bs_bs_banner

Clinical and Experimental Immunology

O R I G I N A L A RT I C L E

doi:10.1111/cei.12415

Successful immunotherapy of autoimmune cholangitis by adoptive transfer of forkhead box protein 3+ regulatory T cells

H. Tanaka,*† W. Zhang,* G.-X. Yang,* Y. Ando,‡ T. Tomiyama,*‡ K. Tsuneyama,§ P. Leung,* R. L. Coppel,¶ A. A. Ansari,** Z. X. Lian,†† W. M. Ridgway,‡‡ T. Joh† and M. E. Gershwin* *Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis, Davis, CA, USA, **Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA, ‡‡Division of Immunology, Allergy and Rheumatology, University of Cincinnati College of Medicine, Cincinnati, OH, USA, †Department of Gastroenterology and Metabolism, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan, ‡Third Department of Internal Medicine, Division of Gastroenterology and Hepatology, Kansai Medical University, Osaka, Japan, §Department of Diagnostic Pathology, Graduate School of Medicine and Pharmaceutical Science for Research, University of Toyama, Toyama, Japan, ¶Department of Microbiology, Monash University, Melbourne, Vic, Australia, and ††Institute of Immunology and School of Life Sciences, University of Science and Technology of China, Hefei, China Accepted for publication 7 July 2014 Correspondence: M. E. Gershwin, Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, 451 Health Sciences Drive, Suite 6510, Davis, CA 95616, USA. E-mail: [email protected]

Summary Treatment of primary biliary cirrhosis (PBC) has lagged behind that of other autoimmune diseases. In this study we have addressed the potential utility of immunotherapy using regulatory T cells (Treg) to treat murine autoimmune cholangitis. In particular, we have taken advantage of our ability to produce portal inflammation and bile duct cell loss by transfer of CD8+ T cells from the dominant negative form of transforming growth factor beta receptor type II (dnTGF-βRII) mice to recombination-activating gene (Rag)1–/– recipients. We then used this robust established adoptive transfer system and co-transferred CD8+ T cells from dnTGF-βRII mice with either C57BL/6 or dnTGF-βRII forkhead box protein 3 (FoxP3+) T cells. Recipient mice were monitored for histology, including portal inflammation and intralobular biliary cell damage, and also included a study of the phenotypical changes in recipient lymphoid populations and local and systemic cytokine production. Importantly, we report herein that adoptive transfer of Treg from C57BL/6 but not dnTGF-βRII mice significantly reduced the pathology of autoimmune cholangitis, including decreased portal inflammation and bile duct damage as well as down-regulation of the secondary inflammatory response. Further, to define the mechanism of action that explains the differential ability of C57BL/6 Treg versus dnTGF-βRII Treg on the ability to downregulate autoimmune cholangitis, we noted significant differential expression of glycoprotein A repetitions predominant (GARP), CD73, CD101 and CD103 and a functionally significant increase in interleukin (IL)-10 in Treg from C57BL/6 compared to dnTGF-βRII mice. Our data reflect the therapeutic potential of wild-type CD4+ FoxP3+ Treg in reducing the excessive T cell responses of autoimmune cholangitis, which has significance for the potential immunotherapy of PBC. Keywords: autoimmunity, cholangitis, colitis, murine models, primary biliary cirrhosis

Introduction Regulatory T cells (Treg) play a pivotal role in control of the immune response in vivo and in vitro both directly by cell– cell contact and indirectly through the production of antiinflammatory cytokines, including interleukin (IL)-10 and transforming growth factor (TGF)-β; functional impairment and absence leads to uncontrolled inflammation [1–5]. The transcription factor forkhead box protein 3

(FoxP3), a specific marker for Treg, is expressed in two Treg subsets: natural Treg (nTreg) cells and adaptive or induced Treg (iTreg) cells. Both subsets display similar phenotypical characteristics and comparable inhibitory activity against the immune response, although subtle differences in mRNA transcripts, protein expression, epigenetic modification and stability have been documented, although not universally accepted [6]. Reduction of Treg suppressive function has been reported in the presence of inflammatory cytokines,

© 2014 British Society for Immunology, Clinical and Experimental Immunology, 178: 253–261

253

H. Tanaka et al.

including IL-1β and IL-6 [7]. Treg can also assume an activated/memory phenotype; such cells have been described in humans with type 1 diabetes [8]. We have previously reported the spontaneous appearance of a robust autoimmune cholangitis in the dominant negative form of TGF-β receptor type II (dnTGF-βRII) mice [9]. Adoptive transfer of CD8+ T cells from dnTGF-βRII mice to recombination-activating gene (Rag)1–/– recipients leads to the transfer of autoimmune chalangitis [10]. Interestingly, CD8+ T cells from OT-I/dnTGF-βRII/Rag1–/– mice, in which the entire T cell repertoire was replaced with ovalbumin (OVA)-specific CD8, failed to transfer disease [11]. Importantly, mixed (dnTGF-βRII and B6) bone marrow chimeric mice were protected from biliary disease compared to dnTGF-βRII single bone marrow chimerics [12]. Therefore, we focused our attention on the CD8+ T cell population and T regulatory compartments in the development of autoimmune cholangitis. We report herein evaluation of the potential therapeutic efficacy of Treg. In particular, co-transfer of CD4+FoxP3+ regulatory T cells from B6 mice [wild-type (WT) Treg] prevents inflammatory cytokine production, portal inflammation and interlobular biliary cell damage induced by adoptive transfer of CD8 cells from dnTGF-βRII mice to Rag1–/– mice. Importantly, comprehensive analysis of phenotypical markers led to the identification of effector/memory Treg cells associated with increased activation markers that include CD25, CD39, CD101, OX40, PD-L1 and Helios, and decreased production of IL-10 in FoxP3GFP/dnTGF-βRII mice. Indeed, correction of this phenotype was successful despite the intrinsic CD8 T cell defects mediated by the dnTGF-βRII transgene. Treg cells are now within the scope of clinical use to treat autoimmune diseases and control physiological and pathological immune responses [13]. Thus, our current data provide the foundation for the potential use of normal Treg to regulate aberrant autoimmune T cell responses by adoptive transfer in autoimmune cholangitis.

Materials and methods Animals Our colony of dnTGF-βRII mice onto a C57BL/6 (B6) strain background was bred at the animal facilities of the University of California at Davis [9]. Mice were fed a sterile rodent helicobacter medicated dosing system (three-drug combination) diet (Bio-Serv, Frenchtown, NJ, USA) and maintained in individually ventilated cages under specific pathogen-free conditions. Sulfatrim (Hi-tech Pharmacal, Amityville, NY, USA) was delivered through drinking water. B6.Cg-FoxP3tm2 GFP /B6 mice) were bred at the animal (EGFP) Tch/J mice (FoxP3 facilities of the University of California at Davis. FoxP3GFP/ dnTGF-βRII mice were generated as follows. Male dnTGFβRII mice were mated with female FoxP3GFP/B6 mice to obtain FoxP3GFP/dnTGF-βRII male mice, which were subse254

Donor CD8+ T cells

Recipient CD4+ FoxP3+ Tregs

Figs. 2, 3, 4 Rag1–/–

Liver histology

A: dnTGFβRII 8 wks

B: dnTGFβRII + FoxP3/GFP/dnTGFβRII C: dnTGFβRII + FoxP3/GFP/B6

Cell in spleen Flow cytometry IL-10 production Fig. 5

Cytokine profile IFN-γ, TNF-α IL12/23p40, IL-6 Cells in blood, LN liver, spleen Phenotype Cytokine profile

Fig. 1. Schematic illustration of the experimental protocol.

quently back-crossed with female FoxP3GFP/B6 mice to obtain FoxP3GFP/dnTGF-βRII female mice. The parental dnTGFβRII and the derived FoxP3GFP/dnTGF-βRII mice at 3–4 weeks of age were genotyped to confirm the dnTGF-βRII gene in their genomic DNA [9] and the peripheral blood from the derived FoxP3GFP/dnTGF-βRII mice was analysed by flow cytometry to confirm the expression of the FoxP3 green fluorescent protein (GFP) gene. Male hemizygous FoxP3GFP/ dnTGF-βRII mice were back-crossed onto female FoxP3GFP/ B6 mice, respectively. All protocols were approved by the University of California Animal Care and Use Committee.

Isolation and adoptive transfer of CD8+ T cells from dnTGF-βRII mice and CD4+FoxP3+ regulatory T cells from FoxP3GFP/dnTGF-βRII mice or FoxP3GFP/B6 mice The experimental protocol is summarized in Fig. 1. Female recipient Rag1–/– mice at 8 weeks of age underwent adoptive transfer with purified splenic CD8+ T cells from donor dnTGF-βRII mice (dnRII CD8) and splenic CD4+FoxP3+ regulatory T cells from FoxP3GFP/dnTGF-βRII mice or FoxP3GFP/B6 mice. Purified CD8+ T cells were prepared using CD8 microbeads (Miltenyi Biotec, Auburn, CA, USA) and the purity of CD8+ T cells was greater than 90%. Purified CD4+FoxP3+ regulatory T cells were prepared using Cytomation MoFlo Cell Sorter (Beckman Coulter, Brea, CA, USA) after negative selection by CD8α, CD19, CD11c, Gr-1 Dynabeads® (Life Technologies, Grand Island, NY, USA). Single-cell suspensions in 100 μl phosphate-buffered saline (PBS) (1 × 106 of CD8 T cells with or without 1 × 106 of FoxP3 Treg per mouse) were transferred adoptively via orbital vein injection. Eight weeks following adoptive transfer, all recipients were killed and blood, liver, spleen and mesenteric lymph node were collected. The liver specimens were examined for histopathology. Splenic and hepatic MNCs were analysed by flow cytometry [11].

Flow cytometry analysis Liver and spleen infiltrating mononuclear cells (MNCs) were isolated from FoxP3GFP/dnTGF-βRII and FoxP3GFP/B6

© 2014 British Society for Immunology, Clinical and Experimental Immunology, 178: 253–261

Regulatory T cells and PBC

mice [9,10]. The cells were resuspended in staining buffer [0·2% bovine serum albumin (BSA), 0·04% ethylenediamine tetraacetic acid (EDTA) and 0·05% sodium azide in PBS], divided into 25-μl aliquots and incubated with anti-mouse FcR blocking reagent (BioLegend, San Diego, CA, USA) for 15 min at 4°C. Cells were washed and individual aliquots stained for 30 min at 4°C with a panel of fluorochrome-conjugated monoclonal antibody (mAb) that included the cell surface markers CD4, CD44, CD62L, CD103, CD69, OX40, CD73 and PD-1 (Biolegend), T cell receptor (TCR)-β, CD101, CD39, glucocorticoidinduced TNF-receptor (GITR), glycoprotein A repetitions predominant (GARP), programmed death-ligand 1 [PD-L1, or CD274(B7-H1)], TCR-β and CD25 (eBioscience, San Diego, CA, USA). The cells were then washed with PBS containing 0·2% BSA. For intracellular cytokine staining, cells were resuspended in 10% FBS (RPMI-FBI) and stimulated at 37°C for 4 h with leucocyte activation cocktail in the presence of BD GolgiPlug (BD Pharmingen, San Diego, CA, USA). The cells were stained for surface CD8α and TCR-β, fixed and permeabilized with BD Cytofix/Cytoperm Solution (BD Biosciences, San Diego, CA, USA), then stained for intracellular interferon (IFN)-γ (eBioscience), cytotoxic T lymphocyte antigen-4 (CTLA-4), Helios, IL-10 and tumour necrosis factor (TNF)-α (BioLegend) [14]. A fluorescence activated cell sorter (FACS)can flow cytometer (BD Immunocytometry Systems, San Jose, CA, USA) upgraded for detection of five colours by Cytek Development (Fremont, CA, USA) was used to acquire data, which were analysed with Cellquest PRO software (BD Immunocytometry Systems) [14,15].

Histopathology The liver and spleen of Rag1–/– recipient mice were fixed in 4% paraformaldehyde, embedded in paraffin, cut into 4-μm sections, deparaffinized and stained with haematoxylin and eosin (H&E) [16]. Portal inflammation, lobular inflammation and bile duct damage were evaluated by a ‘blinded’ pathologist. The severity of portal inflammation and lobular inflammation were scored as follows: 0, normal liver histology; 1, minimal inflammation; 2, mild inflammation; 3, moderate inflammation; and 4, severe inflammation. The severity of bile duct damage was graded as: 0, no significant changes of bile duct; 1, epithelial damage (cytoplasmic change); 2, epithelial damage with nuclear change; 3, chronic non-suppurative destructive cholangitis (CNSDC); and 4, bile duct loss. Frequency was scored as follows: 0, none; 1, 1–10%; 2, 11–20%; 3, 21–50%; and 4, more than 50%.

Cell culture and cytokine analysis Splenic CD8+ T cells were isolated and sorted from Rag1–/– recipient mice with CD8a (Ly-2) MicroBeads (Miltenyi

Biotec Inc., Auburn, CA, USA). Aliquots of 2·0 × 10 [5] CD8+ T cells were cultured in 96-well round-bottomed plates in 200 μl of RPMI media supplemented with 10% heat-inactivated fetal bovine serum (FBS) (GibcoInvitrogen Corp., Grand Island, NY, USA), 100 μg/ml streptomycin, 100 U/ml penicillin and 0·5 μg/ml each of anti-CD3 (BioLegend) and anti-CD28 (BioLegend). The cells were incubated for 72 h at 37°C in a humidified 5% CO2 incubator, then centrifuged. The supernatant was collected and analysed for concentration of cytokines [14,15]. In addition, total protein was extracted from 30 mg of frozen liver tissue by homogenization in T-Per® Tissue Protein Extraction buffer (Thermo, Rockford, IL, USA) containing a protease inhibitor cocktail (Roche, Indianapolis, IN, USA). The homogenized tissue suspension was centrifuged at 12 096 g for 20 min at 4°C and supernatant fluid stored at −80°C. Using serum, tissue lysate and supernatants from cell cultures, levels of IFN-γ, TNF-α, IL-6 and IL-10 were quantified using a cytokine bead array assay termed the T helper type 1 (Th1)/Th2/Th17 cytokine kit (BD Biosciences). The protein concentration of liver extracts was measured using the BCA protein assay kit (Thermo Fisher Scientific, Waltham, MA, USA) [15]. Sera were tested for IL-12/23p40 levels at a dilution of 1:4 utilizing the Quantikine enzyme-linked immunosorbent assay (ELISA) kit (R&D Systems, Minneapolis, MN, USA).

Statistical analysis A two-tailed unpaired t-test and Kruskal–Wallis test [nonparametric analysis of variance (anova)] were used to analyse the data; P-values < 0·05 were considered statistically significant.

Results FoxP3+ regulatory T cells from B6 mice (WT Treg) prevent autoimmune cholangitis Liver tissues from each of the studies were obtained at 8 weeks post-adoptive transfer of cells. As seen in Fig. 2, the transfer of dnRII CD8 cells into Rag1–/– mice as a positive control induced cholangitis with portal inflammation, cholangiocyte damage and bile duct loss. Co-transfer of dnRII CD8 and FoxP3+ dnRII Treg did not ameliorate this damage (Fig. 2). In contrast, however, adoptive transfer of dnRII CD8 together with FoxP3+ WT Treg resulted in a significant reduction in both the severity and frequency of portal pathology and interlobular biliary cell damage (Fig. 2). We note that although four of seven mice in the dnRII CD8+WT Treg group exhibited portal inflammation, the inflammatory scores were minimal and there was no interlobular biliary cell damage (Fig. 2b). Hence, WT Treg, but not dnTGF-βRII Treg, successfully reduced dnRII CD8+ T cell-mediated autoimmune cholangitis (Fig. 2).

© 2014 British Society for Immunology, Clinical and Experimental Immunology, 178: 253–261

255

H. Tanaka et al. (a)

dnRII CD8 + dnRII Treg

dnRII CD8 only

dnRII CD8 + WT Treg

H&E ×400

H&E ×100

H&E ×400

Histological score

D

g

C

8

D

8

+

+

dn

W

R

T

II

T

T

re

re

ly on 8 D C II R

dn

II

R

II

C 2 1 0

re g

T W

8 D C

8 dn

R II

D II

T

R II

on +

C

dn

D 8

T

ly

re g

dn R dn

**

3

II dn R dn R

C II

** 4

re g

W + D

8

+ 8 D dn R

g

0

T

T II

dn R

8

on l

y

re g

0

C

1

Histological score

1

D

2

T

+ 8 D C

II R dn

2

C

3

T W

R dn +

8 D II

*

3

II

4

re

re

T II

on 8 D C II R

C

dn R dn

***

4

II dn R

g

g

0

Frequency *** *

+

1

Histological score

Bile duct damage

To further determine whether the improved autoimmune cholangitis was due to decreased hepatic accumulation of dnRII CD8+ T cells in the liver of recipients, CD8+ T cells from blood, spleen, liver and mesenteric lymph node of adoptive transfer recipients were analysed and compared among the three groups. Our data revealed a significantly decreased frequency of effector memory population (defined as CD44+CD62L−) in recipients of the dnRII CD8 + WT Treg group when compared to recipients of either the dnRII CD8+ T cells only or dnRII CD8 + dnRII Treg groups in spleen and liver. A reduced frequency of effector 256

2

dn R

Fig. 2. Splenic CD4 forkhead box protein 3 (FoxP3)+ regulatory T cells (Treg) from FoxP3GFP/B6 mice suppresses cholangitis. (a) Representative haematoxylin and eosin (H&E)-stained liver sections from recombination-activating gene (Rag)1–/– mice at 8 weeks of age underwent adoptive transfer with purified splenic CD8+ T cells from donor dominant negative form of transforming growth factor beta receptor type II (dnTGF-βRII) mice (n = 11) and splenic CD4+ FoxP3+ regulatory T cells from FoxP3GFP/dnTGF-βRII (n = 7) or FoxP3GFP/B6 mice (n = 7). (b) Severity and frequency of portal inflammation and bile duct destruction scores. Data are expressed as individual scores. *P < 0·05; **P < 0·01; Kruskal–Wallis test [non-parametric analysis of variance (anova)].

3

ly

+

4

T

Portal inflammation

Severity ** *

C

Histological score

(b)

memory cells was also found in the peripheral blood and mlNs of dnRII CD8 + WT Treg recipients, but the changes did not reach statistical significance when compared with dnRII CD8 + dnRII Treg recipients (Fig. 3a). Furthermore, the total count of intrahepatic MNC, CD8+ T cells and CD8+ effector memory T cells decreased significantly in the dnRII CD8 + WT Treg group in comparison to either the dnRII CD8 or dnRII CD8 + dnRII Treg groups. It has been reported previously that dnTGF-βRII mice still retain a low level of functional TGF-βRII that can induce some levels of TGF-β signalling in Treg [17,18]. In agreement with this,

© 2014 British Society for Immunology, Clinical and Experimental Immunology, 178: 253–261

Regulatory T cells and PBC *** *** *** n.s.

***

*** *** n.s.

dnRll CD8 + WT Treg

40 20

n.s.

Liver

** *

10 5

*

*

6 4 2

20 15 10 5 0

*

0·8 0·6

n.s.

80

n.s. TNF-α (%)

**

TNF-α (pg/mg)

*

20 0

(b) n.s.

*

*

1·0 0·5 0·0

**

***

n.s.

150 100 50 0 100 80 60 40 20 0

n.s. *

*

IL-12/23P40 (pg/ml)

IFN-γ (pg/ml) IL-6 (pg/ml)

n.s.

6

** *

*

4 2 0

n.s.

dnRll CD8 only dnRll CD8 + dnRll Treg dnRll CD8 + WT Treg

*

*

0·4 0·2 0·0

(c) 200

8

reduced significantly in dnRII CD8 + WT Treg transfer recipients compared to the dnRII CD8 or dnRII CD8 + WT Treg groups (Fig. 4b). A similar set of results was obtained in serum samples from the adoptive transfer of WT Treg but not dnRII Treg, exemplified by the suppressed production of inflammatory cytokines IFN-γ, TNF-α, IL-6 and IL-12/ 23p40 by dnRII CD8 (Fig. 4c). Our data clearly support the view that a therapeutic potential of WT Treg in suppressing dnRII CD8-induced autoimmune cholangitis exists and indicates that in the presence of defects in both Treg and T effector cells (as seen in the dnTGF-βRII mice), correction

40

1·5

**

8

0

0

60

mLN

n.s.

*

IL-6 (pg/mg)

15

Spleen

CD44+ CD62L– (×106)

Blood

CD8 (×106)

0

(a)

IFN-γ (pg/mg)

dnRll CD8 + dnRll Treg

60

reduced intrahepatic CD8+ T cells and CD8+ effector memory T cells were also detected in the dnRII CD8 + dnRII Treg group when compared to the dnRII CD8 group, indicating that dnRII Treg retained some ability to limit the accumulation of dnRII CD8 effectors. dnRII Treg, however, could not prevent disease progression (Fig. 3b). Intracellular staining analysis demonstrated that the percentages of IFN-γ and TNF-α-producing CD8+ T cells were decreased in liver tissues from dnRII CD8 + WT Treg transfer recipients (Fig. 4a). The hepatic production of the proinflammatory cytokines IFN-γ, TNF-α and IL-6 was

Fig. 4. Splenic CD4+ forkhead box protein 3 (FoxP3)+ regulatory T cells (Treg) from FoxP3GFP/B6 mice [wild-type (WT) Treg] successfully suppresses inflammatory cytokine production by CD8 T cells. (a) Intracellular staining analysis for interferon (IFN)-γ and tumour necrosis factor (TNF)-α-expressing CD8 T cells in liver. (b) Level of IFN-γ, TNF-α and interleukin (IL)-6 in liver protein extracts from recipients. (c) Serum levels of IFN-γ, TNF-α, IL12/IL23p40 and IL-6 in recipient mice. dnRII CD8 group (n = 11), dnRII CD8 + dnRII Treg group (n = 7) and dnRII CD8 + WT Treg group (n = 7). Data are expressed as mean ± standard error of the mean. *P < 0·05; **P < 0·01; ***P < 0·001; two-tailed unpaired t-test.

dnRll CD8 only

*** *

80

(b)

IFN-γ (%)

*** *

TNF-α (pg/ml)

Fig. 3. Splenic CD4 forkhead box protein 3 (FoxP3)+ regulatory T cells (Treg) from FoxP3GFP/B6 mice [wild-type (WT) Treg] successfully suppresses CD8 T cell activation. (a) Frequency of effector memory (CD44+ CD62L–) subpopulations in CD8 T cell subsets in liver and the periphery. (b) Absolute number of intra-hepatic mononuclear cells (MNCs), CD8 T cells, and effector memory (CD44+CD62L–) subpopulations in CD8 T cell subsets. dnRII CD8 group (n = 11), dnRII CD8 + dnRII Treg group (n = 7) and dnRII CD8 + WT Treg group (n = 7). Data are expressed as mean ± standard error of the mean. *P < 0·05; **P < 0·01; ***P < 0·001; two-tailed unpaired t-test.

MNCs (×106)

+

CD44+CD62L– in CD8 (%)

(a) 100

1·5

n.s.

*

*

1·0 0·5 0·0

*

60 40 20 0

1500

n.s. * * *

1000 500 0

© 2014 British Society for Immunology, Clinical and Experimental Immunology, 178: 253–261

257

H. Tanaka et al. 100

15

5

60 40

reg

dn

T WT

**

4

2

20 0

T Rll

6

(C)

80

IL-10 (%)

* 10

0

(b)

FoxP3+ in CD4 (MFI)

FoxP3+ in CD4 (%)

(a)

reg

ll nR

T reg

d

T WT

0

reg

Rll

T reg

WT

dn

dnRll Treg

Spleen

***

**

**

WT Treg

100

in FoxP3+ CD4 (%)

T reg

***

80

*

**

**

***

***

*

60 40 20 0

CD101 CD103 CD69 OX40 CD73 CD39 GARP PD-1

LN

GITR PD-L1 CTLA-4 Helios CD25 *

***

**

**

***

80 60

*** ***

*

*

**

**

20 CD101 CD103 CD69 OX40 CD73 CD39 GARP PD-1 GITR PD-L1 CTLA-4 Helios CD25

Liver

**

**

in FoxP3+ CD4 (%)

100 ** ***

80 *** 60

*

*** ***

* ***

**

40 20 0

CD101 CD103 CD69 OX40 CD73 CD39 GARP PD-1

GITR PD-L1 CTLA-4 Helios CD25

Thymus

80

***

***

*

60

*

***

40

* **

20 0

**

***

100

CD101 CD103 CD69 OX40 CD73

of the Treg defect alone may be sufficient to modify disease successfully.

The phenotype of splenic dnRII Treg compared to WT Treg: increased IL-10 production by WT Treg To address why dnTGF-βRII Treg were not sufficient to suppress the activation of dnRII CD8 and protect recipients from dnRII CD8-induced portal inflammation compared to 258

**

**

40

0

in FoxP3+ CD4 (%)

Fig. 5. Phenotypical analysis of splenic dnRII and wild-type (WT) Treg. (a) Frequency and mean fluorescence intensity (MFI) of CD4+ forkhead box protein 3 (FoxP3)+ regulatory T cells (Treg) in spleen from FoxP3GFP/donor dominant negative form of transforming growth factor beta receptor type II (dnTGF-βRII) (n = 7) and FoxP3GFP/B6 mice (n = 7). (b) Expression of Treg activation markers were compared between donor FoxP3GFP/dnTGF-βRII and FoxP3GFP/B6 in spleen, lymph node (LN), liver and thymus. CD101, CD103, CD69, OX40, CD73, CD39, glycoprotein A repetitions predominant (GARP), programmed death (PD)-1, glucocorticoid-induced TNF-receptor-related protein (GITR), CD274 (B7-H1) (PD-L1), cytotoxic T lymphocyte antigen-4 (CTLA-4), Helios and CD25 as an activation marker. (c) Frequency of IL-10-positive cells in splenic CD4+ FoxP3+ Treg from FoxP3GFP/dnTGF-βRII (n = 3) and FoxP3GFP/B6 mice. Data are expressed as mean ± standard error of the mean. *P < 0·05; **P < 0·01; ***P < 0·001; two-tailed unpaired t-test.

in FoxP3+ CD4 (%)

100

CD39 GARP PD-1

GITR PD-L1 CTLA-4 Helios CD25

WT Treg, we examined Treg frequency, FoxP3 expression and production of IL-10 in splenic FoxP3 + Treg. We found that the splenic CD4+FoxP3+ Treg population was increased significantly in FoxP3GFP/dnTGF-βRII mice, while the mean fluorescence intensity (MFI) of FoxP3+ was not significantly different between dnRII Treg and WT Treg (Fig. 5a). Furthermore, the production of the inhibitory cytokine IL-10 was significantly lower in splenic Treg from FoxP3GFP/dnTGFβRII mice in comparison with FoxP3GFP/B6 mice (Fig. 5c).

© 2014 British Society for Immunology, Clinical and Experimental Immunology, 178: 253–261

Regulatory T cells and PBC

We next evaluated the phenotype of Treg at the site of inflammation (liver), as well as thymus and secondary lymphoid organs. The expression of CD101, CD69, CD39, GARP and Helios were significantly higher, but the expression of CD73 was significantly lower on dnRII Treg from thymus, spleen, LN and liver compared with WT Treg (Fig. 5b). The Treg within the spleen and LN from dnRII mice showed increased expression of GITR and PD-1 but significantly decreased expression of PD-1 in the thymus compared with WT Treg (Fig. 5b).

Discussion The TGF-β family of cytokines encompass TGF-β1, TGF-β2 and TGF-β3, that are highly pleiotropic with diverse effects on many developmental and physiological processes. TGF-β1 is most abundant in lymphoid organs and has multiple of effects on immunity, including inhibition of T cell proliferation and differentiation and negative effects on macrophage activation and dendritic cell (DC) maturation [19]. Active TGF-β mediates its biological function by binding to TGF-β type I (TGF-βRI) and type II (TGF-βRII) receptors, both of which are serine/threonine kinases. TGF-β engagement with a tetrameric receptor complex consisting of two TGF-βRI molecules and two TGF-βRII molecules activates these receptor kinases, allowing them to phosphorylate downstream targets and to activate different signalling pathways [20]. TGF-β1 has been shown to act as a co-stimulatory factor for the expression of FoxP3 [21], leading to the differentiation of Treg cells from naive T cells [20,22,23]. Therefore, the disruption of TGFR signalling would lead to Treg defects such as we have demonstrated herein [12,24–26]. We have reported previously that peripheral blood CD4+ CD25high Treg were reduced in primary biliary cirrhosis (PBC) patients compared with healthy controls, and the level of FoxP3-expressing Treg was markedly lower in affected portal tracts of PBC patients compared with patients with chronic hepatitis C (CHC) and autoimmune hepatitis (AIH) [27]. We have postulated that murine autoimmune cholangitis requires defects in both the cytotoxic CD8+ T cell effector cells and Treg cells [12]. Moreover, mixed dnTGF-βRII and B6 bone marrow chimeric mice were protected from biliary disease compared to dnTGFβRII single bone marrow chimerics [12]. To assess the therapeutic manipulation of CD4+FoxP3+ Treg in autoimmune cholangitis, we generated FoxP3GFP/dnTGF-βRII mice and performed adoptive CD8+ T cell transfer from dnTGFβRII mice together with either dnRII Treg or WT Treg. In the present study, we evaluated the suppressive function of WT Treg in vivo and demonstrated that WT Treg were sufficient to control abnormal dnRII CD8+ T cell activation (Fig. 3a,b) and suppress the proinflammatory cytokine production in peripheral blood and at the inflammatory hepatic site (Fig. 4). Mechanisms underlying Treg immune suppressive

function include direct cell–cell contact and the production of IL-10 and TGF-β. The successful immune suppressive function of WT Treg in our autoimmune cholangitis model suggests that WT Treg controlled the disease driven by intrinsic defects within the CD8+ T cells via an immunoregulatory pathway, probably including IL-10. We performed a comprehensive examination of the phenotype of CD4+FoxP3+ regulatory T cells in FoxP3GFP/ dnTGFβ-RII and FoxP3GFP/B6 donors. In secondary lymphoid organs such as spleen and LN, there were differences in the expression of several activation markers between dnRII Treg and WT Treg. For example, CD101, CD69, CD39, GARP, PD-1, GITR, PD-L1 and CTLA-4 were significantly lower in WT Treg than in dnRII Treg mice. CTLA-4 is an important surface molecule linked to the suppressive function of Treg [28,29]. FoxP3 drives the constitutive expression of CTLA-4 by Treg. Increased expression level of FoxP3 may promote the expression of CTLA-4 in Treg from dnTGF-βRII mice. However, the reasons why defective Treg are associated with increased expression levels of FoxP3 and CTLA-4 need to be further addressed. GARP is essential for the surface expression of latent TGF-β on activated Treg that represent subsets with highly potent regulatory function [30,31]. The surface expression of GARP on Treg is greatly enhanced by either TGF-β signal blockage [32] or TCR stimulation [33]. Interestingly, it should be noted that the expression of GARP on the Treg was not required for suppressive function of Treg in vitro [33]. Decreased CD73 and CD103 were detected in thymus, liver, spleen and LN from dnRII Treg compared to WT Treg (Fig. 2b). It has been documented that adenosine generation catalyzed by CD73 expressed by regulatory T cells mediates immune suppression [34], and CD103+ Treg are potent suppressors of tissue inflammation in several autoimmune diseases [35]. The expression of CD73 [36,37] and CD103 [38–40] on Treg is known to be inducible by TGF-β. Our findings that CD73 and CD103 expression are reduced on CD4+FoxP3+ Treg in dnTGF-βRII mice further emphasizes the regulatory role of TGF-β signalling on the expression of CD73 and CD103. The reduction of CD73 and CD103 in Treg at the inflammatory site and other organs may be partially responsible for the defective suppressive function of dnRII Treg. It is of interest to note that while this is the first preclinical evidence of the effectiveness of a cellular-based therapeutic approach in PBC, current data on the role of Treg abnormalities in PBC remain speculative. Indeed, the effector mechanisms in PBC, as with other examples of mucosal mediated diseases, may have significant individual variations [11,41–48]. We note in the current study the unique and different profile of the phenotypical expression of CD39 and CD73 on dnTGF-βRII Treg. This differential expression of the two ectonucleotidases may be important for the immunosuppressive Treg activities. CD39 and CD73 are ectonucleotidases that hydrolyze adenosine 5′-triphosphate (ATP), adenosine 5′-diphosphate (ADP)

© 2014 British Society for Immunology, Clinical and Experimental Immunology, 178: 253–261

259

H. Tanaka et al.

and adenosine 5′-phosphate (AMP) to adenosine, creating a ‘purinergic halo’ surrounding immune cells. While this has not been studied in PBC, we note a variety of previous data which have highlighted differential expressions of a variety of effector pathways in PBC, and we suggest that this is an important area for future research [11,14,15,49–53]. We suggest that further studies are necessary to determine the mechanisms of regulation by CD73 and CD103 on Treg function.

Conclusion Taken together, our data demonstrate a distinct Treg phenotype in dnTGF-βRII mice and show that correction of these Treg defects with physiologically normal Treg, was associated with enhanced IL-10 effects, decreased inflammatory cytokines and prevention of disease. Thus, wild-type CD4+FoxP3+ Treg have a therapeutic potential in limiting the excessive effector T cell response in autoimmune cholangitis.

Acknowledgements The authors thank Bridget McLaughlin, Jonathan Van Dyke, Kazuhito Kawata, Masanobu Tsuda, Bin Liu, Jinjun Wang, Jun Zhang, Chen-Yen Yang, Kerstien Padgett and Tom P. Kenny for technical support in this experiment. We also thank Ms. Nikki Phipps for support in preparing this paper. Financial support was provided in part by National Institutes of Health grant DK090019.

Disclosure There are no competing interests.

Author contributions H.T., W. Z., G.-X. Y., Y. A., T. T., K. T., P. L., Z. X. L. and T. J. performed the experiments. M. E. G., W. M. R., R. L. C. and A. A. A. designed the experiments. M. E. G., A. A. A., W. M. R. and H. T. wrote the paper.

References 1 Fort MM, Narayanan PK. Manipulation of regulatory T-cell function by immunomodulators: a boon or a curse? Toxicol Sci 2010; 117:253–62. 2 Sakaguchi S, Sakaguchi N, Shimizu J et al. Immunologic tolerance maintained by CD25+ CD4+ regulatory T cells: their common role in controlling autoimmunity, tumor immunity, and transplantation tolerance. Immunol Rev 2001; 182:18–32. 3 Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell 2008; 133:775–87. 4 Maloy KJ, Powrie F. Regulatory T cells in the control of immune pathology. Nat Immunol 2001; 2:816–22.

260

5 Schneider-Hohendorf T, Stenner MP, Weidenfeller C et al. Regulatory T cells exhibit enhanced migratory characteristics, a feature impaired in patients with multiple sclerosis. Eur J Immunol 2010; 40:3581–90. 6 Lin X, Chen M, Liu Y et al. Advances in distinguishing natural from induced FoxP3(+) regulatory T cells. Int J Clin Exp Pathol 2013; 6:116–23. 7 Beriou G, Costantino CM, Ashley CW et al. IL-17-producing human peripheral regulatory T cells retain suppressive function. Blood 2009; 113:4240–9. 8 Sakuraba K, Shibata K, Iwamoto Y, Yoshikai Y, Yamada H. Naturally occurring PD-1+ memory phenotype CD8 T cells belong to nonconventional CD8 T cells and are cyclophosphamide-sensitive regulatory T cells. J Immunol 2013; 190:1560–6. 9 Oertelt S, Lian ZX, Cheng CM et al. Anti-mitochondrial antibodies and advances in distinguishing natural from induced FoxP3(+) regulatory T cellsprimary biliary cirrhosis in TGF-beta receptor II dominant-negative mice. J Immunol 2006; 177:1655–60. 10 Yang GX, Lian ZX, Chuang YH et al. Adoptive transfer of CD8(+) T cells from transforming growth factor beta receptor type II (dominant negative form) induces autoimmune cholangitis in mice. Hepatology 2008; 47:1974–82. 11 Kawata K, Yang GX, Ando Y et al. Clonality, activated antigenspecific CD8(+) T cells, and development of autoimmune cholangitis in dnTGFbetaRII mice. Hepatology 2013; 58:1094– 104. 12 Huang W, Kachapati K, Adams D et al. Murine autoimmune cholangitis requires two hits: cytotoxic KLRG1 CD8 effector cells and defective T regulatory cells. J Autoimmun 2014; 50:123–34. 13 d’Hennezel E, Piccirillo CA. Analysis of human FOXP3+ Treg cells phenotype and function. Methods Mol Biol 2011; 707:199– 218. 14 Tsuda M, Zhang W, Yang GX et al. Deletion of interleukin (IL)12p35 induces liver fibrosis in dominant-negative TGFbeta receptor type II mice. Hepatology 2013; 57:806–16. 15 Ando Y, Yang GX, Tsuda M et al. The immunobiology of colitis and cholangitis in interleukin-23p19 and interleukin-17A deleted dominant negative form of transforming growth factor beta receptor type II mice. Hepatology 2012; 56:1418–26. 16 Yoshida K, Yang GX, Zhang W et al. Deletion of interleukin-12p40 suppresses autoimmune cholangitis in dominant negative transforming growth factor beta receptor type II mice. Hepatology 2009; 50:1494–500. 17 Ishigame H, Mosaheb MM, Sanjabi S, Flavell RA. Truncated form of TGF-betaRII, but not its absence, induces memory CD8+ T cell expansion and lymphoproliferative disorder in mice. J Immunol 2013; 190:6340–50. 18 Li MO, Sanjabi S, Flavell RA. Transforming growth factor-beta controls development, homeostasis, and tolerance of T cells by regulatory T cell-dependent and -independent mechanisms. Immunity 2006; 25:455–71. 19 Gorelik L, Flavell RA. Transforming growth factor-beta in T-cell biology. Nat Rev Immunol 2002; 2:46–53. 20 Li MO, Flavell RA. TGF-beta: a master of all T cell trades. Cell 2008; 134:392–404. 21 Cobbold SP, Castejon R, Adams E et al. Induction of FoxP3+ regulatory T cells in the periphery of T cell receptor transgenic mice tolerized to transplants. J Immunol 2004; 172:6003–10. 22 Chen W, Jin W, Hardegen N et al. Conversion of peripheral CD4+CD25– naive T cells to CD4+CD25+ regulatory T cells by

© 2014 British Society for Immunology, Clinical and Experimental Immunology, 178: 253–261

Regulatory T cells and PBC

23

24

25

26

27

28

29

30

31

32

33

34

35

36

TGF-beta induction of transcription factor FoxP3. J Exp Med 2003; 198:1875–86. Fantini MC, Becker C, Monteleone G, Pallone F, Galle PR, Neurath MF. Cutting edge: TGF-beta induces a regulatory phenotype in CD4+CD25– T cells through FoxP3 induction and downregulation of Smad7. J Immunol 2004; 172:5149–53. Ouyang W, Beckett O, Ma Q, Li MO. Transforming growth factorbeta signaling curbs thymic negative selection promoting regulatory T cell development. Immunity 2010; 32:642–53. Peng Y, Laouar Y, Li MO, Green EA, Flavell RA. TGF-beta regulates in vivo expansion of FoxP3-expressing CD4+CD25+ regulatory T cells responsible for protection against diabetes. Proc Natl Acad Sci USA 2004; 101:4572–7. Marie JC, Liggitt D, Rudensky AY. Cellular mechanisms of fatal early-onset autoimmunity in mice with the T cell-specific targeting of transforming growth factor-beta receptor. Immunity 2006; 25:441–54. Lan RY, Cheng C, Lian ZX et al. Liver-targeted and peripheral blood alterations of regulatory T cells in primary biliary cirrhosis. Hepatology 2006; 43:729–37. Paust S, Lu L, McCarty N, Cantor H. Engagement of B7 on effector T cells by regulatory T cells prevents autoimmune disease. Proc Natl Acad Sci USA 2004; 101:10398–403. Sojka DK, Hughson A, Fowell DJ. CTLA-4 is required by CD4+CD25+ Treg to control CD4+ T-cell lymphopenia-induced proliferation. Eur J Immunol 2009; 39:1544–51. Tran DQ, Andersson J, Wang R, Ramsey H, Unutmaz D, Shevach EM. GARP (LRRC32) is essential for the surface expression of latent TGF-beta on platelets and activated FOXP3+ regulatory T cells. Proc Natl Acad Sci USA 2009; 106:13445–50. Dhamne C, Chung Y, Alousi AM, Cooper LJ, Tran DQ. Peripheral and thymic FoxP3(+) regulatory T cells in search of origin, distinction, and function. Front Immunol 2013; 4:253. Zhou AX, Kozhaya L, Fujii H, Unutmaz D. GARP-TGF-beta complexes negatively regulate regulatory T cell development and maintenance of peripheral CD4+ T cells in vivo. J Immunol 2013; 190:5057–64. Edwards JP, Fujii H, Zhou AX, Creemers J, Unutmaz D, Shevach EM. Regulation of the expression of GARP/latent TGF-beta1 complexes on mouse T cells and their role in regulatory T cell and Th17 differentiation. J Immunol 2013; 190:5506–15. Deaglio S, Dwyer KM, Gao W et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. J Exp Med 2007; 204:1257– 65. Chang LY, Lin YC, Kang CW et al. The indispensable role of CCR5 for in vivo suppressor function of tumor-derived CD103+ effector/memory regulatory T cells. J Immunol 2012; 189:567– 74. Regateiro FS, Howie D, Nolan KF et al. Generation of antiinflammatory adenosine by leukocytes is regulated by TGF-beta. Eur J Immunol 2011; 41:2955–65.

37 Takimoto T, Wakabayashi Y, Sekiya T et al. Smad2 and Smad3 are redundantly essential for the TGF-beta-mediated regulation of regulatory T plasticity and Th1 development. J Immunol 2010; 185:842–55. 38 Reynolds LA, Maizels RM. Cutting edge: in the absence of TGFbeta signaling in T cells, fewer CD103+ regulatory T cells develop, but exuberant IFN-gamma production renders mice more susceptible to helminth infection. J Immunol 2012; 189:1113–7. 39 Robinson PW, Green SJ, Carter C, Coadwell J, Kilshaw PJ. Studies on transcriptional regulation of the mucosal T-cell integrin alphaEbeta7 (CD103). Immunology 2001; 103:146–54. 40 Suffia I, Reckling SK, Salay G, Belkaid Y. A role for CD103 in the retention of CD4+CD25+ Treg and control of Leishmania major infection. J Immunol 2005; 174:5444–55. 41 Invernizzi P. Liver auto-immunology: the paradox of autoimmunity in a tolerogenic organ. J Autoimmun 2013; 46:1–6. 42 Trivedi PJ, Adams DH. Mucosal immunity in liver autoimmunity: a comprehensive review. J Autoimmun 2013; 46:97–111. 43 Hudspeth K, Pontarini E, Tentorio P et al. The role of natural killer cells in autoimmune liver disease: a comprehensive review. J Autoimmun 2013; 46:55–65. 44 Yang CY, Ma X, Tsuneyama K et al. IL-12/Th1 and IL-23/Th17 biliary microenvironment in primary biliary cirrhosis: implications for therapy. Hepatology 2014; 59:1944–53. 45 Tanaka H, Yang GX, Iwakoshi N et al. Anti-CD40 ligand monoclonal antibody delays the progression of murine autoimmune cholangitis. Clin Exp Immunol 2013; 174:364–71. 46 Rong GH, Yang GX, Ando Y et al. Human intrahepatic biliary epithelial cells engulf blebs from their apoptotic peers. Clin Exp Immunol 2013; 172:95–103. 47 Leung PS, Wang J, Naiyanetr P et al. Environment and primary biliary cirrhosis: electrophilic drugs and the induction of AMA. J Autoimmun 2013; 41:79–86. 48 Chen RC, Naiyanetr P, Shu SA et al. Antimitochondrial antibody heterogeneity and the xenobiotic etiology of primary biliary cirrhosis. Hepatology 2013; 57:1498–508. 49 Yao Y, Yang W, Yang YQ et al. Distinct from its canonical effects, deletion of IL-12p40 induces cholangitis and fibrosis in interleukin-2Ralpha(–/–) mice. J Autoimmun 2014; 51:99–108. 50 Chang CH, Chen YC, Yu YH et al. Innate immunity drives xenobiotic-induced murine autoimmune cholangitis. Clin Exp Immunol 2014; 177:373–80. 51 Lleo A, Zhang W, McDonald WH et al. Shotgun proteomics: identification of unique protein profiles of apoptotic bodies from biliary epithelial cells. Hepatology 2014. doi: 10.1002/hep.27230. 52 Jin Q, Moritoki Y, Lleo A et al. Comparative analysis of portal cell infiltrates in antimitochondrial autoantibody-positive versus antimitochondrial autoantibody-negative primary biliary cirrhosis. Hepatology 2012; 55:1495–506. 53 Takahashi T, Miura T, Nakamura J et al. Plasma cells and the chronic nonsuppurative destructive cholangitis of primary biliary cirrhosis. Hepatology 2012; 55:846–55.

© 2014 British Society for Immunology, Clinical and Experimental Immunology, 178: 253–261

261

Successful immunotherapy of autoimmune cholangitis by adoptive transfer of forkhead box protein 3(+) regulatory T cells.

Treatment of primary biliary cirrhosis (PBC) has lagged behind that of other autoimmune diseases. In this study we have addressed the potential utilit...
623KB Sizes 0 Downloads 4 Views