Categories
Articles

Cooperation of invariant NKT cells and CD4

Original Paper

Pdf

file on Synergy OPEN

omments

Acta Biochim Biophys

Sin 2008, 40: 381-390

doi:10.1111/j.1745-7270.2008.00410.x

Cooperation of invariant NKT cells

and CD4+CD25+ T regulatory cells in

prevention of autoimmune diabetes in non-obese diabetic mice treated with a-galactosylceramide

Weipeng Li1,2#, Fang Ji1#, Yong Zhang1, Ying Wang1, Neng Yang1, Hailiang Ge1, and Fuqing Wang1*

1 Shanghai Institute

of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai

200025, China

2 First

Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China

Received: December

1, 2007       

Accepted: February

20, 2008

#These authors contributed

equally to this work

Abbreviations: a-GalCer, a-galactosylceramide;

CY, cyclophosphamide; DC, dendritic cell; ELISA, enzyme-linked immunosorbent

assay; FITC, fluorescein-isothiocyanate; Foxp3, forkhead/winged helix

transcription factor; IFN, interferon; Ig, immunoglobulin; IL, interleukin;

iNKT, invariant natural killer; NKT, natural killer T; NOD, non-obese diabetic;

PLN, pancreatic lymph node; T1D, type 1 diabetes; TCR, T cell receptor; Th1/2,

T helper type 1/type 2; Treg, regulatory T

*Corresponding

author: Tel, 86-21-63846590, ext 776632; Fax, 86-21-63846383; E-mail,

[email protected]

CD1d-restricted

natural killer T (NKT) cells and CD4+CD25+ regulatory T

(Treg) cells are two thymus-derived subsets of regulatory T cells that play an

important role in the maintenance of self-tolerance. Yet the functional changes

of the two subsets of regulatory T cells in the development of diabetes in

non-obese diabetic (NOD) mice remain unclear, and how NKT cells and CD4+CD25+ Treg cells

cooperate functionally in the regulation of autoimmune diabetes is also

uncertain. We provide evidence that in NOD mice, an animal model of human type

1 diabetes, the functions of both NKT cells and CD4+CD25+ Treg cells

decrease in an age-dependent manner. We show that treatment with a-galactosylceramide

increases the size of the CD4+CD25+ Treg cell

compartment in NOD mice, and augments the expression of forkhead/winged helix

transcription factor and the potency of CD4+CD25+ Treg cells

to inhibit proliferation of CD4+CD25 T cells. Our

data indicate that NKT cells and CD4+CD25+ Treg cells

might cooperate in the prevention of autoimmune diabetes in NOD mice treated

with a-galactosylceramide.

Induced cooperation of NKT cells and CD4+CD25+ Treg cells

could serve as a strategy to treat human autoimmune disease, such as type 1

diabetes.

Keywords        invariant NKT cell; Treg; Foxp3; type 1

diabetes; a-galactosylceramide

CD1d-restricted invariant natural killer T (iNKT) cells are unique

in that they share receptor structures with conventional T cells and NK cells.

The majority of murine iNKT cells use an invariant Va14Ja18 T cell receptor (TCR)

chain paired preferentially with a Vb8.2, Vb7, or Vb2 chain and

recognize lipid antigens presented by CD1d, a major histocompatibility complex

class I-like molecule [1]. The distinctive feature of iNKT cells is their

ability to secrete large amounts of cytokines upon activation. Importantly,

activation of iNKT cells with a superagonist glycosphingolipid such as a-galactosylceramide

(a-GalCer)

can transactivate B cells, NK cells, dendritic cells (DCs), and conventional T

cells, indicating that a-GalCer can act as an adjuvant to promote many antigen-specific

responses during innate and adaptive immunity [26].Characterization of CD1d-restricted iNKT cells in humans with

autoimmune disease and autoimmuneprone mouse strains has suggested that

defective NKT cell function relates to the emergence of autoimmunity. In many

experimental models of autoimmunity [e.g., type 1 diabetes (T1D),

encephalomyelitis], intentional activation of NKT cells by the synthetic

glycolipid agonist, a-GalCer, can elicit the regulatory functions of NKT cells and

prevent autoimmunity. Induction of T helper type 2 (Th2) deviation and the

generation of tolerogenic DCs have been suggested as mechanisms governing the

protective function of NKT cells in these models [7]. However, induction of Th2

deviation or generation of tolerogenic DC has not been confirmed in all model

systems tested to date. In addition, Th2 deviation in response to therapeutic

intervention is sometimes an outcome rather than the cause of disease

protection [7]. Therefore, additional mechanisms underlying the regulatory role

of NKT cells in autoimmune disease must be involved.The unique features of NKT cells are reminiscent of another T cell

population, CD4+CD25+ regulatory T (Treg) cells. They comprise 5%10% of murine peripheral

CD4+ T cells and express many surface markers including CD62L, CTLA-4,

GITR, and CD45RB [8]. To date, the most definitive lineage marker for naturally

occurring CD4+CD25+ Treg is the forkhead/winged helix transcription factor (Foxp3) [912]. These Tregs

are reduced in NOD mice deficient in CD80/86 or CD28 expression, which

contributes to accelerated T1D in these strains [13]. Both NKT cells and CD4+CD25+

Treg cells are thymus-derived subsets of Treg cells that play an important role

in the maintenance of self-tolerance. Whether NKT cells and Treg cells

cooperate functionally in the regulation of autoimmunity is not known. We have

explored this possibility in NOD mice by repeated injections of a-GalCer. NOD

mouse serves as an animal model of human T1D, a classic T cell-mediated

destruction of insulin-producing pancreatic islet b cells. In this study, we

show that a-GalCer-activated NKT cells can induce expansion of CD4+CD25+

Treg cells and enhance their suppressing function, which in turn mediates the therapeutic

effects of a-GalCer in NOD mice.

Materials and Methods

Mice

NOD mice were purchased from the Seed Animal Center of the Chinese

Academy of Sciences (Shanghai, China). Female NOD mice, aged 46 weeks at the initiation

of the experiments, were used. All mice were bred and maintained in specific

pathogen-free conditions. Animal experimental procedures were in compliance

with institutional guidelines.

Antibodies

Anti-CD3, anti-CD16/32, PE/Cy5-anti-CD4, PE-anti-CD25,

FITC-anti-TCR-b, PE/Cy5-anti-IL-2, PE/Cy5-anti-IL-4, PE/Cy5-anti-IL-10,

PE/Cy5-anti-IFN-g and FITC-anti-Foxp3 monoclonal antibodies were from eBioscience

(San Diego, USA). IgG1, CD1d, anti-CD25 antibody (clone PC61), and anti-mouse

IgG1-PE monoclonal antibody were from BD Pharmingen (San Diego, USA).

Treatment of mice with a-GalCer

A synthetic form of a-GalCer, KNR7000, was obtained from Axxora (Lausen, Switzerland) for

this study. Injections of a-GalCer (2 mg/mouse/injection) or vehicle (0.025% polysorbate-20 in

phosphate-buffered saline) were carried out every 3 d when the mice reached 6

weeks of age, and continued for 18 weeks. In another protocol, for protection

studies against cyclophosphamide (CY)-induced diabetes, a 2 mg/dose a-GalCer was

injected i.p. on days 0, 3, 6, 9, 12, 15, 18, and 21. In addition, 500 mg anti-CD25

(PC61) or control rat IgG was injected i.v. on days 0, 6, 12, and 18.

In vitro stimulation of splenocytes

with a-GalCer

Approximately 2?105 splenocytes

were incubated with 100 ng/ml a-GalCer in RPMI 1640 medium supplemented with 10% fetal calf serum,

50 mM

2-mercaptoethanol, 2 mM glutamine, 100 U/ml penicillin, 100 mg/ml

streptomycin, and 10 mM HEPES at 37 ?C in 5% CO2, for 72 h. For

proliferation assays, 1 mCi of [3H]thymidine was then added to each well, and after an additional 16

h of culture, cells were collected with a cell harvester and uptake of

radioactivity was measured with a Betaplate reader (Wallac, Gaithersburg, USA).Approximately 2?105 splenocytes

were incubated with 100 ng/ml a-GalCer in RPMI 1640 medium supplemented with 10% fetal calf serum,

50 mM

2-mercaptoethanol, 2 mM glutamine, 100 U/ml penicillin, 100 mg/ml

streptomycin, and 10 mM HEPES at 37 ?C in 5% CO2, for 72 h. For

proliferation assays, 1 mCi of [3H]thymidine was then added to each well, and after an additional 16

h of culture, cells were collected with a cell harvester and uptake of

radioactivity was measured with a Betaplate reader (Wallac, Gaithersburg, USA).

Cytokine secretion following in

vivo a-GalCer treatment

Mice were injected with either a-GalCer or vehicle alone (2

mg/dose,

i.v.) and were bled 2 h later. Cytokine levels (IL-4, IL-2, IL-10, and IFN-g) in the serum

were measured by enzyme-linked immunosorbent assay (ELISA; eBioscience).

Generation of a-GalCer-loaded CD1d-IgG1 dimers

Loading of CD1d-IgG1 dimers with a-GalCer was carried out as

previously described [14]. Briefly, CD1d-IgG1 dimers and a-GalCer were mixed

at neutral pH at a molar ratio of 1:9 (CD1d-IgG1 dimer:a-GalCer), followed by

overnight incubation at 37 ?C.

CY challenge, assessment of

diabetes, and evaluation of insulitis

Prediabetic female NOD mice (1012-week-old) were

challenged with one dose (300 mg/kg) of CY (Sigma, Oakville, Canada). Diabetes

was assessed by monitoring blood glucose levels every week using a OneTouch

Horizon one-step blood glucose meter (LifeScan, Mumbai, India). In CY-induced

mice, blood glucose levels were detected every 2 d. Mice with two consecutive

blood glucose measurements greater than 250 mg/dl were considered diabetic. For

evaluation of insulitis, mice were killed and pancreases were prepared, fixed

with 4% paraformaldehyde, and sectioned. Sections were stained with

hematoxylin-eosin to evaluate insulitis. Multiple hematoxylin-eosin-stained

pancreatic sections were scored in a blinded fashion. Insulitis was graded as

described [15]: 0, no inflammation; 1, peri-insulitis but no intra-insulitis;

2, 0%50% intra-insulitis; 3, more than 50% intra-insulitis.

Flow cytometry

Mononuclear cells from the spleen and pancreatic lymph node (PLN)

were isolated by Ficoll-Hypaque density gradient centrifugation. For staining

of Va14 NKT cells, mononuclear cells were first treated with antibodies

directed against Fc receptor g, then incubated with a-GalCer-loaded CD1d-IgG1 dimers, followed by

incubations with anti-mouse IgG1-PE monoclonal antibody (A85-1) and

FITC-anti-TCR-b. CD4+CD25+ cells were identified by staining with PE/Cy5-anti-CD4 and

PE-anti-CD25. For analysis of intracellular Foxp3, cells were fixed with

fixation/permeabilization solution (eBioscience) and incubated with

FITC-anti-Foxp3 monoclonal antibody. Isotype-matched antibodies were used as

controls. Flow cytometric analysis was carried out with an FACSCalibur

instrument using CellQuest software (Becton Dickinson, San Jose, USA).

Cell preparation and

suppression assay

CD4+ T cells were prepared by Dynal beads (negative selection;

Invitrogen, Oslo, Norway). CD4+ T cells were incubated with PE-anti-CD25

antibody, followed by anti-PE beads. CD4+CD25+ T

cells were isolated by positive selection over an MS column (Miltenyi Biotec,

Bergisch Gladbach, Germany), and the counterparts (CD4+CD25 cells) were also collected. In all experiments, 90%95% of these

cells were positive for both the CD4 and CD25 markers. For the in vitro

CD4+CD25+ T cell suppression assay, CD4+CD25+ T

cells (sorted from PLNs) were co-cultured in 96-well plates with CD4+CD25 T cells (sorted from PLNs) in the presence of 1 mg/ml anti-CD3

antibody (145-2C11) and 1?105

irradiated T cell depleted splenocytes. The percentage of inhibition was

determined by the following equation:

Eq.

Cytokine ELISA

Single cell suspensions of spleen cells were cultured in 96-well

plates in the presence of a-GalCer (or vehicle). The supernatants were collected after 72 h.

IFN-g, IL-2, IL-4, and IL-10 production in culture supernatants was

measured by ELISA kits [IFN-g and IL-10 from U-CyTech (Utrecht, The Netherlands), and IL-2 and

IL-4 from eBioscience].

mRNA analysis

Total RNA was isolated from PLN cells using the RNAiso reagent

(TaKaRa, Tokyo, Japan). Two micrograms of total RNA was reverse transcribed

with random 6-mers and ExScript-RTase (TaKaRa). Quantitative real-time RT-PCR

was carried out in a LightCyler (Roche Diagnostics, Mannheim, Germany) using an

SYBR Green PCR kit from TaKaRa. A threshold was set in the linear part of the

amplification curve, and the number of cycles needed to reach the threshold was

calculated for each gene. Relative mRNA levels were determined using standard

curves for each individual gene and further normalization to HPRT. Melting

curves established the purity of the amplified band. Primer sequences used

were: T-bet (5-TCAACCAGCAC­C­­A­GACAGAGATG-3, 5-GTAATGGCTT­GTGGGCTCC­AG-3);

GATA-3 (5-ATGGTACCGGGCACTACCTTTG-3, 5-TGACAGTTCGCGCAGGATG-3); and HPRT (5-A­G­CCTAAGATGAGCGCAAGT-3,

5-TTACTAGGCA­GATGGCCACA-3).

Statistical analysis

Data were expressed as the mean±SEM (or, mean±SD). Differences

between groups were analyzed by Student’s t-test. Differences between

the groups with respect to disease incidence were carried out using log-rank

tests. The level of significance was set at P<0.05.

Results

Age-dependent loss of function

of NKT cells in female NOD mice

Previous studies indicated the quantitative and functional

deficiency of NKT cells in NOD mice when compared with other strains of mice

[1619].

Here we determined the quantity and function of NKT cells in female NOD mice at

6, 12, and 18 weeks of age and the time when the mice became diabetic (most of

the mice developed diabetes after 20 weeks of age). No significant changes in

the frequency of NKT cells were observed [Fig. 1(A)]. But when treated

with 100 ng/ml a-GalCer in vitro, the NKT cells showed a decrease in

proliferation [Fig. 1(B)]. In vitro cytokine production by NKT

cells in response to a-GalCer also decreased with time [Fig. 1(C)], in agreement

with stimulation of NKT cells in vivo [Fig. 1(D,E)]. The ratio of

IFN-g to IL-4 increased dramatically, indicating a deviation to a

Th1-type immune response [Fig. 1(C)].

Suppressor function of PLN CD4+CD25+ Treg cells declines in an

age-dependent manner

To investigate the change in the number of CD4+CD25+ T

cells during the course of diabetes development, we observed this T cell

population at different time points by flow cytometry analysis. Consistent with

other reports [20], CD4+CD25+ T cells did

not change significantly in PLN [Fig. 2(A)]. However, CD25 is not a

unique marker for Tregs [21], but is also found on activated CD4+ T

cells. To confirm the true nature of the CD4+CD25+ T

cell population, we carried out intracellular staining with an antibody against

the natural Treg-specific marker Foxp3. No significant changes over time in the

proportion of NOD CD4+CD25+ T cells

expressing Foxp3 were observed [Fig. 2(B)]. While the mean fluorescence

intensity of Foxp3 in CD4+CD25+ Foxp3+ T

cells decreased with time [Fig. 2(C)]. In the in vitro

suppression assay, PLN CD4+CD25+ T cells

decreased in their ability to suppress the proliferation of CD4+CD25 T cells [Fig. 2(D)]. Some correlation exists between the

impairment of in vitro suppression and the reduction in protein

expression levels of Foxp3 on a per-cell basis.  

Functional abnormality of NKT

cells is corrected in female NOD mice treated with a-GalCer

In agreement with previous studies [15,22], a-GalCer treatment caused a

significant drop in the diabetes incidence of female NOD mice [Fig. 3(A)]

and the a-GalCer-treated mice do not develop severe insulitis [Fig. 3(B)].

We examined whether repeated injections of a-GalCer influences NKT cell

responsiveness. Female NOD mice were injected every 3 d with a-GalCer or

vehicle at the beginning of 6 weeks of age. This procedure lasted until 24

weeks of age. Two days after the last injection, we measured in vitro

proliferation and cytokine responses of splenocytes to a-GalCer. The a-GalCer-stimulated

splenocytes from a-GalCer-injected mice proliferated more vigorously than those from

vehicle-injected mice [Fig. 3(C)]. And the former produced substantial

amounts of IL-2, IL-4, and IL-10 [Fig. 3(D)]. In contrast, spleen cell

cultures from a-GalCer-injected mice produced smaller amounts of IFN-g than

vehicle-injected mice [Fig. 3(D)], indicating that these NKT cells from a-GalCer-injected

mice had a strengthened capacity to produce IL-2, IL-4, and IL-10 on their own,

or to induce their synthesis by other cell types, whereas NKT cells from a-GalCer-injected

mice lost the capacity to produce IFN-g. To evaluate the effects of long-term

treatment with a-GalCer on the immune response of PLN cells, we determined the mRNA

levels of T-bet and GATA-3 in PLN cells. As mentioned above, compared with

vehicle-injected mice, GATA-3 was up-regulated and T-bet was down-regulated in

PLNs of a-GalCer-injected mice [Fig. 3(E)], indicating a Th2-polarized

immune response.

Suppressor function of PLN CD4+CD25+ Treg cells is strengthened in

female NOD mice treated with a-GalCer

Previous studies showed that the therapeutic effect of a-GalCer did not

entirely depend on the Th2 cytokine IL-4 [23], so an alternative mechanism

other than Th1/Th2 deviation might be operating. One possibility is that the a-GalCer-activated

NKT cells function through the induction of CD4+CD25+

Treg cells. To verify this possibility, we first quantified the frequency of

CD4+CD25+ Treg cells from female NOD mice treated with a-GalCer or vehicle.

Compared with the vehicle-injected NOD mice, a-GalCer-treated recipients

had almost the same percentage of CD4+CD25+

cells among CD4+ cells in the PLNs as their vehicle-treated counterparts [Fig.

4(A)]. However, the percentage in the proportion of NOD CD4+CD25+ T

cells expressing Foxp3 increased [Fig. 4(B)], meaning that the number of

CD4+CD25+ Treg cells clearly increased. Furthermore, the mean fluorescence

intensity of Foxp3 in CD4+CD25+ T cells

expressing Foxp3 was enhanced in a-GalCer-injected mice [Fig. 4(C)]. We

also assessed the suppressive potential of CD4+CD25+

Treg cells in a-GalCer-injected mice. In co-culture experiments, CD4+CD25+

Treg cells inhibited proliferation of CD4+CD25 cells, confirming their suppressive activity. CD4+CD25+ Treg

cells from a-GalCer-treated NOD mice showed more potency in inhibiting responses

of CD4+CD25 cells to anti-CD3

stimulation when compared with those from vehicle-injected NOD mice [Fig.

4(D)]. To investigate whether these Treg cells contribute to the therapeutic

effects induced by a-GalCer, we compared the effects of anti-CD25 or control antibody on

the development of T1D in CY-challenged and a-GalCer-treated female NOD

mice. Compared with the control group, the incidence of T1D in the anti-CD25

treatment group was largely diminished [Fig. 4(E)]. These data strongly

suggest that the therapeutic effect of a-GalCer is through

collaboration of NKT cells with Treg cells.

Discussion

Previous studies have shown that NKT cells and CD4+CD25+

Treg cells from NOD mice are deficient in both quantity and function when

compared with those from other strains of mice. Here we investigated whether

these two subsets of Treg cells change in different stages of disease

development. In our study, we found no significant changes in the quantity of

NKT cells and CD4+CD25+ Treg cells in the lifetime of female NOD mice. We did find that the

loss of function of both NKT cells and CD4+CD25+

Treg cells is age-dependent in these mice. And the immune response showed a Th1

deviation. Our data indicated that the balance between Treg cells and

autoreactive T cells is destroyed.Functional abnormality of NKT cells is rectified in female NOD mice

treated with a-GalCer. Mice injected with a-GalCer produced smaller

amounts of IFN-g than vehicle-injected mice [Fig. 3(D)], indicating that

these NKT cells from a-GalCer-injected mice had a strengthened capacity to produce IL-2,

IL-4, and IL-10 on their own, or to induce their synthesis by other cell types,

whereas NKT cells from a-GalCer-injected mice lost the capacity to produce IFN-g. Compared with

vehicle-injected mice, GATA-3 was up-regulated and T-bet was

down-regulated in PLNs of a-GalCer-injected mice [Fig. 3(E)], indicating a Th2-polarized

immune response in PLNs. As mentioned above, in a-GalCer-treated mice, the

cytokine profiles of splenocytes were changed. This might be due to regulation

of cytokine secretion. The detailed mechanism of this process is to be further

investigated.Many studies have shown that CD4+CD25+

Treg cells suppress effector T cells under pathological conditions, such as

inflammation, autoimmunity, cancer, and organ transplantation [24,25]. However,

few studies have investigated whether Treg cells can be regulated by other

kinds of cells. In our study, we have verified that the functions of CD4+CD25+

Treg cells in T1D are controlled by the Treg cell subset, NKT cells. a-GalCer-activated

NKT cells promote the development/expansion and function of CD4+CD25+

Treg cells [23].NKT cell-induced CD4+CD25+ Treg cells in

turn contribute to the therapeutic effects of a-GalCer-activated NKT

cells. IL-2 plays an important role in the activation and maintenance of CD4+CD25+

Treg cells [26]. The principal physiological source of IL-2 for the maintenance

of CD4+CD25+ Treg cells has been verified to be CD4+CD25

low-activated T cells. There is some evidence that IL-2 gene

transcription and/or IL-2 protein expression can be detected in human and

murine NKT cells with or without a-GalCer stimulation, raising the possibility

that NKT cells contribute to IL-2 production in a pathophysiological state. We

have provided further evidence that a-GalCer-activated NKT cells can produce a

great amount of IL-2 [Fig. 3(C)]. During initiation of T1D, IL-2

released by a-GalCer-activated NKT cells could serve as a primary source of IL-2

that supports CD4+CD25+ Treg cells, because islet-reactive T cells have not yet been fully

activated at this stage and therefore, can not provide a source of IL-2 [23].

The a-GalCer-treated mice sustained a high level of IL-2 (data not

shown). CD4+CD25+ Treg cells from a-GalCer-treated NOD mice, in the same numbers as from

vehicle-treated mice, were more potent in their suppression of the function and

proliferation of CD4+CD25 T cells [Fig. 3(D)], consistent with the report of Liu et

al [23]. In Fig. 4(D), it seems that our data is not consistent with

the report of Ly et al [27]. However, for the following reasons, this is

not an accurate observation. Ly et al. treated the NOD mice (810 weeks old)

with a-GalCer for only 2 weeks before the mice were killed. In our

experiment, we treated NOD mice with a-GalCer (2 mg/mouse/injection) every 3

d from the age of 6 weeks, for a total of 18 weeks. At the age of 24 weeks,

spleens and PLNs were harvested from the treated mice. In our preliminary

experiment, there was no significant difference in the function of Treg cells

between mice treated with a-GalCer for 23 weeks and the controls. This is consistent with the

report of Ly et al. However, if the NOD mice were treated with a-GalCer for a

long time, for example, 6 weeks or longer, the functions of Treg cells were

strengthened, consistent with the findings of Liu et al. This might be

due to the roles played by IL-2. On stimulation, NKT cells secrete IL-2, with

the latter sustaining and/or strengthening the function of Treg cells. So, IL-2

released by NKT cells induces CD4+CD25+ Treg cells and

thus prevents the development of diabetes in NOD mice. Other factors, in

addition to IL-2, could also contribute to the enhanced functions of Treg

cells, for example, in a-GalCer-treated mice, the function and/or phenotype of DCs might be

changed. These DCs could contribute to the CD4+CD25+

Treg cells, however, this issue is not clear. The combined effects of

up-regulated Foxp3 and IL-2, induced by a-GalCer-activated NKT

cells, would underlie the strengthened function of CD4+CD25+

Treg cells. The emergence and progression of autoimmunity results from the

imbalance between autoreactive immune cells and Treg cells. Under some circumstances,

the function of Treg cells (including CD4+CD25+

Treg cells and NKT cells) can be destroyed or blocked. Pasare and Medzhitov

reported that a microbe-induced Toll pathway exists that can block the

suppressive effect of CD4+CD25+ Treg cells,

allowing activation of pathogen-specific adaptive immune responses [28]. Humans

with autoimmune diseases such as T1D often have defective Treg cell functions.

Therefore, finding ways to restore and/or enhance the function and/or the

frequency of Treg cells would have significant implications in improving the

efficacy of current immunomodulatory drugs for autoimmune disorders such as

human T1D.a-GalCer is

verified to be an ideal compound because it can stimulate both NKT cells and

CD4+CD25+ Treg cells and induce functional cooperation between them to

disrupt pathogenic responses in autoimmune disease. Compared with control mice,

mice treated with a-GalCer did not develop severe insulitis [Fig. 3(B)], and the

incidence of diabetes was lower than the control group [Fig. 3(A)]. We

observed that a-GalCer is effective in preventing the development of diabetes when

treatment is initiated after 6 weeks of age [Fig. 3(A)]. There are

several possible explanations for this. First, a-GalCer-activated NKT cells

shift the type of immune response from Th1 to Th2. Second, IL-2 produced by a-GalCer-activated

NKT cells might be the primary source of IL-2 in supporting CD4+CD25+

Treg cells. The expanded/enhanced CD4+CD25+

Treg cells prevent the autoreactive T cells from destroying the islets. To

address whether CD4+CD25+ Treg cells are necessary in the prevention of T1D in a-GalCer-treated

NOD mice [27], anti-CD25 antibody (clone PC61) was used in our experiment to

block the function of CD4+CD25+ Treg cells.

Our data indicated that a-GalCer shows nearly no preventive effect when used together with

anti-CD25 antibody (clone PC61) [Fig. 4(E)].In the present study, we showed that cytokines produced by activated

NKT cells might promote the generation and maintenance of CD4+CD25+

Treg cells in NOD mice. Subsequently, CD4+CD25+

Treg cells could enhance the therapeutic effect of a-GalCer-actived NKT cells.

Because we do not have NKT cell-deficient (CD1d knockout) mice in our

laboratory, our data only indicated the relationship between the two subsets of

regulatory T cells. If CD1d knockout mice were used in our study, our data

would be more convincing. As a-GalCer can stimulate both murine and human NKT cells, our results

further verified the implications for the use of a-GalCer in the treatment of

human autoimmune diseases, such as T1D. However, previous published reports

indicated that a-GalCer produces adverse side-effects in human liver. It would be

interesting to find another compound to optimize the effects of a-GalCer in the

prevention and treatment of human T1D.

References

 1   Godfrey DI, MacDonald HR, Kronenberg M, Smyth

MJ, Van KL. NKT cells: what’s in a name? Nat Rev Immunol 2004, 4: 231237

 2   Parekh VV, Wilson MT, Olivares-Villagomez D,

Singh AK, Wu L, Wang CR, Joyce S et al. Glycolipid antigen induces

long-term natural killer T cell anergy in mice. J Clin Invest 2005, 115: 25722583

 3   Burdin N, Brossay L, Kronenberg M.

Immunization with a-galactosylceramide polarizes CD1-reactive NK T cells

towards Th2 cytokine synthesis. Eur J Immunol 1999, 29: 20142025

 4   Fujii S, Shimizu K, Smith C, Bonifaz L,

Steinman RM. Activation of natural killer T cells by a-galactosylceramide

rapidly induces the full maturation of dendritic cells in vivo and

thereby acts as an adjuvant for combined CD4 and CD8 T cell immunity to a

coadministered protein. J Exp Med 2003, 198: 267279

 5   Taniguchi M, Harada M, Kojo S, Nakayama T,

Wakao H. The regulatory role of Va14 NKT cells in innate and

acquired immune response. Annu Rev Immunol 2003, 21: 483513

 6   Silk JD, Hermans IF, Gileadi U, Chong TW,

Shepherd D, Salio M, Mathew B et al. Utilizing the adjuvant properties

of CD1d-dependent NK T cells in T cell-mediated immunotherapy. J Clin Invest

2004, 114: 18001811

 7   Van Kaer L. a-Galactosylceramide

therapy for autoimmune diseases: prospects and obstacles. Nat Rev Immunol 2005,

5: 3142

 8   Shevach EM. CD4+ CD25+ suppressor T cells:

more questions than answers. Nat Rev Immunol 2002, 2: 389400

 9   Hori S, Nomura T, Sakaguchi S. Control of

regulatory T cell development by the transcription factor Foxp3. Science 2003,

299: 10571061

10  Fontenot JD, Rudensky AY. A well adapted

regulatory contrivance: regulatory T cell development and the forkhead family

transcription factor Foxp3. Nat Immunol 2005, 6: 331337

11  Fontenot JD, Rasmussen JP, Williams LM, Dooley

JL, Farr AG, Rudensky AY. Regulatory T cell lineage specification by the

forkhead transcription factor foxp3. Immunity 2005, 22: 329341

12  Fontenot JD, Gavin MA, Rudensky AY. Foxp3

programs the development and function of CD4+CD25+ regulatory T cells. Nat

Immunol 2003, 4: 330336

13  Salomon B, Lenschow DJ, Rhee L, Ashourian N,

Singh B, Sharpe A, Bluestone JA. B7/CD28 costimulation is essential for the

homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune

diabetes. Immunity 2000, 12: 431440

14  Schumann J, Voyle RB, Wei BY, MacDonald HR.

Cutting edge: influence of the TCR V b domain on the avidity of

CD1d: a-galactosylceramide binding by invariant V a 14 NKT cells. J

Immunol 2003, 170: 58155819

15  Hong S, Wilson MT, Serizawa I, Wu L, Singh N,

Naidenko OV, Miura T et al. The natural killer T-cell ligand a-galactosylceramide

prevents autoimmune diabetes in non-obese diabetic mice. Nat Med 2001, 7: 10521056

16  Gombert JM, Tancrede-Bohin E, Hameg A,

Leite-de-Moraes MC, Vicari A, Bach JF, Herbelin A. IL-7 reverses NK1+ T

cell-defective IL-4 production in the non-obese diabetic mouse. Int Immunol

1996, 8: 17511758

17  Falcone M, Yeung B, Tucker L, Rodriguez E,

Sarvetnick N. A defect in interleukin 12-induced activation and interferon g

secretion

of peripheral natural killer T cells in nonobese diabetic mice suggests new

pathogenic mechanisms for insulin-dependent diabetes mellitus. J Exp Med 1999,

190: 963972

18  Godfrey DI, Kinder SJ, Silvera P, Baxter AG.

Flow cytometric study of T cell development in NOD mice reveals a deficiency in

abTCR+CDR-CD8- thymocytes. J Autoimmun 1997, 10: 279285

19  Poulton LD, Smyth MJ, Hawke CG, Silveira P,

Shepherd D, Naidenko OV, Godfrey DI et al. Cytometric and functional

analyses of NK and NKT cell deficiencies in NOD mice. Int Immunol 2001, 13: 887896

20  Pop SM, Wong CP, Culton DA, Clarke SH, Tisch

R. Single cell analysis shows decreasing FoxP3 and TGFb1 coexpressing

CD4+CD25+ regulatory T cells during autoimmune diabetes. J Exp Med 2005, 201:

13331346

21  Fehervari Z, Sakaguchi S. Control of Foxp3+

CD25+CD4+ regulatory cell activation and function by dendritic cells. Int

Immunol 2004, 16: 17691780

22  Sharif S, Arreaza GA, Zucker P, Mi QS, Sondhi

J, Naidenko OV, Kronenberg M et al. Activation of natural killer T cells

by -galactosylceramide treatment prevents the onset and recurrence of

autoimmune type 1 diabetes. Nat Med 2001, 7: 10571062

23  Liu R, La Cava A, Bai XF, Jee Y, Price M,

Campagnolo DI, Christadoss P et al. Cooperation of invariant NKT cells

and CD4+CD25+ T regulatory cells in the prevention of autoimmune myasthenia. J

Immunol 2005, 175: 78987904

24  Sakaguchi S. Naturally arising Foxp3-expressing

CD25+CD4+ regulatory T cells in immunological tolerance to self and non-self.

Nat Immunol 2005, 6: 345352

25  Shevach EM. Regulatory T cells in

autoimmmunity. Annu Rev Immunol 2000, 18: 423449

26  Setoguchi R, Hori S, Takahashi T, Sakaguchi S.

Homeostatic maintenance of natural Foxp3(+) CD25(+) CD4(+) regulatory T cells

by interleukin (IL)-2 and induction of autoimmune disease by IL-2

neutralization. J Exp Med 2005, 201: 723735

27  Ly D, Mi QS, Hussain S, Delovitch TL.

Protection from type 1 diabetes by invariant NK T cells requires the activity

of CD4+CD25+ regulatory T cells. J Immunol 2006, 177: 36953704

28  Pasare C, Medzhitov R. Toll pathway-dependent

blockade of CD4+CD25+ T cell-mediated suppression by dendritic cells. Science

2003, 299: 10331036