Download Jenny Walldén Studies of immunological risk factors in type 1 diabetes

Document related concepts

Phagocyte wikipedia , lookup

Human leukocyte antigen wikipedia , lookup

Immune system wikipedia , lookup

Lymphopoiesis wikipedia , lookup

T cell wikipedia , lookup

Hygiene hypothesis wikipedia , lookup

Adaptive immune system wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Autoimmunity wikipedia , lookup

Innate immune system wikipedia , lookup

Diabetes mellitus type 1 wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Molecular mimicry wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Sjögren syndrome wikipedia , lookup

Immunomics wikipedia , lookup

Transcript
Linköping University Medical Dissertations
No 1075
Studies of immunological risk factors
in type 1 diabetes
Jenny Walldén
Division of Pediatrics and Diabetes Research Centre
Department of Clinical and Experimental Medicin
Faculty of Health Sciences, Linköping University
SE-581 85 Linköping, Sweden
Linköping 2008
Jenny Walldén, 2008
Cover design: Jenny Walldén
ISBN: 978-91-7393-824-2
ISSN: 0345-0082
Paper I and III have been reprinted with permission from Blackwell Publishing
Ltd, Oxford, UK.
Paper II has been printed with the permission from Dove Medical Press Ltd.
During the course of the research underlying this thesis, Jenny Walldén was
enrolled in Forum Scientium, a multidisciplinary doctoral programme at
Linköping University, Sweden.
Printed in Sweden by LIU-tryck, Linköping 2008
” The greatest virtue of man is perhaps curiosity”
Anatole France
To my family,
with all my love
ABSTRACT
Background: Type 1 diabetes (T1D) is a chronic, autoimmune disease caused
by a T cell mediated destruction of -cells in pancreas. The development of T1D
is determined by a combination of genetic susceptibility genes and
environmental factors involved in the pathogenesis of T1D.
This thesis aimed to investigate diverse environmental and immunological risk
factors associated with the development of T1D. This was accomplished by
comparing autoantibody development, T cell responses and the function of
CD4+CD25+ regulatory T cells between healthy children, children at risk of T1D
and T1D patients.
Results: Induction of autoantibodies in as young children as one year old, was
associated with previously identified environmental risk factors of T1D, such as
maternal gastroenteritis during pregnancy and early introduction of cow’s milk.
We did not see any general increase in the activity of peripheral blood TH
subtypes in children with HLA class II risk haplotypes associated with T1D, nor
were HLA class II risk haplotypes associated with any aberrant cytokine
production in response to antigenic stimulation of peripheral blood mononuclear
cells. However children with a HLA class II protective haplotype showed an
increased production of IFN- in response to enteroviral stimulation.
CTLA-4 polymorphisms connected with a risk of autoimmune disease were
associated with enhanced production of IFN-.
Healthy children with -cell autoantibodies had a lower expression level of
GATA-3 compared to health children with HLA risk genotype or children
without risk. Instead, children with manifest T1D showed lower expression
levels of T-bet, IL-12R1 and IL-4R.
Both T1D and healthy children showed the same expression of the regulatory
markers Foxp3, CTLA-4 and ICOS in peripheral blood mononuclear cells, and
the amount of CD4+CD25high T cells did neither reveal any differences. The
regulatory T cells seemed also to be functional in children with T1D, since
increased proliferation after depletion of CD4+CD25high cells from PBMC was
demonstrated in T1D as well as in healthy children.
However, T1D children did have more intracellular CTLA-4 per CD4+CD25high
T cell, increased levels of serum C-reactive protein and higher spontaneous
expression of IFN- in CD25depleted PBMC, all which are signs of activation of
the immune system. This suggests a normal or enhanced functional activity of
regulatory T cells in T1D at diagnosis.
Conclusions: Our findings emphasize that environmental risk factors do have a
role in the development of -cell autoimmunity. Our results do not support a
systemic activation of the immune system in pre-diabetes or T1D, but instead a
possible up-regulation of regulatory mechanisms seems to occur after diagnosis
of T1D, which probably tries to dampen the autoimmune reaction taking place.
SAMMANFATTNING
Bakgrund: Typ 1 diabetes är en autoimmun sjukdom som orsakas av förstörelse
av de insulinproducerande cellerna i bukspottkörteln (pankreas). Det finns flera
olika faktorer som påverkar destruktionen av cellerna i pankreas och därmed
utvecklingen mot diabetes. Genetiska faktorer spelar en stor roll, men
inflytandet av olika miljöfaktorer har också en stor del i detta samspel, som leder
den slutliga sjukdomsutvecklingen. Den här avhandlingen syftar till att
undersöka den inverkan miljöfaktorer och immunologiska komponenter har på
utvecklingen mot diabetes. Detta gjordes genom att utforska T cellers
immunsvar och autoantikroppsutveckling hos friska barn, barn med ökad risk
för diabetes och barn som har en manifest sjukdom.
Resultat: Produktionen av autoantikroppar kunde detekteras redan vid ett års
ålder och relateras till olika miljöfaktorer i barnets omgivning. Till exempel
finns det associationer mellan maginfluensa hos mamman under graviditet eller
om barnet tidigt i livet får komjölksbaserad välling och utvecklingen av
antikroppar hos barnet vid ett års ålder. Dessa autoantikroppar leder dock sällan
till manifestation av diabetes.
Barn med genetisk risk för diabetes verkar inte ha någon generell obalans i det
perifera immunsystemet och inte heller någon rubbad produktion av cytokiner
efter stimulering av perifera mononucleära celler. Däremot verkar barn med en
skyddande genetisk profil ha ett ökat IFN- svar mot enterovirus. CTLA-4
polymorfier associerade med ökad risk av autoimmuna sjukdomar gav en ökad
produktion av IFN-.
En pågående autoimmun process mot de insulinproducerande cellerna i pankreas
verkar påverka nivån av transkriptionsfaktorn GATA-3, då barn med
autoantikroppar hade lägre nivåer av GATA-3. Barn med manifest diabetes hade
istället en lägre nivå av IL-12R1, IL-4R och T-bet. Friska barn och barn med
diabetes hade lika nivåer av de regulatoriska markörerna Foxp3, CTLA-4 och
ICOS i perifera mononucleära celler, och även nivån av de regulatoriska
CD4+CD25+ cellerna var lika. De regulatoriska T cellerna verkar dessutom vara
funktionella, då celler från friska barn och diabetesbarn svarade på liknande sätt
vid proliferationstester.
Vid insjuknandet av diabetes hade barnen ett aktiverat immunsystem, sett som
högre nivåer av intracellulärt CTLA-4 per CD4+CD25high T cell, högre nivåer av
akutfasproteinet C reaktivt protein och ett högre uttryck av IFN- hos lymfocyter
efter att CD25+ celler tagits bort.
Slutsatser: Våra fynd betonar vikten av miljöfaktorers påverkan av
immunsystemet och utvecklingen av autoantikroppar. Vi kan däremot inte säga
att det finns en systemisk aktivering av immun systemet innan sjukdomen
framträder, men när väl diabetes utvecklats är immunsystemet aktiverat och
denna aktivering verkar regulatoriska celler försöka dämpa.
ORIGINAL PUBLICATIONS
This thesis is based on the following papers, which are referred to in the text by
their roman numerals:
I. Wahlberg, J., Fredriksson J., Nikolic E., Vaarala O., Ludvigsson J. and the
ABIS-study group
Environmental factors related to the induction of beta-cell autoantibodies in
1-yr-old healthy children
Pediatric Diabetes 2005: 6: 199-205
II. Walldén J., Honkanen J., Ilonen J., Ludvigsson J. and Vaarala O.
No evidence for activation of TH1 of TH17 pathways in unstimulated
peripheral blood mononuclear cells from children with β-cell autoimmunity
or T1D
Journal of Inflammation Research, In press
III. Walldén J., Ilonen J., Roivainen M., Ludvigsson J., Vaarala O. and the
ABIS-study group
The effect of HLA genotypes or CTLA-4 polymorphism on cytokine
response in healthy children
Scandinavian Journal of Immunology 2008:68 (3):345-350
IV. Walldén J., Lundberg A., Ludvigsson J. and Vaarala O.
Regulatory T-cells in children with type 1 diabetes
Manuscript
CONTENTS
ABBREVIATIONS .......................................................... 13
REVIEW OF THE LITERATURE ......................................... 15
Introduction to immunology ............................................... 15
Basic immunology ............................................................................... 15
Cytotoxic T cells ....................................................................................... 15
Helper T cells ............................................................................................ 16
B cells and antibodies ............................................................................... 17
The immunological synapse ............................................................... 17
Down-regulation of response.............................................................. 18
T helper cells........................................................................................ 20
Cytokines, cytokine receptors and transcription factors of TH
subtypes ................................................................................................ 21
Naturally occurring regulatory T cells ............................................... 22
Introduction to diabetes mellitus ........................................ 25
Definition and diagnosis of diabetes .................................................. 25
Classification of diabetes mellitus ...................................................... 26
Epidemiology of type 1 diabetes.......................................................... 27
Aetiology of type 1 diabetes ................................................................. 28
Pathogenesis of type 1 diabetes ........................................... 29
Genetic risk of type 1 diabetes ............................................................ 29
Human leukocyte antigen ......................................................................... 29
Cytotoxic T lymphocyte associated antigen 4 .......................................... 31
Inducible co-stimulator ............................................................................. 32
Lymphoid tyrosine phosphatase ............................................................... 32
Autoantibodies against β-cell antigens .............................................. 33
Insulin ........................................................................................................ 33
Glutamic acid decarboxylase .................................................................... 34
Tyrosine phosphatase–like insulinoma antigen-2 ..................................... 34
Environmental factors ........................................................................ 35
Viral infections .......................................................................................... 35
Cow’s milk ................................................................................................ 36
Gluten ........................................................................................................ 36
β-cell stress ................................................................................................ 37
T cells and cytokines in the pathogenesis of type 1 diabetes ............. 37
AIM OF THE THESIS ...................................................... 39
SUBJECTS & METHODS ................................................. 41
Study populations ................................................................ 41
Healthy school children ...................................................................... 41
The ABIS study.................................................................................... 41
Type 1 diabetic patients ....................................................................... 42
Methods.............................................................................. 43
Measurement of autoantibodies ......................................................... 43
HLA genotyping .................................................................................. 44
Single nucleotide polymorphism ........................................................ 45
Preparation of peripheral mononuclear cells .................................... 46
Real-time polymerase chain reaction ................................................. 46
Stimulation of peripheral mononuclear cells .................................... 47
Cell proliferation ................................................................................. 47
Enzyme-linked immunsorbent assay .................................................. 48
Multiplex cytokine assay ..................................................................... 49
Flow cytometry .................................................................................... 50
Statistical methods ............................................................. 50
Ethical considerations ......................................................... 52
RESULTS AND DISCUSSION ............................................ 55
Methodological aspects ....................................................... 55
Autoantibodies ..................................................................................... 55
Real-time PCR ..................................................................................... 57
Optimization of antigen stimulations ................................................. 59
Gating stategy ...................................................................................... 59
Magnetic depletion .............................................................................. 60
Influence of environment .................................................... 61
Effect of genetic risk factors on immune responses .............65
Immunological balance ....................................................... 74
SUMMARY AND CONCLUDING REMARKS .......................... 85
ACKNOWLEDGEMENTS ................................................. 87
REFERENCES............................................................... 91
Abbreviations
ABBREVIATIONS
ABIS
all babies in south-east Sweden
APC
antigen presenting cell
CD
cluster of differentiation
cDNA
complementary DNA
CI
confidence interval
CRP
C-reactive protein
cSMAC
central supra-molecular activation cluster
CTLA-4
cytotoxic T lymphocyte associates antigen 4
CV
coefficient of variation
CVB4
coxackie virus B4
DASP
diabetes autoantibody standardization program
DC
dendritic cell
ELISA
enzyme-linked immunosorbent assay
Foxp3
Forkhead/winged helix transcription factor
GAD65(A)
glutamic acid decarboxylase (autoantibody)
HLA
human leukocyte antigen
IA-2(A)
tyrosine phosphatase-like insulinoma antigen-2 (autoantibody)
IA-2ic
intracellular part of the IA-2 protein
IAA
insulin autoantibody
ICOS
inducible co-stimulator
IFN
interferon
Ig
immunoglobulin
IL
interleukin
IL-4R
interleukin-4 receptor
IL-12R
interleukin-12 receptor
iTreg
inducible regulatory T cell
kDa
kilo Dalton
LYP
lymphoid tyrosine phosphatase
MFI
mean fluorescent intensity
13
Abbreviations
MHC
major histocompability complex
mRNA
messenger RNA
NFAT
Nuclearfactor of activated T cells
NK cell
natural killer cell
nTreg
naturally regulatory T cell
OGTT
oral glucose tolerance test
OR
odds ratio
PBMC
peripheral blood mononuclear cells
PCR
polymerase chain reaction
PHA
phytohemagglutinin
pSMAC
peripheral supra-molecular activation cluster
RNA
ribonucleic acid
rRNA
ribosomal RNA
SI
stimulation index
SNP
single nucleotide polymorphisms
SPSS
statistical package for the social sciences
STAT
signal transducers and activator of transcription
T1D
type 1 diabetes
T2D
type 2 diabetes
T-bet
T-box expressed in T cells
TC
cytotoxic T cell
TCR
T-cell receptor
TGF
transforming growth factor
TH
helper T cells
TNF
tumor necrosis factor
Tr1
regulatory T cells type 1
TT
tetanus toxoid
WHO
World Health Organization
14
Review of the literature
REVIEW OF THE LITERATURE
Introduction to immunology
Basic immunology
Immunology is the science of our immune system, which provides defence
against infections, and is often divided into innate immunity and adaptive
immunity 1. Innate immunity is the first line of defence against infections with
microbial pathogens and every time an infectious agent is encountered the
response is similar. It consists of many interacting systems, including epithelial
barriers, antimicrobial peptides and effector cells. Distinct patterns on the
pathogen are recognized by cells like neutrophils, monocytes, dendritic cells
(DC) and natural killer cells (NK cells), clearing the infection by the mechanism
of phagocytosis and by secreting inflammatory mediators.
If the innate immune system is bypassed or overwhelmed by the pathogens, the
adaptive immune response is required. The adaptive immune response provides
a specific response, with an enormous ability to recognize different antigens,
and the responsiveness is improved on repeated exposure, providing
immunological memory. The cells involved are antigen-specific T and B cells
which, when activated, proliferate and develop into effector cells secreting
cytokines and antibodies, respectively 1. T cells can be divided into two different
populations based on their expression of the cell surface proteins cluster of
differentiation (CD) 4 or CD8. 1. B cells differentiates into antibody producing
plasma cells when activated by an antigen via its B cell receptor 1.
Cytotoxic T cells
Some intracellular bacteria and all viruses infecting different cell types multiply
in the cytoplasm and once they have entered the cells they are not susceptible for
extracellular antibodies. These pathogens can only be eliminated by the
15
Review of the literature
destruction of the host cell, which is mediated by cytotoxic T cells (TC).
Cytotoxic T cells are CD8+ T cells recognizing human leukocyte antigen (HLA)
class I molecules expressed on all nucleated cells. The HLA class I express
fragments from the infectious pathogens activating the TC cell which induces
cell death by the release of lytic enzymes or by introducing apoptosis 1.
Helper T cells
Helper T cells (TH) are CD4+ T cells recognizing HLA class II molecules
expressed on professional antigen presenting cells (APC), i.e. DCs,
macrophages and B cells. Helper T cells differentiate into different subtypes
depending on released mediators from the APC. The APC recognize distinct
patterns among pathogens, inducing the production of mediators resulting in
differentiation of naïve T cells into TH1, TH2 or TH17 cells. These subtypes
differ in the cytokines patterns (Figure 1) and thereby also in function.
Figure 1 Schematic overview of the possible differentiation factors for T helper (TH) cells
16
Review of the literature
B cells and antibodies
Antigen bound to the B cell receptor is internalized and presented upon HLA
class II for TH cells. Signals from the bound antigen and from the TH cell induce
the B cell to proliferate and differentiate into plasma cells, secreting antigen
specific antibodies 1. The main function of the antibodies are to neutralize
pathogens, binding the pathogen for enable phagocytosis or complement
activation 1. The antibodies can be divided into five major isotypes:
immunoglobulin M (IgM), IgD, IgA, IgE and IgG.
The immunological synapse
A central event in the development of the adaptive immunity is the activation of
T cells. The initiation of this process is the triggering of T cells by APC. The
contact molecules between T cells and APC form the so called “immunological
synapse”, a place for signalling 2.
The APC degrades foreign pathogens into peptides, which are expressed
together with HLA molecules on the APC surface. A T cell with a T cell
receptor (TCR) specific for the presented antigen will bind to the HLA/peptide
complex, the CD8 or CD4 binds to the HLA class I or II molecules,
respectively, strengthening HLA/TCR interaction, and the T cell will receive its
first signal for activation. This will lead to the rearrangement of receptors into a
central supra-molecular activation cluster (cSMAC) 3, 4, enriched with TCRs,
CD28 and CD2 molecules, surrounded by a peripheral SMAC (pSMAC),
enriched with adhesion molecules 3, 4. CD28 binds to CD80 and CD86 expressed
on the APC, generating a signal lowering the threshold for T cell activation 5.
The binding of adhesion molecules will extend the contact between APC and
T cells, mediating a longer HLA/TCR interaction. Efficient T cell activation
requires one signal from the HLA/TCR complex and second signals from the
17
Review of the literature
co-stimulatory (Figure 2) and adhesion molecules present in the cSMAC and
pSMAC.
Figure 2 A schematic overview of the immunological synapse. The antigen presenting cell
(APC) presents a degraded protein as peptides, bound to human leukocyte antigen (HLA)
molecules. The HLA/peptide complex is recognized by the T cell receptor (TCR) and delivers
the first signal for T cell activation via CD3. The T cell also receives a second signal though
the binding of CD28-CD80/86. Once activated, cytotoxic T lymphocyte associated antigen
(CTLA-4) is up-regulated and, binding to CD80/86, confers an inhibitory effect.
Down-regulation of response
Once a successful T cell activation has occurred and the pathogen is rendered
harmless, the immune response must be down regulated to avoid damage in the
surrounding tissue. The mechanisms in terminating a lymphocyte response
include T cell inhibition by cytotoxic T lymphocyte associated antigen (CTLA4, CD152), activation-induced cell death or down regulation mediated by
regulatory T cells 6.
18
Review of the literature
Cytotoxic T lymphocyte associated antigen-4 (CD152) is primarily an
intracellular membrane protein and it is up regulated on the T cell surface during
activation 7, 8. The peak of messenger RNA (mRNA) expression appears after six
to 24 hours and maximal protein expression two to three days after initial T cell
activation 7, 8. The ligands for CTLA-4 are the same as for CD28 (Figure 2), but
the affinity of CTLA-4 for both CD80 and CD86 is between 20- and 100-fold
higher than that of CD28, with a strong bias towards CD80 9. Several
mechanisms have been proposed for the down-regulation of immune response
by CTLA-4, including competition with CD28 for ligands and/or interference
with intracellular signalling pathways for TCR and /or CD28 signalling, thereby
inhibiting proliferation and interleukin (IL)-2 secretion by T cells 10, 11.
Cytotoxic T lymphocyte associated antigen-4 may also be involved in
disassociation of the cSMAC 12 or lead to a more transient interaction between T
cells and APC, diminishing T cell activation 13.
Regulatory T cells have been proposed to have a role in suppression of immune
response and maintenance of self- tolerance 14, 15. Both inducible and naturally
regulatory T cells (iTreg and nTreg, respectively) are involved, but these
populations differ in their mechanisms of action. Inducible regulatory T cells
develop in the periphery from conventional CD4+ T cells under the influence of
cytokines and are defined by their secretion of IL-10 (regulatory T cells type 1
(Tr1)) and transforming growth factor (TGF)-β (TH3) 14-17. Suppression by
naturally occurring CD4+CD25+ T cells is mediated by cell contact-dependent
mechanisms, e.g. by “outside-in” through CD80/86, inhibiting IL-2 production
in responder cells 18-20 or by the secretion of IL-35 21.
In conclusion, the immune system defends us against infections in different
ways, adapting its response depending on type of pathogen. The innate immunity
serves as the first line of defence and the adaptive immunity serves a more
19
Review of the literature
specific response. The down regulation of the immune response is important in
suppressing activated cells after pathogen clearance and preventing tissue
damage.
T helper cells
The TH1 and TH2 cells were originally characterized in mice 22 and later also in
humans 23. Murine and human T cells produce cytokines in similar patterns, with
TH1 cells producing IL-2, interferon (IFN) - and tumor necrosis factor (TNF)-β,
whereas TH2 cells produce IL-4, IL-5, IL-9 and IL-13 (Figure 1). TH1 and TH2
cells play different roles in the response against invading pathogens. TH1 cells
are effective in cell-mediated inflammatory reactions, recruiting and activating
macrophages and TC cells to fight against intracellular infections and stimulating
B cells to produce opsonising and neutralizing antibodies. TH2 cells are on the
other hand involved in the humoral immune response by activating antibody
secreting B cells, eosinophils and basophils in the defence against parasites and
helminths.
The TH1/TH2 paradigm is a rather generalized and simplified picture, but this
concept has been, and is still today, rather useful. However, recently a subset
distinct from TH1 and TH2 was discovered and named TH17 according to the
production of the highly pro-inflammatory cytokines of the IL-17 family
(Figure 1) 24. The receptors for IL-17 are expressed on epithelial cells and other
cell types, and IL-17 is therefore believed to promote tissue inflammation. The
complete mechanistic function of these TH17 cells is however not fully
understood.
20
Review of the literature
Cytokines, cytokine receptors and transcription factors of TH subtypes
One of the most critical cytokine inducing a TH1 immune response is IL-12 25.
Interleukin-12 is a heterodimer composed by the two subunits IL-12α (p35) and
IL-12β (p40) signalling through the IL-12 receptor (IL-12R). The IL-12R is
composed of a β1 and a β2 subunit. Both β1 and β2 are necessary for binding IL12, but the β2 subunit is responsible for transferring the signal 26. Interleukin-12
acts on cells of the innate and the adaptive immune system, inducing significant
levels of IFN-. Interferon- induces the expression of the transcription factor
T-box expressed in T cells (T-bet), via signal transducers and activator of
transcription (STAT) 1 27. This even further increases the production of IFN-,
but also induces IL-12Rβ2 chain expression 27-29. Interleukin-12 strongly induces
STAT4 phosphorylation in human TH1 cells 30, augmenting the IFN-
production 28 as well as increasing the expression of IL-12Rβ2 31, completing the
TH1 developmental commitment process. Once activated, T-bet, together with
IFN-, forms an auto-regulatory positive feedback loop to maintain a type 1mediated response 27 and inhibit IL-5 production 32.
The TH2 lineage is predominantly induced by the IL-4/STAT6 signalling
pathway. Although several types of cells have been shown to secrete IL-4, the
initial source of IL-4 is still unknown. Interleukin-4 binds to the IL-4R,
composed of the common  chain and the IL-4Rα chain. The phosphorylated IL4R can further recruit and activate STAT6, which in turn induces GATA-3 and
c-maf 33. GATA-3 has been demonstrated to be selectively expressed in TH2
cells 34 and both GATA-3 and c-maf are important transcription factors for IL-5
and IL-4 35, 36. Retroviral introduction of GATA-3 or STAT6 into TH1 cells
introduced TH2 specific cytokines and suppressed IFN- production, further
stressing the importance of GATA-3 in TH2 development 33, 37.
21
Review of the literature
The relatively newly discovered TH17 cells produce the pro-inflammatory
cytokines IL-17A, IL-17F, IL-21, IL-22 and IL-6 24, 38-40. T helper 17 cells were
previously believed to develop from naïve T cells in response to IL-23 41-44.
Later TGF-β and IL-6 were shown to elicit TH17 cells in mice 45 while IL-23
was found to retain the TH17 commitment. Recent data suggest that TGF-β
together with pro-inflammatory cytokines like IL-6 and IL-1β is necessary also
in humans for the up-regulation of the transcription factor RORc (the human
ortholog of mouse RORt) 40, 46, which is required for the induction of IL-17
production 40, 47.
Figure 3 summarize the important factors involved in the differentiation of naïve
TH cells into different subtypes.
Naturally occurring regulatory T cells
Naturally occurring regulatory T cells is a subpopulation of TH cells with low
proliferative capacity and is specialized in down regulation of the immune
response. Naturally regulatory T cells develops in thymus and are found in the
peripheral lymphoid tissue, where the induction of suppressor capacity requires
activation through TCR, but once activated their function can be nonspecific.
Suppression by nTreg cells is mediated by cell contact-dependent mechanisms,
e.g. by “outside-in” through CD80/86, by inhibiting IL-2 production in
responder cells 18-20, or by the secretion of IL-35 21.
Naturally occurring regulatory T cells have been characterized by the expression
of CD25 (IL2Rα) 48, 49, CTLA-4 50, 51, glucocorticoid-induced TNF receptor
(GITR) 52 and inducible co-stimulator (ICOS) 53 together with the absence of
CD127 54.
22
Review of the literature
The forkhead/winged helix transcription factor Foxp3 is the transcription factor
for nTreg 55, 56 and IL-2 is an important cytokine for maintaining Foxp3
expression 57, signaling through STAT5 58. Transforming growth factor-β also
supports the maintenance of Foxp3 59. The importance of Foxp3 in nTreg
function has been demonstrated in mice and humans with mutations in the
Foxp3 gene. 60, 61. These mutations are in humans responsible for a disease
called immunodysfunction polyendocrinopathy enteropathy X-linked syndrome
(IPEX), causing an impairment of nTreg cells. This disease is associated with
several autoimmune diseases, such as type 1 diabetes (T1D).
Recent studies have shown Foxp3 expression in activated CD4+CD25- T effector
cells 62-64. The Foxp3 expression is making them hyporesponsive, suggesting
that Foxp3 induction turn of T cell activation 62, 65. Stimulation of effector T
cells, resulting in a transient up-regulation of Foxp3, does not result in
suppressive activity, however. For the conversion of effector T cells to
regulatory T cells, a high and stable expression of Foxp3 is required 62. The
function of Foxp3 in effector T cells and the molecular factors that regulate its
expression remains to be elucidated. However, the transcription factor nuclear
factor of activated T cells (NFAT)-c2, together with STAT5, have been found to
be involved in Foxp3 up-regulation 58, 66.
Figure 3 summarize the important factors involved in the differentiation and
function of nTreg cells.
In conclusion, the fate of TH lymphocytes is controlled by antigen receptors and
secreted cytokines from APCs, influencing the development of different subtypes.
The nTreg is important in controlling the action of activated effector T cells,
although the mechanisms are not fully understood.
23
Review of the literature
Figure 3 Schematic picture of TH1, TH2, TH17 and nTreg cells together with cytokines,
receptors and transcription factors important for their development. Markers studied in Paper
II are indicated in bold.
24
Review of the literature
Introduction to diabetes mellitus
Definition and diagnosis of diabetes
Diabetes mellitus comprises a group of metabolic disorders characterized by a
chronic hyperglycaemia caused by defects in insulin secretion, insulin action or
both 67. The effects of the chronic hyperglycaemia in diabetes are damage,
dysfunction and failure of several organs, in particular eyes, kidneys, nerves,
blood vessels and heart.
The symptoms associated with diabetes are caused by the hyperglycaemia,
including thirst, polyuria, blurring of vision and weight loss 67. These symptoms
are often not severe and may even be absent, leading to pathological and
functional changes long before diagnosis is made 67. Diabetic children are often
presenting severe symptoms at onset with very high blood glucose levels,
glycosuria and ketonuria 67.
The diagnosis is based on measurements of plasma/blood glucose in
combination with the symptoms mentioned above. If a person is asymptomatic,
the diagnosis should be made only after repeated plasma/blood glucose tests
where the value should be in the diabetic range, or the diagnostic criteria for
diabetes is met in the oral glucose tolerance test (OGTT) 67. The diagnostic
criteria are the same in children and adults but in most children the diagnosis is
made by plasma/blood glucose test, without an OGTT 67, since the symptoms
are severe. The diagnosis can be confirmed by plasma glucose value ≥ 12.2
mmol/L or fasting plasma glucose  7.0 mmol/L. The diagnostic criterion for
fasting whole blood glucose is  6.1mmol/L. If the diagnosis still is uncertain,
an OGTT is used as confirmation of the diagnosis. In children a plasma glucose
value ≥ 11.1 mmol/L in capillary blood or ≥ 10.0 mmol/L in venous blood are
25
Review of the literature
considered as diagnostic cut-off values for diabetes two hours after an oral
glucose load of 1.75 g per kg body weight.
Classification of diabetes mellitus
According to the World Health Organization (WHO), diabetes mellitus can be
divided into four groups depending on aetiology.
Type 1 diabetes is due to pancreatic islet β-cell destruction 67 and thus “insulin
is required for survival, to prevent the development of ketoacidosis, coma and
death” 67. At least ten percent of all cases of diabetes are T1D. Most cases are
autoimmune, characterized by the presence of autoantibodies against antigens
present in the pancreatic islets i.e. insulin, glutamic acid decarboxylase (GAD65)
and/or tyrosine phosphatase-like insulinoma antigen-2 (IA-2), identifying an
ongoing autoimmune process. Insulin dependent T1D without signs of
autoimmunity, where no autoantibodies are found but β-cell destruction is
evident, is classified as a type 1 idiopathic diabetes 67. In children the disease
process usually is rapid, but more gradual in adults. The slowly progressive form
among adults, with phenotypic type 2 diabetes (T2D) but a slowly developing
autoimmune process, is referred to as latent autoimmune diabetes in adults
(LADA).
Type 2 diabetes is the most common type of diabetes, including the forms
caused by defects in insulin secretion and/or insulin resistance. Exogenous
insulin is usually not required for survival 67 and the treatment consist of diet,
increased physical activity and weight loss. Type 2 diabetes is still rare in
children and adolescents in Sweden, but is becoming increasingly common in
e.g. USA 68. The symptoms of the hyperglycaemia are usually not severe, but the
disease increases the risk of severe late complications.
26
Review of the literature
Gestational diabetes means pronounced carbohydrate intolerance with onset
during pregnancy resulting in hyperglycaemia 67.
Other specific types include less common types where the cause can be
identified in a relatively specific manner 67. This group of diabetes includes for
example genetic defects on -cell function or insulin action, e.g. different forms
of maturity onset diabetes in the young (MODY). There are also rare cases of
diabetes caused by diseases of exocrine pancreas, drug- or chemical induced or
caused by infections 69.
Epidemiology of type 1 diabetes
The incidence of T1D is increasing all over the world, with significant variation
in the incidence in different parts of the world. The highest incidence is seen
among Caucasians and the lowest in Asia and South America. The WHO has
tried to estimate the incidence of T1D in 57 countries around the world during a
10-year period, between 1990-1999 70, despite the difficulties in several
countries with high child mortality. The incidence rates varied between
0.1/100,000 per year in China and Venezuela to 40.9/100,000 in Finland 70,
where Finland had a peak of 48.5 cases/100,000 in 1998 71. Sweden had an
average incidence of 30.0/100,000 per year during 1990-1999 70. Since then,
there has been a steady increase in incidence up to >60/100 000 in Finland 72 and
>40/100 000 in Sweden, according to the national Swedish registry (personal
communication).
The mean annual increase worldwide between 1995-1999 was 3.4% and the
corresponding figure for Sweden between 1990-1999 was 3.6% 70. The
increasing incidence of T1D in Swedish children may perhaps to some extent be
explained by a shift to a younger age at diagnosis 73.
27
Review of the literature
Aetiology of type 1 diabetes
The aetiology of T1D is largely unknown but the development is believed to be
determined by a combination of genetic susceptibility genes, immune
dysregulation and environmental factors. The resulting autoimmune process
may be initiated several years before clinical onset of T1D (Figure 4). The
autoimmune process causes a decrease in β-cell mass, leading to diminishing
insulin production. Several genetic susceptibility loci have been proposed to be
involved in the development of T1D 74 and a number of environmental factors
have been suggested to additional contribution 75.
Figure 4 Schematic illustration over T1D development. Interactions between genes, the
immune system and environmental factors may trigger an autoimmune response leading to the
loss of β-cell mass and the progression to T1D (Adapted from Atkinson & Eisenbarth 76).
In conclusion, T1D is a serious disease, dependent of lifelong support of insulin
and the incidence of T1D in Sweden is among the highest in the world and is
still increasing. The mechanisms behind the development are still largely
unknown, but there seems to be interactions between several parameters.
28
Review of the literature
Pathogenesis of type 1 diabetes
Genetic risk of type 1 diabetes
Several genes are involved in the pathogenesis of T1D. These genes are neither
necessary nor sufficient for disease to develop, but they modify the risk. The
HLA is the major locus associated with T1D 74, 77, 78, but also CTLA-4 74, 79, 80, as
well as other loci 74, 80-82 have been found to modulate the susceptibility.
Human leukocyte antigen
The first loci identified to increase the risk for T1D was in the HLA region 83.
This region is located on chromosome 6p21 and has been implicated as a major
genetic risk factor. The genes in this area are arranged in three sub-regions, class
I, II and III, where class II has the major influence on T1D risk. HLA class IIDQ alleles have the strongest genetic association with T1D 84, but DR alleles
have the ability to modify the risk conferred by the DQ locus, suggesting that
both DR and DQ are likely to be important for determining the risk for T1D 85,
86
. Each HLA molecule consists of one alpha (A) and one beta (B) chain, which
together form a heterodimer (Figure 5). These chains of DQ are encoded by the
DQA1 and DQB1 genes, some of which are in linkage disequilibrium with each
other. This means that these alleles are showing a non-random association with
each other, whereby it is possible to deduce a DQA1 allele based on the DQB1
and vice versa.
An asterisk is used to separate the gene name from the alleles and the alleles for
each gene are numbered using four digits. The first two describe the most
closely associated serologic specificity and the latter two describe the subtypes
within this serologic specificity. Different DR/DQ molecules might have
varying degrees of binding affinity to specific peptides, but as the binding
affinity of a given DR or DQ molecule to a particular epitope is identical in all
29
Review of the literature
ethnic groups, the effect of the same HLA molecule on T1D risk should not vary
across ethnic groups if the primary auto-antigens are identical.
Figure 5 The HLA class II molecule.
Most studies regarding genetic risk factors are done in Caucasian populations
and HLA allele and haplotypes frequencies vary considerably across ethnic
groups 87. For instance, Asian populations show much lower frequencies of the
T1D high risk DR-DQ haplotypes relatively common among Caucasian
populations; and the disease prevalence in these two ethnic groups might mirror
these differences. Certain HLA class II haplotypes show strong associations
with the development of T1D and are found more often among T1D children
than controls. Others are more often found among controls than T1D children
and are then considered to be protective (Table 1).
Table 1 A selection of HLA haplotypes and the association with T1D risk
Risk of T1D
DR
DQA1
DQB1
High
*0401/2/4/5
*0301
*0302
High
*03
*05
*02
Neutral
*0101
*0101
*0501
Protective
*1501
*0102
*0602
Protective
*05
*05
*0301
30
Abbreviation
DR4-DQ8
DR3-DQ2
DR1-DQ5
DR2-DQ6
DR5-DQ7
Review of the literature
The HLA class II is responsible for the presentation of antigenic peptides to
T cells. Appearance of β-cell autoantibodies have been associated with HLA
haplotypes connected with T1D 88, 89 and aberrant cytokine responses have been
reported in association with HLA class II risk haplotypes 90-92. Thus, peptide
presentation may affect T1D susceptibility.
Cytotoxic T lymphocyte associated antigen 4
Cytotoxic T lymphocyte associated antigen-4 is an important regulator of the
immune system as it, when interacting with its ligands CD80/86, is responsible
for the down-regulation of T cell activation. The gene of CTLA-4 is located on
chromosome 2q33 93 and consist of four different exons where several single
nucleotide polymorphisms (SNP) have been identified 79, 80, 94.
The CTLA-4 polymorphism + 49 A/G (rs231775), where a shift from an A to an
G results in an amino acid substitution, is the SNP most associated with
autoimmune disease, but also other SNPs in the gene of CTLA-4, like CT60
(+6230) A/G (rs3087243) and CTBC217_1 C/T (rs2033171), are related to the
development of autoimmune diseases 80, 94-98. Relatively little is known about the
effect of these susceptibility polymorphisms.
The +49 A/G polymorphism has been associated with an altered function of
CTLA-4, resulting in a higher cytokine secretion and proliferative response
upon T cell activation 99, 100, perhaps due to a reduced up-regulation of CTLA-4
in the individuals with susceptibility SNP 101.
The CT60 SNP has also been associated with disease susceptibility and may
correlate to a lower mRNA of the soluble form of CTLA-4 80, 102, although this
has not been confirmed by others 97. The CT60 susceptibility SNP has also been
shown to alter the signalling threshold of CD4+ T cells 103.
31
Review of the literature
Inducible co-stimulator
The ICOS is a co-stimulatory molecule induced on T cells during activation,
enhancing T cell responses to foreign antigens and induces the production of
several cytokines, including IL-4, IL-5, IFN- and IL-10, from T cells 104.
The ICOS gene is mapped to the same region as CTLA-4 on chromosome 2q33
and the polymorphism in the ICOS gene may contribute to disease susceptibility
too. Several SNPs have been found in the ICOS gene 105. The CTIC154_1C/T
(c.1624, rs10932037) is a SNP located in the 3´ UTR of ICOS gene 80 and
individuals homozygous for the CTIC154_1 C/C have higher expression of
ICOS than heterozygous individuals 102.
Lymphoid tyrosine phosphatase
The gene of PTPN22, encoding the protein Lymphoid Tyrosine Phosphatase
(LYP), is located on chromosome 1p13 and has been implicated in the
pathogenesis of T1D 81, 82, 106. Lymphoid Tyrosine Phosphatase is a protein
found in the cytosol and nucleus of haemapoetic cells and is a strong negative
regulator of T cell activation. For T cells to become activated several proteins
inside the cells are phosphorylated and LYP is causing dephosphorylation of
these proteins, e.g. the TCR/CD3 complex, thereby inhibiting T cell activation
107
. A SNP has been identified within PTPN22, the gene encoding LYP, at
position 1858(rs2476601), where a shift from a C to a T results in a replacement
of an amino acid 81. Individuals homozygous for 1858T/T have been shown to
have a defect in TCR response, which results in a gain of inhibitory function 108,
i.e. dephosphorylation of the signalling proteins occurs much more efficiently.
The variant of PTPN22 1858T has been associated with an increased risk for
autoimmune diseases, including T1D 81, 82, 106. One hypothesis postulated is that
1858T allele predisposes to autoimmune disease due to a more active
32
Review of the literature
suppression of TCR signalling during thymic development, allowing survival of
autoreactive T cells that normally would be deleted 107.
In conclusion, several genetic factors are important regulators of the
immunological response, also making them important in the pathogenesis of
T1D. More detailed studies are required to understand the mechanisms
underlying these effects, however.
Autoantibodies against β-cell antigens
Before clinical manifestation of T1D, autoantibodies against β-cell antigens can
be detected in most individuals. These autoantibodies can be used as markers of
the autoimmune process and may be present years before clinical T1D. The
presence of multiple autoantibodies can be used to identify individuals at risk of
T1D 109. Today, three major islet autoantibodies are used for prediction of T1D.
Insulin
Insulin was the first β-cell antigen detected in newly diagnosed T1D patients 110
and is the only β-cell specific auto antigen. Insulin autoantibodies (IAA) are
often the first autoantibody to appear in individuals who develop autoimmunity
to islet antigens and the levels of IAA correlate inversely with age since levels
of IAA decline at older ages 111. The frequency of IAA positive patients seems
to be more common in children from areas with high incidence of T1D and the
presence of IAA appears to give a clinically milder disease 112.
Insulin autoantibodies are found in 40-70% of newly diagnosed patients 112-114
and in 0.9-3% of a healthy population 115, 116.
33
Review of the literature
Glutamic acid decarboxylase
Glutamic acid decarboxylase was originally detected as a 64 kilo Dalton (kDa)
protein from islet homogenates to which sera from newly diagnosed T1D
patients were shown to immunoprecipitate 117. These autoantibodies were also
found before clinical onset of T1D 118. Later, this 64 kDa protein was identified
as the enzyme GAD and was found to be expressed in pancreatic islet cells and
in peripheral nerves 119. In terms of action, GAD is involved in the conversion of
glutamic acid to GABA, an inhibitory neurotransmitter, but the function in the
pancreatic islets is however not clear 120. There are two isoforms of GAD,
GAD65 and GAD67, but only GAD65 has been shown to be expressed in human
islets and is responsible for the autoantibody response 121.
Autoantibodies against GAD (GADA) are found in about 50-80% of newly
diagnosed T1D patients 112, 113, 120, 122. In contrast 0-3% of the general population
has GADA 115, 116, 120, 123.
Tyrosine phosphatase–like insulinoma antigen-2
Besides the 64 kDa protein identified as GAD, sera from T1D patients were
found to bind another protein that is cleaved into a 37 kDa and 40 kDa
fragments by trypsin. The 40 kDa and 37 kDa proteins were subsequently
identified as the proteins IA-2/ICA512 and IA-2β, respectively 124-127. Both IA-2
and IA-2β are transmembrane proteins and autoantibodies to have been shown
to be directed against the intracellular part of the IA-2 protein (IA-2ic) 124, 128
where 95% of T1D patients with autoantibodies against IA-2 react with the
carboxyl-terminus (amino acids 771-979) 120.
About 55-80% of newly diagnosed T1D patients have autoantibodies against
IA-2 (IA-2A) 113, 120 while 0-2.5% of the general population has IA-2A 115, 116, 120,
123
.
34
Review of the literature
In conclusion, the emergence of autoantibodies against insulin, GAD and IA-2
are markers for an ongoing autoimmune process and can be used to identify
individuals at risk of T1D.
Environmental factors
The genetic predisposition can not entirely explain the development of T1D in
certain individuals, since there is approximately 40-50% concordance of
developing autoantibodies or T1D in identical twins 78, 129, 130 and only about six
to 20% of siblings to probands are affected 131-133. Furthermore the rapidly
increasing incidence is unlikely to depend on changes of genetic predispositions
in the population but may be a consequence from environmental changes.
Several environmental factors have been suggested to trigger the autoimmune
response and the development of T1D and some of them are presented more in
details below.
Viral infections
Viral infections have been suggested as risk factors for the development of T1D
134-140
, and several mechanisms how viruses can trigger autoimmunity have been
proposed 141. Enteroviruses, and in particular Coxsackie virus B4 (CVB4), have
been proposed as triggers of β-cell autoimmunity and T1D 142, 143. Enteroviral
infections during pregnancy have been associated with increased risk for T1D in
the offspring 134, 135, although discrepant results exist 143-145. Also, increased
numbers of enterovirus infections have been demonstrated in children later
progressing to T1D 134. Children with T1D have, compared to healthy children,
also been shown to have an impaired immune response against CVB4 146, 147.
The hygiene hypothesis suggest that increased hygiene may cause changes in the
gut bacterial flora, influencing the maturation of the immune system, facilitating
35
Review of the literature
imbalance and thereby autoimmune reactions in genetically predisposed
individuals 148 and thus the increased hygiene might lead to low immunity
against certain viruses. This fits with the idea that low frequencies of enteroviral
infections increase the susceptibility of diabetogenic effect of enteroviruses 149.
Cow’s milk
Associations between cow’s milk exposure and β-cell autoimmunity and later
T1D development have been shown in several studies 150, 151, even though
contradicting results exist 152. Early introduction of cow’s milk based formula
and high consumption of cow’s milk have been associated with higher levels of
β-cell autoantibodies 153. An important question is what component in cow’s
milk that can trigger β-cell autoimmunity and several suggestions have been
made, including different cow milk proteins and particularly bovine insulin.
Enhanced immune response to cow milk proteins have been shown in children
later developing T1D 154 suggesting a primary failure in the gut immune system
in children at risk of T1D. This could result in aberrant immune response to
dietary proteins, such as bovine insulin. Early exposure to bovine insulin can
enhance the humoral and cellular immune response to insulin in infants 155, and
thus be the link between β-cell autoimmunity and cow’s milk.
Gluten
Celiac disease is a chronic inflammation in the small intestine resulting in villus
atrophy and this inflammation is triggered by wheat gluten. The association of
celiac disease and T1D have been shown in several studies 156, 157, possibly
partially explained by the shared genetic background of DQA1*05-DQB1*02
(DR3-DQ2) 158. Early introduction of gluten has been reported as a risk factor
for β-cell autoimmunity 159, but the mechanisms mediating the risk are not
understood.
36
Review of the literature
β-cell stress
Increased weight gain in infancy 75, 160 and psychological stress 161 may induce
extra pressure on the β-cells and can be regarded as risk factors for the
development of T1D. During infancy and puberty, periods of rapid growth, the
need for insulin increases, causing β-cells work hard to produce the required
insulin. The increased insulin production may result in stimulation of the
autoimmune process leading to overt T1D 148. Psychosocial stress in families
may also affect insulin secretion and insulin sensitivity, although the biological
mechanisms are still not known. Several psychological factors can influence the
development of autoantibodies as high parental stress and serious life events can
induce IA-2A in one or two and a half-year old children of a general population,
respectively 161, 162.
In conclusion, an immunological imbalance may result in multiple
environmental factors triggering the development of β-cell autoimmunity and
T1D, although the detailed mechanisms are unknown.
T cells and cytokines in the pathogenesis of type 1 diabetes
Clinical, autoimmune diabetes is caused by a loss of immunological tolerance,
causing a destruction of the insulin producing β-cells in pancreas, called
insulitis. The initial event triggering this autoimmune reaction is still unknown,
however the immune reaction to β-cells can be divided into three stages 163. The
first stage is the primary sensitization against β-cell antigens, the second stage is
APC presentation of auto-antigens causing an immune reaction and consequent
inflammation in the islet. The third and last stage includes a cycle of harmful
interactions between immune cells and β-cells.
37
Review of the literature
Autoimmune insulitis has been investigated in several animal models like the
nonobese diabetic (NOD) mice and BioBreeding (BB) rats 163, 164, greatly
enhancing our knowledge in pathogenesis of T1D. Activated T cells, specific for
β-cell auto-antigens, home to the pancreas, expand clonally and start an
autoimmune reaction culminating in β-cell destruction. The release of cytokines
from TH cells recruit macrophages into the tissue, activating them, eliminating
the antigen-bearing cell 165. The cytokines also activate TC cells to destroy target
cells expressing the antigen presented together with major histocompability
complex (MHC) class I and NK cells to destroy target cells independently of
MHC expression. Also other mediators released, like free radicals and nitrix
oxide, support the destruction since they are highly toxic to the β-cell.
In human insulitis, immunostaining has revealed infiltrating CD4+, CD8+ T cells
as well as macrophages 166, 167 and the role of cytokines have been investigated
in several studies, demonstrating discrepant results. The model of TH1 and TH2,
and subsequently IFN- and IL-4, has been used in several studies exploring the
possible changes in immune response in T1D. Peripheral blood mononuclear
cells (PBMC) have been used for these studies and the expression and secretion
of various cytokines have been explored in unstimulated PBMC as well as after
various stimulations. Peripheral blood mononuclear cells been shown to produce
higher amounts of the cytokine IFN- 168, 169 as well as lower levels of IL-4 170,
171
. However, others have shown a lower expression and secretion of IFN- 171,
172
, while the level of IL-4 was similar in T1D patients and controls 168, 172. This
low secretion of IFN- has also been shown in individuals at high risk of
developing T1D after stimulation with the three auto-antigens GAD, insulin and
IA-2 173 or with the viral antigen CVB4 146.
In conclusion, the pathogenesis of T1D is believed to result from cytokine and
immunoregulatory imbalance, culminating in β-cell death and subsequent T1D.
38
Aim of the thesis
AIM OF THE THESIS
The general aim of this thesis was to investigate how diverse environmental and
immunological risk factors associated with the development of T1D influence
the immunological system.
The specific aims of the individual papers were:
I.
To study associations between environmental risk factors and T1D-related
autoantibodies at one year of age in a general population.
II.
To investigate the profile of T helper cell polarization markers in
peripheral blood, both in children with increased risk of T1D, either
genetic risk and/or with diabetes related autoantibodies, and in children
with T1D during the first year after manifestation of T1D.
III.
To examine if children with T1D associated gene polymorphisms, such as
HLA class II risk alleles or the CTLA-4 +49 A/G, CTLA-4 CT60 A/G
and CTLA-4 CTBC217_1 C/T polymorphism, are associated with an
aberrant cytokine responses to diverse antigens.
IV.
To study the amount and fluctuation of CD4+CD25high regulatory T cells
during 18 months period from the manifestation of T1D and also to
compare the function of CD4+CD25high regulatory T cells in children with
T1D and healthy children.
39
Subjects & Methods
SUBJECTS & METHODS
Study populations
Three different study populations were used: healthy children, healthy children
with increased risk for T1D and children with manifest T1D.
Healthy school children
Healthy school children were recruited from Rydsskolan in Linköping, Sweden.
The children were asked to fill out a form together with their parents. This form
contained questions about atopic status (eczema, hay fever, asthma or usage of
allergy medicines), presence of type 1 or type 2 diabetes, thyroiditis, rheumatoid
arthritis or celiac disease in the children or in any first degree relative. Children
with any of the conditions above or with first degree relatives with any of the
conditions above were excluded. In Paper II we studied samples from eight
children with the mean age of 11.7 years (range 11.0-12.8 years) and in Paper
IV we studied samples from 17 children with the mean age of 11.7 years (range
5.3-16.3 years).
The ABIS study
In Paper I, II and III we studied samples taken from children participating in the
ABIS study (All Babies in south-east Sweden) which is a population-based
prospective cohort study of 17 055 (out of 21 700 invited) infants born between
1st October 1997 and 1st October 1999. The aim of the study was to study the
importance of environmental factors for the development of T1D, but also of
other autoimmune diseases and allergy in the general population. A second aim
was to identify children with risk of developing T1D, and if effective
intervention appeared, to be able to intervene to prevent some cases of the
disease.
41
Subjects & Methods
The newborns have been followed from birth up to five years of age with
biological samples and questionnaires. Biological samples have been collected
at birth (cord blood, breast milk, hair from mothers), one (blood, stool and hair),
two and a half and five years of age (blood, stool, urine and hair). At the age of
five, venous blood samples were taken, enabling analysis of cell mediated
immunity.
In Paper I we studied capillary blood samples from 6 000 children taken at one
year of age together with at-birth and one year questionnaires. Autoantibodies to
GAD and IA-2 were analyzed in 5 765 and 5 635 children, respectively. In
Paper II we studied venous blood samples from 44 children taken at five years
of age. These children were selected based on their risk for T1D. Fifteen
children as they had T1D associated HLA class II risk haplotypes DQA1*05DQB1*02 (DR3-DQ2) and/or DRB1*0401/2/4/5-DQB1*0302 (DR4-DQ8), 13
children because of their autoantibody positivity for GADA and/or IA-2A and/or
IAA and finally, for comparison, 16 children without any HLA risk haplotypes
or autoantibodies. In Paper III we used randomly collected venous blood
samples from 67 children taken at five years of age.
Type 1 diabetic patients
In Paper II and IV we studied samples taken from children with manifest T1D.
All these children have been recruited at the Pediatric Clinic at
Linköping University hospital and the diagnosis was based on the WHO criteria
from 1999 67.
In Paper II, samples from 17 children were taken at three follow-up visits, ten
days after diagnosis, one to three months after diagnosis and nine to 18 months
after diagnosis. Mean age at diagnosis was 10.4 years (range 6.0-15.6 years).
In Paper IV, samples were taken from 20 children at four follow-up visits, ten
42
Subjects & Methods
days after diagnosis, three month after diagnosis, nine month after diagnosis and
18 month after diagnosis. Mean age at diagnosis was 10.9 years (range 3.5-16.8
years).
Methods
Measurement of autoantibodies
Autoantibodies against GAD, IA-2 and insulin were analyzed by radiobinding
assay. Capillary blood and serum samples were taken at well-baby clinics and
stored in freezers at -20C until analysis. Capillary whole blood samples were
analyzed in Paper I and serum samples in Paper II and III. Serum samples in
Paper II and III were also analyzed for IAA.
The complementary DNA (cDNA) for GAD65 and IA-2ic was extracted from
plasmid-carrying E.Coli (a kind gift from Professor Å. Lernmark, Seattle) and
transcribed and translated in vitro in the presence of 35S-labeled methionine. All
samples analyzed for GADA and IA-2A were incubated with 35S-labeled protein
where after IgG antibodies were precipitated before washing and measurement
of radioactivity in a Microbeta Tri-Lux (Wallac, Turku, Finland). The results
were expressed as concentrations of autoantibodies, calculated in relation to a
standard curve. Sample with values above the highest standard were diluted to
fit the standard curve. The sensitivity limits for quantitative determinations were
7.8-500 units/mL for GADA and 4-176 units/mL for IA-2. Background counts
were subtracted from each well and duplicate samples with a coefficient of
variation (CV) over 20% were re-run.
The samples analyzed for IAA were tested in competition assay with equal
volumes of human recombinant unlabeled insulin (Sigma-Aldrich, Stockholm,
Sweden) and human recombinant insulin labelled with 125I (3-
43
Subjects & Methods
[125I]iodotyrosylA14 insulin) (Amersham Biosciences). The samples were
incubated with recombinant insulin, before precipitation, washing and
measurement of radioactivity in a gamma counter. The results were expressed as
concentrations of IAA, calculated in relation to a standard curve. Sample with
values above the highest standard were diluted to fit the standard curve. The
sensitivity limits for quantitative determinations were 1.96-125 units/mL.
Background counts were subtracted and specific counts were calculated by
subtraction of counts of excess unlabeled insulin from counts of labelled insulin.
Duplicate samples with a CV over 20% were rerun.
In Paper I we used both the 99th percentile and the 90th percentile as cut-off for
autoantibody positivity. The 99th percentile corresponded in our study
population to 37.2 WHO units for GADA and 32.7 WHO units for IA-2A. The
90th percentile corresponded in our study population to 30.9 WHO units for
GADA and 29.9 WHO units for IA-2A. In Paper II and III, the 98th percentile
was the cut-off for positivity for GADA and IAA and 99th percentile for IA-2A.
This corresponds to 33.2 WHO units for GADA, 4.3 WHO units for IA-2A and
4.1 WHO units for IAA.
HLA genotyping
In Paper II and III, HLA genotyping was done with time-resolved fluorometry
based sandwich hybridisation assay (Figure 6), and performed by the research
group of Professor Jorma Ilonen in Turku, Finland. The polymerase chain
reactions (PCR) were carried out using whole blood or a 3 mm in diameter disc
from whole blood spots on filter paper. Specific HLA-DQB1, DQA1 or DRB1
primer pairs, of which one was biotinylated at the 5’-end, were used for
amplification of the genes. The PCR product was transferred into a streptavidincoated microtiter plate together with the detection probe labelled with various
lanthanide chelates (Europium, Samarium and Terbium) as described previously
44
Subjects & Methods
174-176
. After an incubation step, allowing the formation of hybrids and to
simultaneously collect them onto the wells, the time-resolved fluorescence was
detected in a using a Victor 1420 Multi label Counter (Wallac Oy, Turku,
Finland). Different emission wave length and delay times were used for specific
detection of each lanthanide label.
Figure 6 Principle of time-resolved fluorometry. Isolated DNA was amplified with a
biotinylated primer. Hybridization was performed with specific DQB1, DQA1 and
DRB1detection probes on streptavidin coated microtiter plates, thereafter time-resolved
fluorescence was measured.
Single nucleotide polymorphism
In Paper III, CTLA-4, ICOS and PTPN22 SNPs were analyzed by the research
group of Professor Jorma Ilonen in Turku, Finland. Three polymorphisms for
CTLA-4 were detected as +49 A/G (rs231775), CT60 (+6230) A/G (rs3087243)
and CTBC217_1 C/T (rs2033171), one ICOS SNP was detected as CTIC154_1
(c.1624) C/T (rs10932037) and one PTPN22 SNP was detected as +1858 C/T
(rs2476601).
DNA was extracted according to a standard salting-out method 177. Detection of
CTLA-4 +49 A/G and PTPN22+1858 C/T SNPs was done after DNA was
amplified using primer pairs, of which one was biotinylated at the 5’-end. The
biotinylated amplification product was bound to a streptavidin coated microtiter
plates and hybridized to one of two lanthanide labelled reporter probe specific
45
Subjects & Methods
for the polymorphisms of CTLA-4 +49 A/G or PTPN22 +1858 C/T (Figure 6).
Detection was done by time-resolved fluorometry using a Victor 1420 Multi
label Counter (Wallac).
Determination of CT60 A/G (rs3087243), CTBC217_1 C/T (rs2033171) and
CTIC154_1 C/T (r10932037) was done using pre-designed primers and
TaqMan probes in real-time PCR based SNP genotyping assays (Applied
Biosystems, Foster City, CA, USA). Samples are mixed with two target specific
primers and one VIC and one FAM dye-labelled probe, each specific for one
SNP where after genotypes are determined.
Preparation of peripheral mononuclear cells
In Paper II, III and IV, PBMC were isolated from heparinized venous blood by
Ficoll-Paque™ (Amersham Biosciences AB, Uppsala, Sweden) density gradient
centrifugation.
In Paper II, the PBMC were cryopreserved until further analysis. In Paper III,
the isolated PBMC were resuspended to 1x106 viable for stimulation, as
described below and in Paper IV, PBMC were depleted of the CD25+ subset
(CD25depleted PBMC) by using anti-CD25-magnetic beads from Dynabeads
before the PBMC and CD25depleted PBMC were used for proliferation assay
and stimulation, as described below.
Real-time polymerase chain reaction
Total ribonucleic acid (RNA) was extracted from unstimulated, thawed PBMC
(Paper II) or after 72 hours incubation (Paper IV) and converted to cDNA. The
mRNA expression was determined by the TaqMan method of real-time PCR,
with ribosomal RNA (rRNA) as endogenous control. The comparative Ct
46
Subjects & Methods
method described by the manufacturer is used to calculate a relative
transcription value.
Table 2 All analyzed markers in Paper II and Paper IV, purchased from Applied Biosystems
(Foster City, CA, USA).
Marker
Paper II Paper IV
rRNA/18s (Hs99999901_s1)
X
X
GATA-3 (Hs00231122_m1)
X
IL-4Rα (Hs00166237_m1)
X
T-bet (Hs00203436_m1)
X
IL-12Rβ1 (Hs00234651_m1)
X
IL-12Rβ2 (Hs00155486_m1)
X
IL-17A (Hs00174383_m1)
X
X
ICOS (Hs00359999_m1)
X
CTLA-4 (Hs00175480_m1)
X
Foxp3 (Hs00203958_m1)
X
X
X
IFN- (Hs00174143_m1)
IL-6 (Hs00174131_m1)
X
IL-10 (Hs00174086_m1)
X
IL-2 (Hs00174114_m1)
X
Stimulation of peripheral mononuclear cells
In Paper III, the isolated PBMC were resuspended to 1x106 viable cells/mL and
were cultured with medium alone, tetanus toxoid (TT), polio virus type 1 /Sabin,
CVB4, pertussis toxin or phytohemagglutinin (PHA). After 48 hours, the
samples were centrifuged at 400g for 10 minutes and the supernatants were
aspirated and stored at -70C. In Paper IV, the PBMC and CD25depleted PBMC
were resuspended to 1x106 viable cells/mL and cultured with medium alone or
PHA. After 72 hours, the samples were centrifuged at 400g for 10 minutes and
the supernatants were aspirated and stored at -70C.
Cell proliferation
In Paper IV, 1x105 cells/well of PBMC and CD25depleted PBMC were
stimulated in a round bottom, 96-well plate with medium alone or in the
presence of PHA. The cells were cultured for 6 days and were pulsed with
47
Subjects & Methods
[3H]thymidine for the last 16-18 hours. The cells were then harvested and
assessed for [3H]thymidine incorporation in a liquid scintillation counter (1205
Betaplate, Wallac, PerkinElmer Inc, Boston, MA, USA). Results were expressed
as a stimulation index (SI=mean counts/min of quadruplicate wells containing
cells + antigen divided by mean counts/min of quadruplicate wells containing
cells + medium alone). Fifty micro litre cell supernatant was taken from each
well before [3H]thymidine was added, enabling the measurement of cytokines.
Enzyme-linked immunsorbent assay
Cytokine production and C-reactive protein (CRP) were analyzed using enzymelinked immunosorbent assay (ELISA) (Figure 7). Microtiter plates were coated
with monoclonal antibodies against measured proteins and free plastic spaces
were blocked with blocking solution. Standards and samples were added to the
plates and following incubation, detection was performed by using biotinylated
antibodies directed against measured proteins, followed by streptavidin
conjugated polyhorseradish peroxidase. Substrate was added and the reaction
was stopped by adding 1.8 M sulfuric acid. The responses were expressed in
pg/mL and calculated in relation to the standard curve. Those samples that had a
value below the limit for quantitative determinations were assigned a value
corresponding to half of that value and samples with values above the limit was
diluted. Background responses (cultures with medium alone) were subtracted
from the response from cultures with antigen induced cytokine concentrations.
In Paper III and IV, ELISA was used for analyzing levels of cytokines and in
Paper IV, CRP was also measured with ELISA.
48
Subjects & Methods
Figure 7 General principle of enzyme-linked immunosorbent assay (ELISA).
Cellsupernatants are transfered into a microtitre plates coated with antibodies, capturing the
protein of interest. A secondary antibody conjugated with biotin is added and thereafter
streptavidin bond with an enzyme is. The enzyme reacts with a substrate and form a colored
product which can be measured spectrophotometrically. The amount of colored product is
proportional to the amount of the specific protein present in the sample.
Multiplex cytokine assay
In paper IV, secreted cytokine levels were determined by commercially
available multiplex cytokine assays (Bio-Rad Laboratories Inc, Hercules, CA,
USA) on a Luminex 100. Samples or standards were mixed together with a
mixture containing an excess of microspheres coupled with antibodies to each
cytokine. After incubation and washing, biotinylated detection antibodies were
added, followed by streptavidin-phycoerythrin (PE). The fluorescence intensities
of the microsphere and detection antibodies were measured. The responses were
expressed in pg/mL and calculated in relation to a standard curve. Background
responses (cultures with medium alone) were subtracted from the response from
cultures with antigen induced cytokine concentrations.
49
Subjects & Methods
Flow cytometry
Regulatory markers, CD25 and CTLA-4, were analyzed with flow cytometry in
Paper IV. Whole blood was incubated with anti-CD4, CD8, CD25 and CTLA-4
antibodies with attached fluorescent dyes. After an incubation step, the
erythrocytes were lysed and cells were permeabilized for enabling
measurements of intracellular CTLA-4. The cells were analyzed with four-color
flow cytometry using a FACSCalibur (BD Bioscience). Lymphocytes were
gated from forward- and side scatter and two parameter dot plots were created.
CD25high was gated according to a slight decrease in CD4 expression.
Extracellular and intracellular expression of CTLA-4 and mean fluorescent
intensities (MFI) of CTLA-4 was determined in all gated populations.
Statistical methods
In Paper I, all data from the questionnaires were optically scanned and manually
checked for errors. As a marker for induction of autoantibodies, we used the 90th
percentile as cut-off and for identification of children at risk of T1D we used the
99th percentile, calculated from 5 765 children regarding GADA and 5 635
children regarding IA-2. To identify variables which individually was predictive
of the development of autoantibodies, we used a logistic regression model where
we used univariate analysis (stage 1). Variables found significant at the level of
0.05 were then included in the multivariate model (stage 2), where all variables
were included by stepwise forward selection of the most significant and
analyzed simultaneously. Multivariate logistic regression was used to estimate
the odds ratio (OR), with a 95% confidence interval (CI), and an OR above 1.0
indicates increased risk.
In Paper II-IV, data were not normally distributed and non-parametric test,
corrected for ties, were used. Comparisons between paired groups were analyzed
50
Subjects & Methods
with Wilcoxon signed-rank test and more than two paired groups with Friedman
test. More than two unpaired groups were analyzed with Kruskal-Wallis as a
pre-test (correcting for multiple comparisons) with Mann-Whitney U test as
post-hoc. For comparison of two unrelated groups, Mann-Whitney U test was
used. Correlations were analyzed with Spearman’s rank correlation coefficient
test.
Mass significance is a phenomenon due to the fact that statistical analyses are
based on probabilities. The probability of finding a difference is 5%, if p=0.05 is
chosen as significance level. The risk of finding false significances (type 1 error)
increases with the number of statistical analyses performed. One way to adjust
for multiple tests is to use a lower p-value. However, this increases the risk for
missing actual differences (type II error). In Paper I, p<0.05 was used since we
regarded the risk of missing actual differences more problematic than the risk of
finding false significances. Differences found were interpreted for whether they
were theoretically expected and biologically explainable. In Paper II and IV, no
corrections for multiple comparisons were made due to rather few comparisons
in each group, and a p<0.05 was considered significant. However, in Paper III,
adjustments for multiple comparisons were done regarded different HLAgenotypes and the level of significance was lowered to 0.01. For comparisons
between three groups, we used Kruskal-Wallis and the method itself considers
the multiple groups compared.
All statistical calculations were performed using the statistical package for the
social sciences (SPSS) (SPSS Inc., Chicago, Illinois, USA). In Paper I version
11.0 were used, in Paper II version 14 were used, in Paper III version 13 were
used and in Paper IV version 15 were used. A p-value given as 0.000 by the
SPSS program is reported as p<0.001.
51
Subjects & Methods
Ethical considerations
Type 1 diabetes often develops in children, why studying the environmental and
immunological factors influencing the development of T1D among children is
important. Research on children should not be performed if similar results may
be obtained on adults with their informed consent, only if physiological and
pathological states that are particular for childhood will be investigated. This
applies to studies of immune and environmental aspects of childhood diabetes. It
may seem unethical to include individuals who cannot give informed consent in
research projects, but to exclude groups from research can also be unethical and
discriminating.
The society benefits from our research if the results lead to better understanding
of the factors influencing the development of T1D among children, possibly to
eventually enable prediction and prevention of childhood diabetes. The scientific
society gains from increased knowledge of important issues regarding general
functions of T1D associated factors.
All participating families got written and oral information what it should mean
to take part in our studies. They were also informed that the participation was
totally voluntary and could be discontinued at any time point without any
specific reason. The parents were not automatically informed about genetic risk
genotypes or presence of autoantibodies. However, they were told that they
could receive this information upon active request, although no effective
intervention is available at the moment.
Discomfort during blood sampling was minimized by a topical anaesthetic
(EMLA band-aid) and sampling was performed by experienced personnel. All
parents /guardians gave their informed consent to enter the studies. Thus, we
believe that the ethical benefits exceed the risks in these studies.
52
Subjects & Methods
All studies were approved by the Research Ethic committee of the Medical
Faculty of Linköping University. The ABIS study was also approved by the
Research Ethic committee of the Medical Faculty of Lund University.
53
Results & Discussion
RESULTS AND DISCUSSION
Several environmental, e.g. virus infections 134, 136, 140 and early exposure to
bovine protein and insulin from cow’s milk 178, and immunological risk factors
have been associated with the development of T1D. I have hypothesized that
some general immunological mechanisms are missing in children that will
develop T1D, resulting in an incorrect response to various antigen, enhancing
their risk to develop an autoimmune disease like T1D.
In the different projects some aspects of this matter have been studied:
 If environmental factors may induce an autoimmune process in a general
population (Paper I).
 If children with increased risk for T1D or with manifest T1D have a
different balance between subsets of peripheral blood derived CD4+ T
cells (Paper II).
 If children in the general population with T1D associated HLA class II
risk genotypes or CTLA-4 polymorphisms associated with autoimmune
diseases have an altered cytokine response upon T cell stimulation (Paper
III).
 If T1D children have a reduced number of CD4+CD25high regulatory T
cells or if these cells show a defective function (Paper IV).
Methodological aspects
Autoantibodies
In Paper I, II and III, samples from the ABIS-study were used. Three slightly
different cut-off levels were used for the autoantibody measurements. In Paper I,
capillary, lysed whole blood was used and as cut-off the 90th and 99th percentile
was chosen, both for GADA and IA-2A. These cut-offs identified two different
populations of children. The 90th percentile was used to identify children in the
general population in whom β-cell autoimmunity has been induced but without
55
Results & discussion
any strong connection to the development of T1D. The 99th percentile was used
to identify children with an increased risk of developing T1D, proven as a good
cut-off previously 115. Insulin autoantibodies were not analyzed due to
difficulties measuring IAA in whole blood.
In Paper II and III, we used serum samples and the 98th percentile for GADA
and IAA and 99th percentile for IA-2A were used. A high cut-off was chosen
due to an increased risk for T1D. We used the 98th percentile instead of the 99th
percentile due to decreased sensitivity when threshold was increased, but still
with a rather high specificity (Table 3). As very few children had increased
levels of IA-2 and the 98th percentile was below the sensitivity level of the
method, we used the 99th percentile for IA-2A. Due to differences in values of
IA-2A when analyzing whole blood (Paper I) and serum samples (Paper II and
III), whole blood generating higher levels, the 90th percentile could be used in
Paper I, but only 99th percentile in Paper II.
The cut-off for GADA and IA-2A in Paper I were based on all samples analyzed
in our study (5 765 samples for GADA and 5 635 samples for IA-2A) and only
the specificity for the 90th and 99th percentile were calculated. The cut-off for
GADA and IA-2A in Paper II and Paper III are based on 4007 samples analyzed
randomly from the five-year follow-up and the cut-off for IAA is based on 2 481
samples.
Taken part in the Diabetes Autoantibody Standardization Program (DASP) in
both 2002 and 2005 gave the specificity and sensitivity shown in Table 3.
56
Results & Discussion
Table 3 Specificity and sensitivity from Diabetes Autoantibody Standardization Program
(DASP) in 2002 and 2005. n.a.; not analyzed
Percentile Autoantibody
2002
2005
Specificity Sensitivity Specificity Sensitivity
90th
th
98
th
99
GADA
94%
82%
n.a.
n.a.
IA-2A
100%
54%
n.a.
n.a.
GADA
n.a.
n.a.
96%
76%
IA-2A
n.a.
n.a.
n.a.
n.a.
IAA
n.a.
n.a.
99%
30%
GADA
96%
82%
99%
72%
IA-2A
100%
55%
100%
72%
IAA
n.a.
n.a.
100%
24%
Real-time PCR
In Paper II and IV, data from real-time PCR were included, allowing a fast and
sensitive quantification in small samples sizes. However, discrepancies may
exist between the expression of mRNA and protein levels. In general, several
methods can be used to evaluate the expression of a specific marker, for
example expression of specific mRNA, secretion of the protein or expression of
the protein in cells, with flow cytometry.
Real-time PCR was chosen for Paper II due to several reasons. Only small
volumes of blood from the children were received due to ethical and practical
reasons, limiting the methods which can be used. In addition, as expression of
various markers was studied in unstimulated cells, where cytokine secretion is
low, we did not measure the secreted cytokines. Also, most cytokines are not
detectable or close to the detection limits in plasma/serum. Although Western
Blot can be used for analyzing proteins, this did unfortunately not work properly
57
Results & discussion
in unstimulated cells. Also for Paper IV, we chose to measure mRNA expression
as we were interested in the spontaneous expression of cytokines.
In both Paper II and Paper IV the comparative Ct method was used. In each
quantitative PCR method, differences in starting material will introduce errors.
Therefore normalization by a house-keeping gene is required to correct for these
experimental variations. In both Paper II and Paper IV, rRNA was used for this
purpose. Phytohemagglutinin stimulated PBMC from four healthy donors, from
whom cDNA has been pooled, was used as an in-house calibrator, and all
samples were related to this.
The comparative Ct method uses arithmetric formulas to achieve a relative
quantification and the amount of target gene, normalized to an endogenous
reference (rRNA, 18s) and relative to a calibrator, is given by the formula
2-ΔΔCt(User Bulletin #2, Applied Biosystems). For each calculation to be valid,
the efficiency of the target amplification and the efficiency of the endogenous
reference must be approximately equal. This has been tested by looking at how
ΔCt varies with template dilution. Dilutions ranged from 1 to 1/512 and after the
run, ΔCt was plotted against log input amount. The slope should be <0.1 for
each measured target gene. In Figure 8, ICOS is shown as an example and all
target genes, from both Paper II and IV, passes this test.
58
Results & Discussion
ICOS
45
40
y = -1.1283x + 35.078
R2 = 0.9892
35
Ct(ICOS)
Ct (ICOS)
Ct(18s)
Ct values
30
y = -1.0897x + 20.726
25
R2 = 0.9989
20
dCt
Ct (rRNA)
Linear (dCt)
Linear
ΔCt (Ct(18s))
15
Linear (Ct(ICOS))
10 y = -0.0386x + 14.352
R2 = 0.1244
5
0
-8
-6
-4
-2
0
2
4
log2(conc_RNA)
Figure 8 Plot over Ct values against log input amount for ICOS. The dilution ranged from 1
to 1/512 and the absolute value of the slope should be close to 0, but in any case <0.1. ΔCt of
ICOS showed a slope of -0.04, which passes the test
Optimization of antigen stimulations
In Paper III, stimulation of PBMC with antigens was optimized using PBMC
collected from children not participating in the study. The stimulation was
performed as described in Paper III. Different incubation times (24, 48 and 72
hours) were tested and 48 hours were found to be optimal. Different
concentrations were tested for each antigen (TT: 5, 10 and 20 Lf/mL; Polio:
0.1,0.5 and 1.0 µg/mL; CVB4: 0.1, 0.5, 1.0 µg/mL; Pertussis: 0.5, 1.0, 2.0
µg/mL) and the optimal concentrations were found to be 20 Lf/mL of TT, 0.5
µg/mL of Polio, 0.5 µg/mL of CVB4, 2 µg/mL Pertussis. Optimal
concentrations and incubation times of PHA and anti-IL-4R have been
optimized previously.
Gating strategy in flow cytometry analysis
The methods of gating regulatory T cells have been discussed carefully. As the
CD4+CD25+ phenotype is displayed by activated T cells, the ability to identify
regulatory T cells with this marker is limited. Several efforts have been made to
59
Results & discussion
combine different surface markers for a more specific characterization 50, 54, 179182
, and commercially available antibodies against Foxp3 can further support the
identification of regulatory T cells 56, 182. However, at the time of sample
collection we had no possibility to study the expression of Foxp3 or CD127 due
to lack of suitable antibodies. Therefore lymphocytes were identified by their
forward- and side- scatter properties (Figure 9a) and the CD4+CD25high
population was gated according to the highest expression of CD25 and a slight
decrease in CD4 intensity after regions were set individually for each child with
corresponding isotype control (Figure 9b) 183.
a)
b)
Figure 9 Representative flow cytometric plots from a T1D patient showing gating strategy
used to define CD4+CD25high T cells. Lymphocytes were gated according to their forwardand side-scatter properties (a) and examined for expression of CD4 and CD25. CD4+CD25high
T cells were defined as lymphocytes positive for CD4 staining with the highest expression of
CD25 together with a slight decrease in CD4 expression (b).
For intensity of intra- and extra cellular CTLA-4, MFI were calculated after
correction for instrument variation by QC3 ™ microbeads, allowing CV of 15%
for each fluorescent dye.
Magnetic depletion
In Paper IV, we used magnetic sorting for the depletion of CD4+CD25+ T cells.
The optimal way to select a specific population would be by cell sorting on a
60
Results & Discussion
flow cytometer, but since we had no cell sorter available we chose magnetic
depletion for the depletion of CD25+ cells. This is a rather cheap method,
needing nothing else than you sample, magnetic beads and a magnet. However,
magnetic cell sorting don’t always allow for a stringent isolation of cells. The
intention in Paper IV was to remove the CD4+CD25+ regulatory T cells and we
examined the result with flow cytometry, individually for each sample, making
sure that the population of regulatory cells where depleted after treatment.
(Figure 10)
b)
a)
3-10 %
1%
Figure 10 Representative flow cytometric plots from a child showing depletion of
CD4+CD25+ T cells. Lymphocytes were examined for expression of CD4 and CD25.
CD4+CD25high T cells were defined as lymphocytes positive for CD4 staining with the highest
expression of CD25 together with a slight decrease in CD4 expression (a). After magnetic
depletion the CD4+CD25high T cells was absent in the sample, verifying a successful depletion
(b).
Influence of environment
In studies investigating environmental factors, an ordinary set up is using
children with increased genetic risk, who are followed with respect to
autoantibody development and development to clinical manifest T1D. Thus we
investigated whether environmental factors could trigger signs of β-cell
autoimmunity during the first year of life in an unselected population of children
(Paper I). Out of 5 765 samples analyzed for GADA and 5 635 samples
analyzed for IA-2A, 582 had levels of GADA above the 90th percentile and
61
Results & discussion
57 above the 99th percentile. For IA-2A, the corresponding figures are 574 at the
90th and 57 at the 99th percentile. 126 samples had both GADA and IA-2A above
the 90th percentile and 6 samples above the 99th percentile. The appearance of
autoantibodies in young children is a dynamic process and some of the
autoantibody positive children will revert back to negativity 184. As this
fluctuation is more likely to appear among children with lower autoantibody
level than the 99th percentile, the 99th percentile was used as marker for
increased risk of T1D. Children with high levels of two or more autoantibodies
have the highest risk of developing clinical T1D 185 and in Paper I, six children
had multiple antibodies above 99th percentile at one year of age. However, at
later time points all children had reverted back to autoantibody negativity and
none of them had in November 2007 converted to clinical T1D.
The risk for developing autoantibodies above 90th percentile was associated with
several factors analyzed (Table 4).
Table 4 Risk factors related with increased levels of β-cell autoantibodies to IA-2A and
GADA, above the 90th percentile, in an unselected population. The odds ratio (OR),
confidence interval (CI) and p-values from a stepwise logistic regression analysis are shown.
Risk factors
>90th percentile
Gender
Boys/Girls
Celiac disease among
grandparents
Birth month
Autumn
Winter
Spring
Summer
Maternal gastroenteritis
Month 7 during pregnancy
Mother’s education
IA-2A
[OR (95% CI)]
GADA
[OR (95% CI)]
IA-2A and GADA
[OR (95% CI)]
1.3 (1.1-1.6)a
2.2 (1.1-4.4)b
1.0 (ref)
1.3 (1.0-1.8)ns
1.5 (1.1-2.0)c
0.9 (0.7-1.3)ns
1.1 (1.0-1.3)d
1.5 (1.2-1.9)e
1.4 (1.1-1.7)f
a
p=0.007, b p=0.027, c p=0.004, d p=0.028, e p=0.001, f p=0.003
Low/medium vs high
62
Results & Discussion
This suggests that environmental factors might contribute to the development of
β-cell autoimmunity. Interestingly, these factors were the same which previously
have been associated with the development of T1D in several studies. Increased
risk of IA-2A above 90th percentile in children born during spring may suggests
that environmental factors very early in life 150, or even during pregnancy 134, 135,
may play a role in the development of β-cell autoimmunity. The finding of
gastroenteritis during pregnancy as a risk factor for both GADA and IA-2A also
supported the theory of maternal influences of the children’s immune system
during the perinatal period.
Children with levels of GADA and/or IA- 2A above the 99th percentile more
often had a first-degree relative with T1D, OR 2.6 (95% CI 1.0-6.6; p=0.042).
The risk of developing T1D has previously been associated with T1D in first
degree relatives 131. Offspring of mothers with T1D have a risk of approximately
one to five percent of developing T1D, whereas the risk is about five to eight
percent if the father is affected 186-188. In brothers or sisters the risk varies
between six and 20%, depending on their degree of HLA identity 131-133. As we
investigated an unselected population of children at one year of age, we did not
have enough cases for separate analysis of the effect of diabetes in the parents or
siblings. It would however be interesting to investigate this relation in the ABIS
material now, when the children are nine to eleven years old, or in the future
when they are even older, with both β-cell autoimmunity and T1D as primary
outcomes.
The presence of GADA above the 99th percentile was associated with
grandparents having T2D, OR 2.1 (95% CI 1.1-4.3; p=0.033) and the prevalence
of T1D and T2D has also previously been shown to occur rather often in the
same families 131, 189, 190. We have in follow-up analysis confirmed the
association between T2D in first-degree relatives and the occurrence of
63
Results & discussion
autoantibodies 153. This suggests that the etiological factors of T1D and T2D
may, at least partly, be shared, which fits with the β-cell stress hypothesis 148.
Early exposure to cow’s milk (during the first two months in life) was also
associated with high levels of autoantibodies, especially IA-2A, OR 2.9 (95% CI
1.5-5.5; p=0.001). Early introduction of cow’s milk based formula, together with
high consumption of fresh cow’s milk at one year of age, has also been
associated with higher β-cell autoantibodies in older children 153. Associations
between cow’s milk, T1D and induction of IA-2A have previously been shown
151
, even though contradicting results exist 152. As the importance of cow’s milk
in the induction of β-cell autoantibodies and T1D is debated, a large doubleblinded prospective intervention study have been started, aiming to determine
the relationship between early introduction of cow’s milk and the development
of T1D 191.
Our results, together with others, emphasize the complexity around
environmental factors and the importance of these factors at an early stage in the
process against β-cell autoimmunity. However, as most children with
autoantibodies do not develop T1D, other factors likely modify the effect of the
environmental triggers and may provide protection against T1D. One of these
factors may be the HLA class II protective haplotypes. When we studied the
effect of protective haplotypes on response to enterovirus stimulation (Paper III)
we found that the DQB1*0602 (DR2-DQ6) allele was associated with a higher
IFN- and IL-2 response to polio virus and the DQA1*05-DQB1*0301 (DR5DQ7) allele with higher IL-6 and IL-2 response to CVB4 (Figure 12a-d). This
might indicate a stronger immune response against enterovirus and contribute to
protection from enterovirus infections 192.
64
Results & Discussion
Effect of genetic risk factors on immune responses
Human leukocyte antigen class II region on chromosome 6p21encodes the most
important genetic factor in T1D and previous studies have reported aberrant
cytokine responses in association with HLA class II risk haplotypes 90-92,
however multiple non-HLA genes also contribute to disease development. The
gene encoding CTLA-4 on chromosome 2q33 80, 95, ICOS mapped to the same
chromosome, 2q33, as CTLA-4 and recently LYP, encoded by the PTPN22 gene
on chromosome 1p13, have all been implicated in autoimmune diseases like
T1D 81, 82. All of these genes are actively involved in the function of the immune
system, in regulating the activation of T cells. The HLA DQ and DR alleles
influence T1D susceptibility 85, 86 and among Caucasians, DRA1*0401/2/4/5DQB1*0302, encoding DR4-DQ8, and DQA1*0501-DQB1*0201, encoding
DR3-DQ2, have been suggested to have the strongest association with T1D.
Studies of the HLA-peptide binding domain demonstrate that different HLA
alleles bind different epitopes, maybe even from the same antigen 193 and
susceptibility and protective HLA class II molecules could bind and present
non-overlapping peptides 194.
The profile of TH cells in peripheral blood derived unstimulated lymphocytes
was in Paper II investigated in children with HLA class II haplotypes associated
with T1D. In Paper III, children with T1D associated HLA class II haplotypes,
CTLA-4 polymorphisms, ICOS polymorphism or PTPN22 polymorphism
associated with autoimmune diseases were investigated with respect to cytokine
responses upon T cell stimulation. No remarkable up- or down regulation of
markers for different TH subtypes was seen in children with the risk haplotypes
DQA1*05-DQB1*02 (DR3-DQ2) and DRB1*0401/2/4/5-DQB1*0302 (DR4DQ8), either with risk haplotypes in combination with each other or with neutral
haplotypes (Figure 11). This indicates that HLA risk haplotypes do not have an
impact on a general polarization of TH cells in peripheral blood.
65
Results & discussion
Figure 11 The mRNA expression of various immune factors was similar in children with
HLA risk haplotypes DQA1*05-DQB1*02 (DR3-DQ2) and DRB1*0401/2/4/5-DQB1*0302
(DR4-DQ8) (filled squares) and in healthy children without any risk for T1D (open squares).
Horizontal lines indicate median values.
In Paper III, cytokine responses to different antigens were investigated with
respect to different genetic risk factors associated with T1D. As our group
previously explored the relationship between HLA and CTLA-4 +49 SNP risk
factors and cytokine response to T1D related antigens 99, the aim in Paper III
was to study whether additional risk genotypes related to T1D were associated
with an aberrant cytokine response in general.
As most Swedish children follow the recommended vaccination program and are
thus vaccinated against TT, pertussis toxin and polio virus, almost 100% of the
children respond with cytokine secretion to these antigens. Also enterovirus
66
Results & Discussion
infections are common 195 and responses to CVB4 are thus seen in many of the
children.
Children with the risk haplotype DQA1*05-DQB1*02 (DR3-DQ2) had a higher
IL-13 response to TT stimulation (p=0.002, Paper III). Different HLA alleles
have the ability to present different peptides in different ways. The elevated IL13 response seen among children with DQA1*05-DQB1*02 (DR3-DQ2) might
be a result of this. As IL-13 can be regarded as a marker of TH2 response this
might also suggest an impaired polarization against a TH1 immune response in
response to TT. This kind of impaired TH1 response has earlier been shown
among T1D patients 196. As no other differences were seen in children with the
two different HLA risk haplotypes DQA1*05-DQB1*02 (DR3-DQ2) or
DRB1*0401/2/4/5-DQB1*0302 (DR4-DQ8), it seems that children with T1D
associated risk haplotypes do not have disposition for developing aberrant
cytokine responses in general. These results are in accordance with the previous
finding of no general T cell polarization among unstimulated peripheral blood T
cells (Paper II).
Children with the protective haplotype DQB1*0602 (DR2-DQ6) had higher
IFN- (p<0.001) and IL-2 (p=0.005) in response to polio stimulation, compared
to other genotypes (Figure 12a-b). Polio virus belongs to the family of
enteroviruses, which have been associated with T1D 134, 197, 198 and the
seroconversion to -cell autoantibody positivity 142, 143. Interferons have the
ability to induce an antiviral response in human pancreatic islet cells, especially
true for IFN-α, but also IFN- showed the same tendencies 192. Our results may
support this view and indicate that the response seen in children with the
haplotype DQB1*0602 (DR2-DQ6) could contribute to a better protection from
enterovirus infections and minimize the risk of accelerating β-cell destruction.
Stimulation with CVB4 did not however show the same increased IFN-
response in children with DQB1*0602 (DR2-DQ6). Instead, the eleven children
67
Results & discussion
with the protective haplotype DQA1*05-DQB1*0301 (DR5-DQ7) showed a
higher IL-6 (p=0.039) and IL-2 (p=0.015) response against CVB4 than the
children with other genotypes (Figure 12c-d).
a)
b)
c)
d)
Figure 12 The protective HLA haplotypes DQB1*0602 (DR2-DQ6) was associated with
higher production of IFN- (a) and IL-2 (b) in response to polio stimulation while children
with the protective haplotype DQA1*05-DQB1*0301 (DR5-DQ7) was related to increased
production of IL-6 (c) and IL-2 (d) in response to coxsackie virus (CVB4). Horizontal lines
indicate median values and p-values of Mann-Whitney U-test are shown. Note the different
scales for the y-axis.
68
Results & Discussion
The haplotype DQB1*0501 (DR1-DQ5), which is considered as a neutral
haplotype in regard to T1D risk, was associated with increased secretion of
IL-10 in response to polio stimulation (p=0.003) and a higher secretion of IL-6
in response to TT (p=0.001) (Paper III).
The connection between CTLA-4 +49 A/G polymorphism and autoimmune
diseases has been investigated and show a greater proliferative response of T
cells from individuals with +49 G/G genotype 100 and lower expression of
CTLA-4 in CD4+CD25high T cells among children with +49 G/G genotype 199, as
well as an increased expression in +49 A/A individuals 101. The CTLA-4 is also
involved in reducing the contact period between HLA and TCR molecules 13 and
a decreased expression of CTLA-4 on cell surfaces may increase this contact
and induce T cell activation and cytokine release. CT60 G/G has also been
associated with a lower expression of soluble CTLA-4 80, 102, although discrepant
results exist 97. We investigated whether these polymorphisms, together with
CTBC217_1 C/T, were associated with an aberrant cytokine response, as
CTLA-4 inhibits cytokine production in activated T cells. Usually transcription
factors such as activator protein-1 (AP-1), nuclear factor-B (NF-B) and
NFAT cause an up-regulation of the cytokines IL-2, IFN- and IL-4 200, 201 upon
T cell activation. This signal is inhibited by CTLA-4 mediating a downregulated cytokine production 11. Thus, a decreased expression of CTLA-4 due
to polymorphisms 101, 199 could lead to increased T cell activation 100 and higher
secretion of cytokines, which could increase the risk for autoimmune diseases,
like T1D.
69
Results & discussion
a)
b)
Figure 13 Pertussis toxin (PT) induced IFN- in children with CTLA-4 G/G polymorphism
(a) and CTBC217_1 T/T polymorphism (b). Horizontal lines indicate median values and pvalues of Mann-Whitney U-test are shown.
Stimulation of lymphocytes with bacterial toxins and viruses resulted in a higher
IFN- secretion in children with the CTLA-4 genotypes associated with T1D
risk. For instance pertussis stimulation induced a higher IFN- secretion in
children with the +49 G/G SNP (p=0.034, Kruskal-Wallis test) (Figure 13a) and
CTBC217_1 T/T (p=0.015, Kruskal-Wallis test) (Figure 13b).
Also, after PHA stimulation, children with CTBC217_1 T/T had a higher IFN-
secretion (p=0.046, Kruskal-Wallis test) (Figure 14).
70
Results & Discussion
Figure 14 Children with the CTBC217_1 T allele showed higher IFN- production in
response to Phytohemagglutinin (PHA) stimulation. Horizontal lines indicate median values
and p-values of Mann-Whitney U-test are shown.
Similar tendencies were observed for the genotype CT60 G/G, with higher
IFN- production both after PT and PHA stimulation (p=0.055 and p=0.062,
respectively, Kruskal-Wallis test) (Figure 15a-b).
71
Results & discussion
a)
b)
Figure 15 Stimulation with pertussis toxin (PT) (a) and phythohemagglutinin (PHA (b)
tended to induced a higher IFN- production in children with CT60 G/G haplotype.
Horizontal lines indicate median values and p-values of Mann-Whitney U-test are shown.
Note the different scales for the y-axis.
The increased IFN- production was seen as a response to mitogen and antigen
specific stimuli, confirming previous results by our group 99 and fitting with the
view of CTLA-4 down-regulating immune response in general and not as an
antigen-specific mechanism. This is in accordance with the idea of more
activated T cells in association with CTLA-4 polymorphisms 99, 100. However,
the secretion of IL-2 or IL-4 was not affected by any CTLA-4 polymorphism,
suggesting that the CTLA-4 SNPs investigated might regulate T cell inactivation
through other, more general, mechanisms than though AP-1, NF-κB and NFAT,
for instance by the disassociation of cSMAC 12 or in reducing the contact period
between HLA and TCR molecules 13. T helper 2 cells, prominent IL-4
producers, have higher levels of CTLA-4 compared to TH1 cells 202 and
72
Results & Discussion
decreased expression of CTLA-4 may thereby not influence the IL-4 production
as much as the IFN- production.
How may increased IFN- response be both beneficial and harmful, as children
with the HLA class II protective haplotypes DQB1*0602 (DR2-DQ6) and
DQA1*05-DQB1*0301 (DR5-DQ7) and also children with CTLA-4 SNP
associated with T1D risk were found to have higher IFN- secretion (Paper III)?
Enteroviruses have for a long time been associated with the development of T1D
134, 136, 142
and children with HLA class II risk haplotypes have been found to
have a decreased IFN- response to CVB4 146. Our result, with an increased
cytokine response in children with protective haplotypes, indicates a possible
contribution to protection from enteroviruses 192. However, the high IFN-
response with T1D associated CTLA-4 alleles may contribute to a higher
general activation of the immune system 13, 100 and thus be associated with T1D
pathogenesis.
The possible associations between SNPs in the ICOS gene and autoimmune
diseases made us investigate the CTIC154_1 C/T SNP.
Fiftyfive children (83.5%) in Paper III had the C/C genotype, nine (14,5%)
carried the C/T genotype and only two children (3%) had the T/T genotype.
Children with the T/T genotype did show a lower IL-6 response to PHA and a
higher IL-13 response to pertussis stimulation. This is interesting since
polymorphisms in the ICOS gene have been associated with TH2 cytokine
production 203. However, any clear conclusion could not be made since there
were only two individuals with T/T genotype.
Lymphoid Tyrosine Phosphatase is a protein strongly associated with negative
regulator of T cell activation and the SNP at position +1858 in the gene
PTPN22, encoding LYP, is associated with T1D 81, 82, 106. The +1858T allele
73
Results & discussion
gives a gain of function, which means that individuals with this allelic variant
would have an increased inhibition and a lower T cell response to antigenic
stimulation 107. In Paper III, 85.5% of the children had the C/C genotype and
14.5% had the T/C genotype. The T/T genotype is very rare and none of the
children carried this genotype.
The stimulated cytokine responses were similar in children with the C/C
genotype and the T/C genotype, although another study has shown reduced
IL-10 secretion in heterozygous individuals 108. Unfortunately, since none of the
children had the T/T genotype we could not investigate the full effect of this
SNP on cytokine secretion.
Immunological balance
T helper cells and regulatory T cells are important players in the immune
reactions and unbalance between these cellular subtypes could contribute to the
development of autoimmune reactions leading to a chronic autoimmune disease,
such as T1D. Paper II and IV discuss this topic through the examination of
general activity of TH1, TH2, TH17 and regulatory T cells, as well as the function
of regulatory T cells.
Children with an autoimmune process showed a decreased expression of
GATA-3 in unstimulated PBMC compared to healthy children without any risk
factors associated with T1D (p=0.014) and children with HLA risk haplotypes
alone (p=0.032), but the children with genetic risk alone did not differ from the
control children without genetic risk (Figure16).
74
Results & Discussion
Figure 16 The Relative transcription of GATA-3 was lower in children with autoantibodies
(Aab+) above 98th percentile for GADA and IAA or 99th percentile for IA-2A compared to
children without increased risk for T1D (Control) and children with HLA risk genotypes
(HLA risk). Horizontal lines indicate median values and p-values of Mann-Whitney U-test are
shown.
Inhibition of GATA-3 seems to be more important than increased levels of T-bet
for the production of IFN- 204, 205, and thus the low levels of GATA-3 could
support the development of TH1 immune response capable for destruction of cells. No differences in expression levels of IL-4Rα, T-bet or IL-12R were
observed, however (Table 6).
75
Results & discussion
Table 5 The relative transcription of markers associated with TH1 and TH2 in children without
increased risk for T1D (control), children with HLA risk genotypes associated with T1D
(HLA risk) and children with autoantibodies above 98th for GADA and IAA or 99th percentile
for IA-2A (aab+).
Control
HLA risk
Aab+
p-value
median (range)
median (range)
median (range)
(Kruskal-Wallis)
IL-4Rα
17.5 (9.0-31.7)
15.5 (9.6-39.1)
18.3 (7.3-44.4)
0.73
T-bet
5.4 (1.9-19.7)
7.1 (0.5-17.9)
2.9 (1.3-10.7)
0.24
IL-12Rβ1
15.8 (2.7-51.5)
17.1 (2.1-34.2)
15.8 (5.1-27.3)
0.86
IL-12Rβ2
3.9 (0.5-21.7)
2.9 (1.2-6.9)
3.0 (0.5-32.1)
0.98
In contrast to children at risk of T1D, diabetic children showed lower levels of
IL-12R1 (Figure 17a) and IL-4R (Figure 17b) mRNA at the diagnosis of the
disease, as well as in samples taken after three and 12 month duration, when
compared to the healthy control children.
a)
b)
Figure 17 The relative transcription of IL-12Rβ1 (a) and IL-4Rα (b) was lower in T1D
children, at all follow-up times, compared to healthy control children (control). Horizontal
lines indicate median values and p-values of Mann-Whitney U-test are shown. Note the
different scales for the y-axis.
76
Results & Discussion
The expression of T-bet was also significantly lower at the disease diagnosis
compared to controls with similar tendencies in later samples (Figure 18). This
decreased expression of several T cell polarization markers may reflect a general
exhaustion of the immune system at the time of clinical manifestation of T1D.
The decreased expression of IL-4Rα, T-bet and IL-12Rβ1 also supports previous
findings of decreased expression of IL-4 and IFN- in PBMC 171.
Figure 18 The relative transcription of T-bet was lower in T1D children ten days after
diagnose compared to healthy control children (control). Later time points showed the same
tendencies, although not significant. Horizontal lines indicate median values and p-values of
Mann-Whitney U-test are shown.
T helper 17 cells were recently discovered to be a subset of TH cells separate
from TH1 and TH2 cells. These TH17 cells have been associated with several
77
Results & discussion
autoimmune disorders 206-210 and they produce the pro-inflammatory cytokines
IL-17A, IL-17F and IL-22 38, 39. TH17 cells may also be important in the
development of T1D although the association with T1D have not been
confirmed yet. In unstimulated peripheral PBMC, the transcription of IL-17A
was low in general and there were no differences between healthy children,
children at risk or T1D children (data not shown).
Regulatory T cells are important in down-regulation of the immune responses
and controlling the autoreactive T cells present peripherally 211. Regulatory T
cells have been linked with the expression of CD25 48, CTLA-4 50, 51, Foxp3 55, 56
and ICOS 53 and the regulatory capacity of CD4+CD25high regulatory T cells has
been investigated in several different autoimmune diseases 172, 212-216 as well as in
allergy 217, 218.
We did not have the opportunity to do functional studies of regulatory T cells in
children with increased risk of T1D. However, the PBMC from children with
risk of T1D showed similar expression levels of the markers Foxp3, CTLA-4
and ICOS as was seen in healthy control children (Figure 19), suggesting a
comparable number of regulatory T cells in pre-diabetic children. Also in T1D
children, the expression of CTLA-4 and ICOS was similar as in healthy control
children (data not shown).
78
Results & Discussion
Figure 19 The relative transcription of markers associated with regulatory T cells did not
differ between children without any risk (open squares), children with HLA risk genotype
(filled squares) and children with autoantibodies above 98th percentile for GADA and IAA or
99th percentile for IA-2A (filled triangles). Horizontal lines indicate median values and pvalues of Kruskal-Wallis test are shown.
Foxp3 showed similar levels of expression in T1D children and in healthy
control children ten days after diagnosis (Paper II and IV) and three months after
diagnosis (Paper II). However, Foxp3 expression was decreased 12 months after
diagnosis of T1D when compared to healthy children (p=0.027, Paper II). The
Foxp3 expression was lower also in diabetic children 12 months after diagnosis
79
Results & discussion
when compared to the samples taken at diagnosis and at three months (p=0.038
and p=0.056 respectively, Paper II).
As the expression of Foxp3, CTLA-4 and ICOS has been shown to increase as a
response to insulin treatment in newly diagnosed T1D patients 219, this suggests
that the diabetic children have an activation of insulin-specific regulatory T cells
at diagnosis. This may be a reason why no differences were seen in these
markers between T1D children and control children at diagnosis; the insulin
treatment may correct the impaired regulatory function. This might also be a
reason of the decreased expression of IL-4Rα, T-bet and IL-12Rβ1 seen in Paper
II, since the regulatory compartment might be effective in diminishing the
immune response.
Investigating the proportion of CD4+CD25high T cells with flow cytometry
during 18 months follow-up after T1D diagnosis revealed no differences
between diabetic and healthy children at any time point (Paper IV) 54, 212, 213, 220,
221
and the amount of CD4+CD25high T cell expressing extracellular or
intracellular CTLA-4 was similar in both groups. However, the diabetic children
had more intracellular CTLA-4 per CD4+CD25high T cell represented as the MFI
of intracellular CTLA-4, than the healthy children. This was seen at ten days
after diagnosis (Figure 20) and may reflect an ongoing activation of regulatory T
cells during T1D debut. No significant differences were observed in the
expression level of intracellular CTLA-4 during the follow-up among T1D
children, however. A general activation of the immune system was also seen as
increased levels of serum CRP at ten days and three months after diagnosis
(Paper IV). C-reactive protein is induced by IL-6, which is secreted in great
quantity from the innate immune system, and activates macrophages and the
complement system. The increased levels of CRP might reflect the
inflammatory process taking place in pancreas.
80
Results & Discussion
Figure 20 The T1D children had higher expression of intracellular CTLA-4 per
CD4+CD25high T cell than the healthy control children ten days after diagnosis. The horizontal
line indicates median value and p-values of Mann-Whitney U-test are shown.
Interestingly, a negative correlation between insulin dose and the level of Foxp3
was seen at 3 months after diagnosis (R=-0.541; p=0.030, Paper II). This could
support the activation of insulin-specific regulatory T cells and indicates that
activation of regulatory T cells near diagnosis may control the -cell destruction
and thus be related to the insulin dose needed. The activation of T effector cells
increase the Foxp3 expression 62-64, trying to diminish the immune activation 65.
The loss of Foxp3 expression from diagnosis toward 12 month duration (Paper
II) might be a result of diminishing regulatory activity during the fading
autoimmune attack with final loss of β-cells.
A negative correlation of IFN- expression with the production of C-peptide was
observed ten days after diagnosis (r=-0.676, p= 0.004, Paper IV), suggesting that
a more pronounced inflammation may lead to a more rapid loss of -cells and
insulin production.
81
Results & discussion
The effect of regulatory T cells on effector cells was investigated using
depletion of CD4+CD25high T cells. Real-time PCR showed that expression of
Foxp3 was significantly higher in PBMC than in CD25depleted PBMC (Paper
IV), which is reasonable due to the expression of Foxp3 in CD4+CD25high cells.
We also saw a higher expression IL-10 in PBMC compared to CD25depleted
PBMC (Paper IV) and the spontaneous secretion of TGF-β in PBMC was also
higher than in CD25depleted PBMC (Paper IV), although this difference could
not be seen at mRNA level. Transforming growth factor- and IL-10 are
cytokines known to be involved in regulation of immune response and TGF-
has been shown to up-regulate the expression of Foxp3 for the induction iTreg
222
and maintaining suppressor function in nTreg cells 59. Interleukin-10 has been
shown to be important for the induction of the Tr1 cells 223, the regulatory cells
which mediate their suppression by local secretion of IL-10 and TGF-β 224. A
higher production of TGF-β and IL-10 in PBMC than in CD25depleted PBMC
suggests that these cytokines are produced by CD4+CD25high T cells 213, 225. The
results verify that the depletion of regulatory T cells worked using our method
and since the same phenomenom was seen both among T1D and healthy
children the results suggest a normal functional activity of CD4+CD25high
regulatory T cells in T1D 220.
Stimulating PBMC and CD25depleted PBMC with PHA resulted in a more
pronounced proliferative response among CD25depleted PBMC than in whole
PBMC in diabetic children (median SI 25.2 and 13.6, respectively, p= 0.009).
Also the spontaneous expression of IFN- was higher in the CD25depleted
PBMC as compared to whole PBMC population (Paper IV) in the diabetic
children at ten days after diagnosis. Accordingly, the presence of CD4+CD25high
regulatory T cells in PBMC population inhibited proliferation response of the
cells and expression of IFN-. Although we did not find any significant
82
Results & Discussion
difference in the proliferation response of CD25depleted PBMC and PBMC in
the healthy children this might be due to rather small numbers of healthy
children studied since the median SI was somewhat higher in CD25depleted
PBMC compared to PBMC in healthy children (median SI 22.4 and 20.5,
respectively). We did not find any difference in SI between diabetic and healthy
children (data not shown), supporting a normal function of the regulatory T cells
among T1D children.
83
Summary & concluding remarks
SUMMARY AND CONCLUDING REMARKS
In this thesis the association between environmental factors and β-cell
autoimmunity was evaluated, as well as the relationship between possible
immunological aberrancies and increased risk for T1D in children was
elucidated. Different environmental factors, as well as immune dysregulation
have been proposed to interact, triggering an autoimmune response leading to
overt T1D. The mechanism behind this is still largely unknown and maybe some
general immunological mechanisms are missing in children that will develop
T1D, making them more susceptible for the disease.
Several environmental factors such as early nutritional factors and early
infections may induce islet autoantibodies in a general population, but the
mechanisms of the induction are still elusive. It should be emphasised that the
induction of autoantibodies, especially single autoantibodies, may not always
lead to the development of manifest T1D and other factors, protective or
triggering, have to be involved as well. The results in this thesis further showed
that responses to antigens was dependent on HLA genotypes presenting antigens
and that down regulation of immune response was controlled in part of CTLA-4.
The T1D associated HLA haplotypes and different CTLA-4 polymorphisms
may thus be associated with T1D pathogenesis.
The findings do not support a systemic activation of the immune system in
children at risk of T1D. Instead there is a decrease in TH1 and TH2 markers after
T1D manifestation, possibly as a result of activated regulatory mechanisms
trying to dampen the autoimmune reaction taking place. The studies performed
did not reveal impaired function of the peripheral blood derived regulatory T
cells in T1D, but rather activated and functional compartment of CD4+CD25high
regulatory T cells, supporting the theory of regulatory T cells suppressing an
ongoing inflammation.
85
Results & discussion
In summary, several genetic and environmental factors seem to contribute to
T1D and this thesis emphasizes the importance of multiple mechanisms acting
in concert. The results raise the question whether the immunological enigma of
T1D could be revealed by studies of peripheral blood.
86
Acknowledgement
ACKNOWLEDGEMENTS
I am very grateful to everyone, colleagues, family and friends, who have helped
me to complete this thesis and made a dream come true. Without all of you this
would never have happened.
I wish to thank you for all the support, enthusiasm and the fun we have had
during these years. In particular I wish to thank:
All children and their parents, who decided to be a part of my studies and to
contribute to the understanding of T1D.
Outi Vaarala, my main supervisor, who always believed in me, even when my
own beliefs stumble. Thanks for all your enthusiasm during our meetings and
making the work of science feel so fun.
Johnny Ludvigsson, my second supervisor, who believed in my ability to
become a scientist and always being so optimistic and enthusiastic.
Malin Fagerås Böttcher, my first contact to the world of science and whom I
admire. You have always been there, with questions pushing my research
forward, with answers to my endless questions and with laughter at “fika”-time.
Thank you for all your inspiration, I would like to be like you when I grow up!
Sara Tomičić, my dearest friend, who always is there for me. Thank you for
your endless encouragements, having answers to all my stupid questions and for
sharing all your scientific knowledge. Thank you also for all the fun outside of
work with our families, travelling, “vinprovningar” and sleepovers.
My collaborators in different projects, Anna Lundberg, Jeanette Walhberg,
Jarno Honkanen, Jorma Ilonen and Merja Roivainen, for your invaluable
knowledge you shared with me and for valuable comments on manuscripts.
Anki Gilmore-Ellis, for all your help during so many years and for always have
something nice to say. We could not manage without you!
Eva Isaksson, Ann-Marie Sandström, Lena Hanberger, for help with
collecting blood samples and introducing me to the children behind the samples.
Iris Franzén and Christina Larsson for the organization around the ABISproject and your kindness and Gunilla Hallström for recruiting healthy
schoolchildren.
87
Acknowledgement
Lena Berglert, Ammi Fornander and Florence Sjögren for teaching me
methods used in my work, for never ending support when I gore with technical
equipment and for making me feel proud of our profession.
Ingela Johansson, Gosia Smolinska-Konefal and Cecilia Runnqvist for skilful
technical assistance with the autoantibody assessment and for all the fun in the
laboratory.
Further, I wish to thank Hanna Holmberg, Kristina Warstedt, Camilla
Janefjord, Anne Lahdenperä and Anna Kivling, as well as Sara and Anna L.,
for your friendship and sharing the ups and downs in research. You are all dear
to me and I will never forget the fun we have had during the years. The
invaluable discussions we have had regarding our own research as well as
methodological issues, but also sharing frustration over supervisors or rejections
from journals.
All friends and colleagues at Division at Pediatric and the unit of Clinical and
Experimental Research, my “roomies” during the years, Hanna, Kristina, Maria
Faresjö and Maria Jenmalm, for sharing your knowledge whenever you can, to
fellow PhD students and to Kerstin Hagersten, Håkan Wiktander, Pia Karlsson
and Irene Cavalli-Björkman for always being helpful.
To all my friends outside the scientific world who are very important to me,
always making me feel satisfied with my life and who includes me and my
family into yours. Especially I would like to thank “Strömmingarna” and
“vinprovarna” for all discussions about different facts in life, for unforgettable
dinners, wonderful holidays and a lot of laughter.
Thank you all for sharing my life at different time points, and for
helping me grow as a scientist and human being. I love you all!
****************************************************************
Till sist vill jag tacka min familj
Mina föräldrar, Gun och Roland, som alltid funnits där och bistått mig, vad jag
än har gett mig in på. Ni är helt fantastiska och utan er hade jag aldrig kommit så
här långt. Tack för allt!
Till min bror Magnus som stöttat mig hela livet och som varit stolt över sin
syster. Tack också till min svägerska Therese och brorsdotter Stella, som tar
hand om min bror och får honom att må bra.
88
Acknowledgement
Till mormor Lola och morfar Ragnar, ni är helt underbara som med ert
skojfriska humör alltid får mig på gott humör. Jag älskar er och hoppas ni får
vara med oss i många år till.
Familjen Walldén, Roland, Marianne, Sofia och Fredrik, som från första dagen
tog emot mig med öppna armar och inspirerat mig till att följa sina drömmar.
Gurra, du har hela tiden trott på min förmåga att klara av detta och har funnits
vid min sida i både vått och torrt, när jag varit som mest nedstämd och under
den lyckligaste tiden i mitt liv. Du är den klippa jag kan luta mig mot när
världen gungar, jag älskar dig så mycket!
Molly, du är det bästa som har hänt mamma och med dig finns det inte en enda
tråkig minut. Med dig hemma kan jag koppla bort jobbet en stund och bara vara.
Jag älskar dig så otroligt mycket gumman. Jag hoppas du får fortsätta sjunga,
dansa och vara glad hela livet! Puss och Kram på dig!
This work was generously supported by JDRF-Wallenberg foundation, the
Swedish Medical Research Council (MFR, Vetenskapsrådet), the Swedish Child
Diabetes Foundation (Barndiabetesfonden), the Swedish Diabetes Association,
the Söderberg Foundation, the Novo Nordisk Foundation, the Medical Research
Found of the country of Östergötland, the Swedish Samariten Foundation
(Stiftelsen Samariten), the Lions Foundation and the Jerring Foundation
(Jerringfonden).
89
References
REFERENCES
1.
2.
3.
4.
5.
6.
7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
Janeway, C., et al., Immunobiology: the immune system in health and
disease. 6th ed. ed. 2005, London: Garland Science Publishing.
Dustin, M.L. and A.S. Shaw, Costimulation: building an immunological
synapse. Science, 1999. 283(5402): p. 649-50.
Lin, J., M.J. Miller, and A.S. Shaw, The c-SMAC: sorting it all out (or in).
Journal of Cell Biology, 2005. 170(2): p. 177-82.
Monks, C.R., et al., Three-dimensional segregation of supramolecular
activation clusters in T cells. Nature, 1998. 395(6697): p. 82-6.
Viola, A. and A. Lanzavecchia, T cell activation determined by T cell
receptor number and tunable thresholds. Science, 1996. 273(5271): p.
104-6.
Van Parijs, L. and A.K. Abbas, Homeostasis and self-tolerance in the
immune system: turning lymphocytes off. Science, 1998. 280(5361): p.
243-8.
Perkins, D., et al., Regulation of CTLA-4 expression during T cell
activation. Journal of Immunology, 1996. 156(11): p. 4154-9.
Wang, X.B., et al., Regulation of surface and intracellular expression of
CTLA-4 on human peripheral T cells. Scandinavian Journal of
Immunology, 2001. 54(5): p. 453-8.
Collins, A.V., et al., The interaction properties of costimulatory molecules
revisited. Immunity, 2002. 17(2): p. 201-10.
Krummel, M.F. and J.P. Allison, CTLA-4 engagement inhibits IL-2
accumulation and cell cycle progression upon activation of resting T
cells. Journal of Experimental Medicine, 1996. 183: p. 2533-2540.
Fraser, J.H., et al., CTLA4 ligation attenuates AP-1, NFAT and NFkappaB activity in activated T cells. European Journal of Immunology,
1999. 29(3): p. 838-44.
Martin, M., et al., Cytotoxic T lymphocyte antigen 4 and CD28 modulate
cell surface raft expression in their regulation of T cell function. Journal
of Experimental Medicine, 2001. 194(11): p. 1675-81.
Schneider, H., et al., Reversal of the TCR stop signal by CTLA-4. Science,
2006. 313(5795): p. 1972-5.
Bluestone, J.A. and A.K. Abbas, Natural versus adaptive regulatory T
cells. Nature Reviews Immunology, 2003. 3(3): p. 253-7.
Thompson, C. and F. Powrie, Regulatory T cells. Current Opinion in
Pharmacology, 2004. 4(4): p. 408-14.
Taylor, A., et al., Mechanisms of immune suppression by interleukin-10
and transforming growth factor-beta: the role of T regulatory cells.
Immunology, 2006. 117(4): p. 433-42.
91
References
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
92
Weiner, H.L., Induction and mechanism of action of transforming growth
factor-beta-secreting Th3 regulatory cells. Immunological Reviews, 2001.
182: p. 207-14.
Thornton, A.M. and E.M. Shevach, CD4+CD25+ immunoregulatory T
cells suppress polyclonal T cell activation in vitro by inhibiting interleukin
2 production. Journal of Experimental Medicine, 1998. 188(2): p. 287-96.
Paust, S., et al., Engagement of B7 on effector T cells by regulatory T cells
prevents autoimmune disease. Proceedings of the National Academy of
Sciences of the United States of America, 2004. 101(28): p. 10398-403.
von Boehmer, H., Mechanisms of suppression by suppressor T cells.
Nature Immunology, 2005. 6(4): p. 338-44.
Collison, L.W., et al., The inhibitory cytokine IL-35 contributes to
regulatory T-cell function. Nature, 2007. 450(7169): p. 566-9.
Mosmann, T.R., et al., Two types of murine helper T cell clone. I.
Definition according to profiles of lymphokine activities and secreted
proteins. Journal of Immunology, 1986. 136(7): p. 2348-57.
Del Prete, G.F., et al., Purified protein derivative of Mycobacterium
tuberculosis and excretory-secretory antigen(s) of Toxocara canis expand
in vitro human T cells with stable and opposite (type 1 T helper or type 2
T helper) profile of cytokine production. Journal of Clinical Investigation,
1991. 88(1): p. 346-50.
Kurts, C., Th17 cells: a third subset of CD4+ T effector cells involved in
organ-specific autoimmunity. Nephrology, Dialysis, Transplantation,
2008. 23(3): p. 816-9.
Hsieh, C.S., et al., Development of TH1 CD4+ T cells through IL-12
produced by Listeria-induced macrophages. Science, 1993. 260(5107): p.
547-9.
Presky, D.H., et al., A functional interleukin 12 receptor complex is
composed of two beta-type cytokine receptor subunits. Proceedings of the
National Academy of Sciences of the United States of America, 1996.
93(24): p. 14002-7.
Afkarian, M., et al., T-bet is a STAT1-induced regulator of IL-12R
expression in naive CD4+ T cells. Nature Immunology, 2002. 3(6): p.
549-57.
Mullen, A.C., et al., Role of T-bet in commitment of TH1 cells before IL12-dependent selection. Science, 2001. 292(5523): p. 1907-10.
Szabo, S.J., et al., Distinct effects of T-bet in TH1 lineage commitment and
IFN-gamma production in CD4 and CD8 T cells. Science, 2002.
295(5553): p. 338-42.
Rogge, L., et al., Selective expression of an interleukin-12 receptor
component by human T helper 1 cells. Journal of Experimental Medicine,
1997. 185(5): p. 825-31.
References
31.
32.
33.
34.
35.
36.
37.
38.
39.
40.
41.
42.
43.
44.
Becskei, A. and M.J. Grusby, Contribution of IL-12R mediated feedback
loop to Th1 cell differentiation. FEBS Letters, 2007. 581(27): p. 5199206.
Lametschwandtner, G., et al., Sustained T-bet expression confers
polarized human TH2 cells with TH1-like cytokine production and
migratory capacities. Journal of Allergy and Clinical Immunology, 2004.
113(5): p. 987-94.
Kurata, H., et al., Ectopic expression of activated Stat6 induces the
expression of Th2-specific cytokines and transcription factors in
developing Th1 cells. Immunity, 1999. 11(6): p. 677-88.
Zheng, W. and R.A. Flavell, The transcription factor GATA-3 is
necessary and sufficient for Th2 cytokine gene expression in CD4 T cells.
Cell, 1997. 89(4): p. 587-96.
Zhang, D.H., L. Yang, and A. Ray, Differential responsiveness of the IL-5
and IL-4 genes to transcription factor GATA-3. Journal of Immunology,
1998. 161(8): p. 3817-21.
Kim, J.I., et al., The transcription factor c-Maf controls the production of
interleukin-4 but not other Th2 cytokines. Immunity, 1999. 10(6): p. 74551.
Ferber, I.A., et al., GATA-3 significantly downregulates IFN-gamma
production from developing Th1 cells in addition to inducing IL-4 and IL5 levels. Clinical Immunology, 1999. 91(2): p. 134-44.
Aggarwal, S., et al., Interleukin-23 promotes a distinct CD4 T cell
activation state characterized by the production of interleukin-17. Journal
of Biological Chemistry, 2003. 278(3): p. 1910-4.
Zheng, Y., et al., Interleukin-22, a T(H)17 cytokine, mediates IL-23induced dermal inflammation and acanthosis. Nature, 2007. 445(7128): p.
648-51.
Volpe, E., et al., A critical function for transforming growth factor-beta,
interleukin 23 and proinflammatory cytokines in driving and modulating
human T(H)-17 responses. Nature Immunology, 2008. 9(6): p. 650-7.
Harrington, L.E., et al., Interleukin 17-producing CD4+ effector T cells
develop via a lineage distinct from the T helper type 1 and 2 lineages.
Nature Immunology, 2005. 6(11): p. 1123-32.
Park, H., et al., A distinct lineage of CD4 T cells regulates tissue
inflammation by producing interleukin 17. Nature Immunology, 2005.
6(11): p. 1133-41.
Wynn, T.A., T(H)-17: a giant step from T(H)1 and T(H)2. Nature
Immunology, 2005. 6(11): p. 1069-70.
Wilson, N.J., et al., Development, cytokine profile and function of human
interleukin 17-producing helper T cells. Nature Immunology, 2007. 8(9):
p. 950-7.
93
References
45.
46.
47.
48.
49.
50.
51.
52.
53.
54.
55.
56.
57.
58.
94
Mangan, P.R., et al., Transforming growth factor-beta induces
development of the T(H)17 lineage. Nature, 2006. 441(7090): p. 231-4.
Manel, N., D. Unutmaz, and D.R. Littman, The differentiation of human
T(H)-17 cells requires transforming growth factor-beta and induction of
the nuclear receptor RORgammat. Nature Immunology, 2008. 9(6): p.
641-9.
Ivanov, II, et al., The orphan nuclear receptor RORgammat directs the
differentiation program of proinflammatory IL-17+ T helper cells. Cell,
2006. 126(6): p. 1121-33.
Sakaguchi, S., et al., Immunologic self-tolerance maintained by activated
T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a
single mechanism of self-tolerance causes various autoimmune diseases.
Journal of Immunology, 1995. 155(3): p. 1151-64.
Dieckmann, D., et al., Ex vivo isolation and characterization of
CD4+CD25+ T cells with regulatory properties from human blood.
Journal of Experimental Medicine, 2001. 193(11): p. 1303-1310.
Read, S., V. Malmstrom, and F. Powrie, Cytotoxic T lymphocyteassociated antigen 4 plays an essential role in the function of
CD25(+)CD4(+) regulatory cells that control intestinal inflammation.
Journal of Experimental Medicine, 2000. 192(2): p. 295-302.
Jonuleit, H., et al., Identification and functional characterization of human
CD4(+)CD25(+) T cells with regulatory properties isolated from
peripheral blood. Journal of Experimental Medicine, 2001. 193(11): p.
1285-94.
McHugh, R.S., et al., CD4(+)CD25(+) immunoregulatory T cells: gene
expression analysis reveals a functional role for the glucocorticoidinduced TNF receptor. Immunity, 2002. 16(2): p. 311-23.
Herman, A.E., et al., CD4+CD25+ T regulatory cells dependent on ICOS
promote regulation of effector cells in the prediabetic lesion. Journal of
Experimental Medicine, 2004. 199(11): p. 1479-89.
Liu, W., et al., CD127 expression inversely correlates with FoxP3 and
suppressive function of human CD4+ T reg cells. Journal of Experimental
Medicine, 2006. 203(7): p. 1701-11.
Hori, S., T. Nomura, and S. Sakaguchi, Control of regulatory T cell
development by the transcription factor Foxp3. Science, 2003. 299(5609):
p. 1057-61.
Khattri, R., et al., An essential role for Scurfin in CD4+CD25+ T
regulatory cells. Nature Immunology, 2003. 4(4): p. 337-42.
Antony, P.A., et al., Interleukin-2-dependent mechanisms of tolerance and
immunity in vivo. Journal of Immunology, 2006. 176(9): p. 5255-66.
Passerini, L., et al., STAT5-signaling cytokines regulate the expression of
FOXP3 in CD4+CD25+ regulatory T cells and CD4+CD25- effector T
cells. International Immunology, 2008. 20(3): p. 421-31.
References
59.
60.
61.
62.
63.
64.
65.
66.
67.
68.
69.
70.
71.
72.
73.
Marie, J.C., et al., TGF-beta1 maintains suppressor function and Foxp3
expression in CD4+CD25+ regulatory T cells. Journal of Experimental
Medicine, 2005. 201(7): p. 1061-7.
Fontenot, J.D., M.A. Gavin, and A.Y. Rudensky, Foxp3 programs the
development and function of CD4+CD25+ regulatory T cells. Nature
Immunology, 2003. 4(4): p. 330-6.
Bacchetta, R., et al., Defective regulatory and effector T cell functions in
patients with FOXP3 mutations. Journal of Clinical Investigation, 2006.
116(6): p. 1713-22.
Wang, J., et al., Transient expression of FOXP3 in human activated
nonregulatory CD4+ T cells. European Journal of Immunology, 2007.
37(1): p. 129-38.
Morgan, M.E., et al., Expression of FOXP3 mRNA is not confined to
CD4+CD25+ T regulatory cells in humans. Human Immunology, 2005.
66(1): p. 13-20.
Walker, M.R., et al., Induction of FoxP3 and acquisition of T regulatory
activity by stimulated human CD4+CD25- T cells. Journal of Clinical
Investigation, 2003. 112(9): p. 1437-43.
Ziegler, S.F., FOXP3: not just for regulatory T cells anymore. European
Journal of Immunology, 2007. 37(1): p. 21-3.
Mantel, P.Y., et al., Molecular mechanisms underlying FOXP3 induction
in human T cells. Journal of Immunology, 2006. 176(6): p. 3593-602.
WHO, World Health Organization: Definition, Diagnosis and
Classification of Diabetes Mellitus and it's complications: Report of a
WHO consultation Part 1: Diagnosis and Classifications of Diabetes
Mellitus. Geneva, World Health Org. 1999.
Mayer-Davis, E.J., Type 2 diabetes in youth: epidemiology and current
research toward prevention and treatment. Journal of the American
Dietetic Association, 2008. 108(4 Suppl 1): p. S45-51.
ADA, Report of the Expert Committee on the Diagnosis and
Classification of Diabetes Mellitus. Diabetes Care, 1997. 20(7): p. 118397.
DIAMOND, Incidence and trends of childhood Type 1 diabetes
worldwide 1990-1999. Diabetic Medicine, 2006. 23(8): p. 857-66.
Podar, T., et al., Increasing incidence of childhood-onset type I diabetes in
3 Baltic countries and Finland 1983-1998. Diabetologia, 2001. 44 Suppl
3: p. B17-20.
Harjutsalo, V., L. Sjoberg, and J. Tuomilehto, Time trends in the
incidence of type 1 diabetes in Finnish children: a cohort study. Lancet,
2008. 371(9626): p. 1777-82.
Pundziute-Lycka, A., et al., The incidence of Type I diabetes has not
increased but shifted to a younger age at diagnosis in the 0-34 years
group in Sweden 1983-1998. Diabetologia, 2002. 45(6): p. 783-91.
95
References
74.
75.
76.
77.
78.
79.
80.
81.
82.
83.
84.
85.
86.
87.
88.
96
Genome-wide association study of 14,000 cases of seven common diseases
and 3,000 shared controls. Nature, 2007. 447(7145): p. 661-78.
Akerblom, H.K., et al., Environmental factors in the etiology of type 1
diabetes. American Journal of Medical Genetics, 2002. 115(1): p. 18-29.
Atkinson, M.A. and G.S. Eisenbarth, Type 1 diabetes: new perspectives
on disease pathogenesis and treatment. Lancet, 2001. 358(9277): p. 2219.
Davies, J.L., et al., A genome-wide search for human type 1 diabetes
susceptibility genes. Nature, 1994. 371(6493): p. 130-6.
Kelly, M.A., C.H. Mijovic, and A.H. Barnett, Genetics of type 1 diabetes.
Best Practice & Research Clinical Endocrinology and Metabolism, 2001.
15(3): p. 279-91.
Nisticò, L., et al., The CTLA4 gene region of chromosome 2q33 is linked
to, and associated with, type 1 diabetes. Human Molecular Genetics,
1996. 5(7): p. 1075-1080.
Ueda, H., et al., Association of the T-cell regulatory gene CTLA4 with
susceptibility to autoimmune disease. Nature, 2003. 423(6939): p. 506-11.
Bottini, N., et al., A functional variant of lymphoid tyrosine phosphatase is
associated with type I diabetes. Nature Genetics, 2004. 36(4): p. 337-8.
Ladner, M.B., et al., Association of the single nucleotide polymorphism
C1858T of the PTPN22 gene with type 1 diabetes. Human Immunology,
2005. 66(1): p. 60-4.
Singal, D.P. and M.A. Blajchman, Histocompatibility (HL-A) antigens,
lymphocytotoxic antibodies and tissue antibodies in patients with diabetes
mellitus. Diabetes, 1973. 22(6): p. 429-32.
Todd, J.A., et al., The A3 allele of the HLA-DQA1 locus is associated with
susceptibility to type 1 diabetes in Japanese. Proceedings of the National
Academy of Sciences of the United States of America, 1990. 87(3): p.
1094-8.
Cucca, F., et al., The distribution of DR4 haplotypes in Sardinia suggests
a primary association of type I diabetes with DRB1 and DQB1 loci.
Human Immunology, 1995. 43(4): p. 301-8.
Huang, H.S., et al., HLA-encoded susceptibility to insulin-dependent
diabetes mellitus is determined by DR and DQ genes as well as their
linkage disequilibria in a Chinese population. Human Immunology, 1995.
44(4): p. 210-9.
Thomson, G., et al., Relative predispositional effects of HLA class II
DRB1-DQB1 haplotypes and genotypes on type 1 diabetes: a metaanalysis. Tissue Antigens, 2007. 70(2): p. 110-27.
Hermann, R., et al., The effect of HLA class II, insulin and CTLA4 gene
regions on the development of humoral beta cell autoimmunity.
Diabetologia, 2005. 48(9): p. 1766-75.
References
89.
Gullstrand, C., et al., Progression to type 1 diabetes and autoantibody
positivity in relation to HLA-risk genotypes in children participating in
the ABIS study. Pediatric Diabetes, 2008. 9(3 Pt 1): p. 182-90.
90. Faresjo, M.K., et al., Diminished IFN-gamma response to diabetesassociated autoantigens in children at diagnosis and during follow up of
type 1 diabetes. Diabetes/Metabolism Research and Reviews, 2006. 22(6):
p. 462-70.
91. Petrovsky, N. and L.C. Harrison, HLA class II-associated polymorphism
of interferon-gamma production. Implications for HLA-disease
association. Human Immunology, 1997. 53(1): p. 12-6.
92. Luopajarvi, K., et al., Reduced CCR4, interleukin-13 and GATA-3 upregulation in response to type 2 cytokines of cord blood T lymphocytes in
infants at genetic risk of type 1 diabetes. Immunology, 2007. 121(2): p.
189-96.
93. Dariavach, P., et al., Human Ig superfamily CTLA-4 gene: chromosomal
localization and identity of protein sequence between murine and human
CTLA-4 cytoplasmic domains. European Journal of Immunology, 1988.
18(12): p. 1901-5.
94. Anjos, S.M., M.C. Tessier, and C. Polychronakos, Association of the
cytotoxic T lymphocyte-associated antigen 4 gene with type 1 diabetes:
evidence for independent effects of two polymorphisms on the same
haplotype block. Journal of Clinical Endocrinology and Metabolism,
2004. 89(12): p. 6257-65.
95. Haller, K., et al., Type 1 diabetes is insulin -2221 MspI and CTLA-4 +49
A/G polymorphism dependent. European Journal of Clinical Investigation,
2004. 34(8): p. 543-8.
96. Donner, H., et al., CTLA4 alanine-17 confers genetic susceptibility to
Graves' disease and to type 1 diabetes mellitus. Journal of Clinical
Endocrinology and Metabolism, 1997. 82(1): p. 143-6.
97. Mayans, S., et al., CT60 genotype does not affect CTLA-4 isoform
expression despite association to T1D and AITD in northern Sweden.
BMC Medical Genetics, 2007. 8: p. 3.
98. Blomhoff, A., et al., Polymorphisms in the cytotoxic T lymphocyte
antigen-4 gene region confer susceptibility to Addison's disease. Journal
of Clinical Endocrinology and Metabolism, 2004. 89(7): p. 3474-6.
99. Jonson, C.O., et al., The importance of CTLA-4 polymorphism and human
leukocyte antigen genotype for the induction of diabetes-associated
cytokine response in healthy school children. Pediatric Diabetes, 2007.
8(4): p. 185-92.
100. Maurer, M., et al., A polymorphism in the human cytotoxic T-lymphocyte
antigen 4 ( CTLA4) gene (exon 1 +49) alters T-cell activation.
Immunogenetics, 2002. 54(1): p. 1-8.
97
References
101. Ligers, A., et al., CTLA-4 gene expression is influenced by promoter and
exon 1 polymorphisms. Genes and Immunity, 2001. 2(3): p. 145-52.
102. Kaartinen, T., et al., Genetic variation in ICOS regulates mRNA levels of
ICOS and splicing isoforms of CTLA4. Molecular Immunology, 2007.
44(7): p. 1644-51.
103. Maier, L.M., et al., Allelic variant in CTLA4 alters T cell phosphorylation
patterns. Proceedings of the National Academy of Sciences of the United
States of America, 2007. 104(47): p. 18607-12.
104. Hutloff, A., et al., ICOS is an inducible T-cell co-stimulator structurally
and functionally related to CD28. Nature, 1999. 397(6716): p. 263-6.
105. Haimila, K.E., J.A. Partanen, and P.M. Holopainen, Genetic
polymorphism of the human ICOS gene. Immunogenetics, 2002. 53(12):
p. 1028-32.
106. Smyth, D., et al., Replication of an association between the lymphoid
tyrosine phosphatase locus (LYP/PTPN22) with type 1 diabetes, and
evidence for its role as a general autoimmunity locus. Diabetes, 2004.
53(11): p. 3020-3.
107. Vang, T., et al., Protein tyrosine phosphatase PTPN22 in human
autoimmunity. Autoimmunity, 2007. 40(6): p. 453-61.
108. Rieck, M., et al., Genetic variation in PTPN22 corresponds to altered
function of T and B lymphocytes. Journal of Immunology, 2007. 179(7): p.
4704-10.
109. Taplin, C.E. and J.M. Barker, Autoantibodies in type 1 diabetes.
Autoimmunity, 2008. 41(1): p. 11-8.
110. Palmer, J.P., et al., Insulin antibodies in insulin-dependent diabetics
before insulin treatment. Science, 1983. 222(4630): p. 1337-9.
111. Ziegler, A.G., et al., Autoantibody appearance and risk for development of
childhood diabetes in offspring of parents with type 1 diabetes: the 2-year
analysis of the German BABYDIAB Study. Diabetes, 1999. 48(3): p. 4608.
112. Holmberg, H., et al., Higher prevalence of autoantibodies to insulin and
GAD65 in Swedish compared to Lithuanian children with type 1 diabetes.
Diabetes Research and Clinical Practice, 2006. 72(3): p. 308-14.
113. Borg, H., et al., Insulin autoantibodies are of less value compared with
islet antibodies in the clinical diagnosis of autoimmune type 1 diabetes in
children older than 3 yr of age. Pediatric Diabetes, 2002. 3(3): p. 149-54.
114. Williams, A.J., et al., The prevalence of insulin autoantibodies at the
onset of Type 1 diabetes is higher in males than females during
adolescence. Diabetologia, 2003. 46(10): p. 1354-6.
115. LaGasse, J.M., et al., Successful prospective prediction of type 1 diabetes
in schoolchildren through multiple defined autoantibodies: an 8-year
follow-up of the Washington State Diabetes Prediction Study. Diabetes
Care, 2002. 25(3): p. 505-11.
98
References
116. Strebelow, M., et al., Karlsburg Type I diabetes risk study of a general
population: frequencies and interactions of the four major Type I
diabetes-associated autoantibodies studied in 9419 schoolchildren.
Diabetologia, 1999. 42(6): p. 661-70.
117. Baekkeskov, S., et al., Autoantibodies in newly diagnosed diabetic
children immunoprecipitate human pancreatic islet cell proteins. Nature,
1982. 298(5870): p. 167-9.
118. Baekkeskov, S., et al., Antibodies to a 64,000 Mr human islet cell antigen
precede the clinical onset of insulin-dependent diabetes. Journal of
Clinical Investigation, 1987. 79(3): p. 926-34.
119. Baekkeskov, S., et al., Identification of the 64K autoantigen in insulindependent diabetes as the GABA-synthesizing enzyme glutamic acid
decarboxylase. Nature, 1990. 347(6289): p. 151-6.
120. Leslie, D.G., M.A. Atikinson, and A.L. Notkins, Autoantigens IA-2 and
GAD in type 1 (insulin-dependent) diabetes. Diabetologia, 1999. 42: p. 314.
121. Hagopian, W.A., et al., Autoantibodies in IDDM primarily recognize the
65,000-M(r) rather than the 67,000-M(r) isoform of glutamic acid
decarboxylase. Diabetes, 1993. 42(4): p. 631-6.
122. Bonifacio, E., et al., Islet autoantibody markers in IDDM: risk assessment
strategies yielding high sensitivity. Diabetologia, 1995. 38(7): p. 816-22.
123. Marciulionyte, D., et al., A comparison of the prevalence of islet
autoantibodies in children from two countries with differing incidence of
diabetes. Diabetologia, 2001. 44(1): p. 16-21.
124. Payton, M.A., C.J. Hawkes, and M.R. Christie, Relationship of the
37,000- and 40,000-M(r) tryptic fragments of islet antigens in insulindependent diabetes to the protein tyrosine phosphatase-like molecule IA-2
(ICA512). Journal of Clinical Investigation, 1995. 96(3): p. 1506-11.
125. Bonifacio, E., et al., Identification of protein tyrosine phosphatase-like
IA2 (islet cell antigen 512) as the insulin-dependent diabetes-related
37/40K autoantigen and a target of islet-cell antibodies. Journal of
Immunology, 1995. 155(11): p. 5419-26.
126. Lu, J., et al., Identification of a second transmembrane protein tyrosine
phosphatase, IA-2beta, as an autoantigen in insulin-dependent diabetes
mellitus: precursor of the 37-kDa tryptic fragment. Proceedings of the
National Academy of Sciences of the United States of America, 1996.
93(6): p. 2307-11.
127. Notkins, A.L., et al., IA-2 and IA-2 beta are major autoantigens in IDDM
and the precursors of the 40 kDa and 37 kDa tryptic fragments. Journal of
Autoimmunity, 1996. 9(5): p. 677-82.
128. Kawasaki, E., et al., Evaluation of islet cell antigen (ICA) 512/IA-2
autoantibody radioassays using overlapping ICA512/IA-2 constructs.
99
References
129.
130.
131.
132.
133.
134.
135.
136.
137.
138.
139.
140.
141.
142.
100
Journal of Clinical Endocrinology and Metabolism, 1997. 82(2): p. 37580.
Redondo, M.J., et al., Genetic determination of islet cell autoimmunity in
monozygotic twin, dizygotic twin, and non-twin siblings of patients with
type 1 diabetes: prospective twin study. BMJ, 1999. 318(7185): p. 698702.
Barnett, A.H., et al., Diabetes in identical twins. A study of 200 pairs.
Diabetologia, 1981. 20(2): p. 87-93.
Wagener, D.K., et al., The Pittsburgh study of insulin-dependent diabetes
mellitus. Risk for diabetes among relatives of IDDM. Diabetes, 1982.
31(2): p. 136-44.
Tillil, H. and J. Kobberling, Age-corrected empirical genetic risk
estimates for first-degree relatives of IDDM patients. Diabetes, 1987.
36(1): p. 93-9.
Aly, T.A., et al., Extreme genetic risk for type 1A diabetes. Proceedings of
the National Academy of Sciences of the United States of America, 2006.
103(38): p. 14074-9.
Hyoty, H., et al., A prospective study of the role of coxsackie B and other
enterovirus infections in the pathogenesis of IDDM. Childhood Diabetes
in Finland (DiMe) Study Group. Diabetes, 1995. 44(6): p. 652-7.
Dahlquist, G.G., et al., Maternal enteroviral infection during pregnancy
as a risk factor for childhood IDDM. A population-based case-control
study. Diabetes, 1995. 44(4): p. 408-13.
Gamble, D.R. and K.W. Taylor, Seasonal incidence of diabetes mellitus.
British Medical Journal (Clinical Research Ed.), 1969. 3(5671): p. 631-3.
Montgomery, S.M., et al., Pertussis infection in childhood and subsequent
type 1 diabetes mellitus. Diabetic Medicine, 2002. 19(12): p. 986-93.
Makela, M., et al., Enteral virus infections in early childhood and an
enhanced type 1 diabetes-associated antibody response to dietary insulin.
Journal of Autoimmunity, 2006. 27(1): p. 54-61.
Oikarinen, M., et al., Detection of enteroviruses in the intestine of type 1
diabetic patients. Clinical and Experimental Immunology, 2008. 151(1):
p. 71-5.
van der Werf, N., et al., Viral infections as potential triggers of type 1
diabetes. Diabetes/Metabolism Research and Reviews, 2007. 23(3): p.
169-83.
Flodstrom-Tullberg, M., Viral infections: their elusive role in regulating
susceptibility to autoimmune disease. Microbes and Infection, 2003.
5(10): p. 911-21.
Moya-Suri, V., et al., Enterovirus RNA sequences in sera of
schoolchildren in the general population and their association with type
1-diabetes-associated autoantibodies. Journal of Medical Microbiology,
2005. 54(Pt 9): p. 879-83.
References
143. Salminen, K., et al., Enterovirus infections are associated with the
induction of beta-cell autoimmunity in a prospective birth cohort study.
Journal of Medical Virology, 2003. 69(1): p. 91-8.
144. Fuchtenbusch, M., et al., No evidence for an association of coxsackie
virus infections during pregnancy and early childhood with development
of islet autoantibodies in offspring of mothers or fathers with type 1
diabetes. Journal of Autoimmunity, 2001. 17(4): p. 333-40.
145. Viskari, H.R., et al., Maternal first-trimester enterovirus infection and
future risk of type 1 diabetes in the exposed fetus. Diabetes, 2002. 51(8):
p. 2568-71.
146. Skarsvik, S., et al., Decreased in vitro type 1 immune response against
coxsackie virus B4 in children with type 1 diabetes. Diabetes, 2006. 55(4):
p. 996-1003.
147. Juhela, S., et al., T-cell responses to enterovirus antigens in children with
type 1 diabetes. Diabetes, 2000. 49(8): p. 1308-13.
148. Ludvigsson, J., Why diabetes incidence increases--a unifying theory.
Annals of the New York Academy of Sciences, 2006. 1079: p. 374-82.
149. Viskari, H., et al., Relationship between the incidence of type 1 diabetes
and enterovirus infections in different European populations: results from
the EPIVIR project. Journal of Medical Virology, 2004. 72(4): p. 610-7.
150. Virtanen, S.M., et al., Early introduction of dairy products associated with
increased risk of IDDM in Finnish children. The Childhood in Diabetes in
Finland Study Group. Diabetes, 1993. 42(12): p. 1786-90.
151. Kimpimaki, T., et al., Short-term exclusive breastfeeding predisposes
young children with increased genetic risk of Type I diabetes to
progressive beta-cell autoimmunity. Diabetologia, 2001. 44(1): p. 63-9.
152. Norris, J.M., et al., Lack of association between early exposure to cow's
milk protein and beta-cell autoimmunity. Diabetes Autoimmunity Study in
the Young (DAISY). JAMA, 1996. 276(8): p. 609-14.
153. Wahlberg, J., O. Vaarala, and J. Ludvigsson, Dietary risk factors for the
emergence of type 1 diabetes-related autoantibodies in 21/2 year-old
Swedish children. British Journal of Nutrition, 2006. 95(3): p. 603-8.
154. Luopajarvi, K., et al., Enhanced levels of cow's milk antibodies in infancy
in children who develop type 1 diabetes later in childhood. Pediatric
Diabetes, 2008.
155. Paronen, J., et al., Effect of cow's milk exposure and maternal type 1
diabetes on cellular and humoral immunization to dietary insulin in
infants at genetic risk for type 1 diabetes. Finnish Trial to Reduce IDDM
in the Genetically at Risk Study Group. Diabetes, 2000. 49(10): p. 165765.
156. Holmes, G.K., Screening for coeliac disease in type 1 diabetes. Archives
of Disease in Childhood, 2002. 87(6): p. 495-8.
101
References
157. Ludvigsson, J.F., et al., Celiac disease and risk of subsequent type 1
diabetes: a general population cohort study of children and adolescents.
Diabetes Care, 2006. 29(11): p. 2483-8.
158. Sollid, L.M., et al., Evidence for a primary association of celiac disease to
a particular HLA-DQ alpha/beta heterodimer. Journal of Experimental
Medicine, 1989. 169(1): p. 345-50.
159. Ziegler, A.G., et al., Early infant feeding and risk of developing type 1
diabetes-associated autoantibodies. JAMA, 2003. 290(13): p. 1721-8.
160. Johansson, C., U. Samuelsson, and J. Ludvigsson, A high weight gain
early in life is associated with an increased risk of type 1 (insulindependent) diabetes mellitus. Diabetologia, 1994. 37(1): p. 91-4.
161. Sepa, A., et al., Psychological stress may induce diabetes-related
autoimmunity in infancy. Diabetes Care, 2005. 28(2): p. 290-5.
162. Sepa, A., A. Frodi, and J. Ludvigsson, Mothers' experiences of serious life
events increase the risk of diabetes-related autoimmunity in their children.
Diabetes Care, 2005. 28(10): p. 2394-9.
163. Pearl-Yafe, M., et al., Pancreatic islets under attack: cellular and
molecular effectors. Current Pharmaceutical Design, 2007. 13(7): p. 74960.
164. Yoon, J.W. and H.S. Jun, Cellular and molecular pathogenic mechanisms
of insulin-dependent diabetes mellitus. Annals of the New York Academy
of Sciences, 2001. 928: p. 200-11.
165. Rabinovitch, A. and W.L. Suarez-Pinzon, Cytokines and their roles in
pancreatic islet beta-cell destruction and insulin-dependent diabetes
mellitus. Biochemical Pharmacology, 1998. 55(8): p. 1139-49.
166. Moriwaki, M., et al., Fas and Fas ligand expression in inflamed islets in
pancreas sections of patients with recent-onset type 1 diabetes mellitus.
Diabetologia, 1999. 42: p. 1332-1340.
167. Itoh, N., et al., Mononuclear cell infiltration and its relation to the
expression of major histocompatibility complex antigens and adhesion
molecules in pancreas biopsy specimens from newly diagnosed insulindependent diabetes mellitus patients. Journal of Clinical Investigation,
1993. 92(5): p. 2313-22.
168. Kallmann, B.A., et al., Systemic bias of cytokine production toward cellmediated immune regulation in IDDM and toward humoral immunity in
Graves' disease. Diabetes, 1997. 46(2): p. 237-43.
169. Karlsson, M.G. and J. Ludvigsson, Peptide from glutamic acid
decarboxylase similar to coxsackie B virus stimulates IFN-gamma mRNA
expression in Th1-like lymphocytes from children with recent-onset
insulin-dependent diabetes mellitus. Acta Diabetologica, 1998. 35(3): p.
137-44.
102
References
170. Berman, M.A., et al., Decreased IL-4 production in new onset type I
insulin-dependent diabetes mellitus. Journal of Immunology, 1996.
157(10): p. 4690-6.
171. Halminen, M., et al., Cytokine expression in unstimulated PBMC of
children with type 1 diabetes and subjects positive for diabetes-associated
autoantibodies. Scandinavian Journal of Immunology, 2001. 53(5): p.
510-3.
172. Kukreja, A., et al., Multiple immuno-regulatory defects in type-1 diabetes.
Journal of Clinical Investigation, 2002. 109(1): p. 131-40.
173. Karlsson Faresjo, M.G. and J. Ludvigsson, Diminished Th1-like response
to autoantigens in children with a high risk of developing type 1 diabetes.
Scandinavian Journal of Immunology, 2005. 61(2): p. 173-9.
174. Sjoroos, M., et al., Time-resolved fluorometry based sandwich
hybridisation assay for HLA-DQA1 typing. Disease Markers, 1998. 14(1):
p. 9-19.
175. Nejentsev, S., et al., Population-based genetic screening for the
estimation of Type 1 diabetes mellitus risk in Finland: selective
genotyping of markers in the HLA-DQB1, HLA-DQA1 and HLA-DRB1
loci. Diabetic Medicine, 1999. 16(12): p. 985-92.
176. Laaksonen, M., et al., HLA class II associated risk and protection against
multiple sclerosis-a Finnish family study. Journal of Neuroimmunology,
2002. 122(1-2): p. 140-5.
177. Miller, S.A., D.D. Dykes, and H.F. Polesky, A simple salting out
procedure for extracting DNA from human nucleated cells. Nucleic Acids
Research, 1988. 16(3): p. 1215.
178. Wasmuth, H.E. and H. Kolb, Cow's milk and immune-mediated diabetes.
Proceedings of the Nutrition Society, 2000. 59(4): p. 573-9.
179. Viglietta, V., et al., Loss of functional suppression by CD4+CD25+
regulatory T cells in patients with multiple sclerosis. Journal of
Experimental Medicine, 2004. 199(7): p. 971-9.
180. Ruprecht, C.R., et al., Coexpression of CD25 and CD27 identifies
FoxP3+ regulatory T cells in inflamed synovia. Journal of Experimental
Medicine, 2005. 201(11): p. 1793-803.
181. Hartigan-O'Connor, D.J., et al., Human CD4+ regulatory T cells express
lower levels of the IL-7 receptor alpha chain (CD127), allowing
consistent identification and sorting of live cells. Journal of
Immunological Methods, 2007. 319(1-2): p. 41-52.
182. Hori, S. and S. Sakaguchi, Foxp3: a critical regulator of the development
and function of regulatory T cells. Microbes and Infection, 2004. 6(8): p.
745-51.
183. Luhn, K., et al., Increased frequencies of CD4+ CD25(high) regulatory T
cells in acute dengue infection. Journal of Experimental Medicine, 2007.
204(5): p. 979-85.
103
References
184. Kimpimaki, T., et al., Natural history of beta-cell autoimmunity in young
children with increased genetic susceptibility to type 1 diabetes recruited
from the general population. Journal of Clinical Endocrinology and
Metabolism, 2002. 87(10): p. 4572-9.
185. Bingley, P.J., et al., Prediction of IDDM in the general population:
strategies based on combinations of autoantibody markers. Diabetes,
1997. 46(11): p. 1701-10.
186. Warram, J.H., et al., Differences in risk of insulin-dependent diabetes in
offspring of diabetic mothers and diabetic fathers. New England Journal
of Medicine, 1984. 311(3): p. 149-52.
187. Dahlquist, G., et al., The epidemiology of diabetes in Swedish children 014 years--a six-year prospective study. Diabetologia, 1985. 28(11): p.
802-8.
188. Harjutsalo, V., A. Reunanen, and J. Tuomilehto, Differential transmission
of type 1 diabetes from diabetic fathers and mothers to their offspring.
Diabetes, 2006. 55(5): p. 1517-24.
189. Dahlquist, G., et al., The Swedish childhood diabetes study--results from a
nine year case register and a one year case-referent study indicating that
type 1 (insulin-dependent) diabetes mellitus is associated with both type 2
(non-insulin-dependent) diabetes mellitus and autoimmune disorders.
Diabetologia, 1989. 32(1): p. 2-6.
190. Li, H., et al., Possible human leukocyte antigen-mediated genetic
interaction between type 1 and type 2 Diabetes. Journal of Clinical
Endocrinology and Metabolism, 2001. 86(2): p. 574-82.
191. TRIGR, Study design of the Trial to Reduce IDDM in the Genetically at
Risk (TRIGR). Pediatric Diabetes, 2007. 8(3): p. 117-37.
192. Hultcrantz, M., et al., Interferons induce an antiviral state in human
pancreatic islet cells. Virology, 2007. 367(1): p. 92-101.
193. Jones, E.Y., et al., MHC class II proteins and disease: a structural
perspective. Nature Reviews Immunology, 2006. 6(4): p. 271-82.
194. Marttila, J., et al., Epitopes recognized by CBV4 responding T cells: effect
of type 1 diabetes and associated HLA-DR-DQ haplotypes. Virology,
2004. 319(1): p. 27-35.
195. Hyoty, H. and K.W. Taylor, The role of viruses in human diabetes.
Diabetologia, 2002. 45(10): p. 1353-61.
196. Skarsvik, S., J. Ludvigsson, and O. Vaarala, Aberrant regulation of
interleukin-12 receptor beta2 chain on type 1 cytokine-stimulated T
lymphocytes in type 1 diabetes. Immunology, 2005. 114(2): p. 287-93.
197. Roivainen, M., et al., Functional impairment and killing of human beta
cells by enteroviruses: the capacity is shared by a wide range of
serotypes, but the extent is a characteristic of individual virus strains.
Diabetologia, 2002. 45(5): p. 693-702.
104
References
198. Dotta, F., et al., Coxsackie B4 virus infection of beta cells and natural
killer cell insulitis in recent-onset type 1 diabetic patients. Proceedings of
the National Academy of Sciences of the United States of America, 2007.
104(12): p. 5115-20.
199. Jonson, C.O., et al., The association of CTLA-4 and HLA class II
autoimmune risk genotype with regulatory T cell marker expression in 5year-old children. Clinical and Experimental Immunology, 2006. 145(1):
p. 48-55.
200. Murphy, K.M. and S.L. Reiner, The lineage decisions of helper T cells.
Nature Reviews Immunology, 2002. 2(12): p. 933-44.
201. Rao, A., C. Luo, and P.G. Hogan, Transcription factors of the NFAT
family: regulation and function. Annual Review of Immunology, 1997.
15: p. 707-47.
202. Jackman, R.P., F. Balamuth, and K. Bottomly, CTLA-4 differentially
regulates the immunological synapse in CD4 T cell subsets. Journal of
Immunology, 2007. 178(9): p. 5543-51.
203. Shilling, R.A., et al., Cutting edge: polymorphisms in the ICOS promoter
region are associated with allergic sensitization and Th2 cytokine
production. Journal of Immunology, 2005. 175(4): p. 2061-5.
204. Chakir, H., et al., T-bet/GATA-3 ratio as a measure of the Th1/Th2
cytokine profile in mixed cell populations: predominant role of GATA-3.
Journal of Immunological Methods, 2003. 278(1-2): p. 157-69.
205. Zhu, J., et al., Conditional deletion of Gata3 shows its essential function
in T(H)1-T(H)2 responses. Nature Immunology, 2004. 5(11): p. 1157-65.
206. Kotake, S., et al., IL-17 in synovial fluids from patients with rheumatoid
arthritis is a potent stimulator of osteoclastogenesis. Journal of Clinical
Investigation, 1999. 103(9): p. 1345-52.
207. Hwang, S.Y. and H.Y. Kim, Expression of IL-17 homologs and their
receptors in the synovial cells of rheumatoid arthritis patients. Molecules
and Cells, 2005. 19(2): p. 180-4.
208. Kebir, H., et al., Human TH17 lymphocytes promote blood-brain barrier
disruption and central nervous system inflammation. Nature Medicine,
2007. 13(10): p. 1173-5.
209. Annunziato, F., et al., Phenotypic and functional features of human Th17
cells. Journal of Experimental Medicine, 2007. 204(8): p. 1849-61.
210. Lundy, S.K., et al., Cells of the synovium in rheumatoid arthritis. T
lymphocytes. Arthritis Research and Therapy, 2007. 9(1): p. 202.
211. Danke, N.A., et al., Autoreactive T cells in healthy individuals. Journal of
Immunology, 2004. 172(10): p. 5967-72.
212. Lindley, S., et al., Defective suppressor function in CD4(+)CD25(+) Tcells from patients with type 1 diabetes. Diabetes, 2005. 54(1): p. 92-9.
105
References
213. Brusko, T.M., et al., Functional defects and the influence of age on the
frequency of CD4+ CD25+ T-cells in type 1 diabetes. Diabetes, 2005.
54(5): p. 1407-14.
214. Valencia, X., et al., Deficient CD4+CD25high T regulatory cell function
in patients with active systemic lupus erythematosus. Journal of
Immunology, 2007. 178(4): p. 2579-88.
215. Tsutsumi, Y., et al., Phenotypic and genetic analyses of T-cell-mediated
immunoregulation in patients with Type 1 diabetes. Diabetic Medicine,
2006. 23(10): p. 1145-50.
216. Cao, D., et al., CD25brightCD4+ regulatory T cells are enriched in
inflamed joints of patients with chronic rheumatic disease. Arthritis
Research and Therapy, 2004. 6(4): p. R335-46.
217. Grindebacke, H., et al., Defective suppression of Th2 cytokines by
CD4CD25 regulatory T cells in birch allergics during birch pollen
season. Clinical and Experimental Allergy, 2004. 34(9): p. 1364-72.
218. Bellinghausen, I., et al., Human CD4+CD25+ T cells derived from the
majority of atopic donors are able to suppress TH1 and TH2 cytokine
production. Journal of Allergy and Clinical Immunology, 2003. 111(4): p.
862-8.
219. Tiittanen, M., et al., Insulin treatment in patients with type 1 diabetes
induces upregulation of regulatory T-cell markers in peripheral blood
mononuclear cells stimulated with insulin in vitro. Diabetes, 2006. 55(12):
p. 3446-54.
220. Putnam, A.L., et al., CD4+CD25high regulatory T cells in human
autoimmune diabetes. Journal of Autoimmunity, 2005. 24(1): p. 55-62.
221. Brusko, T., et al., No alterations in the frequency of FOXP3+ regulatory
T-cells in type 1 diabetes. Diabetes, 2007. 56(3): p. 604-12.
222. Chen, W., et al., Conversion of peripheral CD4+CD25- naive T cells to
CD4+CD25+ regulatory T cells by TGF-beta induction of transcription
factor Foxp3. Journal of Experimental Medicine, 2003. 198(12): p. 187586.
223. Groux, H., et al., A CD4+ T-cell subset inhibits antigen-specific T-cell
responses and prevents colitis. Nature, 1997. 389(6652): p. 737-42.
224. Roncarolo, M.G., et al., Interleukin-10-secreting type 1 regulatory T cells
in rodents and humans. Immunological Reviews, 2006. 212: p. 28-50.
225. Zhang, X., et al., Activation of CD25(+)CD4(+) regulatory T cells by oral
antigen administration. Journal of Immunology, 2001. 167(8): p. 4245-53.
106