Increased numbers of Th17 cells in the periphery of Ctrp4−/− mice
In our previous study, we already observed preferential expression of Ctrp4 in T cells, which inspired us to further analyse the function of Ctrp4 in T cell development and differentiation. To this end, we first examined the distribution of thymocytes within the four major thymic populations in the Ctrp4−/− mice and WT controls. We found that eight-week-old CTRP4-deficient mice had undetectable changes in the percentage of CD4+ T cells and had normal thymocyte development, suggesting a minimal role of CTRP4 in T cell development(Fig. 1A-B). Considering that the size of the peripheral T cell pool is stably maintained under homeostatic conditions and that the most of peripheral T cells are in a naïve state with CD44loCD62Lhi, a proportion of memory-like T cells, characterized by CD44hi CD62Llo markers, increase with age because of homeostatic proliferation induced by self-peptide/MHC ligands. Notably, a marked increase in memory-like CD4+ T cells in Ctrp4−/− mouse and a reduction in naïve CD4+ T cells were observed, which commonly tend to indicate autoimmune inflammation (Fig. 1C). However, the flow cytometry analysis showed no differences in the proportion of memory-like CD8+ T in the spleen(Fig. 1D).Then, we examined the effector T cell subsets in the periphery of WT and Ctrp4−/−mice. Among the CD44hi memory T cell population in the spleen, the IL-17A-producing Th17 cell subset was profoundly increased in the Ctrp4−/− mice (Fig. 1E), while the numbers of Treg, Th1 and Th2 cells remained unchanged when compared with those in the WT mice (Fig. 1E, supplemental Fig. 1A-B). Changes in different CD4+ T cell subsets were further measured by Th cell signature genes in CD4+ T cells. When Ctrp4−/− T cells were compared with WT cells, we found that the mRNA expression of Th17 cell lineage-specific genes (Il17a, Il17f, Rorc and Cxcl3) was upregulated significantly (Fig. 1F), whereas Tbx21, Gata3 and Foxp3 gene expression was unaltered (supplemental Fig. 1C). In summary, these data suggest that CTRP4 may be involved in T cells differentiation toward the Th17 cell lineage and suggest the possibility that CTRP4 plays a role in autoimmunity.
2. CTRP4 deficiency in mice exacerbates EAE symptom
T cell responses to self-antigens contribute to the development of many autoimmune and inflammatory diseases. To gain further insight into the pathophysiological roles of CTRP4 in the Th17 cell-mediated autoimmune disease, we generated the EAE models with CTRP4-sufficient and -deficient mice by immunizing them with myelin oligodendrocyte glycoprotein (MOG35 − 55) to mimic human MS. Compared with the wild-type mice, disease progression in the Ctrp4−/− mice showed advanced onset and developed more severe clinical symptoms and more body weight loss, suggesting a role for CTRP4 in reducing EAE induction (Fig. 2A). Haematoxylin and eosin or Luxol fast blue staining revealed that more immune cell infiltration and increased demyelination and axon degeneration in the spinal cord (Fig. 2B). To investigate the population of lymphocytes infiltrating the CNS, we isolated mononuclear cells from the CNS. The immunohistological analysis and flow cytometry showed more CD4+ T cells in neuronal tissue in the Ctrp4−/− mice 18 days after immunization (Fig. 2C, supplemental Fig. 2A). The phenotype of Ctrp4-knockout mice was characterized by an increased frequency and absolute number of CNS-infiltrating CD45+F4/80+ macrophages during the peak phase of EAE (Fig. 2D). Additionally, the number of IL17A+ and IL17A+ IFNγ+ double-producing CD4+ T cells, which reflects the disease severity, was increased in Ctrp4-deficient mice as compared with the number in the spinal cords from CTRP4-expression counterparts (Fig. 2E-F, supplemental Fig. 2B). Furthermore, the infiltration of Th1 and Treg cells was not different between WT and CTRP4-deficient mice (Fig. 2E). To investigate whether the augment of CNS-infiltrating CD4+ cells in the Ctrp4−/− mice was due to increased peripheral Th17 cells or not, we further analysed the proportions of CD4+ T cells and Th17 cells in the draining lymph nodes and spleen. Increased frequencies of Th17 cells corresponded with an increased number of CNS-infiltrating Th17 cells at the peak of the EAE symptoms, suggesting that CTRP4 may influence the number of Th17 cells not only in the CNS but also in the peripheral lymphoid tissues (supplemental Fig. 2D-G).
To determine the role of Ctrp4 in regulating the recall response of MOG35 − 55-specific T cells, we stimulated T cells isolated from draining LNs at the early effector phase of EAE induction and cultured them ex vivo. Upon re-stimulation with MOG peptides, we found that MOG35 − 55-stimulated T cells exhibited substantially increased proliferation in the Ctrp4-deficient mice (Fig. 2G). Consistently, the production of IL-17 and IFN-γ was significantly increased in the Ctrp4−/− mice, which correlated with the clinical score (Fig. 2H). To further elucidate the subsets of MOG35 − 55-peptide-specific CD4+ T cells, CFSElow CD4+ T cells were gated, and an increased percentage of MOG35 − 55-peptide-specific Th17 cells was observed in sample obtained from CTRP4-deficient mice (Fig. 2I).We thus speculated that the loss of CTRP4 promotes EAE disease progression primarily by regulating Th17 cells.
3. CTRP4 deficiency facilitates IL-6-driven Th17 cell differentiation
We further examined whether the increased accumulation of Th17 cells in the CNS in Ctrp4−/− mice was attributed to the different abilities of Th17 cells to migrate, expand, survive, or infiltrate in the absence of CTRP4. We first sorted and cultured naïve CD4+ T cells obtained from Ctrp4−/− and WT mice and cultured them under classical Th17 cell-polarizing condition and analysed their cytokine profiles. The loss of Ctrp4 resulted in a significant difference in the percentage of IL17A-producing cells and increased the production of IL-17A in the cultures (Fig. 3A). Consistent with this finding, CD4+ T cells lacking of Ctrp4 exhibited increased Rorc mRNA expression under Th17 cell-differentiating conditions, while Ctrp4−/− T cells remained unchanged under Th0 cell-differentiating conditions (Fig. 3B). Moreover, IL-23 is critical for the differentiation of pathogenic Th17 cells. Exposure of naïve CD4+ T cells to the cytokine combination of IL-1β + IL-6 + IL-23 results in the acquisition of a pathogenic Th17 cell phenotype. Thus, we next examined whether CTRP4 deficiency affected the differentiation of pathogenic Th17 cells. More IL-17-producing CD4+ cells from Ctrp4−/− mice were observed under pathogenic conditions than were observed among the cells obtained from WT mice, and IL-17A was much more abundant in the Ctrp4−/− cultures than in the control cultures (Fig. 3A). Th17-associated genes, such as Rorc, Il17a and Il17f, were significantly upregulated in the Ctrp4−/− CD4+ T cells, implying that elevated IL-17 secretion can be partially attributed to the increased expression of their mRNAs. In addition, IL23R expression on T cells is essential for Th17 cell stability and pathogenicity. The mRNA expression of Il23r and Ifng was also increased in CD4+ T cells from the Ctrp4−/− mice (Fig. 3C).
Next, we examined whether Ctrp4 deficiency affects T cell proliferative capacity. We performed flow cytometry analysis of CFSE dilution in CD4+ T cells stimulated with TCR activation or Th17 cell-polarizing cytokines. Ctrp4−/− CD4+ T cells showed a similar proliferative capacity (Fig. 3D-E). In addition, we evaluated the apoptotic capacity of CTRP4-deficient CD4+ T cells under the same conditions, and similarly, no significant differences was detected (Fig. 3F-G). With regard to Th17 cell migration, Th17 cells highly express the chemokine receptors CCR6 and CCR2, which allow their recruitment to the CNS 21, 22. Our data demonstrate that there was no difference in the proportion of cells that expressed CCR2 and CCR6 after Ctrp4 was depleted, suggesting that Ctrp4 did not alter the ability of Th17 cells to traffic to sites of inflammation (supplemental Fig. 3A). Consistent with this finding, the CNS of EAE-induced Ctrp4-deficient expressed levels of various chemokines that mediate immune-cell recruitment that were similar to levels expressed by WT- immunized mice (supplemental Fig. 3B). Further, the absence of Ctrp4 in Th17 cells did not lead to changes in the expression of the key activation markers CD25, CD44, CD69 or CD103 (supplemental Fig. 3C). Collectively, these findings further support the possibility that Ctrp4 directs CD4+T cell fate choice toward Th17 cell differentiation without affecting Th17 cell proliferation, apoptosis, migration or activation.
4. CTRP4 inhibits IL-6 activation by directly binding with IL-6R
Many studies have reported that IL-6 signaling is required for the differentiation of Th17 cells, and we hypothesized that CTRP4 probably interacts with IL-6R to suppress IL-6-induced Th17 cell differentiation. We used computational molecular docking experiments performed using Discovery Studio software to analyse the interaction modes on the basis of X-ray crystal structure of IL-6R obtained from the PDB database. The docking results supported our hypothesis (supplemental Fig. 4A). To test this hypothesis, we performed confocal immunofluorescence microscopy. The results revealed CTRP4 colocalization with IL-6R in both the cytoplasm and membrane of Jurkat cells (Fig. 4A). The direct interaction was further confirmed by CTRP4 and IL-6R co-immunoprecipitating from primary CD4+T cells, and reverse immunoprecipitation experiments showed the same results (Fig. 4B). To confirm interactional binding, recombinant CTRP4 protein was labelled with an 125I tracer. Saturation-binding assays demonstrated the direct binding between CTRP4 and IL-6R with a KD of 3.941 nM (Fig. 4C,F). We then evaluated the ability of unlabelled CTRP4, IL-6 and OSM of gp130 family to replace 125I -labelled CTRP4 in the competition-binding assays. Indeed, unlabelled CTRP4 and IL-6 strongly competed with radio-iodinated CTRP4 for binding with IL-6R with an IC50 value of 77.25 nM and 5.233 nM, respectively, while OSM did not exert a competitive effect (Fig. 4D). To determine the region of IL-6R necessary for binding CTRP4, HEK293T cells were transfected with Myc-tagged CTRP4 and different FLAG-tagged domains of IL-6R. Because the D3 domain of IL-6R covers the majority of the IL-6 interface area23, 24, the D3 domain was found to be critical for the IL-6R association with CTRP4, as expected (Fig. 4E).
While these results clearly suggest that CTRP4 interacts with IL-6R, we sought to investigate whether CTRP4 disturbed the formation of the IL-6-to-IL-6R complex for further activation. Thus, HEK293T cells transfected with pmCherry-tagged IL-6 and EGFP-tagged IL-6R, and the significant colocalization was observed in the cytoplasm and membrane. However, the association was decreased in the presence of CTRP4 (Fig. 4H). This results was consistent with the ELASA results. The addition of recombinant CTRP4 protein impaired the interaction of IL-6 and IL-6R in an in vitro binding assay (Fig. 4G). Similarly, co-immunoprecipitation demonstrated that CTRP4 interfered with the binding of IL-6 to IL-6R (Fig. 4I).Collectively, these results demonstrated that CTRP4 may function to suppress IL-6/IL-6R signaling pathway activation during Th17 cell differentiation.
5. CTRP4 prevents IL-6-mediated STAT3 phosphorylation by interacting with IL-6R
IL6 induces Th17 cell differentiation through the activation of the STAT3 pathway25, 26, especially the phosphorylation of STAT3 at tyrosine 70527, 28. To delineate the molecular mechanism underlying the observed inhibitory effect of CTRP4 on Th17 cell differentiation, we assessed the signaling activity of JAK/STAT pathways. First, we determined that the expression of IL-6R and gp130 was comparable between WT and Ctrp4-deficient CD4+ T cells upon TCR stimulation (supplemental Fig. 4A). Furthermore, IL6 stimulation led to increased levels of pSTAT3 in WT cells, and we observed significantly higher levels of pSTAT3 at tyrosine 705 in Ctrp4−/− CD4+ T cells stimulated for 5 and 15 min with physiological concentrations of IL6 (Fig. 5A). Consistent with the results, enhanced STAT3 phosphorylation in the Ctrp4−/− T cells was observed by flow cytometry (Fig. 5B). Among the other signaling molecules tested, pJAK2, pERK and pAkt had higher expression than CD4+ T cells from WT mice, suggesting that the IL6-STAT3 signaling pathway is constitutively hyper-activated after the loss of CTRP4 in CD4+ T cells (Fig. 5A). To address the molecular mechanisms by which CTRP4 inhibits the development of EAE in vivo, we assessed the expression of the IL6 receptor in the context of the EAE model. Flow cytometry analysis revealed that the expression of IL6R and gp130 in the CD4+ T cells from CTRP4-deficient mice was similar to that in the CD4+ T cells from WT mice (supplemental Fig. 4B-D). Of note, the deficiency of CTRP4 in CD4+T cells resulted in a significant increase in Y705-phosphorylated STAT3 in the spinal cord tissue of EAE mice compared to the level in the WT mice, while the abundance of phosphorylated STAT1 in the CD4+T cells was not altered (Fig. 5C).
Previous studies have shown that the IL6 signaling cascade is initiated by the binding of IL6 to membrane-bound IL6R and gp130, which is called a classical signal11. First, we used Ba/F3 cells, which are an IL-3-dependent mouse pro-B-cell line that lacks both endogenous IL-6R and gp130 expression29, 30, to establish cells with gp130 and IL-6R through stable retroviral expression, and then we analysed their proliferation rate in response to IL-6 signaling (supplemental Fig. 5A). IL-6 alone induced the proliferation of Ba/F3-gp130-IL-6R cells, while rhCTRP4 substantially reduced IL-6-mediated proliferation, suggesting that CTRP4 affects classical IL-6 signaling (Fig. 5D). Furthermore, we investigated the effect of rhCTRP4 on IL-6 trans-signaling. After establishing stable expression of gp130 in Ba/F3 cells, the cells proliferated in response to hyper-IL-6 but not in response to IL-6 (supplemental Fig. 5B-C). Exogenous CTRP4 suppressed hyper-IL-6-mediated Ba/F3-gp130 growth (Fig. 5E). This results was validated with MEFs obtained from IL-6R-knockout mice, which do not express membrane-bound IL-6R; therefore, IL-6 stimulation relies on trans-signaling (Fig. 5F). Recombinant CTRP4 protein treatment reduced basal pSTAT3 levels, and reversed IL-6- or hyper-IL-6- induced STAT3 activation (Fig. 5G). Together, these results suggested that CTRP4 influences proliferation by competing with IL-6 for membrane-bound or soluble IL-6R.
6. rhCTRP4 is effective for the treatment of EAE
As CTRP4 is a secreted protein, we first verified whether the addition of rhCTRP4 impairs Th17 cell differentiation. The frequency of the IL17-producing cell population in culture was decreased in the presence of rhCTRP4 to the culture in vitro (Fig. 6A). The CD4+ T cells treated with rhCTRP4 under Th17 cell-differentiating conditions significantly reduced the Rorc and Il17a transcript levels (Fig. 6B), as also illustrated by a markedly decreased IL17A generation (Fig. 6C). To determine whether the degree of the suppressive effect correlated with the dose of rhCTRP4, we treated CD4+ T cells with various concentrations of rhCTRP4. The results indicated that higher doses of rhCTRP4 inhibited Th17 cell differentiation to a lesser extent, suggesting that rhCTRP4 impaired the differentiation of naïve CD4+ T cells towards Th17 cells in a dose-dependent manner (Fig. 6D-E). Consistent with the fact that CTRP4 had an impact on the IL-6 downstream pathway, pretreatment with rhCTRP4 abrogated STAT3 activation in naïve CD4+ T cells in response to IL6 (Fig. 6F). Additionally, the IL-6-triggered STAT3 phosphorylation increase was abolished as early as 5 min after exposure to rhCTRP4 and sustained the inhibitory effect through 60 min (Fig. 6G). Similarly, the enhanced phosphorylation of JAK2 and STAT3 by IL-6 can be attenuated in a dose-dependent manner to normal levels by adding rhCTRP4 (Fig. 6H).Taken together, these results further confirmed that the IL6/STAT3 signaling is negatively regulated by CTRP4.
Next, we wondered whether rhCTRP4 is effective in alleviating established EAE disease in Ctrp4−/− mice. We immunized mice with the MOG peptide, followed by daily intraperitoneal injection of rhCTRP4 on the day of immunization. As illustrated in Fig. 7A, treatment with rhCTRP4 showed significant efficacy and resulted in reduced severity of EAE compared with that observed in control mice. The histological analysis of the spinal cord revealed that administration of rhCTRP4 was accompanied by decreased inflammation and demyelination in the affected spinal cord (Fig. 7B). The mice had a marked reduction in the number of CD4+ cells and Th17 cells infiltrating the CNS upon therapeutic rhCTRP4 administration (Fig. 7C-D). The immunohistological analysis of pSTAT3 in the spinal cord revealed a reduced number of pSTAT3-positive cells in rhCTRP4-treated mice (Fig. 7E). Accordingly, treatment with rhCTRP4 suppressed the phosphorylation of STAT3 and JAK2 in CD4+ T cells at the peak of the disease (Fig. 7F). We also isolated T cells from primed mice and detected their reactivity towards antigen. When challenged with the MOG peptide, the T cells derived from the lymph nodes of the EAE mice treated with rhCTRP4 displayed a markedly dampened proliferative response (Fig. 7G).IL-17A and IFNγ generation was reduced in the MOG-primed T cells from rhCTRP4-injected mice (Fig. 7H). In summary, therapeutic delivery of rhCTRP4 ameliorated the clinical severity of EAE associated with reduced encephalitogenic effector T cells responses.
7. CTRP4 prevents EAE disease in a STAT3-dependent manner
We have shown that Th17 cells accumulate in Ctrp4−/− mice in a manner dependent on the activation of amplified IL-6. We wondered whether the suppressive effect of CTRP4 on STAT3 activation contributes to the protection of the host resistant to EAE. To target STAT3 signaling, we used S3I-201,which is a small-molecule inhibitor that interacts with the STAT3-SH2 domain to suppress STAT3 homodimer formation and STAT3 binding to DNA31. After immunizing Ctrp4-Knockout mice with MOG peptide, the mice were treated either with DMSO or with S3I-201. The results showed that S3I-201 administration resulted in the mitigation of the autoimmune disease compared with disease level in the control mice (Fig. 8A), as reflected by a significant reduction in immune cell infiltration in the spinal cord (Fig. 8B). Similar to the inhibitory effects of CTRP4 in vitro, inhibition of STAT3 has a significant impact on ameliorating disease caused by the depletion of CTRP4 in vivo.
8. MOG-reactive T cells that expand in the presence of rhCTRP4 ameliorated EAE
We next determined whether CTRP4 can influence the pathogenicity of the autoantigen-specific CD4+T cell response in vivo. For this purpose, we immunized B6.SJL mice with MOG35 − 55 emulsified in CFA. Ten days later, we isolated lymphoid cells from the spleen and draining lymph nodes before re-stimulating the cells with MOG peptide, IL-23 and either BSA or rhCTRP4. Compared to the CD4+T cells stimulated with BSA, the CD4+T cells treated with rhCTRP4 showed significantly reduced the frequency of MOG-specific Th17 cells. In agreement with the flow cytometry data, IL-17A release was diminished in the culture supernatants of the CD4+T cells treated with IL-23 and rhCTRP4 (Fig. 8C).
To further determine whether rhCTRP4 incubation affects the pathogenicity of MOG-reactive CD4+T cells, expanded MOG-reactive CD4+T cells were adoptively transferred into irradiated C57BL/6J mice. The majority of the recipient mice injected T cells stimulated with rhCTRP4 exhibited significantly milder symptoms than the mice injected with BSA-treated T cells (Fig. 8D). We observed that the numbers of host CD4+ T cells and CD4+ CD45.1+ donor T cells were both comparable in the CNS in the two groups(Fig. 8E, F).Consistent with this finding, the frequency and absolute number of IL-17-producing cells among the CD45.1+ donor T cell population stimulated with IL-23 plus rhCTRP4 were similar to those of the donor T cells stimulated with IL-23 plus BSA. Notably, given that the comparable number of CD45.1+ CD4+ IL-17A+ donor T cells, we speculate that Th17 cell pathogenic phenotype was impaired. Accordingly, the IL-17 producer were able to co-express GM-CSF and IFNγ, an ability that was linked to the encephalitogenic potential of the Th17 cells and that was reduced in the recipients of the T cells treated with exogenous rhCTRP4 (Fig. 8E, G). Collectively, these results showed that pretreatment with rhCTRP4 during the ex vivo expansion of MOG-reactive CD4+T cells rendered the T cells more encephalitogenic and more proficient in inducing autoimmune CNS inflammation.