing Th2 differentiation,
since T cells do not express the IL-13 receptor [48].
IL-10 and TGF-β
The biological effect of IL-10 is anti-inflammatory and immune suppressive. Several
subsets of T cells, monocytes, DCs and B cells produce IL-10. Class switching in B cells can
be induced by IL-10 leading to high amounts of IgG1, IgG3 and IgA [67]. IL-10 indirectly
suppresses activation of NK cells through the inhibition of IL-12-production [96].
Like IL-10, TGF-β is also a cytokine with immune suppressive effects. TGF-β mediates
inhibition of surface expression of NKp30 and NKG2D on the cell surface of NK cells and
also reduces NK-cell cytotoxicity against immature DCs in vitro [115] NK-cell production of
IFN-γ is suppressed by TGF-β [116].
26
NK CELLS
NK-cell definition and distribution
NK cells are described as large granular lymphocytes and in humans they represent
approximately 15 % of circulating lymphocytes in blood [117]. Human NK cells are broadly
distributed and, except from blood, are also found in small numbers in lymphoid tissues like
lymph nodes, spleen and tonsils [118]. In the liver, NK cells make up a large proportion of
lymphocytes [119] and during pregnancy, a predominant proportion of infiltrating leucocytes
in the decidua are NK cells [120]. NK cells belong to innate immunity, as they do not need
prior immunization in order to exert their activity. These cells have even been found to
express mRNA for some TLRs, with relatively high expression of TLR-2, -3. -5 and -6 but
very limited levels of TLR-4, -7, -8 and -9 in healthy adult blood donors [121]. Although NK
cells do not appear to express TLR-7 or -8, the stimulation with related agonists results in
indirect activation of NK cells through cytokines like IL-12 and IL-18 produced by i.e.
myeloid cells [121]. In a recent study, surface expression of TLR-2, -3 and also TLR-4 on NK
cells from cord blood (CB) was detected [122]. Even if these differences in TLR-4 expression
could be due to differences in NK-cell features in CB and adult blood cells, caution should be
taken in the interpretation of mRNA expression as an indication of expression of functional
proteins on the cell surface. In a monocyte study, where TLR4 was evaluated, no consistency
was found between mRNA levels and surface expression of TLR4 [123].
Although NK cells are generally not considered to have memory functions they have been
described to respond better to haptens upon restimulation, in mice devoid of T cells and B
cells. The authors suggested that this might indicate a function where NK cells can mediate
long-lived, antigen-specific adaptive recall responses independent of B cells and T cells [124],
but the concept is controversial and further studies are needed.
Human NK cells are identified as lymphocytes that express CD56 but lack CD3 and can be
further subdivided into two subsets based on the cell surface expression of CD16 and CD56.
The majority of NK cells in peripheral blood expresses low levels of CD56 and high levels of
CD16 and is commonly called CD56dim NK cells (CD56dimCD16+CD3- lymphocytes). The
minority of NK cells in peripheral blood is the CD56bright NK-cell subpopulation, which
expresses high levels of CD56 and lack CD16 (CD56brightCD16-CD3- lymphocytes) [125].
These subpopulations of NK cells are generally considered to differ in their effector functions.
27
The main effector function of the CD56dim subpopulation is cellular cytotoxicity, but they can
also produce low levels of cytokines. The CD56bright NK cells are generally considered as
more potent producers of cytokines and in particular IFN-γ. However, this functional
dichotomy between the two main NK-cell subsets (CD56bright and CD56dim) does not always
comply. The above stated differences in function between the subtypes of human NK cells
appear to be valid upon stimulation with e.g. IL-12, IL-15 and IL-18 [80,126]. Following in
vitro stimulation with malaria (Plasmodium falciparum) infected erythrocytes, CD56dim NK
cells have a high capacity to produce IFN-γ and CD56bright NK cells show increased signs of
cytotoxicity [127]. In another study, where freshly isolated peripheral blood NK cells were
stimulated with MHC class I-deficient tumor cells (K562), a larger fraction of CD56dim NK
cells produced IFN-γ [126]. Anfossi et al suggest a functional definition based on the outcome
of the responses after exposure to target cells or cytokines where the CD56bright NK-cell
subset would be referred to as “cytokine responsive” and the CD56dim NK cells as “target-cell
responsive” [126].
In HIV positive individuals an expanded population of CD56-CD16+ NK cells has been
described and these cells are hyporesponsive [128,129].
NK cell receptors
Through the divergent and unique repertoire of surface receptors, NK cells can recognize
alterations in the expression of major histocompatibility complex (MHC) class I or class I-like
molecules on target cells, respond to stress signals, molecules induced by pathogen
transformation of cells and to cytokines. One of the strategies the NK cells use to recognize
target cells is through detection of “missing-self”, which is dependent on MHC class I
molecules [130]. A balance of negative and positive signals from cell-surface receptors
determines the regulation of NK-cell activity. Cells with a normal or unaltered expression of
MHC class I are protected from NK-cell-mediated killing. During e.g. virus infection, selfMHC molecules can be down regulated resulting in a lack of expression of self-molecules.
The reduced interaction of inhibitory receptors with MHC class I results in a predominant
positive signal that leads to activation of the NK cell with target-cell elimination as a result.
NK cell can also express receptors that have non-MHC-molecule ligands [131]. Some of the
receptors on NK cells will be further described below and are divided into two classes of
structurally distinct receptors: C-type lectin-like receptors and Ig-like receptors.
28
The C-type lectin-like receptors
The CD94 and NKG2 receptors belong to the C-type lectin receptor family and form
heterodimers, with a notably high expression on CD56bright NK cells and γδ T cells [132] but
also on activated CD8+ T cells and to some extent on CD4+ T cells and CD56dim NK cells
[133-135]. Both inhibitory and activating functional forms of this heterodimer exist and the
functional specificity is determined by which NKG2 molecule is associated to the CD94
molecule. CD94 lacks a cytoplasmic domain for signal transduction, while NKG2A and the
splice variant NKG2B mediate inhibitory signals through intracellular immunoreceptor
tyrosine-based inhibition motifs (ITIMs) [136]. All other NKG2 molecules that form
heterodimers with CD94, including NKG2C, NKG2E and the splice variant NKG2H, mediate
activating signals through DAP12 associated to immunoreceptor tyrosine-based activation
motifs (ITAMs) [137,138]. The ligand for CD94/NKG2A/B/C/E/H complexes is the MHC
class Ib molecule HLA-E [139,140].
NKG2D and NKG2F are also C-type lectin receptors, but they do not associate with CD94.
The activating receptor NKG2D is expressed as a homodimer that signals via its associated
adaptor protein DAP10 [141]. In humans, NKG2D is expressed on virtually all NK cells,
CD8+αβ T cells and γδ T cells [142]. The expression levels can be altered by exposure to
cytokines where e.g. the levels of NKG2D increase after exposure to IL-15 [143] and IL-12
[144]. The ligands for NKG2D are MICA and MICB, and human cytomegalovirus UL16
binding protein (ULBP) 1-4 [142,145,146]. These ligands are expressed at low levels on
normal cells but are frequently expressed on tumor cells and can be induced on cells infected
by viruses [147,148]. So, in the surveillance of tumors, expression of MICA and MICB by
leukemic cells increases the susceptibility to cytotoxicity by NKG2D-expressing NK cells
[144,149]. The interaction of NKG2D with its ligand also induces increased expression of
CD25, proliferation and cytokine secretion by NK cells. [150]. NKG2F expression appears to
be restricted to intracellular compartments and does not seem to be associated with CD94.
NKG2F associates with the adaptor protein DAP12 and may provide activating signals by
interacting with ligands present intracellularly. It is possible that NKG2F regulates cellular
activation through competition for DAP12 with other receptors [151].
29
Receptor (alternative Ligands
name)
(alternative name)
Function
Signaling
Refs
CD94/NKG2A
(CD159a)
CD94/NKG2C
(CD159c)
NKG2D
(CD314)
NKG2F
CD161
(NKR-P1A)
HLA-E
Inhibitory
ITIM
[136,139,140]
HLA-E
Activating
DAP12
[137,139,140]
MICA, MICB, ULBPs Activating
DAP10
[141,142,152]
Unknown
LLT-1 (osteoclast
inhibitory lectin,
CLEC2D)
AICL (CLEC2B)
Activating
Inhibitory
DAP12
ITIM
[151]
[153,154]
Activating
Unclear
[155-157]
Tyrosine-based motifs
(untypical ITIMs)
suggested, with
undefined adaptor
proteins
NKp80
(KLRF1, CLEC5C)
Table 2. General summary of some C-type lectin-like receptors on human NK cells.
The Ig-like receptors
The killer Ig-like receptors (KIRs)
The KIRs are expressed on NK cells (preferably CD56dim NK cells) and on a minor
subpopulation of both CD8+αβ and γδT cells [158,159]. The ligands for KIRs are MHC class
I molecules (including HLA-A, -B, -C and -G) [160]. KIR interaction with MHC class I
molecules results in inhibitory or activating signals. The inhibitory KIRs have a long
cytoplasmic tail containing ITIMs that mediate negative signals. The activating KIRs have
short cytoplasmic tails and a positively charged amino acid in the transmembrane domain.
Association of the adaptor protein DAP12 to the charged amino acid in the transmembrane
domain allows for activating signals through ITAMs of DAP12 [161,162].
To date, 16 different KIR genes have been described and the genetic setup differs between
individuals. The KIR genes are highly polymorphic and different NK-cell clones express a
random combination of the present KIR genes, rendering a great diversity of the KIR-receptor
repertoire between individuals and when comparing NK cells within one individual
[160,163]. Although the gene expression is highly variable, there is some degree of
organization. Two main groups, the A haplotype and the B haplotype, have been
characterized with different KIR gene content. The A haplotype only exhibits one gene for an
activating KIR and several inhibitory KIRs, while the B haplotype is more diverse in gene
30
content and contain several activating KIR genes [163]. In nearly all haplotypes, so-called
framework genes are present, including the three KIR genes 3DL3, 2DL4 and 3DL2 [160].
Leucocyte Ig-like receptor-1 (LIR-1)
Another type of Ig-like receptor is LIR-1; also called Ig-like transcript (ILT)-2 or CD85j.
LIR-1 is expressed on NK cells and T cells but also on myeloid cells like B cells, monocytes
and DCs [164]. LIR-1 recognizes MHC class I molecules on surrounding cells. Among MHC
molecules, LIR-1 binds to HLA-A, -B, -C and HLA-G [164]. HLA-G has the highest affinity
while UL18, a virally encoded MHC-like molecule, has by far the highest affinity for LIR-1
[165,166]. The function of the receptor is inhibitory and binding results in negative signaling
mediated by four intracellular ITIMs [167].
The Natural cytotoxicity receptors (NCRs)
The NCRs include NKp30, NKp46 and NKp44, of which the two first mentioned are
constitutively and selectively expressed on all peripheral NK cells. NKp44 is upregulated on
IL-2 activated NK cells and is also expressed on some γδT cells [168,169]. Natural
cytotoxicity receptors (NCRs) are Ig-like receptors that trigger lysis of tumors and virusinfected cells upon interaction with ligands on the target cells. Both NKp44 and NKp46 can
bind viral hemagglutinins, supporting the role of NCRs in recognition of virus-infected cells
[170,171]. HIV infected cells have been reported to express a ligand (still undefined) for
NKp44 [172]. In herpes simplex virus-infected cells, expression of a viral gene product is
recognized by NCRs leading to increased susceptibility to NK-cell mediated lysis [173]. The
NK-cell mediated cytotoxicity has been reported to be inhibited by interaction between the
tegument protein, pp65, from human cytomegalovirus (CMV) and NKp30 [174] perhaps
demonstrating an escape mechanism by the virus. NKp30 has also been showed to be
involved in the NK-cell induced maturation of DCs and in killing of iDCs [175,176].
31
Receptor
(alternative
name)
KIR2DL1
(CD158b)
KIR2DL2
(CD158b1)
KIR2DL3
(CD158b2)
KIR2DL4
(CD158d)
KIR2DL5
(CD158f)
KIR3DL1
(CD158e1)
KIR3DL2
(CD158k)
KIR2DS1
(CD158h)
KIR2DS2
(CD158j)
KIR2DS3
KIR2DS4
(CD158i)
KIR2DS5
KIR3DS1
(CD158e2)
KIR3DL3
LIR-1
(CD85j, ILT2)
LAIR-1
(CD305)
CD244
(2B4)
NKp30
(CD337)
NKp44
(CD336)
NKp46
(CD335)
Ligands
(alternative name)
Function
Signaling
Refs
HLA-C2
Inhibitory
ITIM
[160]
HLA-C1
Inhibitory
ITIM
[160]
HLA-C1
Inhibitory
ITIM
[160]
HLA-G
Activating/inhibitory
ITIM/FcεRI-γ [160,177,178]
Unknown
Inhibitory
ITIM
[160]
HLA-Bw4
Inhibitory
ITIM
[160]
HLA-A3, A11
Inhibitory
ITIM
[160]
HLA-C2
Activating
DAP12
[160,177]
Unknown
Activating
DAP12
[160,177]
Unknown
HLA-Cw4
Activating
Activating
DAP12
[160]
[160,177]
Unknown
HLA-Bw4?
Activating
Activating
DAP12
[160]
[160,177]
Unknown
HLA-class I
Unknown
Inhibitory
ITIM
[160]
[165,167]
Collagen
Inhibitory
ITIM
[179]
CD48
Activating/Inhibitory
ITSM, SAP
[180,181]
Unknown
Activating
FcεR1-γ
[181]
Unknown
Activating
DAP12
[181,182]
Unknown
Activating
FcεR1-γ
[181]
Table 3. General summary of some immunoglobulin (Ig)-like receptors on human NK
cells.
Antibody-dependent cellular cytotoxicity (ADCC) mediated by NK cells
The CD16 receptor is expressed on almost all CD56dim NK cells. Another name for CD16
is FcγRIIIA that is the low affinity receptor for IgG. Ligation of CD16 to IgG bound to
specific antigen on the target cells, IgG-immune complexes or anti-CD16 monoclonal
antibodies (mAb) induces cytolysis through ADCC [183] and also increased expression of the
IL-2 receptor and production of several cytokines [184]. Crosslinking of CD16 has also been
32
shown to induce apoptosis in NK cells [185]. The surface expression of CD16 can be altered
by cytokines. CD16+ NK cells convert to CD16- NK cells in the presence of TGF-β [186].
After in vitro culture with IL-2 and IL-12 the expression of CD16 is decreased and can be reexpressed to some extent by IL-15 [187]. A polymorphism in the extracellular domain 2 of
CD16 has been reported to affect ligand binding by NK cells and may play a role in human
autoimmune disease [188]. It was recently described in a mouse model that NK cells were
activated by IgE through CD16 and responded by both production of IFN-γ and ADCC [189].
Chemokines and NK-cell homing
Chemokines and chemokine receptors are important for the circulation of immune cells to
different organs and tissues. For NK cells to be able to mediate their cytolytic and regulatory
functions, they must be recruited to the site where their effector functions should be applied.
The CD56bright and CD56dim NK-cell subsets express different chemokine receptors,
suggesting that they home to different sites in vivo. CD56bright NK cells express high levels of
e.g. CCR7, involved in homing of immune cells to secondary lymphoid organs [190], and also
the adhesion molecule L-selectin (CD62L) important for the interaction with endothelial cells
[191]. CX3CR1 and the IL-8 receptor (CXCR1) are expressed on CD56dim NK cells but not
on CD56bright NK cells [115]. Thus, the CD56dim NK cell subset migrates in response to IL-8
(chemokine CXCL8) [192] which is released during early responses to infection. Several
chemokines have been shown to be involved in the regulation of NK-cell mediated cytolysis
by promoting cytotoxic granular release [193].
NK-cell generation
NK cells, along with other lymphocytes, originate from CD34+ haematopoietic precursor
cells (HPCs) present in the bone marrow of adults. During fetal life NK cells are generated
from precursors in the liver in humans [194], and in the thymus in mice [195]. The bone
marrow is considered to be the primary site of NK-cell generation in adults because it
provides the required interaction with stromal cells and composes a rich source of cytokines
and growth factors. The importance of the bone marrow for generation of NK cells is shown
by the loss of NK cells upon bone marrow depletion [196]. However, NK cells at different
developmental stages have been found in other organs of the body, including liver, spleen,
lymph nodes and in blood [192,197-200]. Organs like thymus and spleen are not essential in
33
the generation of NK cells, since normal numbers of functional NK cells are present, in
humans and mice, even though they lack functional organs of this type [192]. This does not
preclude that NK cells can develop at these sites. It is unclear whether NK-cell precursors
(NKPs) found at these sites are derived from precursors migrating from the bone marrow or if
these sites can actually support NK-cell differentiation from local HPC.
The ontogeny of the CD56bright and CD56dim NK cells is unclear. It has been suggested that
CD56bright NK cells are precursors for the CD56dim NK cells [201,202]. The CD56bright
subpopulation of human NK cells is preferentially found in secondary lymphoid tissues and at
sites of inflammation [203-205]. These cells might develop within lymphoid tissue as CD34+
HPC isolated from lymph nodes can be induced to differentiate in vitro to CD56bright NK cells
in the presence of IL-2 or IL-15 or after culture with activated T cells alone [198]. NK cells
have been reported to reside in close proximity to T-cell rich regions in lymph nodes in vivo
[198,206]. This could support the theory of a direct interaction between NK cells and T cells
at the site of T-cell activation and thus a possibility of NK cells to affect this process.
Data concerning the life span of mature NK cells in vivo is diverse, perhaps due to
different methods of transfusion and tracking of the cells. Splenic mature NK cells, labelled
with tracking dyes and transferred to normal adult mice, have been estimated to have a halflife of 10 days [207] up to almost 3 weeks [105]. In humans, transfused allogenic NK cells in
patients with cancer have been detected for up to 3 days in blood [208]. In another study,
human blood NK cells in healthy individuals were suggested to have a turnover rate of
approximately 2 weeks [209].
Receptor expression during development
The general consensus is that HPC develop into either common lymphoid progenitors
(CLP), (giving rise to B, T and NK cells) or common myeloid progenitors (CMP), (giving rise
to monocytes, granulocytes, erythrocytes and mast cells). The understanding of human NKcell development is incomplete. To begin with, it is uncertain exactly what factor/s that
determines the commitment of CLPs to the NK-cell linage. Although, in vitro studies have
shown that cytokines like stem cell factor (SCF), fetal liver kinase 2 ligand (FLK2) and IL-7
can promote the development of NKPs, they are not essential for NK-cell commitment.
However, once the CD34+CD45RA+CD117+ CLPs acquire expression of the interleukin-2
receptor-β (IL-2Rβ) they are denoted NKPs and have the potential to develop into mature NK
cells. The further development has been described to take place in different stages where
34
CD161 expression is evident very early on immature NK cells. The immature NK-cell stage is
followed by a sequential process of maturation that seems to be initiated with the acquisition
of CD56 together with features of cytotoxicity [192]. The maturation stages proceed as NK
cells start to express receptors that are specific for self-MHC molecules, such as
CD94/NKG2A or KIRs, which bind to MHC class I molecules. The CD94/NKG2A
heterodimeric receptor complex is more dominantly expressed compared to the KIRs in
humans and most of the analogous Ly-49 inhibitory receptors in mice [210,211] during early
stages of development (Figure 1).
The KIR receptor repertoire on NK cells is mainly determined by gene variations and
polymorphisms, as described above. It has, however, been suggested that MHC class I might
have some influence on the KIR expression on NK cells where many optional MHC class I
ligands is associated to fewer cognate KIRs. This inverse relationship also extends to the
frequency of NK cells expression NKG2A, being inversely correlated to the number of
KIR:MHC class I pairs [212]. In general, developing NK cells express inhibitory receptors
before the activating, as if to assure that they are able to be inhibited by self molecules before
they gain the capacity to be activated [213]. Notably, a subpopulation of human NK cells
without inhibitory receptors has been found and the tolerance to self seems to be achieved
through hyporesponsiveness by these NK cells [126]. Upon in vitro stimulation with IL-15
and stromal cells, these anergic NK cells start to express inhibitory receptors and acquire
effector functions [214].
35
Figure 1. Phenotypic and functional features of developing NK cells. Several NK-cell related
molecular markers are expressed in a sequential manner during NK cell development. NKPs do not express
specific NK-cell markers but give rise to NK cells. Immature NK cells express CD161 together with receptors
required for growth and survival. In the periphery, NK cells can become activated after e.g. detection of missing
or altered self-MHC class I molecules or by cytokines. Adapted by permission from Macmillan Publishers Ltd:
Nature Reviews Immunology 2007; 7 (9) 703-714 [200].
NK-cell education
To prevent autoreactivity, NK cells need to discriminate aberrant cells from normal cells
and this is achieved through a combination of activating and inhibitory receptors. Although
the process is not well understood, host MHC class I molecules are believed to be part of the
development of NK-cell reactivity and self-tolerance. An educational model called
“licensing” has been suggested by Yokoyama et al. [215]. In this model NK cells are induced
to mature only after successful engagement of inhibitory ligands. Another model named “the
disarming theory” that have been proposed by Raulet et al suggests that, NK cells that fail to
receive inhibition become anergic [216].
36
NK receptors on T cells
NK-cell receptors were first described on NK cells, hence the name. However, other
immune cells like T cells, B cells and monocytes also express some of these receptors [217].
When NK receptors are expressed on T cells they can function in a costimulatory or
coinhibitory fashion [218].
On human naïve CD8+ T cells, NKG2D serves as a costimulatory receptor for the TCR
resulting in priming to a type 1 phenotype [219]. CD8+ T cells with specificity for CMV show
increased cytokine and cytolytic responses upon NKG2D ligation [148]. Inhibitory KIRs are
expressed mainly on CD8+ αβT cells [158]. CD8+ T cells expressing KIRs are not found in
CB [220] but memory KIR+CD8+ T cells have been reported to accumulate with increasing
age [221]. The expression of KIRs by T cells is associated with resistance of effector-cellmediated cell death [222]. Of the receptors present in the LIR family, only LIR-1 has been
described on T cells. LIR-1 appears to be primarily expressed on effector T cells that are KIR
negative [223] whereas the KIR expression appears to be more restricted to memory T cells
[224]. The co-ligation of TCRs and inhibitory KIRs or LIRs can reduce or prevent T-cell
effector functions including both cytokine production and cytotoxicity [158,220,225-227].
Although the mechanism is unkown, CD8+ T cells have been reported to be able to mediate
MHC-independent lysis of CMV infected cells through UL18 and LIR-1 interaction,
suggesting alterations in the function of LIR-1 [228].
NK cells bridge to adaptive immunity
The interaction between NK cells and several cell types can have immuno-modulatory
effects on the outcome of the adaptive immunity. Lymph node resident CD56bright NK cells
can, through cell contact together with GM-CSF and CD154, trigger CD14+ monocytes to
differentiate into DCs with a Th1-promoting activity [229]. CD56bright NK cells have been
reported to produce IFN-γ in response to IL-12 derived from LPS activated DCs [230], which
might be important for the priming of adaptive immune cells, as IL-12 exposed NK cells have
been shown to favor the selection of mature DCs that are associated to Th1-cell priming
[231]. In a murine model it was shown, in vivo, that upon antigen stimulation in lymph nodes,
NK cells are recruited and there they provide an early source of IFN-γ needed for Th1 priming
of T cells [232].
37
NK cells have been shown to affect antibody responses by B cells. NK cell knock-out mice
can develop a Th1 response and produce IFN-γ but are deficient in their antigen-specific
IgG2a antibody response [233]. In humans, NK cells are able to induce B-cell differentiation
into antibody-producing cells and to isotype switching to IgG and IgA. This function seems to
be regulated through CD40-CD40 ligand interactions [234]. Human IFN-γ producing NK
cells have also been shown to inhibit in vitro IgE production [90].
NK cells and accessory cells
Many pathogens are recognized by accessory cells like DCs, monocytes and macrophages
that activate NK cells through cytokines and/or by signals that are contact dependent [235].
The effects on NK-cell activity by accessory cell derived cytokines are variable. IL-12
induces IFN-γ production by NK cells [97], increases the cytotoxic activity and have a
proliferative effect [93]. IL-15 is important in the initiation of NK-cell mediated cytotoxicity
by inducing surface expression of the adhesion molecule LFA-1 [106] but is also crucial for
NK-cell development and induction of NK-cell proliferation and survival [97,103-105].
Cytokines delivered to NK cells through synaptic formation is an efficient route of activation
[236]. NK cells also affects accessory cells, suggesting a reciprocal regulation, which might
depend on the nature of the cellular stimuli. In in vitro cultures with LPS, NK cells can
enhance DC maturation and IL-12 production [237] and the maturation of DCs can be
mediated by NK-cell derived TNF and IFN-γ [176]. Ligation of the NKp30 receptor on NK
cells has been suggested to be involved in both the killing of iDCs and in the induction of
IFN-γ production by NK cells [175]. DC-derived transforming growth factor beta 1 (TGF-β1)
can inhibit the expression of NKp30, and thus, decrease the NK-cell mediated DC regulation
[115]. A reciprocal regulation between macrophages and NK cells has also been reported.
Macrophages can induce NK-cell proliferation and functions, while NK cells control
macrophage activity through elimination of overstimualted macrophages [238]. The
importance of accessory cell and NK cell interaction has been demonstrated in relation to
different diseases like malaria [239], rheumatoid arthritis [229] and respiratory allergic
disorders [240].
38
HERPESVIRUSES
The family of herpesviruses (Herpesviridae) includes viruses of the α-, β- and γ-subfamily.
CMV belongs to the β-herpesviruses, which all have a rather restricted host range and a
replicating cycle of days resulting in enlarged cells. Epstein-Barr virus (EBV) is included in
the family of γ-herpesviruses that replicate in lymphoblastoid cells. Both CMV and EBV are
widespread pathogens with a majority of the population living in industrial countries being
seropositive and the prevalence of seropositivity is even higher in populations with low
socioeconomic status. Primary infection usually occurs during early childhood. The infection
is persistent throughout life and asymptomatic in healthy people. However, if primary EBV
infection is acquired later in life it can cause infectious mononucleosis that is associated with
e.g. fever, sore throat and fatigue. Breast milk and saliva is the most common route of
transmission of CMV and for EBV through saliva. Upon primary infection the virus is
amplified in a permissive cell type and during the asymptomatic latent phase of infection the
virus is sustained in B cells for EBV and in cells of the myeloid linage for CMV [241,242].
Herpesvirus infections are in large controlled by T-cell immunity [243] but also B cells and
NK cells are important in combating the virus. During early phases of EBV infection in vitro,
tonsillar NK cells stimulated by EBV-activated DCs produce vast amounts of IFN-γ that
limits the EBV-induced B-cell proliferation. For an in vivo scenario, the authors suggest that
the innate immune response by NK cells and DCs would limit primary EBV infection until
the specific immunity has been recruited [244]. The expression of MHC class I molecules on
B cells during EBV latency protects against NK-cell mediated attack. However, during lytic
EBV infection, the surface expression of MHC class I molecules is reduced, which increases
the risk of NK-cell mediated lysis [245]. CMV in particular has evolved several escape
strategies to evade immune responses, including a whole set of genes responsible for
downmodulation of MHC class I expression and genes responsible for evading NK-cell
recognition [246].
ALLERGY
Allergy is caused by a hypersensitivity reaction to a common antigen (i.e. allergen) and is
mediated by allergen-specific IgE antibodies or allergen-specific lymphocytes causing
symptoms from mucosal membranes. An IgE-sensitized person has allergen-specific IgE
antibodies, but do not display allergic symptoms. The definition of an allergic person is
39
presence of allergen-specific IgE antibodies and allergic symptoms. Atopy is referred to as a
personal and/or familial tendency to become IgE sensitized [247].
The allergic reaction
For some unknown reasons, some individuals start to produce IgE antibodies against
antigens (called allergens) that normally are harmless.
Initially, allergens are taken up by APCs in e.g. the respiratory tract. In the lymph node the
allergen is presented to naive T cells in a peptide-MHC class II complex. Several other types
of immune cells are present (mast cells, NK cells and T cells) that can influence the priming
of the naïve T cell. IL-4 promotes the naïve T cell towards the Th2 type that produces IL-4
and IL-13. The interaction of these Th2 cells with B cells, under the influence of IL-4 and IL13 in addition to ligation of co-stimulatory molecules, induces immunoglobulin class-switch
recombination to the IgE class. The produced IgE antibodies are distributed systemically
through the blood. The allergen-specific IgE antibodies bind to the FcεRI on tissue-resident
mast cells [16]. This is referred to as the sensitization phase and is not symptomatic.
The symptoms may appear upon a second exposure to the allergen. Allergen-binding,
crosslinks the FcεRI-bound IgE antibodies on the mast cell, which results in degranulation of
inflammatory mediators. This immediate phase reaction leads to the recruitment of other cells,
including eosinophils, T cells and macrophages, to the area. At the site, these cells become
activated and the result in a prolonged state of inflammation. This late phase reaction can be
evident around 6 hours after allergen exposure [17,18].
The symptoms, caused by IgE-mediated allergic reactions, vary depending on location and
severity. Allergic eczema/dermatitis syndrome (AEDS) is caused by reactions in the skin,
asthma in the bronchial respiratory tract and rhino-conjunctivitis in the upper respiratory tract
and eyes. In systemic anaphylaxis all the sites are involved simultaneously. If severe enough
it can cause massive vasodilation and anaphylactic shock and be life threatening.
Type 1 and type 2 cytokines in allergy
T-cell responses have traditionally been classified based on two effector scenarios, Th1
and Th2. The Th1 cell and Th2 cell nomenclature was originally suggested by Tokuhisa et al.
[248] and further distinguished in their cytokine profiles in mice by Coffman et al. [249].
Since then it has for long been a general consideration that the function of the immune system
40
is dependent on a Th1/Th2 balance in healthy individuals. This balance is reciprocally
regulated where Th1 responses downmodulate Th2 responses and vice versa. Typical Th1
inducing cytokines are IL-12 and IFN-γ, while Th2 inducing type 2 cytokines include IL-4,
IL-5 and IL-13.
In view of the classical Th1/Th2 paradigm, a Th2 skewed immune response has been
considered to be involved in the risk of early development of allergic disease. Neonates, with
an increased risk of developing allergic disease (i.e. having allergic parents) have a Th2
skewed type of response [250]. In line with this, lower numbers of IL-12- and IFN-γproducing cells in CB is associated with IgE sensitization [251]. However, there are some
contradictory results revealing a mixed Th1/Th2 type of response after allergen stimulation in
young atopic children [252,253].
Also other immune cells, like DCs [254] and NK cells [90,255] have been described as
polarized towards type 1 and type 2 cytokine production, probably affecting the fate of naïve
helper T-cell activation in lymphoid tissue.
The hygiene hypothesis
The reason for the increased incidence of allergic diseases during the last decades is not
fully understood but several possible theories have been proposed. Although there appear to
be a genetic association [256] this does not explain the rapidly increased occurrence. Another
plausible explanation includes environmental factors. Through the observations by Strachan
that more siblings decreased the risk of developing hay fever [257], the theory of the ”hygiene
hypothesis” came forth. The “hygiene hypothesis” states that due to changes in the exposure
pattern to microbial components (both infectious and non-infectious) there are insufficient
qualities and/or quantities of these factors, leading to an altered development of the immune
system with a increased tendency to develop allergic diseases.
Environmental factors in allergy development
Several studies support the hygiene hypothesis. Exposures, during early childhood, to an
environment with higher infection rate seem to have a protective effect against allergy
development. In addition to the protective effect of increasing number of siblings [257,258] it
has been documented that day care attendance in early life is associated with a reduced risk of
developing allergic disease [259,260]. Growing up on a farm is associated to an environment
41
rich in microbial components. LPS or endotoxins are components of the cell wall of Gramnegative bacteria that are highly prevalent in households of farmers [261]. The level of
endotoxin has been shown to be inversely related to the occurrence of allergic disease
[262,263]. Other bacterial components, like N-acetyl-muramic acid (a component of PGN)
have been inversely associated with asthma [264] and the high levels of bacterial DNA found
in farm barns [265] may also be part of the protective effect. In line with this, children
growing up in farm families have a reduced risk of developing allergic disease [266-270]. In
families with part-time farming, the protection is not as strong as in families with full-time
farming [269], suggesting that dosage is important and that exposure should be constant in
order to reach optimal effect.
However, the current scientific evidence also reports on opposing effects by infections on
the development of allergic disease. Helminth and mycobacteria have a protective effect but
depend on at what age the infection occurs, the route and the dose of infection. Infection by
viruses has been reported to be associated to both protection and increased risk to develop
allergic disease [271-273]. It is possible that a decreased exposure to e.g. Th2-inducing
helminths or a delayed infection during early life to Th1-inducing viruses alters the balance of
immune responses to allergens [274].
NK CELLS IN HEALTH AND DISEASE
Implication of NK cells in health and disease include many areas of research. For the
topics involved in this thesis, the focus will be on the role of NK cells in virus infections
(mainly EBV and CMV) and in allergic disease.
NK cells in viral infections
NK cells are important in the defense against viral infections. In humans this is supported
by data from cases of natural infections where NK-cell deficiencies increase susceptibility,
mainly to herpesviruses.
Genetic mutations can cause NK-cell defects. Patients with Chediak-Higashi syndrome
suffer of immunological deviations, such as deficient NK-cell cytotoxicity and ADCC. As a
result these patients have chronic active EBV infection [275]. X-linked lymphoproliferative
syndrome is associated with severe immune deficiencies. NK-cell cytotoxicity is often
markedly reduced and patients are commonly affected by fatal infectious mononucleosis,
42
caused by EBV, or/and lymphoreticular malignancies [276]. The Griscelli syndrome is caused
by another gene mutation and defects in NK-cell cytotoxicity leading to increased
susceptibility to EBV [277]. Gene mutations that cause hemophagocytic lymphohistiocytosis
affect the perforin protein, NK-cell activity or/and the percentages of NK cells. For some
patients this disease is associated with EBV infection [278]. A complete lack of NK cells has
been reported, resulting in increased susceptibility to several herpesviruses and bacterial
infections with fatal outcomes [279,280]. The importance of NK cells for antiviral responses
is further highlighted by a case study where a young girl (3 months old) was able to recover
from CMV disease without treatment, although she virtually lacked T cells. During recovery,
the viral load correlated with the number of NK cells and NK-cell derived cytokines [281].
NK cells in allergy
During infection and/or exposure to non-pathogenic microbes, cells of innate immunity are
probably activated early, leading to initiation of adaptive immune responses. The functions of
NK cells include, lysis of target cells and also regulation of immune responses by the release
of cytokines, like IFN-γ, TNF, GM-CSF, IL-4, IL-5, IL-10 and IL-13 that can affect immune
responses by other cells in different ways.
Several reports suggest a role for NK cells in allergic disease. It has been shown that atopic
asthmatic patients have a higher ratio of IL4-producing NK cells than healthy individuals
[282]. Aberrant NK-cell frequencies and functions have also been observed in patients with
atopic dermatitis, where circulating NK cells were decreased and had a defective ability to
produce TNF and IFN-γ, but not IL-4. These effects were suggested to be due to apoptosis of
NK cells, induced after cell contact with activated monocytes [283]. In a study by Buentke et
al. NK cells were shown to interact with DCs in skin lesions of atopic dermatitis patients
[284]. It has further been shown that peripheral mononuclear cells from atopic patients have a
reduced capacity to produce IFN-γ in response to IL-12 [285]. Although this was not related
to NK cells specifically, another study show reduced numbers of IL-12 producing monocytes
and IFN-γ producing NK cells in adult allergic patients with rhinoconjunctivtis [286]. In a
recent study allergic adult individuals were reported to have a reduced proportion of
CD56bright NK cells and decreased production of IFN-γ in response to DCs in vitro [240].
These data might suggest an aberrant feedback loop of IFN-γ and IL-12 between NK cells and
monocytes/DCs in allergic individuals.
43
In resemblance to the T-cell subsets Th1 or Th2, NK cells have also been shown to be able
to polarize in vitro into two subsets with different cytokine profiles. NK cells with type 1
responses produce IFN-γ and IL-10 while type 2 NK cells mainly produce IL-5 and IL-13
[90,287]. Freshly isolated human NK cells, from both adults and neonates, contain a distinct
subpopulation of immature IL-13+ NK cells that are induced to proliferate to IL-4 in vitro
[288]. In cell cultures with IL-4 and IL-13, mature IFN-γ+ NK cells are prevented to
accumulate [288] and IFN-γ in turn prevents the accumulation of IL-13+ NK cells, suggesting
a negative feedback loop between these two subsets of NK cells [289]. This could be of
importance in an allergy context, where the presence of IL-4 and IL-13 may drive the
progression of IL-13+ NK cells and prevent type 1 responses through inhibition of IFN-γ and
aggravate atopic reactions or reciprocally, in the presence of proinflammatory cytokines
suppress type 2 responses.
THE IMMUNE SYSTEM OF NEONATES AND YOUNG CHILDREN
The ability of neonates to mount an immune response is insufficient in comparison to older
individuals. However, the process of birth is followed by an age-dependent maturation of the
immune system.
The distribution and function of mononuclear cells in blood differs between newborns and
adults. Very low numbers of memory CD4+CD45RO+ T cells are present in CB [290] but the
proportion of the CD4+ [291,292] and CD8+ [293] T-cell memory pool increase with age. CB
cells proliferate less than cells from adults in response to in vitro stimulation with low doses
of the T-cell mitogens concanavalin A (Con A) and phytohaemagglutinin (PHA), but reach
equivalent proliferation levels as adults with higher concentration [294]. The fractions of DCs
and monocytes from CB appear to be similar to adults, but the function of cord-blood DCs as
accessory cells for T-cell responses to mitogens is insufficient, perhaps due to lower
expression of molecules like ICAM-1, MHC class I and II on DCs, needed for antigen
presentation [294]. The highest proportion of NK cells, in relation to lymphocytes, is found in
CB followed by a decline during early childhood years and then an increase again in adults,
although the levels do not reach that of newborns [295]. NK-cell cytotoxicity is low in CB but
is improved quickly during infancy [296].
Cytokine responses by newborns are considered to be skewed towards Th2. The
concentration of IFN-γ in serum from CB is low compared to adults, but increases during the
first days of life, while IL-2 and IL-4 can be found at higher concentration in CB than in
44
adults [297]. This early cytokine profile is also detected in T cells where, relative to adults,
smaller proportions of both CD4+ and CD8+ CB T cells are positive for IFN-γ but not IL-2
after cell culture with phorbol myristate acetate (PMA) and ionomycin [290]. CB NK cells
are, however, readily activated by cytokines. In cell cultures with IL-12 and IL-18 the
activating marker CD69 is more effectively upregulated on NK cells from CB as compared to
adults. This activation is mirrored in the release of IFN-γ by these cells that by far exceed that
of adult NK cells [298]. The production of IL-12p70 and IFN-α by CB cells is impaired in
neonates [299], which may be part in the initial inefficiency to mount Th1 responses.
The mode of delivery affects the frequency of lymphocyte populations and cytokine
production. In full term neonates, NK cells are increased in children that are vaginally
delivered (VD) as compared to children born by elective Cesarean section (ECS) [300]. CB
cells from children born by VD produce more IFN-γ and IL-12 in response to mitogens [301].
PREECLAMPSIA
Preeclampsia is a disorder seen in the later part of pregnancy with symptoms like
proteinuria, hypertension and oedema. If the condition is severe enough the outcome can be
lethal for both mother and child. Approximately 3-5% of all pregnancies are diagnosed with
preeclampsia. The cause for this pregnancy disorder is unknown but the disease is related to
the placenta since the symptoms cease upon removal of placenta and fetus [302]. Further, the
development of the placenta during preeclamptic pregnancies is deficient as seen by altered
placental morphology [303]. This can be a result of the poor penetration by extravillious
trophoblasts into the endometrium and the establishment of fewer placental arteries resulting
in poor utero-placental blood circulation [120].
Healthy pregnancies are associated with a mild inflammation in the circulation, which is
further enhanced in preeclampsia [304]. Cytokines like IL-12p40 and IL-15 are increased in
the circulation of preeclamptic women. Alterations of the cytokine balance are also local in
the placenta where IL-12p70 is practically absent in preeclampsia while it is abundant in the
placenta of healthy pregnancies [303]. A subset of NK cells is found in the human uterus and
is referred to as uterine NK (uNK) cells. During early phases of placentation these cells are
thought to be involved in the regulation of trophoblast invasion in the decidua through NKreceptor and trophoblast-ligand interactions and cytokine production [120]. One mechanism
that have been suggested in the regulation of placenta development is the interaction of
maternal KIRs on the uNK cells and fetal expression of MHC class I molecules on
45
trophoblasts cells. This statement is based on the finding that mothers with the A haplotype
for KIRs, bearing a fetus with a HLA-C2 allotype are at increased risk of preeclampsia [305].
Further, higher numbers of CD56+ cells in the deciduas of preeclamptic women [303] and a
larger proportion of NK cells in CB of neonates from preeclamptic mothers compared to the
newborns from healthy mothers [306,307], has been reported.
46
AIMS OF THE STUDY
During infancy, before adaptive immunity is matured, innate immunity is thought to be
rather important. Human natural killer (NK) cells are innate immune cells involved in the
control of virus-infected cells and can influence adaptive immunity mainly through cytokine
production. The overall aim of this study was to evaluate function and phenotype of NK cells
in children from birth and during early childhood years and if these features are altered in
children that develop early allergy, are latently infected by herpes viruses or are born by
preeclamptic mothers. In more detail, we wanted to evaluate:
Paper I: The general understanding of the maturation kinetics of NK-cell immune functions
after birth is modest. Comparisons are mainly done between CB cells and cells from adult
donors, while data on blood mononuclear cell from birth and young children are scarce. In
paper I we wanted to examine the receptor expression and IFN-γ production by NK cells in
CB and in the same children at the age of 2 years and at 5 years.
Paper II: NK cells are important during early life. Here, we wanted to evaluate early features
of NK cells that could be of importance for immune responses later in life. In paper II, the
aim was to elucidate if NK cells were altered regarding phenotype and/or function at birth in
individuals that developed allergic disease at 5 years of age.
Paper III: Herpes-virus infection associates with a reduced risk of becoming IgE-sensitized,
and NK cells are important in the control of herpes viruses. In paper III we aimed at analyzing
the functional capacity and interaction of innate immune cells (NK cells and monocytes) in
healthy 2-year-old children that were either seropositive for EBV, EBV and CMV or
seronegative for both viruses.
Study IV: Preeclampsia is associated with escalated inflammation in the circulation of the
pregnant woman. In study IV, we wanted to evaluate if the pro-inflammatory immunological
profile of the mother had an influence on the immunological profile in their neonates in
regard to the functional and phenotypic features of CB NK cells from healthy and
preeclamptic pregnancies.
47
SUBJECTS
The three first studies utilize samples from the same longitudinal and prospective study
cohort that was initiated to investigate the parental influence on the immunology of the child.
The cohort has previously been described by Nilsson C. [308]. The subjects included in the
respective studies are described in the individual papers I-III, while the study population for
the preliminary Study IV is described below.
Study population (Study IV, preliminary data)
Eighteen children from healthy mothers and 19 children from preeclamptic mothers were
included in the study. Preeclampsia was divided into moderate (n=11, >0.3 g proteinuria,
systolic/diastolic blood pressure >140/90) and severe preeclampsia (n=8, >3 g proteinuria,
systolic/diastolic blood pressure >160/110). The study included 14 mothers delivered by
vaginal delivery (VD) (n=7 healthy and n=7 preeclamptic mothers) and 23 mothers delivered
by elective caesarean section (ECS) (n=11 healthy and n=12 preeclamptic mothers) (see Table
4). CB was collected at time of delivery at the delivery unit, Karolinska University Hospital
and all women gave their informed consent to participate in the study. The Ethics Committee
of Karolinska Institute, Stockholm, Sweden, approved the study.
Group
Cases
Maternal age
at delivery
Mode of
delivery
Gestational age
Weight (children)
(years)
33 (24-38)
(VD/ECS)*
7/11
(weeks)
39 (37-43)
(g)
3540 (2810-4900)
7/12
38 (31-41)
3270 (1158-4125)
5/6
40 (37-41)
3295 (2510-4096)
2/6
37 (31-40)
2580 (1158-4125)
Neonates from
18
healthy mothers
Neonates from
19
33 (22-44)
preeclamptic mothers
Mild cases of
11
33 (27-44)
preeclampsia
Severe cases of
8
33 (22-41)
preeclampsia
*VD=vaginal delivery; ECS=elective caesarean section
Table 4. Demographic data of all neonates born from healthy and preeclamptic mothers included in the study.
Median (range) values are shown in the table.
48
MATERIAL AND METHODS
The methods used for the individual papers I-III are summarized in short below and
described in detail in the respective papers. For the preliminary Study IV the material and
methods are described in more detail here.
•
Separation of CBMCs and PBMCs (I-III)
•
Clinical evaluation (I-III)
•
Phadiatop/Cap-FEIATM (I-III)
•
Skin prick test (SPT) (I-III)
•
In vitro activation of blood cells (I-III)
•
ELISA (III)
•
Cytometric Bead Array (CBA) (II)
•
Flow cytometry (I-III)
•
Intracellular staining for analysis by flow cytometry (I and III)
•
Statistical analyses (Paper I-III)
Material and methods (Study IV, preliminary data)
Collection and isolation of cord blood mononuclear cells (CBMCs)
CBMC samples were prepared within 24 hours after collection. For preparation of serum,
CB was centrifuged at 1500 rpm for 10 minutes and the serum samples were stored in -20°C
until use. Cells were isolated using Ficoll-PaqueTMPLUS (GE Healthcare Bio-Sciences AB,
Uppsala, Sweden) and stored in RPMI 1640 supplemented with 10% heat-inactivated fetal
calf serum, L-glutamine (2mmol/L), penicillin G sodium (100 units/ml) and streptomycin
sulphate (100µg/ml) (complete tissue culture medium, CTCM) containing 10% Dimethyl
sulphoxide (DMSO). Freezing was performed gradually at 1°C/min to -70°C in a freezing
container (Nalgene Cryo 1°C, Nalge Co, Rochester, NY, USA) and thereafter stored in 135°C until use.
In vitro activation of cells
All CBMCs were thawed quickly and then washed twice in CTCM. CBMCs included in
49
the phenotypic study were aliquoted in 0.5x106 cells/well and incubated for 20 minutes in
37°C prior to surface staining. For stimulation experiments, CBMCs at a concentration of
1x106 cells/mL were cultured in CTCM alone or in the presence of IL-15 (20 ng/mL) and
peptidoglycan (PGN) (1 µg/ml) in 37°C for 24 hours.
Flow cytometry
To prevent non-specific binding of antibodies to FcR, the cells were incubated in 1% BSA
and 0,02% NaN3 in PBS (wash buffer) supplemented with 5% human serum (blocking
buffer) prior to staining with specific antibodies. APC-conjugated anti-CD56, Per-Cpconjugated anti-CD3 and PE-conjugated anti-CD16 monoclonal antibodies were used to
identify NK cells (all from BD Biosciences, San Diego, CA). NK-cell receptor expression
was studied by the following antibodies: Anti-CD69, anti-NKG2D (both PE-conjugated from
BD Biosciences), PE-conjugated anti-NKp30 and anti-NKp46 (both from Beckman Coulter,
Immunotech, Marseille, France), un-conjugated anti-NKG2A, NKG2C (RnD Systems,
Minneapolis, MN, USA) detected with PE-conjugated goat anti-mouse (DakoCytomation). As
negative controls, IgG1 (DakoCytomation, Glostrup, Denmark) and IgG2a (RnD Systems)
were used. To study intracellular cytokine production from NK cells, antibodies for IFN-γ
(FITC-conjugated from BD Biosciences) and IL-4 (PE-conjugated from BD Biosciences)
were used. Flow cytometry was performed on a FACSCalibur (BD Biosciences, Mountain
View, CA, USA) and analyses were made using Cellquest Pro software (BD, version 5.2.1).
Cytometric Bead Array
IL-12p70, TNF, IL-1β, IL-6, IL-8 and IL-10 in sera from children and mothers were
measured with Cytometric Bead Array (CBA) (BD Biociences, San Diego, CA, USA)
according to the manufacturer’s instructions. Calibration of the flow cytometer was performed
using BD FACSCompTM and BD CaliBRITETM Beads and analysis was using BD CBA
Software (all from BD Biociences, San Diego, CA, USA). The detection limits for the
cytokines were, IL-12p70 (1.9 pg/ml), TNF (3.7 pg/ml), IL-1β (7.2 pg/ml), IL-6 (2.5 pg/ml),
IL-8 (3.6 pg/ml) and IL-10 (3.3 pg/ml).
IFN-γ Enzyme-Linked Immuno Sorbent Assay (ELISA)
IFN-γ production in culture supernatants, after 24 hours of stimulation with IL-15 and
PGN, alone or in combination, was studied with ELISA. Ninety-six well plates (Costar 3690,
50
Corning Inc., Corning, NY, USA) were coated with anti-IFN-γ mAbs (2µg/ml, Mab 1-DIK,
from Mabtech, Stockholm, Sweden) and incubated ON at 4°C. Prior to adding the samples in
duplicates blocking with 0.5 % bovine serum albumin (BSA) in PBS was performed. Biotin
labelled mAb (1µg/ml, 7-B6-1-Biotin, Mabtech, Stockholm, Sweden) and streptavidinalkaline phosphatase (Mabtech, Stockholm, Sweden) were added followed by the substrate
(SIGMA-Aldrich, Stockholm, Sweden). All incubations were performed for 1 h in 37°C,
except when adding the substrate, when plates were incubated in room temperature. Standard
curve was used with calculated dilutions of recombinant IFN-γ (NIBSC, Hertfordshire, UK).
Detection limit was 3.9 pg/mL and optical densities were measured at 405 nm.
Statistics
Data is shown as median (range) in the results section. The non-parametric Mann-Whitney
U test was used to evaluate differences between the healthy and preeclamptic children.
Correlations between mother and child were calculated with Spearman Correlation test.
Statistical analyses were performed with Statistica 7.1 (STATISTICA Statsoft Inc., Tulsa,
USA). Statistical significance was assumed when p<0.05.
51
RESULTS AND DISCUSSION
Paper I
The overall understanding of the maturation kinetics of NK-cell immune functions after
birth is modest. Comparisons are mainly done between CB cells and cells from adult donors,
while data on immune characteristics of NK cells from birth and during early childhood years
are limited. In Paper I we examined the receptor expression and IFN-γ production by NK
cells in CB and in the same children at the age of 2 years and at 5 years.
In this study, we demonstrate alterations in the distribution and the phenotype of NK cells
with increasing age. The proportion of NK cells was higher in CB compared to later time
points. In line with our data, a higher fraction of NK cells in neonates compared to older age
groups has been reported [295,300]. Further, the mode of delivery has been shown to
influence the frequency of lymphocyte subsets in CB with a larger proportion of NK cells in
VD full term neonates compared to babies delivered by ECS [300]. All subjects included in
this study were VD. Human labour and delivery are characterised by an inflammatory
response with cytokines [309] that directly or indirectly could influence NK cells. Taken
together, our results and others indicate that the increased proportion of NK cells at birth may
be a consequence of the systemic inflammatory response induced by the process of delivery.
Further, the gradual decrease of peripheral blood NK cells, with age, might indicate that the
frequency drops after birth due to for example retention in secondary lymphoid tissues or at
sites of infection.
We also found that the CD56bright NK-cell population comprised similar proportions in CB
and at five years of age, but increased marginally at two years of age. An increase of
CD56bright NK cells in peripheral blood has been reported in individuals that are healthy but
infected by Mycobacterium tuberculosis [310], as well as during chronic systemic
inflammation [311]. This indicates that ongoing immune activation can result in an altered
distribution of NK cells in the periphery. The marginal increase of CD56bright NK cells at two
years of age may thus be explained by increased exposure to infectious agents during this age.
Concurrent with that theory, many of the children included in this study attended day care
centres, which could be a source of increased infection load.
Further, the in vitro induction of IFN-γ production was slightly lower in CB NK cells
compared to later time points. Although CB NK cells have been reported to have low
52
cytotoxicity [296] they are readily activated by cytokines in vitro and respond by producing
vast amounts of IFN-γ [298]. Our results regarding intracellular IFN-γ in CB NK cells are in
concordance with findings by others where similar levels of IFN-γ was induced in NK cells
from CB and adults after in vitro culture with PMA and ionomycin [312]. Taken together, our
data would agree with reports showing that the capacity of NK cells to respond to stimuli by
producing cytokines is not impaired in healthy neonates. Rather, the lower levels of IFN-γ in
CB NK cells, compared to later time points, may be due to other factors such as impaired IL12p70 production by accessory cells [299], which could result in lower activation of NK cells.
The receptor expression on NK cells changed with increasing age. The proportion of LIR1+ NK cells was found to increase with age. To our knowledge this is the first report about
expression of LIR-1 on NK cells in CB or during the early years of childhood. In adult
peripheral blood approximately 30% of the NK cells express LIR-1 [223]. Cytokines, like IL2 or IL-15, can induce expression of LIR-1 on NK cells in vitro [313]. Thus, the increased
proportion of LIR-1+ NK cells in our study may be explained by an altered cytokine milieu
with increasing age. The proportion of CD94+NKG2C- (NKG2A+) NK cells and the level of
expression of NKG2D and NKp30 decreased with age. The changes in proportion of
CD94+NKG2C-, reflecting CD94+NKG2A+ NK cells, is in line with data showing acquisition
of inhibiting receptors (CD94/NKG2A) in early NK-cell development, in both mice and
humans [314,315]. The inhibitory properties appear to be acquired in an orderly pattern so
that NK cells first can be inhibited, preventing autoreactivity. The expression of the activating
receptors NKG2D and NKp30 can be influenced by cytokines [144,316-318] and the
increased expression on NK cells in CB may be an effect of the pro-inflammatory cytokine
profile induced by labour at delivery [309].
The reason why CB NK cells express more of the activating receptors compared to later in
life, and why inhibitory receptor expression rather increase with age is not clear. One
hypothesis is that NK cells and innate immunity are relatively more important in early life
when adaptive immunity is immature. The first years of life involve an increased adaptation
to a broad variety of external and internal stimuli that may influence the NK-cell receptor
expression and function.
Paper II
The immune responses mediated by innate cells helps to determine the outcome of the
subsequent acquired immune responses, which could be important in the context of allergy
53
development. In Paper II, we wanted to elucidate if NK cells were altered regarding
phenotype and/or function at birth in individuals that later developed allergic disease.
In this study, we show alterations in the NK-cell subpopulation composition and NK-cell
receptor expression in CBMCs from newborns that became allergic at the age of 5. No
variations were detected in the T-cell compartment. Children that became allergic had a
smaller proportion of CD56bright NK cells compared to the children that did not become
allergic. Several reports show alterations in the proportion of CD56bright NK cells in peripheral
blood in pathological conditions. During treatment with IL-2R blockade in uveitis [319] and
multiple sclerosis [320] and also during IFN-β therapy of multiple sclerosis [321] the
proportion of CD56bright NK cells is increased. Furthermore, in primary HIV infection, the
proportion of CD56bright NK cells is reduced while it is expanded in chronic HIV infection
[322]. In a recent study, patients with the autoimmune inflammatory disease, systemic lupus
erythematosus (SLE) were reported to have increased proportions of CD56bright NK cells,
independent of treatment [311]. In the context of SLE being a disease promoted to some
extent by increased production of type 1 interferons and allergies generally being considered
to be driven by type 2 cytokines, it is interesting that the proportions of CD56bright NK cells
are increased in SLE [311] and decreased in children that are developing early allergy but also
in adults with manifested allergic symptoms [240].
Activated CD56bright NK cells are capable of producing IFN-γ that have multiple effects on
the immune system, including activation of APCs [84,85], Th1 differentiation [86]
and inhibition of class switching to IgE [89,90]. The CD56bright NK-cell subset and DCs are
located in close proximity to T cells in human lymph nodes [97], providing prerequisites for
NK-cell effects on T-cell priming. The local cytokine milieu might be of importance for the
contributing effects of NK cells on DC induced polarization of naïve T cells [323]. In a
murine model, injection of mature DCs induced recruitment of NK cells to lymph nodes and
NK-cell derived IFN-γ was necessary for Th1 priming [232]. In conjunction with these
reports, it is tempting to speculate that a decreased proportion of CD56bright NK cells could
possibly reduce Th1 priming, favouring Th2 priming. In line with this, it was recently
reported that also adult allergic individuals have a reduced CD56bright NK-cell subpopulation
in the periphery and that their NK-cell IFN-γ-production in response to stimulation by DCs
was reduced [240].
The group of children developing early allergy also tended to have an increased expression
of the activating receptor NKp30. Cytokines derived from accessory cells like DCs are
54
important for activation of NK cells [324] and NK cells have been reported to regulate DC by
means of the NKp30 mediated killing of immature dendritic cells (iDCs) and maturation of
DCs in vitro [175,176]. Both NK cells and DCs are recruited early to sites of inflammation,
which, besides from lymphoid tissue and priming of T cells as discussed above, could be
another possible place for interaction between the cells types. If the NKp30-mediated
regulation of DCs occurs in vivo, the reduced proportion of CD56bright NK cells in
combination with the increased expression of NKp30 on NK cells could result in an altered
regulation of early immune responses to infections. An altered ability by NK cells to stimulate
DC maturation with subsequent Th1 responses, in pre-allergic individuals, may be one factor
involved in the development of allergy. Further, the proportion of NK cells expressing
CD94/NKG2C, in children that developed early allergy tended to be decreased while both
groups had equally large proportions of NKG2A expressing NK cells. Although the
difference is not big, the allergic group, with lower NKG2C but normal NKG2A, may be
more inhibited than NK cells in the control group. An early alteration in the activating
capacity of NK cell could perhaps contribute to the reduced Th1 priming discussed above.
Whether the altered receptor expression have an effect on NK-cell function remains to be
evaluated. However, upon in vitro stimulation with IL-2 and IL-12, CB cells from the two
groups of children produced similar levels of cytokines (IFN-γ, IL-10 or TNF). Since IL-2 and
IL-12 are cytokines that readily activates NK cells, these results could be indicative of equal
activation of NK cells to cytokines between the groups.
Our results imply that an early imbalance in NK-cell subpopulations may correlate with the
development of allergy even before the onset of disease. However, if NK cells are involved in
the process of disease development the mechanism still remains to be evaluated.
Paper III
Children seropositive (SP) for certain herpesviruses have been suggested to be less likely
to become IgE-sensitized than seronegative (SN) children [271]. Recent findings in mice with
herpesvirus latency, suggest resistance to infection with bacterial pathogens through
prolonged production of the antiviral cytokine IFN-γ and systemic activation of macrophages
[325]. In Paper III, we wanted to evaluate the functional capacities of innate immunity (NK
cells and monocytes) in healthy young children that were either SP for EBV, EBV and CMV
or SN for both. We hypothesized that herpesvirus latency would be associated with a higher
55
activation capacity of innate immune responses, which could be involved in Th1 priming and
perhaps explain the reduced risk of becoming IgE-sensitized, observed by Nilsson et al [271].
In this study we demonstrate that 2-year old children SP for EBV have reduced
proportions of IFN-γ+ NK cells as well as lower intracellular IFN-γ levels upon in vitro
stimulation with IL-15 and PGN, (known to promote NK-cell survival/proliferation [97,326]
and monocyte activation [327], respectively). This reduction was even more pronounced in
children who were SP for both EBV and CMV. Since data on intracellular NK-cell IFN-γ and
levels of IFN-γ detected in cell culture supernatants correlated, kinetic differences is unlikely
the reason for the difference observed between the groups. We rather argue for a difference in
NK-cell and/or monocyte reactivity between the groups of children. Concurrent with our in
vitro data, plasma IFN-γ levels in SP children were significantly lower than in their SN
counterparts, again with the lowest levels in CMV co-infection.
Both CMV and EBV evade the immune system by interfering with the surface expression
of MHC class I on the cell surface [245,328]. The disturbed balance of inhibitory and
activating ligands may lead to direct activation of NK cells, which upon interaction with
accessory cells could induce subsequent activation of adaptive immunity. Thus, our data
could be a direct cause of viral infection, by interaction between infected cells and e.g. NK
cells, or could be indirectly associated to infection with herpesvirus. The fact that some
children are SN may reflect a very potent innate reaction towards viral infections, which
theoretically could block productive viral infections and thus preclude adaptive immune
responses. Indeed, we have observed an overall decreased cytokine production by PHAstimulated PBMCs from SN children (Nilsson et al., unpublished), which could indicate that
adaptive immunity, including T cells, is less activated. On the other hand, if herpes virus
latency confers some kind of suppression of NK-cell immunity, it might be beneficial to the
host for the development of adaptive immunity during early childhood years.
SP children also tended to have a decreased proportion of the CD14+CD16+ monocytes
subpopulation, which are more likely to differentiate into DCs upon stimulation [329]. In line
with this finding there was a tendency towards lower levels of IL-6 in culture supernatants
from SP subjects upon stimulation with IL-15 and PGN, indicating less proinflammatory
monocyte function. Concurrent with the discussion of NK cells above, also alterations in
monocytes subpopulations and function could be either directly due to viral infections, or a
more indirect consequence thereof. The activation of accessory-cell function could be
modified by herpesvirus infections. It has been shown that infection of monocyte-derived
56
DCs with CMV [330] and herpes simplex virus 1 [331] inhibits their maturation process as
well as their release of pro-inflammatory cytokines. We did however, not observe any
statistically significant differences between the groups in monocytes-related cytokine levels
(culture supernatant concentrations of IL-6 and IL-10). It is possible that the quantification of
IL-10 could have been affected by EBV and CMV encoded homologues for human IL-10
[332], although it is unlikely since the frequency of CMV infected monocytes from
seropositive donors is very low. Although, the levels of IL-12p70 in cell culture supernatants
were below the detection limit of our assay, low levels of IL-12p70 may be produced by
monocytes in the cell culture, and delivered to NK cells through synaptic formation [236].
Altered IL-12 levels/delivery could possibly contribute to the differences in NK-cell IFN-γ
responses between the groups.
The fact that the NK-cell IFN-γ production in vitro and plasma levels of IFN-γ correlated,
could imply that NK-cell derived IFN-γ was the major source of IFN-γ in plasma. T cells are
also important in controlling viral infections [242], but here their contribution to the plasma
IFN-γ is unclear. However, since the SP subjects had lower IFN-γ in their plasma, and we
have observed an overall increased cytokine production by PHA-stimulated PBMCs from SP
children (Nilsson et al. unpublished), this strongly argues against T cells as the major
contributors to the differences in plasma IFN-γ between the groups.
At first sight, the present data may seem difficult to reconcile with our previous findings
of a high cytokine production in SP children (Nilsson et al. unpublished). However, in the
study by Nilsson et al., PBMCs were activated in vitro with PHA, which mainly induces Tcell derived cytokines. Thus, the two studies indicate that seropositivity for EBV and CMV is
associated with a lower NK-cell but higher T-cell cytokine response. This is interesting in
relation to allergy development, since exposure to pathogenic and non-pathogenic agents
during childhood has a strong influence on the progressive shift from the allergy-related Th2polarization towards a Th1-Th2 balanced type of immunity [333]. NK-cell derived IFN-γ
regulates Th1 development which is essential for long-term control of many pathogens [116]
and it might also be important in preventing the development of aberrant immune reactions.
Taken together, herpesvirus infections could be one of the contributing factors needed for
maturation of adaptive immune responses. A very potent innate immunity may delay these
infections and thereby also influence the development of allergies.
57
Study IV (preliminary data)
Results
No alterations of NK-cell proportions and the expression of NK-cell receptors in
children from preeclamptic mothers
We analysed the NK-cell populations in neonates born by preeclamptic and healthy
mothers. The proportion of all NK cells and the proportion of CD56bright NK cells did not
differ between the groups. Further, there were no differences in receptor expression of
NKG2A and NKG2C with regard to the proportion of NK cells expressing the respective
receptor [NKG2A; 66% (40-72) in healthy and 63% (51-66) in preeclampsia, NKG2C; 15%
(13-78) in healthy and 19% (17-23) in preeclampsia] or in the level of expression on NK cells
in either of the subsets (data not shown). In the few cases were there was enough cells, NK
cells were further phenotyped for the activating receptors CD69, NKp30 and NKp46
(preeclamptic n=5 and healthy n=3 neonates). No significant differences were seen when
comparing either proportion or expression levels of the receptors on NK cells (data not
shown). Although no statistically significant difference could be demonstrated, there was a
tendency towards a lower expression of NKG2D on NK cells in preeclamptic children [MFI
60 (25-105)] when compared to healthy children [MFI 101 (99-142)] (p= 0.4, Figure 2).
Another study, performed in our group, with new material, that has continued to evaluate the
NKG2D expression in preeclampsia, supports these findings (Ebba Sohlberg, personal
communication).
To study if the mode of delivery could influence NK-cell proportion and receptor
expression, neonates were divided into VD (n=3) and ECS groups (n=5). An increased
number of NK cells were detected in VD (p=0.03). However, there was a statistically
significant difference in gestational age (p=0.012) where pregnancies that ended by ECS were
shorter, which may influence the distribution among lymphocytes.
58
Figure 2. Median fluorescence intensity (MFI) of NKG2D on CB human NK cells from
healthy pregnancies (n=3) and from preeclamptic pregnancies (n=5).
Low capacity of NK cells to produce IFN-γ and IL-4 in both healthy and preeclamptic
neonates
The ability of NK cells to produce IFN-γ and IL-4 in response to in vitro stimulation (IL15 and PGN) was measured by intracellular staining. There was very low spontaneous and
induced production of IFN-γ by NK cells in both the groups [0.3% (0.0-0.5) in the
preeclamptic group and 0.4% (0.0-0.8) in the healthy group]. We confirmed this poor
production of IFN-γ by ELISA, run on culture supernatants and in most cases the levels were
close to the detection limit of the assay (3.9 pg/ml). Both groups did have NK cells positive
for IL-4 although no differences in proportion of IL-4+ NK cells [5.8% (1.7-14.2) in the
preeclamptic group and 6.4% (3.1-11.6) in the healthy group] or expression level [MFI 43.5
(26.5-63.7) in the preeclamptic group and MFI 51.1 (22.3-75.6) in the healthy group] were
detected. To evaluate if the cellular response to stimulation differed at an individual level we
compared the stimulation indexes of the groups. The index is obtained by dividing the value
for stimulated cells with the value for unstimulated cells, for each individual. An index of 1
means no cellular response. A value below 1 is a downregulation, while a value above 1 is an
upregulation of the response by NK cells to IL-15 and PGN. The two groups tended to differ
in their response to in vitro stimulation of IL-15 and PGN where the proportion of IL-4+ NK
cells in neonates from healthy pregnancies decreased but were unchanged in the preeclamptic
group (p=0.14, Figure 3). No differences between the groups were detected in the regulation,
59
measured as MFI of IL-4+ NK cells.
Figure 3. Stimulation index (ratio of IL-15 and PGN stimulated/unstimulated from cell
cultures) of the proportion of IL-4+ NK cells of all lymphocytes.
Altered levels of pro-inflammatory cytokines in sera from neonates born by
preeclamptic mothers
The levels of the cytokines IL-12p70, TNF, IL-1β, IL-6, IL-8 and IL-10, in sera from
neonates born by preeclamptic and healthy mothers were evaluated. In sera from preeclamptic
neonates there were significantly decreased levels of IL-12p70 (p=0.003, Figure 4a) and TNF
(p=0.001, Figure 4b) compared to neonates from healthy mothers, while the levels of IL-8
were increased (p=0.047, Figure 4c). No differences in the levels of IL-1β, IL-6, and IL-10
between the two groups of newborn children were found.
60
Figure 4. Cytokine levels of a) IL-12, b) TNF and c) IL-8 in CB sera from healthy (n=16)
and preeclamptic (n=10) mothers.
Labour can cause elevated levels of pro-inflammatory cytokines (IL-12p70, TNF and IL-8)
in neonates [309]. To evaluate effects of labour all children, healthy and sick, were divided
into VD and ECS. There were statistically significant differences between the two groups
with regard to the levels of IL-6 (p=0.02) and IL-8 (p=0.004) where the VD group had higher
levels of both cytokines. There was a significant difference in gestational age, being higher in
the VD group (p=0.0006, median VD = 40 weeks, median ECS = 38 weeks).
To exclude labour as a confounding factor, only healthy and preeclamptic neonates
delivered by ECS were evaluated next. Levels of TNF were still significantly lower (p=0.03,
Figure 5a) and IL-8 was higher (p=0.03, Figure 5b) in the preeclamptic group. The
preeclamptic group had a significantly lower gestational age compared to the healthy group
(p=0.02) (healthy group, median=38 weeks and preeclamptic group, median=37 weeks). Only
one neonate in the ECS group was not delivered by full term.
61
Figure 5. Cytokine levels of a) TNF and b) IL-8 in CB sera from healthy (n=11) and
preeclamptic (n=4) mothers delivered by ECS.
Positive correlation of serum IL-6 levels between mother and neonate in preeclampsia
The morphology of both moderate and severe cases of preeclamptic placentae is altered,
with disturbed layers of syncytiotrophoblasts and malformed villi [303]. To evaluate if an
altered placental barrier in preeclampsia was associated to abnormal function of maternalfoetal communication, we studied the correlation of cytokines between the children and their
mothers (neonates from healthy mothers n=8, neonates from preeclamptic mothers n=8).
There was a significant correlation between IL-6 levels in the preeclamptic mothers and their
neonates (r= 0.83 p=0.042), but not in the healthy group.
Discussion
In this study we wanted to investigate whether the escalated inflammation in the
circulation of preeclamptic women [334-337] and altered placental morphology [303] had an
influence on the immunological profile in the neonates. The following parameters were
evaluated: NK-cell proportions, NK-cell receptor expression, NK-cell IFN-γ and IL-4
production and serum levels of pro- and anti-inflammatory cytokines.
Maternal preeclampsia had no effect on CB NK-cell proportions and NK-cell-subset
distribution, which is in contrast to what have been reported before [306,307]. Children born
to preeclamptic mothers are more often delivered by ECS [307] and the mode of delivery
62
[300] as well as gestational age [338] can influence NK-cell numbers. In our study we found
increased proportions of NK cells in CB from children delivered by VD, which is in line with
the study by Samelson et al [300].
Preeclampsia did not influence the proportion of NK cells expressing NKG2A, NKG2C,
CD69, NKp30 and NKp46 or the receptor levels on NK cells. To note is the large proportion
of human cord-blood NK cells expressing NKG2A compared to data obtained from analyis of
adult blood donors, reported in Paper I and a recent report [339]. The high prevalence of
NKG2A+ NK cells in CB could possibly explain their low cytotoxicity [296,339], as NKG2A
is an inhibitory receptor with an ubiquitously expressed ligand, i.e. HLA-E. Except for
inhibition of NK-cell activation, co-engagement of NKG2A can suppress CD69-initiated
cytotoxicity [340]. The proportion of cord-blood NK cells expressing NKG2C is also
concurrent with our previous findings (Paper I). Also this receptor is more prevalent in
children than healthy adults although increased proportions of NKG2C+ NK cells are evident
in adult individuals seropositive for CMV [341]. Preliminary data showed a trend towards
reduced levels of NKG2D on NK cells from children with preeclamptic mothers. NKG2D
expression can be influenced by cytokines [143,316,342] where e.g. NK cells treated with IL15 and IL-12 upregulate the expression of NKG2D [143,144], and T-cell expression of
NKG2D is upregulated by TNF [343]. The reduced NKG2D expression on NK cells from
neonates born from preeclamptic mothers could depend on the lower IL-12 and TNF levels
found in their cord sera. Further, pregnancy has also been reported to influence NKG2D
expression. The NKG2D receptor can be downregulated on PBMCs, in the mother, by soluble
MICA and MICB (sMICs) derived from the syncytiotrophoblasts in the placenta [344].
Although we did not substantiate the presence of these molecules in the mothers or their
children we found a strong correlation between IL-6 measured in sera from preeclamptic, but
not healthy, mothers and their neonates indicative of a transfer of IL-6 over the maternalfoetal interface. If sMICs would be transferred to the foetal side by leakage or active transfer,
it could possibly influence the NKG2D levels in the preeclamptic children. Although we did
not detect any differences between the groups regarding the other receptors that were
evaluated, the reduced levels of the activating receptor NKG2D on NK cells from children
born by preeclamptic mothers may indicate a population of NK cells that are less activated
and/or less capable of cytolysis or cytokine production.
The proportions of CB NK cells positive for IFN-γ and IL-4 following in vitro stimulation
was very low, as was the levels of released IFN-γ in culture supernatant with no detectable
differences between groups. The low levels of released IFN-γ is in line with earlier reports
63
[345], but our results regarding intracellular IFN-γ in CB NK cells do not agree with other
findings [312,346], which could be due to the different stimulations used in vitro. When
evaluating the stimulation index, there was a tendency towards a downregulation of IL-4
producing NK cells in children with healthy mothers. In an attempt to explain this finding, we
looked into serum levels of IL-12p70 and TNF.
According to the literature, the immune system of the neonate is skewed towards a type 2profile while the type 1-response is ameliorated during the first days of life [290,297]. This
may be due to an impaired TLR-induced APC production of pro-inflammatory cytokines seen
in neonates [347,348] and defects in intracellular events have been explained for the low IL12p70 and TNF production [349]. However, the production of cytokines in APCs found in
early life might depend on the chosen stimuli [299,345,349]. In our study, we found that CB
serum levels of IL-12p70 and TNF in foetal sera from healthy pregnancies are similar to adult
levels, while children born from preeclamptic mothers had significantly lower levels of these
cytokines. Thus, the downregulation of IL-4+ NK cells possibly reflects a dampened type 2response driven by a more pronounced pro-inflammatory response induced by APCs in
healthy neonates that do not function equally well in neonates born by preeclamptic mothers.
Although the mode of delivery can influence levels of TNF in CB, with higher levels in VD
than in unlaboured ECS [301,309], this did not explain the differences between the groups
observed here.
IL-8 levels were significantly increased in sera from neonates of preeclamptic pregnancies.
IL-8 production can be influenced by labour and therefore we subdivided all children into
mode of delivery. Not surprisingly, IL-6 and IL-8 were elevated in VD children. These
cytokines are produced in higher levels during labour [309]. IL-6 induces oxytocin, an
important hormone believed to induce labour work [350]. IL-8 can also be further escalated
during stressful deliveries like acute ECS and VD with assistance and in preterm ECS
associated with infections [351,352] and it is possible that preeclamptic pregnancies could be
associated with a higher stress during delivery. To investigate if the increased IL-8 levels in
preeclamptic cord sera were influenced by mode of delivery, we further studied neonates from
the two groups delivered by ECS, which still showed increased IL-8 in the preeclamptic
group. The higher IL-8 levels in sera from neonates with preeclamptic mothers could be due
to an overall higher load of maternal stress or to an infection in preeclamptic pregnancies.
Taken together, our results suggest that NK cells in neonates born by preeclamptic mothers
are phenotypically and functionally altered compared to neonates from healthy controls. The
cytokine levels in sera were also altered indicating that preeclampsia has an effect on the
64
immune system of the child. However, the results are preliminary and need to be confirmed
before any firm conclusions can be drawn.
65
CONCLUDING REMARKS AND FUTURE PERSPECTIVES
During early childhood years a continuous exposure to pathogens and harmless microbial
products modulates the immune system and an altered and/or delayed exposure might affect
the immune responses later in life. With age being an important factor for the outcome of both
innate and adaptive immune responses, the mechanisms responsible for disease development
might origin from early immunological variations. This work has focused on evaluating NK
cells at birth and during early childhood years.
According to the results from study I, the NK-cell populations in peripheral blood, defined
by their phenotype, are continuously changing with increasing age in healthy children. This
was paralleled by an increasing capacity to produce IFN-γ. These results indicate that
environmental factors modulate the immune system. The innate and adaptive immune systems
are tightly interrelated and disturbances of the innate responses could affect the outcome of
the adaptive responses. In study II, we found that children that developed early allergy had a
decreased proportion of CD56bright NK cells and a slightly altered receptor expression by NK
cells at birth. We speculate that these early alterations of NK cells could have an influence on
adaptive immunity and subsequently on development of allergic disease. The reason for these
alterations at birth is unknown. However, from the preliminary results of study IV, we show
data that suggest that preeclampsia can have an influence on the innate immunity of the child.
These data indicate a maternal effect on the immune status of the child.
A main finding from study III is that innate immunity seems to be affected by herpesvirus
seropositivity in children at two years of age. This is exhibited by lower production of
cytokines by innate cells that may be one factor that can influence the maturation of the
adaptive immunity.
CB NK cells have been reported to be inefficient in their cytotoxic capacity [296]. For
future evaluations it could be of interest to study their functional capacities, including
cytotoxicity and cytokine production during early childhood years. Infection by herpesvirus,
before the age of 2, associates with decreased function of innate immunity. The future plan is
to evaluate if this effect is sustained at 5 years of age. Since herpesvirus seropositivity has
been suggested to decrease the risk of becoming IgE-sensitized [271], it could be of interest to
evaluate features of adaptive immunity. In study II we found a difference in NK cell
proportions between allergic and non-allergic children, present already in CB. In a pilot study
we found that this difference did not remain at 2 and 5 years (not shown). A recent study
shows that allergic adults have a reduced proportion of CD56bright NK cells in the periphery
66
[240]. Thus, it could be of interest to follow the children from study II to see if the observed
alteration at birth reappears at an older age. For the last study, preliminary data associating
preeclampsia with CB NK-cell phenotype and function and with CB serum levels of
monokines are intriguing and it would be interesting to evaluate if these associations can be
validated in a larger cohort of neonates.
67
ACKNOWLEDGEMENTS
I would like to express my sincere gratitude to everyone that in one way or another has
contributed to this work and especially:
My supervisor Marita Troye-Blomberg for your support and dedication to this project.
My co-supervisor Louise Berg. You are the best supervisor a student could ever wish for!
You are always willing to take the time to discuss questions and I am ever so grateful for your
dedication, patients and encouragement. Your enthusiasm is contagious! 
The seniors at the department of immunology: Klavs Berzins, Carmen Fernandez, Eva
Severinson and Eva Sverremark-Ekström for your support.
Thanks to my co-workers at Sachs’ Children’s Hospital, Södersjukhuset; Caroline Nilsson,
Gunnar Lilja, Monica Nordlund and Anna Stina Ander.
Also great thanks to Klas Kärre at MTC, Karolinska Institutet for your valuable comments
on manuscripts.
Thanks to all students at the Department of Immunology that make the working
environment so warm and friendly! John Arko-Mensah who realized that my blood sugar
was low before I did and sent me away for lunch when we were teaching together! Nancy
Awah, Nora Bachmayer, Halima Balogun, Jacqueline Calla, Olga Daniela Chuquima,
Salah Farouk. Pablo Giusti for your refreshing, happy attitude! Ulrika Holmlund,
Nnaemeka Iriemenam. Elisabeth Israelsson for the extra support during the last few
months. Maria Johansson, Valentina Mangano, Amre Nasr, Jubayer Rahman, Shanie
Saghafian-Hedengren, Ylva Sjögren, Olivia Simone, Ebba Sohlberg, Fernando Sosa,
Stefania Verani.
Former students at the department: Anna Tjärnlund, Camilla Rydström, Ariane
Rodriguez, Manijeh Vafa Homann, Nina-Maria Vasconcelos, Qazi Khaleda Rahman,
68
Anna-Karin Sigfrinius (Larsson), Jacob Minang, Khosro Masjedi. Petra Amodruz, my
former room mate for our discussions and mutual encouragement.
Special thanx to Nora Bachmayer for the occasional brakes during the days to get some
“finkaffe” and for always making me laugh. Anna Tjärnlund, my fellow “norrlänning”, for
your encouraging support and the inspiration you supply by just being the person that you are.
Camilla Rydström the best creator of “chokladpraliner”! It is always great to see you when
you come around for a visit. Ylva Sjögren, the greatest traveling companion. Shanie
Saghafian-Hedengren for brightening up the working days with your lively personality.
Special thanx to Margareta “Maggan” Hagstedt for putting up with all the questions! You
are truly an asset for the department and a nice companion during coffee brakes! Ann
Sjölund for the help with practical things and the plommon-deliveries! Yummy! Thanks to
Gelana Yadeta for all the help. You always have a positive attitude! Anna-Lena Jarva for
all your fun parties and quizzes!
My deepest and warmest thanks to my friends. I treasure our friendship and you mean the
world to me!
Kamilla Östberg, my very dear friend from the childhood years in Gnarp. We have a special
bond that keeps us close even if we are apart.
My “gymnasiepolare” Jenny Elwell, Jenny “Väris” Värhammar, Fredrica Jonsson and
Mathilda Hermansson. I am very privileged to have you as my friends! Jenny E, we have to
go out dancing soon! Seriously! And “Väris”, thank you for taking my to the gym on a more
regular basis than I would have been able to achieve without your company! Especially
during this last year…
Jenny Wolin, that shares my interest in creative activities! Anna Lövgren, the master of
mixing drinks!
My dear “biologkompisar” Lina Enell, Åsa Andersson, Ida von Schéele, Anna Davidsson,
Julia Borg and Agnes Karlson. It always makes my happy to see you! What should we
“leka” next? 
69
During later years my family have been extended to include Bosse (who sadly passed away
recently), Berit, Ulf, Richard, Anneli and Henrik. Thank you for embracing me into your
family and making me feel as a part of it! Thanks also to Lars-Erik and Barbro, Margot and
Harry.
Most importantly I would like to express my warmest gratitude to my family; my sister
Susanne, my brother Niklas, my mother Siv and my father Rolf! Your support and love
throughout my life is invaluable. Mamma och pappa, jag är lyckligt lottad som har er till
föräldrar! You are the best! My sincere thanks also to Sara for you support and understanding
of this process, Emil for directing the focus towards the really important things in life, Stig
for your kindness and Barbro for all our nice chats and wonderful times together.
Fredrik, min älskling! For always focusing on the positive things in life. Without your love
and support I would not have been able to do this.
70
REFERENCES
1.
Wickelgren I. Immunology. Targeting the tolls. Science 2006; 312:184-7.
2.
Dziarski R, Gupta D. Staphylococcus aureus peptidoglycan is a toll-like receptor 2 activator: a
reevaluation. Infect Immun 2005; 73:5212-6.
3.
Gutierrez O, Pipaon C, Inohara N et al. . Induction of Nod2 in myelomonocytic and intestinal epithelial
cells via nuclear factor-kappa B activation. J Biol Chem 2002; 277:41701-5.
4.
Inohara N, Koseki T, del Peso L et al. . Nod1, an Apaf-1-like activator of caspase-9 and nuclear factorkappaB. J Biol Chem 1999; 274:14560-7.
5.
Chamaillard M, Hashimoto M, Horie Y et al. . An essential role for NOD1 in host recognition of bacterial
peptidoglycan containing diaminopimelic acid. Nat Immunol 2003; 4:702-7.
6.
Girardin SE, Boneca IG, Viala J et al. . Nod2 is a general sensor of peptidoglycan through muramyl
dipeptide (MDP) detection. J Biol Chem 2003; 278:8869-72.
7.
Bennouna S, Bliss SK, Curiel TJ, Denkers EY. Cross-talk in the innate immune system: neutrophils
instruct recruitment and activation of dendritic cells during microbial infection. J Immunol 2003;
171:6052-8.
8.
Appelberg R. Neutrophils and intracellular pathogens: beyond phagocytosis and killing. Trends Microbiol
2007; 15:87-92.
9.
Falcone FH, Dahinden CA, Gibbs BF et al. . Human basophils release interleukin-4 after stimulation with
Schistosoma mansoni egg antigen. Eur J Immunol 1996; 26:1147-55.
10.
Aumuller E, Schramm G, Gronow A, Brehm K, Gibbs BF, Doenhoff MJ, Haas H. Echinococcus
multilocularis metacestode extract triggers human basophils to release interleukin-4. Parasite Immunol
2004; 26:387-95.
11.
Charlesworth EN, Hood AF, Soter NA, Kagey-Sobotka A, Norman PS, Lichtenstein LM. Cutaneous latephase response to allergen. Mediator release and inflammatory cell infiltration. J Clin Invest 1989;
83:1519-26.
12.
Gibbs BF, Haas H, Falcone FH et al. . Purified human peripheral blood basophils release interleukin-13
and preformed interleukin-4 following immunological activation. Eur J Immunol 1996; 26:2493-8.
13.
Ganley-Leal LM, Mwinzi PN, Cetre-Sossah CB et al. . Correlation between eosinophils and protection
against reinfection with Schistosoma mansoni and the effect of human immunodeficiency virus type 1
coinfection in humans. Infect Immun 2006; 74:2169-76.
14.
Shi HZ. Eosinophils function as antigen-presenting cells. J Leukoc Biol 2004; 76:520-7.
15.
Kitamura Y, Kasugai T, Arizono N, Matsuda H. Development of mast cells and basophils: processes and
regulation mechanisms. Am J Med Sci 1993; 306:185-91.
16.
Turner H, Kinet JP. Signalling through the high-affinity IgE receptor Fc epsilonRI. Nature 1999;
402:B24-30.
17.
Grimbaldeston MA, Metz M, Yu M, Tsai M, Galli SJ. Effector and potential immunoregulatory roles of
mast cells in IgE-associated acquired immune responses. Curr Opin Immunol 2006; 18:751-60.
18.
Galli SJ, Tsai M, Piliponsky AM. The development of allergic inflammation. Nature 2008; 454:445-54.
71
19.
Malaviya R, Ikeda T, Ross E, Abraham SN. Mast cell modulation of neutrophil influx and bacterial
clearance at sites of infection through TNF-alpha. Nature 1996; 381:77-80.
20.
Di Nardo A, Vitiello A, Gallo RL. Cutting edge: mast cell antimicrobial activity is mediated by
expression of cathelicidin antimicrobial peptide. J Immunol 2003; 170:2274-8.
21.
Maurer M, Wedemeyer J, Metz M et al. . Mast cells promote homeostasis by limiting endothelin-1induced toxicity. Nature 2004; 432:512-6.
22.
Metz M, Piliponsky AM, Chen CC et al. . Mast cells can enhance resistance to snake and honeybee
venoms. Science 2006; 313:526-30.
23.
Grimbaldeston MA, Nakae S, Kalesnikoff J, Tsai M, Galli SJ. Mast cell-derived interleukin 10 limits skin
pathology in contact dermatitis and chronic irradiation with ultraviolet B. Nat Immunol 2007; 8:1095-104.
24.
Nockher WA, Scherberich JE. Expanded CD14+ CD16+ monocyte subpopulation in patients with acute
and chronic infections undergoing hemodialysis. Infect Immun 1998; 66:2782-90.
25.
Belge KU, Dayyani F, Horelt A et al. . The proinflammatory CD14+CD16+DR++ monocytes are a major
source of TNF. J Immunol 2002; 168:3536-42.
26.
Muzio M, Bosisio D, Polentarutti N et al. . Differential expression and regulation of toll-like receptors
(TLR) in human leukocytes: selective expression of TLR3 in dendritic cells. J Immunol 2000; 164:59986004.
27.
Novak N, Allam P, Geiger E, Bieber T. Characterization of monocyte subtypes in the allergic form of
atopic eczema/dermatitis syndrome. Allergy 2002; 57:931-5.
28.
Skinner NA, MacIsaac CM, Hamilton JA, Visvanathan K. Regulation of Toll-like receptor (TLR)2 and
TLR4 on CD14dimCD16+ monocytes in response to sepsis-related antigens. Clin Exp Immunol 2005;
141:270-8.
29.
Verreck FA, de Boer T, Langenberg DM et al. . 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
2004; 101:4560-5.
30.
Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse
forms of macrophage activation and polarization. Trends Immunol 2004; 25:677-86.
31.
Martinez FO, Sica A, Mantovani A, Locati M. Macrophage activation and polarization. Front Biosci
2008; 13:453-61.
32.
Shortman K, Liu YJ. Mouse and human dendritic cell subtypes. Nat Rev Immunol 2002; 2:151-61.
33.
Kaczorowski DJ, Mollen KP, Edmonds R, Billiar TR. Early events in the recognition of danger signals
after tissue injury. J Leukoc Biol 2008; 83:546-52.
34.
Guermonprez P, Valladeau J, Zitvogel L, Thery C, Amigorena S. Antigen presentation and T cell
stimulation by dendritic cells. Annu Rev Immunol 2002; 20:621-67.
35.
Siegal FP, Kadowaki N, Shodell M et al. . The nature of the principal type 1 interferon-producing cells in
human blood. Science 1999; 284:1835-7.
36.
Sallusto F, Lanzavecchia A. 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 1994; 179:1109-18.
37.
Randolph GJ, Inaba K, Robbiani DF, Steinman RM, Muller WA. Differentiation of phagocytic monocytes
into lymph node dendritic cells in vivo. Immunity 1999; 11:753-61.
72
38.
O'Callaghan CA, Tormo J, Willcox BE et al. . Structural features impose tight peptide binding specificity
in the nonclassical MHC molecule HLA-E. Mol Cell 1998; 1:531-41.
39.
Lee N, Goodlett DR, Ishitani A, Marquardt H, Geraghty DE. HLA-E surface expression depends on
binding of TAP-dependent peptides derived from certain HLA class I signal sequences. J Immunol 1998;
160:4951-60.
40.
Chumbley G, King A, Robertson K, Holmes N, Loke YW. Resistance of HLA-G and HLA-A2
transfectants to lysis by decidual NK cells. Cell Immunol 1994; 155:312-22.
41.
Llano M, Lee N, Navarro F, Garcia P, Albar JP, Geraghty DE, Lopez-Botet M. HLA-E-bound peptides
influence recognition by inhibitory and triggering CD94/NKG2 receptors: preferential response to an
HLA-G-derived nonamer. Eur J Immunol 1998; 28:2854-63.
42.
McMaster MT, Librach CL, Zhou Y et al. . Human placental HLA-G expression is restricted to
differentiated cytotrophoblasts. J Immunol 1995; 154:3771-8.
43.
Carosella ED, Paul P, Moreau P, Rouas-Freiss N. HLA-G and HLA-E: fundamental and
Pathophysiological aspects. Immunol Today 2000; 21:532-4.
44.
Collins RW. Human MHC class I chain related (MIC) genes: their biological function and relevance to
disease and transplantation. Eur J Immunogenet 2004; 31:105-14.
45.
Rengarajan J, Szabo SJ, Glimcher LH. Transcriptional regulation of Th1/Th2 polarization. Immunol
Today 2000; 21:479-83.
46.
Cosmi L, De Palma R, Santarlasci V et al. . Human interleukin 17-producing cells originate from a
CD161+CD4+ T cell precursor. J Exp Med 2008; 205:1903-16.
47.
Abbas AK, Murphy KM, Sher A. Functional diversity of helper T lymphocytes. Nature 1996; 383:787-93.
48.
de Vries JE. The role of IL-13 and its receptor in allergy and inflammatory responses. J Allergy Clin
Immunol 1998; 102:165-9.
49.
Del Prete G, Maggi E, Parronchi P et al. . IL-4 is an essential factor for the IgE synthesis induced in vitro
by human T cell clones and their supernatants. J Immunol 1988; 140:4193-8.
50.
Yao Z, Painter SL, Fanslow WC, Ulrich D, Macduff BM, Spriggs MK, Armitage RJ. Human IL-17: a
novel cytokine derived from T cells. J Immunol 1995; 155:5483-6.
51.
Kolls JK, Linden A. Interleukin-17 family members and inflammation. Immunity 2004; 21:467-76.
52.
Jovanovic DV, Di Battista JA, Martel-Pelletier J et al. . IL-17 stimulates the production and expression of
proinflammatory cytokines, IL-beta and TNF-alpha, by human macrophages. J Immunol 1998; 160:351321.
53.
Annunziato F, Cosmi L, Santarlasci V et al. . Phenotypic and functional features of human Th17 cells. J
Exp Med 2007; 204:1849-61.
54.
Wilczynski JR, Radwan M, Kalinka J. The characterization and role of regulatory T cells in immune
reactions. Front Biosci 2008; 13:2266-74.
55.
Fantini MC, Becker C, Monteleone G, Pallone F, Galle PR, Neurath MF. Cutting edge: TGF-beta induces
a regulatory phenotype in CD4+CD25- T cells through Foxp3 induction and down-regulation of Smad7. J
Immunol 2004; 172:5149-53.
56.
Nakamura K, Kitani A, Strober W. Cell contact-dependent immunosuppression by CD4(+)CD25(+)
regulatory T cells is mediated by cell surface-bound transforming growth factor beta. J Exp Med 2001;
194:629-44.
73
57.
Baecher-Allan C, Anderson DE. Regulatory cells and human cancer. Semin Cancer Biol 2006; 16:98-105.
58.
Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell 2008;
133:775-87.
59.
Bendelac A, Savage PB, Teyton L. The biology of NKT cells. Annu Rev Immunol 2007; 25:297-336.
60.
Carding SR, Egan PJ. Gammadelta T cells: functional plasticity and heterogeneity. Nat Rev Immunol
2002; 2:336-45.
61.
Wesch D, Hinz T, Kabelitz D. Analysis of the TCR Vgamma repertoire in healthy donors and HIV-1infected individuals. Int Immunol 1998; 10:1067-75.
62.
Kabelitz D, Glatzel A, Wesch D. Antigen recognition by human gammadelta T lymphocytes. Int Arch
Allergy Immunol 2000; 122:1-7.
63.
Groh V, Steinle A, Bauer S, Spies T. Recognition of stress-induced MHC molecules by intestinal
epithelial gammadelta T cells. Science 1998; 279:1737-40.
64.
Zhao J, Huang J, Chen H, Cui L, He W. Vdelta1 T cell receptor binds specifically to MHC I chain related
A: molecular and biochemical evidences. Biochem Biophys Res Commun 2006; 339:232-40.
65.
Brandes M, Willimann K, Moser B. Professional antigen-presentation function by human gammadelta T
Cells. Science 2005; 309:264-8.
66.
Roessner K, Wolfe J, Shi C, Sigal LH, Huber S, Budd RC. High expression of Fas ligand by synovial
fluid-derived gamma delta T cells in Lyme arthritis. J Immunol 2003; 170:2702-10.
67.
Poulsen LK, Hummelshoj L. Triggers of IgE class switching and allergy development. Ann Med 2007;
39:440-56.
68.
Muller U, Steinhoff U, Reis LF, Hemmi S, Pavlovic J, Zinkernagel RM, Aguet M. Functional role of type
I and type II interferons in antiviral defense. Science 1994; 264:1918-21.
69.
Rogge L, Barberis-Maino L, Biffi M, Passini N, Presky DH, Gubler U, Sinigaglia F. Selective expression
of an interleukin-12 receptor component by human T helper 1 cells. J Exp Med 1997; 185:825-31.
70.
Korngold R, Blank KJ, Murasko DM. Effect of interferon on thoracic duct lymphocyte output: induction
with either poly I:poly C or vaccinia virus. J Immunol 1983; 130:2236-40.
71.
Ishikawa R, Biron CA. IFN induction and associated changes in splenic leukocyte distribution. J Immunol
1993; 150:3713-27.
72.
Salazar-Mather TP, Ishikawa R, Biron CA. NK cell trafficking and cytokine expression in splenic
compartments after IFN induction and viral infection. J Immunol 1996; 157:3054-64.
73.
Tough DF, Borrow P, Sprent J. Induction of bystander T cell proliferation by viruses and type I interferon
in vivo. Science 1996; 272:1947-50.
74.
Biron CA, Nguyen KB, Pien GC, Cousens LP, Salazar-Mather TP. Natural killer cells in antiviral
defense: function and regulation by innate cytokines. Annu Rev Immunol 1999; 17:189-220.
75.
Swann JB, Hayakawa Y, Zerafa N et al. . Type I IFN contributes to NK cell homeostasis, activation, and
antitumor function. J Immunol 2007; 178:7540-9.
76.
Bandyopadhyay S, Miller DS, Matsumoto-Kobayashi M, Clark SC, Starr SE. Effects of interferons and
interleukin 2 on natural killing of cytomegalovirus-infected fibroblasts. Clin Exp Immunol 1987; 67:37282.
74
77.
Nguyen KB, Salazar-Mather TP, Dalod MY et al. . Coordinated and distinct roles for IFN-alpha beta, IL12, and IL-15 regulation of NK cell responses to viral infection. J Immunol 2002; 169:4279-87.
78.
Street NE, Mosmann TR. Functional diversity of T lymphocytes due to secretion of different cytokine
patterns. FASEB J 1991; 5:171-7.
79.
Balashov KE, Khoury SJ, Hafler DA, Weiner HL. Inhibition of T cell responses by activated human
CD8+ T cells is mediated by interferon-gamma and is defective in chronic progressive multiple sclerosis.
J Clin Invest 1995; 95:2711-9.
80.
Cooper MA, Fehniger TA, Turner SC et al. . Human natural killer cells: a unique innate
immunoregulatory role for the CD56(bright) subset. Blood 2001; 97:3146-51.
81.
Bach EA, Aguet M, Schreiber RD. The IFN gamma receptor: a paradigm for cytokine receptor signaling.
Annu Rev Immunol 1997; 15:563-91.
82.
Valente G, Ozmen L, Novelli F, Geuna M, Palestro G, Forni G, Garotta G. Distribution of interferongamma receptor in human tissues. Eur J Immunol 1992; 22:2403-12.
83.
Santoli D, Trinchieri G, Koprowski H. Cell-mediated cytotoxicity against virus-infected target cells in
humans. II. Interferon induction and activation of natural killer cells. J Immunol 1978; 121:532-8.
84.
Freedman AS, Freeman GJ, Rhynhart K, Nadler LM. Selective induction of B7/BB-1 on interferongamma stimulated monocytes: a potential mechanism for amplification of T cell activation through the
CD28 pathway. Cell Immunol 1991; 137:429-37.
85.
Farrar MA, Schreiber RD. The molecular cell biology of interferon-gamma and its receptor. Annu Rev
Immunol 1993; 11:571-611.
86.
Macatonia SE, Hsieh CS, Murphy KM, O'Garra A. Dendritic cells and macrophages are required for Th1
development of CD4+ T cells from alpha beta TCR transgenic mice: IL-12 substitution for macrophages
to stimulate IFN-gamma production is IFN-gamma-dependent. Int Immunol 1993; 5:1119-28.
87.
Hardy KJ, Sawada T. Human gamma interferon strongly upregulates its own gene expression in
peripheral blood lymphocytes. J Exp Med 1989; 170:1021-6.
88.
Bach EA, Szabo SJ, Dighe AS, Ashkenazi A, Aguet M, Murphy KM, Schreiber RD. Ligand-induced
autoregulation of IFN-gamma receptor beta chain expression in T helper cell subsets. Science 1995;
270:1215-8.
89.
Pene J, Rousset F, Briere F et al. . IgE production by normal human B cells induced by alloreactive T cell
clones is mediated by IL-4 and suppressed by IFN-gamma. J Immunol 1988; 141:1218-24.
90.
Aktas E, Akdis M, Bilgic S et al. . Different natural killer (NK) receptor expression and immunoglobulin
E (IgE) regulation by NK1 and NK2 cells. Clin Exp Immunol 2005; 140:301-9.
91.
Idriss HT, Naismith JH. TNF alpha and the TNF receptor superfamily: structure-function relationship(s).
Microsc Res Tech 2000; 50:184-95.
92.
Gupta S. A decision between life and death during TNF-alpha-induced signaling. J Clin Immunol 2002;
22:185-94.
93.
Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev
Immunol 2003; 3:133-46.
94.
Snijders A, Kalinski P, Hilkens CM, Kapsenberg ML. High-level IL-12 production by human dendritic
cells requires two signals. Int Immunol 1998; 10:1593-8.
95.
Gerosa F, Paganin C, Peritt D et al. . Interleukin-12 primes human CD4 and CD8 T cell clones for high
production of both interferon-gamma and interleukin-10. J Exp Med 1996; 183:2559-69.
75
96.
D'Andrea A, Aste-Amezaga M, Valiante NM, Ma X, Kubin M, Trinchieri G. Interleukin 10 (IL-10)
inhibits human lymphocyte interferon gamma-production by suppressing natural killer cell stimulatory
factor/IL-12 synthesis in accessory cells. J Exp Med 1993; 178:1041-8.
97.
Ferlazzo G, Pack M, Thomas D et al. . Distinct roles of IL-12 and IL-15 in human natural killer cell
activation by dendritic cells from secondary lymphoid organs. Proc Natl Acad Sci U S A 2004;
101:16606-11.
98.
Giri JG, Ahdieh M, Eisenman J et al. . Utilization of the beta and gamma chains of the IL-2 receptor by
the novel cytokine IL-15. EMBO J 1994; 13:2822-30.
99.
Giri JG, Kumaki S, Ahdieh M et al. . Identification and cloning of a novel IL-15 binding protein that is
structurally related to the alpha chain of the IL-2 receptor. EMBO J 1995; 14:3654-63.
100. Caligiuri MA, Zmuidzinas A, Manley TJ, Levine H, Smith KA, Ritz J. Functional consequences of
interleukin 2 receptor expression on resting human lymphocytes. Identification of a novel natural killer
cell subset with high affinity receptors. J Exp Med 1990; 171:1509-26.
101. Nagler A, Lanier LL, Phillips JH. Constitutive expression of high affinity interleukin 2 receptors on
human CD16-natural killer cells in vivo. J Exp Med 1990; 171:1527-33.
102. Zhou J, Zhang J, Lichtenheld MG, Meadows GG. A role for NF-kappa B activation in perforin expression
of NK cells upon IL-2 receptor signaling. J Immunol 2002; 169:1319-25.
103. Cooper MA, Bush JE, Fehniger TA et al. . In vivo evidence for a dependence on interleukin 15 for
survival of natural killer cells. Blood 2002; 100:3633-8.
104. Prlic M, Blazar BR, Farrar MA, Jameson SC. In vivo survival and homeostatic proliferation of natural
killer cells. J Exp Med 2003; 197:967-76.
105. Jamieson AM, Isnard P, Dorfman JR, Coles MC, Raulet DH. Turnover and proliferation of NK cells in
steady state and lymphopenic conditions. J Immunol 2004; 172:864-70.
106. Barao I, Hudig D, Ascensao JL. IL-15-mediated induction of LFA-1 is a late step required for cytotoxic
differentiation of human NK cells from CD34+Lin- bone marrow cells. J Immunol 2003; 171:683-90.
107. Granucci F, Vizzardelli C, Pavelka N et al. . Inducible IL-2 production by dendritic cells revealed by
global gene expression analysis. Nat Immunol 2001; 2:882-8.
108. Setoguchi R, Hori S, Takahashi T, Sakaguchi S. Homeostatic maintenance of natural Foxp3(+) CD25(+)
CD4(+) regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2
neutralization. J Exp Med 2005; 201:723-35.
109. Musso T, Calosso L, Zucca M et al. . Human monocytes constitutively express membrane-bound,
biologically active, and interferon-gamma-upregulated interleukin-15. Blood 1999; 93:3531-9.
110. Mattei F, Schiavoni G, Belardelli F, Tough DF. IL-15 is expressed by dendritic cells in response to type I
IFN, double-stranded RNA, or lipopolysaccharide and promotes dendritic cell activation. J Immunol
2001; 167:1179-87.
111. Punnonen J, Aversa G, Cocks BG et al. . Interleukin 13 induces interleukin 4-independent IgG4 and IgE
synthesis and CD23 expression by human B cells. Proc Natl Acad Sci U S A 1993; 90:3730-4.
112. Punnonen J, Yssel H, de Vries JE. The relative contribution of IL-4 and IL-13 to human IgE synthesis
induced by activated CD4+ or CD8+ T cells. J Allergy Clin Immunol 1997; 100:792-801.
113. Defrance T, Vanbervliet B, Aubry JP et al. . B cell growth-promoting activity of recombinant human
interleukin 4. J Immunol 1987; 139:1135-41.
76
114. Mosmann TR, Sad S. The expanding universe of T-cell subsets: Th1, Th2 and more. Immunol Today
1996; 17:138-46.
115. Castriconi R, Cantoni C, Della Chiesa M et al. . Transforming growth factor beta 1 inhibits expression of
NKp30 and NKG2D receptors: consequences for the NK-mediated killing of dendritic cells. Proc Natl
Acad Sci U S A 2003; 100:4120-5.
116. Laouar Y, Sutterwala FS, Gorelik L, Flavell RA. Transforming growth factor-beta controls T helper type
1 cell development through regulation of natural killer cell interferon-gamma. Nat Immunol 2005; 6:6007.
117. Cooper MA, Fehniger TA, Caligiuri MA. The biology of human natural killer-cell subsets. Trends
Immunol 2001; 22:633-40.
118. Ferlazzo G, Thomas D, Lin SL et al. . The abundant NK cells in human secondary lymphoid tissues
require activation to express killer cell Ig-like receptors and become cytolytic. J Immunol 2004;
172:1455-62.
119. Norris S, Collins C, Doherty DG et al. . Resident human hepatic lymphocytes are phenotypically different
from circulating lymphocytes. J Hepatol 1998; 28:84-90.
120. Moffett-King A. Natural killer cells and pregnancy. Nat Rev Immunol 2002; 2:656-63.
121. Gorski KS, Waller EL, Bjornton-Severson J et al. . Distinct indirect pathways govern human NK-cell
activation by TLR-7 and TLR-8 agonists. Int Immunol 2006; 18:1115-26.
122. Prescott SL, Noakes P, Chow BW et al. . Presymptomatic differences in Toll-like receptor function in
infants who have allergy. J Allergy Clin Immunol 2008; 122:391,9, 399.e1-5.
123. Adib-Conquy M, Cavaillon JM. Gamma interferon and granulocyte/monocyte colony-stimulating factor
prevent endotoxin tolerance in human monocytes by promoting interleukin-1 receptor-associated kinase
expression and its association to MyD88 and not by modulating TLR4 expression. J Biol Chem 2002;
277:27927-34.
124. O'leary JG, Goodarzi M, Drayton DL, von Andrian UH. T cell- and B cell-independent adaptive immunity
mediated by natural killer cells. Nat Immunol 2006;7:507-16.
125. Farag SS, Caligiuri MA. Human natural killer cell development and biology. Blood Rev 2006; 20:123-37.
126. Anfossi N, Andre P, Guia S et al. . Human NK cell education by inhibitory receptors for MHC class I.
Immunity 2006; 25:331-42.
127. Korbel DS, Newman KC, Almeida CR, Davis DM, Riley EM. Heterogeneous human NK cell responses
to Plasmodium falciparum-infected erythrocytes. J Immunol 2005; 175:7466-73.
128. Mavilio D, Lombardo G, Benjamin J et al. . Characterization of CD56-/CD16+ natural killer (NK) cells: a
highly dysfunctional NK subset expanded in HIV-infected viremic individuals. Proc Natl Acad Sci U S A
2005; 102:2886-91.
129. Hu PF, Hultin LE, Hultin P et al. . Natural killer cell immunodeficiency in HIV disease is manifest by
profoundly decreased numbers of CD16+CD56+ cells and expansion of a population of CD16dimCD56cells with low lytic activity. J Acquir Immune Defic Syndr Hum Retrovirol 1995; 10:331-40.
130. Karre K. On the Immunobiology of Natural Killer Cell. Doctoral thesis, Karolinska Institute 1981.
131. Kumar V, McNerney ME. A new self: MHC-class-I-independent natural-killer-cell self-tolerance. Nat
Rev Immunol 2005; 5:363-74.
77
132. Aramburu J, Balboa MA, Izquierdo M, Lopez-Botet M. A novel functional cell surface dimer (Kp43)
expressed by natural killer cells and gamma/delta TCR+ T lymphocytes. II. Modulation of natural killer
cytotoxicity by anti-Kp43 monoclonal antibody. J Immunol 1991; 147:714-21.
133. Mingari MC, Ponte M, Bertone S et al. . HLA class I-specific inhibitory receptors in human T
lymphocytes: interleukin 15-induced expression of CD94/NKG2A in superantigen- or alloantigenactivated CD8+ T cells. Proc Natl Acad Sci U S A 1998; 95:1172-7.
134. Guma M, Busch LK, Salazar-Fontana LI, Bellosillo B, Morte C, Garcia P, Lopez-Botet M. The
CD94/NKG2C killer lectin-like receptor constitutes an alternative activation pathway for a subset of
CD8+ T cells. Eur J Immunol 2005; 35:2071-80.
135. Jacobs R, Hintzen G, Kemper A et al. . CD56bright cells differ in their KIR repertoire and cytotoxic
features from CD56dim NK cells. Eur J Immunol 2001; 31:3121-7.
136. Le Drean E, Vely F, Olcese L et al. . Inhibition of antigen-induced T cell response and antibody-induced
NK cell cytotoxicity by NKG2A: association of NKG2A with SHP-1 and SHP-2 protein-tyrosine
phosphatases. Eur J Immunol 1998; 28:264-76.
137. Lanier LL, Corliss B, Wu J, Phillips JH. Association of DAP12 with activating CD94/NKG2C NK cell
receptors. Immunity 1998; 8:693-701.
138. Bellon T, Heredia AB, Llano M, Minguela A, Rodriguez A, Lopez-Botet M, Aparicio P. Triggering of
effector functions on a CD8+ T cell clone upon the aggregation of an activatory CD94/kp39 heterodimer.
J Immunol 1999; 162:3996-4002.
139. Braud VM, Allan DS, O'Callaghan CA et al. . HLA-E binds to natural killer cell receptors CD94/NKG2A,
B and C. Nature 1998; 391:795-9.
140. Kaiser BK, Barahmand-Pour F, Paulsene W, Medley S, Geraghty DE, Strong RK. Interactions between
NKG2x immunoreceptors and HLA-E ligands display overlapping affinities and thermodynamics. J
Immunol 2005; 174:2878-84.
141. Wu J, Song Y, Bakker AB, Bauer S, Spies T, Lanier LL, Phillips JH. An activating immunoreceptor
complex formed by NKG2D and DAP10. Science 1999; 285:730-2.
142. Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, Spies T. Activation of NK cells and T cells by
NKG2D, a receptor for stress-inducible MICA. Science 1999; 285:727-9.
143. Raulet DH. ROLES OF THE NKG2D IMMUNORECEPTOR AND ITS LIGANDS. Nat Rev Immunol
2003; 3:781-90.
144. Zhang C, Zhang J, Niu J, Zhou Z, Zhang J, Tian Z. Interleukin-12 improves cytotoxicity of natural killer
cells via upregulated expression of NKG2D. Hum Immunol 2008; 69:490-500.
145. Cosman D, Mullberg J, Sutherland CL et al. . ULBPs, novel MHC class I-related molecules, bind to CMV
glycoprotein UL16 and stimulate NK cytotoxicity through the NKG2D receptor. Immunity 2001; 14:12333.
146. Chalupny NJ, Sutherland CL, Lawrence WA, Rein-Weston A, Cosman D. ULBP4 is a novel ligand for
human NKG2D. Biochem Biophys Res Commun 2003; 305:129-35.
147. Salih HR, Antropius H, Gieseke F, Lutz SZ, Kanz L, Rammensee HG, Steinle A. Functional expression
and release of ligands for the activating immunoreceptor NKG2D in leukemia. Blood 2003; 102:1389-96.
148. Groh V, Rhinehart R, Randolph-Habecker J, Topp MS, Riddell SR, Spies T. Costimulation of
CD8alphabeta T cells by NKG2D via engagement by MIC induced on virus-infected cells. Nat Immunol
2001; 2:255-60.
78
149. Kato N, Tanaka J, Sugita J et al. . Regulation of the expression of MHC class I-related chain A, B (MICA,
MICB) via chromatin remodeling and its impact on the susceptibility of leukemic cells to the cytotoxicity
of NKG2D-expressing cells. Leukemia 2007; 21:2103-8.
150. Andre P, Castriconi R, Espeli M et al. . Comparative analysis of human NK cell activation induced by
NKG2D and natural cytotoxicity receptors. Eur J Immunol 2004; 34:961-71.
151. Kim DK, Kabat J, Borrego F, Sanni TB, You CH, Coligan JE. Human NKG2F is expressed and can
associate with DAP12. Mol Immunol 2004; 41:53-62.
152. Rosen DB, Araki M, Hamerman JA, Chen T, Yamamura T, Lanier LL. A Structural basis for the
association of DAP12 with mouse, but not human, NKG2D. J Immunol 2004; 173:2470-8.
153. Rosen DB, Bettadapura J, Alsharifi M, Mathew PA, Warren HS, Lanier LL. Cutting edge: lectin-like
transcript-1 is a ligand for the inhibitory human NKR-P1A receptor. J Immunol 2005; 175:7796-9.
154. Rosen DB, Cao W, Avery DT, Tangye SG, Liu YJ, Houchins JP, Lanier LL. Functional consequences of
interactions between human NKR-P1A and its ligand LLT1 expressed on activated dendritic cells and B
cells. J Immunol 2008; 180:6508-17.
155. Welte S, Kuttruff S, Waldhauer I, Steinle A. Mutual activation of natural killer cells and monocytes
mediated by NKp80-AICL interaction. Nat Immunol 2006; 7:1334-42.
156. Roda-Navarro P, Arce I, Renedo M, Montgomery K, Kucherlapati R, Fernandez-Ruiz E. Human KLRF1,
a novel member of the killer cell lectin-like receptor gene family: molecular characterization, genomic
structure, physical mapping to the NK gene complex and expression analysis. Eur J Immunol 2000;
30:568-76.
157. Vitale M, Falco M, Castriconi R et al. . Identification of NKp80, a novel triggering molecule expressed by
human NK cells. Eur J Immunol 2001; 31:233-42.
158. Ferrini S, Cambiaggi A, Meazza R, Sforzini S, Marciano S, Mingari MC, Moretta L. T cell clones
expressing the natural killer cell-related p58 receptor molecule display heterogeneity in phenotypic
properties and p58 function. Eur J Immunol 1994; 24:2294-8.
159. Vivier E, Anfossi N. Inhibitory NK-cell receptors on T cells: witness of the past, actors of the future. Nat
Rev Immunol 2004; 4:190-8.
160. Gardiner CM. Killer cell immunoglobulin-like receptors on NK cells: the how, where and why. Int J
Immunogenet 2008; 35:1-8.
161. Lanier LL. NK cell receptors. Annu Rev Immunol 1998; 16:359-93.
162. Lanier LL, Corliss BC, Wu J, Leong C, Phillips JH. Immunoreceptor DAP12 bearing a tyrosine-based
activation motif is involved in activating NK cells. Nature 1998; 391:703-7.
163. Uhrberg M, Valiante NM, Shum BP et al. . Human diversity in killer cell inhibitory receptor genes.
Immunity 1997; 7:753-63.
164. Colonna M, Navarro F, Bellon T et al. . A common inhibitory receptor for major histocompatibility
complex class I molecules on human lymphoid and myelomonocytic cells. J Exp Med 1997; 186:1809-18.
165. Chapman TL, Heikeman AP, Bjorkman PJ. The inhibitory receptor LIR-1 uses a common binding
interaction to recognize class I MHC molecules and the viral homolog UL18. Immunity 1999; 11:603-13.
166. Vitale M, Castriconi R, Parolini S et al. . The leukocyte Ig-like receptor (LIR)-1 for the cytomegalovirus
UL18 protein displays a broad specificity for different HLA class I alleles: analysis of LIR-1 + NK cell
clones. Int Immunol 1999; 11:29-35.
79
167. Cosman D, Fanger N, Borges L, Kubin M, Chin W, Peterson L, Hsu ML. A novel immunoglobulin
superfamily receptor for cellular and viral MHC class I molecules. Immunity 1997; 7:273-82.
168. Cantoni C, Bottino C, Vitale M et al. . NKp44, a triggering receptor involved in tumor cell lysis by
activated human natural killer cells, is a novel member of the immunoglobulin superfamily. J Exp Med
1999; 189:787-96.
169. von Lilienfeld-Toal M, Nattermann J, Feldmann G, Sievers E, Frank S, Strehl J, Schmidt-Wolf IG.
Activated gammadelta T cells express the natural cytotoxicity receptor natural killer p 44 and show
cytotoxic activity against myeloma cells. Clin Exp Immunol 2006; 144:528-33.
170. Arnon TI, Lev M, Katz G, Chernobrov Y, Porgador A, Mandelboim O. Recognition of viral
hemagglutinins by NKp44 but not by NKp30. Eur J Immunol 2001; 31:2680-9.
171. Mandelboim O, Lieberman N, Lev M et al. . Recognition of haemagglutinins on virus-infected cells by
NKp46 activates lysis by human NK cells. Nature 2001; 409:1055-60.
172. Ward J, Bonaparte M, Sacks J, Guterman J, Fogli M, Mavilio D, Barker E. HIV modulates the expression
of ligands important in triggering natural killer cell cytotoxic responses on infected primary T-cell blasts.
Blood 2007; 110:1207-14.
173. Chisholm SE, Howard K, Gomez MV, Reyburn HT. Expression of ICP0 is sufficient to trigger natural
killer cell recognition of herpes simplex virus-infected cells by natural cytotoxicity receptors. J Infect Dis
2007; 195:1160-8.
174. Arnon TI, Achdout H, Levi O et al. . Inhibition of the NKp30 activating receptor by pp65 of human
cytomegalovirus. Nat Immunol 2005; 6:515-23.
175. Ferlazzo G, Tsang ML, Moretta L, Melioli G, Steinman RM, Munz C. Human dendritic cells activate
resting natural killer (NK) cells and are recognized via the NKp30 receptor by activated NK cells. J Exp
Med 2002; 195:343-51.
176. Vitale M, Della Chiesa M, Carlomagno S, Pende D, Arico M, Moretta L, Moretta A. NK-dependent DC
maturation is mediated by TNFalpha and IFNgamma released upon engagement of the NKp30 triggering
receptor. Blood 2005; 106:566-71.
177. Lanier LL. Up on the tightrope: natural killer cell activation and inhibition. Nat Immunol 2008; 9:495502.
178. Rajagopalan S, Fu J, Long EO. Cutting edge: induction of IFN-gamma production but not cytotoxicity by
the killer cell Ig-like receptor KIR2DL4 (CD158d) in resting NK cells. J Immunol 2001; 167:1877-81.
179. Meyaard L. The inhibitory collagen receptor LAIR-1 (CD305). J Leukoc Biol 2008; 83:799-803.
180. Chlewicki LK, Velikovsky CA, Balakrishnan V, Mariuzza RA, Kumar V. Molecular basis of the dual
functions of 2B4 (CD244). J Immunol 2008; 180:8159-67.
181. Moretta A, Bottino C, Vitale M et al. . Activating receptors and coreceptors involved in human natural
killer cell-mediated cytolysis. Annu Rev Immunol 2001; 19:197-223.
182. Campbell KS, Yusa S, Kikuchi-Maki A, Catina TL. NKp44 triggers NK cell activation through DAP12
association that is not influenced by a putative cytoplasmic inhibitory sequence. J Immunol 2004;
172:899-906.
183. Trinchieri G, Valiante N. Receptors for the Fc fragment of IgG on natural killer cells. Nat Immun 1993;
12:218-34.
184. Anegon I, Cuturi MC, Trinchieri G, Perussia B. Interaction of Fc receptor (CD16) ligands induces
transcription of interleukin 2 receptor (CD25) and lymphokine genes and expression of their products in
human natural killer cells. J Exp Med 1988; 167:452-72.
80
185. Jewett A, Cacalano NA, Head C, Teruel A. Coengagement of CD16 and CD94 receptors mediates
secretion of chemokines and induces apoptotic death of naive natural killer cells. Clin Cancer Res 2006;
12:1994-2003.
186. Keskin DB, Allan DS, Rybalov B et al. . TGFbeta promotes conversion of CD16+ peripheral blood NK
cells into CD16- NK cells with similarities to decidual NK cells. Proc Natl Acad Sci U S A 2007;
104:3378-83.
187. Loza MJ, Perussia B. The IL-12 signature: NK cell terminal CD56+high stage and effector functions. J
Immunol 2004; 172:88-96.
188. Wu J, Edberg JC, Redecha PB et al. . A novel polymorphism of FcgammaRIIIa (CD16) alters receptor
function and predisposes to autoimmune disease. J Clin Invest 1997; 100:1059-70.
189. Arase N, Arase H, Hirano S, Yokosuka T, Sakurai D, Saito T. IgE-mediated activation of NK cells
through Fc gamma RIII. J Immunol 2003; 170:3054-8.
190. Berahovich RD, Lai NL, Wei Z, Lanier LL, Schall TJ. Evidence for NK cell subsets based on chemokine
receptor expression. J Immunol 2006; 177:7833-40.
191. Frey M, Packianathan NB, Fehniger TA et al. . Differential expression and function of L-selectin on
CD56bright and CD56dim natural killer cell subsets. J Immunol 1998; 161:400-8.
192. Colucci F, Caligiuri MA, Di Santo JP. What does it take to make a natural killer? Nat Rev Immunol 2003;
3:413-25.
193. Robertson MJ. Role of chemokines in the biology of natural killer cells. J Leukoc Biol 2002; 71:173-83.
194. Jaleco AC, Blom B, Res P, Weijer K, Lanier LL, Phillips JH, Spits H. Fetal liver contains committed NK
progenitors, but is not a site for development of CD34+ cells into T cells. J Immunol 1997; 159:694-702.
195. Ikawa T, Kawamoto H, Fujimoto S, Katsura Y. Commitment of common T/Natural killer (NK)
progenitors to unipotent T and NK progenitors in the murine fetal thymus revealed by a single progenitor
assay. J Exp Med 1999; 190:1617-26.
196. Seaman WE, Gindhart TD, Greenspan JS, Blackman MA, Talal N. Natural killer cells, bone, and the bone
marrow: studies in estrogen-treated mice and in congenitally osteopetrotic (mi/mi) mice. J Immunol 1979;
122:2541-7.
197. Vosshenrich CA, Samson-Villeger SI, Di Santo JP. Distinguishing features of developing natural killer
cells. Curr Opin Immunol 2005; 17:151-8.
198. Freud AG, Becknell B, Roychowdhury S et al. . A human CD34(+) subset resides in lymph nodes and
differentiates into CD56bright natural killer cells. Immunity 2005; 22:295-304.
199. Freud AG, Yokohama A, Becknell B, Lee MT, Mao HC, Ferketich AK, Caligiuri MA. Evidence for
discrete stages of human natural killer cell differentiation in vivo. J Exp Med 2006; 203:1033-43.
200. Huntington ND, Vosshenrich CA, Di Santo JP. Developmental pathways that generate natural-killer-cell
diversity in mice and humans. Nat Rev Immunol 2007; 7:703-14.
201. Romagnani C, Juelke K, Falco M et al. . CD56brightCD16- killer Ig-like receptor- NK cells display
longer telomeres and acquire features of CD56dim NK cells upon activation. J Immunol 2007; 178:494755.
202. Chan A, Hong DL, Atzberger A et al. . CD56bright human NK cells differentiate into CD56dim cells: role
of contact with peripheral fibroblasts. J Immunol 2007; 179:89-94.
81
203. Dalbeth N, Gundle R, Davies RJ, Lee YC, McMichael AJ, Callan MF. CD56bright NK cells are enriched
at inflammatory sites and can engage with monocytes in a reciprocal program of activation. J Immunol
2004; 173:6418-26.
204. Schierloh P, Yokobori N, Aleman M et al. . Increased susceptibility to apoptosis of CD56dimCD16+ NK
cells induces the enrichment of IFN-gamma-producing CD56bright cells in tuberculous pleurisy. J
Immunol 2005; 175:6852-60.
205. Katchar K, Soderstrom K, Wahlstrom J, Eklund A, Grunewald J. Characterisation of natural killer cells
and CD56+ T-cells in sarcoidosis patients. Eur Respir J 2005; 26:77-85.
206. Fehniger TA, Cooper MA, Nuovo GJ, Cella M, Facchetti F, Colonna M, Caligiuri MA. CD56bright
natural killer cells are present in human lymph nodes and are activated by T cell-derived IL-2: a potential
new link between adaptive and innate immunity. Blood 2003; 101:3052-7.
207. Ranson T, Vosshenrich CA, Corcuff E, Richard O, Muller W, Di Santo JP. IL-15 is an essential mediator
of peripheral NK-cell homeostasis. Blood 2003; 101:4887-93.
208. Meller B, Frohn C, Brand JM et al. . Monitoring of a new approach of immunotherapy with allogenic
(111)In-labelled NK cells in patients with renal cell carcinoma. Eur J Nucl Med Mol Imaging 2004;
31:403-7.
209. Zhang Y, Wallace DL, de Lara CM et al. . In vivo kinetics of human natural killer cells: the effects of
ageing and acute and chronic viral infection. Immunology 2007; 121:258-65.
210. Miller JS, McCullar V. Human natural killer cells with polyclonal lectin and immunoglobulinlike
receptors develop from single hematopoietic stem cells with preferential expression of NKG2A and
KIR2DL2/L3/S2. Blood 2001; 98:705-13.
211. Stevenaert F, Van Beneden K, De Creus A, Debacker V, Plum J, Leclercq G. Ly49E expression points
toward overlapping, but distinct, natural killer (NK) cell differentiation kinetics and potential of fetal
versus adult lymphoid progenitors. J Leukoc Biol 2003; 73:731-8.
212. Parham P. Taking license with natural killer cell maturation and repertoire development. Immunol Rev
2006; 214:155-60.
213. Smith HR, Chuang HH, Wang LL, Salcedo M, Heusel JW, Yokoyama WM. Nonstochastic coexpression
of activation receptors on murine natural killer cells. J Exp Med 2000; 191:1341-54.
214. Cooley S, Xiao F, Pitt M et al. . A subpopulation of human peripheral blood NK cells that lacks inhibitory
receptors for self MHC is developmentally immature. Blood 2007; 110:578-86.
215. Yokoyama WM, Kim S. How do natural killer cells find self to achieve tolerance? Immunity 2006;
24:249-57.
216. Raulet DH. Missing self recognition and self tolerance of natural killer (NK) cells. Semin Immunol 2006;
18:145-50.
217. Lanier LL. NK cell recognition. Annu Rev Immunol 2005; 23:225-74.
218. Raulet DH. Interplay of natural killer cells and their receptors with the adaptive immune response. Nat
Immunol 2004; 5:996-1002.
219. Maasho K, Opoku-Anane J, Marusina AI, Coligan JE, Borrego F. NKG2D is a costimulatory receptor for
human naive CD8+ T cells. J Immunol 2005; 174:4480-4.
220. Mingari MC, Ponte M, Cantoni C et al. . HLA-class I-specific inhibitory receptors in human cytolytic T
lymphocytes: molecular characterization, distribution in lymphoid tissues and co-expression by individual
T cells. Int Immunol 1997; 9:485-91.
82
221. Anfossi N, Pascal V, Vivier E, Ugolini S. Biology of T memory type 1 cells. Immunol Rev 2001;
181:269-78.
222. Chwae YJ, Chang MJ, Park SM, Yoon H, Park HJ, Kim SJ, Kim J. Molecular mechanism of the
activation-induced cell death inhibition mediated by a p70 inhibitory killer cell Ig-like receptor in Jurkat T
cells. J Immunol 2002; 169:3726-35.
223. Young NT, Uhrberg M, Phillips JH, Lanier LL, Parham P. Differential expression of leukocyte receptor
complex-encoded Ig-like receptors correlates with the transition from effector to memory CTL. J
Immunol 2001; 166:3933-41.
224. Mingari MC, Schiavetti F, Ponte M et al. . Human CD8+ T lymphocyte subsets that express HLA class Ispecific inhibitory receptors represent oligoclonally or monoclonally expanded cell populations. Proc Natl
Acad Sci U S A 1996; 93:12433-8.
225. D'Andrea A, Chang C, Phillips JH, Lanier LL. Regulation of T cell lymphokine production by killer cell
inhibitory receptor recognition of self HLA class I alleles. J Exp Med 1996; 184:789-94.
226. Saverino D, Fabbi M, Ghiotto F et al. . The CD85/LIR-1/ILT2 inhibitory receptor is expressed by all
human T lymphocytes and down-regulates their functions. J Immunol 2000; 165:3742-55.
227. Guerra N, Guillard M, Angevin E et al. . Killer inhibitory receptor (CD158b) modulates the lytic activity
of tumor-specific T lymphocytes infiltrating renal cell carcinomas. Blood 2000; 95:2883-9.
228. Saverino D, Ghiotto F, Merlo A et al. . Specific recognition of the viral protein UL18 by CD85j/LIR1/ILT2 on CD8+ T cells mediates the non-MHC-restricted lysis of human cytomegalovirus-infected cells.
J Immunol 2004; 172:5629-37.
229. Zhang AL, Colmenero P, Purath U et al. . Natural killer cells trigger differentiation of monocytes into
dendritic cells. Blood 2007; 110:2484-93.
230. Vitale M, Della Chiesa M, Carlomagno S, Romagnani C, Thiel A, Moretta L, Moretta A. The small subset
of CD56brightCD16- natural killer cells is selectively responsible for both cell proliferation and
interferon-gamma production upon interaction with dendritic cells. Eur J Immunol 2004; 34:1715-22.
231. Marcenaro E, Della Chiesa M, Bellora F, Parolini S, Millo R, Moretta L, Moretta A. IL-12 or IL-4 prime
human NK cells to mediate functionally divergent interactions with dendritic cells or tumors. J Immunol
2005; 174:3992-8.
232. Martin-Fontecha A, Thomsen LL, Brett S, Gerard C, Lipp M, Lanzavecchia A, Sallusto F. Induced
recruitment of NK cells to lymph nodes provides IFN-[gamma] for TH1 priming. Nat Immunol 2004;
5:1260-5.
233. Satoskar AR, Stamm LM, Zhang X, Okano M, David JR, Terhorst C, Wang B. NK cell-deficient mice
develop a Th1-like response but fail to mount an efficient antigen-specific IgG2a antibody response. J
Immunol 1999; 163:5298-302.
234. Blanca IR, Bere EW, Young HA, Ortaldo JR. Human B cell activation by autologous NK cells is
regulated by CD40-CD40 ligand interaction: role of memory B cells and CD5+ B cells. J Immunol 2001;
167:6132-9.
235. Newman KC, Riley EM. Whatever turns you on: accessory-cell-dependent activation of NK cells by
pathogens. Nat Rev Immunol 2007; 7:279-91.
236. Borg C, Jalil A, Laderach D et al. . NK cell activation by dendritic cells (DCs) requires the formation of a
synapse leading to IL-12 polarization in DCs. Blood 2004; 104:3267-75.
237. Gerosa F, Baldani-Guerra B, Nisii C, Marchesini V, Carra G, Trinchieri G. Reciprocal activating
interaction between natural killer cells and dendritic cells. J Exp Med 2002; 195:327-33.
83
238. Nedvetzki S, Sowinski S, Eagle RA et al. . Reciprocal regulation of human natural killer cells and
macrophages associated with distinct immune synapses. Blood 2007; 109:3776-85.
239. Newman KC, Korbel DS, Hafalla JC, Riley EM. Cross-Talk with Myeloid Accessory Cells Regulates
Human Natural Killer Cell Interferon-gamma Responses to Malaria. PLoS Pathog 2006; 2:e118.
240. Scordamaglia F, Balsamo M, Scordamaglia A et al. . Perturbations of natural killer cell regulatory
functions in respiratory allergic diseases. J Allergy Clin Immunol 2008; 121:479-85.
241. Landolfo S, Gariglio M, Gribaudo G, Lembo D. The human cytomegalovirus. Pharmacol Ther 2003;
98:269-97.
242. Hislop AD, Taylor GS, Sauce D, Rickinson AB. Cellular responses to viral infection in humans: lessons
from Epstein-Barr virus. Annu Rev Immunol 2007; 25:587-617.
243. Sylwester AW, Mitchell BL, Edgar JB et al. . Broadly targeted human cytomegalovirus-specific CD4+
and CD8+ T cells dominate the memory compartments of exposed subjects. J Exp Med 2005; 202:673-85.
244. Strowig T, Brilot F, Arrey F, Bougras G, Thomas D, Muller WA, Munz C. Tonsilar NK cells restrict B
cell transformation by the Epstein-Barr virus via IFN-gamma. PLoS Pathog 2008; 4:e27.
245. Pappworth IY, Wang EC, Rowe M. The switch from latent to productive infection in epstein-barr virusinfected B cells is associated with sensitization to NK cell killing. J Virol 2007; 81:474-82.
246. Rajagopalan S, Long EO. Viral evasion of NK-cell activation. Trends Immunol 2005; 26:403-5.
247. Johansson SG, Bieber T, Dahl R et al. . Revised nomenclature for allergy for global use: Report of the
Nomenclature Review Committee of the World Allergy Organization, October 2003. J Allergy Clin
Immunol 2004; 113:832-6.
248. Tada T, Takemori T, Okumura K, Nonaka M, Tokuhisa T. Two distinct types of helper T cells involved in
the secondary antibody response: independent and synergistic effects of Ia- and Ia+ helper T cells. J Exp
Med 1978; 147:446-58.
249. Mosmann TR, Cherwinski H, Bond MW, Giedlin MA, Coffman RL. Two types of murine helper T cell
clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J Immunol 1986;
136:2348-57.
250. Gabrielsson S, Soderlund A, Nilsson C, Lilja G, Nordlund M, Troye-Blomberg M. Influence of atopic
heredity on IL-4-, IL-12- and IFN-gamma-producing cells in in vitro activated cord blood mononuclear
cells. Clin Exp Immunol 2001; 126:390-6.
251. Nilsson C, Larsson AK, Hoglind A, Gabrielsson S, Troye BM, Lilja G. Low numbers of interleukin-12producing cord blood mononuclear cells and immunoglobulin E sensitization in early childhood. Clin Exp
Allergy 2004; 34:373-80.
252. Kimura M, Yamaide A, Tsuruta S, Okafuji I, Yoshida T. Development of the capacity of peripheral blood
mononuclear cells to produce IL-4, IL-5 and IFN-gamma upon stimulation with house dust mite in
children with atopic dermatitis. Int Arch Allergy Immunol 2002; 127:191-7.
253. Larsson AK, Nilsson C, Hoglind A, Sverremark-Ekstrom E, Lilja G, Troye-Blomberg M. Relationship
between maternal and child cytokine responses to allergen and phytohaemagglutinin 2 years after
delivery. Clin Exp Immunol 2006; 144:401-8.
254. Kapsenberg ML, Hilkens CM, Wierenga EA, Kalinski P. The paradigm of type 1 and type 2 antigenpresenting cells. Implications for atopic allergy. Clin Exp Allergy 1999; 29 Suppl 2:33-6.
255. Loza MJ, Perussia B. Final steps of natural killer cell maturation: a model for type 1-type 2
differentiation? Nat Immunol 2001; 2:917-24.
84
256. Dold S, Wjst M, von Mutius E, Reitmeir P, Stiepel E. Genetic risk for asthma, allergic rhinitis, and atopic
dermatitis. Arch Dis Child 1992; 67:1018-22.
257. Strachan DP. Hay fever, hygiene, and household size. BMJ 1989; 299:1259-60.
258. von Mutius E, Martinez FD, Fritzsch C, Nicolai T, Reitmeir P, Thiemann HH. Skin test reactivity and
number of siblings. BMJ 1994; 308:692-5.
259. Celedon JC, Wright RJ, Litonjua AA, Sredl D, Ryan L, Weiss ST, Gold DR. Day care attendance in early
life, maternal history of asthma, and asthma at the age of 6 years. Am J Respir Crit Care Med 2003;
167:1239-43.
260. Hoffjan S, Nicolae D, Ostrovnaya I et al. . Gene-environment interaction effects on the development of
immune responses in the 1st year of life. Am J Hum Genet 2005; 76:696-704.
261. von Mutius E, Braun-Fahrlander C, Schierl R et al. . Exposure to endotoxin or other bacterial components
might protect against the development of atopy. Clin Exp Allergy 2000; 30:1230-4.
262. Gehring U, Bolte G, Borte M et al. . Exposure to endotoxin decreases the risk of atopic eczema in infancy:
a cohort study. J Allergy Clin Immunol 2001; 108:847-54.
263. Braun-Fahrlander C, Riedler J, Herz U et al. . Environmental exposure to endotoxin and its relation to
asthma in school-age children. N Engl J Med 2002; 347:869-77.
264. van Strien RT, Engel R, Holst O et al. . Microbial exposure of rural school children, as assessed by levels
of N-acetyl-muramic acid in mattress dust, and its association with respiratory health. J Allergy Clin
Immunol 2004; 113:860-7.
265. Roy SR, Schiltz AM, Marotta A, Shen Y, Liu AH. Bacterial DNA in house and farm barn dust. J Allergy
Clin Immunol 2003; 112:571-8.
266. Braun-Fahrlander C, Gassner M, Grize L et al. . Prevalence of hay fever and allergic sensitization in
farmer's children and their peers living in the same rural community. SCARPOL team. Swiss Study on
Childhood Allergy and Respiratory Symptoms with Respect to Air Pollution. Clin Exp Allergy 1999;
29:28-34.
267. Kilpelainen M, Terho EO, Helenius H, Koskenvuo M. Farm environment in childhood prevents the
development of allergies. Clin Exp Allergy 2000; 30:201-8.
268. Riedler J, Eder W, Oberfeld G, Schreuer M. Austrian children living on a farm have less hay fever,
asthma and allergic sensitization. Clin Exp Allergy 2000; 30:194-200.
269. Von Ehrenstein OS, Von Mutius E, Illi S, Baumann L, Bohm O, von Kries R. Reduced risk of hay fever
and asthma among children of farmers. Clin Exp Allergy 2000; 30:187-93.
270. Downs SH, Marks GB, Mitakakis TZ, Leuppi JD, Car NG, Peat JK. Having lived on a farm and
protection against allergic diseases in Australia. Clin Exp Allergy 2001; 31:570-5.
271. Nilsson C, Linde A, Montgomery SM, Gustafsson L, Nasman P, Blomberg MT, Lilja G. Does early EBV
infection protect against IgE sensitization? J Allergy Clin Immunol 2005; 116:438-44.
272. Kamradt T, Goggel R, Erb KJ. Induction, exacerbation and inhibition of allergic and autoimmune diseases
by infection. Trends Immunol 2005; 26:260-7.
273. Sidorchuk A, Wickman M, Pershagen G, Lagarde F, Linde A. Cytomegalovirus infection and
development of allergic diseases in early childhood: interaction with EBV infection? J Allergy Clin
Immunol 2004; 114:1434-40.
274. Yazdanbakhsh M, Kremsner PG, van Ree R. Allergy, parasites, and the hygiene hypothesis. Science
2002; 296:490-4.
85
275. Merino F, Henle W, Ramirez-Duque P. Chronic active Epstein-Barr virus infection in patients with
Chediak-Higashi syndrome. J Clin Immunol 1986; 6:299-305.
276. Sullivan JL, Byron KS, Brewster FE, Purtilo DT. Deficient natural killer cell activity in x-linked
lymphoproliferative syndrome. Science 1980; 210:543-5.
277. Plebani A, Ciravegna B, Ponte M, Mingari MC, Moretta L. Interleukin-2 mediated restoration of natural
killer cell function in a patient with Griscelli syndrome. Eur J Pediatr 2000; 159:713-4.
278. Kogawa K, Lee SM, Villanueva J, Marmer D, Sumegi J, Filipovich AH. Perforin expression in cytotoxic
lymphocytes from patients with hemophagocytic lymphohistiocytosis and their family members. Blood
2002; 99:61-6.
279. Biron CA, Byron KS, Sullivan JL. Severe herpesvirus infections in an adolescent without natural killer
cells. N Engl J Med 1989; 320:1731-5.
280. Wendland T, Herren S, Yawalkar N, Cerny A, Pichler WJ. Strong alpha beta and gamma delta TCR
response in a patient with disseminated Mycobacterium avium infection and lack of NK cells and
monocytopenia. Immunol Lett 2000; 72:75-82.
281. Kuijpers TW, Baars PA, Dantin C, van den Burg M, van Lier RA, Roosnek E. Human NK cells can
control CMV infection in the absence of T cells. Blood 2008; 112:914-5.
282. Wei H, Zhang J, Xiao W, Feng J, Sun R, Tian Z. Involvement of human natural killer cells in asthma
pathogenesis: natural killer 2 cells in type 2 cytokine predominance. J Allergy Clin Immunol 2005;
115:841-7.
283. Katsuta M, Takigawa Y, Kimishima M, Inaoka M, Takahashi R, Shiohara T. NK cells and gamma delta+
T cells are phenotypically and functionally defective due to preferential apoptosis in patients with atopic
dermatitis. J Immunol 2006; 176:7736-44.
284. Buentke E, Heffler LC, Wilson JL et al. . Natural killer and dendritic cell contact in lesional atopic
dermatitis skin--Malassezia-influenced cell interaction. J Invest Dermatol 2002; 119:850-7.
285. Matsui E, Kaneko H, Teramoto T et al. . Reduced IFNgamma production in response to IL-12 stimulation
and/or reduced IL-12 production in atopic patients. Clin Exp Allergy 2000; 30:1250-6.
286. Plewako H, Wosinska K, Arvidsson M, Bjorkander J, Hakansson L, Rak S. Production of interleukin-12
by monocytes and interferon-gamma by natural killer cells in allergic patients during rush
immunotherapy. Ann Allergy Asthma Immunol 2006; 97:464-8.
287. Deniz G, Akdis M, Aktas E, Blaser K, Akdis CA. Human NK1 and NK2 subsets determined by
purification of IFN-gamma-secreting and IFN-gamma-nonsecreting NK cells. Eur J Immunol 2002;
32:879-84.
288. Loza MJ, Peters SP, Zangrilli JG, Perussia B. Distinction between IL-13+ and IFN-gamma+ natural killer
cells and regulation of their pool size by IL-4. Eur J Immunol 2002; 32:413-23.
289. Loza MJ, Perussia B. Differential regulation of NK cell proliferation by type I and type II IFN. Int
Immunol 2004; 16:23-32.
290. Gasparoni A, Ciardelli L, Avanzini A, Castellazzi AM, Carini R, Rondini G, Chirico G. Age-related
changes in intracellular TH1/TH2 cytokine production, immunoproliferative T lymphocyte response and
natural killer cell activity in newborns, children and adults. Biol Neonate 2003; 84:297-303.
291. Pirruccello SJ, Collins M, Wilson JE, McManus BM. Age-related changes in naive and memory CD4+ T
cells in healthy human children. Clin Immunol Immunopathol 1989; 52:341-5.
292. Osugi Y, Hara J, Kurahashi H et al. . Age-related changes in surface antigens on peripheral lymphocytes
of healthy children. Clin Exp Immunol 1995; 100:543-8.
86
293. Saule P, Trauet J, Dutriez V, Lekeux V, Dessaint JP, Labalette M. Accumulation of memory T cells from
childhood to old age: central and effector memory cells in CD4(+) versus effector memory and terminally
differentiated memory cells in CD8(+) compartment. Mech Ageing Dev 2006; 127:274-81.
294. Hunt DW, Huppertz HI, Jiang HJ, Petty RE. Studies of human cord blood dendritic cells: evidence for
functional immaturity. Blood 1994; 84:4333-43.
295. Hannet I, Erkeller-Yuksel F, Lydyard P, Deneys V, DeBruyere M. Developmental and maturational
changes in human blood lymphocyte subpopulations. Immunol Today 1992; 13:215, 218.
296. Yabuhara A, Kawai H, Komiyama A. Development of natural killer cytotoxicity during childhood:
marked increases in number of natural killer cells with adequate cytotoxic abilities during infancy to early
childhood. Pediatr Res 1990; 28:316-22.
297. Rizos D, Protonotariou E, Malamitsi-Puchner A, Sarandakou A, Trakakis E, Salamalekis E. Cytokine
concentrations during the first days of life. Eur J Obstet Gynecol Reprod Biol 2006; 131:32-5.
298. Nomura A, Takada H, Jin CH, Tanaka T, Ohga S, Hara T. Functional analyses of cord blood natural killer
cells and T cells: a distinctive interleukin-18 response. Exp Hematol 2001; 29:1169-76.
299. De Wit D, Tonon S, Olislagers V, Goriely S, Boutriaux M, Goldman M, Willems F. Impaired responses
to toll-like receptor 4 and toll-like receptor 3 ligands in human cord blood. J Autoimmun 2003; 21:27781.
300. Samelson R, Larkey DM, Amankwah KS, McConnachie P. Effect of labor on lymphocyte subsets in fullterm neonates. Am J Reprod Immunol 1992; 28:71-3.
301. Brown MA, Rad PY, Halonen MJ. Method of birth alters interferon-gamma and interleukin-12 production
by cord blood mononuclear cells. Pediatr Allergy Immunol 2003; 14:106-11.
302. Walker JJ. Pre-eclampsia. Lancet 2000; 356:1260-5.
303. Bachmayer N, Rafik Hamad R, Liszka L, Bremme K, Sverremark-Ekstrom E. Aberrant uterine natural
killer (NK)-cell expression and altered placental and serum levels of the NK-cell promoting cytokine
interleukin-12 in pre-eclampsia. Am J Reprod Immunol 2006; 56:292-301.
304. Sargent IL, Borzychowski AM, Redman CW. NK cells and human pregnancy--an inflammatory view.
Trends Immunol 2006; 27:399-404.
305. Hiby SE, Walker JJ, O'shaughnessy KM, Redman CW, Carrington M, Trowsdale J, Moffett A.
Combinations of maternal KIR and fetal HLA-C genes influence the risk of preeclampsia and
reproductive success. J Exp Med 2004; 200:957-65.
306. Darmochwal-Kolarz D, Leszczynska-Gorzelak B, Rolinski J, Oleszczuk J. Pre-eclampsia affects the
immunophenotype of neonates. Immunol Lett 2001; 77:67-71.
307. Bujold E, Chaiworapongsa T, Romero R et al. . Neonates born to pre-eclamptic mothers have a higher
percentage of natural killer cells (CD3-/CD56+16+) in umbilical cord blood than those without preeclampsia. J Matern Fetal Neonatal Med 2003; 14:305-12.
308. Nilsson C. Cytokine profiles, infections and IgE sensitisation in childhood. Doctoral thesis, Karolinska
Institute 2006.
309. Patni S, Flynn P, Wynen LP, Seager AL, Morgan G, White JO, Thornton CA. An introduction to Toll-like
receptors and their possible role in the initiation of labour. BJOG 2007; 114:1326-34.
310. Barcelos W, Sathler-Avelar R, Martins-Filho OA et al. . Natural killer cell subpopulations in putative
resistant individuals and patients with active Mycobacterium tuberculosis infection. Scand J Immunol
2008; 68:92-102.
87
311. Schepis D, Gunnarsson I, Eloranta ML, Lampa J, Jacobson SH, Karre K, Berg L. Increased proportion of
CD56(bright) natural killer cells in active and inactive systemic lupus erythematosus. Immunology 2008
[Epub ahead of print].
312. Krampera M, Tavecchia L, Benedetti F, Nadali G, Pizzolo G. Intracellular cytokine profile of cord blood
T-, and NK- cells and monocytes. Haematologica 2000; 85:675-9.
313. Wagner CS, Riise GC, Bergstrom T, Karre K, Carbone E, Berg L. Increased Expression of Leukocyte IgLike Receptor-1 and Activating Role of UL18 in the Response to Cytomegalovirus Infection. J Immunol
2007; 178:3536-43.
314. Sivakumar PV, Gunturi A, Salcedo M et al. . Cutting edge: expression of functional CD94/NKG2A
inhibitory receptors on fetal NK1.1+Ly-49- cells: a possible mechanism of tolerance during NK cell
development. J Immunol 1999; 162:6976-80.
315. Van Beneden K, Stevenaert F, De Creus A, Debacker V, De Boever J, Plum J, Leclercq G. Expression of
Ly49E and CD94/NKG2 on fetal and adult NK cells. J Immunol 2001; 166:4302-11.
316. Zhang C, Zhang J, Sun R, Feng J, Wei H, Tian Z. Opposing effect of IFNgamma and IFNalpha on
expression of NKG2 receptors: negative regulation of IFNgamma on NK cells. Int Immunopharmacol
2005; 5:1057-67.
317. Mavoungou E, Bouyou-Akotet MK, Kremsner PG. Effects of prolactin and cortisol on natural killer (NK)
cell surface expression and function of human natural cytotoxicity receptors (NKp46, NKp44 and
NKp30). Clin Exp Immunol 2005; 139:287-96.
318. Zwirner NW, Fuertes MB, Girart MV, Domaica CI, Rossi LE. Cytokine-driven regulation of NK cell
functions in tumor immunity: role of the MICA-NKG2D system. Cytokine Growth Factor Rev 2007;
18:159-70.
319. Li Z, Lim WK, Mahesh SP, Liu B, Nussenblatt RB. Cutting edge: in vivo blockade of human IL-2
receptor induces expansion of CD56(bright) regulatory NK cells in patients with active uveitis. J Immunol
2005; 174:5187-91.
320. Bielekova B, Catalfamo M, Reichert-Scrivner S et al. . Regulatory CD56(bright) natural killer cells
mediate immunomodulatory effects of IL-2Ralpha-targeted therapy (daclizumab) in multiple sclerosis.
Proc Natl Acad Sci U S A 2006; 103:5941-6.
321. Saraste M, Irjala H, Airas L. Expansion of CD56Bright natural killer cells in the peripheral blood of
multiple sclerosis patients treated with interferon-beta. Neurol Sci 2007; 28:121-6.
322. Titanji K, Sammicheli S, De Milito A et al. . Altered distribution of natural killer cell subsets identified by
CD56, CD27 and CD70 in primary and chronic human immunodeficiency virus-1 infection. Immunology
2008; 123:164-70.
323. Agaugue S, Marcenaro E, Ferranti B, Moretta L, Moretta A. Human natural killer cells exposed to IL-2,
IL-12, IL-18, or IL-4 differently modulate priming of naive T cells by monocyte-derived dendritic cells.
Blood 2008; 112:1776-83.
324. Fitzgerald-Bocarsly P, Feng D. The role of type I interferon production by dendritic cells in host defense.
Biochimie 2007; 89:843-55.
325. Barton ES, White DW, Cathelyn JS et al. . Herpesvirus latency confers symbiotic protection from
bacterial infection. Nature 2007; 447:326-9.
326. Kennedy MK, Glaccum M, Brown SN et al. . Reversible defects in natural killer and memory CD8 T cell
lineages in interleukin 15-deficient mice. J Exp Med 2000; 191:771-80.
88
327. Saghafian-Hedengren S, Holmlund U, Amoudruz P, Nilsson C, Sverremark-Ekstrom E. Maternal allergy
influences p38-mitogen-activated protein kinase activity upon microbial challenge in CD14+ monocytes
from 2-year-old children. Clin Exp Allergy 2008; 38:449-57.
328. Orange JS, Fassett MS, Koopman LA, Boyson JE, Strominger JL. Viral evasion of natural killer cells. Nat
Immunol 2002; 3:1006-12.
329. Randolph GJ, Sanchez-Schmitz G, Liebman RM, Schakel K. The CD16(+) (FcgammaRIII(+)) subset of
human monocytes preferentially becomes migratory dendritic cells in a model tissue setting. J Exp Med
2002; 196:517-27.
330. Moutaftsi M, Mehl AM, Borysiewicz LK, Tabi Z. Human cytomegalovirus inhibits maturation and
impairs function of monocyte-derived dendritic cells. Blood 2002; 99:2913-21.
331. Salio M, Cella M, Suter M, Lanzavecchia A. Inhibition of dendritic cell maturation by herpes simplex
virus. Eur J Immunol 1999; 29:3245-53.
332. Moore KW, de Waal Malefyt R, Coffman RL, O'Garra A. Interleukin-10 and the interleukin-10 receptor.
Annu Rev Immunol 2001; 19:683-765.
333. Romagnani S. Immunologic influences on allergy and the TH1/TH2 balance. J Allergy Clin Immunol
2004; 113:395-400.
334. Jonsson Y, Ruber M, Matthiesen L et al. . Cytokine mapping of sera from women with preeclampsia and
normal pregnancies. J Reprod Immunol 2006; 70:83-91.
335. Dudley DJ, Hunter C, Mitchell MD, Varner MW, Gately M. Elevations of serum interleukin-12
concentrations in women with severe pre-eclampsia and HELLP syndrome. J Reprod Immunol 1996;
31:97-107.
336. Saito S, Shiozaki A, Nakashima A, Sakai M, Sasaki Y. The role of the immune system in preeclampsia.
Mol Aspects Med 2007; 28:192-209.
337. Sharma A, Satyam A, Sharma JB. Leptin, IL-10 and inflammatory markers (TNF-alpha, IL-6 and IL-8) in
pre-eclamptic, normotensive pregnant and healthy non-pregnant women. Am J Reprod Immunol 2007;
58:21-30.
338. Perez A, Gurbindo MD, Resino S, Aguaron A, Munoz-Fernandez MA. NK cell increase in neonates from
the preterm to the full-term period of gestation. Neonatology 2007; 92:158-63.
339. Wang Y, Xu H, Zheng X, Wei H, Sun R, Tian Z. High expression of NKG2A/CD94 and low expression
of granzyme B are associated with reduced cord blood NK cell activity. Cell Mol Immunol 2007; 4:37782.
340. Zingoni A, Palmieri G, Morrone S et al. . CD69-triggered ERK activation and functions are negatively
regulated by CD94 / NKG2-A inhibitory receptor. Eur J Immunol 2000; 30:644-51.
341. Guma M, Angulo A, Vilches C, Gomez-Lozano N, Malats N, Lopez-Botet M. Imprint of human
cytomegalovirus infection on the NK cell receptor repertoire. Blood 2004; 104:3664-71.
342. Burgess SJ, Marusina AI, Pathmanathan I, Borrego F, Coligan JE. IL-21 down-regulates NKG2D/DAP10
expression on human NK and CD8+ T cells. J Immunol 2006; 176:1490-7.
343. Groh V, Bruhl A, El Gabalawy H, Nelson JL, Spies T. Stimulation of T cell autoreactivity by anomalous
expression of NKG2D and its MIC ligands in rheumatoid arthritis. Proc Natl Acad Sci U S A 2003;
100:9452-7.
344. Mincheva-Nilsson L, Nagaeva O, Chen T et al. . Placenta-derived soluble MHC class I chain-related
molecules down-regulate NKG2D receptor on peripheral blood mononuclear cells during human
pregnancy: a possible novel immune escape mechanism for fetal survival. J Immunol 2006; 176:3585-92.
89
345. Lee SM, Suen Y, Chang L et al. . Decreased interleukin-12 (IL-12) from activated cord versus adult
peripheral blood mononuclear cells and upregulation of interferon-gamma, natural killer, and lymphokineactivated killer activity by IL-12 in cord blood mononuclear cells. Blood 1996; 88:945-54.
346. Fan YY, Yang BY, Wu CY. Phenotypic and functional heterogeneity of natural killer cells from umbilical
cord blood mononuclear cells. Immunol Invest 2008; 37:79-96.
347. Levy O, Zarember KA, Roy RM, Cywes C, Godowski PJ, Wessels MR. Selective impairment of TLRmediated innate immunity in human newborns: neonatal blood plasma reduces monocyte TNF-alpha
induction by bacterial lipopeptides, lipopolysaccharide, and imiquimod, but preserves the response to R848. J Immunol 2004; 173:4627-34.
348. Levy O, Coughlin M, Cronstein BN, Roy RM, Desai A, Wessels MR. The adenosine system selectively
inhibits TLR-mediated TNF-alpha production in the human newborn. J Immunol 2006; 177:1956-66.
349. Levy O. Innate immunity of the newborn: basic mechanisms and clinical correlates. Nat Rev Immunol
2007; 7:379-90.
350. Rauk PN, Friebe-Hoffmann U, Winebrenner LD, Chiao JP. Interleukin-6 up-regulates the oxytocin
receptor in cultured uterine smooth muscle cells. Am J Reprod Immunol 2001; 45:148-53.
351. Yektaei-Karin E, Moshfegh A, Lundahl J, Berggren V, Hansson LO, Marchini G. The stress of birth
enhances in vitro spontaneous and IL-8-induced neutrophil chemotaxis in the human newborn. Pediatr
Allergy Immunol 2007; 18:643-51.
352. Joyner JL, Augustine NH, Taylor KA, La Pine TR, Hill HR. Effects of group B streptococci on cord and
adult mononuclear cell interleukin-12 and interferon-gamma mRNA accumulation and protein secretion. J
Infect Dis 2000; 182:974-7.
90