<<

European Journal of Endocrinology (2005) 152 635–643 ISSN 0804-4643

EXPERIMENTAL STUDY CXCR6 within T-helper (Th) and T-cytotoxic (Tc) type 1 lymphocytes in Graves’ disease (GD) G Aust, M Kamprad1, P Lamesch2 and E Schmu¨cking Institute of Anatomy, 1Department of Clinical Immunology and Transfusion Medicine and 2Department of Surgery, University of Leipzig, Phillipp-Rosenthal-Str. 55, Leipzig, 04103, Germany (Correspondence should be addressed to G Aust; Email: [email protected])

Abstract Objective: In Graves’ disease (GD), stimulating anti-TSH antibodies are responsible for hyperthyroidism. T-helper 2 (Th2) cells were expected to be involved in the underlying immune mech- anism, although this is still controversial. The aim of this study was to examine the expression of CXCR6, a receptor that marks functionally specialized T-cells within the Th1 and T-cyto- toxic 1 (Tc1) cell pool, to gain new insights into the running immune processes. Methods: CXCR6 expression was examined on peripheral blood lymphocytes (PBLs) and thyroid- derived lymphocytes (TLs) of GD patients in flow cytometry. CXCR6 cDNA was quantified in thyroid tissues affected by GD (n ¼ 16), Hashimoto’s thyroiditis (HT; n ¼ 2) and thyroid autonomy (TA; n ¼ 11) using real-time reverse transcriptase PCR. Results: The percentages of peripheral CXCR6þ PBLs did not differ between GD and normal subjects. CXCR6 was expressed by small subsets of circulating T-cells and natural killer (NK) cells. CXCR6þ cells were enriched in thyroid-derived T-cells compared with peripheral CD4þ and CD8þ T-cells in GD. The increase was evident within the Th1 (CD4þ -gþ (IFN-gþ)) and Tc1 (CD8þIFN- gþ) subpopulation and CD8þ granzyme Aþ T-cells (cytotoxic effector type). Thyroid-derived fibro- blasts and thyrocytes were CXCR62. There was no significant difference between the CXCR6 mRNA levels in GD compared with HT and normal TA tissues. The lowest CXCR6 mRNA levels were obtained from thyroid nodules from TA patients and GD patients with low thyroid peroxidase autoantibody levels. Conclusions: CXCR6 was overexpressed in Th1 and Tc1 TLs compared with PBLs in GD. CXCR6 could be a marker for lymphocytes that have migrated into the thyroid and assist in the thyroid, indepen- dently of the bias of the underlying disease.

European Journal of Endocrinology 152 635–643

Introduction type 2 , respectively. In addition to secretion, Tc1 and Tc2 populations mediate cytolytic Graves’ disease (GD) and Hashimoto’s thyroiditis (HT) effects, with Tc1 cells utilizing both perforin- and Fas- are organ-specific autoimmune diseases characterized based killing pathways, whereas Tc2 cells primarily by diffuse or focal lymphocytic infiltration. In GD, stim- utilize perforin-mediated cytolysis (1). ulating anti-thyroid-stimulating hormone (TSH) recep- However, recent results from clinical studies and tor (TSHR) autoantibodies overstimulating the TSHR newly established mouse models (2–7) have clearly are responsible for hyperthyroidism. In HT, normal shown that the assumption that GD and HT are thyroid tissue is replaced mainly by lymphocytes orga- immune responses dominated by Th2 or Th1/Tc1, nized in ectopic follicles. The destruction of thyrocytes respectively, is an oversimplification. The role of the causes hypothyroidism. Th1/Th2 balance is especially controversial in murine The expected underlying immune mechanisms of models of Graves’ hyperthyroidism. Suppression of both diseases led to the application of the common TSHR-specific Th1 immune responses by exogenous CD4þ T-helper 1 (Th1)/Th2 paradigm of immunology. IL-4 was associated with inhibition of hyperthyroidism Th2 cells producing interleukin 4 (IL-4), IL-5, IL-10 in BALB/c mice injected with adenovirus-expressing and IL-13 are involved in humoral immunity, as was TSHR (TSHR-adeno model), indicating the critical role first expected for GD. In HT, Th1 cells are thought to of Th1 cytokines (4). In contrast, BALB/c IL-4-knockout produce interferon-g (IFN-g) and act on cell-mediated but not IFN-g-knockout mice failed to develop Graves’ immune responses leading to thyrocyte destruction. hyperthyroidism when injected with TSHR-expressing Similarly to CD4þ T-cells, the CD8þ type 1 cytotoxic M12 B lymphoma cells, suggesting the importance of (Tc1) and Tc2 cells differentially secrete type 1 and endogenous IL-4. In the IL-4- and IFN-g-knockout

q 2005 Society of the European Journal of Endocrinology DOI: 10.1530/eje.1.01892 Online version via www.eje-online.org

Downloaded from Bioscientifica.com at 09/27/2021 12:37:12PM via free access 636 G Aust and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2005) 152

TSHR-adeno model, both IL-4 and IFN-g are implicated blood lymphocytes (PBLs) from the same patients. in the development of hyperthyroidism (7). The potential thyroid-cell sources of the receptor and Besides determining the ratio of type 1 and type 2 its were defined. specific cytokine responses – in most cases IFN-g/IL- 4 – or the immunoglobulin subclasses of TSHR anti- Materials and methods bodies (IgG2a/IgG1), analysis of chemokine receptors and their ligands allows insights into the running auto- Patients immune process (2, 3). Among the over 50 and 20 chemokine receptors, only a few are thought to Thyroid diseases were diagnosed on the basis of clinical, mark functionally specialized T-cells within the total biochemical and immunological features as well as Th1 and Tc1 T-cell pool (8, 9). from scintiscans. Antibodies against the TSHR (TSH- CXCR6 (Bonzo, STRL33, TYMSTR) (10–12) and its binding-inhibiting immunoglobulin; TBII) and thyroid ligand CXCL16 (SR-PSOX) belong to the few unique peroxidase (TPO) were measured in serum obtained chemokine/receptor pairs that suggest special up to 2 weeks before operation with commercial RIA w immune functions, whereas most other pairs show kits (TRAKhuman DYNOTEST , anti-TPOn DYNO- w promiscuity and overlap in cell and tissue expression. TEST ; Brahms Diagnostica GmbH, Berlin, Germany). CXCR6 seems to mark functionally specialized T-cells The numbers of focal lymphocytic infiltrates and ecto- within the total Th1 and Tc1 cell pool (13). In contrast, pic germinal centers were counted on hematoxilin/ the is displayed by very few (1–2%) eosin-stained sections of thyroid samples within an 2 Th2 or Tc2 cells. CXCL16 (13–15) is structurally area of 226 mm as described in (21). different from most other chemokines. It is a type I For mRNA quantitation, we used thyroid tissues from transmembrane with four distinct domains: GD (n ¼ 16, all females; mean age^S.E.M. 35.6^3.7 one chemokine domain tethered to the cell surface via years), HT (n ¼ 2, both females; 18 and 34 years) a mucin-like stack, which in turn is attached to trans- and TA (n ¼ 11, five females; 55.7^5.2 years) patients membrane and cytoplasmic domains. CXCL16 acts as (same collective as described in detail in (22)). Four an adhesion molecule when expressed on the cell out of these 16 GD patients used for mRNA analysis surface (16). Upon cleavage from the cell surface, the were selected for CXCR6 expression analysis (patients soluble chemokine shows chemoattractive activity (17). 13–16). PBLs from six additional GD patients were Recently published studies indicate a unique role for further included for flow-cytometric analysis (Table 1). CXCR6 and its ligand in attracting activated lympho- All GD patients were treated with anti-thyroid drugs cyte subsets during inflammation, while also facilita- but not corticosteroids at the time of surgery. The dur- ting immune responses via cell contact (16, 18, 19). ation of GD until enrolment was longer than 15 There are no data on the distribution of CXCR6 in months in most cases. Peripheral blood samples from thyroid autoimmunity and few on autoimmune dis- 10 normal adults (two male, eight female; mean age eases at all (20). The aim of our study was to investi- 33.7^2.7 years) without any history of autoimmune gate CXCR6 and CXCL16 mRNA expression in disease were used as controls. The study was approved thyroids affected by GD compared with thyroid tissue by the local Committee of Medical Ethics. from patients with HT and non-autoimmune thyroid autonomy (TA) to gain new insights into the running Isolation of cells and cell lines used immune processes within the thyroid and to character- in this study ize the lymphocytic subpopulations involved. In GD, we isolated thyroid-derived lymphocytes (TLs) and com- Thyroid samples were obtained during operation. TLs pared the expression of CXCR6 with that of peripheral were isolated after mechanical tissue disaggregation

Table 1 Age, sex, duration of disease until enrolment, signs of Graves’ ophthalmopathy (GO), treatment with anti-thyroid drugs and corticosteroids of patients with GD analyzed for CXCR6 expression on PBLs and TLs in flow cytometry.

Patient Patient no. in Duration of Anti-thyroid drug Corticosteroid Signs no. reference (22) Age Sex disease (months) treatment treatment of GO

1 27 Female #15 Yes No Yes 2 22 Female $15 Yes No No 3 49 Male $15 Yes No Yes 4 62 Male $15 Yes No No 5 35 Male $15 Yes No No 6 59 Male $15 Yes No Yes 7 13 18 Female $15 Yes No Yes 8 14 25 Female $15 Yes No Yes 9 15 38 Female #5 Yes No Yes 10 16 50 Female $15 Yes No No

www.eje-online.org

Downloaded from Bioscientifica.com at 09/27/2021 12:37:12PM via free access EUROPEAN JOURNAL OF ENDOCRINOLOGY (2005) 152 CXCR6 in thyroid autoimmunity 637 and Ficoll density-gradient centrifugation. Thyrocytes and (3) CXCR6-PE; CD8-RPE-Cy5; granzyme A-FITC. were enriched after enzymatic digestion of the tissue The cells were analyzed by flow cytometry (FAC- as described in (23). Outgrowing fibroblasts were Scanw; Becton Dickinson, Mountain View, CA, USA) obtained by culturing small pieces of thyroid samples. using electronic gating on lymphocyte subpopulations. PBLs were isolated by Ficoll density-gradient centrifu- The Mann–Whitney test was used to determine statisti- gation. Anaplastic thyroid carcinoma cell lines SW- cal significance. 1736, Hth-74, C-634 (kindly provided by N.-E. Heldin, University of Uppsala, Uppsala, Sweden) and 8505 C, follicular lines FTC-133 and FTC-238 and Real-time reverse transcriptase (RT)-PCR papillary line B-CPAP (DSMZ, Braunschweig, Total cellular RNA was isolated with the Qiagen total Germany), as well as Chinese hamster ovary (CHO) RNA isolation (Qiagen GmbH, Hilden, Germany). cells (ATCC) were cultured in Dulbecco’s modified cDNA was synthesized from 1 mgRNAina20ml stan- Eagle’s medium/10% fetal calf serum. dard reaction mixture containing 200 U Superscript II RNaseH reverse transcriptase (Invitrogen). Quantitative Examination of the CXCR6 antibody PCR was performed on a Rotorgene (Corbett Research, Mortlake, Australia) real-time machine by using Since we always measured a lower number of T-cells SYBRw Green I as a double-strand DNA-specific binding expressing CXCR6 than described in (10), the specificity dye and continuous fluorescence monitoring. Amplifi- and sensitivity of the monoclonal antibody (mAb) used cation was carried out in a total volume of 20 ml con- was examined. First, PBLs were cultured with taining 0.5 mM of each primer, 1–3 mM MgCl2,1U 100 ng/ml IL-15 (Immunotools, Friesoythe, Germany) Taq polymerase (Roche), 10 £ Taq buffer (500 mM over 6 days, and the number of CXCR6þ cells was KCl/100 mM Tris/HCl, pH 8.3; Roche), 2 mM of each determined by flow cytometry. Secondly, the CXCR6- dNTP and 1 ml SYBRw Green (1:2000; Molecular encoding cDNA was cloned in the vector pcDNA3.1 Probes Europe, Leiden, The Netherlands) dissolved in Zeo(þ) (Invitrogen GmbH, Karlsruhe, Germany) using DMSO and 1 ml cDNA diluted 1:5. the primer pair 50-CCG GAA GCT TAC CAT GGC AGA The PCR primers were CXCR6 sense (50-CCT GCC GCA TGA TTA CC-30 (sense) and 50-GGA ATT CGA TTA ACC CTG TGC TCT AT-30) and antisense (50-ATG ACA TGC TGG TGG CCT CCA C-30 (antisense). CHO TCA TCC CCC TTG GTT TCA-30), CXCL16 sense (50- cells were transiently transfected with the construct GGG GGC AGT CAC CGC AGT CCT-30) and antisense using Lipofectamine (Invitrogen). mAb binding was (50-ATT AGC CGG GTG TGG TGG TGA GCA-30)or measured after 48 h by flow cytometry. CD18 sense (50-GGC GCA CAA GCT GGC TGA AAA CAA-30) and antisense (50-AGC GCC CGG ATG ACA 0 Analysis by flow cytometry AAC GAC TG- 3 ), respectively. CD18 mRNA was measured in order to quantify leukocytic infiltration Directly fluorochrome-labeled mAbs were supplied by (25). Sizes of PCR products were: CXCR6 459 bp; R & D Systems GmbH (Wiesbaden, Germany; CXCR6- CXCL16 518 bp, and CD18 372 bp. Glyceraldehyde PE, IFN-g-FITC), DakoCytomation Denmark A/S 3-phosphatase dehydrogenase (GAPDH) transcripts (Glostrup, Denmark; CD3-RPE-Cy5, CD8-RPE-Cy5, were measured with a ready-to-use quantitative PCR CD62L-FITC, CD4-FITC), Immunotech (Marseilles, test kit (Roboscreen, Leipzig, Germany) (26). External France; CD45R0-FITC) or BD Biosciences Europe (Hei- cDNA standards were constructed by blunt-end cloning delberg, Germany; CD4-Cy5, CD19-PerCP, granzyme of PCR fragments into pGEM-T vector (Promega, A-FITC). Mannheim, Germany). Plasmid DNA was prepared To enrich intracellular cytokines, cells were treated (Plasmid Mini Kit; Qiagen), quantified and stabilized with PMA (10 ng/ml), ionomycin (1 mmol) and Golgi- in the PCR tubes (27). Amounts were calculated from Plug (1 ml/ml; BD Biosciences) in RPMI 1640/10% a reference curve obtained from the simultaneously fetal calf serum for 4 h. Since ionomycin markedly processed standard curve. Data were expressed as zep- diminishes CXCR6 expression, cells must be pre-stained tomoles (zmol; 10221 mol) of cDNA per attomole with the anti-CXCR6 antibody before treatment (24). (amol; 10218 mol) of GAPDH cDNA. Cells were fixed and permeabilized using Cytofix/ Cytoperm solutions (BD Biosciences) before staining to analyze intracellular cytokines and granzyme A. This Results procedure is not necessary if only cell-surface antigens Flow cytometry are analyzed. Unfixed or fixed PBLs or TLs (3 £ 105) were incubated with mAbs at the desired concentration We first examined whether the available CXCR6 mAb for 30 min at 4 8C. The following mAb combinations was suitable for flow cytometry. IL-15 increased the were used: (1) CXCR6-PE; CD3-RPE-Cy5; CD8-FITC, number of CXCR6þ PBLs by about 5-fold as shown in CD4-FITC, CD45RO-FITC, CD62L-FITC or CD56-FITC; a typical case in Fig. 1A. Moreover, transiently (2) CXCR6-PE; CD8-RPE-Cy5 or CD4-Cy5; IFN-g-FITC transfected CHO cells expressing human CXCR6 were

www.eje-online.org

Downloaded from Bioscientifica.com at 09/27/2021 12:37:12PM via free access 638 G Aust and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2005) 152

CXCR6 expression. All results are summarized in Table 2. Table 2A contains a general overview on the cellular composition of PBLs and TLs. The percentage of memory T-cells (CD3þCD45R0þ) increased in the thyroid compared with periphery. TLs differed from their PBLs counterparts in terms of lymph-node-homing capabilities since CD62L was expressed at only 27% of TLs but 79% of PBLs. The percentage of T-cells capable to produce IFN-g was higher in the thyroid compared the periphery. IFN-gþ cells were more present in CD8þ than CD4þ PBLs and TLs. The expression pattern of CXCR6 within these lym- phocytic subpopulations was also characterized (Table 2B, Fig. 2). The percentage of CD3þ and, further subdivided, CD4þ and CD8þ T-lymphocytes expressing CXCR6 in PBLs did not vary between normal subjects and patients with GD. However, these CD4þCXCR6þ and CD8þCXCR6þ T-cells were enriched 3-5-fold in Figure 1 Examination of the CXCR6 mAb. (A) IL-15 increased the the thyroid compared with the periphery. The increase number of CXCR6þ PBLs by about 5-fold. (B) Transiently CXCR6 in CXCR6þ cells was evident within the Th1 þ cDNA-transfected CHO cells were CXCR6 (left, direct-labeled (CD4þIFN-gþ) and Tc1 (CD8þIFN-gþ) T-cell subpopu- CXCR6 mAb; right, indirect detection system). lations (Fig. 2A and B). A higher percentage of peripheral as well as thyroid-derived CD8þ T-cells, com- CXCR6þ. The number of positive cells varied non-sig- pared with CD4þ T-cells, expressed CXCR6. Since the nificantly whether we used a direct or indirect CXCR6 percentage of CXCR6þ T-cells producing IL-4 was labeling system (Fig. 1B). very low in PBLs and TLs (,1%; not shown), we do Secondly, PBLs of normal subjects and GD patients not suggest a function for CXCR6þ Th2 or Tc2 cells as well as TLs of GD patients were analyzed for in the thyroid.

Table 2 CXCR6 expression of TLs and PBLs from patients with GD and normal controls (n ¼ 10) analyzed by flow cytometry using multicolor analysis and lymphocyte gating.

Relative number of cells expressing the marker Marker (%) in each lymphocyte subpopulation expression TLs (GD) PBLs (GD) PBLs (controls)

Characterization of lymphocytes þ þ 100% CD3 CD62L 27.1^6.5 (*P , 0.0001) 79.0^4.6 77.2^2.5 þ CD45RO 61.0^4.5 (*P , 0.02) 42.0^4.8 39.7^2.9 CD4þ 64.4^5.4 65.2^6.5 62.9^3.7 CD8þ 37.1^3.8 33.0^3.3 36.9^3.8 100% CD8þ Granzyme Aþ 47.3^5.3 41.4^5.9 44.0^11.0 þ IFN-g 59.8^6.6 (*P , 0.02) 30.3^4.9 43.9^4.9 þ þ 100% CD4 IFN-g 20.1^3.7 (*P , 0.01) 5.6^0.6 (**P , 0.02) 17.1^2.9 Characterization of CXCR6þ lymphocytes þ þ 100% CD3 CXCR6 21.0^1.9 (*P , 0.0001) 4.7^0.4 4.1^0.5 þ þ þ 100% CD3 CD45RO CXCR6 25.1^2.5 (*P , 0.0001) 8.8^1.7 7.1^0.9 þ þ þ 100% CD3 CXCR6 CD62L 10.5^1.9 (*P , 0.0001) 45.4^6.5 39.8^6.1 100% CD3þCD56þ CXCR6þ 32.1^3.3 32.9^6.7 14.3^3.5 þ þ 100% CD32CD56 CXCR6 8.6^2.2 (*P , 0.05) 3.4^0.7 2.0^0.5 þ þ þ 100% CD3 CD8 CXCR6 28.0^3.4 (*P , 0.0001) 6.5^1.4 6.5^1.1 þ þ þ 100% CD8 IFN-g CXCR6 15.7^2.7 (*P , 0.01) 3.7^1.1 1.5^0.3 þ þ þ 100% CD8 CXCR6 IFN-g 52.6^7.7 (*P , 0.01) 30.4^2.3 44.4^5.7 þ þ þ 100% CD8 granzymeA CXCR6 18.5^4.2 (*P , 0.01) 3.7^0.9 1.5^0.2 100% CD8þCXCR6þ Granzyme Aþ 66.9^7.0 67.9^7.5 80.1^1.4 þ þ þ 100% CD3 CD4 CXCR6 11.7^1.7 (*P , 0.001) 3.4^0.8 2.4^0.2 þ þ þ 100% CD4 IFN-g CXCR6 8.3^2.4 (*P , 0.01) 1.6^0.3 1.2^0.3

*Significant differences between TLs and PBLs in GD patients; **significant differences between PBLs from GD patients and controls; percentage of labeled cells (means^S.E.M.).

www.eje-online.org

Downloaded from Bioscientifica.com at 09/27/2021 12:37:12PM via free access EUROPEAN JOURNAL OF ENDOCRINOLOGY (2005) 152 CXCR6 in thyroid autoimmunity 639

Figure 2 FACS analysis of PBLs and TLs from the same GD patient. CXCR6 was expressed on a higher percentage of (A) CD4þ and (B) CD8þ TLs than PBLs. (C) The percen- tage of CXCR6þCD8þ granzyme Aþ cells was also higher in the thyroid compared with the periphery. (D) 60% of periph- eral CXCR6þ T-cells failed to express CD62L, whereas approximately 90% of the corresponding thyroid-derived cells lacked CD62L.

Granzymes are important effector molecules for cyto- Most peripheral CD3þCXCR6þ cells have extralym- toxic lymphocytes and induce granule-mediated apopto- phoid tissue-homing potential since 60% failed to sis in target cells. Some 40–50% of CD8þ cells expressed express CD62L. Approximately 90% of thyroid-derived intracellular granzyme A in both PBLs and TLs from GD CXCR6þCD3þ T-cells lack CD62L (Table 2, Fig. 2D). patients. Within these peripheral CD8þ granzyme Aþ Peripheral and thyroid-derived B-cells did not express cells, only a few (,4%) expressed CXCR6, whereas CXCR6 (not shown). nearly 20% carried the receptor in the thyroid (Fig. 2C). The majority of CD8þCXCR6þ peripheral and thyr- oid-derived cells were positive for granzyme A. CXCR6 and CXCL16 mRNA in thyroid tissues CD3þCD56þ T-cells are effector cells with strong and thyroid-derived cells cytotoxic activity against target cells. In this regard, CXCR6 was expressed on higher percentages of Real-time PCR revealed that thyroid tissues from all GD CD3þCD56þ than on less cytotoxic CD32CD56þ cells. patients with high TPO levels and both HT patients

www.eje-online.org

Downloaded from Bioscientifica.com at 09/27/2021 12:37:12PM via free access 640 G Aust and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2005) 152 were positive for CXCR6 mRNA, whereas no signal was circulating Th1 and Tc1 cells, is expressed on a present within some tissues from GD patients with low higher percentage of TLs than PBLs from GD patients. or no TPO levels and TA tissues (Fig. 3A). We found a The data raise the question of the CXCR6’s function slight correlation between CXCR6 and (1) CD18 mRNA in the thyroid. The result agrees with our own previous levels (r ¼ 0.51; P ¼ 0.044) and (2) TPO antibody study on CXCR3 and CCR5 (29), chemokine receptors levels (r ¼ 0.66; P ¼ 0.05) in GD patients. There was that are thought to be predominantly, but not exclu- no correlation between CXCR6 mRNA levels and TBII sively, found on Th1 cells. Both receptors were (P ¼ 0.42) and the number of lymphocytic infiltrates expressed on a higher percentage of TLs compared (P ¼ 0.60) or germinal centers (P ¼ 0.60) in these with PBLs, and may be involved in the recruitment of patients. We found no difference in CXCR6 mRNA lymphocytes to the thyroid in GD (30, 31). We suggest levels in GD compared with normal TA tissue. Lowest that, in addition to CXCR3 and CCR5, CXCR6 may be a levels were present in TA nodules. Here, three out of marker for cells that have migrated into thyroid tissue nine samples were negative for CXCR6 mRNA. and assist there, independently of the bias of the under- Using simple RT-PCR, PBLs and TLs were CXCR6 lying disease. This is confirmed by the fact that most of mRNA-positive, whereas no signal was obtained from the CXCR6þ T-cells in the thyroid lost their lymph- thyroid-derived fibroblasts (Fig. 3C). Thyrocytes node-homing capabilities since they were CD62L2. showed only a very faint signal. Thyroid carcinoma The antigen is downregulated in response to migration cell lines were positive for CXCR6 mRNA. The and activation by proteolytically cleaving from the cell expression level varied considerably. surface (32). Moreover, our assumption is supported Quantifying CXCL16 cDNA in thyroid tissues by recent studies demonstrating that CXCR6þ T-cells (Fig. 3B), the levels were somewhat high compared were enriched in tissue sites of inflammation, such as with other chemokines quantified (21, 25, 28). Surpris- rheumatoid joints and inflamed livers (13, 18). ingly, CXCL16 cDNA levels within the two HT tissues Although a higher percentage of TLs compared with were within the same range as those of many GD and PBLs were CXCR6þ in GD, we did not find a difference normal TA tissues. This suggests that cells other than between CXCR6 mRNA levels in the thyroid tissues the known CXCL16 producers such as dendritic cells from different patients groups. Mistakes in CXCR6 and may express CXCL16 mRNA. mRNA determination were unlikely since quantitation Indeed, besides TL, thyrocytes were strongly positive of the chemokine receptor/ligand pair CXCR5/CXCL13 for this chemokine mRNA (Fig. 3C). Thus, the in the same cDNAs samples resulted in significantly CXCL16 mRNA levels tend to represent the presence higher levels in GD compared with TA samples and of this chemokine within several cell types of the thyroid the highest levels in HT tissues (21). Our results than a specific expression by immunocompetent cells. suggest that the number of CXCR6þ and thus Th1 and Tc1 cells in HT may not be higher than in other thyroids, including non-autoimmune TA. There are Discussion several possible explanations for this result. As in GD tissues, the chronic lymphocytic infiltrate does not We have demonstrated here that CXCR6, a chemokine reflect the initiation, but rather the maintenance or receptor present on a small subset of specialized perturbation, of the immune process in HT tissues

Figure 3 Quantitation of (A) CXCR6 and (B) CXCL16 mRNA in thyroid tissues of GD patients with high (.1000 U/ml; TPOhigh; n ¼ 8) and low or no antibodies against TPO (TPOlow; n ¼ 8), HT patients (HT; n ¼ 2) and in TA patients (normal and nodular tissue; n ¼ 11), *P , 0.005. (C) Simple RT-PCR for CXCR6 and CXCL16 mRNA in thyroid- derived cell populations (thyrocytes, fibroblasts, lymphocytes ¼ TLs), thyroid carcinoma cell lines and PBLs.

www.eje-online.org

Downloaded from Bioscientifica.com at 09/27/2021 12:37:12PM via free access EUROPEAN JOURNAL OF ENDOCRINOLOGY (2005) 152 CXCR6 in thyroid autoimmunity 641 since a substantial number of thyrocytes are always chemokine/receptor pair in the thyroid? CXCL16 not destroyed at the time of surgery (33, 34). The time of only attracts CXCR6 expressing T-cells, natural killer action of specialized Th1 and Tc1 cells may be over (NK) cells and NK/T-cells to the site of inflammation during HT. This assumption is confirmed by reports and injury, but also mediates firm adhesion of showing a significant increase in IFN-g-inducible che- CXCR6-expressing cells in its transmembrane form mokines CXCL10 (IP-10) and CXCL9 (Mig) strongly (16, 19). The chemokine is induced by pro-inflamma- associated with Th1-mediated immune responses in tory stimuli such as tumour necrosis factor-a and thyroid tissue specimens obtained from subjects IFN-g (17). Surprisingly, we did not find any differences affected by recent-onset GD (2, 35, 36). An increase between CXCL16 mRNA levels in the thyroid tissues in serum CXCL10 levels has only been found in analyzed. Normally, CXCL16 is only expressed by newly diagnosed GD patients, compared with healthy macrophages, dendritic cells and CD19þ B-cells (14, subjects and patients with long-standing disease. 42). In our study, a strong CXCL16 mRNA signal was An inverse correlation between circulating CXCL10 obtained from thyrocytes and thyroid carcinoma cell levels and disease duration has been demonstrated lines. Thus, CXCL16 also seems to be produced by epi- (37). This means that changes in the composition thelial cells, which confirms older studies showing that of regulating T-cell subsets involved, and thus thyrocytes were potential cytokine producers (43). chemokines and their receptors, occur within the Untreated thyrocytes secrete CXCL8 (IL-8) and the thyroid but also at the periphery, depending on the other chemokine that binds to the CXCR2, CXCL1 duration of GD. (GRO-a) (28). CCL2 (monocyte chemoattractant pro- Data on CXCR6 in autoimmunity are very limited tein-1; MCP-1) is a chemokine that attracts T-lympho- (20). An increase of CXCR6þ cells was shown in cytes as well as monocytes, and can be also produced arthritic synovial fluid, which was discussed as playing by thyrocytes (44). Thyrocytes from autoimmune thyr- a role of the receptor in the traffic of effector T-cells that oid disorders produce the chemokines CXCL9 and mediate type 1 inflammation. The paucity of data CXCL10 after stimulation (36). All these studies suggest published on CXCR6 may be due to the discrepant a possible role for thyrocytes on accumulation of leuko- results obtained with the available antibodies and the cytes in the tissue from the blood. Unfortunately, useful first data published (10). The percentages of CXCR6þ CXCL16 antibodies for immunohistology or establish- T-cells in normal subjects in our study were lower, ment of protein screening assays were not available. although we used the same mAb and staining tech- In summary, our study demonstrates that the per- nique as in (10) (15 versus 4.1% CXCR6þ cells centages of Tc1 (CD8þIFN-gþ) and Th1 (CD4þIFN- within CD3þ). Thus, we first showed that the mAb gþ) cells as well as cytotoxic effector cells (CD8þ used indeed detects CXCR6 and that the percentage of granzymeAþ) expressing CXCR6 were increased in CXCR6þ T-cells does not depend on direct or indirect TLs compared with PBLs in GD patients. However, we labeling methods. Moreover, IL-15 increased the did not find any difference between CXCR6 mRNA number of CXCR6þ PBLs by about 5-fold, which was levels in thyroid tissues from different patient groups. in accordance to data obtained by Unutmaz et al. (10). CXCR6 could be a marker for cells that have migrated CXCR6þ T-cells are thought to represent type 1 cells, into the thyroid and assist there, independently of the which mainly regulate cellular but not humoral bias of the underlying disease. immune responses. This might explain the missing cor- relation of CXCR6 mRNA with TBII and the slight cor- relation of CXCR6 mRNA with TPO antibodies. TPO Acknowledgements antibodies better reflect the immune responses in long-standing disease compared with TBII (38). The authors are thankful to D Sittig for her excellent Additionally, thyroid autoantibodies might not be pro- technical assistance. This study was supported by the duced exclusively by the thyroid itself (39–41). Deutsche Forschungsgemeinschaft (project AU-132-2). The correlation between CXCR6 mRNA and CD18 mRNA is weak. CD18 mRNA also includes the mRNA of those lymphocytes that were not located within infil- References trates or germinal centers, but scattered throughout the tissue and, additionally, the mRNA of non-lymphoid 1 Mosmann TR, Li L & Sad S. Functions of CD8 T-cell subsets secret- leukocytes, the macrophages. We do not know where ing different cytokine patterns. Seminars in Immunology 1997 9 þ 87–92. CXCR6 lymphocytes are located within Graves’ 2 Rotondi M, Lazzeri E, Romagnani P & Serio M. Role for interferon- thyroids or whether or not they accumulate on gamma inducible chemokines in endocrine autoimmunity: an certain structures. This may explain why we missed a expanding field. Journal of Endocrinological Investigation 2003 26 correlation between CXCR6 and defined lymphocytic 177–180. 3 Dogan RN, Vasu C, Holterman MJ & Prabhakar BS. Absence of infiltrates and germinal centers. IL-4, and not suppression of the Th2 response, prevents develop- Does the ligand of CXCR6, CXCL16, yield any ment of experimental autoimmune Graves’ disease. Journal of additional information on the function of this Immunology 2003 170 2195–2204.

www.eje-online.org

Downloaded from Bioscientifica.com at 09/27/2021 12:37:12PM via free access 642 G Aust and others EUROPEAN JOURNAL OF ENDOCRINOLOGY (2005) 152

4 Nagayama Y, Mizuguchi H, Hayakawa T, Niwa M, McLachlan SM 20 Kim CH, Kunkel EJ, Boisvert J, Johnston B, Campbell JJ, & Rapoport B. Prevention of autoantibody-mediated Graves’-like Genovese MC, Greenberg HB & Butcher EC. Bonzo/CXCR6 hyperthyroidism in mice with IL-4, a Th2 cytokine. Journal of expression defines type 1-polarized T-cell subsets with extralym- Immunology 2003 170 3522–3527. phoid tissue homing potential. Journal of Clinical Investigation 5 Nagayama Y, McLachlan SM, Rapoport B & Oishi K. Graves’ 2001 107 595–601. hyperthyroidism and the hygiene hypothesis in a mouse model. 21 Aust G, Sittig D, Becherer L, Anderegg U, Schu¨tz A, Lamesch P & Endocrinology 2004 145 5075–5079. Schmu¨cking E. The role of CXCR5 and its ligand CXCL13 in the 6 Nagayama Y, Watanabe K, Niwa M, McLachlan SM & Rapoport B. compartimentalization of lymphocytes in thyroids affected by Schistosoma mansoni and alpha-galactosylceramide: prophyl- Graves’ disease. European Journal of Endocrinology 2004 150 actic effect of Th1 Immune suppression in a mouse model of 225–234. Graves’ hyperthyroidism. Journal of Immunology 2004 173 22 Hamann J, Eichler W, Vogel B, Stortelers C, Hamann D, Horejsi V 2167–2173. & van Lier RA. CD97 workshop panel report. In Leucocyte Typing 7 Nagayama Y, Saitoh O, McLachlan SM, Rapoport B, Kano H & VI. White Cell Differentiation Antigens, pp 527–529. Eds T Kumazawa Y. TSH receptor-adenovirus-induced Graves’ Kishimoto, H Kikutani & AEGKr von dem Borne et al. New York: hyperthyroidism is attenuated in both interferon-gamma and Garland Publishing, 1997. interleukin-4 knockout mice; implications for the Th1/Th2 para- 23 Aust G, Heuer M, Laue S, Lehmann I, Hofmann A, Heldin N-E & digm. Clinical and Experimental Immunology 2004 138 417–422. Scherbaum WA. Expression of TNF-alpha mRNA and protein in 8 Sallusto F, Lenig D, Mackay CR & Lanzavecchia A. Flexible pro- pathological thyroid tissue and carcinoma cell lines. Clinical and grams of chemokine receptor expression on human polarized T Experimental Immunology 1996 105 148–154. helper 1 and 2 lymphocytes. Journal of Experimental Medicine 24 Koprak S, Matheravidathu S, Springer M, Gould S & Dumont FJ. 1998 187 875–883. Down-regulation of cell surface CXCR6 expression during 9 Annunziato F, Cosmi L, Galli G, Beltrame C, Romagnani P, activation is predominantly mediated by calcineurin. Cell Immu- Manetti R, Romagnani S & Maggi E. Assessment of chemokine nology 2003 223 1–12. receptor expression by human Th1 and Th2 cells in vitro and 25 Aust G, Steinert M, Kiessling S, Kamprad M & Simchen C. in vivo. Journal of Leukocyte Biology 1999 65 691–699. Reduced expression of Stromal-derived factor 1 in autonomous 10 Unutmaz D, Xiang W, Sunshine MJ, Campbell J, Butcher E & thyroid adenomas and its regulation in thyroid-derived cells. Littman DR. The primate lentiviral receptor Bonzo/STRL33 is Journal of Clinical Endocrinology and Metabolism 2001 86 coordinately regulated with CCR5 and its expression pattern is 3368–3376. conserved between human and mouse. Journal of Immunology 26 Ko¨hler T, Schill C, Deininger MW, Krahl R, Borchert S, 2000 165 3284–3292. Hasenclever D, Leiblein S, Wagner O & Niederwieser D. High Bad and Bax mRNA expression correlate with negative outcome 11 Deng HK, Unutmaz D, KewalRamani VN & Littman DR. in acute myeloid leukemia (AML). Leukemia 2002 16 22–29. Expression cloning of new receptors used by simian and human immunodeficiency . Nature 1997 388 296–300. 27 Ko¨hler T, Rost AK & Remke H. Calibration and storage of DNA competitors used for contamination-protected competitive PCR. 12 Liao F, Alkhatib G, Peden KW, Sharma G, Berger EA & Farber JM. Biotechniques 1997 23 722–726. STRL33, A novel chemokine receptor-like protein, functions as a 28 Aust G, Steinert M, Boltze C, Kiessling S & Simchen C. GRO-alpha fusion cofactor for both -tropic and T cell line-tropic in normal and pathological thyroid tissues and its regulation in HIV-1. Journal of Experimental Medicine 1997 185 2015–2023. thyroid-derived cells. Journal of Endocrinology 2001 170 13 Wilbanks A, Zondlo SC, Murphy K, Mak S, Soler D, Langdon P, 513–520. Andrew DP, Wu L & Briskin M. Expression cloning of the 29 Aust G, Sittig D, Steinert M, Lamesch P & Lohmann T. Graves’ STRL33/BONZO/TYMSTRligand reveals elements of CC, CXC, disease is associated with an altered CXCR3 and CCR5 expression and CX3C chemokines. Journal of Immunology 2001 166 in thyroid-derived compared with peripheral blood lymphocytes. 5145–5154. Clinical and Experimental Immunology 2002 127 479–485. 14 Shimaoka T, Kume N, Minami M, Hayashida K, Kataoka H, Kita T 30 Qin S, Rottman JB, Myers P, Kassam N, Weinblatt M, Loetscher M, & Yonehara S. Molecular cloning of a novel scavenger receptor Koch AE, Moser B & Mackay CR. The chemokine receptors CXCR3 for oxidized low density lipoprotein, SR-PSOX, on macrophages. and CCR5 mark subsets of T cells associated with certain inflam- Journal of Biological Chemistry 2000 275 40663–40666. matory reactions. Journal of Clinical Investigation 1998 101 15 Matloubian M, David A, Engel S, Ryan JE & Cyster JG. A trans- 746–754. membrane CXC chemokine is a ligand for HIV-coreceptor 31 Bonecchi R, Bianchi G, Bordignon PP, D’Ambrosio D, Lang R, Bonzo. Nature Immunology 2000 1 298–304. Borsatti A, Sozzani S, Allavena P, Gray PA, Mantovani A & 16 Shimaoka T, Nakayama T, Fukumoto N, Kume N, Takahashi S, Sinigaglia F. Differential expression of chemokine receptors and Yamaguchi J, Minami M, Hayashida K, Kita T, Ohsumi J et al. chemotactic responsiveness of type 1 T helper cells (Th1s) and Cell surface-anchored SR-PSOX/CXC chemokine ligand 16 medi- Th2s. Journal of Experimental Medicine 1998 187 129–134. ates firm adhesion of CXC chemokine receptor 6-expressing 32 Kahn J, Ingraham RH, Shirley F, Migaki GI & Kishimoto TK. cells. Journal of Leukocyte Biology 2004 75 267–274. Membrane proximal cleavage of L-: identification of the 17 Abel S, Hundhausen C, Mentlein R, Schulte A, Berkhout TA, cleavage site and a 6-kD transmembrane peptide fragment of Broadway N, Hartmann D, Sedlacek R, Dietrich S, Muetze B L-selectin. Journal of Cell Biology 1994 125 461–470. et al. The transmembrane CXC-chemokine ligand 16 is induced 33 Armengol MP, Juan M, Lucas-Martin A, Fernandez-Figueras MT, by IFN-gamma and TNF-alpha and shed by the activity of the dis- Jaraquemada D, Gallart T & Pujol-Borrell R. Thyroid autoimmune -like metalloproteinase ADAM10. Journal of Immunology disease: demonstration of thyroid antigen-specific B cells and 2004 172 6362–6372. recombination-activating expression in chemokine-contain- 18 Wang J, Holmes TH, Cheung R, Greenberg HB & He XS. Expression ing active intrathyroidal germinal centers. American Journal of of chemokine receptors on intrahepatic and peripheral lympho- Pathology 2001 159 861–873. cytes in chronic hepatitis C infection: its relationship to liver 34 Stassi G & De Maria R. Autoimmune thyroid disease: new models inflammation. Journal of Infectious Diseases 2004 190 989–997. of cell death in autoimmunity. Nature Reviews Immunology 2002 2 19 Chandrasekar B, Bysani S & Mummidi S. CXCL16 Signals via Gi, 195–204. Phosphatidylinositol 3-Kinase, Akt, IkB Kinase, and Nuclear 35 Kemp EH, Metcalfe RA, Smith KA, Woodroofe MN, Watson PF & Factor-kB and Induces Cell-Cell Adhesion and Aortic Smooth Weetman AP. Detection and localization of chemokine gene Muscle Cell Proliferation. Journal of Biological Chemistry 2004 expression in autoimmune thyroid disease. Clinical Endocrinology 279 3188–3196. 2003 59 207–213.

www.eje-online.org

Downloaded from Bioscientifica.com at 09/27/2021 12:37:12PM via free access EUROPEAN JOURNAL OF ENDOCRINOLOGY (2005) 152 CXCR6 in thyroid autoimmunity 643

36 Garcia-Lopez MA, Sancho D, Sanchez-Madrid F & Marazuela M. 41 Voorby HA, Kabel PJ, de Haan M, Jeucken PH, van der Gaag RD, Thyrocytes from autoimmune thyroid disorders produce the che- de Baets MH & Drexhage HA. Dendritic cells and class II MHC mokines IP-10 and Mig and attract CXCR3 þ lymphocytes. Journal expression on thyrocytes during the autoimmune thyroid disease of Clinical Endocrinology and Metabolism 2001 86 5008–5016. of the BB rat. Clinical Immunology and Immunopathology 1990 55 37 Romagnani P, Rotondi M, Lazzeri E, Lasagni L, Francalanci M, 9–22. Buonamano A, Milani S, Vitti P, Chiovato L, Tonacchera M, 42 Minami M, Kume N, Shimaoka T, Kataoka H, Hayashida K, Bellastella A & Serio M. Expression of IP-10/CXCL10 and Akiyama Y, Nagata I, Ando K, Nobuyoshi M, Hanyuu M et al. MIG/CXCL9 in the thyroid and increased levels of IP-10/CXCL10 Expression of SR-PSOX, a novel cell-surface scavenger receptor in the serum of patients with recent-onset Graves’ disease. American for phosphatidylserine and oxidized LDL in human atherosclerotic Journal of Pathology 2002 161 195–206. lesions. Arteriosclerosis Thrombosis and Vascular Biology 2001 21 38 Paschke R, Vogg M, Swillens S & Usadel KH. Correlation of micro- 1796–1800. somal antibodies with the intensity of the intrathyroidal auto- 43 Aust G & Scherbaum WA. Expression of cytokines in the thyroid: immune process in Graves’ disease. Journal of Clinical thyrocytes as potential cytokine producers. Experimental and Clini- Endocrinology and Metabolism 1993 77 939–943. cal Endocrinology & Diabetes 1996 104 64–67. 39 Weetman AP, McGregor AM, Wheeler MH & Hall R. Extrathyroi- 44 Kasai K, Banba N, Motohashi S, Hattori Y, Manaka K & dal sites of autoantibody synthesis in Graves’ disease. Clinical and Shimoda SI. Expression of monocyte chemoattractant protein-1 Experimental Immunology 1984 56 330–336. mRNA and protein in cultured human thyrocytes. FEBS Letters 40 McLachlan SM, Fawcett J, Atherton MC, Thompson P, Baylis P & 1996 394 137–140. Smith BR. Thyroid autoantibody synthesis by cultures of thyroid and peripheral blood lymphocytes. II. Effect of thyroglobulin on thyroglobulin antibody synthesis. Clinical and Experimental Immu- Received 28 October 2004 nology 1983 52 620–628. Accepted 17 January 2005

www.eje-online.org

Downloaded from Bioscientifica.com at 09/27/2021 12:37:12PM via free access