Oestrogen receptor β ligand acts on CD11c+ cells to mediate protection in experimental autoimmune encephalomyelitis

Roy Y Kim, Darian Mangu, Alexandria S Hoffman, Rojan Kavosh, Eunice Jung, Noriko Itoh, Rhonda Voskuhl, Roy Y Kim, Darian Mangu, Alexandria S Hoffman, Rojan Kavosh, Eunice Jung, Noriko Itoh, Rhonda Voskuhl

Abstract

Oestrogen treatments are neuroprotective in a variety of neurodegenerative disease models. Selective oestrogen receptor modifiers are needed to optimize beneficial effects while minimizing adverse effects to achieve neuroprotection in chronic diseases. Oestrogen receptor beta (ERβ) ligands are potential candidates. In the multiple sclerosis model chronic experimental autoimmune encephalomyelitis, ERβ-ligand treatment is neuroprotective, but mechanisms underlying this neuroprotection remain unclear. Specifically, whether there are direct effects of ERβ-ligand on CD11c+ microglia, myeloid dendritic cells or macrophages in vivo during disease is unknown. Here, we generated mice with ERβ deleted from CD11c+ cells to show direct effects of ERβ-ligand treatment in vivo on these cells to mediate neuroprotection during experimental autoimmune encephalomyelitis. Further, we use bone marrow chimeras to show that ERβ in peripherally derived myeloid cells, not resident microglia, are the CD11c+ cells mediating this protection. CD11c+ dendritic cell and macrophages isolated from the central nervous system of wild-type experimental autoimmune encephalomyelitis mice treated with ERβ-ligand expressed less iNOS and T-bet, but more IL-10, and this treatment effect was lost in mice with specific deletion of ERβ in CD11c+ cells. Also, we extend previous reports of ERβ-ligand’s ability to enhance remyelination through a direct effect on oligodendrocytes by showing that the immunomodulatory effect of ERβ-ligand acting on CD11c+ cells is necessary to permit the maturation of oligodendrocytes. Together these results demonstrate that targeting ERβ signalling pathways in CD11c+ myeloid cells is a novel strategy for regulation of the innate immune system in neurodegenerative diseases. To our knowledge, this is the first report showing how direct effects of a candidate neuroprotective treatment on two distinct cell lineages (bone marrow derived myeloid cells and oligodendrocytes) can have complementary neuroprotective effects in vivo.awx315media15688130498001.

Keywords: experimental autoimmune encephalomyelitis; macrophage; multiple sclerosis; neuroprotection; oestrogen receptor beta.

© The Author (2017). Published by Oxford University Press on behalf of the Guarantors of Brain.

Figures

Figure 1
Figure 1
ERβ-specific deletion in CD11c+ cells during EAE. (A) Flow cytometry dot plots of CNS mononuclear immune cells isolated from brains and spinal cords of EAE mice. CNS mononuclear immune cells were gated based on FSC and CD45 expression. CNS CD45+ gated cells included two separate populations, CD45hi and CD45int, within the R1 gate. CD45+ cells were further gated based on CD11c expression into R2: CD45+CD11c− and R3: CD45+CD11c+. CD11b staining revealed that R2 included CD45hiCD11c−CD11b− lymphocytes, CD45hiCD11c−CD11b+ macrophages, and CD45intCD11c−CD11b+ resident microglia, and R3 included CD45hiCD11c+CD11b+ peripherally derived myeloid dendritic cells and macrophages (DC/Mφ) and CD45intCD11c+CD11b+ resident microglia. FSC = forward scatter. (B) Immunofluorescence images of spinal cord tissues stained with CD11c-GFP (green), ERβ (red) and nuclear stain DAPI (blue) with merged images for co-localization (yellow) on right. Top row: CD11c-cre-GFP+ control mice showed co-localization (white arrows) of CD11c-GFP and ERβ. Bottom row: CD11c-cre-GFP+;ERβfl/fl CKO mice did not show co-localization (white arrows). Orange arrows represent other cells in the CNS expressing ERβ. Scale bar = 20 μm. (C) Quantitative analysis of CD11c-GFP and ERβ co-localization in immunofluorescence images of CD11c-cre-GFP+ (Control) and CD11c ERβ CKO (CKO) mice with EAE. (D) Representative flow cytometry plots of isolated CNS mononuclear immune cells from a pool of three individual mice. CNS mononuclear immune cells were gated based on SSC and FSC (left), and subpopulations were identified using CD11c and CD45 staining (right). Cell populations labelled as CD11c+ microglia, CD11c+ myeloid dendritic cells and macrophages, and CD11c− cells were FACS sorted for mRNA isolation and quantitative PCR analysis. (E) Quantitative analysis of ERβ (Esr2) mRNA expression in sorted CNS CD11c+ microglia, CD11c+ myeloid dendritic cells and macrophages cells, and CD11c− cells from mice with ERβ deleted in CD11c+ cells (CKO) and wild-type mice, each with EAE.
Figure 2
Figure 2
ERβ expression in CD11c+ cells is necessary for neuroprotection in EAE. (A) Breeding scheme for creating wild-type (WT) and CKO mice of ERβ in CD11c+ cells. Briefly, CD11c-Cre-GFP+ mice were crossed with mice carrying an ERβ (Esr2) gene flanked by LoxP sites (ERβfl/fl). Homozygous ERβfl/fl mice without (WT) or with (CKO) Cre were generated. Each genotype was separated into two groups and received either vehicle or ERβ-ligand, DPN, treatment (tx). EAE was induced and animals were monitored daily and scored using the standard EAE 0–5 scale. ERβ-ligand treated wild-type EAE mice (WT-ERβ, blue solid) had significantly better clinical scores compared to vehicle treated wild-type EAE mice (WT-V, blue clear), ***P (WT-V versus WT-ERβ) = 0.0002, after Day 20 of EAE. In contrast, ERβ-ligand mediated protection did not occur in ERβ-ligand treated CKO EAE mice (CKO-ERβ, black solid) when compared to vehicle treated CKO EAE mice (CKO-V, black clear), P (CKO-V versus CKO-ERβ) > 0.9999. Detailed EAE statistics are in Supplementary Tables 1 and 2. (B) Representative images and quantitative analyses of NF200+, SMI32+, and βAPP+ axons in dorsal white matter of the spinal cord. Images were taken at 40× magnification. Scale bar = 20 μm. (C) Representative images and quantitative analyses of MBP+ and CNPase+ mean intensity in dorsal white matter of the spinal cord. Scale bar = 100 μm. (D) Representative electron microscopy images of ultraresolution of axons and myelin thickness in the dorsal white matter of spinal cord. Myelin thickness in the wild-type and CKO EAE mice treated with vehicle and ERβ-ligand was measured using the g-ratio (axon diameter) / (axon + outer myelin diameter) and shown in comparison with normal. Quantitative analysis showed that ERβ-ligand treated wild-type EAE mice had a decrease in g-ratio due to increased outer myelin diameter, while CD11c ERβ CKO mice with EAE did not. Scale bar = 1 μm. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Data are representative of three repeated experiments.
Figure 3
Figure 3
Qualitative effects on inflammatory markers on CNS resident and infiltrated Iba1+ myeloid cells. (A) Representative images of spinal cord tissues stained with MHCII (red) and Iba1 (green), (B) iNOS (red) and Iba1 (green), and (C) ARG1 (red) and Iba1 (green). Scale bar = 50 μm and 10 μm (inset). Inset: white arrows indicate co-localization. (D) MHCII+Iba1+ myeloid cells were increased in vehicle treated wild-type EAE mice (WT-V) compared to healthy controls (N), while ERβ-ligand treated wild-type EAE mice (WT-ERβ) showed a reduction in per cent MHCII+Iba1+ myeloid cells. In contrast, ERβ-ligand treated CKO EAE mice (CKO-ERβ) were no different from vehicle treated CKO EAE mice (CKO-V). (E) iNOS+Iba1+ myeloid cells were also reduced in ERβ-ligand treated wild-type EAE, but not in CKO EAE mice. (F) ARG1+Iba1+ myeloid cells were no different between groups. *P < 0.05; **P < 0.01; ***P < 0.001. Data are representative of two repeated experiments.
Figure 4
Figure 4
ERβ-ligand treatment acts on CD11c+ cells to permit increases in mature oligodendrocytes during EAE. (A) Immunofluorescence images of spinal cord tissues stained with Olig2 (red), and co-stained with GSTπ (green), CC1 (green), and NG2 (green). On the left is a representative image of Olig2+ oligodendrocyte lineage cells (OLC) in dorsal white matter of the spinal cord. On the right are representative images of each co-stain; Olig2-GSTπ (top), Olig2-CC1 (middle) and Olig2-NG2 (bottom). Scale bar = 50 μm (left) and 20 μm (right). White box indicates the area where the co-stains were imaged. Quantitative analyses of (B) Olig2+GSTπ+ mature OLCs, (C) Olig2+CC1+ immature/mature OLCs, and (D) Olig2+NG2+ oligodendrocyte precursor cells, each in dorsal white matter of the spinal cord. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Data are representative of two repeated experiments.
Figure 5
Figure 5
ERβ expression on Olig1+ cells is necessary for neuroprotection in EAE. (A) Breeding scheme for creating wild-type (WT) and CKO mice of ERβ in Olig1+ cells. Briefly, Olig1-Cre + mice were crossed with mice carrying an ERβ (Esr2) gene flanked by LoxP sites (ERβfl/fl). Homozygous ERβfl/fl mice without (Olig1-WT) or with (Olig1-CKO) Cre were generated. Each genotype was separated into two groups and received either vehicle or ERβ-ligand treatment (tx), EAE was induced and mice were scored for EAE severity as above. ERβ-ligand treated Olig1-WT EAE mice (Olig1-WT-ERβ, blue solid) had significantly better clinical scores compared to vehicle treated Olig1-WT EAE mice (Olig1-WT-V, blue clear), **P (Olig1-WT-V versus Olig1-WT-ERβ) = 0.0095, after Day 20 of EAE. In contrast, ERβ-ligand mediated protection did not occur in ERβ-ligand treated Olig1-CKO EAE mice (Olig1-CKO-ERβ, black solid) when compared to vehicle treated Olig1-CKO EAE mice (Olig1-CKO-V, black clear), P (Olig1-CKO-V versus Olig1-CKO-ERβ) = 0.7967. Detailed EAE statistics are in Supplementary Tables 1 and 2. Quantitative analyses of (B) NF200+, SMI32+, and βAPP+ axonal counts, (C) MBP+ and CNPase+ myelin intensity, and (D) MHCII+Iba1+ myeloid cells in dorsal white matter of the spinal cord. (E) Quantitative analyses of total Olig2+ OLCs, and the percentage of subpopulations: Olig2+NG2+ OPCs, Olig2+CC1+ immature/mature oligodendrocytes, and Olig2+GSTπ+ mature oligodendrocytes in dorsal white matter of the spinal cord. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Data are representative of three repeated experiments.
Figure 6
Figure 6
CD11c+ microglial ERβ expression is not necessary for neuroprotection in EAE. (A) Diagram for creating CD11c+ microglia-CKO using BMC. Briefly, CD45.1 wild-type mice were used as donors for bone marrow cells, and CD45.2 wild-type (WT→WT) or CKO (WT→CKO) were used as irradiated recipients. Each genotype was separated into two groups and received either vehicle or ERβ-ligand treatment (tx), EAE was induced and mice were scored for EAE severity. (B) ERβ-ligand treated wild-type (WT→WT-ERβ, blue solid) EAE mice had significantly better scores compared to vehicle treated wildtype (WT→WT-V, blue clear) EAE mice, *P (WT→WT-V versus WT→WT-ERβ) = 0.0188. Similarly, ERβ-ligand treated CKO (WT→CKO-ERβ, black solid) EAE mice also had significantly better scores compared to vehicle treated conditional knockout (WT→CKO-V, black clear) EAE mice, **P (WT→CKO-V versus WT→CKO-ERβ) = 0.0077. Detailed EAE statistics are in Supplementary Tables 1 and 2. Quantitative analysis of (C) NF200+ axonal count, (D) βAPP+ axonal count, (E) MBP+ myelin intensity, and (F) MHCII expression on Iba1+ myeloid derived cells (percentage) in dorsal white matter of the spinal cord. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001. Data are representative of two repeated experiments.
Figure 7
Figure 7
ERβ expression on CD11c+ myeloid dendritic cells and macrophages is necessary for neuroprotection in EAE. (A) Diagram for creating CD11c+ myeloid dendritic cells and macrophages (DC/MΦ) CKO using bone marrow chimeras. Briefly, CD45.2 wild-type (WT→WT) versus CKO (CKO→WT) mice were used as donors for bone marrow cells, and CD45.1 wild-type mice were used as irradiated recipients. Each genotype was separated into two groups and received either vehicle or ERβ-ligand treatment (tx), EAE was induced and mice were scored for EAE severity. (B) ERβ-ligand treated wild-type (WT→WT-ERβ, blue solid) EAE mice had significantly better EAE scores compared to vehicle treated wild-type (WT→WT-V, blue open) EAE mice, ***P (WT→WT-V versus WT→WT-ERβ) = 0.0005, after Day 20 of EAE, whereas ERβ-ligand mediated protection did not occur in ERβ-ligand treated conditional knockout (CKO→WT-ERβ, black solid) compared to vehicle treated CKO (CKO→WT-V, black clear) EAE mice, P (CKO→WT-V versus CKO→WT-ERβ) > 0.9999. Detailed EAE statistics are in Supplementary Tables 1 and 2. Quantitative analysis of (C) NF200+ axonal count, (D) βAPP+ axonal count, (E) MBP+ myelin intensity, and (F) MHCII expression on Iba1+ myeloid derived cells (percentage) in dorsal white matter of the spinal cord. *P < 0.05; **P < 0.01; ***P < 0.001; ****P < 0.0001.
Figure 8
Figure 8
Gene expression profiles of CD11c+ microglia and CD11c+ dendritic cells and macrophages cells from the CNS of ERβ-ligand or vehicle treated mice with EAE. (A) Representative flow cytometry plots of isolated CNS mononuclear immune cells from a pool of two to four individual mice. CNS mononuclear immune cells were gated based on SSC and FSC (left), and subpopulations were identified using CD11c and CD45 staining (right). Cell populations labelled as CD11c+ microglia and CD11c+ myeloid dendritic cells and macrophages were FACS sorted for mRNA isolation and quantitative PCR analysis. (B) Quantitative analysis of iNOS, T-bet, IL-10, CCR2, ARG1, and YM-1 mRNA expression levels of sorted CD11c+ microglia and CD11c+ myeloid dendritic cells and macrophages from wild type (WT) (left) and CD11c ERβ CKO (right) mice with EAE that were treated with vehicle or ERβ-ligand. Data are from three separate experiments, with error bar representing variation between experiments. *P < 0.05.

References

    1. Antal MC, Krust A, Chambon P, Mark M. Sterility and absence of histopathological defects in nonreproductive organs of a mouse ERbeta-null mutant. Proc Natl Acad Sci USA 2008; 105: 2433–8.
    1. Bailey SL, Schreiner B, McMahon EJ, Miller SD. CNS myeloid dendritic cells presenting endogenous myelin peptides ‘preferentially' polarize CD4+ T(H)-17 cells in relapsing EAE. Nat Immunol 2007; 8: 172–80.
    1. Bennett ML, Bennett FC, Liddelow SA, Ajami B, Zamanian JL, Fernhoff NB, et al.New tools for studying microglia in the mouse and human CNS. Proc Natl Acad Sci USA 2016; 113: E1738–46.
    1. Bulloch K, Miller MM, Gal-Toth J, Milner TA, Gottfried-Blackmore A, Waters EM, et al.CD11c/EYFP transgene illuminates a discrete network of dendritic cells within the embryonic, neonatal, adult, and injured mouse brain. J Comp Neurol 2008; 508: 687–710.
    1. Clarkson BD, Walker A, Harris MG, Rayasam A, Sandor M, Fabry Z. CCR2-dependent dendritic cell accumulation in the central nervous system during early effector experimental autoimmune encephalomyelitis is essential for effector T cell restimulation in situ and disease progression. J Immunol 2015; 194: 531–41.
    1. De Angelis M, Stossi F, Carlson KA, Katzenellenbogen BS, Katzenellenbogen JA. Indazole estrogens: highly selective ligands for the oestrogen receptor beta. J Med Chem 2005; 48: 1132–44.
    1. Dogan RN, Elhofy A, Karpus WJ. Production of CCL2 by central nervous system cells regulates development of murine experimental autoimmune encephalomyelitis through the recruitment of TNF- and iNOS-expressing macrophages and myeloid dendritic cells. J Immunol 2008; 180: 7376–84.
    1. Du S, Sandoval F, Trinh P, Umeda E, Voskuhl R. Oestrogen receptor-beta ligand treatment modulates dendritic cells in the target organ during autoimmune demyelinating disease. Eur J Immunol 2011; 41: 140–50.
    1. Frasor J, Barnett DH, Danes JM, Hess R, Parlow AF, Katzenellenbogen BS. Response-specific and ligand dose-dependent modulation of oestrogen receptor (ER) alpha activity by ERbeta in the uterus. Endocrinology 2003; 144: 3159–66.
    1. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S, et al.Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 2010; 330: 841–5.
    1. Gold R, Linington C, Lassmann H. Understanding pathogenesis and therapy of multiple sclerosis via animal models: 70 years of merits and culprits in experimental autoimmune encephalomyelitis research. Brain 2006; 129(Pt 8): 1953–71.
    1. Greter M, Heppner FL, Lemos MP, Odermatt BM, Goebels N, Laufer T, et al.Dendritic cells permit immune invasion of the CNS in an animal model of multiple sclerosis. Nat Med 2005; 11: 328–34.
    1. Harris HA, Katzenellenbogen JA, Katzenellenbogen BS. Characterization of the biological roles of the oestrogen receptors, ERalpha and ERbeta, in oestrogen target tissues in vivo through the use of an ERalpha-selective ligand. Endocrinology 2002; 143: 4172–7.
    1. Itoh N, Kim R, Peng M, DiFilippo E, Johnsonbaugh H, MacKenzie-Graham A, et al.Bedside to bench to bedside research: oestrogen receptor beta ligand as a candidate neuroprotective treatment for multiple sclerosis. J Neuroimmunol 2017; 304: 63–71.
    1. Itoh Y, Voskuhl RR. Cell specificity dictates similarities in gene expression in multiple sclerosis, Parkinson's disease, and Alzheimer's disease. PLoS One 2017; 12: e0181349.
    1. Karman J, Ling C, Sandor M, Fabry Z. Initiation of immune responses in brain is promoted by local dendritic cells. J Immunol 2004; 173: 2353–61.
    1. Khalaj AJ, Yoon J, Nakai J, Winchester Z, Moore SM, Yoo T, et al.Estrogen receptor (ER) beta expression in oligodendrocytes is required for attenuation of clinical disease by an ERbeta ligand. Proc Natl Acad Sci USA 2013; 110: 19125–30.
    1. Kim RY, Hoffman AS, Itoh N, Ao Y, Spence R, Sofroniew MV, et al.Astrocyte CCL2 sustains immune cell infiltration in chronic experimental autoimmune encephalomyelitis. J Neuroimmunol 2014; 274: 53–61.
    1. Kim S, Liva SM, Dalal MA, Verity MA, Voskuhl RR. Estriol ameliorates autoimmune demyelinating disease: implications for multiple sclerosis. Neurology 1999; 52: 1230–8.
    1. King IL, Dickendesher TL, Segal BM. Circulating Ly-6C+ myeloid precursors migrate to the CNS and play a pathogenic role during autoimmune demyelinating disease. Blood 2009; 113: 3190–7.
    1. Kuiper GG, Carlsson B, Grandien K, Enmark E, Haggblad J, Nilsson S, et al.Comparison of the ligand binding specificity and transcript tissue distribution of estrogen receptors alpha and beta. Endocrinology 1997; 138: 863–70.
    1. Kumar S, Patel R, Moore S, Crawford DK, Suwanna N, Mangiardi M, et al.Estrogen receptor beta ligand therapy activates PI3K/Akt/mTOR signaling in oligodendrocytes and promotes remyelination in a mouse model of multiple sclerosis. Neurobiol Dis 2013; 56: 131–44.
    1. Lariosa-Willingham KD, Rosler ES, Tung JS, Dugas JC, Collins TL, Leonoudakis D. A high throughput drug screening assay to identify compounds that promote oligodendrocyte differentiation using acutely dissociated and purified oligodendrocyte precursor cells. BMC Res Notes 2016; 9: 419.
    1. Lauritzen C. Results of a 5 years prospective study of estriol succinate treatment in patients with climacteric complaints. Horm Metab Res 1987; 19: 579–84.
    1. Lelu K, Laffont S, Delpy L, Paulet PE, Perinat T, Tschanz SA, et al.Estrogen receptor alpha signaling in T lymphocytes is required for estradiol-mediated inhibition of Th1 and Th17 cell differentiation and protection against experimental autoimmune encephalomyelitis. J Immunol 2011; 187: 2386–93.
    1. Lindberg BS, Johansson ED, Nilsson BA. Plasma levels of nonconjugated oestrone, oestradiol-17beta and oestriol during uncomplicated pregnancy. Acta Obstet Gynecol Scand Suppl 1974; 32: 21–36.
    1. Lu QR, Sun T, Zhu Z, Ma N, Garcia M, Stiles CD, et al.Common developmental requirement for Olig function indicates a motor neuron/oligodendrocyte connection. Cell 2002; 109: 75–86.
    1. Mackenzie-Graham AJ, Rinek GA, Avedisian A, Morales LB, Umeda E, Boulat B, et al.Estrogen treatment prevents gray matter atrophy in experimental autoimmune encephalomyelitis. J Neurosci Res 2012; 90: 1310–23.
    1. McFarland K, Price DL, Davis CN, Ma JN, Bonhaus DW, Burstein ES, et -186, a selective nonsteroidal estrogen receptor beta agonist, shows gender specific neuroprotection in a Parkinson's disease rat model. ACS Chem Neurosci 2013; 4: 1249–55.
    1. Mei F, Fancy SP, Shen YA, Niu J, Zhao C, Presley B, et al.Micropillar arrays as a high-throughput screening platform for therapeutics in multiple sclerosis. Nat Med 2014; 20: 954–60.
    1. Meyers MJ, Sun J, Carlson KE, Marriner GA, Katzenellenbogen BS, Katzenellenbogen JA. Estrogen receptor-beta potency-selective ligands: structure-activity relationship studies of diarylpropionitriles and their acetylene and polar analogues. J Med Chem 2001; 44: 4230–51.
    1. Miller SD, McMahon EJ, Schreiner B, Bailey SL. Antigen presentation in the CNS by myeloid dendritic cells drives progression of relapsing experimental autoimmune encephalomyelitis. Ann N Y Acad Sci 2007; 1103: 179–91.
    1. Miron VE, Boyd A, Zhao JW, Yuen TJ, Ruckh JM, Shadrach JL, et al.M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nat Neurosci 2013; 16: 1211–18.
    1. Mishra MK, Yong VW. Myeloid cells - targets of medication in multiple sclerosis. Nat Rev Neurol 2016; 12: 539–51.
    1. Moore SM, Khalaj AJ, Kumar S, Winchester Z, Yoon J, Yoo T, et al.Multiple functional therapeutic effects of the estrogen receptor beta agonist indazole-Cl in a mouse model of multiple sclerosis. Proc Natl Acad Sci USA 2014; 111: 18061–6.
    1. Morales LB, Loo KK, Liu HB, Peterson C, Tiwari-Woodruff S, Voskuhl RR. Treatment with an estrogen receptor alpha ligand is neuroprotective in experimental autoimmune encephalomyelitis. J Neurosci 2006; 26: 6823–33.
    1. Nilsson S, Koehler KF, Gustafsson JA. Development of subtype-selective oestrogen receptor-based therapeutics. Nat Rev Drug Discov 2011; 10: 778–92.
    1. Paharkova-Vatchkova V, Maldonado R, Kovats S. Estrogen preferentially promotes the differentiation of CD11c+ CD11b(intermediate) dendritic cells from bone marrow precursors. J Immunol 2004; 172: 1426–36.
    1. Palaszynski KM, Liu H, Loo KK, Voskuhl RR. Estriol treatment ameliorates disease in males with experimental autoimmune encephalomyelitis: implications for multiple sclerosis. J Neuroimmunol 2004; 149: 84–9.
    1. Paterni I, Granchi C, Katzenellenbogen JA, Minutolo F. Estrogen receptors alpha (ERalpha) and beta (ERbeta): subtype-selective ligands and clinical potential. Steroids 2014; 90: 13–29.
    1. Prodinger C, Bunse J, Kruger M, Schiefenhovel F, Brandt C, Laman JD, et al.CD11c-expressing cells reside in the juxtavascular parenchyma and extend processes into the glia limitans of the mouse nervous system. Acta Neuropathol 2011; 121: 445–58.
    1. Prokai L, Nguyen V, Szarka S, Garg P, Sabnis G, Bimonte-Nelson HA, et al.The prodrug DHED selectively delivers 17beta-estradiol to the brain for treating estrogen-responsive disorders. Sci Transl Med 2015; 7: 297ra113.
    1. Rawji KS, Yong VW. The benefits and detriments of macrophages/microglia in models of multiple sclerosis. Clin Dev Immunol 2013; 2013: 948976.
    1. Richardson TI, Dodge JA, Durst GL, Pfeifer LA, Shah J, Wang Y, et al.Benzopyrans as selective estrogen receptor beta agonists (SERBAs). Part 3: synthesis of cyclopentanone and cyclohexanone intermediates for C-ring modification. Bioorg Med Chem Lett 2007; 17: 4824–8.
    1. Saijo K, Collier JG, Li AC, Katzenellenbogen JA, Glass CK. An ADIOL-ERbeta-CtBP transrepression pathway negatively regulates microglia-mediated inflammation. Cell 2011; 145: 584–95.
    1. Samantaray S, Matzelle DD, Ray SK, Banik NL. Physiological low dose of estrogen-protected neurons in experimental spinal cord injury. Ann N Y Acad Sci 2010; 1199: 86–9.
    1. Shen FW, Saga Y, Litman G, Freeman G, Tung JS, Cantor H, et al.Cloning of Ly-5 cDNA. Proc Natl Acad Sci USA 1985; 82: 7360–3.
    1. Sicotte NL, Liva SM, Klutch R, Pfeiffer P, Bouvier S, Odesa S, et al.Treatment of multiple sclerosis with the pregnancy hormone estriol. Ann Neurol 2002; 52: 421–8.
    1. Spence RD, Hamby ME, Umeda E, Itoh N, Du S, Wisdom AJ, et al.Neuroprotection mediated through estrogen receptor-{alpha} in astrocytes. Proc Natl Acad Sci USA 2011; 108: 8867–72.
    1. Spence RD, Voskuhl RR. Neuroprotective effects of estrogens and androgens in CNS inflammation and neurodegeneration. Front Neuroendocrinol 2012; 33: 105–15.
    1. Spence RD, Wisdom AJ, Cao Y, Hill HM, Mongerson CR, Stapornkul B, et al.Estrogen mediates neuroprotection and anti-inflammatory effects during EAE through ERalpha signaling on astrocytes but not through ERbeta signaling on astrocytes or neurons. J Neurosci 2013; 33: 10924–33.
    1. Srinivasan K, Friedman BA, Larson JL, Lauffer BE, Goldstein LD, Appling LL, et al.Untangling the brain's neuroinflammatory and neurodegenerative transcriptional responses. Nat Commun 2016; 7: 11295.
    1. Steinman L. Multiple sclerosis: a coordinated immunological attack against myelin in the central nervous system. Cell 1996; 85: 299–302.
    1. Stranges PB, Watson J, Cooper CJ, Choisy-Rossi CM, Stonebraker AC, Beighton RA, et al.Elimination of antigen-presenting cells and autoreactive T cells by Fas contributes to prevention of autoimmunity. Immunity 2007; 26: 629–41.
    1. Suzuki S, Brown CM, Wise PM. Neuroprotective effects of estrogens following ischemic stroke. Front Neuroendocrinol 2009; 30: 201–11.
    1. Takahashi K, Manabe A, Okada M, Kurioka H, Kanasaki H, Miyazaki K. Efficacy and safety of oral estriol for managing postmenopausal symptoms. Maturitas 2000; 34: 169–77.
    1. Tiwari-Woodruff S, Morales LB, Lee R, Voskuhl RR. Differential neuroprotective and antiinflammatory effects of estrogen receptor (ER)alpha and ERbeta ligand treatment. Proc Natl Acad Sci USA 2007; 104: 14813–8.
    1. Tulchinsky D, Hobel CJ, Yeager E, Marshall JR. Plasma estrone, estradiol, estriol, progesterone, and 17-hydroxyprogesterone in human pregnancy. I. Normal pregnancy. Am J Obstet Gynecol 1972; 112: 1095–100.
    1. Uchoa MF, Moser VA, Pike CJ. Interactions between inflammation, sex steroids, and Alzheimer's disease risk factors. Front Neuroendocrinol 2016; 43: 60–82.
    1. Vainchtein ID, Vinet J, Brouwer N, Brendecke S, Biagini G, Biber K, et acute experimental autoimmune encephalomyelitis, infiltrating macrophages are immune activated, whereas microglia remain immune suppressed. Glia 2014; 62: 1724–35.
    1. Voskuhl RR, Wang H, Wu TC, Sicotte NL, Nakamura K, Kurth F, et al.Estriol combined with glatiramer acetate for women with relapsing-remitting multiple sclerosis: a randomised, placebo-controlled, phase 2 trial. Lancet Neurol 2016; 15: 35–46.
    1. Warner M, Gustafsson JA. Estrogen receptor beta and Liver X receptor beta: biology and therapeutic potential in CNS diseases. Mol Psychiatry 2015; 20: 18–22.
    1. Wisdom AJ, Cao Y, Itoh N, Spence RD, Voskuhl RR. Estrogen receptor-beta ligand treatment after disease onset is neuroprotective in the multiple sclerosis model. J Neurosci Res 2013; 91: 901–8.
    1. Wlodarczyk A, Cedile O, Jensen KN, Jasson A, Mony JT, Khorooshi R, et al.Pathologic and protective roles for microglial subsets and bone marrow- and blood-derived myeloid cells in central nervous system inflammation. Front Immunol 2015; 6: 463.
    1. Wlodarczyk A, Lobner M, Cedile O, Owens T. Comparison of microglia and infiltrating CD11c(+) cells as antigen presenting cells for T cell proliferation and cytokine response. J Neuroinflammation 2014; 11: 57.
    1. Wu WF, Tan XJ, Dai YB, Krishnan V, Warner M, Gustafsson JA. Targeting estrogen receptor beta in microglia and T cells to treat experimental autoimmune encephalomyelitis. Proc Natl Acad Sci USA 2013; 110: 3543–8.
    1. Ziehn MO, Avedisian AA, Dervin SM, O'Dell TJ, Voskuhl RR. Estriol preserves synaptic transmission in the hippocampus during autoimmune demyelinating disease. Lab Invest 2012; 92: 1234–45.

Source: PubMed

3
Subskrybuj