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:

Tetraspanin-enriched membrane domains regulate vascular leakage by altering membrane cholesterol accessibility to balance antagonistic GTPases

A Publisher Correction to this article was published on 21 August 2025

This article has been updated

Abstract

Tetraspanins affect metastasis, stemness and angiogenesis, but their roles in inflammation remain to be further clarified. Here we show that endothelial ablation of tetraspanin Cd82 markedly reduces vascular inflammation by mitigating endothelial leakage. Mechanistically, by limiting the anchorages of Cdc42 activator FARP1 and RhoA inhibitor Rnd3 to the plasma membrane (PM), CD82 confines Cdc42 but maintains RhoA activity in endothelial cells, to facilitate endothelium activation. These signaling regulatory effects depend on the ability of CD82 to coalesce and retain accessible cholesterol (AC) at the PM, whereas simvastatin overturns CD82 effects by lowering AC. CD82 supports non-vesicular transfer of AC to the PM through oxysterol-binding protein-related proteins (ORPs). Thus, CD82 and AC promote vascular leakage, whereas statin and ORP inhibitor restrain vascular leakage by decreasing AC. These findings reveal an unconventional anti-inflammation role and mechanism for statin and conceptualize tetraspanin-mediated, AC-mediated and cholesterol transfer-mediated balancing of antagonistic GTPase signaling pathways as regulatory mechanisms for vascular leakage.

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: Cd82 ablation reduces vascular inflammation.
Fig. 2: Cd82 removal stabilizes EC–EC contacts.
Fig. 3: CD82 restrains Cdc42 activity by preventing FARP1 coalescence to the PM.
Fig. 4: CD82 sustains RhoA activity by preventing Rnd3 coalescence to the PM.
Fig. 5: CD82 sustains AC on EC for endothelial hyperpermeability.
Fig. 6: Binding of CD82 to AC supports RhoA but confines Cdc42.
Fig. 7: CD82 facilitates non-vesicular transport of cholesterol to the PM.
Fig. 8: Physical interaction of CD82 with ORP2 supports AC presence at cell surface.

Similar content being viewed by others

Data availability

All source data are provided with this research article. TMT proteomics data were deposited in the PRIDE partner repository under accession number PXD065001, and the bulk RNA-seq data were deposited in National Center for Biotechnology Information BioSample under accession number SAMN48951810. All supporting data for the findings of this project are available within either the article or its Supplementary Information. Additional information related to the findings of this project are available from the corresponding author upon reasonable request. Source data are provided with this paper.

Code availability

No code was developed for this study.

Change history

References

  1. Goddard, L. M. & Iruela-Arispe, M. L. Cellular and molecular regulation of vascular permeability. Thromb. Haemost. 109, 407–415 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Komarova, Y. & Malik, A. B. Regulation of endothelial permeability via paracellular and transcellular transport pathways. Annu. Rev. Physiol. 72, 463–493 (2010).

    Article  PubMed  CAS  Google Scholar 

  3. Prasad, M., Leon, M., Lerman, L. O. & Lerman, A. Viral endothelial dysfunction: a unifying mechanism for COVID-19. Mayo Clin. Proc. 96, 3099–3108 (2021).

    Article  PubMed  CAS  Google Scholar 

  4. Giannotta, M., Trani, M. & Dejana, E. VE-cadherin and endothelial adherens junctions: active guardians of vascular integrity. Dev. Cell 26, 441–454 (2013).

    Article  PubMed  CAS  Google Scholar 

  5. Li, X. et al. VEGFR2 pY949 signalling regulates adherens junction integrity and metastatic spread. Nat. Commun. 7, 11017 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  6. Spindler, V., Schlegel, N. & Waschke, J. Role of GTPases in control of microvascular permeability. Cardiovasc. Res. 87, 243–253 (2010).

    Article  PubMed  CAS  Google Scholar 

  7. Beckers, C. M., van Hinsbergh, V. W. & van Nieuw Amerongen, G. P. Driving Rho GTPase activity in endothelial cells regulates barrier integrity. Thromb. Haemost. 103, 40–55 (2010).

    Article  PubMed  CAS  Google Scholar 

  8. Yanez-Mo, M., Barreiro, O., Gordon-Alonso, M., Sala-Valdes, M. & Sanchez-Madrid, F. Tetraspanin-enriched microdomains: a functional unit in cell plasma membranes. Trends Cell Biol. 19, 434–446 (2009).

    Article  PubMed  CAS  Google Scholar 

  9. Charrin, S. et al. Lateral organization of membrane proteins: tetraspanins spin their web. Biochem. J. 420, 133–154 (2009).

    Article  PubMed  CAS  Google Scholar 

  10. Termini, C. M. & Gillette, J. M. Tetraspanins function as regulators of cellular signaling. Front. Cell Dev. Biol. 5, 34 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Hemler, M. E. Tetraspanin functions and associated microdomains. Nat. Rev. Mol. Cell Biol. 6, 801–811 (2005).

    Article  PubMed  CAS  Google Scholar 

  12. Zhang, F., Kotha, J., Jennings, L. K. & Zhang, X. A. Tetraspanins and vascular functions. Cardiovasc. Res. 83, 7–15 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Bailey, R. L. et al. The emerging role of tetraspanin microdomains on endothelial cells. Biochem. Soc. Trans. 39, 1667–1673 (2011).

    Article  PubMed  CAS  Google Scholar 

  14. Takeda, Y. et al. Deletion of tetraspanin Cd151 results in decreased pathologic angiogenesis in vivo and in vitro. Blood 109, 1524–1532 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Zhang, F. et al. Tetraspanin CD151 maintains vascular stability by balancing the forces of cell adhesion and cytoskeletal tension. Blood 118, 4274–4284 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Wei, Q. et al. CD82 restrains pathological angiogenesis by altering lipid raft clustering and CD44 trafficking in endothelial cells. Circulation 130, 1493–1504 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  17. Xu, C. et al. CD82 endocytosis and cholesterol-dependent reorganization of tetraspanin webs and lipid rafts. FASEB J. 23, 3273–3288 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  18. Zimmerman, B. et al. Crystal structure of a full-length human tetraspanin reveals a cholesterol-binding pocket. Cell 167, 1041–1051 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Susa, K. J., Rawson, S., Kruse, A. C. & Blacklow, S. C. Cryo-EM structure of the B cell co-receptor CD19 bound to the tetraspanin CD81. Science 371, 300–305 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  20. Lipper, C. H., Egan, E. D., Gabriel, K. H. & Blacklow, S. C. Structural basis for membrane-proximal proteolysis of substrates by ADAM10. Cell 186, 3632–3641 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  21. Das, A., Brown, M. S., Anderson, D. D., Goldstein, J. L. & Radhakrishnan, A. Three pools of plasma membrane cholesterol and their relation to cholesterol homeostasis. eLife 3, e02882 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Endapally, S. et al. Molecular discrimination between two conformations of sphingomyelin in plasma membranes. Cell 176, 1040–1053 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Kinnebrew, M. et al. Cholesterol accessibility at the ciliary membrane controls hedgehog signaling. eLife 8, e50051 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Zhou, Q. D. et al. Interferon-mediated reprogramming of membrane cholesterol to evade bacterial toxins. Nat. Immunol. 21, 746–755 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Abrams, M. E. et al. Oxysterols provide innate immunity to bacterial infection by mobilizing cell surface accessible cholesterol. Nat. Microbiol. 5, 929–942 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Wang, S. et al. Cholesterol 25-hydroxylase inhibits SARS-CoV-2 and other coronaviruses by depleting membrane cholesterol. EMBO J. 39, e106057 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  27. Naito, T. et al. Movement of accessible plasma membrane cholesterol by the GRAMD1 lipid transfer protein complex. eLife 8, e51401 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Huang, C., Hays, F. A., Tomasek, J. J., Benyajati, S. & Zhang, X. A. Tetraspanin CD82 interaction with cholesterol promotes extracellular vesicle–mediated release of ezrin to inhibit tumour cell movement. J. Extracell. Vesicles 9, 1692417 (2020).

    Article  PubMed  CAS  Google Scholar 

  29. Charrin, S. et al. A physical and functional link between cholesterol and tetraspanins. Eur. J. Immunol. 33, 2479–2489 (2003).

    Article  PubMed  CAS  Google Scholar 

  30. Wang, Z. et al. 27-Plex tandem mass tag mass spectrometry for profiling brain proteome in Alzheimer’s disease. Anal. Chem. 92, 7162–7170 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. Dejana, E., Orsenigo, F. & Lampugnani, M. G. The role of adherens junctions and VE-cadherin in the control of vascular permeability. J. Cell Sci. 121, 2115–2122 (2008).

    Article  PubMed  CAS  Google Scholar 

  32. Amado-Azevedo, J. et al. A CDC42-centered signaling unit is a dominant positive regulator of endothelial integrity. Sci. Rep. 7, 10132 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Etienne-Manneville, S. & Hall, A. Rho GTPases in cell biology. Nature 420, 629–635 (2002).

    Article  PubMed  CAS  Google Scholar 

  34. Priya, R. et al. Feedback regulation through myosin II confers robustness on RhoA signalling at E-cadherin junctions. Nat. Cell Biol. 17, 1282–1293 (2015).

    Article  PubMed  CAS  Google Scholar 

  35. Wildenberg, G. A. et al. p120-catenin and p190RhoGAP regulate cell-cell adhesion by coordinating antagonism between Rac and Rho. Cell 127, 1027–1039 (2006).

    Article  PubMed  CAS  Google Scholar 

  36. Tall, A. R. & Yvan-Charvet, L. Cholesterol, inflammation and innate immunity. Nat. Rev. Immunol. 15, 104–116 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Radhakrishnan, A., Rohatgi, R. & Siebold, C. Cholesterol access in cellular membranes controls Hedgehog signaling. Nat. Chem. Biol. 16, 1303–1313 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  38. Sakata, A. et al. Acid sphingomyelinase inhibition suppresses lipopolysaccharide-mediated release of inflammatory cytokines from macrophages and protects against disease pathology in dextran sulphate sodium-induced colitis in mice. Immunology 122, 54–64 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Kielczewski, J. L. et al. Free insulin-like growth factor binding protein-3 (IGFBP-3) reduces retinal vascular permeability in association with a reduction of acid sphingomyelinase (ASMase). Invest. Ophthalmol. Vis. Sci. 52, 8278–8286 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Johnson, K. A., Endapally, S., Vazquez, D. C., Infante, R. E. & Radhakrishnan, A. Ostreolysin A and anthrolysin O use different mechanisms to control movement of cholesterol from the plasma membrane to the endoplasmic reticulum. J. Biol. Chem. 294, 17289–17300 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Infante, R. E. & Radhakrishnan, A. Continuous transport of a small fraction of plasma membrane cholesterol to endoplasmic reticulum regulates total cellular cholesterol. eLife 6, e25466 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Gay, A., Rye, D. & Radhakrishnan, A. Switch-like responses of two cholesterol sensors do not require protein oligomerization in membranes. Biophys. J. 108, 1459–1469 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Heisler, D. B. et al. A concerted mechanism involving ACAT and SREBPs by which oxysterols deplete accessible cholesterol to restrict microbial infection. eLife 12, e83534 (2023).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Ikenouchi, J. & Aoki, K. Membrane bleb: a seesaw game of two small GTPases. Small GTPases 8, 85–89 (2017).

    Article  PubMed  CAS  Google Scholar 

  45. Fackler, O. T. & Grosse, R. Cell motility through plasma membrane blebbing. J. Cell Biol. 181, 879–884 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Li, Z., Mintzer, E. & Bittman, R. First synthesis of free cholesterol–BODIPY conjugates. J. Org. Chem. 71, 1718–1721 (2006).

    Article  PubMed  CAS  Google Scholar 

  47. Holtta-Vuori, M. et al. BODIPY-cholesterol: a new tool to visualize sterol trafficking in living cells and organisms. Traffic 9, 1839–1849 (2008).

    Article  PubMed  CAS  Google Scholar 

  48. Sezgin, E. et al. A comparative study on fluorescent cholesterol analogs as versatile cellular reporters. J. Lipid Res. 57, 299–309 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  49. Sandhu, J. et al. Aster proteins facilitate nonvesicular plasma membrane to ER cholesterol transport in mammalian cells. Cell 175, 514–529 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Wang, H. et al. ORP2 delivers cholesterol to the plasma membrane in exchange for phosphatidylinositol 4, 5-bisphosphate (PI(4,5)P2). Mol. Cell 73, 458–473 (2019).

    Article  PubMed  CAS  Google Scholar 

  51. Zhao, K., Foster, J. & Ridgway, N. D. Oxysterol-binding protein-related protein 1 variants have opposing cholesterol transport activities from the endolysosomes. Mol. Biol. Cell 31, 793–802 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Burgett, A. W. et al. Natural products reveal cancer cell dependence on oxysterol-binding proteins. Nat. Chem. Biol. 7, 639–647 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Zhou, B., Liu, L., Reddivari, M. & Zhang, X. A. The palmitoylation of metastasis suppressor KAI1/CD82 is important for its motility- and invasiveness-inhibitory activity. Cancer Res. 64, 7455–7463 (2004).

    Article  PubMed  CAS  Google Scholar 

  54. Jain, M. K. & Ridker, P. M. Anti-inflammatory effects of statins: clinical evidence and basic mechanisms. Nat. Rev. Drug Discov. 4, 977–987 (2005).

    Article  PubMed  CAS  Google Scholar 

  55. Li, D., Dammer, E. B., Lucki, N. C. & Sewer, M. B. cAMP-stimulated phosphorylation of diaphanous 1 regulates protein stability and interaction with binding partners in adrenocortical cells. Mol. Biol. Cell 24, 848–857 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Kentala, H. et al. Analysis of ORP2-knockout hepatocytes uncovers a novel function in actin cytoskeletal regulation. FASEB J. 32, 1281–1295 (2018).

    Article  PubMed  CAS  Google Scholar 

  57. Broman, M. T., Mehta, D. & Malik, A. B. Cdc42 regulates the restoration of endothelial adherens junctions and permeability. Trends Cardiovasc. Med. 17, 151–156 (2007).

    Article  PubMed  CAS  Google Scholar 

  58. Huang, C. et al. Tetraspanin-enriched microdomains regulate digitation junctions. Cell. Mol. Life Sci. 75, 3423–3439 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Uchtmann, K. et al. Homozygous loss of mouse tetraspanin CD82 enhances integrin αIIbβ3 expression and clot retraction in platelets. Exp. Cell. Res. 339, 261–269 (2015).

    Article  PubMed  CAS  Google Scholar 

  60. Benedito, R. et al. Notch-dependent VEGFR3 upregulation allows angiogenesis without VEGF–VEGFR2 signalling. Nature 484, 110–114 (2012).

    Article  PubMed  CAS  Google Scholar 

  61. Gon, Y. et al. S1P3 receptor-induced reorganization of epithelial tight junctions compromises lung barrier integrity and is potentiated by TNF. Proc. Natl Acad. Sci. USA 102, 9270–9275 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Scheppke, L. et al. Retinal vascular permeability suppression by topical application of a novel VEGFR2/Src kinase inhibitor in mice and rabbits. J. Clin. Invest. 118, 2337–2346 (2008).

    PubMed  PubMed Central  CAS  Google Scholar 

  63. Balda, M. S. et al. Functional dissociation of paracellular permeability and transepithelial electrical resistance and disruption of the apical-basolateral intramembrane diffusion barrier by expression of a mutant tight junction membrane protein. J. Cell Biol. 134, 1031–1049 (1996).

    Article  PubMed  CAS  Google Scholar 

  64. Endapally, S., Infante, R. E. & Radhakrishnan, A. Monitoring and modulating intracellular cholesterol trafficking using ALOD4, a cholesterol-binding protein. Methods Mol. Biol. 1949, 153–163 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Newby, Z. E. et al. A general protocol for the crystallization of membrane proteins for X-ray structural investigation. Nat. Protoc. 4, 619–637 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Bartoschik, T. et al. Near-native, site-specific and purification-free protein labeling for quantitative protein interaction analysis by MicroScale Thermophoresis. Sci. Rep. 8, 4977 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  67. Jumper, J. et al. Highly accurate protein structure prediction with AlphaFold. Nature 596, 583–589 (2021).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  68. Practice and Experience in Advanced Research Computing 2017: Sustainability, Success and Impact (Association for Computing Machinery, 2017).

  69. Mirabello, C. & Wallner, B. DockQ v2: improved automatic quality measure for protein multimers, nucleic acids, and small molecules. Bioinformatics 40, btae586 (2024).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  70. Jurrus, E. et al. Improvements to the APBS biomolecular solvation software suite. Protein Sci. 27, 112–128 (2018).

    Article  PubMed  CAS  Google Scholar 

  71. Reed, L. J. & Muench, H. A simple method of estimating fifty per cent endpoints. Am. J. Epidemiol. 27, 493–497 (1938).

    Article  Google Scholar 

  72. Ideker, T. & Krogan, N. J. Differential network biology. Mol. Syst. Biol. 8, 565 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  73. Wren, J. D. A global meta-analysis of microarray expression data to predict unknown gene functions and estimate the literature–data divide. Bioinformatics 25, 1694–1701 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  74. Wren, J. D., Bekeredjian, R., Stewart, J. A., Shohet, R. V. & Garner, H. R. Knowledge discovery by automated identification and ranking of implicit relationships. Bioinformatics 20, 389–398 (2004).

    Article  PubMed  CAS  Google Scholar 

  75. Szklarczyk, D. et al. STRING v10: protein–protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 43, D447–D452 (2015).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by National Institutes of Health grants HL132553, HL137819 and GM135547 and American Heart Association grants 13GRNT17040028 and 15GRNT25700426 to X.A.Z. The study was also supported by NIH grant R37CA276924 to K.M. We thank A. Radhakrishnan of The University of Texas Southwestern Medical Center for lipid probes, Y. Zheng of Cincinnati Children’s Hospital for Cdc42-floxed mouse line and P. Caswell of the University of Manchester for AHPH constructs. We are grateful to A. Radhakrishnan of The University of Texas Southwestern Medical Center, H. Chen of Harvard Medical School and C. Griffin and L. Xia of the Oklahoma Medical Research Foundation for helpful suggestions and valuable discussions.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: Y.D. and X.A.Z. Methodology/Investigation: Y.D., J.C., S.L., J.M.H., X.G., D.N.R., B.L., F.H., X.W., Q.W., J.W., B.S. and A.M.R. Dataset Analysis: Y.D., J.D.W., C.G. and W.M.F. Resources: Z.W., K.K., C.K.M., A.C., Z.U., K.M., M.H.E., J.P. and T.K. Writing: Y.D., J.C., J.D.W., M.S.W., J.F.P., F.A.H. and X.A.Z. Supervision: X.A.Z.

Corresponding author

Correspondence to Xin A. Zhang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Cardiovascular Research thanks Victoria Bautch 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 Upregulation of CD82 expression in inflammation.

(a) GSE159585 analysis of lung biopsies from Covid-19 patients and controls. UMAP visualization of all cell clusters profiled and CD82 gene expression in human lung tissues from GSE159585 (left panel). Levels of CD82 mRNA (Transcripts per million, TPM) are presented as mean±SD (right panel). (b) Increased expression of CD82 in the pulmonary endothelium of COPD patients. Immunohistochemistry of CD82 was examined in lung sections of COPD patients and controls. Arrows indicate CD82 staining in endothelia. (c) Increased CD82 expression in the endothelia within atherosclerosis lesions. Mouse aortas were harvested from wild-type (WT) and ApoE/ mice on C57BL/6 background for CD82 immunohistochemical staining. Dashed areas are atherosclerotic lesions. Black arrows show aortic endothelium in non-lesion regions, while red arrows indicate endothelium from lesion areas. (d) From GEO database, analysis on GSE2372 for CD82 expression in the aortas from WT and ApoE/ mouse lines on C57BL/6 background (mean±SD; n=3 mice per group). (e) Upregulation of CD82 expression in endothelia in LPS-induced lung inflammation. Pulmonary tissues were harvested from C57BL/6 mice with or without LPS challenge (5mg/kg, i.p.) for CD82 immunohistochemical staining. Arrows show endothelium of pulmonary vessels. (f) From GEO database, the GSE2411 analysis on the lungs from C57BL/6 mouse model of LPS-induced lung inflammation and injury (mean±SD; n=6 mice per group). (g) Effects of inflammatory stimuli on endothelial CD82 expression. CD82 mRNA and proteins from the MLECs treated with or without LPS (100 ng/mL) for 6 h and 24 h were examined with qRT-PCR and Western blot, respectively (mean±SD; n=3 independent experiments) (left panel). HMECs were treated with LPS (100 ng/mL), TNF-α (10 ng/mL), or IL-4 (20 ng/mL) for 24 h and analyzed with Western blot for CD82 (middle panel). HUVECs were treated with VEGF-A164 (50 ng/mL), LPS (100 ng/mL), TNF-α (10 ng/mL), IL-1β (20 ng/mL), or IL-6 (20 ng/mL) for 48 h and then analyzed Western blot for CD82 (right panel). Relative levels of CD82 mRNA or protein were quantified (mean±SD; n=3 individual experiments).

Source data

Extended Data Fig. 2 CD82 and inflammatory signaling: bioinformatics analyses.

(a) Scheme of inducible deletion of Cd82 in endothelium. Cd82flox/flox;Cdh5-Cre/ERT2+ and Cd82flox/flox;Cdh5-Cre/ERT2 mice were administered with tamoxifen intraperitoneally daily for five days and used as Cd82ΔEC and control (Ctrl.) mice, respectively, for experiments. (b) CD82 mRNA and protein levels in primary MLECs that were isolated from endothelium-specific Cd82-null mice and control littermates. CD82 mRNA was examined in qRT-PCR, and the quantities are presented as the levels relative to the mRNA levels of housekeeping genes (mean±SD; n=3 individual experiments). CD82 proteins were detected in Western blot with murine CD82 pAb M35, and GAPDH was used as a protein loading control. (c) Ingenuity pathway analysis on the bulk RNAseq data of MLECs from Cd82ΔEC mice and their control littermates. The pathways related to inflammatory and immune responses are labeled with red asterisks. Left panel: the pathways were ranked by -log (P-value)s. Ratio denotes the number of altered genes in a given pathway to the total number of genes in that pathway. Right panel: the pathways were ranked by -log (false discovery rate or FDR)s. The dot size presents the number of enriched genes, and the dot color codes the FDR level. The enrichment ratio is defined as the number of observed genes divided by the number of listed genes in an ingenuity category. (d) CD82-inflammation correlation network was generated as described in "Methods" section. CD82 and the inflammation-related genes that are most strongly correlated with Cd82 in gene expression (green) were connected by potential PPIs (gray) using STRING v10.

Source data

Extended Data Fig. 3 CD82 facilitates vascular inflammation.

(a) Representative images of echocardiography from Ctrl. or Cd82ΔEC mice challenged with LPS (5 mg/kg) at 18 h or 48 h after the LPS administration. (b) Cd82 ablation does not alter the basal permeability of skin vessels. Evans blue dye (30 mg/kg in PBS) was injected intravenously through the retro-orbital sinus into 8~12-week-old Cd82/ mice or their WT littermates. The leakage of Evans blue dye into ear skin was assessed at 0, 30, or 60 min after the injection. Extravasated Evans dye was quantified and corrected with ear dry weight (mean±SEM; n=6 mice per group for 60 min and n=11 mice per group for 30 min). (c) Representative images of Evans blue dye leakage into skin of the ears from WT and Cd82/ mice as well as from Ctrl. and Cd82ΔEC mice in Miles assay. (d) Body weight was measured daily after SARS-Cov-2 infection in Ctrl. or Cd82ΔEC mice (mean±SD; n=9 mice for Ctrl. group and n=8 mice for Cd82ΔEC group). (e) SARS-Cov-2 titer in control and Cd82ΔEC mice. Pulmonary tissues were harvested from Ctrl. or Cd82ΔEC mice, and SARS-CoV-2 nucleocapsid (N) gene expression is presented as its relative level after normalization with actin expression using the dCT method for all mice in the experiment (left panel) and only for those mice which succumbed to infection at the time of death (right panel). Nine mice from Ctrl. group and eight mice from Cd82ΔEC group were used for the study.

Source data

Extended Data Fig. 4 CD82 removal from ECs and the effects of CD82 removal on EC-EC contact/adhesion and Cdc42 and Rac1 activities.

(a) Flow cytometry analysis on the cell surface level of CD82 at 48 and 72 h after the ECs were transfected with control or CD82 siRNA, respectively. Human CD82 was detected with its mAb TS82b, with mouse IgG as a negative control mAb. Western blot of CD82 in bEnd.3 transfectant cell (right panel). (b) Basal TEER levels of MLECs, HMECs, HUVECs, and bEnd.3 murine brain ECs. (c) Effects of CD82 removal on the levels of EC-EC adhesion proteins. MLECs from Cd82+/+ and Cd82/ mice and HUVECs transiently transfected with control and CD82 siRNA were lysed with RIPA lysis buffer, and the lysates were examined by Western blot for the indicated proteins. (d) Cd82 ablation and Rac1 activity. MLECs were stimulated with VEGF-A164 (50 ng/mL) after serum-starvation overnight and then lysed with RIPA buffer. Active Rac1 was detected by G-Lisa assay for Rac1-GTP and presented as activities (mean±SD; n=4 experiments). (e) CD82 silencing and Rac1 activity. HUVECs transfected with control or CD82 siRNA were serum-starved overnight, stimulated with VEGF-A (50 ng/ml), and then lysed with RIPA buffer. GTP-bound Rac1 was pulled down from the lysates with GST-PAK Rac1-binding domain and detected in Western blot. Other indicated proteins in the lysates were directly examined with Western blot. The ratios of GTP-bound Rac1/total Rac1 were quantified based on the band densities of Rac1 (mean±SD; n=3 independent experiments). (f) Effects of VEGF and Cd82 ablation on Cdc42 activity. Confluent MLECs were serum-starved overnight and treated with vehicle (PBS) or VEGF-A164 (50 ng/mL) for 15 min. Cdc42 activities were measured by G-LISA assay for Cdc42-GTP and presented as mean±SD (n=7 experiments). (g) Protein and mRNA levels of FARP1, SYDE1, and others in the MLECs were examined with qRT-PCR and Western blot, respectively. The mRNA levels are presented as mean±SD (n=4 experiments). (h) Increased distribution of FARP1 at or near the basal PM of Cd82/ MLECs, revealed by TIRF microscopy analysis on Cd82+/+ and Cd82/ MLECs. Dashed lines show the edge of the MLEC.

Source data

Extended Data Fig. 5 Downregulation of CD82 and upregulation of FARP1-Cdc42 signaling as anti-inflammation therapeutic strategies.

(a) Western blot analysis on the lysates of MLECs isolated from Ctrl., Cd82ΔEC, and Cd82ΔEC;Cdc42ΔEC mice. (b)-(c) Histochemistry (H-E staining) (b) and immunofluorescence (CD45 staining) (c) analyses on lung sections of the Control, Cd82ΔEC, and Cd82ΔEC;Cdc42ΔEC mice treated with LPS (5 mg/kg) for 18 h described in Fig. 3i. (d) Quantifications of acute lung injury (violin plot), Evans Blue extravasation or vascular leakage (mean±SD), and leukocyte infiltration of the lungs (violin plot) from Control, Cd82ΔEC, and Cd82ΔEC;Cdc42ΔEC mice, n=10 mice per group. (e) Western blot analysis of FARP1 in the HUVECs transfected with control and FARP1 siRNA. (f) Efficiency of in vitro FARP1 delivery and expression with endothelium-specific hADv5 viruses. The bEnd.3 cells infected by the viruses encoding control or FARP1 were examined in Western blot. (g-h) Histochemistry analysis (H-E staining) (g) and immunofluorescence analysis (CD45 staining) (h) on lung sections of the mice delivered with HAdV5-control (HAdV5-Vector) or HAdV5-FARP1 viruses. Ten days after the viral injection, the mice were challenged with LPS (5 mg/kg, i.p.) for 18 h prior to euthanasia. (i) Quantifications of acute lung injury (violin plot), vascular leakage or Evans Blue extravasation (mean±SD), and leukocyte infiltration of the lungs (violin plot) from the mice administered with HAdV5-control or HAdV5-FARP1 viruses, n=6 mice/ group.

Source data

Extended Data Fig. 6 CD82 sustains RhoA activity in ECs.

(a) HUVECs transfected with CD82 or control siRNA were serum-starved overnight, stimulated with VEGF-A (50 ng/ml), and then lysed with RIPA buffer. GTP-bound RhoA was pulled down from the lysates with GST-Rhotekin RhoA-binding domain fusion and detected in Western blot. Other indicated proteins in the lysates were directly examined with Western blot. The ratios of GTP-bound RhoA/total RhoA were quantified based on the band densities of RhoA (mean±SD; n=3 individual experiments). (b) Localization of GFP-AHPH. MLECs cultured on coverslips were transfected with GFP-AHPH WT or GFP-AHPH A470D&E758K mutant and processed for confocal microscopic imaging. (c) CD82 silencing up-regulates p190RhoGAP activity in ECs. HUVECs transfected with CD82 siRNAs or control siRNA were serum-starved overnight and then stimulated with VEGF-A (50 ng/ml). After cell lysis with a lysis buffer containing 1% Triton X-100, the p190RhoGAP proteins in the lysates were immunoprecipitated and then immuno-blotted with phosphotyrosine mAb (4G10) and p190RhoGAPmAb. Other indicated proteins in the lysates were directly examined with Western blot. (d) Immunofluorescence staining of VE-cadherin and Rnd3 in the MLEC monolayers. Images were acquired with confocal microscopy. (e) Western blot analysis of Rnd3 in the HUVECs that were transfected with control siRNA and Rnd3 siRNA. (f) Histochemistry analysis (H-E staining) on the effects of CN04 and/or Y27632 on LPS-induced acute lung injuries in WT C57BL/6 mice.

Source data

Extended Data Fig. 7 CD82 sustains AC on ECs.

(a) MLECs were incubated with AlexaFluor488-C5-maleimide-conjugated OlyA and AlexaFluor594-C5-maleimide-conjugated ALOD4 for 10 min at RT. After 3 washes with PBS, the cells were imaged with confocal microscopy. Fluorescence intensity/cell was quantified (violin plot; n=25 cells/group). (b) Effects of CD82 silencing on cell surface levels of the lipids. Top panel: the cells were labeled with fluorescent probes on ice for 1 h and examined by flow cytometry (mean±SD; n=3 experiments). Bottom panel: schematic presentation of the lipids and their probes. (c). Effects of simvastatin on cell surface levels of the lipids. The cells were treated with DMSO or simvastatin (5.0 µM) for 24 h, then incubated with fluorescent probes on ice for 1 h, and examined by flow cytometry (mean±SD; n=3 experiments). (d) MLECs treated with DMSO and cholesterol (0.1 mM) for 24 h were labeled with fluorescent probes on ice for 1 h and examined by flow cytometry (mean±SD; n=3 experiments). (e) The ECs were treated with MβCD (1.0 µM) for 1 h or cholesterol (0.1 mM) for 24 h, then incubated with the fluorescent probes on ice for 1 h, and examined by flow cytometry (mean±SD; n=3 experiments). (f) The cells treated with LPS (100 ng/mL for 1 h) or VEGF-A164 (50 ng/mL for 1 h) were labeled with fluorescent ALOD4 on ice for 1 h and examined by flow cytometry (mean±SD; n=3 experiments). (g) MLEC monolayers were treated with SMase (100 U/mL) for 30 min or simvastatin (5.0 µM) for 24 h and examined for basal permeability with FITC-dextran (250 kDa) diffusion assay (mean±SD; n=4 experiments). (h)-(i) The bEnd.3 monolayers were treated with cholesterol (0.1 mM) for 24 h or MβCD (1.0 µM) for 1 h and then examined for LPS- or VEGF-A164-stimulated permeability (h) or basal permeability (i), with FITC-dextran (250 kDa) diffusion assay (mean±SD; n=4 experiments). (j) The cells were pretreated with DMSO, cholesterol (0.1 mM), or simvastatin (5.0 µM) for 24 h or with SMase (100 U/mL) or ALOD4 (3.0 µM) for 30 min. The cell surface VE-cadherin was measured by flow cytometry (mean±SD; n=3 experiments).

Source data

Extended Data Fig. 8 AC roles in CD82-facilitated vascular leakage and CD82-tuned RhoA-versus-Cdc42 balance.

(a) The monolayers were treated with indicated ceramide species (20 µM) for 24 h and then examined in FITC-dextran (250 kDa) diffusion assay (mean±SD; n=4 experiments). (b) The cells were treated with 25-HC (0.2 µM) for 3 h, followed by flow cytometry analysis on AC levels at the cell surfaces (mean±SD; n=4 experiments). (c)-(d) The monolayers were treated with 25-HC (0.2 µM) for 3 h and examined for endothelial permeability (mean±SD, n=4 experiments) with FITC-dextran (250 kDa) diffusion assay, under VEGF-A164 (50 ng/mL) stimulated (c) or basal (d) condition. (e) The cells were preincubated with intact ALOD4 peptide (3.0 µM) at 37 °C for 30 min and then examined by flow cytometry with Alexa488-conjugated ALOD4 for the AC level (mean±SD; n=3 experiments). (f)-(g) The monolayers were preincubated with intact ALOD4 peptide (3.0 µM) at 37 °C for 30 min, followed by FITC-dextran diffusion assay (mean±SD, n=4 experiments), under VEGF- or LPS-stimulated (f) or basal (g) condition. (h) The cells were treated with PBS, simvastatin (5.0 µM for 24 h), or intact ALOD4 peptide (3.0 µM for 30 min), followed by LPS stimulation for 24 h. TNF-α in the cell culture supernatants were measured by ELISA (mean±SD; n=4 experiments). (i) The cells were pretreated with simvastatin (5 µM) for 24 h and then examined for RhoA and Cdc42 activities with G-LISA assays. Their relative functionality is present as the ratio of Cdc42 activity to RhoA activity (mean±SD). (j) The cells were treated with PBS or simvastatin (5.0 µM) for 24h, stained for Cdc42-GTP and CD44, and imaged with confocal microscopy. Arrows: Cdc42-GTP-localized cell-cell junction. (k) The live cells were stained with fluorescent OlyA and ALOD4, fixed, and imaged with structured illumination microscopy. (l) The cells were treated with DMSO, simvastatin (5.0 µM), or cholesterol (0.1 mM) for 24 h, followed by the Cdc42-FARP1 PLA. (m) MDA-MB-231 cells were incubated with His-tagged ALOD4 at 37 °C for 30 min and lyzed by sonication for immunoprecipitations with the indicated Abs, shown in Fig. 6f, followed by immunoblot with the indicated mAbs.

Source data

Extended Data Fig. 9 Effects of CD82 removal on subcellular distributions of cholesterol, AC, and ORP2, and interactions between tetraspanins and OSBP or ORPs.

(a) Confocal microscopic images of TopFluor® cholesterol (TF-cholesterol) and lysotracker staining in bEnd.3-Control and -CD82 KD transfectant cells. (b) Localization of ORP1 in MLECs. Immunofluorescence of ORP1 was performed with its Ab that recognizes both ORP1L and ORP1S forms. (c) Subcellular distribution and colocalization of ORP2 and AC. Immunofluorescence and fluorescence staining for ORP2 and ALOD4, respectively, in MLECs were imaged with confocal microscopy, and the ORP2-ALOD4 colocalization was quantified and presented as Pearson’s coefficient (Box&Whisker; n=20 cells from 3 independent experiments). (d) Western blot of ORP2 in the bEnd.3 ECs that were transiently transfected with control siRNA or ORP2 siRNA. (e) PLA analysis on the interactions of CD82 with OSBP and ORPs in HUVECs. Shown are representative fluorescence microscopic images of the PLA reactions. (f) PLA analysis on the interactions of ORP2 with tetraspanins in HUVECs. Shown are representative fluorescence microscopic images of the PLA reactions. (g) Representative FRET images of CFP-ORP2 in the MDA-MB-231 cells that transiently express only CFP-ORP2 construct. (h) Representative fluorescence microscopic images of the PLA between ORP2 and CD82 proteins in Du145-Mock, -CD82 WT, and -CD82 CACA mutant transfectant cells. Of note, Du145 cells do not express endogenous CD82.

Source data

Extended Data Fig. 10 Schematic summarization of the findings.

(a) Molecular and cellular mechanisms by which tetraspanin CD82 regulates AC and its non-vesicular transfer and then GEF/GAP-Rho small GTPases signaling, to alter the inflammatory status of cells and tissues. (b) New therapeutic strategies against inflammation arisen from this study. Red thunder signs denote functional blocking, and green asterisk signs indicate functional strengthening.

Supplementary information

Supplementary Information

Supplementary Fig. 1 and Supplementary Tables 1–3.

Reporting Summary

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 1

Unprocessed western blots and/or gels.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots and/or gels.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 6

Unprocessed western blots and/or gels.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed western blots and/or gels.

Source Data Fig. 8

Statistical source data.

Source Data Fig. 8

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 2

Statistical source data.

Source Data Extended Data Fig. 2

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 4

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 5

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots and/or gels.

Source Data Extended Data Fig. 9

Statistical source data.

Source Data Extended Data Fig. 9

Unprocessed western blots and/or gels.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ding, Y., Chen, J., Liu, S. et al. Tetraspanin-enriched membrane domains regulate vascular leakage by altering membrane cholesterol accessibility to balance antagonistic GTPases. Nat Cardiovasc Res 4, 1011–1033 (2025). https://doi.org/10.1038/s44161-025-00686-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s44161-025-00686-2

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