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.

  • Review Article
  • Published:

Hemodynamic shear stress and the endothelium in cardiovascular pathophysiology

Abstract

Endothelium lining the cardiovascular system is highly sensitive to hemodynamic shear stresses that act at the vessel luminal surface in the direction of blood flow. Physiological variations of shear stress regulate acute changes in vascular diameter and when sustained induce slow, adaptive, structural-wall remodeling. Both processes are endothelium-dependent and are systemically and regionally compromised by hyperlipidemia, hypertension, diabetes and inflammatory disorders. Shear stress spans a range of spatiotemporal scales and contributes to regional and focal heterogeneity of endothelial gene expression, which is important in vascular pathology. Regions of flow disturbances near arterial branches, bifurcations and curvatures result in complex spatiotemporal shear stresses and their characteristics can predict atherosclerosis susceptibility. Changes in local artery geometry during atherogenesis further modify shear stress characteristics at the endothelium. Intravascular devices can also influence flow-mediated endothelial responses. Endothelial flow-induced responses include a cell-signaling repertoire, collectively known as mechanotransduction, that ranges from instantaneous ion fluxes and biochemical pathways to gene and protein expression. A spatially decentralized mechanism of endothelial mechanotransduction is dominant, in which deformation at the cell surface induced by shear stress is transmitted as cytoskeletal tension changes to sites that are mechanically coupled to the cytoskeleton. A single shear stress mechanotransducer is unlikely to exist; rather, mechanotransduction occurs at multiple subcellular locations.

Key Points

  • Hemodynamic forces, and in particular shear stresses, are regulators of many physiologic and pathologic aspects of endothelial function in the cardiovascular system

  • In vivo and in vitro global endothelial analyses reveal that endothelial phenotypes are heterogeneous over regional and focal length scales, which links flow characteristics to cardiovascular disease protection, susceptibility and development

  • Endothelial responses are sensitive to variations in the characteristics of flow that generate shear stresses; regions with oscillating shear stress and flow reversal correspond with pathologic changes in the artery wall and are a risk factor for atherosclerosis-susceptibility

  • When shear stresses deform the endothelium, a mechanical perturbation is communicated via the cytoskeleton to multiple sites of mechanotransduction, which include cell–matrix adhesion sites, intercellular junctions and the nuclear membrane

  • Endothelial responses that are specific to shear stress offer potential therapeutic pharmacological targets, although a single mechanosensor is unlikely to exist

  • Beneficial systemic effects include maintenance of arterial hemodynamics within normal limits through antihypertensive therapies, regular exercise to promote continuous adaptive remodeling and inhibition of endothelial dysfunction, and (when intervention is required) better design of intravascular devices to optimize flow characteristics

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Flow separations at an arterial branch can predispose or contribute to pathogenesis.
Figure 2: Flow separations at a stenosis that predispose to or contribute to pathogenesis.
Figure 3: Flow separations around a stent strut that predispose to or contribute to pathogenesis.
Figure 4: The decentralized model of endothelial mechanotransduction by shear stress.

Similar content being viewed by others

References

  1. Pohl U et al. (1986) Crucial role of endothelium in the vasodilator response to increased flow in vivo. Hypertension 8: 37–44

    Article  CAS  Google Scholar 

  2. Moncada S (2006) Adventures in vascular biology: a tale of two mediators. Philos Trans R Soc Lond B Biol Sci 361: 735–759

    Article  CAS  Google Scholar 

  3. Corson MA et al. (1996) Phosphorylation of endothelial nitric oxide synthase in response to fluid shear stress. Circ Res 79: 984–991

    Article  CAS  Google Scholar 

  4. Griffith TM (2002) Endothelial control of vascular tone by nitric oxide and gap junctions: a haemodynamic perspective. Biorheology 39: 307–318

    CAS  PubMed  Google Scholar 

  5. Davies PF (1995) Flow-mediated endothelial mechanotransduction. Physiol Rev 75: 519–560

    Article  CAS  Google Scholar 

  6. Langille BL and O'Donnell F (1986) Reductions in arterial diameter produced by chronic decreases in blood flow are endothelium-dependent. Science 231: 405–407

    Article  CAS  Google Scholar 

  7. Zhang H et al. (2008). Heparin-binding epidermal growth factor-like growth factor signaling in flow-induced arterial remodeling. Circ Res 102: 1275–1285

    Article  CAS  Google Scholar 

  8. Aird WC (Ed.; 2007) Endothelial Biomedicine. New York: Cambridge University Press

    Book  Google Scholar 

  9. Lucitti JL et al. (2007). Vascular remodeling of the mouse yolk sac requires hemodynamic force. Development 134: 3317–3326

    Article  CAS  Google Scholar 

  10. Teichert AM et al. (2008) Endothelial nitric oxide synthase gene expression during murine embryogenesis: commencement of expression in the embryo occurs with the establishment of a unidirectional circulatory system. Circ Res 103: 24–33

    Article  CAS  Google Scholar 

  11. Glagov S et al. (1988) Hemodynamics and atherosclerosis. Insights and perspectives gained from studies of human arteries. Arch Pathol Lab Med 112: 1018–1031

    CAS  PubMed  Google Scholar 

  12. Passerini AG et al. (2004) Coexisting proinflammatory and antioxidative endothelial transcription profiles in a disturbed flow region of the adult porcine aorta. Proc Natl Acad Sci USA 101: 2482–2487

    Article  CAS  Google Scholar 

  13. Volger OL et al. (2007) Distinctive expression of chemokines and transforming growth factor-beta signaling in human arterial endothelium during atherosclerosis. Am J Pathol 171: 326–337

    Article  CAS  Google Scholar 

  14. Cornhill JF and Roach MR (1976) A quantitative study of the localization of atherosclerotic lesions in the rabbit aorta. Atherosclerosis 23: 489–499

    Article  CAS  Google Scholar 

  15. Mattsson EJ et al. (1997) Increased blood flow induces regression of intimal hyperplasia. Arterioscler Thromb Vasc Biol 17: 2245–2249

    Article  CAS  Google Scholar 

  16. Green DJ et al. (2004) Effect of exercise training on endothelium-derived nitric oxide function in humans. J Physiol 561: 1–25

    Article  CAS  Google Scholar 

  17. Adams JA (2007) Therapeutic approaches to altering hemodynamic forces. In Endothelial Biomedicine, 1690–1697 (Ed Aird WC) New York: Cambridge University Press

    Chapter  Google Scholar 

  18. Suo J et al. (2008) Blood flow patterns in the proximal human coronary arteries: relationship to atherosclerotic plaque occurrence. Mol Cell Biomech 5: 9–18

    PubMed  Google Scholar 

  19. Steinman DA and Taylor CA (2005) Flow imaging and computing: large artery hemodynamics. Ann Biomed Eng 33: 1704–1709

    Article  Google Scholar 

  20. Davies PF et al. (1999) A spatial approach to transcriptional profiling: mechanotransduction and the focal origin of atherosclerosis. Trends Biotechnol 17: 347–351

    Article  CAS  Google Scholar 

  21. García-Cardeña G et al (2001) Mechanosensitive endothelial gene expression profiles: scripts for the role of hemodynamics in atherogenesis? Ann NY Acad Sci 947: 1–6

    Article  Google Scholar 

  22. Libby P (2000) Coronary artery injury and the biology of atherosclerosis: inflammation, thrombosis, and stabilization. Am J Cardiol 86: 3J–8J

    Article  Google Scholar 

  23. Folie BJ and McIntire LV (1989) Mathematical analysis of mural thrombogenesis. Concentration profiles of platelet-activating agents and effects of viscous shear flow. Biophys J 56: 1121–1141

    Article  CAS  Google Scholar 

  24. Duraiswamy N et al. (2005) Spatial distribution of platelet deposition in stented arterial models under physiologic flow. Ann Biomed Eng 33: 1767–1777

    Article  Google Scholar 

  25. DePaola N et al. (1992) Vascular endothelium responds to fluid shear stress gradients. Arterioscler Thromb 12: 1254–1257

    Article  CAS  Google Scholar 

  26. Moore JE and Berry JL (2002) Fluid and solid mechanical implications of vascular stenting. Ann Biomed Eng 30: 498–508

    Article  Google Scholar 

  27. Balakrishnan B et al. (2005) Strut position, blood flow, and drug deposition: implications for single and overlapping drug–eluting stents. Circulation 111: 2958–2965

    Article  Google Scholar 

  28. Kastrati A et al. (2001) Intracoronary stenting and angiographic results: strut thickness effect on restenosis outcome (ISAR-STEREO) trial. Circulation 103: 2816–2821

    Article  CAS  Google Scholar 

  29. Loth F et al. (2003) Transitional flow at the venous anastomosis of an arteriovenous graft: potential activation of the ERK1/2 mechanotransduction pathway. J Biomech Eng 125: 49–61

    Article  Google Scholar 

  30. Hajra L et al. (2000) The NF-κB signal transduction pathway in aortic endothelial cells is primed for activation in regions predisposed to atherosclerotic lesion formation. Proc Natl Acad Sci USA 97: 9052–9057

    Article  CAS  Google Scholar 

  31. Caro CG et al. (1969) Arterial wall shear and distribution of early atheroma in man. Nature 223: 1159–1161

    Article  CAS  Google Scholar 

  32. Fry DL (1973) Atherogenesis: initiating factors. CIBA Found Symposium 12: 96–118

    Google Scholar 

  33. Dewey CF et al. (1981) The dynamic response of vascular endothelial cells to fluid shear stress. J Biomech Eng 103: 177–188

    Article  Google Scholar 

  34. Levesque MJ and Nerem RM (1985) The elongation and orientation of cultured endothelial cells in response to shear stress. J Biomech Eng 107: 341–347

    Article  CAS  Google Scholar 

  35. Suo J et al. (2007) Hemodynamic shear stresses in mouse aortas: implications for atherogenesis. Arterioscler Thromb Vasc Biol 27: 346–351

    Article  CAS  Google Scholar 

  36. Wolf S and Werthessen NT (1979) Dynamics of arterial flow. Adv Exp Med Biol 115: 1–472

    Article  CAS  Google Scholar 

  37. Huo Y et al. (2007) Flow patterns in three-dimensional porcine epicardial coronary arterial tre. Am J Physiol Heart Circ Physiol 293: H2959–H2970

    Article  CAS  Google Scholar 

  38. Asakura T and Karino T (1990) Flow patterns and spatial distribution of atherosclerotic lesions in human coronary arteries. Circ Res 66: 1045–1066

    Article  CAS  Google Scholar 

  39. Dekker RJ et al. (2002) Prolonged fluid shear stress induces a distinct set of endothelial cell genes, most specifically lung Krüppel-like factor (KLF2). Blood 100: 1689–1698

    Article  CAS  Google Scholar 

  40. Dai G et al. (2007) Biomechanical forces in atherosclerosis-resistant vascular regions regulate endothelial redox balance via phosphoinositol 3-kinase/Akt-dependent activation of Nrf2. Circ Res 101: 723–733

    Article  CAS  Google Scholar 

  41. Ziegler T et al. (1998) Influence of oscillatory and unidirectional flow environments on the expression of endothelin and nitric oxide synthase in cultured endothelial cells. Arterioscler Thromb Vasc Biol 18: 686–692

    Article  CAS  Google Scholar 

  42. Cheng C et al. (2005) Shear stress affects the intracellular distribution of eNOS: direct demonstration by a novel in vivo technique. Blood 106: 3691–3698

    Article  CAS  Google Scholar 

  43. Cheng C et al. (2006) Atherosclerotic lesion size and vulnerability are determined by patterns of fluid shear stress. Circulation 113: 2744–2753

    Article  Google Scholar 

  44. Iiyama K et al. (1999) Patterns of vascular cell adhesion molecule-1 and intercellular adhesion molecule-1 expression in rabbit and mouse atherosclerotic lesions and at sites predisposed to lesion formation. Circ Res 85: 199–207

    Article  CAS  Google Scholar 

  45. De Nigris F et al. (2003) Beneficial effects of antioxidants and L-arginine on oxidation-sensitive gene expression and endothelial NO synthase activity at sites of disturbed shear stress. Proc Natl Acad Sci USA 100: 1420–1425

    Article  CAS  Google Scholar 

  46. Magid R and Davies PF (2005) Endothelial protein kinase C isoform identity and differential activity of PKCζ in an athero-susceptible region of porcine aorta. Circ Res 97: 443–449

    Article  CAS  Google Scholar 

  47. Zakkar M et al. (2008) Increased endothelial mitogen-activated protein kinase phosphatase-1 expression suppresses proinflammatory activation at sites that are resistant to atherosclerosis. Circ Res 103: 726–732

    Article  CAS  Google Scholar 

  48. Parmar KM et al. (2006) Integration of flow-dependent endothelial phenotypes by Krüppel-like factor 2. J Clin Invest 116: 49–58

    Article  CAS  Google Scholar 

  49. García-Cardeña G and Gimbrone MA (2006) Biomechanical modulation of endothelial phenotype: implications for health and disease. Handb Exp Pharmacol 176: 79–95

    Article  Google Scholar 

  50. Davies PF et al. (1984) Influence of hemodynamic forces on vascular endothelial function. In vitro studies of shear stress and pinocytosis in bovine aortic cells. J Clin Invest 73: 1121–1129

    Article  CAS  Google Scholar 

  51. Davies PF et al. (1986) Turbulent fluid shear stress induces vascular endothelial cell turnover in vitro. Proc Natl Acad Sci USA 83: 2114–2117

    Article  CAS  Google Scholar 

  52. Shyy YJ et al. (1994) Fluid shear stress induces a biphasic response of human monocyte chemotactic protein 1 gene expression in vascular endothelium. Proc Natl Acad Sci USA 91: 4678–4682

    Article  CAS  Google Scholar 

  53. Barbee KA et al. (1995) Subcellular distribution of shear stress at the surface of flow aligned and non-aligned endothelial monolayers. Am J Physiol 268: H1765–H1772

    CAS  PubMed  Google Scholar 

  54. Davies PF and Helmke BP (2008) Endothelial mechanotransduction. In Cellular Mechanotransduction: Diverse Perspectives from Molecules to Tissue, (Eds Mofrad RK and Kamm RD) New York: Cambridge University Press [In press]

    Google Scholar 

  55. Olesen S-P et al. (1988) Hemodynamic shear stress activates a K+ current in vascular endothelial cells. Nature 331: 168–170

    Article  CAS  Google Scholar 

  56. Gautam M et al. (2006) Flow-activated chloride channels in vascular endothelium. Shear stress sensitivity, desensitization dynamics, and physiological implications. J Biol Chem 281: 36492–36500

    Article  CAS  Google Scholar 

  57. Davies PF et al. (1994) Quantitative studies of endothelial cell adhesion: directional remodeling of focal adhesion sites in response to flow forces. J Clin Invest 93: 2031–2038

    Article  CAS  Google Scholar 

  58. Shyy JY and Chien S (2002) Role of integrins in endothelial mechanosensing of shear stress. Circ Res 91: 769–775

    Article  CAS  Google Scholar 

  59. Fujiwara K et al. (2001) Is PECAM-1 a mechanoresponsive molecule? Cell Struct Funct 26: 11–17

    Article  CAS  Google Scholar 

  60. Tzima E et al. (2005) A mechanosensory complex that mediates the endothelial cell response to fluid shear stress. Nature 437: 426–431

    Article  CAS  Google Scholar 

  61. Helmke BP et al. (2003) Spatial concentration of intracellular strain induced by hemodynamic shear stress. Biophys J 84: 2691–2699

    Article  CAS  Google Scholar 

  62. Mott RE and Helmke BP (2007) Mapping the dynamics of shear stress-induced structural changes in endothelial cells. Am J Physiol Cell Physiol. 293: C1616–C1626

    Article  CAS  Google Scholar 

  63. Maniotis AJ et al. (1997) Demonstration of mechanical connections between integrins, cytoskeletal filaments, and nucleoplasm that stabilize nuclear structure. Proc Natl Acad Sci USA 94: 849–854

    Article  CAS  Google Scholar 

  64. Dalby MJ et al. (2007) Nanomechanotransduction and interphase nuclear organization influence on genomic control. J Cell Biochem 102: 1234–1244

    Article  CAS  Google Scholar 

  65. Weinbaum S et al. (2007) The structure and function of the endothelial glycocalyx layer. Annu Rev Biomed Eng 9: 121–167

    Article  CAS  Google Scholar 

  66. Vink H and Duling BR (1996) Identification of distinct luminal domains for macromolecules, erythrocytes, and leukocytes within mammalian capillaries. Circ Res 79: 581–589

    Article  CAS  Google Scholar 

  67. Florian JA et al. (2003) Heparan sulfate proteoglycan is a mechanosensor on endothelial cells. Circ Res 93: e136–e142

    Article  CAS  Google Scholar 

  68. Yao Y et al. (2007) Glycocalyx modulates the motility and proliferative response of vascular endothelium to fluid shear stress. Am J Physiol Heart Circ Physiol 293: H1023–H1030

    Article  CAS  Google Scholar 

  69. Huang H et al. Three-dimensional sub-diffraction-limited single fluorophore imaging of proteins near the cell membrane: application to the endothelial glycocalyx. Proc Natl Acad Sci USA, in press

  70. Secomb TW et al. (2001) Effect of the endothelial surface layer on transmission of fluid shear stress to endothelial cells. Biorheology 38: 143–150

    CAS  PubMed  Google Scholar 

  71. Helmke BP and Davies PF (2002) The cytoskeleton under external fluid mechanical forces: hemodynamic forces acting on the endothelium. Ann Biomed Eng 30: 284–329

    Article  Google Scholar 

  72. Stamatas GN and McIntire LV (2001) Rapid flow-induced responses in endothelial cells. Biotechnol Prog 17: 383–402

    Article  CAS  Google Scholar 

  73. Haust MD (1987) Endothelial cilia in human aortic atherosclerotic lesions. Virchows Arch A Pathol Anat Histopathol 410: 317–326

    Article  CAS  Google Scholar 

  74. Van der Heiden K et al. (2008) Endothelial primary cilia in areas of disturbed flow are at the base of atherosclerosis. Atherosclerosis 196: 542–550

    Article  CAS  Google Scholar 

  75. Iomini C et al. (2004) Primary cilia of human endothelial cells disassemble under laminar shear stress. J Cell Biol 164: 811–817

    Article  CAS  Google Scholar 

  76. Malek AM and Izumo S (1996) Mechanism of endothelial cell shape change and cytoskeletal remodeling in response to fluid shear stress. J Cell Sci 109: 713–726

    CAS  PubMed  Google Scholar 

  77. Hu S and Wang N (2006) Control of stress propagation in the cytoplasm by prestress and loading frequency. Mol Cell Biomech 3: 49–60

    PubMed  Google Scholar 

  78. Desai G et al. (1999) Pressure-jump studies of the folding/unfolding of trp repressor. J Mol Biol 288: 461–475

    Article  CAS  Google Scholar 

  79. Ingber D (2007) Vascular control through tensegrity-based integration of mechanics and chemistry. In Endothelial biomedicine, 1786–1792 (Ed Aird WC) Cambridge: Cambridge University Press

    Chapter  Google Scholar 

  80. Computational imaging of pulsatile flow in a normal human carotid bifurcation [http://www.mie.utoronto.ca/labs/bsl/gallery/carotid.mpg] (accessed 20 October 2008)

  81. Liu Y et al. (2008) Shc coordinates signals from intercellular junctions and integrins to regulate flow-induced inflammation. J Cell Biol 182: 185–196

    Article  CAS  Google Scholar 

  82. Tzima E (2006) Role of small GTPases in endothelial cytoskeletal dynamics and the shear stress response. Circ Res 98: 176–185

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The author's research is supported by grants from the National Heart Lung and Blood Institute of the National Institutes of Health.

Author information

Authors and Affiliations

Authors

Ethics declarations

Competing interests

The author declares no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Davies, P. Hemodynamic shear stress and the endothelium in cardiovascular pathophysiology. Nat Rev Cardiol 6, 16–26 (2009). https://doi.org/10.1038/ncpcardio1397

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncpcardio1397

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