Sunteți pe pagina 1din 13

REVIEWS

Immune cell crosstalk in type 1


diabetes
Agnès Lehuen*, Julien Diana*, Paola Zaccone‡ and Anne Cooke‡
Abstract | The development of type 1 diabetes involves a complex interaction between pancreatic
β-cells and cells of both the innate and adaptive immune systems. Analyses of the interactions
between natural killer (NK) cells, NKT cells, different dendritic cell populations and T cells have
highlighted how these different cell populations can influence the onset of autoimmunity. There
is evidence that infection can have either a potentiating or inhibitory role in the development of
type 1 diabetes. Interactions between pathogens and cells of the innate immune system, and
how this can influence whether T cell activation or tolerance occurs, have been under close
scrutiny in recent years. This Review focuses on the nature of this crosstalk between the innate
and the adaptive immune responses and how pathogens influence the process.

Non-obese diabetic (NOD)


Type 1 diabetes (T1D) is a chronic autoimmune dis- Studies in animal models (BOX 1) , particularly in
mice ease, during which the pancreatic β-cells (which secrete non-obese diabetic (NOD) mice, have identified roles for
Mice that spontaneously insulin) are selectively destroyed. It is thought to be a several different immune cell types in β-cell destruc-
develop type 1 diabetes as a T helper 1 (TH1) cell-mediated disease that involves tion. CD4+ and CD8+ T cells, as well as macrophages,
result of islet antigen-specific
CD8+ T cells and innate immune cells. Individuals with have been shown to have a role in β-cell death. However,
T cell-mediated destruction of
pancreatic β-cells. T1D develop hyperglycaemia and can develop diabetes- other cell types are present in the pancreatic infiltrate
associated complications in several organ systems owing and in the pancreatic draining lymph node, where the
to a lack of insulin. This was a lethal disease until the initial presentation of islet antigen by dendritic cells
1920s when Banting and Best 1 identified insulin as (DCs) to islet antigen-specific T cells occurs6. These
the hormone in the pancreas responsible for maintain- cells include B cells, natural killer (NK) cells and
ing blood glucose homeostasis. The development of T1D NKT cells, as well as DC subsets, and they could also
is under polygenic control, with an additional role for contribute to β-cell death. Conversely, targeting or har-
environmental factors2. This role for environmental fac- nessing the activities of many of these different immune
tors is highlighted by the 40–60% concordance rate for cell types can also be effective in inhibiting β-cell
diabetes onset in identical twins and also by the dramatic destruction. This strongly suggests that there is substan-
increase in the incidence of T1D in recent years3,4. As the tial crosstalk between the immune cells that are involved
development of T1D is increasing in the UK at a rate of in pathogenesis and those involved in immune regula-
3% per year, which is faster than can be accounted for tion. The genetics of T1D or therapeutic approaches
*Institut National de la Santé by genetic change, there has been a considerable effort for disease prevention have been extensively reviewed
et de la Recherche Médicale to identify the environmental factors that predispose an elsewhere7,8 and therefore are not discussed here.
(INSERM) U986, Hôpital
Saint Vincent de Paul,
individual to diabetes onset. There has been substantial Epidemiological studies suggest that environ-
Bâtiment Petit, 82 Avenue focus on identifying agents, such as viruses, that might mental factors influence the development of T1D
Denfert-Rochereau, precipitate diabetes onset and, more recently, there has in humans9,10. Pathogens, particularly viruses, could
75014 Paris, France. been a growing interest in establishing whether exposure accelerate T1D onset through the induction of inflam-

Department of Pathology,
to certain pathogens might have been the reason for a matory responses following direct infection of β-cells,
University of Cambridge,
Tennis Court Rd, Cambridge historically lower incidence of diabetes onset. Improved as well as through other mechanisms (discussed
CB21QP, UK. sanitation and living conditions, together with vaccina- below). In addition, other pathogens might inhibit
Correspondence to A.L. tion strategies, have decreased our exposure to pathogens T1D onset through effects on numerous cell types and
and A.C. and development of infectious disease. It is therefore pos- the induction of mediators that suppress host pathol-
e-mails:
agnes.lehuen@inserm.fr;
sible that a reduced exposure to pathogens might be the ogy. These contrasting effects are mediated through
ac@mole.bio.cam.ac.uk environmental alteration over the last 60 years that has interactions between cells of both the innate and
doi:10.1038/nri2787 had a role in the increased incidence of T1D5. adaptive immune system. In this Review, we discuss

NATURE REvIEwS | Immunology volUmE 10 | jUly 2010 | 501

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Box 1 | Animal models of type 1 diabetes could initiate β-cell apoptosis. Chemokine production
by β-cells results in further recruitment of mononuclear
Animal models have contributed enormously to the understanding of processes cells to the site, thereby enhancing inflammation15. In
leading to development of type 1 diabetes (T1D). T1D can be experimentally induced addition, IFNγ can activate macrophages and induce
in laboratory animals with chemicals (such as streptozotocin and cyclophosphamide in
increased pro-inflammatory cytokine production,
genetically susceptible mouse strains) or by genetic manipulation (such as transgenic
including interleukin-1β (Il-1β) and tumour necrosis
mice). In addition, two spontaneous murine models have been extensively used to
understand the pathophysiology of T1D: the biobreeding (BB) rat and the non-obese factor (TNF). β-cells express high levels of Il-1 recep-
diabetic (NOD) mouse models. In BB rats there is no sex difference in the incidence of tor and seem to be more sensitive to Il-1β-induced
T1D, but female NOD mice develop T1D at a higher incidence than male NOD mice. apoptosis than other endocrine cells in the islet. This
Besides this difference, both models resemble T1D in humans, including the presence crosstalk between T cells and macrophages undoubtedly
of islet antigen-specific CD4+ and CD8+ T cells and genetic linkage to disease. exacerbates the immune-mediated stress on β-cells and
As in humans, specific MHC gene products are involved in T1D susceptibility in both contributes to their destruction. IFNγ, Il-1β and TNF
models: RT1u/u in BB rats and I–Ag7 in NOD mice. In addition to the MHC locus, other also induce the expression of reactive oxygen species
loci contribute to disease development. No less than 12 loci were found to influence (RoS) including nitric oxide by β-cells, and RoS have
the diabetogenic process in BB rats, and more than 20 potential Idd loci have been
the potential to mediate apoptosis.
identified in NOD mice, which include polymorphisms in the genes cytotoxic
Although T cells have a pathological role in T1D
T lymphocyte antigen 4 (Ctla4), interleukin-2 (Il2) and protein tyrosine phosphatase,
non-receptor type 22 (Ptpn22). In the past 20 years, numerous studies have shown that onset, there is also evidence supporting a role for
both genetic and environmental factors contribute to the pathogenesis of the disease T cells in the prevention of β-cell destruction. Patients
in both models. In particular, infection studies have underlined the importance of with IPEX (immunodysregulation, polyendocrinopathy,
co-evolution between pathogens and the mammalian immune system for both the enteropathy, X-linked) syndrome — who have muta-
induction and prevention of T1D. tions in forkhead box P3 (FoXP3; the regulatory
T (TReg) cell-associated transcription factor) — can
develop T1D16, which highlights the importance of
the role of innate immune cells in promoting or pre- TReg cells in controlling the onset of this autoimmune
venting diabetes onset and the potential for infection disease. Studies in NoD mice have shown the impor-
to influence the crosstalk between immune cells and tance of TReg cells in preventing T1D: CD28-deficient
pancreatic β-cells. NoD mice, which lack TReg cells, develop accelerated
disease17. In addition, strategies such as injection of
Lymphocytes and T1D Il-2 to increase TReg cell numbers are seen as a potential
There is considerable evidence that T cells have an therapeutic approach18.
important role in the development and progression of T cells are clearly pivotal for T1D development,
T1D in both humans and animal models. The recur- but there are also data suggesting an involvement of
rence of T1D in recipients of segmental pancreas grafts other cell types such as B cells; however, there is little
from HlA-identical donors showed a clear role for evidence for a role for antibody in the pathogenesis of
T cells — particularly CD8+ T cells — and monocytes, T1D. B cell depletion in NoD mice, either through gene
with little evidence for a humoral immune response, targeting or antibody treatment, impairs the develop-
in β-cell destruction11. β-cell survival could only be ment of T1D19,20. This has led to the treatment of newly
achieved in these transplantation studies if the recipi- diagnosed T1D patients with the B cell-depleting
ent was immunosuppressed, indicating that T1D in monoclonal antibody rituximab (Rituxan/mabthera;
humans was an autoimmune disease. Genentech/Roche/Biogen Idec), resulting in improved
Studies in NoD mice have shown that T1D develop- β-cell function21. However, there is one description
ment depends on both CD4+ and CD8+ T cells: T1D can of a patient with X-linked agammaglobulinaemia who
only be transferred to immunocompromised syngeneic developed T1D22, which questions the role of B cells
recipients by a combination of splenic CD4+ and CD8+ in disease pathogenesis. It may be that B cells have a
T cells from donor NoD mice but not by either T cell role as antigen-presenting cells that maintain islet
subset alone12. The ability of a CD3-specific antibody, antigen-specific T cell activity 19,23.
which induces T cell tolerance, to reverse T1D onset in
NoD mice emphasized the key role of T cells in sus- Innate immune cells in T1D
tained β-cell destruction13. These studies led the way As islet antigen-specific T cells can differentiate into
X-linked
agammaglobulinaemia
to the use of an aglycosyl CD3-specific monoclonal either pathogenic effector T cells (diabetogenic T cells)
A human immunodeficiency antibody in patients with recent-onset T1D, in which or protective TReg cells, many studies have investigated
that is caused by mutations in there was evidence that targeting T cells resulted in the role of innate immune cells in T1D, as these cells usu-
the gene encoding Bruton’s suppression of ongoing β-cell destruction14. ally determine the type of immune response that ensues.
tyrosine kinase (BTK) (which is
There are several ways in which T cell-mediated Innate cells producing pro-inflammatory or suppres-
located on the X chromosome).
These mutations result in a β-cell death might occur (FIG. 1). CD8+ T cells could sive cytokines define the milieu in which islet antigen-
block in B cell maturation and kill pancreatic β-cells through mHC class I-mediated specific T cells are activated and whether a deleterious or
in poor antibody production. cytotoxicity, and both CD4+ and CD8+ T cells produce protective local immune response occurs in the pancreas
A naturally occurring mouse cytokines, such as interferon-γ (IFNγ), that induce (FIG. 2). we first discuss studies pertaining to the patho-
mutant of BTK, X-linked
immune deficiency, is
expression of the death receptor FAS (also known as genic role of innate immune cells, such as macrophages,
associated with less severe CD95) and chemokine production by β-cells. Activation NK cells and DCs, and then review the growing evidence
disease. of FAS by FAS ligand (FASl)-expressing activated T cells for the protective effect of innate immune cells.

502 | jUly 2010 | volUmE 10 www.nature.com/reviews/immunol

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

The pathogenic role of macrophages. Although numer-


ous studies of both humans and NoD mice emphasize a
predominant role for diabetogenic T cells in the patho-
Macrophage genesis of T1D, several studies support the involvement
of macrophages in this disease24–28. Early experiments
described the presence of macrophages in islet infil-
trates of NoD mice, and showed that inhibition of the
Lymphocyte and
macrophage recruitment macrophage influx into the pancreas, by blocking an
by CXCL10, CCL2 and CCL20 adhesion-promoting receptor on this cell, inhibited
IFNγ TNF the development of T1D24. In vitro and in vivo studies
IL-12
IL-1β
in mice and rats showed that the deleterious effect of
macrophages on β-cells can be mediated through the
Apoptosis
production of TNF and Il-1β29,30. Interestingly, pro-
or induction RAE1 NKG2D inflammatory macrophages can be detected in pancre-
Microbial
products of IDO atic islets before T cell infiltration, as well as in NoD/scid
β-cell damage NK cell
(severe combined immunodeficient) mice, which lack
TLR2 Degranulation functional B and T cells30.
NKp46 macrophages have been shown to produce Il-12
β-cell Unknown (ReF. 26) and to promote efficient differentiation of
TLR4 TLR3 TLR9 ligand
diabetogenic CD8+ cytotoxic T lymphocytes (CTls)
Chromogranin A Cytotoxicity leading to T1D onset25. Genetic differences in Il-12 pro-
Insulin duction have been observed in peritoneal macrophages
Pro-insulin MHC class I from susceptible and resistant mouse strains: cells from
Tolerance β-cell NoD mice secrete more Il-12 than those from non-obese
or antigen
resistant (NOR) mice when stimulated with CD40 ligand or
apoptosis TCR
lipopolysaccharide (lPS)26. more recent data suggest that
PDL1 recruitment of macrophages to islets is mediated by the
WE14 peptide PDL1 FAS PD1 secretion of CC-chemokine ligand 1 (CCl1) and CCl2
Diabetogenic
PD1 FASL CD8+ T cell by CD4+ T cells and pancreatic β-cells, respectively 27,31.
macrophages recruited to the pancreas produce Il-1β,
TNF and RoS that can cause β-cell death, revealing an
TCR additional role for macrophages in the destructive phase
Diabetogenic
DC CD4+ T cell of T1D. Finally, TNF- and Il-1β-producing macrophages
MHC and DCs have been observed in pancreatic islet infiltrates
class II from patients with recent-onset T1D32. Together, these
studies support a pathogenic role for macrophages in
both the initiation and destruction phases of T1D.

A role for NK cells. NK cells mediate early protection


against viruses and are involved in the killing of infected
Figure 1 | molecules expressed by pancreatic β-cells involved in their destruction cells and tumours. NK cells are both cytotoxic and pro-
or protection. Toll-like receptors (TLRs) are expressed by pancreatic β-cells to detect ducers of cytokines, particularly IFNγ. Thus, NK cells
Nature Reviews | Immunology
danger from microbial products and can trigger type 1 diabetes (T1D); for example, could contribute directly and indirectly to the destruction
during a localized virus infection in the pancreas. Cytokines, such as interleukin-1β of β-cells. NK cells have been detected in the pancreas of
(IL-1β), IL-12, interferon-γ (IFNγ) and tumour necrosis factor (TNF), secreted by patients with T1D and in many T1D mouse models33–37.
immune cells, such as macrophages, dendritic cells (DCs), CD4+ and CD8+ T cells and
moreover, several reports have described a correlation
natural killer (NK) cells, could cause direct damage (such as apoptosis) of β-cells but
could also induce self-defence mechanisms; for example, IFNγ induces indoleamine between the frequency and/or activation of NK cells with
2,3-dioxygenase (IDO) expression by β-cells. Chemokines such as CXC-chemokine the destructiveness of the pancreatic infiltrate35,38.
ligand 10 (CXCL10), CC-chemokine ligand 2 (CCL2) and CCL20 secreted by β-cells NK cells isolated from the pancreas of NoD mice have
recruit macrophages and other inflammatory cells to the islet area. Natural killer a more activated phenotype than those isolated from the
group 2, member D (NKG2D) expressed by NK cells binds to retinoic acid early spleen and pancreatic lymph node, with higher expres-
transcript 1 (RAE1) expressed by β-cells in non-obese diabetic (NOD) mice, and is sion of CD25, CD69, programmed cell death 1 (PD1)
associated with β-cell damage. NKp46 engagement by an unknown ligand on β-cells and killer cell lectin-like receptor group G, member 1
causes degranulation of NK cells. Negative co-stimulatory molecules, such as (KlRG1)37. They also proliferate more and spontane-
programmed cell death ligand 1 (PDL1) expressed by β-cells can modulate ously produce higher levels of IFNγ and express CD107a
diabetogenic T cell attack. β-cell antigens (such as peptides derived from insulin
on their cell surface (a marker of granule exocytosis),
or pro-insulin) can be presented by MHC class I molecules and recognized by
diabetogenic CD8+ T cells. MHC class II molecules expressed by antigen-presenting reflecting their cytotoxic function39. Interestingly, NK
cells such as DCs can also present β-cell antigens (peptides such as WE14 from cells were observed in the pancreas in NoD mice before
chromogranin A, insulin and pro-insulin) that can induce islet antigen-specific CD4+ T cell infiltration and in the pancreas of NoD–Rag mice
T cell expansion. FASL-expressing T cells can mediate apoptosis through interaction (which lack mature B and T cells), suggesting that they
with FAS expressed on β-cells. TCR, T cell receptor. could have a sentinel role in the pancreas.

NATURE REvIEwS | Immunology volUmE 10 | jUly 2010 | 503

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Phase I: β-cell death and Phase III: immune cell crosstalk and induction or prevention of β-cell death
APC activation
NK cell Macrophage cDC

β-cell damage: apoptosis


and viral infection

IFNγ,
granzymes, TNF, IL-1β and
and nitric oxide
IL-10 and TGFβ
perforin IL-12
β-cell
β-cell IFNγ, granzymes
antigen and perforin TReg cell
β-cell Diabetogenic
T cell
PDL1–PD1 pDC iNKT cell
cDC
Cell death
IDO
Pancreas

Draining lymph node


Promote
recruitment

iNKT cell IDO, IL-10,


TGFβ and ICOSL
Activated
cDC
PDL1–PD1
MHC
Activation
TCR Tolerogenic cDC

IL-12

Phase II: expansion of IDO, IL-10,


Macrophage Pathogenic islet pathogenic or regulatory TGFβ and
antigen-specific T cell islet antigen-specific T cells TReg cell ICOSL pDC
Figure 2 | Cellular and molecular mechanisms in the development or prevention of type 1 diabetes. The
initiation phase of type 1 diabetes (T1D) takes place in the pancreas, where conventional dendritic cells (cDCs) capture
and process β-cell antigens. β-cell damage can occur by ‘natural’ apoptosis or after viral infections. Invariant
Nature Reviews natural
| Immunology
Non-obese resistant (NOR) killer T (iNKT) cells and plasmacytoid DCs (pDCs) control viral replication, preventing subsequent inflammation and
mice T1D (not shown). Activated cDCs prime pathogenic islet antigen-specific T cells after migration to the draining lymph
NOR mice have the identical node, and macrophages promote this activation through interleukin-12 (IL-12) secretion. B cells present β-cell antigen
T cell developmental to diabetogenic T cells and secrete autoantibodies (not shown). The activation of islet antigen-specific T cells can be
background as NOD mice, but inhibited by cDCs through various mechanisms, such as engagement of programmed cell death ligand 1 (PDL1).
they do not spontaneously
iNKT cells can promote the recruitment of tolerogenic cDCs and pDCs that could expand regulatory T (TReg) cells
develop type 1 diabetes.
through the production of indoleamine 2,3-dioxygenase (IDO), IL-10, transforming growth factor-β (TGFβ) and
BDC2.5 TCR-transgenic inducible T cell co-stimulator ligand (ICOSL). In the pancreas, β-cells can be killed by diabetogenic T cells and NK cells
NOD mice through the release of interferon-γ (IFNγ), granzymes and perforin, as well as by macrophages through the production
T cells in these mice express a of tumour necrosis factor (TNF), IL-1β and nitric oxide. IL-12 produced by cDCs sustains the effector functions of
TCR specific for a pancreatic activated diabetogenic T cells and NK cells (not shown). β-cell damage can be inhibited by TReg cells that inhibit
antigen but, interestingly, these diabetogenic T cells and innate immune cells through IL-10 and TGFβ. Tolerogenic pDCs stimulated by iNKT cells could
mice have a decreased also control diabetogenic T cells through IDO production. Lastly, β-cells can inhibit diabetogenic T cells by expressing
incidence of type 1 diabetes PDL1. This complex crosstalk between innate and adaptive immune cells results in the development or the prevention
and develop a non-invasive
of T1D. APC, antigen-presenting cell; TCR, T cell receptor.
insulitis. These mice are
used as donors of islet
antigen-specific CD4+ T cells
and manipulation of these A pathogenic role for NK cells has been suggested in (cytotoxic T lymphocyte antigen 4)-specific antibody
mice, such as injection of various T1D mouse models such as NoD mice, SoCS1 treated BDC2.5 TCR-transgenic NOD mice 34–36 (BOX 1).
blocking CTLA4-specific
antibody and infection with
(suppressor of cytokine signalling 1)-transgenic NoD Depletion of NK cells using various antibodies against
coxsackie virus B4, induces mice infected with coxsackie virus B4, mice trans- NK1.1 and asialo-Gm1 (ReFs 34–36) prevented T1D in
rapid type 1 diabetes. genically expressing IFNβ in their β-cells, and CTlA4 all of these models. Natural killer group 2, member D

504 | jUly 2010 | volUmE 10 www.nature.com/reviews/immunol

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

(NKG2D) and NKp46 are expressed by NK cells, and Plasmacytoid DCs (pDCs) detect viral RNA or DNA
recent studies suggest that their ligands (retinoic acid through TlR7 and TlR9 and respond by secreting
early transcript 1 (RAE1) and NKp46 ligand, respec- large amounts of type I IFNs, Il-12 and pro-inflam-
tively) are expressed by β-cells from NoD mice 40. matory chemokines. In contrast to cDCs, pDCs have a
moreover, NKG2D and NKp46 blockade could pre- decreased capacity to take up, process and present solu-
vent T1D in NoD mice, suggesting that recognition of ble antigens49. A putative role for pDCs in the devel-
β-cells by NK cells may have a role in T1D39. However, opment of T1D is supported by observations in both
although NKp46 expression is restricted to NK cells, human and murine models that type I IFNs, which are
NKG2D is also expressed by CTls, and NKG2D block- usually produced by this cell type, can in some situa-
ade might affect the function of diabetogenic CTls that tions induce or enhance T1D50,51. Antibody-mediated
infiltrate the pancreas 40. The deleterious role of NK blockade of type I IFNs identified a crucial role for these
cells in T1D has been further highlighted in a recent cytokines in the initiation of disease. However, these
study 38: TReg cell depletion of BDC2.5 TCR-transgenic studies did not show that pDCs were the source of this
NoD mice resulted in an increase in the percent- type I IFN. mouse models have been used to examine
age of activated pancreatic NK cells that produced the pDC populations in the pancreas and pancreatic
large amounts of effector molecules such as IFNγ, lymph node at the early stage of T1D development.
which promote the effector function of diabetogenic Two reports suggest a pathogenic role for pDCs in NoD
CD4+ T cells. mice. In one study, the initiation of T1D correlated with
an increased production of type I IFNs by pDCs in the
How do DCs promote T1D? Early studies described pancreatic lymph node52. FmS-like tyrosine kinase 3
RIP–LCMV transgenic the presence of antigen-presenting cells (APCs) in ligand (FlT3l) treatment, which expands both cDC
model transplanted pancreatic islets41 and showed that deple- and pDC populations, accelerated T1D in 15-week-old
A transgenic mouse model of
type 1 diabetes in which
tion of these cells facilitated graft survival in mice42. NoD mice, an age when islet antigen-specific CTls
peptides derived from These data suggested that APCs could take up and were detectable in blood53.
lymphocytic choriomeningitis process β-cell-derived proteins and subsequently ini- However, there have been apparently discrepant data
(LCMV) are expressed in the tiate the diabetogenic response. This hypothesis was regarding the frequency of pDCs in the blood of patients
pancreas under the control of
tested using mice that express the lymphocytic chorio- with T1D, which may reflect differences in the time
the rat insulin promoter (RIP).
Infection of the mice with meningitis virus (lCmv) glycoprotein under the con- when the blood samples were taken54–56. Interestingly,
LCMV leads to the trol of the rat insulin promoter (RIP) (the RIP–LCMV pDCs from early-diagnosed patients were shown to
development of type 1 transgenic model)43. Repeated injection of conventional present immune complexes to T cells more efficiently
diabetes as a result of DCs (cDCs) constitutively expressing the immuno- than cDCs, suggesting a possible detrimental role of
infiltrating CD8+ effector
T cells.
dominant lCmv epitope into these transgenic mice pDCs in T1D onset.
resulted in severe destructive mononuclear infiltra-
Perforin tion of the pancreatic islets and development of T1D Prevention of T1D by DCs. It is known that patients
A component of cytolytic through CTl activation. This development of CTl- with a congenital DC deficiency develop autoimmune
granules that participates in
mediated T1D was perforin independent, as it was also diseases57. This highlights a role for DCs in mediating
the permeabilization of plasma
membranes, allowing seen in perforin-deficient RIP–lCmv mice43. Further peripheral tolerance and, indeed, antigen presentation by
granzymes and other cytotoxic studies showed that self antigens released after β-cell cDCs mediates tolerance by inducing T cell deletion, T cell
components to enter target death could be captured by cDCs in the pancreatic anergy or the expansion of antigen-specific TReg cells58. TReg
cells. islets and presented to islet antigen-specific T cells cells are key players in preventing T1D18, and various stud-
Peripheral tolerance
in the pancreatic lymph node where the diabetogenic ies have investigated whether modulation of cDCs could
The lack of self responsiveness response is initiated6,44. influence TReg cells and T1D development. Treatment of
of mature lymphocytes in the Initial β-cell death could occur physiologically in NoD mice with granulocyte colony-stimulating factor
periphery to specific antigens. NoD mice at two weeks of age during tissue remodel- (G-CSF) to mobilize DCs prevented T1D development
These mechanisms control
ling and at weaning owing to a metabolic change, or and increased the numbers of cDCs and pDCs in the
potentially self-reactive
lymphocytes that have it could occur through injury mediated by viral infec- spleen59. These cells mediated tolerance through effects
escaped central tolerance. tions6,10. Such β-cell death could activate cDCs and on CD4+CD25+ TReg cells that suppress pathogenic T cells
Peripheral tolerance is initiate priming of islet antigen-specific CD4+ T cells, through the production of transforming growth factor-β
associated with the as revealed by the high frequency of cDCs present- (TGFβ)59. FlT3l injection protected NoD mice from
suppression of production of
self-reactive antibodies by
ing β-cell-derived antigens in the pancreatic draining T1D by enhancing CD4+CD25+ TReg cell frequency in the
B cells and inhibition of lymph node6. A role for Toll-like receptor 2 (TlR2) in pancreatic lymph node and by modulating the balance of
self-reactive effector cells, such this process has been suggested45, but a recent study cDC subsets towards CD8+ cDCs, which have potentially
as cytotoxic T lymphocytes. showed similar T1D incidence in TlR2-deficient and tolerogenic functions60,61. of note, FlT3l treatment is pro-
wild-type NoD mice46. However, it is conceivable that tective only when administered in the early stages of T1D
T cell anergy
A state of T cell other pattern recognition receptors (PRRs) may have a development in NoD mice when islet antigen-specific
unresponsiveness to role in the absence of TlR2. Several reports have sug- T cells are still at a low frequency 53. Thus, the opposing
antigen-specific stimulation. It gested that cDCs from NoD mice have increased abil- role of FlT3l depends on the time of injection relative
can be induced by stimulation ity to activate T cells through higher Il-12 production to the disease progression in NoD mice. A recent study
with a large amount of specific
antigen in the absence of the
and co-stimulatory molecule expression47,48. Together, highlighted the in vivo peripheral feedback loop that exists
engagement of co-stimulatory these studies support a diabetogenic role for cDCs in the between TReg cells and DC frequency involving FlT3l.
molecules. initiation of this disease. Deletion of TReg cells induced FlT3l production, which

NATURE REvIEwS | Immunology volUmE 10 | jUly 2010 | 505

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

increased DC frequency and subsequently restored the Increasing the frequency of iNKT cells, either by adop-
normal TReg cell frequency required to prevent autoim- tive transfer or through the introduction of a vα14–jα18
mune diseases62. Importantly, G-CSF and FlT3l could transgene, substantially reduces the incidence of T1D in
prevent T1D not only by increasing the numbers of cDCs NoD mice73,74. A similar protective effect was observed
but also the numbers of pDCs. after specific iNKT cell stimulation with the exogenous
Several molecules expressed by pDCs might be ligand α-galactosylceramide or its analogues75–78. Early
involved in the induction of T cell tolerance, such as reports suggested that iNKT cell-mediated protection
PDl1, inducible T cell co-stimulator ligand (ICoSl) and was associated with the induction of TH2 cell responses
indoleamine 2,3-dioxygenase (IDo). A protective role for to islet autoantigens75,76,79. However, studies using the
pDCs in T1D was first described by Saxena et al.63: by transfer of islet antigen-specific CD4+ and CD8+ T cells
transferring naive BDC2.5 T cells to NoD-Scid mice, showed that iNKT cells inhibit the differentiation of
they showed that pDCs inhibited the CD4+ T cell diabe- these T cells into effector T cells during their priming
togenic response, induced IDo in the pancreas and pre- in the pancreatic lymph node80,81. Interestingly, these
vented T1D onset. IDo catalyses oxidative catabolism of islet antigen-specific CD4+ T cells became anergic and
tryptophan and, in this way, regulates T cell-mediated remained in the pancreatic lymph node and pancreas80
immune responses, as free tryptophan is an essential where they could have a regulatory role. The abortive
nutrient for T cells. Interestingly, a defect in IDo expres- priming of islet antigen-specific T cells in the pancreatic
sion has been described in young NoD mice64, and over- lymph node could be explained by their ability to recruit
expression of IDo prolongs islet graft survival65. Using tolerogenic DCs81. Interestingly, type 2 NKT cells, which
both virus-induced and spontaneous models of T1D, we express variable TCRs, can also prevent T1D in NoD
recently observed that following viral infection, invariant mice, although the precise mechanism is still under
NKT (iNKT) cell–pDC crosstalk inhibits diabetogenic CTl investigation82. Although the number and function of
responses in the pancreas66. In this infectious context, iNKT cells from NoD mice are defective, which could
pDCs induced the conversion of CD4+ T cells to FoXP3+ contribute to T1D susceptibility 67,83, there is no consen-
TReg cells, which are crucial for preventing T1D (j.D. and sus on whether iNKT cell defects occur in humans with
A.l., unpublished observations). Together, these studies T1D84,85. However, manipulations of iNKT cells prevent
support a protective role of pDCs in T1D and strengthen and even cure T1D in various mouse models, encour-
their potential use in new therapeutic strategies. aging the development of new therapeutic strategies to
target iNKT cells.
Protective role of NK cells in T1D. Despite their poten- The dual role of DCs and NK cells in the development
tial pathogenic role in T1D, several studies suggest a of T1D could depend on different parameters: first, the
protective function of NK cells. NK cells from the blood type of cell subsets involved (for example CD8– cDCs,
of patients with T1D and lymphoid tissues from NoD CD8+ cDCs and/or pDCs); second, different pathways
mice exhibit impaired function67–69. As this defect was of activation through surface and cytoplasmic recep-
observed in patients with long-term T1D it could be a tors (such as TlR-mediated recognition of endogenous
consequence of the disease69. However, in NoD mice, the and exogenous ligands); third, a pro-inflammatory or
Indoleamine
2,3-dioxygenase NK cell defect is present before disease onset, suggesting regulatory cytokine milieu; and fourth, genetic defects
(IDO). An intracellular that impaired NK cell function could contribute to dis- such as increased Il-12p40 production by DCs and
haem-containing enzyme ease development 67,68. Consistent with this hypothesis, impaired NK cell function in NoD mice. The integra-
that catalyses the oxidative injection of young NoD mice with complete Freund’s tion of all of these parameters could result in prevention
catabolism of tryptophan.
IDO suppresses T cell
adjuvant (CFA) induces IFNγ production by NK cells or exacerbation of T1D development by DCs and NK
responses and promotes and prevents T1D70. The protective role of NK cells in cells. Interestingly, based on studies of mouse models,
immune tolerance in this setting was shown by their ability to inhibit T1D these innate cells often seem to have a protective role
mammalian pregnancy, in transfer experiments. The beneficial role of NK cells in in the early phases of the disease, whereas increasing
tumour resistance, chronic
islet allograft tolerance has been shown to involve killing their frequency or activating them at later stages, when
infection, autoimmunity
and allergic inflammation. of DCs by NK cells in a perforin-dependent manner 71. diabetogenic T cell frequency have reached a certain
NK cells seem to have a protective role following CFA threshold, may precipitate disease.
Invariant NKT (iNKT) cells immunostimulation of young NoD mice and in islet
Lymphocytes that express allografts, whereas they have a deleterious effect in older Infection and T1D
a particular variable gene
segment, Vα14 (in mice) and
NoD mice that have massive islet infiltrates or in more Potentiating disease. The previous section describes the
Vα24 (in humans), precisely aggressive mouse models of T1D. innate cells involved in the regulation of T1D, and DCs
rearranged to a particular Jα have a pivotal role both in inducing and preventing dis-
(joining) gene segment to yield iNKT cells. iNKT cells are innate-like T cells that express ease, depending on various factors. As these cells express
T cell receptor α-chains that
an invariant TCR α-chain (vα14–jα18 in mice and several pathogen recognition receptors (PRRs) to sense
have an invariant sequence.
Typically, these cells vα24–jα18 in humans). Following activation, iNKT cells microorganisms, it is not surprising that infections can
co-express cell surface promptly produce large amounts of various cytokines and modulate DCs, and other innate immune cells, and influ-
markers that are encoded by chemokines, thereby providing signals to other immune ence islet antigen-specific T cell responses and the devel-
the NK locus, and they are cells, including DCs, NK cells and lymphocytes. Although opment of T1D (FIG. 3). TlR3 and the cytoplasmic proteins
activated by recognition of
CD1d, particularly when
iNKT cells can promote immunity to pathogens, many retinoic acid-inducible gene-I (RIG-I) and melanoma
α-galactosylceramide is studies have shown their role in preventing auto- differentiation-associated gene 5 (mDA5; also known
bound in the groove of CD1d. immune diseases, particularly T1D (reviewed in ReF. 72). as IFIH1) have been identified as crucial intracellular

506 | jUly 2010 | volUmE 10 www.nature.com/reviews/immunol

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Virus Picornavirus encephalomyocarditis virus (EmCv)


Cytoplasmic induces T1D in wild-type mice. Infection with a high
Molecular
PRR dose of the D strain of EmCv results in acute onset
mimicry of T1D in more than 90% of infected animals within
Viral molecule 4 days of infection88. T1D onset in EmCv-infected mice
Viral antigen MHC cDC seemed to depend on macrophages but not T cells, as
TCR shown by blocking and depleting antibody treatments89.
Recruitment The mechanism of β-cell destruction by macrophages
Islet antigen- Inflammatory could be mediated through the release of cytokines
specific T cell molecules (TNF) and RoS89,90. Interestingly, fulminant T1D in
NK cell humans is associated with a macrophage-dominant,
Macrophage
NKG2D
rather than T cell-dominant, infiltrate91. These data show
the role of innate immune cell activation following virus
RAE1
infection in promoting T1D. In most examples in which
viruses induce T1D, infections lead to a cascade of innate
and adaptive immune cell activation with a clear role for
TGFβ Cytotoxicity β-cell diabetogenic T cells (as described below).
IL-10 IFNα

Stimulation of
Activation of autoimmune T cells through molecular
antiviral innate mimicry. Cross-reactive immune responses arising
β-cell Cell death immunity and through regions of sequence similarity between viral-
viral clearance and self-antigen epitopes could theoretically lead to
antigen
TReg cell diabetogenic T cell activation and tissue destruction
TReg cell following viral infection. Numerous mimics between
induction by pDC
tolerogenic DC
β-cell antigens and viral epitopes have been identified,
including coxsackie virus B4 (ReF. 92), rotavirus93, rubella
TCR virus94 and Cmv95. The RIP–lCmv mouse model,
Viral or self antigen OX40–OX40L
MHC which is based on transgenic expression of virus anti-
gens in β-cells, has been extensively used to investigate
Tolerogenic mechanisms that could be involved in virus-induced
cDC
T1D and how self tolerance could be broken96,97. These
mice seemed to be tolerant of viral antigen, as no sponta-
iNKT cell
neous T1D developed. However, following lCmv infec-
Figure 3 | Effects of viral infection on the regulation of type 1 diabetes. Infection
tion all of the transgenic mice became diabetic within a
of the pancreas by viruses triggers the activation and recruitment
NatureofReviews
innate immune cells
| Immunology
such as macrophages, dendritic cells (DCs) and natural killer (NK) cells, which produce
few weeks owing to the destruction of pancreatic β-cells
pro-inflammatory cytokines through cytoplasmic pattern recognition receptor (PRR) by virus-specific CTls. These virus-induced T1D mod-
activation. In parallel, viruses can enhance the release of β-cell antigens through direct or els have help to reveal the key role for cytokines and
indirect β-cell damage. Activated antigen-presenting cells can present either self chemokines, produced by immune cells and even the
antigen or viral antigens (in the case of molecular mimicry) to islet antigen-specific β-cells themselves, in the development of T1D. Blocking
T cells. The pro-inflammatory milieu created by the innate immune cells promotes the or depletion of IFNγ, TNF or CXC-chemokine ligand 10
recruitment and activation of islet antigen-specific T cells in the pancreas. Viral infection (CXCl10) reduced T1D incidence in these mice follow-
can be controlled by invariant NKT (iNKT) cell–plasmacytoid DC (pDC) crosstalk leading ing infection, revealing the crucial role for the inflam-
to a rapid elimination of the virus from the pancreas. In a second step, activated matory milieu in the development of virus-induced
tolerogenic conventional DCs (cDCs) and pDCs loaded with viral and/or self antigens
T1D98–100. These studies show that viral infections could
can induce regulatory T (TReg) cells, which mediate bystander suppression, through
transforming growth factor-β (TGFβ) and interleukin-10 (IL-10). The complex interplay
induce T1D possibly by the activation of diabetogenic
between viruses and immune cells reflects the dual role, beneficial or detrimental, of T cells that are cross-reactive for both viral and self anti-
infections in the development of type 1 diabetes. IFN, interferon; NKG2D, natural killer gen, but more importantly they show that viruses can
group 2, member D; RAE1, retinoic acid early transcript 1; TCR, T cell receptor. induce T1D by creating an inflammatory environment
favourable for diabetogenic T cell activation.

receptors for host responses to viral dsRNA86. Several Activation of diabetogenic T cells through bystander
Molecular mimicry studies have suggested a genetic association between damage. The bystander damage resulting from virus
Resemblance between epitopes mDA5 allelic variants and susceptibility to T1D 87. infection could involve nonspecific activation of auto-
contained in microbial and host
Different viruses can induce or promote T1D through immune cells by a pro-inflammatory milieu and/or
proteins, leading to cross-
reactivity of T cells in the host. various mechanisms, including molecular mimicry, induc- T cell activation through exposure to released islet
tion of bystander damage of β-cells, release of sequestered antigens. The association of bystander damage with
Insulitis antigens and changes in the balance between the num- T1D onset was first revealed after coxsackie virus B4
An infiltration of lymphocytes bers of TReg cells and effector T cells (TABLe 1). There is infection of BDC2.5 TCR-transgenic NoD mice, which
into pancreatic islets during the
progression of type 1 diabetes.
some evidence that viruses can directly destroy pancreatic induced rapid T1D101. T cells in these mice express a
Insulitis can be innocuous or β-cells but in many cases there seems to be a requirement TCR specific for a pancreatic antigen but interestingly,
destructive. for immune cells in the β-cell destruction process. these mice have a lower incidence of T1D and develop

NATURE REvIEwS | Immunology volUmE 10 | jUly 2010 | 507

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

Table 1 | Infections associated with the regulation of type 1 diabetes


Pathogen Effect on mechanisms of action Refs
type 1 diabetes
Viral infections
Coxsackie B3 and B4 viruses Acceleration Bystander damage, release of sequestered antigens and molecular mimicry 92,101,
104–107,
Prevention Upregulation of PDL1 expression and TReg cell-mediated control of
110,111,132
diabetogenic CTL expansion
Cytomegalovirus Acceleration Molecular mimicry and congenital infections 95,133
Encephalomyocarditis virus Acceleration Direct destruction 88–90
Mumps virus Acceleration Unknown 9
Parvovirus Acceleration Change in TReg cell and effector T cell balance 133,134
Rhesus monkey rotavirus Acceleration Acceleration by induction of pro-inflammatory cytokines 108,109
Prevention Prevention mechanism unknown
Rubella virus Acceleration Molecular mimicry and congenital infections 94
Lymphocytic choriomeningitis virus Prevention Upregulation of PDL1 expression and TReg cell-mediated control of 111,135
diabetogenic CTL expansion
Lactate dehydrogenase virus Prevention Unknown (possible role for antigen-presenting macrophages) 136
Mouse hepatitis virus Prevention Unknown (only observational) 137
Murine gammaherpes virus-68 Delay Alteration of self-antigen presentation by DCs 138
Viral antigens
CpG DNA Prevention Decrease in T cell proliferative responses and possible shift to TH2 cell responses 116
CpG DNA No effect Not applicable 115
PolyI:C Prevention Expression of type I IFNs and induction of suppressor T cells 114
Bacterial infections
Salmonella enterica subsp. enterica Prevention TH1 cell- and IFNγ-mediated inhibition of type 1 diabetes and phenotypic 123,124*
serovar Typhimurium changes in DCs and IDO production
Mycobacterium avium Prevention Induction of suppressor T cells 139
Bacterial antigens
Mycobacterium bovis bacille Prevention Unknown (only observational) 120,121
Calmette–Guérin
Complete Freund’s adjuvant Prevention Expansion of natural suppressor cells and NK cells 70,122
(Mycobacteria)
Streptococcal OK-432 Prevention Unknown (only observational) 140
Klebsiella pneumoniae glycoprotein Prevention Expansion of natural suppressor cells 126
extract and Escherichia coli LPS
OM-85 (complex mix of bacterial Prevention Induction of TGFβ, TReg cells and NKT cells 141
extracts)
Fungal antigens
Zymosan Prevention Activation of innate cells, TReg cells 127
Helminth infections
Schistosoma mansoni Prevention Shift to TH2-type response 130
Trichinella spiralis Prevention Shift to TH2-type response 142
Heligmosomoides polygyrus Prevention Shift to TH2-type response 142
Litomosoides sigmodontis Prevention Shift to TH2-type response and TReg cell increase 143
Helminth antigens
L. sigmodontis antigen Prevention Shift to TH2-type response and TReg cell increase 143
Dirofilaria immitis IgE-inducing antigen Prevention Shift to TH2-type response 144
S. mansoni soluble worm antigen or Prevention Expansion of alternatively activated macrophages, tolerogenic DCs and 128,129,
soluble egg antigen iNKT cells, TH2 cells and TReg cells 131
S. mansoni eggs Prevention Induction of TH2 cells 130
APC, antigen-presenting cell; CTL, cytotoxic T lymphocyte; DC, dendritic cell; IFN, interferon; IDO, indoleamine 2,3-dioxygenase; LPS, lipopolysaccharide;
NK, natural killer; PDL1, programmed cell death ligand 1; polyI:C, polyinosinic–polycytidylic acid; TGFβ, transforming-growth factor-β; TH, T helper; TReg, regulatory T.
*Also observed by S. Newland and A.C., unpublished observations.

508 | jUly 2010 | volUmE 10 www.nature.com/reviews/immunol

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

non-invasive insulitis102,103. Coxsackie virus B4 infec- compared with naive TReg cells from uninfected mice.
tion of NoD mice also led to the acceleration of T1D Positive correlations have been observed between lCmv
development but only in mice at least 8 weeks of age infection and PDl1 upregulation, enhanced TReg cell
with a pancreatic infiltrate of islet antigen-specific number and tolerogenic function following infection111. It
T cells; younger mice were resistant to the disease104. would be interesting to determine whether viral infections
In the NoD mouse model, an accumulation of a crucial upregulate PDl1 expression on β-cells, which is already
number of islet antigen-specific T cells was required for expressed in uninfected NoD mice and participates in the
the induction of T1D by coxsackie virus B4 and other control of diabetogenic T cell attack112.
coxsackie virus B strains105. Treatment with the TlR3 The use of RIP–lCmv model has enabled the
ligand polyinosinic–polycytidylic acid (polyI:C), a syn- identification of a new immune cell crosstalk that con-
thetic double-stranded RNA that induces the secretion trols pancreatic viral load and thereby T1D onset 66,113.
of large amounts of IFNα by DCs and macrophages, is Following infection with lCmv, iNKT cells promote
not sufficient to induce T1D in BDC2.5 TCR-transgenic pDCs recruitment into the pancreas and their produc-
NoD mice106. These results suggest that nonspecific acti- tion of type I IFNs. This local iNKT–pDC crosstalk is
vation of T cells mediated by virus-induced inflammatory dependent on the oX40–oX40l pathway. iNKT cells in
cytokines is not sufficient to induce disease. the pancreas, but not in lymphoid tissues, express oX40,
Coxsackie virus B4-specific innate immune and experiments with blocking antibodies, as well as
responses result in pancreatic tissue damage and the the transfer of wild-type iNKT cells to oX40-deficient
capture of released self antigens by APCs. Coxsackie mice, have shown the crucial role of oX40–oX40l in
virus B4 did not induce direct β-cells death after infec- the iNKT cell–pDC interaction. As a result, in the pan-
tion; it triggered the upregulation of cell surface mark- creas (but not in the spleen), iNKT cells enhance IFNα
ers on β-cells associated with cellular stress, particularly production by pDCs and inhibit local viral replication.
death receptor 4, and subsequent engulfment of infected The low viral burden in the pancreas results in a weak
islets by macrophages107. These macrophages induced local adaptive immune response, thereby preventing tis-
T1D following transfer into uninfected BDC2.5 TCR- sue damage and T1D development. These results reveal
transgenic NoD mice, showing their involvement in the that the diabetogenic effect of lCmv can be controlled
induction of T1D following viral infection. Therefore, by efficient innate immune cell crosstalk, limiting the
viral infection could have a role in T1D development by activation of diabetogenic T cells.
potentiating ongoing autoimmune disease through the viral antigens in the absence of infection also prevent
induction of inflammatory cytokines and the stimula- diabetes in NoD mice. For example, polyI:C can pro-
tion of self-antigen-presenting APCs. tect NoD mice from developing T1D114 and, although
controversial115, vaccination using CpG-containing
Inhibiting disease. Although some viruses have been oligodeoxynucleotides (which are TlR9 ligands) also
shown to promote T1D pathogenesis, infections have also inhibited T1D development in NoD mice116. Recent
been shown to have a protective role in T1D (TABLe 1). This data from the streptozotocin model of T1D suggest that
dual role of viruses in T1D depends on the different effects TlR9 agonists might inhibit diabetes onset through the
that the virus might have on β-cells or in the modulation induction of IDo expression by DCs117. TlR3 and TlR9
of molecules and cells involved in controlling T1D. are expressed not only by cells of the immune system but
An example of the dual role of a virus in T1D is the also by β-cells118, raising the possibility that these TlR
rhesus monkey rotavirus (RRv), which provides protec- ligands function at several levels.
tion when inoculated into young NoD mice but exac- There are many studies showing reduced insulitis
erbates disease if administered when insulitis is already and inhibition of T1D development following expo-
established108,109. Similar effects on T1D development are sure of NoD mice to mycobacterial species or anti-
observed with coxsackie virus infection of NoD mice. In gens119–122 and Salmonella enterica subsp. enterica serovar
this case, the virus precipitates disease after a crucial level Typhimurium123. Given that T1D is a TH1 cell-mediated
of islet antigen-specific immune cells has accumulated in disease, this is intriguing as both infections are known
the pancreas, but can also prevent T1D in pre-diabetic to activate macrophages and DCs that induce a typical
NoD mice110. Coxsackie viruses can control the expan- TH1 cell-mediated immune response. S. Typhimurium
sion of islet antigen-specific CTls by upregulating PDl1 infection prevents T1D in NoD mice with an already
expression on lymphocytes. As the interaction between established infiltrate in the pancreas123. The transfer of
PD1 and PDl1 is known to control not only T cell DC-enriched populations from S. Typhimurium-infected
immunity to viral infection but also autoimmune mice to NoD mice injected with the chemotherapeutic
T cell responses, the hypothesis that viral infection can drug cyclophosphamide to accelerate T1D prevented dis-
potentiate regulatory mechanisms is consistent with ease development 124. In this model, the immunomodula-
the protective effects observed on T1D development. tory effect was due to the ability of the transferred DCs to
Increased numbers of CD4+CD25+ TReg cells are also prevent the trafficking of islet antigen-specific T cells into
observed in coxsackie virus infection111. TReg cells from the pancreas. The paradoxical protective effect of TH1
infected NoD mice express high levels of TGFβ, CTlA4 cell-inducing infections (and their antigens (TABLe 1)) on
and glucocorticoid-induced TNF-receptor-related a TH1 cell-mediated autoimmune disease indicates that
protein (GITR), and have increased ability to regulate a classic pro-inflammatory immune response is not suf-
the onset of T1D during the pre-diabetic phase, when ficient to exacerbate disease per se. Intracellular infections

NATURE REvIEwS | Immunology volUmE 10 | jUly 2010 | 509

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

such as S. Typhimurium can induce the expression of Pathogens such as parasites and bacteria are usu-
IFNγ, resulting in induction of anti-inflammatory mol- ally protective against T1D, whereas viruses have a dual
ecules such as IDo, not just in lymphoid tissues but role. The ability of viruses to induce or promote T1D can
also in the pancreas (S. Newland and A.C., unpublished depend on a susceptible genetic background, the tro-
observations). IDo has been shown to be produced by pism of the virus for islet cells and its ability to induce a
human pancreatic β-cells in response to IFNγ and its role strong pro-inflammatory response10. Conversely, virus
has been proposed as a self-defence mechanism against infection in the presence of a limited number or absence
inflammation125. Functional evidence of a tolerogenic of diabetogenic T cells is usually beneficial against T1D.
role of islet IDo in vivo has been shown by prolonged The local pancreatic viral load is a crucial parameter and,
survival of IDo-expressing islets following transplan- therefore, the efficiency of immune cell crosstalk con-
tation65. Furthermore, an inverse correlation has been trolling viral replication, subsequent β-cell damage and
observed between IDo and apoptosis, suggesting that pro-inflammatory cytokine secretion will determine the
IDo may have an additional protective role by reducing outcome of the infection on T1D.
oxidation and mHC class I expression.
PRR ligands of either bacterial126 or fungal origin127 Concluding remarks and future directions
can also prevent diabetes in NoD mice. The fungal cell Studies of the pathogenesis of T1D have largely focused
wall component zymosan is recognized by innate on the analysis of diabetogenic T cells and their control
cells through an interaction with TlR2 and the C-type by TReg cells. However, there is increasing evidence that
lectin receptor dectin 1 (also known as ClEC7A). innate immune cells have crucial roles in T1D patho-
Zymosan induces phenotypic changes in DCs and genesis. Innate cells such as macrophages, DCs and NK
macrophages, which results in the secretion of both cells are required for the development of T1D in vari-
pro-inflammatory cytokines (Il-6 and TNF) and anti- ous mouse models and these cells have been detected
inflammatory cytokines (Il-10 and TGFβ)127. Zymosan in the pancreas of patients with T1D. Innate cells such
also upregulated the expression of Il-10 and TGFβ by as DCs, NK and iNKT cells have also been shown to be
CD4+ T cells127. These changes seem to be involved in involved in protection against this disease. This Review
the suppression of insulitis. highlights the potential for the ambivalent function of
The expression of TlR2 and TlR4 by β-cells adds innate immune cells in T1D. many observations sup-
another site at which these agents can potentially func- port a protective role for these cells following their
tion118. Interestingly, it has been shown that gut microbi- activation, by specific agonist or following microbial
ota may have a crucial role in T1D development46. Studies infection, in younger mice harbouring limited β-cell
in myeloid differentiation primary-response protein 88 destruction and diabetogenic T cell frequency. one of
(myD88)-deficient NoD mice kept in germ-free con- the key questions that remain to be addressed is why
ditions have revealed normal disease development in these innate immune cells are inefficient in preventing
these mice, whereas a decreased incidence was observed T1D in the absence of exogenous triggering but instead
following gut colonization with commensal microorgan- participate in the development of the disease. one
isms. This suggests that some organisms can prevent T1D hypothesis is that T1D could be associated with some
development in a myD88-independent manner but does immune deficiency of innate immune cells render-
not exclude the involvement of other PRRs. ing them unable to induce tolerance to islet antigens,
Perhaps less surprising, but also complex, is the effect whereas chronic low activation of these cells through
of parasitic infections on T1D. Parasitic worms (and continued β-cell death and/or persistent virus activates
their products) can rapidly skew the immune response their pathogenic functions.
of the host towards an anti-inflammatory response, The knowledge gained from the analysis of the pre-
in order to secure their own survival. The common ventative mechanisms induced by some infections sug-
immune response observed during helminth infection gests two main pathways for the development of new
(or immunization with parasite products) is character- therapeutic approaches for T1D. The first could be
ized by the induction of an immature phenotype in DCs based on parasite or bacterial components that are pro-
and the alternative activation of macrophages128, as well tective in mouse models of T1D or in other immune-
as TH2 and TReg cell129 development. Schistosoma mansoni mediated pathologies in humans such as inflammatory
infection or injection of S. mansoni antigens dramatically bowel disease and asthma. The second could focus on
reduces T1D incidence in NoD mice130,131. After exposure specific triggering of innate cells, such as pDCs and
to S. mansoni soluble egg antigen (SEA), for example, the NKT cells, that preferentially exert a protective role
composition of the cellular infiltrate in the pancreas of against T1D. However, as pDCs can also have delete-
NoD mice is markedly changed. In particular, there is rious effects, similar to cDCs, their triggering by PRR
increased expression of FoXP3, Il-4, Il-10 and TGFβ ligands should be restricted to individuals at risk but
by CD4+ T cells128,129. The number of iNKT cells is also still devoid of any sign of islet-specific autoimmunity,
increased in NoD mice exposed to SEA131. Thus in as indicated by the presence of specific autoantibodies
the context of a live infection with S. mansoni, several and islet antigen-specific T cells. Concerning the exog-
different cell types of both the innate and the adaptive enous activation of iNKT cells, many investigations are
immune responses are affected with the potential to currently underway to generate and screen new agonists
influence each other and reinforce their ability to inhibit to obtain molecules that preferentially direct NKT cells
autoimmune pathology. towards regulatory pathways.

510 | jUly 2010 | volUmE 10 www.nature.com/reviews/immunol

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

1. Banting, F. G. & Best, C. H. The internal secretion of 23. Xiu, Y. et al. B lymphocyte depletion by CD20 41. Lacy, P. E., Davie, J. M. & Finke, E. H. Prolongation of
the pancreas. J. Lab. Clin. Med. 7, 465–480 (1922). monoclonal antibody prevents diabetes in nonobese islet allograft survival following in vitro culture (24
2. Redondo, M. J., Fain, P. R. & Eisenbarth, G. S. diabetic mice despite isotype-specific differences in degrees C) and a single injection of ALS. Science 204,
Genetics of type 1A diabetes. Recent Prog. Horm. Res. FcγR effector functions. J. Immunol. 180, 2863–2875 312–313 (1979).
56, 69–89 (2001). (2008). 42. Faustman, D. L. et al. Prevention of rejection of
3. Kyvik, K. O., Green, A. & Beck-Nielsen, H. 24. Hutchings, P. et al. Transfer of diabetes in mice murine islet allografts by pretreatment with anti-
Concordance rates of insulin dependent diabetes prevented by blockade of adhesion-promoting dendritic cell antibody. Proc. Natl Acad. Sci. USA 81,
mellitus: a population based study of young Danish receptor on macrophages. Nature 348, 639–642 3864–3868 (1984).
twins. BMJ 311, 913–917 (1995). (1990). 43. Ludewig, B., Odermatt, B., Landmann, S.,
4. Redondo, M. J., Jeffrey, J., Fain, P. R., Eisenbarth, G. S. The role of macrophages in the pathogenesis of Hengartner, H. & Zinkernagel, R. M. Dendritic cells
& Orban, T. Concordance for islet autoimmunity T1D was first shown by blocking macrophage induce autoimmune diabetes and maintain disease
among monozygotic twins. N. Engl. J. Med. 359, adhesion with a specific monoclonal antibody. This via de novo formation of local lymphoid tissue.
2849–2850 (2008). treatment prevented insulitis and T1D in NOD J. Exp. Med. 188, 1493–1501 (1998).
5. Dunne, D. W. & Cooke, A. A worm’s eye view of the mice. 44. Marleau, A. M., Summers, K. L. & Singh, B.
immune system: consequences for evolution of human 25. Jun, H. S., Yoon, C. S., Zbytnuik, L., van Rooijen, N. & Differential contributions of APC subsets to T cell
autoimmune disease. Nature Rev. Immunol. 5, Yoon, J. W. The role of macrophages in T cell-mediated activation in nonobese diabetic mice. J. Immunol.
420–426 (2005). autoimmune diabetes in nonobese diabetic mice. 180, 5235–5249 (2008).
6. Turley, S., Poirot, L., Hattori, M., Benoist, C. & J. Exp. Med. 189, 347–358 (1999). 45. Kim, H. S. et al. Toll-like receptor 2 senses β-cell death
Mathis, D. Physiological β cell death triggers priming 26. Alleva, D. G., Pavlovich, R. P., Grant, C., Kaser, S. B. & and contributes to the initiation of autoimmune
of self-reactive T cells by dendritic cells in a type-1 Beller, D. I. Aberrant macrophage cytokine production diabetes. Immunity 27, 321–333 (2007).
diabetes model. J. Exp. Med. 198, 1527–1537 is a conserved feature among autoimmune-prone 46. Wen, L. et al. Innate immunity and intestinal
(2003). mouse strains: elevated interleukin (IL)-12 and an microbiota in the development of type 1 diabetes.
This study reveals the role of physiological β-cell imbalance in tumor necrosis factor-α and IL-10 define Nature 455, 1109–1113 (2008).
apoptosis in young NOD mice, which induces the a unique cytokine profile in macrophages from young This study based on the analysis of MYD88-deficient
presentation of islet antigen by DCs and nonobese diabetic mice. Diabetes 49, 1106–1115 NOD mice suggests that the gut microbiota have a
subsequent islet antigen-specific T cell priming in (2000). crucial role in the prevention of T1D.
the draining lymph nodes. 27. Martin, A. P. et al. Increased expression of CCL2 in 47. Poligone, B., Weaver, D. J. Jr, Sen, P., Baldwin, A. S. Jr
7. Barrett, J. C. et al. Genome-wide association study insulin-producing cells of transgenic mice promotes & Tisch, R. Elevated NF-κB activation in nonobese
and meta-analysis find that over 40 loci affect risk of mobilization of myeloid cells from the bone marrow, diabetic mouse dendritic cells results in enhanced
type 1 diabetes. Nature Genet. 41, 703–707 marked insulitis, and diabetes. Diabetes 57, APC function. J. Immunol. 168, 188–196 (2002).
(2009). 3025–3033 (2008). 48. Steptoe, R. J., Ritchie, J. M. & Harrison, L. C.
8. Reimann, M. et al. An update on preventive and 28. Yang, L. J. Big mac attack: does it play a direct role Increased generation of dendritic cells from myeloid
regenerative therapies in diabetes mellitus. for monocytes/macrophages in type 1 diabetes? progenitors in autoimmune-prone nonobese diabetic
Pharmacol. Ther. 121, 317–331 (2009). Diabetes 57, 2922–2923 (2008). mice. J. Immunol. 168, 5032–5041 (2002).
9. Hyoty, H. et al. Decline of mumps antibodies in 29. Arnush, M., Scarim, A. L., Heitmeier, M. R., 49. Lande, R. & Gilliet, M. Plasmacytoid dendritic cells:
type 1 (insulin-dependent) diabetic children and a Kelly, C. B. & Corbett, J. A. Potential role of resident key players in the initiation and regulation of immune
plateau in the rising incidence of type 1 diabetes islet macrophage activation in the initiation of responses. Ann. NY Acad. Sci. 1183, 89–103
after introduction of the mumps–measles–rubella autoimmune diabetes. J. Immunol. 160, 2684–2691 (2010).
vaccine in Finland. Childhood Diabetes in Finland (1998). 50. Stewart, T. A. et al. Induction of type I diabetes by
Study Group. Diabetologia 36, 1303–1308 30. Dahlen, E., Dawe, K., Ohlsson, L. & Hedlund, G. interferon-α in transgenic mice. Science 260,
(1993). Dendritic cells and macrophages are the first and 1942–1946 (1993).
10. von Herrath, M. G., Fujinami, R. S. & Whitton, J. L. major producers of TNF-α in pancreatic islets in the 51. Huang, Y., Blatt, L. M. & Taylor, M. W. Type 1
Microorganisms and autoimmunity: making the nonobese diabetic mouse. J. Immunol. 160, interferon as an antiinflammatory agent: inhibition
barren field fertile? Nature Rev. Microbiol. 3585–3593 (1998). of lipopolysaccharide-induced interleukin-1β and
1, 151–157 (2003). 31. Cantor, J. & Haskins, K. Recruitment and activation induction of interleukin-1 receptor antagonist.
11. Sibley, R. K., Sutherland, D. E., Goetz, F. & of macrophages by pathogenic CD4 T cells in type 1 J. Interferon Cytokine Res. 15, 317–321 (1995).
Michael, A. F. Recurrent diabetes mellitus in the diabetes: evidence for involvement of CCR8 and CCL1. 52. Li, Q. et al. Interferon-α initiates type 1 diabetes in
pancreas iso- and allograft. A light and electron J. Immunol. 179, 5760–5767 (2007). nonobese diabetic mice. Proc. Natl Acad. Sci. USA
microscopic and immunohistochemical analysis of 32. Uno, S. et al. Macrophages and dendritic cells 105, 12439–12444 (2008).
four cases. Lab. Invest. 53, 132–144 (1985). infiltrating islets with or without β cells produce 53. Van Belle, T. L., Juntti, T., Liao, J. & von Herrath, M. G.
12. Phillips, J. M. et al. Type 1 diabetes development tumour necrosis factor-α in patients with recent-onset Pre-existing autoimmunity determines type 1 diabetes
requires both CD4+ and CD8+ T cells and can be type 1 diabetes. Diabetologia 50, 596–601 (2007). outcome after Flt3-ligand treatment. J. Autoimmun.
reversed by non-depleting antibodies targeting both 33. Dotta, F. et al. Coxsackie B4 virus infection of β cells 34, 445–452 (2009).
T cell populations. Rev. Diabet. Stud. 6, 97–103 and natural killer cell insulitis in recent-onset type 1 54. Vuckovic, S. et al. Decreased blood dendritic cell
(2009). diabetic patients. Proc. Natl Acad. Sci. USA 104, counts in type 1 diabetic children. Clin. Immunol.
13. Chatenoud, L., Thervet, E., Primo, J. & Bach, J. F. 5115–5120 (2007). 123, 281–288 (2007).
Anti-CD3 antibody induces long-term remission of This report describes the isolation of coxsackie 55. Chen, X. et al. Type 1 diabetes patients have
overt autoimmunity in nonobese diabetic mice. virus B4 in the pancreas of patients with T1D, significantly lower frequency of plasmacytoid dendritic
Proc. Natl Acad. Sci. USA 91, 123–127 (1994). which was associated with lower β-cell function and cells in the peripheral blood. Clin. Immunol. 129,
14. Chatenoud, L. Immune therapy for type 1 with NK cell infiltrates. 413–418 (2008).
diabetes mellitus—what is unique about anti-CD3 34. Flodstrom, M. et al. Target cell defense prevents the 56. Allen, J. S. et al. Plasmacytoid dendritic cells are
antibodies? Nature Rev. Endocrinol. 6, 149–157 development of diabetes after viral infection. Nature proportionally expanded at diagnosis of type 1
(2010). Immunol. 3, 373–382 (2002). diabetes and enhance islet autoantigen presentation
15. Eizirik, D. L., Colli, M. L. & Ortis, F. The role of 35. Poirot, L., Benoist, C. & Mathis, D. Natural killer cells to T-cells through immune complex capture. Diabetes
inflammation in insulitis and β cell loss in type 1 distinguish innocuous and destructive forms of 58, 138–145 (2009).
diabetes. Nature Rev. Endocrinol. 5, 219–226 pancreatic islet autoimmunity. Proc. Natl Acad. Sci. 57. Ohnmacht, C. et al. Constitutive ablation of dendritic
(2009). USA 101, 8102–8107 (2004). cells breaks self-tolerance of CD4 T cells and results in
16. Wildin, R. S. & Freitas, A. IPEX and FOXP3: clinical 36. Alba, A. et al. Natural killer cells are required for spontaneous fatal autoimmunity. J. Exp. Med. 206,
and research perspectives. J. Autoimmun. 25, accelerated type 1 diabetes driven by interferon-β. 549–559 (2009).
S56–S62 (2005). Clin. Exp. Immunol. 151, 467–475 (2008). 58. Ueno, H. et al. Dendritic cell subsets in health and
17. Salomon, B. et al. B7/CD28 costimulation is essential 37. Brauner, H. et al. Distinct phenotype and function of disease. Immunol. Rev. 219, 118–142 (2007).
for the homeostasis of the CD4+CD25+ NK cells in the pancreas of nonobese diabetic mice. 59. Kared, H. et al. Treatment with granulocyte colony-
immunoregulatory T cells that control autoimmune J. Immunol. 184, 2272–2280 (2010). stimulating factor prevents diabetes in NOD mice by
diabetes. Immunity 12, 431–440 (2000). 38. Feuerer, M., Shen, Y., Littman, D. R., Benoist, C. & recruiting plasmacytoid dendritic cells and functional
18. Tang, Q. & Bluestone, J. A. The Foxp3+ regulatory Mathis, D. How punctual ablation of regulatory T cells CD4+CD25+ regulatory T-cells. Diabetes 54, 78–84
T cell: a jack of all trades, master of regulation. unleashes an autoimmune lesion within the pancreatic (2005).
Nature Immunol. 9, 239–244 (2008). islets. Immunity 31, 654–664 (2009). 60. Chilton, P. M. et al. Flt3-ligand treatment prevents
19. Hu, C. Y. et al. Treatment with CD20-specific antibody This report shows that TReg cell depletion enhances diabetes in NOD mice. Diabetes 53, 1995–2002
prevents and reverses autoimmune diabetes in mice. T1D development, which is associated with an (2004).
J. Clin. Invest. 117, 3857–3867 (2007). increased frequency of activated NK cells in the 61. O’Keeffe, M. et al. Fms-like tyrosine kinase 3 ligand
20. Serreze, D. V. et al. B lymphocytes are essential for pancreas. administration overcomes a genetically determined
the initiation of T cell-mediated autoimmune 39. Gur, C. et al. The activating receptor NKp46 is dendritic cell deficiency in NOD mice and protects
diabetes: analysis of a new “speed congenic” stock essential for the development of type 1 diabetes. against diabetes development. Int. Immunol. 17,
of NOD.Igμnull mice. J. Exp. Med. 184, 2049–2053 Nature Immunol. 11, 121–128 (2010). 307–314 (2005).
(1996). This study highlights a role for NKp46, an 62. Darrasse-Jeze, G. et al. Feedback control of regulatory
21. Pescovitz, M. D. et al. Rituximab, B-lymphocyte activating receptor on NK cells, in the development T cell homeostasis by dendritic cells in vivo.
depletion, and preservation of β-cell function. of T1D and provides evidence for a direct J. Exp. Med. 206, 1853–1862 (2009).
N. Engl. J. Med. 361, 2143–2152 (2009). interaction between NK cells and β-cells. This study describes a feedback regulatory
22. Martin, S. et al. Development of type 1 diabetes 40. Ogasawara, K. et al. NKG2D blockade prevents loop between TReg cells and DCs. Increasing DC
despite severe hereditary B-lymphocyte deficiency. autoimmune diabetes in NOD mice. Immunity 20, frequency increases TReg cell frequency thereby
N. Engl. J. Med. 345, 1036–1040 (2001). 757–767 (2004). preventing T1D.

NATURE REvIEwS | Immunology volUmE 10 | jUly 2010 | 511

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

63. Saxena, V., Ondr, J. K., Magnusen, A. F., Munn, D. H. 84. Oikawa, Y. et al. High frequency of Vα24+ Vβ11+ 105. Drescher, K. M., Kono, K., Bopegamage, S.,
& Katz, J. D. The countervailing actions of myeloid and T-cells observed in type 1 diabetes. Diabetes Care 25, Carson, S. D. & Tracy, S. Coxsackievirus B3 infection
plasmacytoid dendritic cells control autoimmune 1818–1823 (2002). and type 1 diabetes development in NOD mice:
diabetes in the nonobese diabetic mouse. J. Immunol. 85. Kukreja, A. et al. Multiple immuno-regulatory defects insulitis determines susceptibility of pancreatic islets
179, 5041–5053 (2007). in type-1 diabetes. J. Clin. Invest. 109, 131–140 to virus infection. Virology 329, 381–394 (2004).
This article shows the contrasting roles of DC (2002). 106. Horwitz, M. S., Ilic, A., Fine, C., Rodriguez, E. &
subsets in the development of T1D: cDCs promote 86. Kato, H. et al. Differential roles of MDA5 and RIG-I Sarvetnick, N. Presented antigen from damaged
T1D, whereas pDCs are protective and induce IDO helicases in the recognition of RNA viruses. Nature pancreatic β cells activates autoreactive T cells in
production in the pancreatic islets. 441, 101–105 (2006). virus-mediated autoimmune diabetes. J. Clin. Invest.
64. Grohmann, U. et al. A defect in tryptophan 87. Nejentsev, S., Walker, N., Riches, D., Egholm, M. & 109, 79–87 (2002).
catabolism impairs tolerance in nonobese diabetic Todd, J. A. Rare variants of IFIH1, a gene implicated in 107. Horwitz, M. S., Ilic, A., Fine, C., Balasa, B. &
mice. J. Exp. Med. 198, 153–160 (2003). antiviral responses, protect against type 1 diabetes. Sarvetnick, N. Coxsackieviral-mediated diabetes:
65. Alexander, A. M. et al. Indoleamine 2, 3-dioxygenase Science 324, 387–389 (2009). induction requires antigen-presenting cells and is
expression in transplanted NOD Islets prolongs graft 88. Yoon, J. W., McClintock, P. R., Onodera, T. & accompanied by phagocytosis of β cells. Clin. Immunol.
survival after adoptive transfer of diabetogenic Notkins, A. L. Virus-induced diabetes mellitus. 110, 134–144 (2004).
splenocytes. Diabetes 51, 356–365 (2002). XVIII. Inhibition by a nondiabetogenic variant of 108. Graham, K. L. et al. Rotavirus infection of infant and
66. Diana, J. et al. NKT Cell-plasmacytoid dendritic cell encephalomyocarditis virus. J. Exp. Med. 152, young adult nonobese diabetic mice involves
cooperation via OX40 controls viral infection in a 878–892 (1980). extraintestinal spread and delays diabetes onset.
tissue-specific manner. Immunity 30, 289–299 89. Baek, H. S. & Yoon, J. W. Direct involvement of J. Virol. 81, 6446–6458 (2007).
(2009). macrophages in destruction of β-cells leading to 109. Graham, K. L. et al. Rotavirus infection accelerates
This study describes the cooperation between development of diabetes in virus-infected mice. type 1 diabetes in mice with established insulitis.
iNKT cells and pDCs in the pancreas to control viral Diabetes 40, 1586–1597 (1991). J. Virol. 82, 6139–6149 (2008).
replication and dampen local T cell responses. 90. Lee, Y. S., Li, N., Shin, S. & Jun, H. S. Role of nitric 110. Serreze, D. V. et al. Diabetes acceleration or
67. Carnaud, C., Gombert, J., Donnars, O., Garchon, H. & oxide in the pathogenesis of encephalomyocarditis prevention by a coxsackievirus B4 infection: critical
Herbelin, A. Protection against diabetes and improved virus-induced diabetes in mice. J. Virol. 83, requirements for both interleukin-4 and γ interferon.
NK/NKT cell performance in NOD.NK1.1 mice 8004–8011 (2009). J. Virol. 79, 1045–1052 (2005).
congenic at the NK complex. J. Immunol. 166, 91. Shibasaki, S. et al. Expression of Toll-like receptors 111. Filippi, C. M., Estes, E. A., Oldham, J. E. & von
2404–2411 (2001). in the pancreas of recent-onset fulminant type 1 Herrath, M. G. Immunoregulatory mechanisms
68. Ogasawara, K. et al. Impairment of NK cell function by diabetes. Endocr. J. 23, 211–219 (2009). triggered by viral infections protect from type 1
NKG2D modulation in NOD mice. Immunity 18, 92. Atkinson, M. A. et al. Cellular immunity to a diabetes in mice. J. Clin. Invest. 119, 1515–1523
41–51 (2003). determinant common to glutamate decarboxylase (2009).
69. Rodacki, M. et al. Altered natural killer cells in type 1 and coxsackie virus in insulin-dependent diabetes. 112. Ansari, M. J. et al. The programmed death-1 (PD-1)
diabetic patients. Diabetes 56, 177–185 (2007). J. Clin. Invest. 94, 2125–2129 (1994). pathway regulates autoimmune diabetes in nonobese
70. Lee, I. F., Qin, H., Trudeau, J., Dutz, J. & Tan, R. 93. Honeyman, M. C., Stone, N. L., Falk, B. A., Nepom, G. diabetic (NOD) mice. J. Exp. Med. 198, 63–69
Regulation of autoimmune diabetes by complete & Harrison, L. C. Evidence for molecular mimicry (2003).
Freund’s adjuvant is mediated by NK cells. J. Immunol. between human T cell epitopes in rotavirus and 113. Diana, J. & Lehuen, A. NKT cells: Friend or foe during
172, 937–942 (2004). pancreatic islet autoantigens. J. Immunol. 184, viral infections? Eur. J. Immunol. 39, 3283–3291
71. Beilke, J. N., Kuhl, N. R., Van Kaer, L. & Gill, R. G. 2204–2210 (2010). (2009).
NK cells promote islet allograft tolerance via a 94. Ou, D., Mitchell, L. A., Metzger, D. L., Gillam, S. & 114. Serreze, D. V., Hamaguchi, K. & Leiter, E. H.
perforin-dependent mechanism. Nature Med. 11, Tingle, A. J. Cross-reactive rubella virus and glutamic Immunostimulation circumvents diabetes in NOD/Lt
1059–1065 (2005). acid decarboxylase (65 and 67) protein determinants mice. J. Autoimmun. 2, 759–776 (1989).
72. Novak, J., Griseri, T., Beaudoin, L. & Lehuen, A. recognised by T cells of patients with type I diabetes 115. Lee, B. J. et al. Limited effect of CpG ODN in preventing
Regulation of type 1 diabetes by NKT cells. Int. Rev. mellitus. Diabetologia 43, 750–762 (2000). type 1 diabetes in NOD mice. Yonsei Med. J. 46,
Immunol. 26, 49–72 (2007). 95. Pak, C. Y., Cha, C. Y., Rajotte, R. V., McArthur, R. G. & 341–346 (2005).
73. Hammond, K. J. et al. α/β-T cell receptor (TCR)+CD4– Yoon, J. W. Human pancreatic islet cell specific 38 116. Quintana, F. J., Rotem, A., Carmi, P. & Cohen, I. R.
CD8– (NKT) thymocytes prevent insulin-dependent kilodalton autoantigen identified by cytomegalovirus- Vaccination with empty plasmid DNA or CpG
diabetes mellitus in nonobese diabetic (NOD)/Lt mice induced monoclonal islet cell autoantibody. oligonucleotide inhibits diabetes in nonobese diabetic
by the influence of interleukin (IL)-4 and/or Diabetologia 33, 569–572 (1990). mice: modulation of spontaneous 60-kDa heat shock
IL-10. J. Exp. Med. 187, 1047–1056 (1998). 96. Oldstone, M. B., Nerenberg, M., Southern, P., Price, J. protein autoimmunity. J. Immunol. 165, 6148–6155
74. Lehuen, A. et al. Overexpression of natural killer & Lewicki, H. Virus infection triggers insulin- (2000).
T cells protects Vα14–Jα281 transgenic nonobese dependent diabetes mellitus in a transgenic model: 117. Fallarino, F. et al. IDO mediates TLR9-driven
diabetic mice against diabetes. J. Exp. Med. 188, role of anti-self (virus) immune response. Cell 65, protection from experimental autoimmune diabetes.
1831–1839 (1998). 319–331 (1991). J. Immunol. 183, 6303–6312 (2009).
References 73 and 74 are the first reports 97. Ohashi, P. S. et al. Ablation of “tolerance” and 118. Wen, L., Peng, J., Li, Z. & Wong, F. S. The effect of
describing the role of iNKT cells in preventing T1D. induction of diabetes by virus infection in viral antigen innate immunity on autoimmune diabetes and the
75. Sharif, S. et al. Activation of natural killer T cells by transgenic mice. Cell 65, 305–317 (1991). expression of Toll-like receptors on pancreatic islets.
α-galactosylceramide treatment prevents the onset 98. Seewaldt, S. et al. Virus-induced autoimmune J. Immunol. 172, 3173–3180 (2004).
and recurrence of autoimmune type 1 diabetes. diabetes: most β-cells die through inflammatory 119. Bras, A. & Aguas, A. P. Diabetes-prone NOD mice are
Nature Med. 7, 1057–1062 (2001). cytokines and not perforin from autoreactive (anti- resistant to Mycobacterium avium and the infection
76. Hong, S. et al. The natural killer T-cell ligand viral) cytotoxic T-lymphocytes. Diabetes 49, prevents autoimmune disease. Immunology 89,
α-galactosylceramide prevents autoimmune diabetes 1801–1809 (2000). 20–25 (1996).
in non-obese diabetic mice. Nature Med. 7, 99. Christen, U. et al. A dual role for TNF-α in type 1 120. Baxter, A. G., Healey, D. & Cooke, A. Mycobacteria
1052–1056 (2001). diabetes: islet-specific expression abrogates the precipitate autoimmune rheumatic disease in NOD
77. Mizuno, M. et al. Synthetic glycolipid OCH prevents ongoing autoimmune process when induced late but mice via an adjuvant-like activity. Scand. J. Immunol.
insulitis and diabetes in NOD mice. J. Autoimmun 23, not early during pathogenesis. J. Immunol. 166, 39, 602–606 (1994).
293–300 (2004). 7023–7032 (2001). 121. Harada, M., Kishimoto, Y. & Makino, S. Prevention of
78. Forestier, C. et al. Improved outcomes in NOD mice 100. Christen, U., McGavern, D. B., Luster, A. D., von overt diabetes and insulitis in NOD mice by a single
treated with a novel Th2 cytokine-biasing NKT cell Herrath, M. G. & Oldstone, M. B. Among CXCR3 BCG vaccination. Diabetes Res. Clin. Pract. 8, 85–89
activator. J. Immunol. 178, 1415–1425 (2007). chemokines, IFN-γ-inducible protein of 10 kDa (CXC (1990).
79. Laloux, V., Beaudoin, L., Jeske, D., Carnaud, C. & chemokine ligand (CXCL) 10) but not monokine 122. Sadelain, M. W., Qin, H. Y., Lauzon, J. & Singh, B.
Lehuen, A. NK T cell-induced protection against induced by IFN-γ (CXCL9) imprints a pattern for the Prevention of type I diabetes in NOD mice by
diabetes in Vα14-Jα281 transgenic nonobese subsequent development of autoimmune disease. adjuvant immunotherapy. Diabetes 39, 583–589
diabetic mice is associated with a Th2 shift J. Immunol. 171, 6838–6845 (2003). (1990).
circumscribed regionally to the islets and functionally 101. Horwitz, M. S. et al. Diabetes induced by coxsackie 123. Zaccone, P. et al. Salmonella typhimurium infection
to islet autoantigen. J. Immunol. 166, 3749–3756 virus: initiation by bystander damage and not halts development of type 1 diabetes in NOD mice.
(2001). molecular mimicry. Nature Med. 4, 781–785 (1998). Eur. J. Immunol. 34, 3246–3256 (2004).
80. Beaudoin, L., Laloux, V., Novak, J., Lucas, B. & This report describes the role of bystander 124. Raine, T., Zaccone, P., Mastroeni, P. & Cooke, A.
Lehuen, A. NKT cells inhibit the onset of diabetes by activation of T cells leading to T1D onset following Salmonella typhimurium infection in nonobese
impairing the development of pathogenic T cells coxsackie virus B4 infection. diabetic mice generates immunomodulatory dendritic
specific for pancreatic β cells. Immunity 17, 102. Katz, J. D., Wang, B., Haskins, K., Benoist, C. & cells able to prevent type 1 diabetes. J. Immunol.
725–736 (2002). Mathis, D. Following a diabetogenic T cell from 177, 2224–2233 (2006).
81. Chen, Y. G. et al. Activated NKT cells inhibit genesis through pathogenesis. Cell 74, 1089–1100 125. Sarkar, S. A. et al. Induction of indoleamine 2,
autoimmune diabetes through tolerogenic recruitment (1993). 3-dioxygenase by interferon-γ in human islets.
of dendritic cells to pancreatic lymph nodes. 103. Stadinski, B. D. et al. Chromogranin A is an Diabetes 56, 72–79 (2007).
J. Immunol. 174, 1196–1204 (2005). autoantigen in type 1 diabetes. Nature Immunol. 11, 126. Sai, P. & Rivereau, A. S. Prevention of diabetes in the
82. Duarte, N. et al. Prevention of diabetes in nonobese 225–231 (2010). nonobese diabetic mouse by oral immunological
diabetic mice mediated by CD1d-restricted nonclassical 104. Serreze, D. V., Ottendorfer, E. W., Ellis, T. M., treatments. Comparative efficiency of human insulin
NKT cells. J. Immunol. 173, 3112–3118 (2004). Gauntt, C. J. & Atkinson, M. A. Acceleration of type 1 and two bacterial antigens, lipopolysacharide from
83. Jordan, M. A., Fletcher, J. M., Pellicci, D. & Baxter, A. G. diabetes by a coxsackievirus infection requires a Escherichia coli and glycoprotein extract from
Slamf1, the NKT cell control gene Nkt1. J. Immunol. preexisting critical mass of autoreactive T-cells in Klebsiella pneumoniae. Diabetes Metab. 22,
178, 1618–1627 (2007). pancreatic islets. Diabetes 49, 708–711 (2000). 341–348 (1996).

512 | jUly 2010 | volUmE 10 www.nature.com/reviews/immunol

© 2010 Macmillan Publishers Limited. All rights reserved


REVIEWS

127. Karumuthil-Melethil, S., Perez, N., Li, R. & Vasu, C. 135. Oldstone, M. B. Viruses as therapeutic agents. I. gastrointestinal helminth infection. Infect. Immun.
Induction of innate immune response through TLR2 Treatment of nonobese insulin-dependent diabetes 75, 397–407 (2007).
and dectin 1 prevents type 1 diabetes. J. Immunol. mice with virus prevents insulin-dependent diabetes 143. Hubner, M. P., Stocker, J. T. & Mitre, E. Inhibition of
181, 8323–8334 (2008). mellitus while maintaining general immune type 1 diabetes in filaria-infected non-obese diabetic
128. Zaccone, P. et al. Immune modulation by Schistosoma competence. J. Exp. Med. 171, 2077–2089 mice is associated with a T helper type 2 shift and
mansoni antigens in NOD mice: effects on both innate (1990). induction of FoxP3+ regulatory T cells. Immunology
and adaptive immune systems. J. Biomed. Biotechnol. This paper documents the ability of LCMV infection 127, 512–522 (2009).
2010, 795210 (2010). to prevent T1D in NOD mice without the general 144. Imai, S., Tezuka, H. & Fujita, K. A factor of inducing
129. Zaccone, P. et al. Schistosoma mansoni egg antigens ablation of T cell function. IgE from a filarial parasite prevents insulin-dependent
induce Treg that participate in diabetes prevention 136. Takei, I. et al. Suppression of development of diabetes diabetes mellitus in nonobese diabetic mice. Biochem.
in NOD mice. Eur. J. Immunol. 39, 1098–1107 in NOD mice by lactate dehydrogenase virus infection. Biophys. Res. Commun. 286, 1051–1058 (2001).
(2009). J. Autoimmun. 5, 665–673 (1992).
These data show the importance of TReg cells in the 137. Wilberz, S., Partke, H. J., Dagnaes-Hansen, F. & Acknowledgements
prevention of T1D in NOD mice by S. mansoni SEA Herberg, L. Persistent MHV (mouse hepatitis virus) We apologize to all the authors whose work we could not cite
through the induction of both indirect (DC infection reduces the incidence of diabetes mellitus in owing to space constrictions. The Cooke laboratory is sup-
mediated) and direct functional changes in non-obese diabetic mice. Diabetologia 34, 2–5 ported by The Wellcome Trust, Medical Research Council
diabetogenic CD4+ T cells. (1991). (MRC) and Diabetes UK. P.Z. is supported by the MRC. The
130. Cooke, A. et al. Infection with Schistosoma mansoni 138. Smith, K. A., Efstathiou, S. & Cooke, A. Murine Lehuen laboratory is supported by the Institut National de la
prevents insulin dependent diabetes mellitus in non- gammaherpesvirus-68 infection alters self-antigen Santé et de la Recherche Médicale (INSERM), Agence
obese diabetic mice. Parasite Immunol. 21, 169–176 presentation and type 1 diabetes onset in NOD mice. Nationale de la Recherche (ANR-GENOPAT) and the European
(1999). J. Immunol. 179, 7325–7333 (2007). Foundation for the Study of Diabetes (EFSD).
131. Zaccone, P. et al. Schistosoma mansoni antigens 139. Martins, T. C. & Aguas, A. P. A role for CD45RBlow
modulate the activity of the innate immune response CD38+ T cells and costimulatory pathways of T-cell Competing interests statement
and prevent onset of type 1 diabetes. Eur. J. Immunol. activation in protection of non-obese diabetic (NOD) The authors declare no competing financial interests.
33, 1439–1449 (2003). mice from diabetes. Immunology 96, 600–605
132. Yoon, J. W., Austin, M., Onodera, T. & Notkins, A. L. (1999).
Isolation of a virus from the pancreas of a child with 140. Toyota, T., Satoh, J., Oya, K., Shintani, S. & Okano, T. DATABASES
diabetic ketoacidosis. N. Engl. J. Med. 300, Streptococcal preparation (OK-432) inhibits UniProtKB: http://www.uniprot.org
1173–1179 (1979). development of type I diabetes in NOD mice. dectin 1 | FLT3L | ICOSL | IDO | IFNγ | IL-1β | KLRG1 | MDA5 |
133. Tirabassi, R. S. et al. Infection with viruses from Diabetes 35, 496–499 (1986). NKG2D | PD1 | PDL1 | RIG-I | TLR2 | TLR3 | TLR7 | TLR9 | TNF
several families triggers autoimmune diabetes in 141. Alyanakian, M. A. et al. Transforming growth factor-
LEW*1WR1 rats: prevention of diabetes by maternal beta and natural killer T-cells are involved in the FURTHER INFORMATION
immunization. Diabetes 59, 110–118 (2010). protective effect of a bacterial extract on type 1 Anne Cooke’s homepage: http://www.path.cam.ac.uk/
134. Zipris, D. et al. Infections that induce autoimmune diabetes. Diabetes 55, 179–185 (2006). research/investigators/cooke/
diabetes in BBDR rats modulate CD4+CD25+ T cell 142. Saunders, K. A., Raine, T., Cooke, A. & Lawrence, C. E. All lInks ARE ACtIvE In thE onlInE Pdf
populations. J. Immunol. 170, 3592–3602 (2003). Inhibition of autoimmune type 1 diabetes by

NATURE REvIEwS | Immunology volUmE 10 | jUly 2010 | 513

© 2010 Macmillan Publishers Limited. All rights reserved

S-ar putea să vă placă și