Regulatory T cells ameliorate intracerebral hemorrhage-induced inflammatory injury by modulating microglia/macrophage polarization through the IL-10/GSK3β/PTEN axis

Kai Zhou, Qi Zhong, Yan-Chun Wang, Xiao-Yi Xiong, Zhao-You Meng, Ting Zhao, Wen-Yao Zhu, Mao-Fan Liao, Li-Rong Wu, Yuan-Rui Yang, Juan Liu, Chun-Mei Duan, Jie Li, Qiu-Wen Gong, Liang Liu, Mei-Hua Yang, Ao Xiong, Jian Wang, Qing-Wu Yang, Kai Zhou, Qi Zhong, Yan-Chun Wang, Xiao-Yi Xiong, Zhao-You Meng, Ting Zhao, Wen-Yao Zhu, Mao-Fan Liao, Li-Rong Wu, Yuan-Rui Yang, Juan Liu, Chun-Mei Duan, Jie Li, Qiu-Wen Gong, Liang Liu, Mei-Hua Yang, Ao Xiong, Jian Wang, Qing-Wu Yang

Abstract

Inflammation mediated by the peripheral infiltration of inflammatory cells plays an important role in intracerebral hemorrhage (ICH) induced secondary injury. Previous studies have indicated that regulatory T lymphocytes (Tregs) might reduce ICH-induced inflammation, but the precise mechanisms that contribute to ICH-induced inflammatory injury remain unclear. Our results show that the number of Tregs in the brain increases after ICH. Inducing Tregs deletion using a CD25 antibody or Foxp3DTR-mice increased neurological deficient scores (NDS), the level of inflammatory factors, hematoma volumes, and neuronal degeneration. Meanwhile, boosting Tregs using a CD28 super-agonist antibody reduced the inflammatory injury. Furthermore, Tregs depletion shifted microglia/macrophage polarization toward the M1 phenotype while boosting Tregs shifted this transition toward the M2 phenotype. In vitro, a transwell co-culture model of microglia and Tregs indicated that Tregs changed the polarization of microglia, decreased the expression of MHC-II, IL-6, and TNF-α and increased CD206 expression. IL-10 originating from Tregs mediated the microglia polarization by increasing the expression of Glycogen Synthase Kinase 3 beta (GSK3β), which phosphorylates and inactivates Phosphatase and Tensin homologue (PTEN) in microglia, TGF-β did not participate in this conversion. Thus, Tregs ameliorated ICH-induced inflammatory injury by modulating microglia/macrophage polarization toward the M2 phenotype through the IL-10/GSK3β/PTEN axis.

Keywords: Intracerebral hemorrhage; T-cells; inflammation; macrophages; microglia.

Figures

Figure 1.
Figure 1.
The infiltration of Tregs into the CNS after ICH. (a) Representative FACS plots showing that CD45hiCD3+CD4+ cells were defined as brain-invading T helper cells and further analyzed to identify Tregs. FSC-A indicates the forward scatter channel area, and SSC-A indicates side scatter channel area. (b) Temporal changes in brain-invading CD4+T cells and Tregs after ICH, and Sham-operated mice that were analyzed on day 4 post-hemorrhage. (c) Representative FACS plots showing that CD4+Foxp3+ cells are considered Tregs in spleen. (d) The proportions of Foxp3+ cells in CD4+ cells in the brain and the spleen were separately analyzed at 1, 4, 7 and 14 d after ICH (4 individual experiments with four pooled animals per group in each experiment; *p < 0.05 versus the sham group in brain, #p < 0.05 versus the sham group in spleen).
Figure 2.
Figure 2.
Foxp3DTR mice exhibited increased inflammatory injury after ICH. (a) The NDS of WT and Foxp3DTR mice at 1, 3, 5, and 7 d after ICH (*p < 0.05 versus the WT-ICH group at the corresponding time points, n = 12 for each time point). (b) Brain water content at 4 d after hemorrhage or sham operation. (c) Serial coronal sections of mouse brain tissues at 4 d after ICH. (d) Brain sections were used to measure the hematoma volume. (e) Brain homogenate was used to measure the hemoglobin level (**p < 0.01, *p < 0.05 versus the WT-ICH group, n = 4). (f) Representative FJB-positive cells in peri-hematomal tissue of WT and Foxp3DTR groups (bars=20 µm). (g) FJB-positive cells were quantified and compared (**p < 0.01 versus WT-ICH, n = 6). (h) Global changes in M1-related inflammatory factors. (i) Global changes in M2-related inflammatory factors at 4 d after hemorrhage or sham operation were analyzed using RT-PCR (*p < 0.05, **p < 0.01 versus the WT-ICH group, n = 4). Cereb, cerebellum; Cont BG, contralateral basal ganglia; Cont CX, contralateral cortex; Ipsi BG, ipsilateral basal ganglia; Ipsi CX, ipsilateral cortex.
Figure 3.
Figure 3.
CD25 antibodies increased inflammatory injury after ICH. (a) The NDSs in the IgG and AntiCD25 groups at 1, 3, 5, and 7 d after ICH (*p < 0.05 versus the IgG group at the corresponding time points, n = 12 for each time point). (b) Brain water content at 4 d after hemorrhage or sham operation. (c) Serial coronal sections of mouse brain tissues at 4 d after ICH. (d) Brain sections were used to measure the hematoma volume. (e) Brain homogenate was used to measure the hemoglobin level (**p < 0.01, *p < 0.05 versus the WT-ICH group, n = 4). (f) Representative FJB-positive cells in the AntiCD25 and IgG groups (bars=20 µm). (g) FJB-positive cells were quantified and compared (**p < 0.01 versus IgG, n = 6). (h) Global changes in M1-related inflammatory factors. (i) Global changes in M2-related inflammatory factors at 4 d after hemorrhage or sham operation were analyzed using RT-PCR (*p < 0.05, **p < 0.01 versus the IgG group, n = 4).
Figure 4.
Figure 4.
CD28-SA ameliorated inflammatory injury after ICH. (a) The NDSs in the IgG and CD28-SA groups at 1, 3, 5, and 7 d after ICH (*p < 0.05 versus the IgG group at the corresponding time points, n = 12 for each time point). (b) Brain water content at 4 d after hemorrhage or sham operation. (c) Serial coronal sections of mouse brain tissues at 4 d after ICH. (d) Brain sections were used to measure the hematoma volume. (e) Brain homogenate was used to measure the hemoglobin level (**p < 0.01, *p < 0.05 versus the WT-ICH group, n = 4). (f) Representative FJB-positive cells in the CD28-SA and IgG groups (bars=20 µm). (g) FJB-positive cells were quantified and compared (**p < 0.01 versus IgG, n = 6). (h) Global changes in M1-related inflammatory factors. (i) Global changes in M2-related inflammatory factors at 4 d after hemorrhage or sham operation were analyzed using RT-PCR (*p < 0.05, **p < 0.01 versus the IgG group, n = 4).
Figure 5.
Figure 5.
IL-10 originating from Tregs influenced microglia polarization. In vivo experiments. (a) Representative FACS plots for the sham-operated, WT-ICH, Foxp3DTR-ICH, and CD28SA-ICH groups showing that microglia/macrophage were gated by CD45+CD11b+ from brain inflammatory cells and that the expression levels of MHC-II and CD206 were determined by analyzing mean fluorescence intensity (MFI). (b) The absolute number of microglia/macrophages was calculated using flow cytometry, and the MFI of MHC-II and CD206 on these cells were analyzed (*p < 0.05, **p < 0.01 versus the sham group, four individual experiments with three pooled animals per group are shown for each experiment). The in vitro co-culture model further indicated the function of Tregs. (c) TNF-α and IL-6 were detected using RT-PCR in microglia (d) The numbers of microglia and MFI of CD206 and MHC-II on microglia. (e) The MFI was calculated for CD206 and MHC-II (##p < 0.01 versus the vehicle; *p < 0.05, **p < 0.01 versus the microglia + Hb group, statistics were from four separate experiments).
Figure 6.
Figure 6.
GSK3β/PTEN axis mediated the IL-10 induced microglia polarization. (a) A representative Western-blot shows the expression of GSK3b, P-GSK3b, PTEN, and P-PTEN in microglia 40 h after co-culture. (b) The expression of GSK3β was calculated. (c) Representative Western-blot shows the expression of P-PTEN and P-GSK3β in Lentiviral shRNA-infected microglia. (d) The expression of P-PTEN was calculated (##p < 0.01 versus the vehicle; *p < 0.05, **p < 0.01 versus the microglia + Hb group; statistics were from four separate experiments with four wells per trail).

Source: PubMed

3
購読する