Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Treatment with Vitamin D/MOG Association Suppresses Experimental Autoimmune Encephalomyelitis

Correction

17 Jul 2015: The PLOS ONE Staff (2015) Correction: Treatment with Vitamin D/MOG Association Suppresses Experimental Autoimmune Encephalomyelitis. PLOS ONE 10(7): e0131260. https://doi.org/10.1371/journal.pone.0131260 View correction

Abstract

Experimental autoimmune encephalomyelitis (EAE) is an animal model to study multiple sclerosis (MS). Considering the tolerogenic effects of active vitamin D, we evaluated the therapeutic effect of myelin oligodendrocyte glycoprotein (MOG) associated with active vitamin D in EAE development. EAE was induced in female C57BL/6 mice by immunization with MOG emulsified with Complete Freund’s Adjuvant plus Mycobacterium tuberculosis. Animals also received two intraperitoneal doses of Bordetella pertussis toxin. One day after immunization, mice were treated with 0,1μg of 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) every other day during 15 days (on days 1, 3, 5, 7, 9, 11, 13 and 15). MOG (150μg) was co-administered on days 3 and 11. The administration of 1,25(OH) 2D3 or MOG determined significant reduction in EAE incidence and in clinical scores. When MOG was associated with 1,25(OH) 2D3 the animals did not develop EAE. Spleen and central nervous system (CNS) cell cultures from this group produced less IL-6 and IL-17 upon stimulation with MOG in comparison to the EAE control group. In addition, this treatment inhibited dendritic cells maturation in the spleen and reduced inflammatory infiltration in the CNS. The association of MOG with 1,25(OH) 2D3 was able to control EAE development.

Introduction

Multiple sclerosis (MS) is a demyelinating disease mediated by an immune response against self-antigens from the central nervous system (CNS). This immune response triggers an initial inflammation in brain and spinal cord that is then followed by demyelination, axonal damage and scar formation [1]. Experimental autoimmune encephalomyelitis (EAE) is an induced inflammatory demyelinating disease of the CNS that is largely employed as a model to study MS [2,3]. The pathogenic immune response observed in MS and EAE CNS is mainly mediated by Th1 and Th17 cells whose main signature cytokines are IFN-γ and IL-17, respectively [46]. Current MS treatment consists in general suppression of the immune response which can increase susceptibility to infections. Therefore, an antigen-specific tolerogenic strategy is highly desirable to treat this disease. Administration of antigens derived from myelin by intravenous, transdermal and oral routes protected animals from EAE development [79]. The transdermal application of myelin peptides in MS patients was also effective, being able to reduce magnetic resonance imaging and clinical parameters of disease activity [10]. Many of these strategies induced tolerogenic dendritic cells (DCs) whose tolerogenic properties can also be achieved by the use of pharmacological agents as corticosteroids, rapamycin, mycophenolate mofetil, prostaglandin E2 and vitamin D [11,12].

The active form of vitamin D, 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3), is mainly obtained after sunlight exposure. 1,25(OH)2D3 has an important role in bone metabolism, in calcium and phosphorus homeostasis and also plays a regulatory role in the immune system [13]. Its activity is mediated by the vitamin D receptor which is expressed in many cell types including immune cells such as monocytes, macrophages, DCs and activated B and T lymphocytes [14]. In the innate immunity, it enhances the antimicrobial properties of monocytes and macrophages [13]. The regulatory role of 1,25(OH)2D3 in the adaptive immunity has been mainly attributed to its effects on DCs. In vitro exposure to 1,25(OH)2D3 inhibits DC maturation by reducing expression of markers such as CD1a, MHC II, CD40, CD80 and CD86 [15]. In addition, 1,25(OH)2D3 modulates cytokine production by DCs, inhibiting, for example, the production of IL-12 and IL-23 that induce Th1 and Th17 differentiation, respectively. Classically, it has been described that 1,25(OH)2D3 also increases the secretion of IL-10, a cytokine with very well established anti-inflammatory properties [16]. The effects of 1,25(OH)2D3 on T cells include inhibition of their proliferation, of IL-2 and IFN-γ production [17] and also of Th17 cells differentiation [18]. Besides, in vitro and in vivo studies show that 1,25(OH)2D3 usually induces Foxp3+ regulatory T cells [19]. The beneficial role of 1,25(OH)2D3 in experimental models of autoimmune diseases, as experimental autoimmune uveitis, EAE and diabetes have been clearly demonstrated [2024]. A great deal of attention has been dedicated to reveal the detailed mechanism of 1,25(OH)2D3 over the immune system. Chang et al., 2010 [22], for example, showed that the beneficial effect of this substance in EAE is mediated throughout inhibition of differentiation and also migration of Th17 cells to the CNS. The contribution of tolerogenic DCs induced by 1,25(OH)2D3 to control autoimmunity was clearly showed by Farias et al., 2013 [23]. These authors demonstrated that both, in vivo administration of 1,25(OH)2D3 and transfer of 1,25(OH)2D3-induced IDO(+) immature DCs, significantly reduced EAE severity.

The aim of this study was to evaluate the efficacy of a precocious treatment with MOG/1,25(OH)2D3 association on EAE development.

Materials and Methods

Ethics statement

All animal studies and procedures were approved by local Ethics Committee for animal experimentation (CEEA), Universidade Estadual Paulista (protocol number 314). The animals were anesthetized by intraperitoneal injection of a solution containing ketamine and xylasin.

Animals

Female C57BL/6 mice with 6–9 weeks old were purchased from University of São Paulo (USP, Ribeirão Preto, SP, Brazil). The animals received sterilized food and water ad libitum.

EAE induction

MOG35–55 peptide (MEVGWYRSPFSRVVHLYRNGK) was synthesized by Genemed Synthesis Inc. (San Antonio, Texas, USA). Mice were immunized subcutaneously with 150 μg of MOG35-55 peptide emulsified in Complete Freund’s Adjuvant containing 4mg/mL of Mycobacterium tuberculosis. Mice also received two intraperitoneal doses, 0 and 48 h after immunization, of 250 ng of Bordetella pertussis toxin (Sigma Aldrich, St. Louis, MO, USA). Clinical assessment of EAE was daily performed according to the following criteria: 0—no disease, 1—limp tail, 2—weak hind legs, 3—partially paralyzed hind legs, 4—complete hind leg paralysis, and 5—complete paralysis/death.

Treatment with 1,25(OH)2D3, MOG or 1,25(OH)2D3 associated with MOG

One day after EAE induction, mice were treated, by intraperitoneal route, with 0,1 μg of 1α,25-dihydroxyvitamin D3 (1,25(OH)2D3) from Sigma Aldrich, every other day during 15 days (on days 1, 3, 5, 7, 9, 11, 13 and 15); MOG (150μg) was co-administered, also by this route, on days 3 and 11.

CNS-mononuclear cells isolation

Ten and eighteen days after EAE induction, mice were anesthetized with ketamine/xylazine and perfused with 10 mL of saline solution. Mononuclear cells from CNS were isolated as previously described [25]. Briefly, brain and spinal cord were entirely collected, macerated and digested with collagenase D (2.5mg/mL, Roche Diagnostics, Indianapolis, IN, USA) in 4mL of RPMI (Sigma Aldrich, St. Louis, MO, USA) at 37°C for 45 min. Suspensions were then washed in RPMI and centrifuged at 450 ×g for 15min at 4°C. Cells were resuspended in percoll (GE Healthcare, Uppsala, Sweden) 37% and laid over percoll 70%. The material was centrifuged at 950 ×g for 20 min with centrifuge breaks turned off and the mononuclear cell ring was collected, washed in RPMI, and centrifuged at 450 ×g for 10 min. Cells were then resuspended in complete RPMI medium, counted, and analyzed. To get enough cells to perform the experiments we used pools from 3 animals.

Cell culture conditions and cytokine quantification

Eighteen days after EAE induction, spleen and CNS-isolated cells, whose viability was over 95%, were adjusted to 5 x 106 cells/mL and 2 x 105 cells/mL, respectively and cultured in RPMI medium containing 10% of fetal calf serum and 2 mM of glutamine. Spleen and CNS cultures were stimulated with 20 μg/mL and 50 μg/mL of MOG, respectively. Cytokine levels were evaluated 48h later by enzyme-linked immunosorbent assay (ELISA) using IFN-γ and IL-10 BD OptEIA Sets (BD Biosciences, San Jose, CA, USA) and IL-6 and IL-17 Duosets (R&D Systems, Minneapolis, MN, USA). The assays were performed according to the manufacturer’s instructions.

Flow cytometry

Spleen and lymph nodes (popliteal + inguinal) cells were collected and the red blood cells were lysed with buffer containing NH4Cl. For regulatory T cells analysis, spleen, lymph nodes and CNS-isolated cells were incubated with 0.5 μg of FITC labeled anti-mouse CD4 (clone GK1.5) and 0.25 μg of APC labeled anti-mouse CD25 (clone PC61.5) for 20 min at room temperature. Intracellular Foxp3 transcription factor was detected using Foxp3 PE Staining Set (eBiosciences, San Diego, CA, USA) according to manufacturer's instructions. For DCs analysis, splenocytes were incubated with 0.25 μg of FITC labeled anti-mouse CD11c (clone N418), 0.03 μg of APC labeled anti-mouse MHCII (clone M5/114.15.2) and 0.125 μg of PE labeled anti-mouse CD86 (clone GL1) for 30 min at 4°C. The cells were then washed, resuspended in flow cytometry buffer and fixed in paraformaldehyde 1%. The cells were analyzed by flow cytometry using the FACSCanto II (BD Biosciences, San Jose, CA, USA). Regulatory T cells and DCs analyses were performed by FlowJo software (TreeStar, Ashland, OR, EUA) and Infinicyt software (Cytognos, Salamanca, Spain), respectively. These experiments were performed 18 days after EAE induction except Foxp3+ regulatory T cells (Treg) evaluation in the spleen and lymph nodes that was also assessed at the preclinical phase.

Evaluation of inflammatory infiltrates in the CNS

The histological analysis was performed during EAE acute phase, that is, 18 days after disease induction. After euthanasia, lumbar spinal cord samples were removed and fixed in 10% formaldehyde. Paraffin slides with 5 μm were stained with hematoxylin and eosin (H&E) and analyzed with a Nikon microscope.

Calcium and phosphorus levels

Calcium and phosphorus serum levels were analyzed using Cálcio Arsenazo III commercial kit (Bioclin, Minas Gerais, Brazil) and Quimifos-Fósforo commercial kit (Ebram, São Paulo, Brazil). The assays were performed according to the manufacturer’s instruction.

Statistical analysis

Results were expressed as mean ± standard deviation or with median and interquartile (25–75%) ranges. Comparisons between groups were made by t-test or one way ANOVA followed by Tukey’s test for parametric variables and by Kruskal-Wallis followed by Dunn’s test for non-parametric variables. Chi-square was performed for EAE incidence. Statistical analysis was accomplished with SigmaPlot software version 12.0 (Systat Software Inc., San Jose, CA, USA) and p<0.05 was considered significant. See S1 File for full dataset.

Results

MOG/1,25(OH)2D3 association inhibits EAE development

A 100% disease incidence was observed in the positive control group. These animals also presented very typical signs of disease characterized by elevated clinical score and accentuated body weight loss. On the other hand, groups treated with MOG, 1,25(OH)2D3 or 1,25(OH)2D3+MOG showed a decreased incidence and also a much less severe clinical disease. Distinctly from MOG and 1,25(OH)2D3 treated groups that showed a residual disease, the group treated with 1,25(OH)2D3+MOG was completely protected and did not show any sign of paralysis. The kinetics of clinical scores and the maximum clinical score in all experimental groups is illustrated in Fig 1A and 1C, respectively. Maximum clinical score determination confirmed that MOG and 1,25(OH)2D3 alone similarly reduced disease severity whereas their association completely avoided any clinical disease manifestation. EAE incidence is depicted in Table 1 and clearly indicates that treatment with 1,25(OH)2D3 or MOG were also able to reduce disease incidence. However, their combination reduced disease incidence to zero. As expected, mice from the positive control group lost body weight during the acute disease phase. Mice treated with MOG did not lost weight whereas the ones that received 1,25(OH)2D3 alone or combined with MOG, lost weight even before paralysis manifestations (Fig 1B).

thumbnail
Table 1. EAE incidence in mice treated with 1,25(OH)2D3+MOG.

https://doi.org/10.1371/journal.pone.0125836.t001

thumbnail
Fig 1. Effect of treatment with 1,25(OH)2D3+MOG association on EAE development.

Kinetics of clinical scores (A), maximum clinical score (B) and weight variation (C). Comparisons between groups were made by one way ANOVA followed by Tukey’s test for parametric variables (A and C) and by Kruskal-Wallis followed by Dunn’s test for non-parametric variables (B). Results were expressed as mean or medians (25–75% ranges) of 12 animals per group. * p<0.05 compared with EAE+MOG, EAE+vitD and EAE/vitD+MOG groups. Data are representative of two independent experiments.

https://doi.org/10.1371/journal.pone.0125836.g001

Quantification of DCs in the spleen

As depicted in Fig 2A, there was a significant increase in the absolute number of mature DCs in the EAE group compared with normal control. Contrarily, there was a significant decrease in the number of mature DCs in the groups treated with 1,25(OH)2D3 or 1,25(OH)2D3+MOG compared with EAE and EAE/MOG groups. The same profile was observed in the percentage of these cells among the groups (Fig 2B).

thumbnail
Fig 2. Quantification of splenic dendritic cells in C57BL/6 mice treated with 1,25(OH)2D3+MOG.

Absolute number (A) and percentage (B) of CD11c+MHCII+CD86+ cells in 500,000 acquired events. Comparisons between groups were made by one way ANOVA followed by Tukey’s test. Results are expressed as mean ± SD of 5 animals per group. * p<0.05 compared with EAE and EAE+MOG group. Data are representative of two independent experiments.

https://doi.org/10.1371/journal.pone.0125836.g002

Cytokine evaluation in the spleen and CNS

The recall response was evaluated with cell cultures from the spleen and the CNS stimulated with MOG. Similar amounts of IFN-γ were produced by all experimental groups (Fig 3A). IL-17 production was statistically down-modulated in the EAE/1,25(OH)2D3+MOG compared with the EAE group (Fig 3B). IL-6 was down-modulated in this and also in the EAE/1,25(OH)2D3 group (Fig 3C). The levels of IL-10 were similar in EAE, EAE/1,25(OH)2D3 and EAE/1,25(OH)2D3+MOG groups. However, mice treated only with MOG produced higher levels of IL-10 compared with the three other experimental groups (Fig 3D).

thumbnail
Fig 3. Cytokine production by spleen and CNS cell cultures.

IFN-γ (A and E), IL-17 (B and F), IL-6 (C and G) and IL-10 (D and H) levels were measured in spleen and CNS cell cultures stimulated with MOG. Comparisons between groups were made by one way ANOVA followed by Tukey’s test for parametric variables (D, F and G) and by Kruskal-Wallis followed by Dunn’s test for non-parametric variables (A, B, C, E and H). Data were presented by mean ± SE or medians (25–75% ranges) of 9 animals per group in spleen cultures or 4 pools (each pool contains cells from brain and spinal cord of 3 mice) per group in CNS cultures. * p<0.05. Data are representative of two independent experiments.

https://doi.org/10.1371/journal.pone.0125836.g003

Lower levels of IFN-γ, IL-17, IL-6 and IL-10 were produced by cells eluted from the CNS of treated mice (Fig 3E, 3F, 3G and 3H). However, treatment with 1,25(OH)2D3+MOG determined the most striking modulation, significantly reducing the production of IFN-γ, IL-17 and IL-6 as shown in Fig 3E, 3F and 3G, respectively.

Quantification of regulatory T cells in peripheral lymphoid organs

The frequency of Foxp3+ Treg cells was evaluated at the pre-clinical and clinical phases, that is, 10 and 18 days after EAE induction respectively. No differences were observed in the spleen (Fig 4A). However, the frequency of CD4+CD25+Foxp3+ T cells was significantly reduced in the lymph nodes of mice treated with 1,25(OH)2D3+MOG compared with the EAE untreated group (Fig 4B).

thumbnail
Fig 4. Quantification of regulatory T cells in peripheral lymphoid organs from C57BL/6 mice treated with 1,25(OH)2D3+MOG.

Percentage of CD4+CD25+Foxp3+ T cells in total CD4+ T cell. Spleen (A) and lymph nodes (B) were collected 10 and 18 days after EAE induction. Analyses were performed in 100,000 acquired events. Comparisons between groups were made by t-test followed by Tukey’s test for parametric variables. Results were expressed as mean of 6–9 animals per group. Data are representative of two independent experiments.

https://doi.org/10.1371/journal.pone.0125836.g004

Inflammatory process in the CNS

As expected, the EAE control group showed typical inflammatory foci, with a clear predominance of mononuclear cells in the lumbar spinal cord (Fig 5B). A similar picture was observed in sections from the EAE+MOG group (Fig 5C). Otherwise, samples from EAE+1,25(OH)2D3 (Fig 5D) and EAE/1,25(OH)2D3+MOG (Fig 5E) groups showed no inflammation. The absence of inflammation in normal mice is illustrated in Fig 5A. Similar results were observed in brain analysis (not shown). Inflammation was also assessed by CD4+ and CD4+CD25+Foxp3+ T cells quantification in cells eluted from the CNS. The total amount of CD4+ T cells was similar in EAE and EAE/MOG groups. However, a striking reduction was detected in the EAE/1,25(OH)2D3 and EAE/1,25(OH)2D3+MOG groups. A similar profile was found when CD4+CD25+Foxp3+ T cells were quantified. In both evaluations, a statistical comparison between EAE/1,25(OH)2D3 and EAE/1,25(OH)2D3+MOG did not indicate statistical significance even though down-modulation by the associated treatment was apparently more efficient (Fig 5F).

thumbnail
Fig 5. Infiltrating cells in the CNS after treatment of 1,25(OH)2D3+MOG.

Inflammatory process in lumbar spinal cord sections stained with H&E in control (A), EAE (B), EAE+MOG (C), EAE+vitD (D) and EAE/vitD+MOG (E) groups and absolute number of CD4+ T cells and CD4+CD25+Foxp3+ T cells in CNS-mononuclear cells (F). Regulatory T cells were evaluated in the total CD4+ T cell population and analyses were performed in 20,000 acquired events. Comparisons between groups were made by one way ANOVA followed by Tukey’s test. Data were presented by mean ± SE of 4 pools (each pool contains cells from brain and spinal cord of 3 mice) per group in CNS cultures cells. * p<0.05 in compared with EAE and EAE+MOG group. Scale bar = 100μm. Micrographs are representative of 5 animals per group. Data are representative of two independent experiments.

https://doi.org/10.1371/journal.pone.0125836.g005

Serum levels of calcium and phosphorus

1,25(OH)2D3 treated groups presented significantly higher serum levels of calcium (Fig 6A) but not of phosphorus (Fig 6B) in the moment of euthanasia.

thumbnail
Fig 6. Effect of 1,25(OH)2D3 (alone or combined with MOG) treatment in calcium and phosphorus levels.

Serum calcium (A) and phosphorus (B) levels were quantified 18 days after EAE induction. Comparisons between groups were made by one way ANOVA followed by Tukey’s test. Data were presented by mean ± SE of 4–6 animals per group. * p<0.05 compared with Control, EAE, and EAE+MOG group. Data are representative of two independent experiments.

https://doi.org/10.1371/journal.pone.0125836.g006

Discussion

Considering the immunomodulatory properties of active vitamin D and the epidemiological observations that low serum levels of this vitamin are probably associated with increased MS risk [26], we hypothesized that the association of 1,25(OH)2D3 with MOG could have a protective effect on EAE. The success of MOG+1,25(OH)2D3 treatment was clearly demonstrated by clinical parameters, that is, disease incidence and maximum clinical score were zero in these animals. However, MOG and 1,25(OH)2D3 by themselves were also partially effective, being 1,25(OH)2D3 more efficient than MOG. The observed efficacy of 1,25(OH)2D3 administration (75% reduction in incidence and a significant decrease in the maximum clinical score) was expected and supported by recent work with similar experimental models [23]. A variety of strategies employing MOG has also been devised, tested and proved promising. MOG can be used alone by nasal administration [27] and is also effective by delivery as a DNA vaccine [28] or as part of fusion proteins [29]. The strong immunomodulatory activity of this association was clearly revealed by the analysis of cytokine production by spleen cells and also by the quantification of DCs in this organ. A significant reduction was detected in IL-6 and IL-17 levels even though IFN-γ and IL-10 levels were not affected. Reduced IL-17 production is particularly relevant because it is considered a highly encephalitogenic cytokine in EAE and MS patients [30,31]. In the group that received this combined treatment there was a clear reduction in the amount of mature DCs in the spleen. This effect was attributed solely to 1,25(OH)2D3 because MOG alone increased, even though not substantially, the amount of these cells. Much of the knowledge in this area relates to the in vitro effect of 1,25(OH)2D3 on DCs. In the presence of 1,25(OH)2D3 these cells differentiate into a phenotype that resembles semi-mature DCs that are poor IL-12 producers and express low amounts of co-stimulatory and MHCII molecules. These 1,25(OH)2D3-modulated DCs usually induce tolerance characterized by a blockage in Th1 and Th17 differentiation together with recruiting and differentiating regulatory T cells [32,33]. The adoptive transfer of 1,25(OH)2D3 modified DCs has been considered the most promising immunotherapeutic approach for autoimmune diseases. Our results strongly suggest that a similar immunomodulatory effect can be achieved in vivo by the combined treatment with 1,25(OH)2D3 and the specific antigen.

Unexpectedly, no increase was found in the amount of CD4+CD25+Foxp3+ T cells in spleen and lymph nodes. We even found, at the preclinical stage, a significant decrease in their percentage in the draining lymph nodes. Our interpretation is that protection in this case is independent of Treg induction relying mostly on the concomitant presence of immature DCs and the self-antigen. In this scenario, MOG specific T cells could be deleted by the contact with immature DCs as has been demonstrated in various experimental scenarios [15,34,35]. Even though Treg induction is being predominantly linked to 1,25(OH)2D3 immunoregulatory effects [36, 37] a few reports also show that active vitamin D is able to directly inhibit the expression of Foxp3 [22].

IL-10 has been considered essential to mediate the protection triggered by active vitamin D in EAE [38]. However, our results did not show increased production of this cytokine. A possible explanation for these apparently contradictory findings could be the 1,25(OH)2D3 administration route and/or its association with the neuroantigen. Spach et al (2006) [38], used only 1,25(OH)2D3 by oral route whereas we employed an association of active vitamin D + MOG by intraperitoneal route.

The higher efficacy of 1,25(OH)2D3+MOG treatment and, to a lesser extension, of 1,25(OH)2D3 alone, was confirmed when sections from the CNS, local cytokine production, and the characteristics of the CD4+ T cells eluted from this tissue were analyzed. Distinctly from EAE and EAE/MOG groups that presented clear cell infiltrates in the CNS, no inflammation was found in the CNS of 1,25(OH)2D3+MOG treated animals. However, it was possible to elute cells from the CNS of all experimental groups. One of the major differences was the amount of cells; EAE and EAE/MOG groups had a significantly higher number of CD4+ T cells than EAE/1,25(OH)2D3 and EAE/1,25(OH)2D3+MOG groups. In addition, the total number of CD4+CD25+Foxp3+ T cells was significantly lower in the two groups that received 1,25(OH)2D3.

Altogether, these results strongly suggest that 1,25(OH)2D3 is working as a tolerogenic adjuvant. The potential usefulness of other immunosuppressants as tolerogenic adjuvants was initially suggested by Kang et al. 2008 [39]. These authors demonstrated that BALB/c mice, with a pre-established delayed type hypersensitivity reaction to OVA, could be desensitized by treatment with an OVA-peptide associated with dexamethasone. The tolerogenic mechanism included a blockage in DCs maturation and also an expansion of CD4+CD25+Foxp3+ regulatory T cells. This drug was also tolerogenic and prophylactic when associated with an insulin peptide in the NOD diabetes model. The efficacy of this concept was also tested in EAE. Kang et al. 2009 [40], used FK506 as a tolerogenic adjuvant combined with a DNA vaccine encoding MOG to prevent EAE development.

Based on the above results we postulate that the combined therapy with specific antigens associated with 1,25(OH)2D3 is potentially effective to block the development of autoimmune diseases. Our data, that is a new contribution to this field, warrant additional studies to support the potential indication of 1,25(OH)2D3 + MOG in the clinical setting.

Supporting Information

S1 File. Data used in the analyses of “Treatment with vitamin D/MOG association suppresses experimental autoimmune encephalomyelitis”.

https://doi.org/10.1371/journal.pone.0125836.s001

(XLSX)

Author Contributions

Conceived and designed the experiments: FCM AS. Performed the experiments: FCM SFGZP TGDF LLWI LANM TFCFS. Analyzed the data: FCM AS. Contributed reagents/materials/analysis tools: CM MRVI. Wrote the paper: FCM AS.

References

  1. 1. Loleit V, Biberacher V, Hemmer B. Current and future therapies targeting the immune system in multiple sclerosis. Curr Pharm Biotechnol. 2014;15: 276–296. pmid:24938888
  2. 2. Fletcher JM, Lalor SJ, Sweeney CM, Tubridy N, Mills KH. T cells in multiple sclerosis and experimental autoimmune encephalomyelitis. Clin Exp Immunol. 2010;162: 1–11. pmid:20682002
  3. 3. Mitchell KM, Dotson AL, Cool KM, Chakrabarty A, Benedict SH, LeVine SM. Deferiprone, an orally deliverable iron chelator, ameliorates experimental autoimmune encephalomyelitis. Mult Scler. 2007;13: 1118–1126. pmid:17967839
  4. 4. Domingues HS, Mues M, Lassmann H, Wekerle H, Krishnamoorthy G. Functional and pathogenic differences of Th1 and Th17 cells in experimental autoimmune encephalomyelitis. PLoS One. 2010;5: e15531. pmid:21209700
  5. 5. Chen SJ, Wang YL, Fan HC, Lo WT, Wang CC, Sytwu HK. Current status of the immunomodulation and immunomediated therapeutic strategies for multiple sclerosis. Clin Dev Immunol. 2012;2012: 970789. pmid:22203863
  6. 6. Zorzella-Pezavento SFG, Chiuso-Minicucci F, França TGD, Ishikawa LLW, da Rosa LC, Marques C, et al. Persistent inflammation in the CNS during chronic EAE despite local absence of IL-17 production. Mediators Inflamm. 2013;2013: 519627. pmid:23970813
  7. 7. Higgins PJ, Weiner HL. Suppression of experimental autoimmune encephalomyelitis by oral administration of myelin basic protein and its fragments. J Immunol. 1988;140: 440–445. pmid:2447178
  8. 8. Hilliard B, Ventura ES, Rostami A. Effect of timing of intravenous administration of myelin basic protein on the induction of tolerance in experimental allergic encephalomyelitis. Mult Scler. 1999;5: 2–9. pmid:10096096
  9. 9. Byone M, Evans JT, Viret C, Janeway CA Jr. Epicutaneous immunization with autoantigenic peptides induces T suppressor cells that prevent experimental allergic encephalomyelitis. Immunity. 2003;19: 317–328. pmid:14499108
  10. 10. Walczak A, Siger M, Ciach A, Szczepanik M, Selmaj K. Transdermal application of myelin peptides in multiple sclerosis treatment. JAMA Neurol. 2013;70: 1105–1109 pmid:23817921
  11. 11. Hackstein H, Thomson AW. Dendritic cells: emerging pharmacological targets of immunosuppressive drugs. Nat Rev Immunol. 2014;4: 24–34.
  12. 12. Adorini L, Giarratana N, Penna G. Pharmacological induction of tolerogenic dendritic cells and regulatory T cells. Semin Immunol. 2004;6: 127–134.
  13. 13. Hart PH, Gorman S, Finlay-Jones JJ. Modulation of the immune system by UV radiation: more than just the effects of vitamin D? Nat Rev Immunol. 2011;11: 584–596 pmid:21852793
  14. 14. Carlberg C, Polly P. Gene regulation by vitamin D3. Crit Rev Eukaryot Gene Expr. 1998;8: 19–42. pmid:9673449
  15. 15. Penna G, Adorini L. 1 Alpha,25-dihydroxyvitamin D3 inhibits differentiation, maturation, activation, and survival of dendritic cells leading to impaired alloreactive T cell activation. J Immunol. 2000;164: 2405–2411. pmid:10679076
  16. 16. Mathieu C. Vitamin D and the Immune system: getting it right. IBMS BoneKEy. 2011;8: 178–186.
  17. 17. Dimeloe S, Nanzer A, Ryanna K, Hawrylowicz C. Regulatory T cells, inflammation and the allergic response-The role of glucocorticoids and Vitamin D. J Steroid Biochem Mol Biol. 2010;120: 86–95. pmid:20227496
  18. 18. Tian Y, Wang C, Ye Z, Xiao X, Kijlstra A, Yang P. Effect of 1,25-dihydroxyvitamin D3 on Th17 and Th1 response in patients with Behçet's disease. Invest Ophthalmol Vis Sci. 2012;53: 6434–6441. pmid:22918640
  19. 19. Chambers ES, Hawrylowicz CM. The impact of vitamin D on regulatory T cells. Curr Allergy Asthma Rep. 2011;11: 29–36. pmid:21104171
  20. 20. Gregori S, Giarratana N, Smiroldo S, Uskokovic M, Adorini L. A 1alpha,25-dihydroxyvitamin D(3) analog enhances regulatory T-cells and arrests autoimmune diabetes in NOD mice. Diabetes. 2002;51: 1367–1374. pmid:11978632
  21. 21. Tang J, Zhou R, Luger D, Zhu W, Silver PB, Grajewski RS, et al. Calcitriol Suppresses Antiretinal Autoimmunity through Inhibitory Effects on the Th17 Effector Response. J Immunol. 2009;182: 4624–4632. pmid:19342637
  22. 22. Chang JH, Cha HR, Lee DS, Seo KY, Kweon MN. 1,25-Dihydroxyvitamin D3 inhibits the differentiation and migration of T(H)17 cells to protect against experimental autoimmune encephalomyelitis. PLoS One. 2010;5: e12925. pmid:20886077
  23. 23. Farias AS, Spagnol GS, Bordeaux-Rego P, Oliveira CO, Fontana AG, de Paula RF, et al. Vitamin D3 induces IDO+ tolerogenic DCs and enhances Treg, reducing the severity of EAE. CNS Neurosci Ther. 2013;19: 269–277. pmid:23521914
  24. 24. Li B, Baylink DJ, Deb C, Zannetti C, Rajaallah F, Xing W, et al. 1,25-Dihydroxyvitamin D3 suppresses TLR8 expression and TLR8-mediated inflammatory responses in monocytes in vitro and experimental autoimmune encephalomyelitis in vivo. PLoS One. 2013;8: e58808. pmid:23516559
  25. 25. Zorzella-Pezavento SFG, Chiuso-Minicucci F, França TGD, Ishikawa LLW, da Rosa LC, Colavite PM, et al. Downmodulation of peripheral MOG-specific immunity by pVAXhsp65 treatment during EAE does not reach the CNS. J Neuroimmunol. 2014;268: 35–42. pmid:24439542
  26. 26. Martinelli V, Dalla Costa G, Colombo B, Dalla Libera D, Rubinacci A, Filippi M, et al. Vitamin D levels and risk of multiple sclerosis in patients with clinically isolated syndromes. Mult Scler. 2014;20: 147–155. pmid:23836877
  27. 27. Levy Barazany H, Barazany D, Puckett L, Blanga-Kanfi S, Borenstein-Auerbach N, Yang K, et al. Brain MRI of nasal MOG therapeutic effect in relapsing-progressive EAE. Exp Neurol. 2014;255: 63–70. pmid:24552689
  28. 28. Fissolo N, Costa C, Nurtdinov RN, Bustamante MF, Llombart V, Mansilla MJ, et al. Treatment with MOG-DNA vaccines induces CD4+CD25+FoxP3+ regulatory T cells and up-regulates genes with neuroprotective functions in experimental autoimmune encephalomyelitis. J Neuroinflammation. 2012;9: 139. pmid:22727044
  29. 29. Divekar RD, Haymaker CL, Cascio JA, Guloglu BF, Ellis JS, Tartar DM, et al. T cell dynamics during induction of tolerance and suppression of experimental allergic encephalomyelitis. J Immunol. 2011;187: 3979–3986. pmid:21911603
  30. 30. Ishizu T, Osoegawa M, Mei FJ, Kikuchi H, Tanaka M, Takakura Y, et al. Intrathecal activation of the IL-17/IL-8 axis in opticospinal multiple sclerosis. Brain. 2005;128: 988–1002. pmid:15743872
  31. 31. Petković F, Blaževski J, Momčilović M, Timotijević G, Zocca MB, Mijatović S, et al. Saquinavir-NO inhibits S6 kinase activity, impairs secretion of the encephalytogenic cytokines interleukin-17 and interferon-gamma and ameliorates experimental autoimmune encephalomyelitis. J Neuroimmunol. 2013;259: 55–65. pmid:23602714
  32. 32. Pedersen AW, Claesson MH, Zocca MB. Dendritic cells modified by vitamin D: future immunotherapy for autoimmune diseases. Vitam Horm. 2011;86: 63–82. pmid:21419267
  33. 33. Yang CY, Leung PS, Adamopoulos IE, Gershwin ME. The implication of vitamin D and autoimmunity: a comprehensive review. Clin Rev Allergy Immunol. 2013;45: 217–226. pmid:23359064
  34. 34. Nikolic T, Roep BO. Regulatory multitasking of tolerogenic dendritic cells—lessons taken from vitamin d3-treated tolerogenic dendritic cells. Front Immunol. 2013;14: 4–113.
  35. 35. van Halteren AG, Tysma OM, van Etten E, Mathieu C, Roep BO. 1alpha,25-dihydroxyvitamin D3 or analogue treated dendritic cells modulate human autoreactive T cells via the selective induction of apoptosis. J Autoimmun. 2004;23: 233–239. pmid:15501394
  36. 36. Ishikawa LL, Shoenfeld Y, Sartori A. Immunomodulation in human and experimental arthritis: including vitamin D, helminths and heat-shock proteins. Lupus. 2014;23: 577–587. pmid:24763541
  37. 37. Gordon JR, Ma Y, Churchman L, Gordon SA, Dawicki W. Regulatory dendritic cells for immunotherapy in immunologic diseases. Front Immunol. 2014;31: 5–7.
  38. 38. Spach KM, Nashold FE, Dittel BN, Hayes CE. IL-10 signaling is essential for 1,25-dihydroxyvitamin D3-mediated inhibition of experimental autoimmune encephalomyelitis. J Immunol. 2006;177: 6030–6037. pmid:17056528
  39. 39. Kang Y, Xu L, Wang B, Chen A, Zheng G. Cutting Edge: Immunosuppressant as Adjuvant for Tolerogenic Immunization. J Immunol. 2008;180: 5172–5176. pmid:18390698
  40. 40. Kang Y, Zhao J, Liu Y, Chen A, Zheng G, Yu Y, et al. FK506 as an adjuvant of tolerogenic DNA vaccination for the prevention of experimental autoimmune encephalomyelitis. J Gene Med. 2009;11:1064–1070. pmid:19688809