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:

The endothelial protein PLVAP in lymphatics controls the entry of lymphocytes and antigens into lymph nodes

An Erratum to this article was published on 21 April 2015

This article has been updated

Abstract

In the lymphatic sinuses of draining lymph nodes, soluble lymph-borne antigens enter the reticular conduits in a size-selective manner and lymphocytes transmigrate to the parenchyma. The molecular mechanisms that control these processes are unknown. Here we unexpectedly found that PLVAP, a prototypic endothelial protein of blood vessels, was synthesized in the sinus-lining lymphatic endothelial cells covering the distal conduits. In PLVAP-deficient mice, both small antigens and large antigens entered the conduit system, and the transmigration of lymphocytes through the sinus floor was augmented. Mechanistically, the filtering function of the lymphatic sinus endothelium was dependent on diaphragms formed by PLVAP fibrils in transendothelial channels. Thus, in the lymphatic sinus, PLVAP forms a physical sieve that regulates the parenchymal entry of lymphocytes and soluble antigens.

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

Figure 1: Activation of B lymphocytes and T lymphocytes is induced in the lymph nodes of Plvap−/− mice.
Figure 2: PLVAP is expressed in LECs in lymph node sinus.
Figure 3: PLVAP diaphragms overlay the distal terminals of the conduit system in lymph nodes.
Figure 4: PLVAP determines the size-selective entry of antigens into conduits in the PLNs.
Figure 5: PLVAP diaphragms in transendothelial channels regulate the filling of conduits.
Figure 6: PLVAP-dependent migration of lymph-borne lymphocytes through the floor of lymphatic sinus to the PLN parenchyma.
Figure 7: PLVAP is concentrated in low-resistance areas of sinusoidal LECs that support transcellular lymphocyte migration.

Similar content being viewed by others

Change history

  • 20 March 2015

    In the version of this article initially published, the gray box in the key in the left graph in Figure 6a is labeled incorrectly. The correct label should be Plvap–/–. The error has been corrected in the HTML and PDF versions of the article.

References

  1. Girard, J.P., Moussion, C. & Forster, R. HEVs, lymphatics and homeostatic immune cell trafficking in lymph nodes. Nat. Rev. Immunol. 12, 762–773 (2012).

    Article  CAS  PubMed  Google Scholar 

  2. Masopust, D. & Schenkel, J.M. The integration of T cell migration, differentiation and function. Nat. Rev. Immunol. 13, 309–320 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Germain, R.N., Robey, E.A. & Cahalan, M.D. A decade of imaging cellular motility and interaction dynamics in the immune system. Science 336, 1676–1681 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Nourshargh, S., Hordijk, P.L. & Sixt, M. Breaching multiple barriers: leukocyte motility through venular walls and the interstitium. Nat. Rev. Mol. Cell Biol. 11, 366–378 (2011).

    Article  CAS  Google Scholar 

  5. Lämmermann, T. & Sixt, M. The microanatomy of T-cell responses. Immunol. Rev. 221, 26–43 (2008).

    Article  PubMed  Google Scholar 

  6. Gretz, J.E., Norbury, C.C., Anderson, A.O., Proudfoot, A.E.I. & Shaw, S. Lymph-borne chemokines and other low molecular weight molecules reach high endothelial venules via specialized conduits while a functional barrier limits access to the lymphocyte microenvironments in lymph node cortex. J. Exp. Med. 192, 1425–1440 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sixt, M. et al. The conduit system transports soluble antigens from the afferent lymph to resident dendritic cells in the T cell area of the lymph node. Immunity 22, 19–29 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Bajénoff, M. & Germain, R.N. B-cell follicle development remodels the conduit system and allows soluble antigen delivery to follicular dendritic cells. Blood 114, 4989–4997 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Roozendaal, R. et al. Conduits mediate transport of low-molecular-weight antigen to lymph node follicles. Immunity 30, 264–276 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Palframan, R.T. et al. Inflammatory chemokine transport and presentation in HEV: a remote control mechanism for monocyte recruitment to lymph nodes in inflamed tissues. J. Exp. Med. 194, 1361–1373 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Py, B.F. et al. Cochlin produced by follicular dendritic cells promotes antibacterial innate immunity. Immunity 38, 1063–1072 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Itano, A.A. et al. Distinct dendritic cell populations sequentially present antigen to CD4 T cells and stimulate different aspects of cell-mediated immunity. Immunity 19, 47–57 (2003).

    Article  CAS  PubMed  Google Scholar 

  13. Braun, A. et al. Afferent lymph-derived T cells and DCs use different chemokine receptor CCR7-dependent routes for entry into the lymph node and intranodal migration. Nat. Immunol. 12, 879–887 (2011).

    Article  CAS  PubMed  Google Scholar 

  14. Ulvmar, M.H. et al. The atypical chemokine receptor CCRL1 shapes functional CCL21 gradients in lymph nodes. Nat. Immunol. 15, 623–630 (2014).

    Article  CAS  PubMed  Google Scholar 

  15. Hallmann, R., Mayer, D.N., Berg, E.L., Broermann, R. & Butcher, E.C. Novel mouse endothelial cell surface marker is suppressed during differentiation of the blood brain barrier. Dev. Dyn. 202, 325–332 (1995).

    Article  CAS  PubMed  Google Scholar 

  16. Stan, R.V. Endothelial stomatal and fenestral diaphragms in normal vessels and angiogenesis. J. Cell. Mol. Med. 11, 621–643 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Schlingemann, R.O. et al. Monoclonal antibody PAL-E specific for endothelium. Lab. Invest. 52, 71–76 (1985).

    CAS  PubMed  Google Scholar 

  18. Aird, W.C. Phenotypic heterogeneity of the endothelium: I. Structure, function, and mechanisms. Circ. Res. 100, 158–173 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Baluk, P. & McDonald, D.M. Markers for microscopic imaging of lymphangiogenesis and angiogenesis. Ann. NY Acad. Sci. 1131, 1–12 (2008).

    Article  PubMed  Google Scholar 

  20. Ioannidou, S. et al. An in vitro assay reveals a role for the diaphragm protein PV-1 in endothelial fenestra morphogenesis. Proc. Natl. Acad. Sci. USA 103, 16770–16775 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Stan, R.V. et al. The diaphragms of fenestrated endothelia: gatekeepers of vascular permeability and blood composition. Dev. Cell 23, 1203–1218 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Herrnberger, L. et al. Lack of endothelial diaphragms in fenestrae and caveolae of mutant Plvap-deficient mice. Histochem. Cell Biol. 138, 709–724 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Sainte-Marie, G., Peng, F.S. & Belisle, C. Overall architecture and pattern of lymph flow in the rat lymph node. Am. J. Anat. 164, 275–309 (1982).

    Article  CAS  PubMed  Google Scholar 

  24. Farr, A.G., Cho, Y. & De Bruyn, P.P. The structure of the sinus wall of the lymph node relative to its endocytic properties and transmural cell passage. Am. J. Anat. 157, 265–284 (1980).

    Article  CAS  PubMed  Google Scholar 

  25. Van Vliet, E., Melis, M., Foidart, J.M. & Van Ewijk, W. Reticular fibroblasts in peripheral lymphoid organs identified by a monoclonal antibody. J. Histochem. Cytochem. 34, 883–890 (1986).

    Article  CAS  PubMed  Google Scholar 

  26. Stan, R.V., Tkachenko, E. & Niesman, I.R. PV1 is a key structural component for the formation of the stomatal and fenestral diaphragms. Mol. Biol. Cell 15, 3615–3630 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Shvets, E., Ludwig, A. & Nichols, B.J. News from the caves: update on the structure and function of caveolae. Curr. Opin. Cell Biol. 29, 99–106 (2014).

    Article  CAS  PubMed  Google Scholar 

  28. Razani, B. et al. Caveolin-1 null mice are viable but show evidence of hyperproliferative and vascular abnormalities. J. Biol. Chem. 276, 38121–38138 (2001).

    Article  CAS  PubMed  Google Scholar 

  29. Tkachenko, E. et al. Caveolae, fenestrae and transendothelial channels retain PV1 on the surface of endothelial cells. PLoS ONE 7, e32655 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Junt, T. et al. Subcapsular sinus macrophages in lymph nodes clear lymph-borne viruses and present them to antiviral B cells. Nature 450, 110–114 (2007).

    Article  CAS  PubMed  Google Scholar 

  31. Phan, T.G., Grigorova, I., Okada, T. & Cyster, J.G. Subcapsular encounter and complement-dependent transport of immune complexes by lymph node B cells. Nat. Immunol. 8, 992–1000 (2007).

    Article  CAS  PubMed  Google Scholar 

  32. Katakai, T. Marginal reticular cells: a stromal subset directly descended from the lymphoid tissue organizer. Front. Immunol. 3, 200 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Debes, G.F. et al. Chemokine receptor CCR7 required for T lymphocyte exit from peripheral tissues. Nat. Immunol. 6, 889–894 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Ledgerwood, L.G. et al. The sphingosine 1-phosphate receptor 1 causes tissue retention by inhibiting the entry of peripheral tissue T lymphocytes into afferent lymphatics. Nat. Immunol. 9, 42–53 (2008).

    Article  CAS  PubMed  Google Scholar 

  35. Murphy, D.A. & Courtneidge, S.A. The 'ins' and 'outs' of podosomes and invadopodia: characteristics, formation and function. Nat. Rev. Mol. Cell Biol. 12, 413–426 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Bearer, E.L. & Orci, L. Endothelial fenestral diaphragms: a quick-freeze, deep-etch study. J. Cell Biol. 100, 418–428 (1985).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Schnitzer, J.E., Shen, C.P. & Palade, G.E. Lectin analysis of common glycoproteins detected on the surface of continuous microvascular endothelium in situ and in culture: identification of sialoglycoproteins. Eur. J. Cell Biol. 52, 241–251 (1990).

    CAS  PubMed  Google Scholar 

  38. Malhotra, D. et al. Transcriptional profiling of stroma from inflamed and resting lymph nodes defines immunological hallmarks. Nat. Immunol. 13, 499–510 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Johnson, L.A. et al. An inflammation-induced mechanism for leukocyte transmigration across lymphatic vessel endothelium. J. Exp. Med. 203, 2763–2777 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Pfeiffer, F. et al. Distinct molecular composition of blood and lymphatic vascular endothelial cell junctions establishes specific functional barriers within the peripheral lymph node. Eur. J. Immunol. 38, 2142–2155 (2008).

    Article  CAS  PubMed  Google Scholar 

  41. Baluk, P. et al. Functionally specialized junctions between endothelial cells of lymphatic vessels. J. Exp. Med. 204, 2349–2362 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Carman, C.V. et al. Transcellular diapedesis is initiated by invasive podosomes. Immunity 26, 784–797 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Martinelli, R. et al. Probing the biomechanical contribution of the endothelium to lymphocyte migration: diapedesis by the path of least resistance. J. Cell Sci. 127, 3720–3734 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Buchholz, F., Angrand, P.O. & Stewart, A.F. Improved properties of FLP recombinase evolved by cycling mutagenesis. Nat. Biotechnol. 16, 657–662 (1998).

    Article  CAS  PubMed  Google Scholar 

  45. Schaft, J., Ashery-Padan, R., van der Hoeven, F., Gruss, P. & Stewart, A.F. Efficient FLP recombination in mouse ES cells and oocytes. Genesis 31, 6–10 (2001).

    Article  CAS  PubMed  Google Scholar 

  46. Sakai, K. & Miyazaki, J. A transgenic mouse line that retains Cre recombinase activity in mature oocytes irrespective of the cre transgene transmission. Biochem. Biophys. Res. Commun. 237, 318–324 (1997).

    Article  CAS  PubMed  Google Scholar 

  47. Keuschnigg, J. et al. PV-1 is recognized by the PAL-E antibody and forms complexes with NRP-1. Blood 120, 232–235 (2012).

    Article  CAS  PubMed  Google Scholar 

  48. Weber, M. et al. Interstitial dendritic cell guidance by haptotactic chemokine gradients. Science 339, 328–332 (2013).

    Article  CAS  PubMed  Google Scholar 

  49. Yokomizo, T. et al. Whole-mount three-dimensional imaging of internally localized immunostained cells within mouse embryos. Nat. Protoc. 7, 421–431 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank E. Butcher (Stanford University) for antibody MECA-367; The Cell Imaging Core at the Centre of Biotechnology and Laboratory of Electron Microscopy in University of Turku for help in imaging; E.-L. Väänänen, R. Sjöroos, S. Mäki and M. Pohjansalo for technical assistance; and A. Sovikoski-Georgieva for secretarial assistance. This work benefitted from data assembled by the Immunological Genome Project consortium. Supported by the Finnish Academy and Sigrid Juselius Foundation.

Author information

Authors and Affiliations

Authors

Contributions

P.R. and K.A. planned and performed experiments and analyzed data; N.J., M.Kap., J.V., M.Kar., H.G., I.I.-E.-K., J.K. and E.U. performed experiments; K.T. performed scanning electron microscopy; K.E. supervised the generation of Plvap−/− mice; M.M. and S.J. contributed to the preparation of the manuscript; and M.S. conceived of the project, directed the research and wrote the manuscript.

Corresponding author

Correspondence to Marko Salmi.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Generation of Plvap−/− mice.

(a) Schematic presentation of the targeting construct as well as the wild-type and targeted Plvap alleles]. PuroR, puromycin resistance. The location of the probes (5ext1, 3ext2, CAG) used in hybridizations, and the PCR primers (A,B,C) are depicted. (b) Southern blot analysis of Ssp1-digested genomic DNA isolated from wild-type (Plvap+/+) and heterozygous (Plvap+/−) embryonic stem cell clones (G7, G11, C9 and H4) with the indicated probes. The sizes of the hybridization bands corresponding wild-type (WT) and targeted (Targ) alleles are indicated. (c) Genotyping of the WT (Plvap+/+), Plvap+/− and Plvap−/− mice from heterozygous matings by PCR with the primers A-C. The sizes of the expected WT and targeted products are indicated. Primers A-B: lanes1,3,5,7; Primers B-C: lanes 2,4,6,8. (d) Immunoblotting of spleen, kidney and PLN lysates from littermate WT (Plvap+/+) and Plvap−/− mice with anti-PLVAP (MECA-32) mAb. The specific PLVAP band (105 kDa) is indicated by an arrow. GAPDH is a loading control. Data are representative of three or more independent experiments.

Supplementary Figure 2 The phenotype of lymphocytes and subcapsular sinus in lymph nodes of wild-type and Plvap−/− mice.

(a-c) Flow cytometry of (a) surface immunoglobulin receptors IgM and IgD, (b) chemokine receptor CXCR5 and (c) chemoattractant receptor SIP1R on B220+ B-cells in PLNs of WT and Plvap−/− mice. Representative flow cytometry plots and quantification of data are shown. Grey histograms in (b,c) represent stainings with isotype controls. MFI, mean fluorescence intensity. (d-g) Immunoreactivity of PLVAP (MECA-32) and (d) Prox-1, (e,f) podoplanin or (g) Lyve-1 in PLNs. C, ceiling; F, floor. The red arrow in f and g points to a sinus-traversing strand, and the white arrow to a blood vessel in d. (h) Immunoreactivity of PLVAP (MECA-32) and Prox-1 in WT mice of pure BALB/c, C57BL/6N and NMRI strains and in F1 hybrids of C57BL/6;129. White arrows point to blood vessels. C, ceiling; F, floor. Scale bars, 20 µm (d,e: tile scans), 10 µm (d,e (insets), f-h). Data in a-d are from littermate WT and Plvap−/− mice. Data are representative of or are pooled (all quantifications, mean ± s.e.m.) from 3 independent experiments (n=6 mice/genotype (a), 3-4 mice/genotype (b,c,d,e,f,g) or 2 mice/genotype (h)).

Supplementary Figure 3 Endothelial phenotypes of wild-type and Plvap−/− mice and humans.

(a) Immunoreactivity of mouse mesenteric lymph nodes for PLVAP (MECA-32) and Prox-1. C, ceiling; F, floor. White arrowheads point to representative blood vessels. (b) Plvap mRNA expression in blood endothelial cells (BEC) and lymphatic endothelial cells (LEC) in mesenteric lymph nodes of WT mouse (from the ImmGen data). Expression values >120 (red dashed line) indicate positivity. (c) Immunoreactivity of human PLNs for PLVAP (PAL-E) and Lyve-1. In control stainings an isotype-matched negative control antibody was used instead of PAL-E. C, ceiling, F, floor, white arrows point to HEVs. (d) Immunoreactivity of PLN subcapsular sinus of WT and Plvap−/− mice for CD31, VCAM-1, ICAM-1, VE-cadherin, CCRL1 and Lyve-1. C, ceiling, F, floor. White arrowheads point to blood vessels. (e) Immunoreactivity of skin vasculature for PLVAP (MECA-32) and CD31 (blood vessels, arrowheads) in whole-mount split ear preparations. (f) Immunoreactivity afferent lymphatic vasculature of skin for PLVAP (MECA-32) and Lyve-1 determined by confocal microscopy in ear sections. White arrowheads point to blood vessels, and red arrowheads to a lymphatic vessel. Blue is DAPI. Scale bars, 10 µm (a,b), 20 µm (c,d,f), 100 µm (e). Data in a and d-f are from littermate WT and Plvap−/− mice. Data are representative of three or more independent experiments with a total of 3-5 mice/genotype (a,d,e,f), are pooled (mean) from the ImmGen database (b) and of 3 different individuals (c).

Supplementary Figure 4 Ultrastructure of subcapsular sinus of PLNs.

(a-g) Electron micrographs of WT sinus. (a) The overall structure of the sinus showing highly elongated ceiling and floor LECs (indicated with a 20 % opacity magenta overlay), intrasinusoidal leukocytes (indicated with a 20 % opacity turquoise overlay) and a subcapsular sinus macrophage (M) partially extending to the lumen. Higher magnifications show (b) a transendothelial channel (outlined by arrows) closed by diaphragms (arrowheads), (c) vesicular bodies composed of multiple caveolae with diaphragms (arrowheads), (d) a tight junction (arrows) between 2 subcapsular sinus LECs, (e) a transection of a conduit ensheated by a floor (F) LEC, (f) highly abundant caveolae and vesicular bodies in the attenuated part of floor LECs (arrowheads point to diaphragms), and (g) tubular and canalicular structures (arrows) of LECs. (h,i) Electron micrographs of subcapsular sinus in Plvap−/− mice showing (h) a tight LEC junction (arrows), and (i) tubular and canalicular structures (arrows) within a LEC cytoplasm. SCS, lumen of the subcapsular sinus; N, nucleus. Scale bars, 5 µm (a), 100 nm (b,c), 200 nm (d,f,g,h), and 500 nm (e,i). Data are representative of three or more independent experiments with a total of 3-5 mice/genotype.

Supplementary Figure 5 The conduit network and sinus permeability to different antigens in wild-type and Plvap−/− mice.

(a) Co-localization of subcutaneously administered fluorescent dextrans and perlecan (a conduit marker) immunoreactivity in the conduits of WT and Plvap−/− mice. (b) Localization of a 20 nm quantum dots (Qtracker) in the subcapsular sinus (SCS) and medulla in PLNs of WT and Plvap−/− mice 10 min after subcutaneous injections. Quantitation of the fluorescence signal in the cortical area was determined using image analyses. (c) Schematic drawing showing the main components and molecular markers of the conduits. (d-f) Localization of (d) ER-TR7 and collagen type I, (e) perlecan, and (f) smooth muscle actin (SMA) in the conduits in the interfollicular parenchymal area of PLNs in WT and Plvap−/− mice. Scale bars, 50 µm (a,b) and 10 µm (d-f). All data are from littermate WT and Plvap−/− mice. Data are representative of 2 independent experiments with a total of 4-5 (a,b) and 3 (c-f) and mice/genotype.

Supplementary Figure 6 Conduit filtering function is intact in Cav1−/− mice and in the absence of subcapsular macrophages.

(a-c) Electron micrographs of subcapsular sinus in Cav1−/− mice showing (a) tubular and canalicular structures (arrows), (b) a transendothelial channel (white arrows) guarded by luminal and abluminal diaphragms (arrowheads) and (c) a conduit covered by LEC cytoplasm. SCS, lumen of the subcapsular sinus. (d) Flow cytometry of distribution of CD8+, CD4+ and B220+ lymphocytes in WT and Cav1−/− mice. (e) Immunostainings and quantitative analyses of ER-TR7 reactivity in the follicles (outlined by the white dashed line) and interfollicular areas of WT and Cav1−/− mice. (f) Microscopic analyses of the filling of the conduits 2 min after a subcutaneous injection of Pacific-Blue-labeled 10 kDa, TRITC-labeled 70 kDa and Alexa488-labeled 500 kDa dextrans in WT and Cav1−/− mice. (g,h) Immunoreactivity of cells in the subcapsular sinus (SCS) and medulla for (g) CD169 and Lyve-1 and (h) MAdCAM-1, Lyve and PLVAP (MECA-32) in WT and Plvap−/− mice. C, ceiling; F, floor. (i) Filling of the conduits with the fluorescent dextrans of different molecular weights after 1 min of subcutaneous injections in WT mice pretreated with chlodronate or control liposomes 7 d earlier. White arrows point to medullary macrophages. Scale bars, Scale bars, 500 nm (a), 200 nm (b), 2 µm (c), 100 µm (e,f), 20 µm (g), 10 µm (h) and 50 µm (i). Data are representative of or are pooled (all quantifications, mean ± s.e.m.) from experiments with a total of 3-5 mice/genotype.

Supplementary Figure 7 PLVAP-dependent lymphocyte migration through the LECs at sinus-cortex interface in PLNs.

(a) Immunofluorescence imaging of a representative CFSE+ cell (green arrow; injected subcutaneously 4 h earlier) that is partially positioned within the Lyve-1+ LEC floor in PLN sinus. (b-d) CFSE-labeled lymphocytes (isolated from WT spleens) and non-conjugated MECA-32 or isotype-matched control antibodies were co-administered subcutaneously to WT mice and the cells were allowed to migrate for 8 h. The phenotype of CD8+ T lymphocytes among the injected cells and among the donor cells recovered from the draining PLNs was determined using flow cytometry. Shown are representative flow cytometry dot plots and quantification of the data. Naive T cells are CD44loCD62Lhi, activated T-lymphocytes CD44hiCD62Llo, and central memory T cells CD44hiCD62Lhi. (e) Immunoreactivity of cultured LECs isolated from human PLNs for VE-cadherin and an isotype-matched negative control antibody for PLVAP. (f,g) Immunostainings of an endogenous lymphocyte (CD45+) that is attached to the sinusoidal LEC floor for F-actin, Lyve-1 and PLVAP in WT mice. C, ceiling; F, floor. The correlation of F-actin and PLVAP (MECA-32) staining intensities along the sinusoidal floor (arrow line) was measured using image analyses. Scale bars, 10 µm (a,e), and 5 µm (f,g). Data are representative of or are pooled (all quantifications, mean ± s.e.m.) from 2-3 independent experiments (n=8 WT mice (a), injected cells as a pool from 10 mice and migrated cells from 3 WT mice/ treatment (b-d), 4 stainings (e), 2 WT mice (f) or from 3 WT mice (g)). * P < 0.05 (Mann-Whitney U-test).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 (PDF 8498 kb)

Plvap is expressed in the Prox-1 positive floor and ceiling LECs in subcapsular sinus of WT mice.

Immunoreactivity of Plvap (MECA-32, green) and Prox-1 (white) in co-stained whole mount popliteal lymph node preparations after optical clearing. Note that Plvap is present in the LECs at the floor (F) and ceiling (C) of the subcapsular sinus (SCS), and in the cortical blood vessels (BECs). Nuclear Prox-1 staining defines LECs. Scale bar, 30 μm (at the beginning of the video). (MOV 23986 kb)

Plvap is expressed in the podoplanin positive floor and ceiling LECs in subcapsular sinus of WT mice.

Immunoreactivity of Plvap (MECA-32, green) and podoplanin (white) in co-stained whole mount popliteal lymph node preparations after optical clearing. Plvap and podoplanin signals are shown on individual channels (left video: Plvap channel only; right video: podoplanin channel only). Note that podoplanin stains both the LECs and FRCs. FRC staining is strongest above the ceiling LECs in the capsule of the lymph node. Scale bar, 30 μm (at the beginning of the video) (MOV 16037 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rantakari, P., Auvinen, K., Jäppinen, N. et al. The endothelial protein PLVAP in lymphatics controls the entry of lymphocytes and antigens into lymph nodes. Nat Immunol 16, 386–396 (2015). https://doi.org/10.1038/ni.3101

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3101

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