CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature

Antoine Louveau, Jasmin Herz, Maria Nordheim Alme, Andrea Francesca Salvador, Michael Q Dong, Kenneth E Viar, S Grace Herod, James Knopp, Joshua C Setliff, Alexander L Lupi, Sandro Da Mesquita, Elizabeth L Frost, Alban Gaultier, Tajie H Harris, Rui Cao, Song Hu, John R Lukens, Igor Smirnov, Christopher C Overall, Guillermo Oliver, Jonathan Kipnis, Antoine Louveau, Jasmin Herz, Maria Nordheim Alme, Andrea Francesca Salvador, Michael Q Dong, Kenneth E Viar, S Grace Herod, James Knopp, Joshua C Setliff, Alexander L Lupi, Sandro Da Mesquita, Elizabeth L Frost, Alban Gaultier, Tajie H Harris, Rui Cao, Song Hu, John R Lukens, Igor Smirnov, Christopher C Overall, Guillermo Oliver, Jonathan Kipnis

Abstract

Neuroinflammatory diseases, such as multiple sclerosis, are characterized by invasion of the brain by autoreactive T cells. The mechanism for how T cells acquire their encephalitogenic phenotype and trigger disease remains, however, unclear. The existence of lymphatic vessels in the meninges indicates a relevant link between the CNS and peripheral immune system, perhaps affecting autoimmunity. Here we demonstrate that meningeal lymphatics fulfill two critical criteria: they assist in the drainage of cerebrospinal fluid components and enable immune cells to enter draining lymph nodes in a CCR7-dependent manner. Unlike other tissues, meningeal lymphatic endothelial cells do not undergo expansion during inflammation, and they express a unique transcriptional signature. Notably, the ablation of meningeal lymphatics diminishes pathology and reduces the inflammatory response of brain-reactive T cells during an animal model of multiple sclerosis. Our findings demonstrate that meningeal lymphatics govern inflammatory processes and immune surveillance of the CNS and pose a valuable target for therapeutic intervention.

Conflict of interest statement

Competing interests: J.K. is an Advisor to PureTech Health/Ariya.

Figures

Figure 1:. Meningeal lymphatic sub-arachnoid extensions uptake…
Figure 1:. Meningeal lymphatic sub-arachnoid extensions uptake molecules and immune cells from the cerebrospinal fluid.
a, General scheme of meningeal lymphatic vascular organization. b, Scheme of the experiment presented in (c). Prox1GFP mice were injected into the cisterna magna (i.c.m.) with 5µl of Qdot655. The transverse sinus was imaged through a thinned skull. c, Representative image of meningeal lymphatics adjacent to the transverse sinus (Prox1GFP – green) filled with the i.c.m. injected Qdot655 (red) 60min after injection. The inset represents the coronal view of the lymphatic vessel filled with Qdot655. Scale bar = 65 µm. Representative of 3 independent animals. d, Representative images of the lymphatic vessels co-labeled with i.c.m. injected anti-Lyve-1A488 and exogenously applied anti-Lyve-1A660 at different time points after i.c.m. injection. Arrows in inset at 5 and 15 min illustrate the initial points where the i.c.m. injected anti-Lyve-1A488 labelled the meningeal lymphatics. Scale bar = 1000 µm (upper panel); 250 µm (bottom left panel) 60 µm (bottom right panel). e, Quantification of the percentage of lymphatic vessels labeled by the i.c.m. injected antibody and total lymphatic area at different time points post injection (mean ± s.e.m.; n=4 mice/group). f, Representative images of OVA594 and fluorescent bead accumulation along the lymphatics (Lyve-1 - red) are shown. Arrows point to lymphatic extensions in the area of microbeads and OVA accumulation. Scale bar = 300 µm (upper panel), 120 µm (bottom panels). Representative of 5 independent animals. g, Representative images of the accumulation of exogenously injected T cells (CFSE – green) at the extension-rich regions of lymphatic vessels adjacent to the transverse sinuses 12h after i.c.m. injection. Scale bar = 1000µm (right panel), 200µm (insets) . Representative of 3 independent animals. h, Representative images of exogenously injected T cells (CFSE – green) located within the meningeal lymphatics (Lyve-1 – white) associated with the transverse sinus (CD31 – blue). Scale bar = 35 µm. Representative images of 5 independent animals. i, Representative images of the lymphatics of the superior sagittal sinus (left panel) and the transverse sinus (right panel) with i.c.m. injected Qdot655. Arrows point to the subarachnoid space (SAS). Scale bar = 25 µm. Representative of 2 independent animals. j, Representative images of lymphatic sprout at the extension-rich region of lymphatic vessels along the transverse sinus, immunostained with Lyve-1 (grey) and junction proteins, VE-Cadherin (green) and Claudin-5 (red). Scale bar = 25µm. Representative of 2 independent animals. k, Representative image of the meningeal lymphatic vessels adjacent to the transverse sinus of Prox1GFP mice. Arrows point toward sprouting/extensions along the lymphatic vessels. Scale bar = 350 µm. Representative of 5 mice. l, Quantification of the length of lymphatics and number of lymphatic extensions in adjacent sections of meningeal lymphatics associated with the transverse sinus starting from the pineal gland (mean ± s.e.m.; n = 5 mice; n = 2 transverse sinus/mouse).
Figure 2:. Meningeal T cells migrate into…
Figure 2:. Meningeal T cells migrate into the cervical lymph nodes in a CCR7 dependent manner.
a, Scheme of the experiments in (b-d). C57Bl6 mice were reconstituted with bone marrow from KiKGR mice after irradiation. Ten weeks after reconstitution, meninges were converted for 2 min with a violet light (through the intact skull) every twelve hours for 3 days. Ten hours after the last conversion, tissues were harvested and analyzed by FACS. b, Representative density plot of converted T cells (KiKR+) in the meninges and deep cervical (dCLN) of control and converted mice. c, Quantification of the percentage of converted CD4 T cells (KiKR+) in the meninges, blood and nasal mucosa of control and converted mice (mean ± s.e.m.; F(1,26)=388.8; 2-way ANOVA with Sidak’s multiple comparisons test). d, Quantification of the percentage of KiKR+ CD4 T cells in the dCLN, sCLN and ILN of control and converted mice (mean ± s.e.m.; F(1,33)=4.862; mice pooled from 2 independent experiments, 2-way ANOVA with Sidak’s multiple comparisons test). e, Representative images of i.c.m. injected naïve T cells in the dCLN of mice at 6 and 12h post injection. Scale bar = 200 µm, 50µm (insets). f, Quantification of the density of naïve T cells per mm2 of dCLN, sCLN, brachial, and ILN at different time points post injection (mean ± s.e.m.; n=2–7 mice per group pooled from 2 independent experiments). g, Representative images of CCR7-WT (red) and CCR7-KO (green) CD4 T cells in the dCLN 12h post injection. Scale bar = 200 µm, 75µm (inset). h, i, Quantification of the density of CCR7-WT and CCR7-KO cells per mm2 of total lymph nodes (h) or per T cell zone (i) at 12h post injection (mean ± s.e.m.; t=3.687 df=6 (h) t=5.586 df=6 (i); pooled from 2 independent experiments; two-tailed paired t-test). j, Representative dot plot of GFP expression by CD4 T cells in the meninges of C57Bl6 mice and CCR7GFP mice. Representative of 3 independent mice. k, Representative contour plot of phenotype of CCR7+ and CCR7-CD4 T cells in the meninges of CCR7GFP mice. Representative of 3 independent mice. l, Representative images of CCR7 expression (CCR7GFP – green) in and around the meningeal lymphatics (Lyve-1 – white and CCL21 – red) along the transverse and superior sagittal sinuses. Arrows point to leukocyte-shaped cells expressing CCR7 located inside of the meningeal lymphatics. Scale bar = 120 µm. CCR7 expressing T cells (CD3e – red) are found in the meningeal lymphatics (Lyve-1 – grey). Scale bar = 40 µm, 15µm (insets). Representative of 3 independent mice. m, Representative dot plots of meningeal T cells (both CD4 and CD8) in the meninges of adult CCR7-WT and CCR7-KO mice. n, Quantification of the number of total, CD4 effector and CD8 T cells in the meninges of CCR7-WT and CCR7-KO mice (mean ± s.e.m.; representative of 2 independent experiments; t=5.744 df=7 (T Cells), t=2.897 df=7 (CD4 Eff), t=6.961 df=7 (CD8); two-tailed unpaired t-test). o, Representative images of T cells (CD3e – red) in and around the meningeal lymphatics (Lyve-1 – blue) of the superior sagittal sinus in CCR7-WT and CCR7-KO mice. Scale bar = 120 µm, 15µm (inset). p, Quantification of the density of T cells in the proximity of the sinuses of CCR7-WT and CCR7-KO mice (top panel) and percentage of T cells localized inside of the lymphatics (bottom panel) (mean ± s.e.m.; t=5.998 df=4 (density), t=4.633 df=4 (percentage); two-tailed unpaired t-test).
Figure 3:. Meningeal lymphatics as the main…
Figure 3:. Meningeal lymphatics as the main route for immune cell and macromolecule drainage from the CSF.
a, Representative images of exogenously injected T cells (CellTracker Deep Red Dye – red) in the dCLN at 12h after i.c.m. injection in sham-operate or ligated mice (24h post-surgery). Scale bar = 150 µm. b, Quantification of the density of T cells per mm2 of dCLN and sCLN in sham-operate or ligated mice (mean ± s.e.m.; F(1,14)=7.676; two-way ANOVA with Sidak’s multiple comparison test). c, Representative images of exogenously injected fluorescent microbeads (0.5µm in diameter – green) in the dCLN of sham-operated or ligated mice. Scale bar = 150 µm. d, Quantification of the percentage of bead coverage in the dCLN of sham-operated or ligated mice ((5µl of beads were injected); expressed as percentage of the control condition; mean ± s.e.m.; t=4.603 df=8; two-tailed unpaired t-test). e, Quantification of the size of the dCLNs of sham-operated or ligated mice (mean ± s.e.m). f, Representative contour plot of T cells in the meninges of sham-operated or ligated mice. Quantification of the number of T cells (TCRb+) in the meninges sham-operated or ligated mice (mean ± s.e.m.; representative of 2 independent experiment; t=3.813 df=8; two-tailed unpaired t-test). g, Representative images of i.c.m. injected T cells (Deep Red Cell Tracker – red) in the lymphatics of the cribriform plate (a) and in and around the lymphatics at the base of the nose (b and c). White arrowheads point to intra-lymphatic T cells while yellow arrowheads point to peri-lymphatic T cells. Scale bar = 1000 µm, 50µm (insets). Representative of 2 independent mice. h, Representative images of the cribriform plate region after 2 and 12h post i.c.m. injection of CFSE-labeled T cells (green). Arrowhead points to a CFSE-labeled T cell localized on the nasal side of the cribriform plate. Scale bar = 230 µm. Representative of 4 independent mice. i, Quantification of the number of exogenously injected T cells in the meninges (grey curve), in the meningeal lymphatics (orange curve), and in the nasal mucosa (green curve) of mice at different time post i.c.m. injection (mean ± s.e.m.; n=2–8 mice per group, pooled from 2 independent experiments). j, Representative images of the meningeal lymphatics (Lyve-1 – red) and blood (CD31 – blue) vasculature of laser alone, Visudyne (i.c.m.) alone and Visudyne (i.c.m.) + laser treated mice 4 days after photoconversion. Scale bar = 1000 µm, 400µm (insets). k, l, Quantification of the Lyve-1 (k) and CD31 (l) coverage on the superior sagittal and transverse sinuses of laser alone, Visudyne (i.c.m.) alone and Visudyne (i.c.m.) + laser treated mice 4 days after photoconversion (mean ± s.e.m.; representative of 3 independent experiments; F(2,18)=10.67; two-way ANOVA with Sidak’s multiple comparison test). m, Representative images of the nasal lymphatics (Prox1GFP – green, Lyve1 – red) 24h after laser or intranasal (i.n.) injection of Visudyne. The inset illustrates the lymphatic bundle at the base of the skull that is ablated after Visudyne treatment. Scale bar = 500 µm, 200µm (insets). Representative of 6 mice/group from 2 independent experiments. n, o, Representative images of i.c.m. injected T cells (CFSE-labeled, green) in the dCLNs (n) or sCLNs (o) of laser alone, Visudyne alone, Visudyne i.c.m. + laser, and Visudyne (i.n.) + laser treated mice 12h after i.c.m. injection. Scale bar = 150µm, 30µm (insets) (n), 300µm, 50µm (insets) (o). p, Quantification of the density of T cells per mm2 of dCLNs (i) and size of dCLNs (ii) in the laser alone, Visudyne alone, Visudyne i.c.m. + laser, and Visudyne (i.n.) + laser treated mice 12h after i.c.m. injection (mean ± s.e.m.; pooled from 4 independent experiments (i) pooled from 3 independent experiments (ii); Kruskal-Wallis test with Dunn’s multiple comparison test). q, Quantification of the density of T cells per mm2 of sCLNs (i) and sCLNs size (ii) in the laser alone, Visudyne alone, Visudyne i.c.m. + laser, and Visudyne (i.n.) + laser treated mice 12h after i.c.m. injection (mean ± s.e.m.; pooled from 2 independent experiment (i) and from a single experiment (ii); F(3,49)=4.282; one-way ANOVA with Sidak’s multiple comparison test). r, Representative images of CSF-injected beads drained to the dCLNs in Visudyne (i.n.) + laser and Visudyne (i.c.m.) + laser treated mice at 2h after injection. Scale bar = 150µm, 35µm (insets). s, Quantification of the percentage of bead coverage of the dCLNs of laser, Visudyne (i.c.m.) + laser and Visudyne (i.n.) + laser treated mice at 2h after CSF injection (mean ± s.e.m.; F(2,23)=9.122; one-way ANOVA with Tukey’s multiple comparisons test). t, Scheme of the proposed model of the route of drainage into the cervical lymph nodes. u, Representative contour plot of T cells in the meninges of laser and Visudyne (i.c.m.) + laser treated mice at 7 days post ablation. Quantification of the number of T cells in the meninges of laser and Visudyne (i.c.m.) + laser treated mice (mean ± s.e.m.; pooled from 3 independent experiments; t=3.939 df=23; two-tailed unpaired t-test).
Figure 4:. Transcriptomic analysis of the meningeal…
Figure 4:. Transcriptomic analysis of the meningeal lymphatic endothelial cells.
a, Principal component analysis of the transcriptome of the lymphatic endothelial cells (LEC) of the diaphragm (blue), meninges (green) and skin (red – ear) (n=3 biological replicate per group each pooled from 10 individual mice). b, Upset plot showing the set intersection between the differentially up-regulated genes in meningeal, diaphragm, and skin LEC. The orange bar represents the 304 up-regulated genes in the meningeal LEC compared to the diaphragm and skin LEC. c, Heat map of the significantly up– and down– regulated genes in the meningeal LEC compared to the diaphragm and skin LEC. d, Heat map of the relative expression of the LEC-related genes in the meningeal, diaphragm, and skin LEC. e, Representation of significantly up– and down– regulated gene pathways in the meningeal LEC compared to diaphragm and skin LEC (Fisher’s exact test). f, Quantification of the normalized counts for Sema3a, Ephb2, Thsp1, and Klf4 in the diaphragm, meningeal and skin LEC. Genes were selected from the significantly up-or down-regulated genes (mean ± s.e.m.). g, Quantification of the normalized counts from diaphragm, meninges and skin LECs for genes previously shown to be down-or up-regulated by LECs when cultured on high stiffness surfaces (mean ± s.e.m.; F(2,42)=12.14; two-way ANOVA with Tukey’s multiple comparison test).
Figure 5:. Ablation of lymphatic drainage modulates…
Figure 5:. Ablation of lymphatic drainage modulates T cell activation and ameliorates disease development.
a, EAE clinical symptoms development in laser, Visudyne (i.n.) + laser, and Visudyne (i.c.m.) + laser treated mice (on the day of EAE induction; mean ± s.e.m.; pooled from 3 independent experiments; F(2,79)=23.07; repeated measures two-way ANOVA with Tukey’s multiple comparisons test). b, Incidence of EAE development (the day mice reach a score of 1 or above) in laser, Visudyne (i.n.) + laser, and Visudyne (i.c.m.) + laser treated mice (pooled from 3 independent experiments, Log-rank (Mantel-Cox) test). c, Representative dot plots of CD4 and CD8 T cells in the spinal cord, brain meninges and spinal cord meninges of laser and Visudyne (i.c.m.) + laser mice during late onset EAE (D17). d, Quantification of the number of CD4 and CD8 T cells in the spinal cord, brain meninges and spinal cord meninges of laser and Visudyne (i.c.m.) + laser treated mice at D17 post immunization (mean ± s.e.m.; pooled from 2 independent experiments; F(1,26)=5.5990 (Brain Meninges), F(1,26)=10.91; two-way ANOVA with Sidak’s multiple comparisons test). e-h, Adult wild-type mice were injected i.v. with 1:1 ratio of 2D2TdTOMATO and OTIIGFP T cells (4 millions total). One day after injection, mice were laser or Visudyne (i.c.m.) + laser treated and EAE was induced by immunization with MOG35–55. The dCLNs were harvested at day 8 of EAE development and the interaction of the 2D2 and OTII T cells with CD11c+ cells were analyzed. e, Representative images of MOG-specific T cells (2D2 – red) and OVA-specific T cells (OTII - green) in the dCLNs of laser and Visudyne (i.c.m.) + laser treated mice at D8 post EAE induction. Green arrowheads points to OTII T cells. Yellow arrowheads points to 2D2 non in contact with a CD11c+ cells, and white arrowheads points to 2D2 in contact with a CD11c+ cells. Scale bar = 150µm, 25µm (insets). Representative of 5 independent mice per group. f, Representative images and associated profile plot of a MOG-specific T cells (2D2 – red) in close contact (upper panel) or not (lower panel) to a CD11c+ expressing cells (cyan) in the dCLNs of laser and Visudyne (i.c.m.) + laser treated mice at D8 post EAE induction. Scale bar = 10µm. Representative of 5 independent mice per group. g, Quantification of the density of OTII and 2D2 T cells in the dCLNs of laser and Visudyne (i.c.m.) + laser treated mice at D8 post EAE induction (mean ± s.e.m.). h, Quantification of the percentage of OTII and 2D2 T cells in contact with a CD11c+ cells in the dCLNs of laser and Visudyne (i.c.m.) + laser treated mice at D8 post EAE induction (mean ± s.e.m.; F(1,8)=4.204; two-way ANOVA with Sidak’s multiple comparisons test). i, Heat map of the significantly up-and down-regulated genes in the CD44+ 2D2 T cells obtained from dCLN and spleen of Visudyne (i.c.m.) + laser vs. control laser treated mice (Fisher’s exact test with p value > 0.05). j, Volcano plot of the significantly up– and down– regulated genes in the CD44+ 2D2 T cells obtained from dCLN of Visudyne (i.c.m.) + laser vs. control laser treated mice (n=3 samples per group; P values were corrected for multiple hypothesis testing with the Benjamini-Hochberg false-discovery rate procedure). k, Representation of some of the significantly enriched pathways in the CD44+ 2D2 T cells obtained from dCLN of Visudyne (i.c.m.) + laser vs. control laser treated mice (P values were corrected for multiple hypothesis testing with the Benjamini-Hochberg false-discovery rate procedure). l, Dot plot analysis of the active miRNA in the CD44+ 2D2 T cells obtained from dCLN of the Visudyne (i.c.m.) + laser vs. control laser treated mice (data was analyzed using the hypergeometric distribution with a significance at p<0.05).

References

    1. Louveau A et al. Structural and functional features of central nervous system lymphatic vessels. Nature 523, 337–341, doi:10.1038/nature14432 (2015).
    1. Kida S, Pantazis A & Weller RO CSF drains directly from the subarachnoid space into nasal lymphatics in the rat. Anatomy, histology and immunological significance. Neuropathol Appl Neurobiol 19, 480–488 (1993).
    1. Cserr HF, Harling-Berg CJ & Knopf PM Drainage of brain extracellular fluid into blood and deep cervical lymph and its immunological significance. Brain Pathol 2, 269–276 (1992).
    1. Iliff JJ et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med 4, 147ra111, doi:10.1126/scitranslmed.3003748 (2012).
    1. Go KG, Houthoff HJ, Hartsuiker J, Blaauw EH & Havinga P Fluid secretion in arachnoid cysts as a clue to cerebrospinal fluid absorption at the arachnoid granulation. J Neurosurg 65, 642–648, doi:10.3171/jns.1986.65.5.0642 (1986).
    1. Ma Q, Ineichen BV, Detmar M & Proulx ST Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat Commun 8, 1434, doi:10.1038/s41467-017-01484-6 (2017).
    1. Kipnis J Multifaceted interactions between adaptive immunity and the central nervous system. Science 353, 766–771, doi:10.1126/science.aag2638 (2016).
    1. Aspelund A et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J Exp Med 212, 991–999, doi:10.1084/jem.20142290 (2015).
    1. Engelhardt B, Vajkoczy P & Weller RO The movers and shapers in immune privilege of the CNS. Nat Immunol 18, 123–131, doi:10.1038/ni.3666 (2017).
    1. Louveau A et al. Understanding the functions and relationships of the glymphatic system and meningeal lymphatics. J Clin Invest 127, 3210–3219, doi:10.1172/JCI90603 (2017).
    1. Andres KH, von During M, Muszynski K & Schmidt RF Nerve fibres and their terminals of the dura mater encephali of the rat. Anat Embryol (Berl) 175, 289–301 (1987).
    1. Absinta M et al. Human and nonhuman primate meninges harbor lymphatic vessels that can be visualized noninvasively by MRI. Elife 6, doi:10.7554/eLife.29738 (2017).
    1. Kamphorst AO, Guermonprez P, Dudziak D & Nussenzweig MC Route of antigen uptake differentially impacts presentation by dendritic cells and activated monocytes. J Immunol 185, 3426–3435, doi:10.4049/jimmunol.1001205 (2010).
    1. Baluk P et al. Functionally specialized junctions between endothelial cells of lymphatic vessels. J Exp Med 204, 2349–2362, doi:10.1084/jem.20062596 (2007).
    1. Castren E & Antila H Neuronal plasticity and neurotrophic factors in drug responses. Mol Psychiatry 22, 1085–1095, doi:10.1038/mp.2017.61 (2017).
    1. Foldi M et al. New contributions to the anatomical connections of the brain and the lymphatic system. Acta Anat (Basel) 64, 498–505 (1966).
    1. Antila S et al. Development and plasticity of meningeal lymphatic vessels. J Exp Med 214, 3645–3667, doi:10.1084/jem.20170391 (2017).
    1. Hennet T, Hagen FK, Tabak LA & Marth JD T-cell-specific deletion of a polypeptide N-acetylgalactosaminyl-transferase gene by site-directed recombination. Proc Natl Acad Sci U S A 92, 12070–12074 (1995).
    1. Derecki NC et al. Regulation of learning and memory by meningeal immunity: a key role for IL-4. J Exp Med 207, 1067–1080, doi:10.1084/jem.20091419 (2010).
    1. Ziarek JJ et al. Structural basis for chemokine recognition by a G protein-coupled receptor and implications for receptor activation. Sci Signal 10, doi:10.1126/scisignal.aah5756 (2017).
    1. Debes GF et al. Chemokine receptor CCR7 required for T lymphocyte exit from peripheral tissues. Nat Immunol 6, 889–894, doi:10.1038/ni1238 (2005).
    1. Weber M et al. Interstitial dendritic cell guidance by haptotactic chemokine gradients. Science 339, 328–332, doi:10.1126/science.1228456 (2013).
    1. Randolph GJ, Ivanov S, Zinselmeyer BH & Scallan JP The Lymphatic System: Integral Roles in Immunity. Annu Rev Immunol 35, 31–52, doi:10.1146/annurev-immunol-041015-055354 (2017).
    1. Clarkson BD et al. CCR7 deficient inflammatory Dendritic Cells are retained in the Central Nervous System. Sci Rep 7, 42856, doi:10.1038/srep42856 (2017).
    1. Wigle JT & Oliver G Prox1 function is required for the development of the murine lymphatic system. Cell 98, 769–778 (1999).
    1. Harvey NL et al. Lymphatic vascular defects promoted by Prox1 haploinsufficiency cause adult-onset obesity. Nat Genet 37, 1072–1081, doi:10.1038/ng1642 (2005).
    1. Lavado A & Oliver G Prox1 expression patterns in the developing and adult murine brain. Dev Dyn 236, 518–524, doi:10.1002/dvdy.21024 (2007).
    1. Tammela T et al. Photodynamic ablation of lymphatic vessels and intralymphatic cancer cells prevents metastasis. Sci Transl Med 3, 69ra11, doi:10.1126/scitranslmed.3001699 (2011).
    1. Kim H, Kataru RP & Koh GY Inflammation-associated lymphangiogenesis: a double-edged sword? J Clin Invest 124, 936–942, doi:10.1172/JCI71607 (2014).
    1. Dendrou CA, Fugger L & Friese MA Immunopathology of multiple sclerosis. Nat Rev Immunol 15, 545–558, doi:10.1038/nri3871 (2015).
    1. Ajami B et al. Single-cell mass cytometry reveals distinct populations of brain myeloid cells in mouse neuroinflammation and neurodegeneration models. Nat Neurosci 21, 541–551, doi:10.1038/s41593-018-0100-x (2018).
    1. Proescholdt MA, Jacobson S, Tresser N, Oldfield EH & Merrill MJ Vascular endothelial growth factor is expressed in multiple sclerosis plaques and can induce inflammatory lesions in experimental allergic encephalomyelitis rats. J Neuropathol Exp Neurol 61, 914–925 (2002).
    1. Cursiefen C et al. Thrombospondin 1 inhibits inflammatory lymphangiogenesis by CD36 ligation on monocytes. J Exp Med 208, 1083–1092, doi:10.1084/jem.20092277 (2011).
    1. Jurisic G et al. An unexpected role of semaphorin3a-neuropilin-1 signaling in lymphatic vessel maturation and valve formation. Circ Res 111, 426–436, doi:10.1161/CIRCRESAHA.112.269399 (2012).
    1. Makinen T et al. PDZ interaction site in ephrinB2 is required for the remodeling of lymphatic vasculature. Genes Dev 19, 397–410, doi:10.1101/gad.330105 (2005).
    1. Frye M et al. Matrix stiffness controls lymphatic vessel formation through regulation of a GATA2-dependent transcriptional program. Nature Communications 9, doi: ARTN 1511 10.1038/s41467-018-03959-6 (2018).
    1. Yeh YT et al. Matrix Stiffness Regulates Endothelial Cell Proliferation through Septin 9. Plos One 7, doi:ARTN e46889 10.1371/journal.pone.0046889 (2012).
    1. Furtado GC et al. Swift entry of myelin-specific T lymphocytes into the central nervous system in spontaneous autoimmune encephalomyelitis. J Immunol 181, 4648–4655 (2008).
    1. Phillips MJ, Needham M & Weller RO Role of cervical lymph nodes in autoimmune encephalomyelitis in the Lewis rat. J Pathol 182, 457–464, doi:10.1002/(SICI)1096-9896(199708)182:4<457::AID-PATH870>;2-Y (1997).
    1. van Zwam M et al. Surgical excision of CNS-draining lymph nodes reduces relapse severity in chronic-relapsing experimental autoimmune encephalomyelitis. J Pathol 217, 543–551, doi:10.1002/path.2476 (2009).
    1. Rottman JB et al. Leukocyte recruitment during onset of experimental allergic encephalomyelitis is CCR1 dependent. Eur J Immunol 30, 2372–2377, doi:10.1002/1521-4141(2000)30:8<2372::AID-IMMU2372>;2-D (2000).
    1. Guan YJ et al. Phospho-SXXE/D motif mediated TNF receptor 1-TRADD death domain complex formation for T cell activation and migration. J Immunol 187, 1289–1297, doi:10.4049/jimmunol.1003399 (2011).
    1. Wu X, Lahiri A, Haines GK 3rd, Flavell RA & Abraham C NOD2 regulates CXCR3-dependent CD8+ T cell accumulation in intestinal tissues with acute injury. J Immunol 192, 3409–3418, doi:10.4049/jimmunol.1302436 (2014).
    1. Jeker LT & Bluestone JA MicroRNA regulation of T-cell differentiation and function. Immunol Rev 253, 65–81, doi:10.1111/imr.12061 (2013).
    1. Brinker T, Stopa E, Morrison J & Klinge P A new look at cerebrospinal fluid circulation. Fluids Barriers CNS 11, 10, doi:10.1186/2045-8118-11-10 (2014).
    1. Cai R, P. C, Ghasemigharagoz A, Torodov M,I, Foerstera B, Zhao S, Bhatia H,S, Mrowka L, Theodorou D, Rempfler M, Xavier A, Kress B,T, Benakis C, Liesz A, Menze B, Kerschensteiner M, Nedergaard M, Eturk A. Panoptic vDISCO imaging reveals neuronal connectivity, remote trauma effects and meningeal vessels in intact transparent mice. bioRxiv, doi:10.1101/374785 (2018).
    1. Mollanji R, Bozanovic-Sosic R, Zakharov A, Makarian L & Johnston MG Blocking cerebrospinal fluid absorption through the cribriform plate increases resting intracranial pressure. Am J Physiol Regul Integr Comp Physiol 282, R1593–1599, doi:10.1152/ajpregu.00695.2001 (2002).
    1. Odoardi F et al. T cells become licensed in the lung to enter the central nervous system. Nature 488, 675–679, doi:10.1038/nature11337 (2012).
    1. Mora JR et al. Reciprocal and dynamic control of CD8 T cell homing by dendritic cells from skin-and gut-associated lymphoid tissues. J Exp Med 201, 303–316, doi:10.1084/jem.20041645 (2005).
    1. Zozulya AL et al. Intracerebral dendritic cells critically modulate encephalitogenic versus regulatory immune responses in the CNS. J Neurosci 29, 140–152, doi:10.1523/JNEUROSCI.2199-08.2009 (2009).
    1. Kilarski WW et al. Optimization and regeneration kinetics of lymphatic-specific photodynamic therapy in the mouse dermis. Angiogenesis 17, 347–357, doi:10.1007/s10456-013-9365-6 (2014).
    1. Wachowska M et al. Investigation of cell death mechanisms in human lymphatic endothelial cells undergoing photodynamic therapy. Photodiagnosis Photodyn Ther 14, 57–65, doi:10.1016/j.pdpdt.2016.02.004 (2016).
    1. Ensari S et al. Venous outflow of the brain after bilateral complete jugular ligation. Turk Neurosurg 18, 56–60 (2008).
    1. Kawajiri H, Furuse M, Namba R, Kotani J & Oka T Effect of internal jugular vein ligation on resorption of cerebrospinal fluid. J Maxillofac Surg 11, 42–45 (1983).
    1. Vogh BP, Godman DR & Maren TH Effect of AlCl3 and other acids on cerebrospinal fluid production: a correction. J Pharmacol Exp Ther 243, 35–39 (1987).
    1. Ning B et al. Ultrasound-aided Multi-parametric Photoacoustic Microscopy of the Mouse Brain. Sci Rep 5, 18775, doi:10.1038/srep18775 (2015).
    1. Coles JA et al. Intravital imaging of a massive lymphocyte response in the cortical dura of mice after peripheral infection by trypanosomes. PLoS Negl Trop Dis 9, e0003714, doi:10.1371/journal.pntd.0003714 (2015).
    1. Nowotschin S & Hadjantonakis AK Use of KikGR a photoconvertible green-to-red fluorescent protein for cell labeling and lineage analysis in ES cells and mouse embryos. BMC Dev Biol 9, 49, doi:10.1186/1471-213X-9-49 (2009).
    1. Oshio K, Watanabe H, Song Y, Verkman AS & Manley GT Reduced cerebrospinal fluid production and intracranial pressure in mice lacking choroid plexus water channel Aquaporin-1. FASEB J 19, 76–78, doi:10.1096/fj.04-1711fje (2005).
    1. Cyster JG & Goodnow CC Pertussis toxin inhibits migration of B and T lymphocytes into splenic white pulp cords. J Exp Med 182, 581–586 (1995).
    1. Da Mesquita S et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature, doi:10.1038/s41586-018-0368-8 (2018).

Source: PubMed

3
Iratkozz fel