Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Meningeal lymphatic vessels mediate neurotropic viral drainage from the central nervous system

Abstract

Recent studies have demonstrated that brain meningeal lymphatic vessels (MLVs) act as a drainage path directly into the cervical lymph nodes (CLNs) for macromolecules contained in the cerebrospinal fluid (CSF). However, the role of MLVs during CNS viral infection remains unexplored. Here, we found that infection with several neurotropic viruses in mice promotes MLV expansion but also causes impaired MLV-mediated drainage of macromolecules. Notably, MLVs could drain virus from the CNS to CLNs. Surgical ligation of the lymph vessels or photodynamic ablation of dorsal MLVs increased neurological damage and mortality of virus-infected mice. By contrast, pretreatment with vascular endothelial growth factor C promoted expansion of functional MLVs and alleviated the effects of viral infection. Together, these data indicate that functional MLVs facilitate virus clearance, and MLVs represent a critical path for virus spreading from the CNS to the CLNs. MLV-based therapeutic strategies may thus be useful for alleviating infection-induced neurological damage.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Viral CNS infections induce dorsal meningeal lymphangiogenesis.
Fig. 2: Three-dimensional imaging of meningeal lymphangiogenesis after viral infection.
Fig. 3: Impaired MLV ability to drain biological molecules following acute viral CNS infection.
Fig. 4: Virus infection of the CNS causes MLV damage.
Fig. 5: MLVs drain neurotropic viral particles from the CNS to CLNs.
Fig. 6: Ablation of lymphatic drainage enhances, but VEGF-C pretreatment reduces, mortality of JEV-infected mice.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available in this manuscript and the Supplementary Information. Source data are provided with this paper.

Code availability

Customized code or algorithms were not used to generate results in the present study.

References

  1. Ahn, J. H. et al. Meningeal lymphatic vessels at the skull base drain cerebrospinal fluid. Nature 572, 62–66 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Louveau, A. et al. Structural and functional features of central nervous system lymphatic vessels. Nature 523, 337–341 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Aspelund, A. et al. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J. Exp. Med. 212, 991–999 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Louveau, A. et al. CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat. Neurosci. 21, 1380–1391 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Louveau, A., Da Mesquita, S. & Kipnis, J. Lymphatics in neurological disorders: a neuro–lympho–vascular component of multiple sclerosis and Alzheimer’s disease? Neuron 91, 957–973 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Da Mesquita, S. et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature 560, 185–191 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Song, E. et al. VEGF-C-driven lymphatic drainage enables immunosurveillance of brain tumours. Nature 577, 689–694 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Hu, X. T. et al. Meningeal lymphatic vessels regulate brain tumor drainage and immunity. Cell Res. 30, 229–243 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Hsu, M. et al. Neuroinflammation-induced lymphangiogenesis near the cribriform plate contributes to drainage of CNS-derived antigens and immune cells. Nat. Commun. 10, 229 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Silva, M. T. T. Viral encephalitis. Arq. Neuropsiquiatr. 71, 703–709 (2013).

    Article  PubMed  Google Scholar 

  11. Solomon, T. Flavivirus encephalitis—reply. N. Engl. J. Med. 351, 1804–1804 (2004).

    Article  CAS  Google Scholar 

  12. Tyler, K. L. Herpes simplex virus infections of the central nervous system: encephalitis and meningitis, including Mollaret’s. Herpes 11, 57A-64A (2004).

    PubMed  Google Scholar 

  13. Moseman, E. A., Blanchard, A. C., Nayak, D. & McGavern, D. B. T cell engagement of cross-presenting microglia protects the brain from a nasal virus infection. Sci Immunol. 5, eabb1817 (2020).

  14. Patabendige, A., Michael, B. D., Craig, A. G. & Solomon, T. Brain microvascular endothelial–astrocyte cell responses following Japanese encephalitis virus infection in an in vitro human blood–brain barrier model. Mol. Cell Neurosci. 89, 60–70 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mustafa, Y. M., Meuren, L. M., Coelho, S. V. A. & de Arruda, L. B. Pathways exploited by flaviviruses to counteract the blood–brain barrier and invade the central nervous system. Front. Microbiol. 10, 525 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Luo, Z. C. et al. Dual role of Toll-like receptor 7 in the pathogenesis of rabies virus in a mouse model. J. Virol. 94, e00111-20 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  17. Wang, W. et al. Zika virus infection induces host inflammatory responses by facilitating NLRP3 inflammasome assembly and interleukin-1β secretion. Nat. Commun. 9, 106 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Renier, N. et al. Mapping of brain activity by automated volume analysis of immediate early genes. Cell 165, 1789–1802 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Liu, T. et al. Local sympathetic innervations modulate the lung innate immune responses. Sci. Adv. 6, eaay1497 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cao, Y., Wang, H. & Zeng, W. Whole-tissue 3D imaging reveals intra-adipose sympathetic plasticity regulated by NGF–TrkA signal in cold-induced beiging. Protein Cell 9, 527–539 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lutter, S., Xie, S., Tatin, F. & Makinen, T. Smooth muscle-endothelial cell communication activates Reelin signaling and regulates lymphatic vessel formation. J. Cell Biol. 197, 837–849 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wang, Y. et al. Molecular and cellular basis of the regulation of lymphatic contractility and lymphatic absorption. Int. J. Biochem. Cell Biol. 53, 134–140 (2014).

    Article  CAS  PubMed  Google Scholar 

  23. Muthuchamy, M. & Zawieja, D. Molecular regulation of lymphatic contractility. Ann. N. Y. Acad. Sci. 1131, 89–99 (2008).

    Article  PubMed  Google Scholar 

  24. Petrova, T. V. et al. Defective valves and abnormal mural cell recruitment underlie lymphatic vascular failure in lymphedema distichiasis. Nat. Med. 10, 974–981 (2004).

    Article  CAS  PubMed  Google Scholar 

  25. Sabine, A. et al. Mechanotransduction, PROX1, and FOXC2 cooperate to control connexin37 and calcineurin during lymphatic-valve formation. Dev. Cell 22, 430–445 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Norrmen, C. et al. FOXC2 controls formation and maturation of lymphatic collecting vessels through cooperation with NFATc1. J. Cell Biol. 185, 439–457 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Srinivasan, R. S. & Oliver, G. Prox1 dosage controls the number of lymphatic endothelial cell progenitors and the formation of the lymphovenous valves. Gene Dev. 25, 2187–2197 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Kazenwadel, J. et al. GATA2 is required for lymphatic vessel valve development and maintenance. J. Clin. Invest. 125, 2979–2994 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Kazenwadel, J. et al. Loss-of-function germline GATA2 mutations in patients with MDS/AML or MonoMAC syndrome and primary lymphedema reveal a key role for GATA2 in the lymphatic vasculature. Blood 119, 1283–1291 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Tatin, F. et al. Planar cell polarity protein Celsr1 regulates endothelial adherens junctions and directed cell rearrangements during valve morphogenesis. Dev. Cell 26, 31–44 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Alders, M. et al. Hennekam syndrome can be caused by FAT4 mutations and be allelic to Van Maldergem syndrome. Hum. Genet. 133, 1161–1167 (2014).

    Article  CAS  PubMed  Google Scholar 

  32. Betterman, K. L. et al. Atypical cadherin FAT4 orchestrates lymphatic endothelial cell polarity in response to flow. J. Clin. Invest. 130, 3315–3328 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Coxam, B. et al. Pkd1 regulates lymphatic vascular morphogenesis during development. Cell Rep. 7, 623–633 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Yoo, J. et al. Opposing regulation of PROX1 by interleukin-3 receptor and NOTCH directs differential host cell fate reprogramming by Kaposi sarcoma herpes virus. PLoS Pathog. 8, e1002770 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Gramolelli, S. et al. Oncogenic herpesvirus engages endothelial transcription factors SOX18 and PROX1 to increase viral genome copies and virus production. Cancer Res. 80, 3116–3129 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Xiao, J. et al. Targeting 7-dehydrocholesterol reductase integrates cholesterol metabolism and IRF3 activation to eliminate infection. Immunity 52, 109–122 (2020).

    Article  CAS  PubMed  Google Scholar 

  37. Zhang, C. et al. Targeting lysophospholipid acid receptor 1 and ROCK kinases promotes antiviral innate immunity. Sci. Adv. 7, eabb5933 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. YanboZhang et al. Activation of vascular endothelial growth factor receptor-3 in macrophages restrains TLR4–NF-κB signaling and protects against endotoxin shock. Immunity 40, 501–514 (2014).

    Article  CAS  Google Scholar 

  39. Ma, L. et al. FLT4/VEGFR3 activates AMPK to coordinate glycometabolic reprogramming with autophagy and inflammasome activation for bacterial elimination. Autophagy 2021, 1–16, (2021).

  40. Cle, M. et al. Zika virus infection promotes local inflammation, cell adhesion molecule upregulation, and leukocyte recruitment at the blood–brain barrier. mBio 11, e01183-20 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  41. Carvalho, G. C. D. et al. RAGE and CCR7 mediate the transmigration of Zika-infected monocytes through the blood–brain barrier. Immunobiology 224, 792–803 (2019).

    Article  PubMed  CAS  Google Scholar 

  42. Weller, R. O., Galea, I., Carare, R. O. & Minagar, A. Pathophysiology of the lymphatic drainage of the central nervous system: implications for pathogenesis and therapy of multiple sclerosis. Pathophysiology 17, 295–306 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Raper, D., Louveau, A. & Kipnis, J. How do meningeal lymphatic vessels drain the CNS? Trends Neurosci. 39, 581–586 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Noé, F. M. & Marchi, N. Central nervous system lymphatic unit, immunity, and epilepsy: is there a link? Epilepsia Open 4, 30–39 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  45. Brioschi, S. et al. Heterogeneity of meningeal B cells reveals a lymphopoietic niche at the CNS borders. Science 373, eabf9277 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Shen, T., Liu, K., Miao, D. N., Cao, R. B. & Chen, P. Y. Effective inhibition of Japanese encephalitis virus replication by shRNAs targeting various viral genes in vitro and in vivo. Virology 454, 48–59 (2014).

    Article  PubMed  CAS  Google Scholar 

  47. Chen, J. M. et al. Meningeal lymphatics clear erythrocytes that arise from subarachnoid hemorrhage. Nat. Commun. 11, 3159 (2020).

Download references

Acknowledgements

This work was supported by grants from the National Natural Science Foundation of China (81630043 to B.W., 81825011 to H.W., 81930038 to H.W. and 81961160738 to B.W.), the Ministry of Science and Technology of China (2018YFA0800702 to H.W.), the Strategic Priority Research Program of the Chinese Academy of Sciences (XDB19030200 to H.W.), Shanghai Municipal Science and Technology Major Project and Key National Science and Technology Programs in Shanghai (218014204 to B.W.). We thank the Core Facility and Technical Support in the Shanghai University and Wuhan Institute of Virology, CAS. We thank the Core Facility of Molecular Biology and Core Facility of Cell Biology in the Shanghai Institute of Biochemistry and Cell Biology for their technical support and help. We thank C. Gu of Huazhong Agricultural University for providing anti-JEV E. We thank M. Jin of Huazhong Agricultural University for providing VSV–GFP. We thank the laboratory of Z. Qian of Institute Pasteur of Shanghai, CAS, for providing HSV-1. We thank the laboratory of H. Wang of the Wuhan Institute of Virology, CAS, for providing ZIKV. We thank Q. Leng of the Institute Pasteur of Shanghai, CAS, for providing IFN receptor-deficient mice. We thank Q. Liang of Shanghai University of Traditional Chinese Medicine for providing the non-thermal 689-nm laser. We thank S. Cao of Huazhong Agricultural University for providing the BHK-21 cell line. We thank H. Wang of Shanghai Institute of Biochemistry and Cell Biology, CAS, for providing the 293T cell line. We thank H. Yan of Wuhan Institute of Virology, CAS, for providing the C6/36 cell line.

Author information

Authors and Affiliations

Authors

Contributions

X.L. and L.Q. performed most of the experiments and statistical analysis. D.Y. performed the two-photon imaging. F.Z. and S.H. performed hLECs experiments. S.H. and X.Z. participated in animal experiments. Y.S. performed volume fluorescence imaging. C.C. performed RABV infection. B.W. and H.W. conceptualized and designed experiments. J. Ye., L.Z., J. Yang and S.C. helped with technical support. X.L., L.Q., B.W., D.M.A. and H.W. wrote the paper. All authors approved the final version of the manuscript.

Corresponding authors

Correspondence to Hongyan Wang or Bin Wei.

Ethics declarations

Competing interests

The authors declare no competing interest.

Peer review

Peer review information

Nature Neuroscience thanks Robyn Klein and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 JEV and VSV infections induce dorsal meningeal lymphangiogenesis.

(a-f). C57BL/6 mice were intravenously (i.v.) or intracerebrally (i.c.) infected with JEV or PBS. a. Representative images of LYVE-1 and VEGFR-3 staining and quantification of the area of LYVE-1+ MLVs in TS at 7 Dpi (n = 5). b. Representative image and quantification of the diameter of LYVE-1+ MLVs along the SSS in JEV (i.v.) infected mice (n = 7 PBS; n = 5 JEV). c. Representative images and quantification of the mean intensity of LYVE-1+ MLVs in COS after JEV (i.c.) infection (n = 7 PBS; n = 6 JEV). d. VEGF-C concentrations in the brain after JEV (i.c.) injection (n = 4 PBS; n = 5 JEV). e. The daily behavior scores of mice (n = 6) and the relative mRNA levels of JEV-C gene (n = 8 PBS; n = 4-8 JEV) in the brain at different time points after JEV (i.v.) infection. f. Representative images and quantification of the area of LYVE-1 staining in COS and TS at day 1, day 3 and day 5 after JEV (i.v.) injection (n = 5). (g-i). C57BL/6 mice were intranasally infected with VSV. g. The daily body weight of mice after VSV infection (n = 6). h. The relative mRNA levels of VSV (n = 6 PBS; n = 5 JEV) and Ccl5 (n = 4 PBS; n = 6 JEV) in the brain at 8 Dpi. i. Representative images and the quantification of meningeal LYVE-1, VEGFR-3, PDPN staining in the TS at 8 Dpi (n = 6). Data are presented as Mean ± SD. ns, not significant (p > 0.05), *p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001, two-tailed, unpaired Student’s t test (a right, b right, c right, d, g, h and i right), one-way ANOVA with Holm-Sidak’s multiple comparisons test (e and f right). Data from at least 3 independent experiments. Detailed statistical information is shown in Supplementary Table 2.

Source data

Extended Data Fig. 2 HSV-1, ZIKV and RABV infections induce dorsal meningeal lymphangiogenesis.

(a-c). Mice were corneally infected with HSV-1. a. Left, the daily body weight of mice (n = 7 PBS; n = 6 HSV-1). Right, mRNA levels of HSV-1 in the brain at 8 Dpi (n = 6 PBS; n = 5 HSV-1). b. Representative meningeal LYVE-1 staining and quantification of the area and diameter of LYVE-1+ MLVs in COS or TS (n = 6). c. mRNA levels of Ccl5 in the brain (n = 4 PBS; n = 5 HSV-1). (d-f). Mice were intramuscularly infected with RABV. Representative meningeal LYVE-1 staining (d left, e left) and quantification of lymphatic branches (white arrowheads) (d right, e right) in TS (n = 6 PBS; n = 8 RABV). f. mRNA levels of RABV (n = 5 PBS; n = 9 RABV), Ccl5, Il1b and Il10 (n = 4 PBS; n = 7 RABV) in the brain at 8 Dpi. (g-j). AG6 mice were intraperitoneally infected with ZIKV. g. The daily body weight of mice (n = 5 PBS; n = 8 ZIKV). h. Viral titers were detected in the brain at 8 Dpi (n = 7). i. Representative images and quantification of the area and diameter of LYVE-1+ MLVs in COS or TS (n = 6 or 7 PBS; n = 6 or 7 ZIKV). j. mRNA levels of Il1b and Il10 in the brain at 8 dpi (n = 4 PBS; n = 5 ZIKV). k. Mice were intravenously infected with JEV or intramuscularly infected with RABV. Representative images and quantification of PDPN+ basal MLVs of JEV-infected mice at 7 dpi (n = 5). Representative images and quantification of LYVE-1+ basal MLVs of RABV-infected mice at 8 dpi (n = 4 PBS; n = 8 RABV). l. mRNA levels of PDPN, FOS, JUN and BCL-2 in hLECs treated with 0.1 MOI JEV, inactivated JEV or untreated for 72 h (n = 3). Data are presented as Mean ± SD. not significant (p > 0.05), *p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001, a two-tailed, unpaired Student’s t test (a, b right, c, d right, e right, f, g, h, i right, j, k), one-way ANOVA with Holm-Sidak’s multiple comparisons test (l). Data from at least 2 independent experiments. Detailed statistical information is shown in Supplementary Table 2.

Source data

Extended Data Fig. 3 MLVs function to drain biological molecules upon virus infection.

a. Representative images of LYVE-1 staining and OVA-647 in tissue around sCLN and the superficial skin of neck at 180 min after i.c.m. injection of OVA-647. b. Representative images of LYVE-1 staining and OVA-647 in TS of meninges after i.c.m. injection of OVA-647 (2 μg/μl) at 3, 4, and 5 Dpi of JEV. c. The ventral images show the biodistribution of OVA-647 in the mice at the indicated time points after i.c.m. or intranasally injection with OVA-647. (d-f). Mice were corneally infected with HSV-1. d. The daily body weight of mice in PBS group (n = 5), the acute infection group (n = 6;) and the resolution of viral infection group (n = 4). (e, f). The ventral images show the biodistribution of OVA-647 and the retention of OVA-647 in cervical region was quantified at 10 days (e) or 28 days (f) (n = 4). g. Representative images of LYVE-1 staining and quantification of the area of LYVE-1+ MLVs in COS at 28 Dpi (n = 6 PBS; n = 4 HSV-1). (h-j). C57BL/6 mice were (i.v.) infected with JEV or PBS. 5 μl QDot655 was injected in i.c.m. of JEV or PBS-injected mice at 4 Dpi. Change of QDot655 in MLVs was monitored through a thinned skull preparation by multiphoton microscopy system. h. Schematic of QDot655 treatment. i. Representative images and the quantification of Qdot655 in MLVs were captured at different timepoints (n = 6). See also video S3. j. Representative images of Qdot655 and Lectin-488 (labeled blood vessel) were captured by multiphoton microscopy system. See also video S4. The data are presented as Mean ± SEM (d, e, f) or as Mean ± SD (g). ns, not significant (p > 0.05), **p < 0.01 and ****p < 0.0001, a two-tailed, unpaired Student’s t test (g), one-way ANOVA with Holm- Sidak’s multiple comparisons test (d) and two-way ANOVA with Holm- Sidak’s multiple comparisons test (e right, f right, i). Detailed statistical information is shown in Supplementary Table 2.

Source data

Extended Data Fig. 4 The expression of genes that regulated cell polarization of LECs.

a. The relative mRNA levels of Celsr1, Pkd1, Sema3a, Egfl7, Fat4, Nrp1 in meninges of JEV (n = 6) or PBS (n = 5 or 6) injected mice. The data are presented as Mean ± SEM. b. hLECs treated with 0.1 MOI JEV, inactivated JEV or untreated for 72 h. The relative mRNA levels of BAD and CASP9 in hLECs (n = 3). The data are presented as Mean ± SD. ns, not significant (p > 0.05), a two-tailed, unpaired Student’s t test (a) and one-way ANOVA with Holm- Sidak’s multiple comparisons test (b). Detailed statistical information is shown in Supplementary Table 2.

Source data

Extended Data Fig. 5 Meningeal lymphatic vessels drain neurotropic viral particles from CNS to CLNs.

a. Immunohistochemical analysis of JEV E protein in AxLN sections in JEV or PBS (i.c.) injected mice with severe JE cases. b. 5-week-old C57BL/6 mice were ligation of dCLVs, and 7 days later, VSV-GFP was i.c.m. injected. The viral titers of VSV-GFP in dCLNs were measured by plaque assay after 3 h injection (n = 4). c. Representative images of the colocalization of VSV-GFP (green) and LYVE-1+ MLVs in TS and COS at 3 h, 6 h, 9 h after i.c.m. injection of VSV-GFP. d. Representative images of JEV E protein staining in hLECs treated with 1 MOI JEV for 48 h. (e, f). VSV-GFP was injected in i.c.m. of C57BL/6 mice. e. Representative images of DAPI, VSV-GFP, CD11b staining in COS of meninges at 9 h after VSV-GFP injection. f. Representative images of DAPI, VSV-GFP, CD206 staining in COS of meninges at 9 h after VSV-GFP injection. (g-i). FACS analysis of the CD45+ cells, macrophages, monocytes and DCs in sCLNs and AxLNs of mice (i.c.) injected with JEV and PBS. g. Gating strategy employed to identify CD45+ cells, B cells (MHC II+ B220+), macrophages (F4/80+ CD11b+), monocytes (F4/80 Ly6c+ CD11b+) and DCs (CD11C+ MHC II+). (h, i). MFI of CD86 and CD80 in B cells, macrophages, DCs or monocytes in sCLNs (n = 5 PBS; n = 8 JEV) or AxLNs (n = 6 PBS; n = 7 JEV) of JEV-infected mice. Data are presented as mean ± SD. not significant (p > 0.05), *p < 0.05, **p < 0.01, a two-tailed, unpaired Student’s t test (b right, h and i). Data from at least 2 independent experiments. Detailed statistical information is shown in Supplementary Table 2.

Source data

Extended Data Fig. 6 Gating strategy of cells in brain of JEV-infected mice.

The gating strategy of CD45+ cells, neutrophils, macrophages, monocytes, CD8+ T cells, CD4+ T cells, NK cells, microglia and B cells in the brain of JEV-infected mice.

Supplementary information

Supplementary Information

Supplementary Table 1.

Reporting Summary

Supplementary Table 2

Detailed statistical information.

Supplementary Video 1

Three-dimensional reconstruction of meningeal lymphatics along the COS in PBS- or JEV-infected mice.

Supplementary Video 2

Three-dimensional projections of meningeal lymphatics in whole-mount meninges of PBS- or JEV-infected mice.

Supplementary Video 3

Flow of QD in MLVs of PBS- or JEV-infected mice.

Supplementary Video 4

Three-dimensional projections of the MLVs and blood by two-photon microscopy.

Source data

Source Data Fig. 1

Unprocessed western blots, image, statistical source data.

Source Data Fig. 2

Unprocessed image, statistical source data.

Source Data Fig. 3

Unprocessed image, statistical source data.

Source Data Fig. 4

Unprocessed western blots, image, statistical source data.

Source Data Fig. 5

Unprocessed image, statistical source data.

Source Data Fig. 6

Unprocessed image, statistical source data.

Source Data Extended Data Fig. 1

Unprocessed image, statistical source data.

Source Data Extended Data Fig. 2

Unprocessed image, statistical source data.

Source Data Extended Data Fig. 3

Unprocessed image, statistical source data.

Source Data Extended Data Fig. 4

Unprocessed statistical source data.

Source Data Extended Data Fig. 5

Unprocessed image, statistical source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, X., Qi, L., Yang, D. et al. Meningeal lymphatic vessels mediate neurotropic viral drainage from the central nervous system. Nat Neurosci 25, 577–587 (2022). https://doi.org/10.1038/s41593-022-01063-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41593-022-01063-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing