CIMB, Vol. 44, Pages 5728-5740: Oral Administration of Myelin Oligodendrocyte Glycoprotein Attenuates Experimental Autoimmune Encephalomyelitis through Induction of Th2/Treg Cells and Suppression of Th1/Th17 Immune Responses

Figure 1. Oral administration of MOG inhibited the development of EAE in C57BL/6 mice. Prevention group received 250 μg of MOG35–55 orally two weeks before EAE induction until 14 days after EAE induction, every day, and treatment group received MOG35–55 orally after EAE induction until 14 days post-immunization. Mice were monitored for signs of EAE, and the results for all mice, were presented as (a) mean clinical score, and (b) body weight. Results were expressed as mean ± SEM. ** p < 0.01, *** p < 0.001, compared with control group. Mice were divided into three groups: 1. Control group (CTRL, n = 9); 2. Prevention group with MOG (n = 9); 3. Treatment group with MOG (n = 9).

Figure 1. Oral administration of MOG inhibited the development of EAE in C57BL/6 mice. Prevention group received 250 μg of MOG35–55 orally two weeks before EAE induction until 14 days after EAE induction, every day, and treatment group received MOG35–55 orally after EAE induction until 14 days post-immunization. Mice were monitored for signs of EAE, and the results for all mice, were presented as (a) mean clinical score, and (b) body weight. Results were expressed as mean ± SEM. ** p < 0.01, *** p < 0.001, compared with control group. Mice were divided into three groups: 1. Control group (CTRL, n = 9); 2. Prevention group with MOG (n = 9); 3. Treatment group with MOG (n = 9).

Cimb 44 00388 g001

Figure 2. MOG administration ameliorates CNS inflammation and demyelination. Histopathological evaluation of CNS from all the mice was performed. Brain sections from each group were collected on day 25 post-immunization, fixed in paraformaldehyde, and embedded in paraffin. Five µm sections from different regions of the brain from each of the groups were stained with (a) Hematoxylin and eosin, to enumerate infiltrating leukocytes, and (b) Luxol fast blue to assess demyelination. Scale bars: 100 μm. (c) CNS inflammatory foci and infiltrating inflammatory cells were quantified. Pathological scores, including inflammation and demyelination, were analyzed and shown as mean scores of pathological inflammation or demyelination ± SEM. *** p < 0.001, when compared with control group. Mice were divided into three groups: 1. Control group (CTRL, n = 9); 2. Prevention group with MOG (n = 9); 3. Treatment group with MOG (n = 9).

Figure 2. MOG administration ameliorates CNS inflammation and demyelination. Histopathological evaluation of CNS from all the mice was performed. Brain sections from each group were collected on day 25 post-immunization, fixed in paraformaldehyde, and embedded in paraffin. Five µm sections from different regions of the brain from each of the groups were stained with (a) Hematoxylin and eosin, to enumerate infiltrating leukocytes, and (b) Luxol fast blue to assess demyelination. Scale bars: 100 μm. (c) CNS inflammatory foci and infiltrating inflammatory cells were quantified. Pathological scores, including inflammation and demyelination, were analyzed and shown as mean scores of pathological inflammation or demyelination ± SEM. *** p < 0.001, when compared with control group. Mice were divided into three groups: 1. Control group (CTRL, n = 9); 2. Prevention group with MOG (n = 9); 3. Treatment group with MOG (n = 9).

Cimb 44 00388 g002

Figure 3. Oral administration of MOG suppresses T-cell proliferation. Splenocytes and lymph nodes were harvested on day 25 post-immunization and cultured in PHA (20 μg/mL), as a positive control, or with MOG (20 μg/mL) for 72 h on 96-well plates. Proliferation responses were tested using a Cell Proliferation ELISA, BrdU (colorimetric) kit (Roche Applied Science, Indianapolis, USA). The proliferation assay was conducted in triplicates. Data presented as mean optical density ± SEM. *** p < 0.001, when compared with control group. Mice were divided into three groups: 1. Control group (CTRL, n = 9); 2. Prevention group with MOG (n = 9); 3. Treatment group with MOG (n = 9).

Figure 3. Oral administration of MOG suppresses T-cell proliferation. Splenocytes and lymph nodes were harvested on day 25 post-immunization and cultured in PHA (20 μg/mL), as a positive control, or with MOG (20 μg/mL) for 72 h on 96-well plates. Proliferation responses were tested using a Cell Proliferation ELISA, BrdU (colorimetric) kit (Roche Applied Science, Indianapolis, USA). The proliferation assay was conducted in triplicates. Data presented as mean optical density ± SEM. *** p < 0.001, when compared with control group. Mice were divided into three groups: 1. Control group (CTRL, n = 9); 2. Prevention group with MOG (n = 9); 3. Treatment group with MOG (n = 9).

Cimb 44 00388 g003

Figure 4. Oral administration of MOG suppressed pro-inflammatory cytokines production and enhanced anti-inflammatory cytokines production in splenocytes and lymph nodes from EAE mice. Splenocytes and lymph nodes from immunized mice from all groups were isolated on day 25 post-immunization and re-stimulated with MOG35–55 (20 μg/mL) for 72 h. Cell culture supernatants were collected and indicated cytokine levels were measured by ELISA. Cytokine assays were conducted in duplicate wells. (a) Anti-inflammatory cytokines IL-4, IL-10, and TGF-β; (b) pro-inflammatory cytokines IFN-γ, TNF-α, and IL-17 released by splenocytes in culture, ±SEM. Results from lymph nodes were similar to splenocytes. ** p < 0.01, *** p < 0.001, when compared with control group. Mice were divided into three groups: 1. Control group (CTRL, n = 9); 2. Prevention group with MOG (n = 9); 3. Treatment group with MOG (n = 9).

Figure 4. Oral administration of MOG suppressed pro-inflammatory cytokines production and enhanced anti-inflammatory cytokines production in splenocytes and lymph nodes from EAE mice. Splenocytes and lymph nodes from immunized mice from all groups were isolated on day 25 post-immunization and re-stimulated with MOG35–55 (20 μg/mL) for 72 h. Cell culture supernatants were collected and indicated cytokine levels were measured by ELISA. Cytokine assays were conducted in duplicate wells. (a) Anti-inflammatory cytokines IL-4, IL-10, and TGF-β; (b) pro-inflammatory cytokines IFN-γ, TNF-α, and IL-17 released by splenocytes in culture, ±SEM. Results from lymph nodes were similar to splenocytes. ** p < 0.01, *** p < 0.001, when compared with control group. Mice were divided into three groups: 1. Control group (CTRL, n = 9); 2. Prevention group with MOG (n = 9); 3. Treatment group with MOG (n = 9).

Cimb 44 00388 g004

Figure 5. Gene expression of cytokines and transcription factors in CNS. On day 25 post-immunization, brains and spinal cords were collected, and mRNA levels of cytokines and transcription factors were assessed by real-time quantitative PCR. The assay was run in triplicates, fold change expression of genes was determined, and then compared to the control group. (a) Th1- and Th17-related cytokines and transcription factors IFN-γ, T-bet, IL-17, and ROR-γt; (b) Th2- and Treg-related cytokines and transcription factors IL-4, GATA3, TGF- β, and FoxP3. Results were expressed as fold change normalized to the control group. ** p < 0.01, *** p < 0.001. Mice were divided into three groups: 1. Control group (CTRL, n = 9); 2. Prevention group with MOG (n = 9); 3. Treatment group with MOG (n = 9).

Figure 5. Gene expression of cytokines and transcription factors in CNS. On day 25 post-immunization, brains and spinal cords were collected, and mRNA levels of cytokines and transcription factors were assessed by real-time quantitative PCR. The assay was run in triplicates, fold change expression of genes was determined, and then compared to the control group. (a) Th1- and Th17-related cytokines and transcription factors IFN-γ, T-bet, IL-17, and ROR-γt; (b) Th2- and Treg-related cytokines and transcription factors IL-4, GATA3, TGF- β, and FoxP3. Results were expressed as fold change normalized to the control group. ** p < 0.01, *** p < 0.001. Mice were divided into three groups: 1. Control group (CTRL, n = 9); 2. Prevention group with MOG (n = 9); 3. Treatment group with MOG (n = 9).

Cimb 44 00388 g005

Table 1. Sequences of primers that were used in the study.

Table 1. Sequences of primers that were used in the study.

GenesForwardReverseIFN-γCCAAGTTTGAGGTCAACACTGGCAGAATTATTCTTATTGGT-betTGTGGTCCAAGTTCAACCCATCCTGTAATGGCTTGTGIL-17CCTCAGACTACCTCAACCCCAGATCACAGAGGGATAROR-γtGGATGAGATTGCCCTCTACCTTGTCGATGAGTCTTGIL-4CTGGATTCATCGATAAGCGATGCTCTTTAGGCTTTCGATA3CTGCGGACTCTACCATAAGTGGTGGTCTGACAGTTCTGF-βCGGACTACTATGCTAAAGACTGTGTGAGATGTCTTTGFoxp3CAGAGTTCTTCCACAACACATGCGAGTAAACCAATGB2mCCTGTATGCTATCCAGAAGTAGCAGTTCAGTATGTTC

Table 2. Clinical features of EAE in the administration of MOG, both as prevention and treatment.

Table 2. Clinical features of EAE in the administration of MOG, both as prevention and treatment.

GroupDay of OnsetMaximal Score
(Score at Peak)Mean Score
(Last Day)Cumulative Disease Index (CDI)CTRL 19.5 ± 0.44.3 ± 0.133.4 ± 0.1151.2 ± 1.27Prevention 211.2 ± 0.3 **1.9 ± 0.14 ***0.8 ± 0.1217 ± 0.61 ***Treatment 310.6 ± 0.4 *2.5 ± 0.14 ***1.5 ± 0.12 ***25.3 ± 0.78 ***

留言 (0)

沒有登入
gif