Abstract
Toll-like receptors (TLRs), the innate immune receptors, act as sentinels bridging both innate and adaptive arms of immunity. In the present study, we estimated TLR-induced secretion of IL-27, IL-12, IL-23, IL-8, IP-10, IL-17, IL-6 and TNF-α (by ELISA) and expression of Human Leukocyte Antigen- (Human Leukocyte Antigen - antigen D Related (HLA-DR), CD69, CD80 (also known as B7–1) (by flowcytometry) and Activating Transcription Factor 3(ATF3) (by qRT-PCR) in whole blood cultures of control and type-2 diabetic (both newly diagnosed/NDD and known/KDM) subjects. TLR-induced secretion of IL-27 was significantly reduced in the NDD group compared to the control (Normal Glucose Tolerance (NGT)) and KDM groups. On the other hand, the expression of CD80 was significantly upregulated in both the monocytes and B cells in KDM group. This was associated with increased T cell activation (CD3+CD69+HLA-DR+) with increased IL-17 and reduced TNF-α secretion in this group. Impaired TLR-induced IL-27 secretion and augmented expression of antigen presentation molecules result in chronic T cell activation which may fuel T cell-mediated inflammation in type-2 diabetes.
Keywords: TLR, D80, HLA-DR, IL-27, Type-2 diabetes
1. Introduction
Chronic inflammation and impaired immunity are two key defects in type-2 diabetes mellitus [1]. Toll-like receptors (TLRs) are innate immune receptors which act as sentinels bridging both innate and adaptive arms of immunity [2]. Out of the 10 TLRs known in humans, TLR2 and 4 have been implicated in T-cell activation wherein they serve as co-stimulatory receptors directly modulating T cell activity [3,4]. On the other hand, TLRs present in professional antigen presenting cells (APCs), when activated, can induce antigen presentation machinery (HLA-DR and CD80/B7–1) and cytokines/chemokine secretion, thereby orchestrating the recruitment and activation of T cells [5]. Aberrant expression of these molecules can result in inappropriate T cell activation leading to inflammation and impaired immunity which has been reported in diabetic nephropathy [6,7]. TLR induced secretion of Th polarizing cytokines especially those belonging to the IL-12 family (IL-12, IL-23 and IL-27) have gained much importance since this marks the transition from innate to adaptive immune responses [8]. Th polarizing cytokines, T cell attracting chemokines and antigen presentation machinery together determine the phenotype of the adaptive immune response [8]. Recently, ATF3 a bZib transcription factor which was previously implicated in the negative regulation of TLR signalling has now been identified as a major hub of adaptive immunity [9].
IL-27, a recently described novel cytokine, belonging to the IL-12 cytokine family, has gained major attention in recent years because of its therapeutic potential in treating diabetes [10]. Compared to other IL-12 family members, the role of IL-27 in inflammation is enigmatic since it has been shown to promote both pro- and anti-inflammatory responses [11]. At one end, it promote IFN-γ secretion from Th1 cells [12] and on the other end it inhibits Th1, Th2, and Th17 differentiation [13]. Even though IL-27 has largely been implicated in autoimmune diseases [11], its role in chronic inflammatory conditions like type-2 diabetes is largely unknown. Serum levels of IL-12 [14], IL-23 [15], IL-27 [16], IL-8 [17] and IL-17 [18] were found to be altered in Type-2 diabetes. However, the effect of type-2 diabetes on TLR-induced secretion of these cytokines is not known. Thus, in the present study TLR-induced secretion of IL-27 along with other Th polarising cytokines (IL-12 and IL-23) and T-cell attracting chemokines (IL-8 and IP-10) in newly diagnosed and known type-2 diabetes (chronic) subjects was estimated and was compared with age and gender matched normoglycemic controls. We also evaluated the expression of TLR-induced expression of antigen presentation molecules (HLA-DR and CD80) and T cell activation (HLA-DR and CD69) in these subjects.
2. Materials and methods
2.1. Study subjects
The participants for this study were selected from the outpatients visiting Dr. Mohan’s Diabetes Specialities Centre, Chennai based on the inclusion and exclusion criteria as reported previously [19]. Institutional ethical approval (Ref No-MDRF-EC/SOC/2009//05) was obtained for this study. All the subjects had given their informed consent in writing. As a pilot experiment, controls and age matched diabetic subjects (newly diagnosed and known), 20 per group were included in the study. On the basis of the preliminary results, with a confidence interval of 95%, an estimated p value < 0.05, and a power of 80%, we derived at a sample size of 30 per group. Furthermore, based on our earlier studies we increased the numbers under each group to accommodate for the wide variation generally seen in clinical studies.
The study subjects include: 1. Healthy subjects with Normal Glucose Tolerance (NGT) (n = 42) who served as control; 2. Subjects with newly diagnosed diabetes and not under anti-diabetic medication (NDD) (n = 34) and 3. Subjects with known diabetes and were under anti-diabetic medication (KDM) (n = 53). The in vitro cytokine analysis was performed for all the samples, while the other experiments were done using few samples from NGT (n = 13), NDD (n = 14) and KDM (n = 15) groups which were randomly selected. The study was conducted as per the declaration of Helsinki.
2.2. Diagnosis of type-2 diabetes
Type 2 Diabetes was diagnosed as per WHO guidelines. Subjects were allotted into groups as Control (NGT) and newly diagnosed diabetes based on the Oral Glucose Tolerance Test (OGTT).
2.3. Anthropometric measurements and biochemical parameters
Anthropometric measurements (height, weight and waist circumference) were acquired using standard methods. Serum biochemical parameters including blood glucose, serum cholesterol, serum triglycerides, HDL-cholesterol, urea and creatinine were estimated using a Hitachi-912 Autoanalyser (Hitachi, Mannheim, Germany) [19]. Glycated hemoglobin (HbA1 c) was measured using high pressure liquid chromatography (Bio-Rad, Hercules, CA) [19].
2.4. Peripheral blood Leukocyte cultures
Immediately after the withdrawal of the peripheral blood, plasma was separated and the Packed Cell Volume (PCV) was diluted with RPMI medium (1:1 ratio) containing 10% FCS. Cells were either stimulated with TLR2 ligand-PAM3 (100 ng/ml) or TLR4 ligand-LPS (100 ng/ml) or were left unstimulated for 24 h in parallel cultures. Supernatants were collected for cytokine estimation from all samples. The cell pellets were either solubilized in RNAzol or fixed with 4% paraformaldehyde for gene expression and flowcytometry analysis respectively. All the processed samples were stored at −80 °C until analysis.
2.5. Real-time PCR analysis
RNA extraction from cell pellets solubilised in RNAzol was performed using RNeasy Mini Kit (Qiagen). 1 μg of RNA was used for cDNA conversion and real-time PCR was performed using TaqMan probes (Applied Biosystems, US) specific for ATF3 (Assay ID: Hs00231069_m1) and 18 S (control) (Assay ID: Hs99999901_s1).
2.6. Measurement of cytokines by ELISA
The levels of IL-27 (R & D, US), IL-12, IL-17, IL-23, IL-8 and IP-10 in the cell supernatant were estimated using ELISA (Invitrogen, US) as per the protocol given by the manufacturer. The lower detection limits were: IL-12–1.95 pg/ml, IL-17–0.97 pg/ml, IL-23–1.95 pg/ml, IL-2719.5 pg/ml, TGF-β−0.97 pg/ml, IL-8–3.91 pg/ml and IP-10–0.97 pg/ml. The coefficient of variance was found to be < 10%.
2.7. Flowcytometry analysis
Cells were stained with flurochrome conjugated monoclonal antibodies specific for CD3, CD14, CD19, CD80, CD69, HLA-DR, IL-6 and TNF-α (BD Biosciences, US) and were analyzed on a FACS Canto (BD Biosciences, US). The monocytes and lymphocytes were first gated on the FSC Vs SSC plot. Monocytes were further gated based on CD14 bright cell population on a CD14 Vs SSC plot (S.Fig. 1). B and T cells were gated on a CD3 Vs CD19 plot and were identified as single positive population (B cells-CD3−CD19+; T cells- CD3+CD19−) (S.Figs. 2 and 3). The expression of CD80 and HLA-DR on monocytes and B cells were analysed on CD80/HLA-DR Vs SSC plots on gated monocytes and B cells. The expression of TNF-α and CD69 on T cells were analysed on CD69/IL-6 Vs SSC plots on gated T cells. Both the percentage of cells and mean fluorescence intensity (MFI) were quantified within the gated population of cells. A minimum of 50,000 events were acquired for each tube to arrive at a statistical significant result.
2.8. Statistical analysis
Continuous variables among the groups were compared using Student t-test, whereas proportions were compared using χ2 test or Fisher exact test (as appropriate). Parameters which did not show normal distribution were compared using Kruskal-Wallis test. Holm’s correction was performed in multiple comparisons. GraphPad Prism version 5.0 (GraphPad Software, US) software was used to perform all the above mentioned statistical analysis. p < 0.05 was considered to be significant.
3. Results
3.1. Clinical characteristics of the study subjects
The clinical and biochemical characteristics of the study subjects have been described previously [19]. The average duration of diabetes in KDM group is approximately two to ten years. None of them had microvascular (diabetic retinopathy, nephropathy and neuropathy) and macrovascular (diabetic cardiomyopathy and peripheral arterial disease) complications. With respect to medication: 35% were on insulin, 41% on metformin, 18% on thiazolidinedione, 41% on sulfonylurea, 18% on medlitide, 18% on alpha-glucosidase inhibitor and 18% on mecobalamin. 20% of subjects were on more than one drug combination.
3.2. TLR-induced secretion of IL-27, IL-12, IL-23, IL-8 and IP-10 in the supernatant of PBL cultures
In the present study, we measured TLR-induced secretion of IL-27, IL-12, IL-23, IL-8 and IP-10 in the supernatant of PBL cultures (Fig. 1). While significantly increased secretion of IL-12 and IL-27 was seen in both the TLR2 and TLR4 stimulated cultures (Fig. 1b and c), moderate levels of IL-23 secretion was seen only in the TLR4 stimulated cultures (Fig. 1d). IL-27 was the only cytokine which was significantly downregulated in the NDD compared to NGT and KDM groups. However, serum IL-27 showed no difference between the groups (Fig. 1a). With respect to chemokines, while significantly increased IL-8 secretion was seen in both the TLR2 and 4 stimulated cultures (Fig. 1e), IP-10 secretion was seen only in the TLR4 stimulated cultures (Fig. 1f). However no significant difference was seen in the secretion between the groups. Both IL-33 and TGF-β secretion could not be detected in the culture supernatants (data not shown).
Fig. 1. TLR-induced secretion of Th polarizing cytokines and T cell attracting chemokines in PBL cultures of type-2 diabetic subjects as determined by ELISA.
Histogram showing serum levels of IL-27 (a). Histogram showing TLR2 and 4 induced secretion of IL-27 (b), IL-12 (c), IL-23 (d), IL-8 (e) and IP-10 (f) in the supernatants of PBL cultures in NGT/control (n = 42), NDD (n = 34) and KDM (n = 53) subjects. Statistical significance was determined by non-parametric Mann-Whitney U test and p < 0.05 was considered significant. *p < 0.05; **p < 0.01; ***p < 0.001. NGT- normal glucose tolerance, NDD- newly diagnosed diabetic, KDM- known diabetic subjects, PBL- Peripheral blood leukocytes.
3.3. TLR-induced expression of CD80 and HLA-DR in monocytes and B cells
Next, we studied TLR-induced expression of CD80 and HLA-DR in monocytes (Fig. 2a and b) and B cells (Fig. 2c, d, e and f). Both the basal and TLR-induced level of CD80 was significantly upregulated in both the monocytes and B cells in the KDM group compared to NGT and NDD groups (Fig. 2a, c and e). No significant difference was noted in HLA-DR expression in monocytes and B cells under basal level between groups (Fig. 2b, d, f). Following TLR stimulation, upregulation of HLA-DR was seen only in monocytes (but not in B cells) which was comparable between the groups (Fig. 2b and d).
Fig. 2. TLR-induced expression of CD80 and HLA-DR in monocytes and B cells in PBL cultures of type-2 diabetic subjects as determined by flowcytometry.
Histogram showing the percentage of monocytes, expressing CD80 (a) and HLA-DR (b), and B cells, expressing CD80 (c) and HLA-DR (d), in NGT/control (n = 13), NDD (n = 14) and KDM (n = 15) subjects. Histogram analysis showing the upregulation of CD80 (e) but not HLA-DR (f) in TLR4 stimulated B cells in KDM group (Shaded area-Isotype control; dotted line-NGT; dashed line-NDD and grey line-KDM). Statistical significance was determined by non-parametric Mann-Whitney U test and p < 0.05 was considered significant. *p < 0.05. NGT- normal glucose tolerance, NDD- newly diagnosed diabetic, KDM- known diabetic subjects.
3.4. TLR-induced expression of HLA-DR, CD69, TNF-α and IL-17 in T-cells
Next, we analyzed TLR-induced expression of HLA-DR, CD69, TNF-α and IL-17 in T-cells (Fig. 3). At the basal level, significant upregulation of HLA-DR (Fig. 3a and b), CD69 (Fig. 3c) and secretion of IL-17 (Fig. 3e) was seen in T cells from the KDM group. Upon TLR stimulation strong upregulation of HLA-DR (Fig. 3a) and CD69 (Fig. 3c) was noted. The upregulation of HLA-DR was significantly higher in both the NDD and KDM groups compared to NGT group (Fig. 3b). T cells from the NDD group failed to upregulate CD69 following TLR stimulation (Fig. 3c). CD69 was the only marker which showed significant difference between the NGT and NDD groups. Similarly, T cells from diabetic subjects (both NDD and KDM) failed to upregulate TNF-α following TLR stimulation (Fig. 3d). On the contrary, the basal level secretion of IL-17 was strongly upregulated in the KDM group compared to NDD even in the absence of TLR stimulation (Fig. 3e). No T cell apoptosis (expression of activated caspase-3) or IL-6 secretion were detected (data not shown). Unlike TNF-α, no immunophenotyping was carried out for IL-17 since it is well known that this cytokine is predominantly secreted by T cells in PBL cultures. Finally, ATF-3 showed significantly reduced expression in the KDM group, compared to NDD group (Fig. 3f). Spearman’s correlation analysis revealed a negative correlation between TLR2 induced IL-27 secretion and CD3+HLA-DR expression and positive correlation between TLR2 induced IL-27 secretion (Fig. 3g) and CD3+CD69 expression (Fig. 3h) in the NDD group.
Fig. 3. TLR-induced activation of T cells in PBL cultures of type-2 diabetic subjects as determined by flowcytometry and real-time PCR.
Long standing diabetes is characterized by increased TLR-induced HLA-DR expression and decreased TNF-α expression in T cells. Histogram showing the percentage of T cells expressing CD80 (a), HLA-DR (b), CD69 (c) and TNF-α (d). Histogram showing the levels of IL-17 (e) in the supernatants of PBL cultures and the mRNA expression of ATF3 (f) shown as dct following TLR2 and 4 stimulation in NGT/control (n = 42), NDD (n = 34) and KDM (n = 53) subjects. Spearman’s correlation analysis showing the association between IL-27 secretion and HLA-DR (g) and CD69 (h) expression in T-cells. Statistical significance was determined by non-parametric Mann-Whitney U test and p < 0.05 was considered significant. *p < 0.05; **p < 0.01. PBL- Peripheral blood leukocytes, NGT- normal glucose tolerance, NDD- newly diagnosed diabetic, KDM- known diabetic subjects, dct - delta cycle threshold.
4. Discussion
Even though TLR-induced secretion of cytokines is well studied, comparatively the antigen processing/presentation pathway is less well studied in type-2 diabetic subjects. Monocytes and B cells serve as major APCs which when activated with TLRs express HLA-DR and CD80 molecules and secrete cytokines and chemokines [20,21]. The chemokines secreted by the APCs attract T cells, which then recognize antigen bound MHC and CD80 molecules and get activated/polarized by the Th polarizing cytokines [22]. The cumulative effect of these three signals namely 1. Peptide-MHC (which is recognised by TCR), 2. CD80 (which is recognized by CD28) and 3. Th polarizing cytokines (which is recognized by cytokine receptors) emanating from APCs determine the activation/polarizing status of the T cells. IL-12 (for Th1), IL-23 (for Th17), IL-33 (for Th2) and TGF-β (for Treg) have now been identified as the major Th polarizing cytokines [23], while IL-8 and IP-10 are the major T cell attracting chemokines, secreted by APCs [24]. IL-27 is a recently identified cytokine which belongs to the IL-12 family. However, unlike its other family members, it does not polarize Th cells, but augments the secretion of IFN-γ from Th1 cells and inhibits other Th cells viz IL-10 secretion [25]. Thus, in the present study we looked at the complete TLR-induced antigen processing/ presentation machinery operating in the transition of innate and adaptive immune responses in type-2 diabetic subjects. The major findings of the study are 1. Out of three major T cell polarizing IL-12 family members, only IL-27 secretion was found to be defective in the newly diagnosed type-2 diabetic subjects, 2. TLR-induced expression of CD80 was found to be significantly upregulated in both the monocytes and B cells in chronic type-2 diabetic subjects and 3. Finally, TLR-induced activated T cells (CD3+HLA-DR+) secreted increased IL-17 and reduced TNF-α in chronic type-2 diabetic subjects.
Out of three major T cell polarizing IL-12 family members, only IL-27 secretion was found to be defective in newly diagnosed type-2 diabetic subjects. However, serum IL-27 levels were not significantly different between the groups studied. The defective secretion of IL-27 was seen only in the NDD group and resembles TLR-induced secretion of pro- and anti-inflammatory cytokines (as we reported previously) [19]. Previous reports have indicated significantly increased levels of IL-27 in subjects with diabetes and coronary artery disease (DM-CAD) [26] and reduced levels in subjects with proliferative diabetic retinopathy (PDR) [16]. The ability of IL-27 to suppress diabetogenic Th17 cells has created renewed interest in using this cytokine for immunetherapy against several autoimmune diseases including diabetes [27]. At least in animal models, neutralization of IL-27 inhibited disease progression in NOD mice [27]. However, IL-27 treatment itself reduced hyperglycemia and pancreatic beta cell loss in streptozotocin induced diabetes, making its role in diabetes highly controversial [28]. The disparity could be due to the ability of IL-27 to suppress IL-22 producing Th17 cells under certain conditions [29] and not under other conditions [30].
Next, we looked at the expression of HLA-DR and CD80 in professional antigen presenting cells (monocytes and B cells). TLR-induced expression of CD80 was found to be significantly upregulated in both the monocytes and B cells in chronic type-2 diabetic subjects. It is important to note that this immunophenotype was seen only in the KDM group and not in NDD group indicating impaired metabolic memory in the chronic activation of these APCs. Previously, we have reported significant upregulation of CD80 in dentritic cells from type-2 diabetic subjects, which was due to the high levels of serum GM-CSF as seen in these subjects [31]. Our results are in agreement with Giulietti et al., who reported increased expression of CD80 in macrophages obtained from type-2 diabetic subjects [32]. Further, type-2 diabetic subjects were found to have aberrant soluble co-stimulatory molecule profile in the serum with increased sCD28 and decreased sCTLA-4 indicating augmented CD80 pathway [6]. To summarise, a chronically activated state with enhanced CD80 expression was noted in both monocytes and B cells in the known diabetic group.
Next, we looked at the activation status of T cells in newly diagnosed and chronic diabetic subjects. TLR mediated activation of T cells is less well studied compared to the activation of APCs. Since IL-27 has a dual effect of both activating and inhibiting T cells, we looked at TLR-induced T cell activation using both early (CD69) and late (HLA-DR) activation markers. Previously, our attempt to activate naïve T cells with TLR ligands was not successful. Hence, in the present work, we used the whole blood cultures to activate T cells. Significant upregulation of CD69 and HLA-DR was seen in T cells even at the basal level in the KDM group which was further upregulated by TLR stimulation. In the present study, we selected CD69 instead of CD25 since the former is an early activation marker and is predominantly expressed in effector T-cells while the latter is expressed in both effector and regulatory T-cells. While CD69 represent recent activation, expression of HLA-DR in T cells represents chronic activation and has been implicated in antigenic cross-presentation of T cells [33]. HLA-DR+ T cells have been reported in type-1 diabetic subjects and serves as a major risk factor for disease progression [34]. However, as far as we know this is the first report documenting HLA-DR+ T cells in type-2 diabetes. Following TLR stimulation, these chronically activated T cells, failed to secrete TNF-α, but constitutively secreted high levels of IL-17. The exact mechanism associated with the defective TLR-induced IL-27 secretion in the NDD group is not known but could be due to the upregulation of ATF3 since this transcription factor is the major negative regulator of TLR signalling [9]. Further, in the NDD group, strong positive correlation between CD69 and IL-27 secretion and negative correlation between HLA-DR and IL-27 secretion indicates that IL-27 might be more associated with chronic T cell activation [12,25]. Unlike TLR2 and 4 other TLRs were poorly expressed in T cells and were not studied in this report. Because of the cross-sectional design, no direct cause and effect relationship could be drawn from this study which is a major limitation. Nevertheless, this study gains importance in that it identifies major defects in an important component of TLR signalling pathway in type-2 diabetic subjects. Impaired TLR-induced IL-27 secretion and augmented expression of antigen presentation machinery results in chronic T cell activation which may fuel T cell-mediated inflammation in type-2 diabetes. Infiltration of these T cells into adipose tissue can worsen insulin resistance. At the same time, defective secretion of TNF-α by these T cells can impair immunity against infections as frequently seen among Type-2 diabetic subjects.
Supplementary Material
Acknowledgement
We thank Sajid Bhat and N. Pavankumar for their technical support. The project was funded by Fast Track Scheme for Young Scientists, Department of Science & Technology, New Delhi (SR/FT/LS-105/2009). The Dept of Genetics, University of Madras has received funds for infrastructural support from DST-FIST and UGC-SAP programs.
Footnotes
Conflict of interest
None.
Appendix A. Supplementary material
Supplementary data associated with this article can be found, in the online version, at http://dx.doi.org/10.1016/j.cyto.2017.09.032.
References
- [1].Kawai T, Akira S, Toll-like receptors and their crosstalk with other innate receptors in infection and immunity, Immunity 34 (2011) 637–650. [DOI] [PubMed] [Google Scholar]
- [2].De Nardo D, Toll-like receptors: Activation, signalling and transcriptional modulation, Cytokine 74 (2015) 181–189. [DOI] [PubMed] [Google Scholar]
- [3].Rahman AH, Taylor DK, Turka LA, The contribution of direct TLR signaling to T cell responses, Immunol. Res 45 (2009) 25–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [4].Komai-Koma M, Jones L, Ogg GS, Xu D, Liew FY, TLR2 is expressed on activated T cells as a costimulatory receptor, Proc. Natl. Acad. Sci. USA 101 (2004) 3029–3034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [5].Iwasaki A, Medzhitov R, Toll-like receptor control of the adaptive immune responses, Nat. Immunol 5 (2004) 987–995. [DOI] [PubMed] [Google Scholar]
- [6].Wong CK, Ho AW, Tong PC, Yeung CY, Chan JC, Kong AP, Lam CW, Aberrant expression of soluble co-stimulatory molecules and adhesion molecules in type 2 diabetic patients with nephropathy, J. Clin. Immunol 28 (2008) 36–43. [DOI] [PubMed] [Google Scholar]
- [7].Fiorina P, Vergani A, Bassi R, Niewczas MA, Altintas MM, Pezzolesi MG, D’Addio F, Chin M, Tezza S, Ben Nasr M, Mattinzoli D, Ikehata M, Corradi D, Schumacher V, Buvall L, Yu CC, Chang JM, La Rosa S, Finzi G, Solini A, Vincenti F, Rastaldi MP, Reiser J, Krolewski AS, Mundel PH, Sayegh MH, Role of podocyte B7–1 in diabetic nephropathy, J. Am. Soc. Nephrol 25 (2014) 1415–1429. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [8].Abdelsadik A, Trad A, Toll-like receptors on the fork roads between innate and adaptive immunity, Hum. Immunol 72 (2011) 1188–1193. [DOI] [PubMed] [Google Scholar]
- [9].Gilchrist M, Thorsson V, Li B, Rust AG, Korb M, Roach JC, Kennedy K, Hai T, Bolouri H, Aderem A, Systems biology approaches identify ATF3 as a negative regulator of Toll-like receptor 4, Nature 441 (2006) 173–178. [DOI] [PubMed] [Google Scholar]
- [10].Zamani F, Almasi S, Kazemi T, Jahanban Esfahlan R, Aliparasti MR, New Approaches to the Immunotherapy of Type 1 Diabetes Mellitus Using Interleukin-27, Adv. Pharm. Bull 5 (2016) 599–603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [11].Meka RR, Venkatesha SH, Dudics S, Acharya B, Moudgil KD, IL-27-induced modulation of autoimmunity and its therapeutic potential, Autoimmun. Rev 14 (2015) 1131–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [12].Pflanz S, Timans JC, Cheung J, Rosales R, Kanzler H, Gilbert J, Hibbert L, Churakova T, Travis M, Vaisberg E, Blumenschein WM, Mattson JD, Wagner JL, To W, Zurawski S, McClanahan TK, Gorman DM, Bazan JF, de Waal Malefyt R, Rennick D, Kastelein RA, IL-27, a heterodimeric cytokine composed of EBI3 and p28 protein, induces proliferation of naive CD4+ T cells, Immunity 16 (2002) 779–790. [DOI] [PubMed] [Google Scholar]
- [13].Iwasaki Y, Fujio K, Okamura T, Yamamoto K, Interleukin-27 in T cell immunity, Int. J. Mol. Sci 16 (2015) 2851–2863. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [14].Wegner M, Winiarska H, Bobkiewicz-Kozlowska T, Dworacka M, IL-12 serum levels in patients with type 2 diabetes treated with sulphonylureas, Cytokine 42 (2008) 312–316. [DOI] [PubMed] [Google Scholar]
- [15].Roohi A, Tabrizi M, Abbasi F, Ataie-Jafari A, Nikbin B, Larijani B, Qorbani M, Meysamie A, Asgarian-Omran H, Nikmanesh B, Bajouri A, Shafiey N, Maleki A, Serum IL-17, IL-23, and TGF-beta levels in type 1 and type 2 diabetic patients and age-matched healthy controls, Biomed. Res. Int 2014 (2014) 718946. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [16].Yan A, You H, Zhang X, Levels of Interleukin 27 and Interleukin 35 in the Serum and Vitreous of Patients with Proliferative Diabetic Retinopathy, Ocul. Immunol. Inflamm (2016) 1–7. [DOI] [PubMed] [Google Scholar]
- [17].Doganay S, Evereklioglu C, Er H, Turkoz Y, Sevinc A, Mehmet N, Savli H, Comparison of serum NO, TNF-alpha, IL-1beta, sIL-2R, IL-6 and IL-8 levels with grades of retinopathy in patients with diabetes mellitus, Eye (Lond) 16 (2002) 163–170. [DOI] [PubMed] [Google Scholar]
- [18].Vasanthakumar R, Mohan V, Anand G, Deepa M, Babu S, Aravindhan V, Serum IL-9, IL-17, and TGF-beta levels in subjects with diabetic kidney disease (CURES-134), Cytokine 72 (2014) 109–112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [19].Madhumitha H, Mohan V, Kumar NP, Pradeepa R, Babu S, Aravindhan V, Impaired toll-like receptor signalling in peripheral B cells from newly diagnosed type-2 diabetic subjects, Cytokine 76 (2015) 253–259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [20].Randolph GJ, Jakubzick C, Qu C, Antigen presentation by monocytes and monocyte-derived cells, Curr. Opin. Immunol 20 (2008) 52–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [21].Inaba K, Metlay JP, Crowley MT, Witmer-Pack M, Steinman RM, Dendritic cells as antigen presenting cells in vivo, Int. Rev. Immunol 6 (1990) 197–206. [DOI] [PubMed] [Google Scholar]
- [22].Steinman RM, Hemmi H, Dendritic cells: translating innate to adaptive immunity, Curr. Top. Microbiol. Immunol 311 (2006) 17–58. [DOI] [PubMed] [Google Scholar]
- [23].Raphael I, Nalawade S, Eagar TN, Forsthuber TG, T cell subsets and their signature cytokines in autoimmune and inflammatory diseases, Cytokine 74 (2015) 5–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [24].Ansel KM, McHeyzer-Williams LJ, Ngo VN, McHeyzer-Williams MG, Cyster JG, In vivo-activated CD4 T cells upregulate CXC chemokine receptor 5 and reprogram their response to lymphoid chemokines, J. Exp. Med 190 (1999) 1123–1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [25].Villarino A, Hibbert L, Lieberman L, Wilson E, Mak T, Yoshida H, Kastelein RA, Saris C, Hunter CA, The IL-27R (WSX-1) is required to suppress T cell hyperactivity during infection, Immunity 19 (2003) 645–655. [DOI] [PubMed] [Google Scholar]
- [26].Gong F, Wu J, Zhou P, Zhang M, Liu J, Liu Y, Lu X, Liu Z, Interleukin-22 Might Act as a Double-Edged Sword in Type 2 Diabetes and Coronary Artery Disease, Mediators Inflamm 2016 (2016) 8254797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [27].Wang R, Han G, Wang J, Chen G, Xu R, Wang L, Li X, Shen B, Li Y, The pathogenic role of interleukin-27 in autoimmune diabetes, Cell. Mol. Life Sci 65 (2008) 3851–3860. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [28].Fujimoto H, Hirase T, Miyazaki Y, Hara H, Ide-Iwata N, Nishimoto-Hazuku A, Saris CJ, Yoshida H, Node K, IL-27 inhibits hyperglycemia and pancreatic islet inflammation induced by streptozotocin in mice, Am. J. Pathol 179 (2011) 2327–2336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [29].Bellemore SM, Nikoopour E, Krougly O, Lee-Chan E, Fouser LA, Singh B, Pathogenic T helper type 17 cells contribute to type 1 diabetes independently of interleukin-22, Clin. Exp. Immunol 183 (2015) 380–388. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [30].El-Behi M, Dai H, Magalhaes JG, Hwang D, Zhang GX, Rostami A, Ciric B, Committed Tc17 cells are phenotypically and functionally resistant to the effects of IL-27, Eur. J. Immunol 44 (2014) 3003–3014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [31].Surendar J, Mohan V, Pavankumar N, Babu S, Aravindhan V, Increased levels of serum granulocyte-macrophage colony-stimulating factor is associated with activated peripheral dendritic cells in type 2 diabetes subjects (CURES-99), Diabetes Technol. Ther 14 (2011) 344–349. [DOI] [PubMed] [Google Scholar]
- [32].Giulietti A, van Etten E, Overbergh L, Stoffels K, Bouillon R, Mathieu C, Monocytes from type 2 diabetic patients have a pro-inflammatory profile. 1,25-Dihydroxyvitamin D(3) works as anti-inflammatory, Diabetes Res. Clin. Pract 77 (2007) 47–57. [DOI] [PubMed] [Google Scholar]
- [33].Ko HS, Fu SM, Winchester RJ, Yu DT, Kunkel HG, Ia determinants on stimulated human T lymphocytes. Occurrence on mitogen- and antigen-activated T cells, J. Exp. Med 150 (1979) 246–255. [DOI] [PMC free article] [PubMed] [Google Scholar]
- [34].Baker C, Chang L, Elsegood KA, Bishop AJ, Gannon DH, Narendran P, Leech NJ, Dayan CM, Activated T cell subsets in human type 1 diabetes: evidence for expansion of the DR+ CD30+ subpopulation in new-onset disease, Clin. Exp. Immunol 147 (2007) 472–482. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.



