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:

Interaction between integrin α5 and PDE4D regulates endothelial inflammatory signalling

Abstract

Atherosclerosis is primarily a disease of lipid metabolism and inflammation; however, it is also closely associated with endothelial extracellular matrix (ECM) remodelling, with fibronectin accumulating in the laminin–collagen basement membrane. To investigate how fibronectin modulates inflammation in arteries, we replaced the cytoplasmic tail of the fibronectin receptor integrin α5 with that of the collagen/laminin receptor integrin α2. This chimaera suppressed inflammatory signalling in endothelial cells on fibronectin and in knock-in mice. Fibronectin promoted inflammation by suppressing anti-inflammatory cAMP. cAMP was activated through endothelial prostacyclin secretion; however, this was ECM-independent. Instead, cells on fibronectin suppressed cAMP via enhanced phosphodiesterase (PDE) activity, through direct binding of integrin α5 to phosphodiesterase-4D5 (PDE4D5), which induced PP2A-dependent dephosphorylation of PDE4D5 on the inhibitory site Ser651. In vivo knockdown of PDE4D5 inhibited inflammation at athero-prone sites. These data elucidate a molecular mechanism linking ECM remodelling and inflammation, thereby identifying a new class of therapeutic targets.

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: Integrin α subunit cytoplasmic tails determine ECM-specific inflammatory signalling.
Figure 2: Integrin chimaera knock-in mouse showed reduced inflammation in artery.
Figure 3: Prostacyclin mediates shear-dependent PKA activation but is ECM-independent.
Figure 4: Involvement of PDE4D in ECM-dependent inflammatory signalling.
Figure 5: Mapping the integrin binding site on PDE4D5.
Figure 6: ECM-dependent regulation of PDE4D phosphorylation.
Figure 7: In vivo PDE4D knockdown reduces flow-dependent inflammation.

Similar content being viewed by others

References

  1. Galkina, E. & Ley, K. Immune and inflammatory mechanisms of atherosclerosis. Annu. Rev. Immunol. 27, 165–197 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Zarins, C. K. et al. Carotid bifurcation atherosclerosis. Quantitative correlation of plaque localization with flow velocity profiles and wall shear stress. Circ. Res. 53, 502–514 (1983).

    Article  CAS  PubMed  Google Scholar 

  3. Caro, C. G., Fitz-Gerald, J. M. & Schroter, R. C. Arterial wall shear and distribution of early atheroma in man. Nature 223, 1159–1161 (1969).

    Article  CAS  PubMed  Google Scholar 

  4. Libby, P., Ridker, P. M. & Hansson, G. K. Progress and challenges in translating the biology of atherosclerosis. Nature 473, 317–325 (2011).

    Article  CAS  PubMed  Google Scholar 

  5. Conway, D. E. & Schwartz, M. A. Flow-dependent cellular mechanotransduction in atherosclerosis. J. Cell Sci. 126, 5101–5109 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gaudet, A. D. & Popovich, P. G. Extracellular matrix regulation of inflammation in the healthy and injured spinal cord. Exp. Neurol. 258, 24–34 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Bollyky, P., Bogdani, M., Bollyky, J., Hull, R. & Wight, T. The role of hyaluronan and the extracellular matrix in islet inflammation and immune regulation. Curr. Diab. Rep. 12, 471–480 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Papageorgiou, A.-P. & Heymans, S. Interactions between the extracellular matrix and inflammation during viral myocarditis. Immunobiology 217, 503–510 (2012).

    Article  CAS  PubMed  Google Scholar 

  9. Grant, D. S., Kleinman, H. K. & Martin, G. R. The role of basement membranes in vascular development. Ann. NY Acad. Sci. 588, 61–72 (1990).

    Article  CAS  PubMed  Google Scholar 

  10. Kim, S., Bell, K., Mousa, S. A. & Varner, J. A. Regulation of angiogenesis in vivo by ligation of integrin α5β1 with the central cell-binding domain of fibronectin. Am. J. Pathol. 156, 1345–1362 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Chiang, H.-Y., Korshunov, V. A., Serour, A., Shi, F. & Sottile, J. Fibronectin is an important regulator of flow-induced vascular remodeling. Arterioscler. Thromb. 29, 1074–1079 (2009).

    Article  CAS  Google Scholar 

  12. Chiu, C.-H., Chou, C.-W., Takada, S. & Liu, Y.-W. Development and fibronectin signaling requirements of the zebrafish interrenal vessel. PLoS ONE 7, e43040 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Orr, A. W. et al. The subendothelial extracellular matrix modulates NF-κB activation by flow: a potential role in atherosclerosis. J. Cell Biol. 169, 191–202 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Tan, M. H. et al. Deletion of the alternatively spliced fibronectin EIIIA domain in mice reduces atherosclerosis. Blood 104, 11–18 (2004).

    Article  CAS  PubMed  Google Scholar 

  15. Babaev, V. R. et al. Absence of regulated splicing of fibronectin EDA exon reduces atherosclerosis in mice. Atherosclerosis 197, 534–540.

  16. Rohwedder, I. et al. Plasma fibronectin deficiency impedes atherosclerosis progression and fibrous cap formation. EMBO Mol. Med. 4, 564–576 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Nagel, T., Resnick, N., Atkinson, W. J., Dewey, C. F. Jr & Gimbrone, M. A. Jr Shear stress selectively upregulates intercellular adhesion molecule-1 expression in cultured human vascular endothelial cells. J. Clin. Invest. 94, 885–891 (1994).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Bao, X., Lu, C. & Frangos, J. A. Temporal gradient in shear but not steady shear stress induces PDGF-A and MCP-1 expression in endothelial cells: role of NO, NFκB, and egr-1. Arterioscler. Thromb. 19, 996–1003 (1999).

    Article  CAS  Google Scholar 

  19. Orr, A. W. et al. Matrix-specific p21-activated kinase activation regulates vascular permeability in atherogenesis. J. Cell Biol. 176, 719–727 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Orr, A. W., Hahn, C., Blackman, B. R. & Schwartz, M. A. p21-activated kinase signaling regulates oxidant-dependent NF-κB activation by flow. Circ. Res. 103, 671–679 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Hahn, C., Orr, A. W., Sanders, J. M., Jhaveri, K. A. & Schwartz, M. A. The subendothelial extracellular matrix modulates JNK activation by flow. Circ. Res. 104, 995–1003 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Funk, S. D. et al. Matrix-specific protein kinase a signaling regulates p21-activated kinase activation by flow in endothelial cells. Circ. Res. 106, 1394–1403 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Yurdagul, A. et al. Altered nitric oxide production mediates matrix-specific PAK2 and NF-κB activation by flow. Mol. Biol. Cell 24, 398–408 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yurdagul, A. et al. α5β1 integrin signaling mediates oxidized low-density lipoprotein-induced inflammation and early atherosclerosis. Arterioscler. Thromb. 34, 1362–1373 (2014).

    Article  CAS  Google Scholar 

  25. Orr, A. W., Ginsberg, M. H., Shattil, S. J., Deckmyn, H. & Schwartz, M. A. Matrix-specific suppression of integrin activation in shear stress signaling. Mol. Biol. Cell 17, 4686–4697 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Madamanchi, A., Santoro, S. A. & Zutter, M. M. in I Domain Integrins (ed. Gullberg, D.) 41–60 (Springer, 2014).

    Google Scholar 

  27. Wang, C., Baker, B. M., Chen, C. S. & Schwartz, M. A. Endothelial cell sensing of flow direction. Arterioscler. Thromb. 33, 2130–2136 (2013).

    Article  CAS  Google Scholar 

  28. Khachigian, L. M., Resnick, N., Gimbrone, M. A. Jr & Collins, T. Nuclear factor-kappa B interacts functionally with the platelet-derived growth factor B-chain shear-stress response element in vascular endothelial cells exposed to fluid shear stress. J. Clin. Invest. 96, 1169–1175 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Glagov, S., Zarins, C., Giddens, D. & Ku, D. N. Hemodynamics and atherosclerosis. Insights and perspectives gained from studies of human arteries. Arch. Pathol. Lab. Med. 112, 1018–1031 (1988).

    CAS  PubMed  Google Scholar 

  30. Frangos, S. G., Gahtan, V. & Sumpio, B. Localization of atherosclerosis: role of hemodynamics. Arch. Surg. 134, 1142–1149 (1999).

    Article  CAS  PubMed  Google Scholar 

  31. Frangos, J. A., Eskin, S. G., McIntire, L. V. & Ives, C. L. Flow effects on prostacyclin production by cultured human endothelial cells. Science 227, 1477–1479 (1985).

    Article  CAS  PubMed  Google Scholar 

  32. Fetalvero, K. M., Martin, K. A. & Hwa, J. Cardioprotective prostacyclin signaling in vascular smooth muscle. Prostaglandins Other Lipid Mediat. 82, 109–118 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. Stitham, J., Midgett, C., Martin, K. & Hwa, J. Prostacyclin: an inflammatory paradox. Front. Pharmacol. 2, 24 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Tsai, M.-C. et al. Shear stress induces synthetic-to-contractile phenotypic modulation in smooth muscle cells via peroxisome proliferator-activated receptor α/δ activations by prostacyclin released by sheared endothelial cells. Circ. Res. 105, 471–480 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Maurice, D. H. et al. Advances in targeting cyclic nucleotide phosphodiesterases. Nat. Rev. Drug Discov. 13, 290–314 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Baillie, G. S. Compartmentalized signalling: spatial regulation of cAMP by the action of compartmentalized phosphodiesterases. FEBS J. 276, 1790–1799 (2009).

    Article  CAS  PubMed  Google Scholar 

  37. Muzaffar, S., Jeremy, J. Y., Angelini, G. D. & Shukla, N. NADPH oxidase 4 mediates upregulation of type 4 phosphodiesterases in human endothelial cells. J. Cell. Physiol. 227, 1941–1950 (2012).

    Article  CAS  PubMed  Google Scholar 

  38. Wang, J., Bingaman, S. & Huxley, V. H. Intrinsic sex-specific differences in microvascular endothelial cell phosphodiesterases. Am. J. Physiol. Heart Circ. Physiol. 298, H1146–H1154 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Netherton, S. J. & Maurice, D. H. Vascular endothelial cell cyclic nucleotide phosphodiesterases and regulated cell migration: implications in angiogenesis. Mol. Pharmacol. 67, 263–272 (2005).

    Article  CAS  PubMed  Google Scholar 

  40. Thompson, W. J. et al. Regulation of cyclic AMP in rat pulmonary microvascular endothelial cells by rolipram-sensitive cyclic AMP phosphodiesterase (PDE4). Biochem. Pharmacol. 63, 797–807 (2002).

    Article  CAS  PubMed  Google Scholar 

  41. McCormick, K. & Baillie, G. S. Compartmentalisation of second messenger signalling pathways. Curr. Opin. Genet. Dev. 27, 20–25 (2014).

    Article  CAS  PubMed  Google Scholar 

  42. Pullamsetti, S. S. et al. cAMP phosphodiesterase inhibitors increases nitric oxide production by modulating dimethylarginine dimethylaminohydrolases. Circulation 123, 1194–1204 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Hildebrand, J. D., Schaller, M. D. & Parsons, J. T. Identification of sequences required for the efficient localization of the focal adhesion kinase, pp125FAK, to cellular focal adhesions. J. Cell Biol. 123, 993–1005 (1993).

    Article  CAS  PubMed  Google Scholar 

  44. Pfaff, M., Liu, S., Erle, D. J. & Ginsberg, M. H. Integrin β cytoplasmic domains differentially bind to cytoskeletal proteins. J. Biol. Chem. 273, 6104–6109 (1998).

    Article  CAS  PubMed  Google Scholar 

  45. MacKenzie, S. J., Baillie, G. S., McPhee, I., Bolger, G. B. & Houslay, M. D. ERK2 mitogen-activated protein kinase binding, phosphorylation, and regulation of the PDE4D cAMP-specific phosphodiesterases: The involvement of COOH-terminal docking sites and NH2-terminal UCR regions. J. Biol. Chem. 275, 16609–16617 (2000).

    Article  CAS  PubMed  Google Scholar 

  46. Dahlman, J. E. et al. In vivo endothelial siRNA delivery using polymeric nanoparticles with low molecular weight. Nat. Nanotech. 9, 648–655 (2014).

    Article  CAS  Google Scholar 

  47. Plump, A. S. et al. Severe hypercholesterolemia and atherosclerosis in apolipoprotein E-deficient mice created by homologous recombination in ES cells. Cell 71, 343–353 (1992).

    Article  CAS  PubMed  Google Scholar 

  48. Smith, J. A. Neutrophils, host defense, and inflammation: a double-edged sword. J. Leukoc. Biol. 56, 672–686 (1994).

    Article  CAS  PubMed  Google Scholar 

  49. Morganti-Kossmann, M. C., Rancan, M., Stahel, P. F. & Kossmann, T. Inflammatory response in acute traumatic brain injury: a double-edged sword. Curr. Opin. Crit. Care 8, 101–105 (2002).

    Article  PubMed  Google Scholar 

  50. Arroyo, A. G. & Iruela-Arispe, M. L. Extracellular matrix, inflammation, and the angiogenic response. Cardiovasc. Res. 86, 226–235 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Sorokin, L. The impact of the extracellular matrix on inflammation. Nat. Rev. Immunol. 10, 712–723 (2010).

    Article  CAS  PubMed  Google Scholar 

  52. Stratman, A. N., Malotte, K. M., Mahan, R. D., Davis, M. J. & Davis, G. E. Pericyte recruitment during vasculogenic tube assembly stimulates endothelial basement membrane matrix formation. Blood 114, 5091–5101 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Davis, G. E. & Senger, D. R. Endothelial extracellular matrix: biosynthesis, remodeling, and functions during vascular morphogenesis and neovessel stabilization. Circ. Res. 97, 1093–1107 (2005).

    Article  CAS  PubMed  Google Scholar 

  54. Hahn, C. & Schwartz, M. A. The role of cellular adaptation to mechanical forces in atherosclerosis. Arterioscler. Thromb. 28, 2101–2107 (2008).

    Article  CAS  Google Scholar 

  55. Jongstra-Bilen, J. et al. Low-grade chronic inflammation in regions of the normal mouse arterial intima predisposed to atherosclerosis. J. Exp. Med. 203, 2073–2083 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Kakolyris, S., Karakitsos, P., Tzardi, M. & Agapitos, E. Immunohistochemical detection of fibronectin in early and advanced atherosclerosis. In Vivo 9, 35–40 (1995).

    CAS  PubMed  Google Scholar 

  57. Ghosh, S. et al. Systems genetics analysis of genome-wide association study reveals novel associations between key biological processes and coronary artery disease. Arterioscler. Thromb. 35, 1712–1722 (2015).

    Article  CAS  Google Scholar 

  58. Lee, G.-S. et al. The calcium-sensing receptor regulates the NLRP3 inflammasome through Ca2+ and cAMP. Nature 492, 123–127 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Sokolowska, M. et al. Prostaglandin E2 inhibits NLRP3 inflammasome activation through EP4 receptor and intracellular cyclic AMP in human macrophages. J. Immunol. 194, 5472–5487 (2015).

    Article  CAS  PubMed  Google Scholar 

  60. Yan, Y. et al. Dopamine controls systemic inflammation through inhibition of NLRP3 inflammasome. Cell 160, 62–73 (2015).

    Article  CAS  PubMed  Google Scholar 

  61. Libby, P., Okamoto, Y., Rocha, V. Z. & Folco, E. Inflammation in atherosclerosis transition from theory to practice. Circ. J. 74, 213–220 (2010).

    Article  CAS  PubMed  Google Scholar 

  62. Ni, H. et al. Plasma fibronectin promotes thrombus growth and stability in injured arterioles. Proc. Natl Acad. Sci. USA 100, 2415–2419 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Sakai, T. et al. Plasma fibronectin supports neuronal survival and reduces brain injury following transient focal cerebral ischemia but is not essential for skin-wound healing and hemostasis. Nat. Med. 7, 324–330 (2001).

    Article  CAS  PubMed  Google Scholar 

  64. Allport, J. R. et al. Neutrophils from MMP-9- or neutrophil elastase-deficient mice show no defect in transendothelial migration under flow in vitro. J. Leukoc. Biol. 71, 821–828 (2002).

    CAS  PubMed  Google Scholar 

  65. Tian, J., Alimperti, S., Lei, P. & Andreadis, S. T. Lentiviral microarrays for real-time monitoring of gene expression dynamics. Lab Chip 10, 1967–1975 (2010).

    Article  CAS  PubMed  Google Scholar 

  66. Kim, H. W. et al. Cyclic AMP controls mTOR through regulation of the dynamic interaction between Rheb and phosphodiesterase 4D. Mol. Cell. Biol. 30, 5406–5420 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Love, K. T. et al. Lipid-like materials for low-dose, in vivo gene silencing. Proc. Natl Acad. Sci. USA 107, 1864–1869 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  68. Nam, D. et al. Partial carotid ligation is a model of acutely induced disturbed flow, leading to rapid endothelial dysfunction and atherosclerosis. Am. J. Physiol. Heart Circ. Physiol. 297, H1535–H1543 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Paulucci-Holthauzen, A. A. & O’Connor, K. L. Use of pseudosubstrate affinity to measure active protein kinase A. Anal. Biochem. 355, 175–182 (2006).

    Article  CAS  PubMed  Google Scholar 

  70. Yang, T.-T., Sinai, P. & Kain, S. R. An acid phosphatase assay for quantifying the growth of adherent and nonadherent cells. Anal. Biochem. 241, 103–108 (1996).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank K. Yamada (NIH, USA), D. Calderwood (Yale University, USA), H. Kim (POSTECH, Korea) and A. Jayaraman (Texas A&M University, USA) for providing reagents, and J. Hwa (Yale University, USA) for advice on prostacyclin experiments. Lipid analysis was done by the Yale Mouse Phenotypic Center, supported by a U24 DK059635 grant. This work was funded by a National Institutes of Health grant 5R01HL75092 to M.A.S. G.B. is funded by an MRC project grant (MR/J007412/1). We are grateful to R. Webber and N. Copeland for maintaining the mouse colonies used in this study.

Author information

Authors and Affiliations

Authors

Contributions

S.Y. and M.A.S. designed the project. S.Y. performed in vitro experiments and M.B. designed and performed in vivo experiments with the aid of S.Y. J.E.D. prepared and provided nanoparticles. B.G.C. contributed PDE4D5 imaging. R.T.C. performed in vitro PDE assay. R.L. and D.G.A. provided advice on nanoparticle formulation. S.Y. and M.A.S. wrote the manuscript with the contribution of all the authors.

Corresponding author

Correspondence to Martin A. Schwartz.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Characterization of α5/2 chimera endothelial cells.

(a) BAECs expressing human integrin wild type α5 or the α5/2 chimera were stained with mAb16 that recognizes human integrin α5 extracellular domain. (b) Wild type α5 or the α5/2 chimera cells were lysed and immunoprecipitated with mAb16 to isolate exogenous human integrin α proteins. Western blots with antibodies against the α5 and α2 cytoplasmic tails confirm the sequences (upper and middle panels), and show similar pairing with the β1 subunit (lower panel). (c,d) Wild type α5 and α5/2 chimera cells spreading on fibronectin. BAECs expressing wild type integrin α5 or the chimera were detached and replated on dishes coated with fibronectin (10 μg/ml) for the indicated times. The cells were either fixed and stained with wheat germ agglutinin (c) or lysed and subjected to immunoblotting for FAK phosphorylation (Y397) (d). (e) Wild type α5 or α5/2 chimera cells were plated on fibronectin and grown to monolayer and then kept in low-serum media (1% FBS) for two days to induce fibronectin fibrillogenesis. The cells were fixed and stained for fibronectin. For each condition, n = 30 images pooled across three independent experiments were averaged. The box plot shows the median, with upper and lower percentiles, and the bars show maxima and minima values. (f) Wild type α5 or α5/2 chimera cells were plated on fibronectin and sheared for 36 hrs (20dynes/cm2). Cells were stained with phalloidin and Hoechst and alignment in the direction of flow was quantified (±30°). Scale bars: 50 μm. n = 10 images (60-100 cells/field) were used for quantification for each condition. Error bars are SEM.

Supplementary Figure 2 PDE4D5 is responsible for ECM-dependent inflammation in endothelial cells.

(a) BAECs on FN or collagen were treated with rolipram (1 μM) and assayed for AMPK activation. (b) Immunoblotting HUVEC lysate with pan PDE4D antibody detects the major band co-migrating with reference PDE4D5 (left panel). The band was diminished by transfection with PDE4D siRNA, demonstrating specificity. Similarly, PDE4D5 was the only isoform detectable in BAEC lysate with immunobotting using pan PDE4D antibody or PDE4D5 specific antibody (right two panels). indicates non-specific bands. (c) BAECs expressing wild type PDE4D5 or FAT-PDE4D5 were plated on MG for 1hr to monitor GFP signal.

Supplementary Figure 3 PP2A regulates PDE4D5 phosphorylation and inflammation.

(a) Identification of PP2A as a PDE4D5 binding protein. FLAG-PDE4D5 expressing BAECs on FN were lysed and immunoprecipitated with FLAG antibody. Bound protein with size of 35 kDa was submitted for mass analysis and identified as PP2A catalytic subunit. (b) BAECs expressing FLAG-tagged PDE4D5 were plated on either FN or matrigel for indicated times. PDE4D5 was immunoprecipitated with FLAG antibody and eluted with FLAG peptides. Similar results were obtained in 3 experiments. (c) BAECs expressing PDE4D5 wild type were kept in suspension for 90 min then replated on FN-coated dishes for the indicated times. For okadaic acid treatment, cells in suspension were added with 5 nM OA for last 20 min before replating on FN. S651 phosphorylation was assayed by Western blotting (n = 4 independent experiments). (d) BAECs expressing PDE4D5 were plated on FN for 5 hr then pretreated with DMSO or okadaic acid (OA, 5 nM) and then stimulated with IL1β for 30 min. S651 phosphorylation was assayed by Western blotting (n = 3 independent experiments). (e) BAECs expressing PDE4D5 were transfected with a siRNAs targeting PP2A catalytic subunit. The cells were replated on FN then subject to laminar shear for 30 min. (f) BAECs were plated on FN for 5 hr then pretreated with DMSO or okadaic acid (OA, 5 nM) and then stimulated with IL1β for 30 min. NFκB activity was assayed by Western blotting (n = 3 independent experiments). (g) BAECs were transfected with two different siRNAs targeting the PP2A catalytic subunit. The cells were replated on FN then subject to oscillatory shear for 2 hrs (n = 3 independent experiments). Data are represented as means ± SEM. p < 0.05 by one way ANOVA (d,g) or two-tailed t-test (c,f). Source data are provided in Supplementary Table 1.

Supplementary Figure 4 Effect of PP2A inhibition on endothelial cell adhesion.

(a) BAECs were transfected with PP2A siRNA or treated with okadaic acid (OA, 5 nM, 1hr) and plated on FN (10 μg/ml) for 1hr. Cell adhesion was quantified as described in Supplementary Fig. 2 (n = 3 independent experiments) and cell morphology was examined after fixation and phalloidin and nuclear staining. Error bars are SEM. Scale bar: 100 μm. (b) BAECs treated as (a) were plated on FN (10 μg/ml) for indicated times and FAK phosphorylation was measured by Western blotting.

Supplementary Figure 5 In vivo knock-down of endothelial PDE4D.

(a) NIH3T3 cells were transfected with either luciferase siRNA or mouse PDE4D siRNA used for in vivo knock-down. After 72h, PDE4D5 was assayed by Western blotting with tubulin as a loading control. (b) Nano-particles containing PDE4D siRNA or luciferase siRNA (1 mg/kg) were injected intravenously. After two weeks, mouse aortas were isolated, and endothelial expression of PDE4D was assayed by qPCR (n = 4). (c) Serum lipid profile of α5/2; ApoE(-/-) mice after high fat diet (n = 6 mice). Data are represented as means ± SEM. p < 0.05 by two-tailed t-test. Source data are provided in Supplementary Table 1.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1657 kb)

Supplementary Table 1

Supplementary Information (XLS 47 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yun, S., Budatha, M., Dahlman, J. et al. Interaction between integrin α5 and PDE4D regulates endothelial inflammatory signalling. Nat Cell Biol 18, 1043–1053 (2016). https://doi.org/10.1038/ncb3405

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3405

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