Targeting the shift from M1 to M2 macrophages in experimental autoimmune encephalomyelitis mice treated with fasudil

Chunyun Liu, Yanhua Li, Jiezhong Yu, Ling Feng, Shaowei Hou, Yueting Liu, Mingfang Guo, Yong Xie, Jian Meng, Haifei Zhang, Baoguo Xiao, Cungen Ma, Chunyun Liu, Yanhua Li, Jiezhong Yu, Ling Feng, Shaowei Hou, Yueting Liu, Mingfang Guo, Yong Xie, Jian Meng, Haifei Zhang, Baoguo Xiao, Cungen Ma

Abstract

We observed the therapeutic effect of Fasudil and explored its mechanisms in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). Fasudil, a selective Rho kinase (ROCK) inhibitor, was injected intraperitoneally at 40 mg/kg/d in early and late stages of EAE induction. Fasudil ameliorated the clinical severity of EAE at different stages, and decreased the expression of ROCK-II in spleen, accompanied by an improvement in demyelination and inhibition of inflammatory cells. Fasudil mainly inhibited CD4(+)IL-17(+) T cells in early treatment, but also elevated CD4(+)IL-10(+) regulatory T cells and IL-10 production in late treatment. The treatment of Fasudil shifted inflammatory M1 to anti-inflammatory M2 macrophages in both early and late treatment, being shown by inhibiting CD16/32, iNOS, IL-12, TLR4 and CD40 and increasing CD206, Arg-1, IL-10 and CD14 in spleen. By using Western blot and immunohistochemistry, iNOS and Arg-1, as two most specific markers for M1 and M2, was inhibited or induced in splenic macrophages and spinal cords of EAE mice treated with Fasudil. In vitro experiments also indicate that Fasudil shifts M1 to M2 phenotype, which does not require the participation or auxiliary of other cells. The polarization of M2 macrophages was associated with the decrease of inflammatory cytokine IL-1β, TNF-α and MCP-1. These results demonstrate that Fasudil has therapeutic potential in EAE possibly through inducing the polarization of M2 macrophages and inhibiting inflammatory responses.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. Fasudil delays onset, and ameliorates…
Figure 1. Fasudil delays onset, and ameliorates severity of EAE.
Chronic EAE was induced in C57BL/6 mice with MOG35–55. Fasudil was injected (i.p.) at 40 mg/kg/d every other day on day 3 p.i. (Fasudil early treatment, n = 13) or on day 11 p.i. (Fasudil late treatment, n = 13). The horizontal line is treatment period of Fasudil. The injection of normal saline was set up as control (EAE control, n = 13) in a similar manner. a) Mean clinical score, b) Mean body weight. The comparison in each time point was separately analyzed by Mann Whitney U test after nonparametric Kruskal-Wallis test. *p<0.05; **p<0.01.
Figure 2. Fasudil inhibits inflammation and improves…
Figure 2. Fasudil inhibits inflammation and improves myelination in spinal cords.
Chronic EAE was induced in C57BL/6 mice with MOG35–55. Fasudil was injected (i.p.) at 40 mg/kg/d every other day on day 3 p.i. (Fasudil early treatment, n = 9) or on day 11 p.i. (Fasudil late treatment, n = 9). The injection of normal saline was set up as control (EAE control, n = 12) in a similar manner. On day 21 p.i., spinal cords were used for Luxol fast blue and H&E staining. Left) Demyelination stained with Luxol Fast Blue. Total white matter in Luxol Fast Blue was manually outlined, and pixel area (%) of demyelination in total white matter was calculated by Image-Pro Plus software. Right) Inflammation stained with H&E. Color photo in H&E stain was automatically converted to black and white, and number of inflammatory foci (>20 mononuclear cells/focus) in whole spinal cords were calculated by Image-Pro Plus software. Quantitative results are analyzed for 6 mice in each group and are representative of 3 experiments with similar results. *p<0.05, **p<0.01.
Figure 3. Fasudil inhibits the expression of…
Figure 3. Fasudil inhibits the expression of ROCK-II in spleen.
Chronic EAE was induced in C57BL/6 mice with MOG35–55. Fasudil was injected (i.p.) at 40 mg/kg/d every other day on day 3 and 11 p.i. On day 21 p.i., spleens were collected, and the expression of ROCK II was measured by immunohistochemistry. Spleen from healthy mice was measured as control. Up) Immunohistochemistry. Green = ROCK-II; Blue = Hoechst nuclear staining. As a negative control, additional section was treated similarly, but the primary antibodies were omitted. Quantitative results are analyzed by Western blot. Down) Western blot. Left down = representative bands from one of three experiments with similar results; Right down = quantitative results are analyzed for 5 mice in each group and expressed as fold change relative to GAPDH as the loading control *p<0.05.
Figure 4. Fasudil induces the regulatory T…
Figure 4. Fasudil induces the regulatory T cells and inhibits IL-17 T cells.
Chronic EAE was induced in C57BL/6 mice with MOG35–55. Fasudil was injected (i.p.) at 40 mg/kg/d every other day on days 3 and 11 p.i. Mice were sacrificed and splenic MNCs were prepared on days 9 (a) and 21 (b) p.i. Cells were stained with T cell marker CD4 and CD25, IL-10 and IL-17, and subsets of T cells were analyzed using flow cytometry. Scatterplot, representative dot plots from one of three experiments with similar results. Histogram, quantitative results are analyzed for 5 mice in each group. Results are expressed as the percentage of double positive cells in four-quadrant diagram or within rectangle. *p<0.05, **p<0.01, ***p<0.001.
Figure 5. Fasudil shifts M1 to M2…
Figure 5. Fasudil shifts M1 to M2 phenotype.
Chronic EAE was induced in C57BL/6 mice with MOG35–55. Fasudil was injected (i.p.) at 40 mg/kg/d every other day on day 3 and 11 p.i. Mice were sacrificed and splenic MNCs were prepared on day 9 (a) and 21 (b) p.i.. Cells were stained with macrophage marker F4/80 and M1/M2 markers, and the polarization of M1/M2 was analyzed using flow cytometry. Scatterplot, representative dot plots from one of three experiments with similar results. Histogram, quantitative results are analyzed for 5 mice in each group. Results are expressed as the percentage of double positive cells in four-quadrant diagram. *p<0.05, **p<0.01, ***p<0.001.
Figure 6. Fasudil enhances the ratio of…
Figure 6. Fasudil enhances the ratio of Arg-1/iNOS.
Chronic EAE was induced in C57BL/6 mice with MOG35–55. Fasudil was injected (i.p.) at 40 mg/kg/d every other day on days 3 and 11 p.i. On day 21 p.i., mice were sacrificed, and protein was extracted from splenic macrophages and spinal cords. a) Representative bands of Western blot for iNOS and Arg-1 from one of two experiments with similar results. Quantitative analysis of iNOS and Arg-1 expression in splenic macrophages and spinal cords are analyzed for 4–5 mice in each group. Results are expressed as fold change relative to GAPDH as the loading control. *p<0.05, **p<0.01, ***p<0.001. b) The double staining of CD68 (green) and iNOS/Arg-1 (red) by immunohistochemistry. Representative patterns were obtained from one of three experiments with similar results.
Figure 7. Fasudil directly shifts M1 to…
Figure 7. Fasudil directly shifts M1 to M2 phenotype in macrophages.
Macrophages from healthy mice were purified and stimulated with IFN-γ. Fasudil was added for 12 h immediately after IFN-γ treatment. The levels of iNOS and Arg-1 were measured by Western blot (a) and flow cytometry (b). The results were analyzed from three independent experiments. *p<0.05, **p<0.01.
Figure 8. Fasudil inhibits inflammatory molecules.
Figure 8. Fasudil inhibits inflammatory molecules.
Chronic EAE was induced in C57BL/6 mice with MOG35–55. Fasudil was injected (i.p.) at 40 mg/kg/d every other day on days 3 and 11 p.i. On day 21 p.i., spleen was obtained, and splenic MNCs were prepared. Splenic MNCs were incubated for 48 h at 37°C in the presence of MOG35–55 (10 µg/ml). Splenic MNCs from healthy mice were measured as control. The supernatants were collected, and the levels of IL-10, IL-17 (a), IL-1β, IL-6, TNF-α and MCP-1 (b) were measured by ELISA kits. The results were represented as pg/ml, and analyzed from 5 mice in each group. *p<0.05, **p<0.01.

References

    1. Tselis A (2011) Evidence for viral etiology of multiple sclerosis. Semin Neurol 31: 307–316.
    1. Taylor BV (2011) The major cause of multiple sclerosis is environmental: genetics has a minor role–yes. Mult Scler 17: 1171–1173.
    1. Haegert DG (2011) Multiple sclerosis: a disorder of altered T-cell homeostasis. Mult Scler Int 2011: 461304.
    1. Sosa RA, Forsthuber TG (2011) The critical role of antigen-presentation-induced cytokine crosstalk in the central nervous system in multiple sclerosis and experimental autoimmune encephalomyelitis. J Interferon Cytokine Res 31: 753–768.
    1. Denkinger CM, Denkinger M, Kort JJ, Metz C, Forsthuber TG (2003) In vivo blockade of macrophage migration inhibitory factor ameliorates acute experimental autoimmune encephalomyelitis by impairing the homing of encephalitogenic T cells to the central nervous system. J Immunol 170: 1274–1282.
    1. Ukkonen M, Wu K, Reipert B, Dastidar P, Elovaara I (2007) Cell surface adhesion molecules and cytokine profiles in primary progressive multiple sclerosis. Mult Scler 13: 701–707.
    1. Pinto-Medel MJ, García-León JA, Oliver-Martos B, López-Gómez C, Luque G, et al. (2012) The CD4(+) T-cell subset lacking expression of the CD28 costimulatory molecule is expanded and shows a higher activation state in MS. J Neuroimmunol 243: 1–11.
    1. Martin R (2012) Anti-CD25 (daclizumab) monoclonal antibody therapy in relapsing-remitting multiple sclerosis. Clin Immunol 142: 9–14.
    1. Constantinescu CS, Farooqi N, O’Brien K, Gran B (2011) Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol 164: 1079–1106.
    1. Bennett J, Basivireddy J, Kollar A, Biron KE, Reickmann P, et al. (2010) Blood-brain barrier disruption and enhanced vascular permeability in the multiple sclerosis model EAE. J Neuroimmunol 229: 180–191.
    1. Segal BM (2003) Experimental autoimmune encephalomyelitis: cytokines, effector T cells, and antigen-presenting cells in a prototypical Th1-mediated autoimmune disease. Curr Allergy Asthma Rep 3: 86–93.
    1. Becher B, Segal BM (2011) T(H)17 cytokines in autoimmune neuro-inflammation. Curr Opin Immunol 23: 707–712.
    1. Haas J, Korporal M, Balint B, Fritzsching B, Schwarz A, et al. (2009) Glatiramer acetate improves regulatory T-cell function by expansion of naive CD4(+)CD25(+)FOXP3(+)CD31(+) T-cells in patients with multiple sclerosis. J Neuroimmunol 216: 113–117.
    1. Michel L, Berthelot L, Pettré S, Wiertlewski S, Lefrère F, et al. (2008) Patients with relapsing-remitting multiple sclerosis have normal Treg function when cells expressing IL-7 receptor alpha-chain are excluded from the analysis. J Clin Invest 118: 3411–3419.
    1. Mikita J, Dubourdieu-Cassagno N, Deloire MS, Vekris A, Biran M, et al. (2011) Altered M1/M2 activation patterns of monocytes in severe relapsing experimental rat model of multiple sclerosis. Amelioration of clinical status by M2 activated monocyte administration. Mult Scler 17: 2–15.
    1. Lavasani S, Dzhambazov B, Nouri M, Fåk F, Buske S, et al. (2010) A novel probiotic mixture exerts a therapeutic effect on experimental autoimmune encephalomyelitis mediated by IL-10 producing regulatory T cells. PLoS One 5: e9009.
    1. Jadidi-Niaragh F, Mirshafiey A (2011) Regulatory T-cell as orchestra leader in immunosuppression process of multiple sclerosis. Immunopharmacol Immunotoxicol 33: 545–567.
    1. Li H, Ciric B, Yang J, Xu H, Fitzgerald DC, et al... (2009) Intravenous tolerance modulates macrophage classical activation and antigen presentation in experimental autoimmune encephalomyelitis. J. Neuroimmunol. 208, 54–60.
    1. Gordon S (2003) Alternative activation of macrophages. Nat Rev Immunol 3: 23–35.
    1. Schroder K, Sweet MJ, Hume DA (2006) Signal integration between IFNgamma and TLR signalling pathways in macrophages. Immunobiology 211: 511–524.
    1. Martinez FO, Sica A, Mantovani A, Locati M (2008) Macrophage activation and polarization. Front Biosci 13: 453–461.
    1. Street CA, Bryan BA (2011) Rho kinase proteins–pleiotropic modulators of cell survival and apoptosis. Anticancer Res 31: 3645–3657.
    1. Guilluy C, Garcia-Mata R, Burridge K (2011) Rho protein crosstalk: another social network? Trends Cell Biol 21: 718–726.
    1. Bao W, Hu E, Tao L, Boyce R, Mirabile R, et al. (2004) Inhibition of Rho-kinase protects the heart against ischemia/reperfusion injury. Cardiovasc Res 61: 548–558.
    1. Slotta JE, Laschke MW, Menger MD, Thorlacius H (2008) Rho-kinase signalling mediates endotoxin hypersensitivity after partial hepatectomy. Br J Surg 95: 976–984.
    1. Ding RY, Zhao DM, Zhang ZD, Guo RX, Ma XC (2011) Pretreatment of Rho kinase inhibitor inhibits systemic inflammation and prevents endotoxin-induced acute lung injury in mice. J Surg Res 171: e209–214.
    1. Pierot L, Aggour M, Moret J (2010) Vasospasm after aneurysmal subarachnoid hemorrhage: recent advances in endovascular management. Curr Opin Crit Care 16: 110–116.
    1. Rikitake Y, Kim HH, Huang Z, Seto M, Yano K, et al. (2005) Inhibition of Rho kinase (ROCK) leads to increased cerebral blood flow and stroke protection. Stroke 36: 2251–2257.
    1. Hara M, Takayasu M, Watanabe K, Noda A, Takagi T, et al. (2000) Protein kinase inhibition by fasudil hydrochloride promotes neurological recovery after spinal cord injury in rats. J Neurosurg 93: 94–101.
    1. Wang L, Kamath A, Frye J, Iwamoto GA, Chun JL, et al. (2011) Aorta-derived mesoangioblasts differentiate into the oligodendrocytes by inhibition of the Rho Kinase signaling pathway. Stem Cells Dev 21: 1069–1089.
    1. Huang XN, Fu J, Wang WZ (2011) The effects of fasudil on the permeability of the rat blood-brain barrier and blood-spinal cord barrier following experimental autoimmune encephalomyelitis. J Neuroimmunol 239: 61–67.
    1. Sun X, Minohara M, Kikuchi H, Ishizu T, Tanaka M, et al. (2006) The selective Rho-kinase inhibitor Fasudil is protective and therapeutic in experimental autoimmune encephalomyelitis. J Neuroimmunol 180: 126–134.
    1. Yu JZ, Ding J, Ma CG, Sun CH, Sun YF, et al. (2010) Therapeutic potential of experimental autoimmune encephalomyelitis by Fasudil, a Rho kinase inhibitor. J Neurosci Res 88: 1664–1672.
    1. Yamamoto M, Ramirez SH, Sato S, Kiyota T, Cerny RL, et al. (2008) Phosphorylation of claudin-5 and occludin by rho kinase in brain endothelial cells. Am J Pathol 172: 521–533.
    1. Heasman SJ, Ridley AJ (2010) Multiple roles for RhoA during T cell transendothelial migration. Small Gtpases 1: 174–179.
    1. Carson MJ, Thrash JC, Walter B (2006) The cellular response in neuroinflammation: The role of leukocytes, microglia and astrocytes in neuronal death and survival. Clin Neurosci Res 6: 237–245.
    1. Engelhardt B (2006) Molecular mechanisms involved in T cell migration across the blood-brain barrier. J Neural Transm 113: 477–485.
    1. Mensah-Brown EP, Shahin A, Al Shamisi M, Lukic ML (2011) Early influx of macrophages determines susceptibility to experimental allergic encephalomyelitis in Dark Agouti (DA) rats. J Neuroimmunol 232: 68–74.
    1. Ajami B, Bennett JL, Krieger C, McNagny KM, Rossi FM (2011) Infiltrating monocytes trigger EAE progression, but do not contribute to the resident microglia pool. Nat Neurosci 14: 1142–1149.
    1. Benveniste EN (1997) Role of macrophages/microglia in multiple sclerosis and experimental allergic encephalomyelitis. J Mol Med 75: 165–173.
    1. Bauer J, Sminia T, Wouterlood FG, Dijkstra CD (1994) Phagocytic activity of macrophages and microglial cells during the course of acute and chronic relapsing experimental autoimmune encephalomyelitis. J Neurosci Res 38: 365–375.
    1. Benoit M, Desnues B, Mege JL (2008) Macrophage polarization in bacterial infections. J Immunol 181: 3733–3739.
    1. Kou PM, Babensee JE (2011) Macrophage and dendritic cell phenotypic diversity in the context of biomaterials. J Biomed Mater Res A 96: 239–260.
    1. Bosurgi L, Manfredi AA, Rovere-Querini P (2011) Macrophages in injured skeletal muscle: a perpetuum mobile causing and limiting fibrosis, prompting or restricting resolution and regeneration. Front Immunol 2: 62.
    1. Lumeng CN, Bodzin JL, Saltiel AR (2007) Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 117: 175–184.
    1. Wolf NA, Amouzegar TK, Swanborg RH (2007) Synergistic interaction between Toll-like receptor agonists is required for induction of experimental autoimmune encephalomyelitis in Lewis rats. J Neuroimmunol 185: 115–122.
    1. Sinha S, Miller L, Subramanian S, McCarty OJ, Proctor T, et al. (2010) Binding of recombinant T cell receptor ligands (RTL) to antigen presenting cells prevents upregulation of CD11b and inhibits T cell activation and transfer of experimental autoimmune encephalomyelitis. J Neuroimmunol 225: 52–61.
    1. Lutterotti A, Kuenz B, Gredler V, Khalil M, Ehling R, et al. (2006) Increased serum levels of soluble CD14 indicate stable multiple sclerosis. J Neuroimmunol 181: 145–149.
    1. Marchetti V, Yanes O, Aguilar E, Wang M, Friedlander D, et al. (2011) Differential macrophage polarization promotes tissue remodeling and repair in a model of ischemic retinopathy. Sci Rep 1: 76.
    1. Martinez FO, Helming L, Gordon S (2009) Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol 27: 451–83.
    1. Chan T, Pek EA, Huth K, Ashkar AA (2011) CD4(+) T-cells are important in regulating macrophage polarization in C57BL/6 wild-type mice. Cell Immunol 266: 180–186.
    1. Deng B, Wehling-Henricks M, Villalta SA, Wang Y, Tidball JG (2012) IL-10 triggers changes in macrophage phenotype that promote muscle growth and regeneration. J Immunol 189: 3669–3680.
    1. Liu G, Ma H, Qiu L, Li L, Cao Y, et al. (2011) Phenotypic and functional switch of macrophages induced by regulatory CD4+CD25+ T cells in mice. Immunol Cell Biol 89: 130–142.
    1. Jiang HR, Milovanović M, Allan D, Niedbala W, Besnard AG, et al. (2012) IL-33 attenuates EAE by suppressing IL-17 and IFN-γ production and inducing alternatively activated macrophages. Eur J Immunol 42: 1804–1814.

Source: PubMed

3
Se inscrever