High wall shear stress and spatial gradients in vascular pathology: a review

Jennifer M Dolan, John Kolega, Hui Meng, Jennifer M Dolan, John Kolega, Hui Meng

Abstract

Cardiovascular pathologies such as intracranial aneurysms (IAs) and atherosclerosis preferentially localize to bifurcations and curvatures where hemodynamics are complex. While extensive knowledge about low wall shear stress (WSS) has been generated in the past, due to its strong relevance to atherogenesis, high WSS (typically >3 Pa) has emerged as a key regulator of vascular biology and pathology as well, receiving renewed interests. As reviewed here, chronic high WSS not only stimulates adaptive outward remodeling, but also contributes to saccular IA formation (at bifurcation apices or outer curves) and atherosclerotic plaque destabilization (in stenosed vessels). Recent advances in understanding IA pathogenesis have shed new light on the role of high WSS in pathological vascular remodeling. In complex geometries, high WSS can couple with significant spatial WSS gradient (WSSG). A combination of high WSS and positive WSSG has been shown to trigger aneurysm initiation. Since endothelial cells (ECs) are sensors of WSS, we have begun to elucidate EC responses to high WSS alone and in combination with WSSG. Understanding such responses will provide insight into not only aneurysm formation, but also plaque destabilization and other vascular pathologies and potentially lead to improved strategies for disease management and novel targets for pharmacological intervention.

Figures

FIGURE 1
FIGURE 1
Examples of arterial geometries with different WSS characteristics. (a) Carotid bifurcation with disturbed flow in the sinus opposite the apex, characterized by low WSS, and prone to atherogenesis. (b) Cerebral bifurcation with apex experiencing high WSS and WSSG resulting from flow impingement are prone to aneurysm formation. (c) Arterial stenosis, with both high WSS (upstream shoulder) and low WSS (downstream, disturbed flow).
FIGURE 2
FIGURE 2
Hemodynamic-histology co-mapping in the canine de novo bifurcation model reveals specific hemodynamic conditions leading to different remodeling responses. (a) Overlay of flow velocity vectors on the corresponding histological section. (b) Plot of WSS (blue) and WSSG (red) vs. distance from the apex revealing 3 regions with distinct flow patterns and remodeling responses from (a). Region 1: the impingement region with hyperplastic responses; Region II: acceleration region with aneurysmlike remodeling; and Region III: decelerating flow and recovery to baseline flow with no morphological changes. From Meng et al. Reproduced with permission.
FIGURE 3
FIGURE 3
Hemodynamic conditions for aneurysm initiation at the basilar terminus in rabbits that received bilateral CCA ligation. (a) Histology (Van Gieson staining) at 5 days showing early aneurysmal damage in the form of IEL loss in two segments (ends are marked by arrows) flanking the apex. (b) Hemodynamics-histology co-mapping to determine the local hemodynamic conditions of initial aneurysmal damage. (c) WSS (blue) and WSSG (red) distribution along the wall, whereby IEL damage (yellow bands) localized in the flow acceleration zone. (d) “Point cloud” showing microsegments of intact IEL (black open circles) and damaged IEL (red solid dots) plotted against their local WSS and WSSG values. The red dots are clustered to the top right of the coordinate system, indicating that aneurysm-initiating conditions are positive WSSG and high WSS. Reproduced from Metaxa et al. with permission.
FIGURE 4
FIGURE 4
A marble-in-a-bowl metaphor for the role of hemodynamic insult in relation to other risk factors in aneurysm initiation. Homeostasis is represented by the marble at the bottom of the bowl and hemodynamic insult (white arrow) is represented by a push on the marble. (a) A gentle push results in a return to homeostasis. (b) A strong enough push causes the marble to fall out of the bowl causing a departure from homeostasis into pathogenesis. (c) Other risk factors (family history, female gender, genetic diseases, cigarette smoking and hypertension) lower the rim of the bowl, such that a gentle push can result in pathogenesis (i.e., aneurysm formation). Modified from Meng et al.
FIGURE 5
FIGURE 5
Alignment of bovine aortic ECs subjected to high WSS alone (a) or in combination with WSSG (b). (a) In vitro flow chamber subjecting EC monolayers to baseline WSS and high WSS. ECs align and elongate with their long axis parallel to the flow with a time course dependent on the WSS and display a transient perpendicular alignment when WSS is high. (b) In vitro flow system with converging and diverging-segments to produce positive and negative WSSG across EC monolayers. Positive WSSG hinders cell alignment to flow while negative WSS promotes it, even under the same range of WSS. Modified from Dolan et al., Reproduced with permission.
FIGURE 6
FIGURE 6
A conceptual picture of the mechanical forces on ECs in accelerating and decelerating flow: WSS from the flowing blood, net stretching/compression due to WSSG, and cyclic stretching from pulsatile pressure.

References

    1. Abruzzo T, Kendler A, Apkarian R, Workman M, Khoury JC, Cloft HJ. Cerebral aneurysm formation in nitric oxide synthase-3 knockout mice. Curr. Neurovasc. Res. 2007;4:161–169.
    1. Alnaes MS, Isaksen J, Mardal KA, Romner B, Morgan MK, Ingebrigtsen T. Computation of hemodynamics in the circle of Willis. Stroke. 2007;38:2500–2505.
    1. Aoki T, Kataoka H, Ishibashi R, Nozaki K, Egashira K, Hashimoto N. Impact of monocyte chemoattractant protein-1 deficiency on cerebral aneurysm formation. Stroke. 2009;40:942–951.
    1. Aoki T, Kataoka H, Ishibashi R, Nozaki K, Hashimoto N. Cathepsin B, K, and S are expressed in cerebral aneurysms and promote the progression of cerebral aneurysms. Stroke. 2008;39:2603–2610.
    1. Aoki T, Kataoka H, Morimoto M, Nozaki K, Hashimoto N. Macrophage-derived matrix metalloproteinase-2 and-9 promote the progression of cerebral aneurysms in rats. Stroke. 2007;38:162–169.
    1. Aoki T, Kataoka H, Nishimura M, Ishibashi R, Morishita R, Miyamoto S. Ets-1 promotes the progression of cerebral aneurysm by inducing the expression of MCP-1 in vascular smooth muscle cells. Gene Ther. 2010;17:1117–1123.
    1. Aoki T, Kataoka H, Shimamura M, Nakagami H, Wakayama K, Moriwaki T, Ishibashi R, Nozaki K, Morishita R, Hashimoto N. NF-kappaB is a key mediator of cerebral aneurysm formation. Circulation. 2007;116:2830–2840.
    1. Aoki T, Nishimura M, Kataoka H, Ishibashi R, Nozaki K, Miyamoto S. Complementary inhibition of cerebral aneurysm formation by eNOS and nNOS. Lab. Invest. 2011;91:619–626.
    1. Aoki T, Nishimura M, Matsuoka T, Yamamoto K, Furuyashiki T, Kataoka H, Kitaoka S, Ishibashi R, Ishibazawa A, Miyamoto S, Morishita R, Ando J, Hashimoto N, Nozaki K, Narumiya S. PGE(2) -EP(2) signalling in endothelium is activated by haemodynamic stress and induces cerebral aneurysm through an amplifying loop via NF-kappaB. Br. J. Pharmacol. 2011;163:1237–1249.
    1. Bark DL, Jr, Para AN, Ku DN. Correlation of thrombosis growth rate to pathological wall shear rate during platelet accumulation. Biotechnol. Bioeng. 2012;109:2642–2650.
    1. Brown RD, Jr, Wiebers DO, Forbes GS. Unruptured intracranial aneurysms and arteriovenous malformations: frequency of intracranial hemorrhage and relationship of lesions. J Neurosurg. 1990;73:859–863.
    1. Bruno G, Todor R, Lewis I, Chyatte D. Vascular extracellular matrix remodeling in cerebral aneurysms. J Neurosurg. 1998;89:431–440.
    1. Burke A, Fitzgerald GA. Oxidative stress and smoking-induced vascular injury. Prog. Cardiovasc. Dis. 2003;46:79–90.
    1. Caro CG, Fitz-Gerald JM, Schroter RC. Atheroma and arterial wall shear. Observation, correlation and proposal of a shear dependent mass transfer mechanism for atherogenesis. Proc. R. Soc. Lond. B Biol. Sci. 1971;177:109–159.
    1. Castier Y, Brandes RP, Leseche G, Tedgui A, Lehoux S. p47phox-dependent NADPH oxidase regulates flow-induced vascular remodeling. Circ. Res. 2005;97:533–540.
    1. Chatzizisis YS, Jonas M, Coskun AU, Beigel R, Stone BV, Maynard C, Gerrity RG, Daley W, Rogers C, Edelman ER, Feldman CL, Stone PH. Prediction of the localization of high-risk coronary atherosclerotic plaques on the basis of low endothelial shear stress: an intravascular ultrasound and histopathology natural history study. Circulation. 2008;117:993–1002.
    1. Chien S. Mechanotransduction and endothelial cell homeostasis: the wisdom of the cell. Am. J. Physiol. Heart Circ. Physiol. 2007;292:H1209–H1224.
    1. Davies PF. Flow-mediated endothelial mechanotransduction. Physiol. Rev. 1995;75:519–560.
    1. DePaola N, Davies PF, Pritchard WF, Jr, Florez L, Harbeck N, Polacek DC. Spatial and temporal regulation of gap junction connexin43 in vascular endothelial cells exposed to controlled disturbed flows in vitro. Proc. Natl Acad. Sci. USA. 1999;96:3154–3159.
    1. Dolan JM, Meng H, Singh S, Paluch R, Kolega J. High fluid shear stress and spatial shear stress gradients affect endothelial proliferation, survival, and alignment. Ann. Biomed. Eng. 2011;39:1620–1631.
    1. Dolan JM, Sim FJ, Meng H, Kolega J. Endothelial cells express a unique transcriptional profile under very high wall shear stress known to induce expansive arterial remodeling. Am. J. Physiol. Cell Physiol. 2012;302:C1109–C1118.
    1. Dumont O, Loufrani L, Henrion D. Key role of the NO-pathway and matrix metalloprotease-9 in high blood flow-induced remodeling of rat resistance arteries. Arterioscler. Thromb. Vasc. Biol. 2007;27:317–324.
    1. Eldawoody H, Shimizu H, Kimura N, Saito A, Nakayama T, Takahashi A, Tominaga T. Simplified experimental cerebral aneurysm model in rats: comprehensive evaluation of induced aneurysms and arterial changes in the circle of Willis. Brain Res. 2009;1300:159–168.
    1. Frosen J, Tulamo R, Paetau A, Laaksamo E, Korja M, Laakso A, Niemela M, Hernesniemi J. Saccular intracranial aneurysm: pathology and mechanisms. Acta Neuropathol. 2012;123:773–786.
    1. Fujii K, Kobayashi Y, Mintz GS, Takebayashi H, Dangas G, Moussa I, Mehran R, Lansky AJ, Kreps E, Collins M, Colombo A, Stone GW, Leon MB, Moses JW. Intravascular ultrasound assessment of ulcerated ruptured plaques: a comparison of culprit and nonculprit lesions of patients with acute coronary syndromes and lesions in patients without acute coronary syndromes. Circulation. 2003;108:2473–2478.
    1. Fukuda S, Hashimoto N, Naritomi H, Nagata I, Nozaki K, Kondo S, Kurino M, Kikuchi H. Prevention of rat cerebral aneurysm formation by inhibition of nitric oxide synthase. Circulation. 2000;101:2532–2538.
    1. Gao L, Hoi Y, Swartz DD, Kolega J, Siddiqui A, Meng H. Nascent aneurysm formation at the basilar terminus induced by hemodynamics. Stroke. 2008;39:2085–2090.
    1. Gertz SD, Roberts WC. Hemodynamic shear force in rupture of coronary arterial atherosclerotic plaques. Am. J. Cardiol. 1990;66:1368–1372.
    1. Gibbons GH, Dzau VJ. The emerging concept of vascular remodeling. N. Engl. J. Med. 1994;330:1431–1438.
    1. Gijsen FJ, Mastik F, Schaar JA, Schuurbiers JC, van der Giessen WJ, de Feyter PJ, Serruys PW, van der Steen AF, Wentzel JJ. High shear stress induces a strain increase in human coronary plaques over a 6- month period. EuroIntervention. 2011;7:121–127.
    1. Gijsen FJ, Wentzel JJ, Thury A, Mastik F, Schaar JA, Schuurbiers JC, Slager CJ, van der Giessen WJ, de Feyter PJ, van der Steen AF, Serruys PW. Strain distribution over plaques in human coronary arteries relates to shear stress. Am. J. Physiol. Heart Circ. Physiol. 2008;295:H1608–H1614.
    1. Glagov S, Weisenberg E, Zarins CK, Stankunavicius R, Kolettis GJ. Compensatory enlargement of human atherosclerotic coronary arteries. N. Engl. J. Med. 1987;316:1371–1375.
    1. Glagov S, Zarins C, Giddens DP, Ku DN. Hemodynamics and atherosclerosis. Insights and perspectives gained from studies of human arteries. Arch. Pathol. Lab. Med. 1988;112:1018–1031.
    1. Greve JM, Les AS, Tang BT, Draney Blomme MT, Wilson NM, Dalman RL, Pelc NJ, Taylor CA. Allometric scaling of wall shear stress from mice to humans: quantification using cine phase-contrast MRI and computational fluid dynamics. Am. J. Physiol. Heart Circ. Physiol. 2006;291:H1700–H1708.
    1. Groen HC, Gijsen FJ, van der Lugt A, Ferguson MS, Hatsukami TS, van der Steen AF, Yuan C, Wentzel JJ. Plaque rupture in the carotid artery is localized at the high shear stress region: a case report. Stroke. 2007;38:2379–2381.
    1. Guzman RJ, Abe K, Zarins CK. Flow-induced arterial enlargement is inhibited by suppression of nitric oxide synthase activity in vivo. Surgery. 1997;122:273–279. discussion 279–280.
    1. Han HC. Twisted blood vessels: symptoms, etiology and biomechanical mechanisms. J Vasc. Res. 2012;49:185–197.
    1. Hashimoto N, Handa H, Hazama F. Experimentally induced cerebral aneurysms in rats. Surg. Neurol. 1978;10:3–8.
    1. Hashimoto N, Handa H, Nagata I, Hazama F. Experimentally induced cerebral aneurysms in rats: Part V. Relation of hemodynamics in the circle of Willis to formation of aneurysms. Surg. Neurol. 1980;13:41–45.
    1. Hashimoto T, Meng H, Young WL. Intracranial aneurysms: links among inflammation, hemodynamics and vascular remodeling. Neurol. Res. 2006;28:372–380.
    1. Hazama F, Kataoka H, Yamada E, Kayembe K, Hashimoto N, Kojima M, Kim C. Early changes of experimentally induced cerebral aneurysms in rats. Lightmicroscopic study. Am. J. Pathol. 1986;124:399–404.
    1. Hoi Y, Gao L, Tremmel M, Paluch RA, Siddiqui AH, Meng H, Mocco J. In vivo assessment of rapid cerebrovascular morphological adaptation following acute blood flow increase. J Neurosurg. 2008;109:1141–1147.
    1. Hsieh HJ, Li NQ, Frangos JA. Shear-induced platelet-derived growth factor gene expression in human endothelial cells is mediated by protein kinase C. J Cell. Physiol. 1992;150:552–558.
    1. Hyun S, Kleinstreuer C, Archie JP., Jr Hemodynamics analyses of arterial expansions with implications to thrombosis and restenosis. Med. Eng. Phys. 2000;22:13–27.
    1. Ishibashi A, Yokokura Y, Kojima K, Abe T. Acute obstructive hydrocephalus due to an unruptured basilar bifurcation aneurysm associated with bilateral internal carotid occlusion—a case report. Kurume Med. J. 1993;40:21–25.
    1. Jamous MA, Nagahiro S, Kitazato KT, Satomi J, Satoh K. Role of estrogen deficiency in the formation and progression of cerebral aneurysms. Part I: experimental study of the effect of oophorectomy in rats. J Neurosurg. 2005;103:1046–1051.
    1. Jamous MA, Nagahiro S, Kitazato KT, Tamura T, Aziz HA, Shono M, Satoh K. Endothelial injury and inflammatory response induced by hemodynamic changes preceding intracranial aneurysm formation: experimental study in rats. J Neurosurg. 2007;107:405–411.
    1. Jesty J, Yin W, Perrotta P, Bluestein D. Platelet activation in a circulating flow loop: combined effects of shear stress and exposure time. Platelets. 2003;14:143–149.
    1. Jou LD, van Tyen R, Berger SA, Saloner D. Calculation of the magnetization distribution for fluid flow in curved vessels. Magn. Reson. Med. 1996;35:577–584.
    1. Juvela S. Natural history of unruptured intracranial aneurysms: risks for aneurysm formation, growth, and rupture. Acta Neurochir. Suppl. 2002;82:27–30.
    1. Kamiya A, Bukhari R, Togawa T. Adaptive regulation of wall shear stress optimizing vascular tree function. Bull. Math. Biol. 1984;46:127–137.
    1. Kamiya A, Togawa T. Adaptive regulation of wall shear stress to flow change in the canine carotid artery. Am. J. Physiol. 1980;239:H14–H21.
    1. Karwowski JK, Markezich A, Whitson J, Abbruzzese TA, Zarins CK, Dalman RL. Dosedependent limitation of arterial enlargement by the matrix metalloproteinase inhibitor RS-113,456. J Surg. Res. 1999;87:122–129.
    1. Kataoka K, Taneda M, Asai T, Kinoshita A, Ito M, Kuroda R. Structural fragility and inflammatory response of ruptured cerebral aneurysms. A comparative study between ruptured and unruptured cerebral aneurysms. Stroke. 1999;30:1396–1401.
    1. Kojima M, Handa H, Hashimoto N, Kim C, Hazama F. Early changes of experimentally induced cerebral aneurysms in rats: scanning electron microscopic study. Stroke. 1986;17:835–841.
    1. Kolega J, Gao L, Mandelbaum M, Mocco J, Siddiqui AH, Natarajan SK, Meng H. Cellular and molecular responses of the basilar terminus to hemodynamics during intracranial aneurysm initiation in a rabbit model. J Vasc. Res. 2011;48:429–442.
    1. Koskinas KC, Feldman CL, Chatzizisis YS, Coskun AU, Jonas M, Maynard C, Baker AB, Papafaklis MI, Edelman ER, Stone PH. Natural history of experimental coronary atherosclerosis and vascular remodeling in relation to endothelial shear stress: a serial, in vivo intravascular ultrasound study. Circulation. 2010;121:2092–2101.
    1. Kulcsar Z, Ugron A, Marosfoi M, Berentei Z, Paal G, Szikora I. Hemodynamics of cerebral aneurysm initiation: the role of wall shear stress and spatial wall shear stress gradient. AJNR Am. J. Neuroradiol. 2011;32:587–594.
    1. Kwak BR, Silacci P, Stergiopulos N, Hayoz D, Meda P. Shear stress and cyclic circumferential stretch, but not pressure, alter connexin43 expression in endothelial cells. Cell Commun. Adhes. 2005;12:261–270.
    1. LaBarbera M. Principles of design of fluid transport systems in zoology. Science. 1990;249:992–1000.
    1. LaMack JA, Friedman MH. Individual and combined effects of shear stress magnitude and spatial gradient on endothelial cell gene expression. Am. J. Physiol. Heart Circ. Physiol. 2007;293:H2853–H2859.
    1. Langille BL, O’Donnell F. Reductions in arterial diameter produced by chronic decreases in blood flow are endothelium-dependent. Science. 1986;231:405–407.
    1. Leach JR, Rayz VL, Soares B, Wintermark M, Mofrad MR, Saloner D. Carotid atheroma rupture observed in vivo and FSI-predicted stress distribution based on pre-rupture imaging. Ann. Biomed. Eng. 2010;38:2748–2765.
    1. Lehman RM, Owens GK, Kassell NF, Hongo K. Mechanism of enlargement of major cerebral collateral arteries in rabbits. Stroke. 1991;22:499–504.
    1. Lehoux S, Castier Y, Tedgui A. Molecular mechanisms of the vascular responses to haemodynamic forces. J Intern. Med. 2006;259:381–392.
    1. Lehoux S, Tronc F, Tedgui A. Mechanisms of blood flow-induced vascular enlargement. Biorheology. 2002;39:319–324.
    1. Levkau B, Kenagy RD, Karsan A, Weitkamp B, Clowes AW, Ross R, Raines EW. Activation of metalloproteinases and their association with integrins: an auxiliary apoptotic pathway in human endothelial cells. Cell Death Differ. 2002;9:1360–1367.
    1. Li ZY, Taviani V, Tang T, Sadat U, Young V, Patterson A, Graves M, Gillard JH. The mechanical triggers of plaque rupture: shear stress vs pressure gradient. Br. J. Radiol. 2009;82(Spec No. 1):S39–S45.
    1. Lindekleiv HM, Valen-Sendstad K, Morgan MK, Mardal KA, Faulder K, Magnus JH, Waterloo K, Romner B, Ingebrigtsen T. Sex differences in intracranial arterial bifurcations. Gend. Med. 2010;7:149–155.
    1. Lovett JK, Rothwell PM. Site of carotid plaque ulceration in relation to direction of blood flow: an angiographic and pathological study. Cerebrovasc. Dis. 2003;16:369–375.
    1. Malek AM, Alper SL, Izumo S. Hemodynamic shear stress and its role in atherosclerosis. JAMA. 1999;282:2035–2042.
    1. Malek AM, Gibbons GH, Dzau VJ, Izumo S. Fluid shear stress differentially modulates expression of genes encoding basic fibroblast growth factor and platelet- derived growth factor B chain in vascular endothelium. J Clin. Invest. 1993;92:2013–2021.
    1. Masuda H, Zhuang YJ, Singh TM, Kawamura K, Murakami M, Zarins CK, Glagov S. Adaptive remodeling of internal elastic lamina and endothelial lining during flow-induced arterial enlargement. Arterioscler. Thromb. Vasc. Biol. 1999;19:2298–2307.
    1. Meng H, Metaxa E, Gao L, Liaw N, Natarajan SK, Swartz DD, Siddiqui AH, Kolega J, Mocco J. Progressive aneurysm development following hemodynamic insult. J Neurosurg. 2011;114:1095–1103.
    1. Meng H, Swartz DD, Wang Z, Hoi Y, Kolega J, Metaxa EM, Szymanski MP, Yamamoto J, Sauvageau E, Levy EI. A model system for mapping vascular responses to complex hemodynamics at arterial bifurcations in vivo. Neurosurgery. 2006;59:1094–1100. discussion 1100–1101.
    1. Meng H, Wang Z, Hoi Y, Gao L, Metaxa E, Swartz DD, Kolega J. Complex hemodynamics at the apex of an arterial bifurcation induces vascular remodeling resembling cerebral aneurysm initiation. Stroke. 2007;38:1924–1931.
    1. Meng H, Xiang J, Liaw N. The role of hemodynamics in intracranial aneurysm initiation. Int. Rev. Thrombosis. 2012;7:40–57.
    1. Metaxa E, Meng H, Kaluvala SR, Szymanski MP, Paluch RA, Kolega J. Nitric oxide-dependent stimulation of endothelial cell proliferation by sustained high flow. Am. J. Physiol. Heart Circ. Physiol. 2008;295:H736–H742.
    1. Metaxa E, Tremmel M, Natarajan SK, Xiang J, Paluch RA, Mandelbaum M, Siddiqui AH, Kolega J, Mocco J, Meng H. Characterization of critical hemodynamics contributing to aneurysmal remodeling at the basilar terminus in a rabbit model. Stroke. 2010;41:1774–1782.
    1. Mintz GS, Kent KM, Pichard AD, Satler LF, Popma JJ, Leon MB. Contribution of inadequate arterial remodeling to the development of focal coronary artery stenoses. An intravascular ultrasound study. Circulation. 1997;95:1791–1798.
    1. Moore JE, Jr, Burki E, Suciu A, Zhao S, Burnier M, Brunner HR, Meister JJ. A device for subjecting vascular endothelial cells to both fluid shear stress and circumferential cyclic stretch. Ann. Biomed. Eng. 1994;22:416–422.
    1. Moriwaki T, Takagi Y, Sadamasa N, Aoki T, Nozaki K, Hashimoto N. Impaired progression of cerebral aneurysms in interleukin-1beta-deficient mice. Stroke. 2006;37:900–905.
    1. Murray CD. The physiological principle of minimum work: I. The vascular system and the cost of blood volume. Proc. Natl Acad. Sci. USA. 1926;12:207–214.
    1. Nagata I, Handa H, Hashimoto N, Hazama F. Experimentally induced cerebral aneurysms in rats: Part VI. Hypertension. Surg. Neurol. 1980;14:477–479.
    1. Nakatani H, Hashimoto N, Kang Y, Yamazoe N, Kikuchi H, Yamaguchi S, Niimi H. Cerebral blood flow patterns at major vessel bifurcations and aneurysms in rats. J Neurosurg. 1991;74:258–262.
    1. Neelamegham S, Taylor AD, Burns AR, Smith CW, Simon SI. Hydrodynamic shear shows distinct roles for LFA-1 and Mac-1 in neutrophil adhesion to intercellular adhesion molecule-1. Blood. 1998;92:1626–1638.
    1. Nuki Y, Matsumoto MM, Tsang E, Young WL, van Rooijen N, Kurihara C, Hashimoto T. Roles of macrophages in flow-induced outward vascular remodeling. J Cereb. Blood Flow Metab. 2009;29:495–503.
    1. Park JB, Charbonneau F, Schiffrin EL. Correlation of endothelial function in large and small arteries in human essential hypertension. J Hypertens. 2001;19:415–420.
    1. Rajagopalan S, Meng XP, Ramasamy S, Harrison DG, Galis ZS. Reactive oxygen species produced by macrophage-derived foam cells regulate the activity of vascular matrix metalloproteinases in vitro. Implications for atherosclerotic plaque stability. J Clin. Invest. 1996;98:2572–2579.
    1. Rinkel GJ, Djibuti M, Algra A, van Gijn J. Prevalence and risk of rupture of intracranial aneurysms: a systematic review. Stroke. 1998;29:251–256.
    1. Ross R. Atherosclerosis—an inflammatory disease. N. Engl. J. Med. 1999;340:115–126.
    1. Sakamoto N, Saito N, Han X, Ohashi T, Sato M. Effect of spatial gradient in fluid shear stress on morphological changes in endothelial cells in response to flow. Biochem. Biophys. Res. Commun. 2010;395:264–269.
    1. Samady H, Eshtehardi P, McDaniel MC, Suo J, Dhawan SS, Maynard C, Timmins LH, Quyyumi AA, Giddens DP. Coronary artery wall shear stress is associated with progression and transformation of atherosclerotic plaque and arterial remodeling in patients with coronary artery disease. Circulation. 2011;124:779–788.
    1. Schiffrin EL, Park JB, Intengan HD, Touyz RM. Correction of arterial structure and endothelial dysfunction in human essential hypertension by the angiotensin receptor antagonist losartan. Circulation. 2000;101:1653–1659.
    1. Schirmer CM, Malek AM. Computational fluid dynamic characterization of carotid bifurcation stenosis in patient-based geometries. Brain Behav. 2012;2:42–52.
    1. Schirmer CM, Malek AM. Wall shear stress gradient analysis within an idealized stenosis using non- Newtonian flow. Neurosurgery. 2007;61:853–863. discussion 863–864.
    1. Schoenhagen P, Ziada KM, Kapadia SR, Crowe TD, Nissen SE, Tuzcu EM. Extent and direction of arterial remodeling in stable versus unstable coronary syndromes : an intravascular ultrasound study. Circulation. 2000;101:598–603.
    1. Sho E, Komatsu M, Sho M, Nanjo H, Singh TM, Xu C, Masuda H, Zarins CK. High flow drives vascular endothelial cell proliferation during flow-induced arterial remodeling associated with the expression of vascular endothelial growth factor. Exp. Mol. Pathol. 2003;75:1–11.
    1. Sho E, Sho M, Singh TM, Nanjo H, Komatsu M, Xu C, Masuda H, Zarins CK. Arterial enlargement in response to high flow requires early expression of matrix metalloproteinases to degrade extracellular matrix. Exp. Mol. Pathol. 2002;73:142–153.
    1. Shumacker HB., Jr Aneurysm development and degenerative changes in dilated artery proximal to arteriovenous fistula. Surg. Gynecol. Obstet. 1970;130:636–640.
    1. Singh TM, Abe KY, Sasaki T, Zhuang YJ, Masuda H, Zarins CK. Basic fibroblast growth factor expression precedes flow-induced arterial enlargement. J Surg. Res. 1998;77:165–173.
    1. Slager CJ, Wentzel JJ, Gijsen FJ, Schuurbiers JC, van der Wal AC, van der Steen AF, Serruys PW. The role of shear stress in the generation of rupture-prone vulnerable plaques. Nat. Clin. Pract. Cardiovasc. Med. 2005;2:401–407.
    1. Stehbens WE. Aneurysms and anatomical variation of cerebral arteries. Arch. Pathol. 1963;75:45–64.
    1. Stone PH, Coskun AU, Kinlay S, Clark ME, Sonka M, Wahle A, Ilegbusi OJ, Yeghiazarians Y, Popma JJ, Orav J, Kuntz RE, Feldman CL. Effect of endothelial shear stress on the progression of coronary artery disease, vascular remodeling, and in-stent restenosis in humans: in vivo 6-month follow-up study. Circulation. 2003;108:438–444.
    1. Szymanski MP, Metaxa E, Meng H, Kolega J. Endothelial cell layer subjected to impinging flow mimicking the apex of an arterial bifurcation. Ann. Biomed. Eng. 2008;36:1681–1689.
    1. Tada Y, Kitazato KT, Yagi K, Shimada K, Matsushita N, Kinouchi T, Kanematsu Y, Satomi J, Kageji T, Nagahiro S. Statins promote the growth of experimentally induced cerebral aneurysms in estrogendeficient rats. Stroke. 2011;42:2286–2293.
    1. Tada Y, Yagi K, Kitazato KT, Tamura T, Kinouchi T, Shimada K, Matsushita N, Nakajima N, Satomi J, Kageji T, Nagahiro S. Reduction of endothelial tight junction proteins is related to cerebral aneurysm formation in rats. J Hypertens. 2010;28:1883–1891.
    1. Tanweer O, Metaxa E, Liaw N, Sternberg S, Siddiqui A, Kolega J, Meng H. Inhibition of stretch-activated ion channels on endothelial cells disrupts nitric oxide-mediated arterial outward remodeling. J Biorheol. 2011;24:77–83.
    1. Teng Z, Canton G, Yuan C, Ferguson M, Yang C, Huang X, Zheng J, Woodard PK, Tang D. 3D critical plaque wall stress is a better predictor of carotid plaque rupture sites than flow shear stress: an in vivo MRIbased 3D FSI study. J Biomech. Eng. 2010;132:031007.
    1. Thi MM, Tarbell JM, Weinbaum S, Spray DC. The role of the glycocalyx in reorganization of the actin cytoskeleton under fluid shear stress: a “bumper-car” model. Proc. Natl Acad. Sci. USA. 2004;101:16483–16488.
    1. Tohda K, Masuda H, Kawamura K, Shozawa T. Difference in dilatation between endothelium-preserved and -desquamated segments in the flow-loaded rat common carotid artery. Arterioscler. Thromb. 1992;12:519–528.
    1. Torii R, Wood NB, Hadjiloizou N, Dowsey AW, Wright AR, Hughes AD, Davies J, Francis DP, Mayet J, Yang GZ, Thom SA, Xu XY. Stress phase angle depicts differences in coronary artery hemodynamics due to changes in flow and geometry after percutaneous coronary intervention. Am. J. Physiol. Heart Circ. Physiol. 2009;296:H765–H776.
    1. Tremmel M, Xiang J, Natarajan SK, Hopkins LN, Siddiqui AH, Levy EI, Meng H. Alteration of intra-aneurysmal hemodynamics for flow diversion using enterprise and vision stents. World Neurosurg. 2010;74:306–315.
    1. Tronc F, Mallat Z, Lehoux S, Wassef M, Esposito B, Tedgui A. Role of matrix metalloproteinases in blood flow-induced arterial enlargement: interaction with NO. Arterioscler. Thromb. Vasc. Biol. 2000;20:E120–E126.
    1. Tronc F, Wassef M, Esposito B, Henrion D, Glagov S, Tedgui A. Role of NO in flow-induced remodeling of the rabbit common carotid artery. Arterioscler. Thromb. Vasc. Biol. 1996;16:1256–1262.
    1. Tuttle JL, Nachreiner RD, Bhuller AS, Condict KW, Connors BA, Herring BP, Dalsing MC, Unthank JL. Shear level influences resistance artery remodeling: wall dimensions, cell density, and eNOS expression. Am. J. Physiol. Heart Circ. Physiol. 2001;281:H1380–H1389.
    1. Tzima E, Irani-Tehrani M, Kiosses WB, Dejana E, Schultz DA, Engelhardt B, Cao G, DeLisser H, Schwartz MA. A mechanosensory complex that mediates the endothelial cell response to fluid shear stress. Nature. 2005;437:426–431.
    1. van Everdingen KJ, Klijn CJ, Kappelle LJ, Mali WP, van der Grond J. MRA flow quantification in patients with a symptomatic internal carotid artery occlusion. The Dutch EC-IC Bypass Study Group. Stroke. 1997;28:1595–1600.
    1. Van Remmen H, Williams MD, Guo Z, Estlack L, Yang H, Carlson EJ, Epstein CJ, Huang TT, Richardson A. Knockout mice heterozygous for Sod2 show alterations in cardiac mitochondrial function and apoptosis. Am. J. Physiol. Heart Circ. Physiol. 2001;281:H1422–H1432.
    1. Varnava AM, Mills PG, Davies MJ. Relationship between coronary artery remodeling and plaque vulnerability. Circulation. 2002;105:939–943.
    1. Vega C, Kwoon JV, Lavine SD. Intracranial aneurysms: current evidence and clinical practice. Am. Fam. Physician. 2002;66:601–608.
    1. Waga S, Okada M, Kojima T. Saccular aneurysm associated with absence of the left cervical carotid arteries. Neurosurgery. 1978;3:208–212.
    1. Wang Z, Kolega J, Hoi Y, Gao L, Swartz DD, Levy EI, Mocco J, Meng H. Molecular alterations associated with aneurysmal remodeling are localized in the high hemodynamic stress region of a created carotid bifurcation. Neurosurgery. 2009;65:169–177. discussion 177–178.
    1. Weinberg PD, Ross Ethier C. Twenty-fold difference in hemodynamic wall shear stress between murine and human aortas. J Biomech. 2007;40:1594–1598.
    1. Wentzel JJ, Chatzizisis YS, Gijsen FJ, Giannoglou GD, Feldman CL, Stone PH. Endothelial shear stress in the evolution of coronary atherosclerotic plaque and vascular remodeling: current understanding and remaining questions. Cardiovasc. Res. 2012;96(2):234–243.
    1. White SJ, Hayes EM, Lehoux S, Jeremy JY, Horrevoets AJ, Newby AC. Characterization of the differential response of endothelial cells exposed to normal and elevated laminar shear stress. J Cell. Physiol. 2011;226:2841–2848.
    1. Yong-Zhong G, van Alphen HA. Pathogenesis and histopathology of saccular aneurysms: review of the literature. Neurol. Res. 1990;12:249–255.
    1. Zarins CK, Giddens DP, Bharadvaj BK, Sottiurai VS, Mabon RF, Glagov S. Carotid bifurcation atherosclerosis. Quantitative correlation of plaque localization with flow velocity profiles and wall shear stress. Circ. Res. 1983;53:502–514.
    1. Zarins CK, Zatina MA, Giddens DP, Ku DN, Glagov S. Shear stress regulation of artery lumen diameter in experimental atherogenesis. J Vasc. Surg. 1987;5:413–420.
    1. Zhao S, Suciu A, Ziegler T, Moore JE, Jr, Burki E, Meister JJ, Brunner HR. Synergistic effects of fluid shear stress and cyclic circumferential stretch on vascular endothelial cell morphology and cytoskeleton. Arterioscler. Thromb. Vasc. Biol. 1995;15:1781–1786.

Source: PubMed

3
Předplatit