Capillary dysfunction: its detection and causative role in dementias and stroke

Leif Østergaard, Sune Nørhøj Jespersen, Thorbjørn Engedahl, Eugenio Gutiérrez Jiménez, Mahmoud Ashkanian, Mikkel Bo Hansen, Simon Eskildsen, Kim Mouridsen, Leif Østergaard, Sune Nørhøj Jespersen, Thorbjørn Engedahl, Eugenio Gutiérrez Jiménez, Mahmoud Ashkanian, Mikkel Bo Hansen, Simon Eskildsen, Kim Mouridsen

Abstract

In acute ischemic stroke, critical hypoperfusion is a frequent cause of hypoxic tissue injury: As cerebral blood flow (CBF) falls below the ischemic threshold of 20 mL/100 mL/min, neurological symptoms develop and hypoxic tissue injury evolves within minutes or hours unless the oxygen supply is restored. But is ischemia the only hemodynamic source of hypoxic tissue injury? Reanalyses of the equations we traditionally use to describe the relation between CBF and tissue oxygenation suggest that capillary flow patterns are crucial for the efficient extraction of oxygen: without close capillary flow control, "functional shunts" tend to form and some of the blood's oxygen content in effect becomes inaccessible to tissue. This phenomenon raises several questions: Are there in fact two hemodynamic causes of tissue hypoxia: Limited blood supply (ischemia) and limited oxygen extraction due to capillary dysfunction? If so, how do we distinguish the two, experimentally and in patients? Do flow-metabolism coupling mechanisms adjust CBF to optimize tissue oxygenation when capillary dysfunction impairs oxygen extraction downstream? Cardiovascular risk factors such as age, hypertension, diabetes, hypercholesterolemia, and smoking increase the risk of both stroke and dementia. The capillary dysfunction phenomenon therefore forces us to consider whether changes in capillary morphology or blood rheology may play a role in the etiology of some stroke subtypes and in Alzheimer's disease. Here, we discuss whether certain disease characteristics suggest capillary dysfunction rather than primary flow-limiting vascular pathology and how capillary dysfunction may be imaged and managed.

Figures

Fig. 1
Fig. 1
Panel a illustrates how MTT and CTH can be determined by two-photon microscopy, tracking the passage of a fluorescein isothiocyanate (FITC) bolus as it passes through the microvasculature of a mouse brain. The top panels show images obtained through a cranial window 8 and 20 s after injection. Arteries and veins can be identified based on the arrival of the fluorescent dye and their concentration-time curves (CTC) measured over time (lower left panel). The parameters of the transit time distribution (lower right) are then fitted so that the venous outflow curve is accurately predicted (lower left panel). The delay between arterioles and venules defines MTT, and CTH is defined as the standard deviation of the transit time distribution (lower right panel). Previously unpublished data. Panel b shows the probability that CoV correlated with mini-mental state examination (MMSE) scores in 16 patients with clinically suspected possible or probable Alzheimer’s disease (AD). Note the significant, negative correlations between MMSE and cortical CoV. AD was verified by ICD-10, DSM-IV, and NINCDS-ADRDA. The age of the patients was 70.4 ± 6.3 years and their MMSE 24.8 ± 2.7. Note the strong temporoparietal, cingulate, and precuneus involvement. In AD, these regions typically reveal abnormally low fluorodeoxyglucose uptake and cortical thinning. Data were acquired after informed consent in a project approved by the regional Ethics Committee. Previously unpublished data. Panel c shows maps of acute MTT, CTH, CoV, OEFmax, and apparent diffusion coefficient (ADC) in a 74-year-old male with a distal occlusion of the middle cerebral artery, imaged 4 h and 14 min after symptom onset. His NIHSS score was 14. Twenty days later, follow-up (FoUp) FLAIR images were acquired to assess the extent of tissue damage. The extent of hypoperfusion is visualized as areas of prolonged MTT (green, yellow, and orange colors indicate higher values), while disturbed capillary flow patterns can be identified as elevated CTH values. Note that white matter hyperintensities, indicative of cerebral small-vessel disease (SVD) are present in this patient. These can be recognized as confluent hyperintensities in the ADC image and bright lesions in FLAIR images. Reproduced from [••] with permission from the publisher. Panel d shows PET and MR data from two patients with occlusion of their right carotid artery. Both had experienced short episodes of left-sided hemiparesis and right-sided amaurosis fugax (blindness), but experienced no neurological deficits at the time of the study. Occlusion of their carotid arteries was diagnosed by ultrasonic examination, and competing cerebral pathologies were excluded by earlier MRI. The PET protocol, experimental procedures, and PET data have previously been published in [86]. The PWI data presented here were obtained after informed consent as part of the original study protocol which was approved by the local ethics committee. The PET images were manually registered (MNI Register, McConnell Brain Imaging Centre of the Montreal Neurological Institute, McGill University) to patient MRIs using an affine transformation. Then, PET OEF maps were generated for 11 slices, corresponding to the location of 11 midbrain PWI slices in which prolonged MTT could easily be observed. The three panels on the left compares MTT and OEFmax images, obtained by MRI, to OEF maps obtained by PET, at two slice locations in one of the patients. In the two plots to the right, the ability of MTT and OEFmax to predict “true” OEF is compared for the hemispheres ipsi- and contralateral to the stenosis (red and green dots) in 11 slices each of the two patients. The higher OEF in the affected side has traditionally been associated with severe hypoperfusion caused by carotid stenosis. See text. Previously unpublished data

References

    1. Renkin EM. B. W. Zweifach Award lecture. Regulation of the microcirculation. Microvasc Res. 1985;30:251–63. doi: 10.1016/0026-2862(85)90057-3.
    1. Vorstrup S, Henriksen L, Paulson OB. Effect of acetazolamide on cerebral blood flow and cerebral metabolic rate for oxygen. J Clin Invest. 1984;74:1634–9. doi: 10.1172/JCI111579.
    1. Ruitenberg A, den Heijer T, Bakker SL, et al. Cerebral hypoperfusion and clinical onset of dementia: the Rotterdam study. Ann Neurol. 2005;57:789–94. doi: 10.1002/ana.20493.
    1. Hirao K, Ohnishi T, Hirata Y, et al. The prediction of rapid conversion to Alzheimer’s disease in mild cognitive impairment using regional cerebral blood flow SPECT. Neuroimage. 2005;28:1014–21. doi: 10.1016/j.neuroimage.2005.06.066.
    1. Hardy J, Selkoe DJ. The amyloid hypothesis of Alzheimer’s disease: progress and problems on the road to therapeutics. Science. 2002;297:353–6. doi: 10.1126/science.1072994.
    1. Zlokovic BV. Neurovascular pathways to neurodegeneration in Alzheimer’s disease and other disorders. Nat Rev Neurosci. 2011;12:723–38.
    1. Iadecola C. The overlap between neurodegenerative and vascular factors in the pathogenesis of dementia. Acta Neuropathol. 2010;120:287–96. doi: 10.1007/s00401-010-0718-6.
    1. Østergaard L, Kristiansen SB, Angleys H, et al. The role of capillary transit time heterogeneity in myocardial oxygenation and ischemic heart disease. Basic Res Cardiol. 2014;109:409. doi: 10.1007/s00395-014-0409-x.
    1. Jespersen SN, Østergaard L. The roles of cerebral blood flow, capillary transit time heterogeneity and oxygen tension in brain oxygenation and metabolism. J Cereb Blood Flow Metab. 2012;32:264–77. doi: 10.1038/jcbfm.2011.153.
    1. Østergaard L, Sorensen AG, Chesler DA, et al. Combined diffusion-weighted and perfusion-weighted flow heterogeneity magnetic resonance imaging in acute stroke. Stroke. 2000;31:1097–103. doi: 10.1161/01.STR.31.5.1097.
    1. Kleinfeld D, Mitra PP, Helmchen F, Denk W. Fluctuations and stimulus-induced changes in blood flow observed in individual capillaries in layers 2 through 4 of rat neocortex. Proc Natl Acad Sci U S A. 1998;95:15741–6. doi: 10.1073/pnas.95.26.15741.
    1. Villringer A, Them A, Lindauer U, Einhaupl K, Dirnagl U. Capillary perfusion of the rat brain cortex. An in vivo confocal microscopy study. Circ Res. 1994;75:55–62. doi: 10.1161/01.RES.75.1.55.
    1. Stefanovic B, Hutchinson E, Yakovleva V, et al. Functional reactivity of cerebral capillaries. J Cereb Blood Flow Metab. 2008;28:961–72. doi: 10.1038/sj.jcbfm.9600590.
    1. Paulson OB, Hasselbalch SG, Rostrup E, Knudsen GM, Pelligrino D. Cerebral blood flow response to functional activation. J Cereb Blood Flow Metab. 2010;30:2–14. doi: 10.1038/jcbfm.2009.188.
    1. Schulte ML, Wood JD, Hudetz AG. Cortical electrical stimulation alters erythrocyte perfusion pattern in the cerebral capillary network of the rat. Brain Res. 2003;963:81–92. doi: 10.1016/S0006-8993(02)03848-9.
    1. Vogel J, Kuschinsky W. Decreased heterogeneity of capillary plasma flow in the rat whisker-barrel cortex during functional hyperemia. J Cereb Blood Flow Metab. 1996;16:1300–6. doi: 10.1097/00004647-199611000-00026.
    1. Rasmussen PM, Jespersen SN, Østergaard L. The effects of transit time heterogeneity on brain oxygenation during rest and functional activation. J Cereb Blood Flow Metab. 2015;35:432–42. doi: 10.1038/jcbfm.2014.213.
    1. Angleys H, Østergaard L, Jespersen SN. The effects of capillary transit time heterogeneity (CTH) on brain oxygenation. J Cereb Blood Flow Metab. 2015
    1. Isakson BE, Damon DN, Day KH, Liao Y, Duling BR. Connexin40 and connexin43 in mouse aortic endothelium: evidence for coordinated regulation. Am J Physiol Heart Circ Physiol. 2006;290:H1199–205. doi: 10.1152/ajpheart.00945.2005.
    1. Segal SS, Duling BR. Flow control among microvessels coordinated by intercellular conduction. Science. 1986;234:868–70. doi: 10.1126/science.3775368.
    1. Pries AR, Hopfner M, le Noble F, Dewhirst MW, Secomb TW. The shunt problem: control of functional shunting in normal and tumour vasculature. Nat Rev Cancer. 2010;10:587–93. doi: 10.1038/nrc2895.
    1. Armulik A, Abramsson A, Betsholtz C. Endothelial/pericyte interactions. Circ Res. 2005;97:512–23. doi: 10.1161/01.RES.0000182903.16652.d7.
    1. Hall CN, Reynell C, Gesslein B, et al. Capillary pericytes regulate cerebral blood flow in health and disease. Nature. 2014;508:55–60. doi: 10.1038/nature13165.
    1. Vink H, Duling BR. Identification of distinct luminal domains for macromolecules, erythrocytes, and leukocytes within mammalian capillaries. Circ Res. 1996;79:581–9. doi: 10.1161/01.RES.79.3.581.
    1. Secomb TW, Hsu R, Pries AR. A model for red blood cell motion in glycocalyx-lined capillaries. Am J Physiol. 1998;274:H1016–22.
    1. Desjardins C, Duling BR. Heparinase treatment suggests a role for the endothelial cell glycocalyx in regulation of capillary hematocrit. Am J Physiol. 1990;258:H647–54.
    1. Mazzoni MC, Schmid-Schonbein GW. Mechanisms and consequences of cell activation in the microcirculation. Cardiovasc Res. 1996;32:709–19. doi: 10.1016/0008-6363(96)00146-0.
    1. Østergaard L, Aamand R, Gutierrez-Jimenez E, et al. The capillary dysfunction hypothesis of Alzheimer’s disease. Neurobiol Aging. 2013;34:1018–31. doi: 10.1016/j.neurobiolaging.2012.09.011.
    1. Kalaria RN. Cerebral vessels in ageing and Alzheimer’s disease. Pharmacol Ther. 1996;72:193–214. doi: 10.1016/S0163-7258(96)00116-7.
    1. Bell MA, Ball MJ. Morphometric comparison of hippocampal microvasculature in ageing and demented people: diameters and densities. Acta Neuropathol. 1981;53:299–318. doi: 10.1007/BF00690372.
    1. Junker U, Jaggi C, Bestetti G, Rossi GL. Basement membrane of hypothalamus and cortex capillaries from normotensive and spontaneously hypertensive rats with streptozotocin-induced diabetes. Acta Neuropathol. 1985;65:202–8. doi: 10.1007/BF00686999.
    1. Tagami M, Nara Y, Kubota A, Fujino H, Yamori Y. Ultrastructural changes in cerebral pericytes and astrocytes of stroke-prone spontaneously hypertensive rats. Stroke. 1990;21:1064–71. doi: 10.1161/01.STR.21.7.1064.
    1. Schonfelder U, Hofer A, Paul M, Funk RH. In situ observation of living pericytes in rat retinal capillaries. Microvasc Res. 1998;56:22–9. doi: 10.1006/mvre.1998.2086.
    1. Kawamura H, Kobayashi M, Li Q, et al. Effects of angiotensin II on the pericyte-containing microvasculature of the rat retina. J Physiol. 2004;561:671–83. doi: 10.1113/jphysiol.2004.073098.
    1. Johnson PC, Brendel K, Meezan E. Thickened cerebral cortical capillary basement membranes in diabetics. Arch Pathol Lab Med. 1982;106:214–7.
    1. Reske-Nielsen E, Lundbæk K, Rafaelsen OJ. Pathological changes in the central and peripheral nervous system of young long-term diabetics. I. Diabetic encephalopathy. Diabetologia. 1965;1:233–41. doi: 10.1007/BF01257917.
    1. Barnes AJ, Locke P, Scudder PR, Dormandy TL, Dormandy JA, Slack J. Is hyperviscosity a treatable component of diabetic microcirculatory disease? Lancet. 1977;2:789–91. doi: 10.1016/S0140-6736(77)90724-3.
    1. McCuskey PA, McCuskey RS. In vivo and electron microscopic study of the development of cerebral diabetic microangiography. Microcirc Endothel Lymphat. 1984;1:221–44.
    1. Price TO, Eranki V, Banks WA, Ercal N, Shah GN. Topiramate treatment protects blood-brain barrier pericytes from hyperglycemia-induced oxidative damage in diabetic mice. Endocrinology. 2012;153:362–72. doi: 10.1210/en.2011-1638.
    1. Vink H, Constantinescu AA, Spaan JA. Oxidized lipoproteins degrade the endothelial surface layer : implications for platelet-endothelial cell adhesion. Circulation. 2000;101:1500–2. doi: 10.1161/01.CIR.101.13.1500.
    1. Constantinescu AA, Vink H, Spaan JA. Elevated capillary tube hematocrit reflects degradation of endothelial cell glycocalyx by oxidized LDL. Am J Physiol Heart Circ Physiol. 2001;280:H1051–7.
    1. Czarnowska E, Karwatowska-Prokopczuk E. Ultrastructural demonstration of endothelial glycocalyx disruption in the reperfused rat heart. Involvement of oxygen free radicals. Basic Res Cardiol. 1995;90:357–64. doi: 10.1007/BF00788496.
    1. Shah GN, Price TO, Banks WA, et al. Pharmacological inhibition of mitochondrial carbonic anhydrases protects mouse cerebral pericytes from high glucose-induced oxidative stress and apoptosis. J Pharmacol Exp Ther. 2013;344:637–45. doi: 10.1124/jpet.112.201400.
    1. Farkas E, Luiten PG. Cerebral microvascular pathology in aging and Alzheimer’s disease. Prog Neurobiol. 2001;64:575–611. doi: 10.1016/S0301-0082(00)00068-X.
    1. Verbeek MM, de Waal RM, Schipper JJ, Van Nostrand WE. Rapid degeneration of cultured human brain pericytes by amyloid beta protein. J Neurochem. 1997;68:1135–41. doi: 10.1046/j.1471-4159.1997.68031135.x.
    1. Wilhelmus MMM, Otte-Höller I, van Triel JJJ, et al. Lipoprotein receptor-related protein-1 mediates amyloid-β-mediated cell death of cerebrovascular cells. Am J Pathol. 2007;171:1989–99. doi: 10.2353/ajpath.2007.070050.
    1. Albaugh G, Bellavance E, Strande L, Heinburger S, Hewitt CW, Alexander JB. Nicotine induces mononuclear leukocyte adhesion and expression of adhesion molecules, VCAM and ICAM, in endothelial cells in vitro. Ann Vasc Surg. 2004;18:302–7. doi: 10.1007/s10016-004-0030-9.
    1. Yong T, Zheng MQ, Linthicum DS. Nicotine induces leukocyte rolling and adhesion in the cerebral microcirculation of the mouse. J Neuroimmunol. 1997;80:158–64. doi: 10.1016/S0165-5728(97)00151-3.
    1. Østergaard L, Jespersen SN, Mouridsen K, et al. The role of the cerebral capillaries in acute ischemic stroke: the extended penumbra model. J Cereb Blood Flow Metab. 2013;33:635–48. The authors demonstrate that a certain CTH threshold, CTHmaxexists for which any changes in CBF (increases or reductions) or additional capillary flow disturbances (eg. increased blood viscosity) can cause stroke-like symptoms. They propose that severe capillary dysfunction represent a condition of extreme risk of both stroke and dementia, and show that flow metabolism coupling mechanisms will cause CBF to approach the classical ischemic threshold as CTH approaches CTHmax. They argue that irreversible per-ischemic capillary compression or pericyte constrictions are likely to cause severe tissue hypoxia upon reperfusion and underscore the importance of capillary reperfusion in stroke care, irrespective of whether recanalization therapy is attempted or not. They discover that the Tmaxparameter, which is used by some for imaging-based selection for revascularization therapy, show a coincidental correlation with oxygen extraction efficacy, OEFmax.
    1. Dalkara T, Arsava EM. Can restoring incomplete microcirculatory reperfusion improve stroke outcome after thrombolysis? J Cereb Blood Flow Metab. 2012;32:2091–9. doi: 10.1038/jcbfm.2012.139.
    1. Lassen NA. The luxury-perfusion syndrome and its possible relation to acute metabolic acidosis localised within the brain. Lancet. 1966;2:1113–5. doi: 10.1016/S0140-6736(66)92199-4.
    1. Østergaard L, Chesler DA, Weisskoff RM, Sorensen AG, Rosen BR. Modeling cerebral blood flow and flow heterogeneity from magnetic resonance residue data. J Cereb Blood Flow Metab. 1999;19:690–9. doi: 10.1097/00004647-199906000-00013.
    1. Stewart GN. Researches on the circulation time in organs and on the influences which affect it. Parts I.-III. J Physiol. 1893;15:1–89. doi: 10.1113/jphysiol.1893.sp000462.
    1. Iadecola C. Neurovascular regulation in the normal brain and in Alzheimer’s disease. Nat Rev Neurosci. 2004;5:347–60. doi: 10.1038/nrn1387.
    1. Al-Saeedi FJ. Perfusion scanning using 99mTc-HMPAO detects early cerebrovascular changes in the diabetic rat. BMC Med Phys. 2008;8:1. doi: 10.1186/1756-6649-8-1.
    1. Simpson RE, 3rd, Phillis JW, Buchannan J. A comparison of cerebral blood flow during basal, hypotensive, hypoxic and hypercapnic conditions between normal and streptozotocin diabetic rats. Brain Res. 1990;531:136–42. doi: 10.1016/0006-8993(90)90766-5.
    1. Rubin MJ, Bohlen HG. Cerebral vascular autoregulation of blood flow and tissue PO2 in diabetic rats. Am J Physiol. 1985;249:H540–6.
    1. Kim T, Richard Jennings J, Kim SG. Regional cerebral blood flow and arterial blood volume and their reactivity to hypercapnia in hypertensive and normotensive rats. J Cereb Blood Flow Metab. 2014;34:408–14. doi: 10.1038/jcbfm.2013.197.
    1. Scarmeas N, Habeck CG, Stern Y, Anderson KE. APOE genotype and cerebral blood flow in healthy young individuals. JAMA. 2003;290:1581–2. doi: 10.1001/jama.290.12.1581.
    1. Scarmeas N, Habeck CG, Hilton J, et al. APOE related alterations in cerebral activation even at college age. J Neurol Neurosurg Psychiatry. 2005;76:1440–4. doi: 10.1136/jnnp.2004.053645.
    1. Bookheimer SY, Strojwas MH, Cohen MS, et al. Patterns of brain activation in people at risk for Alzheimer’s disease. N Engl J Med. 2000;343:450–6. doi: 10.1056/NEJM200008173430701.
    1. Fleisher AS, Podraza KM, Bangen KJ, et al. Cerebral perfusion and oxygenation differences in Alzheimer’s disease risk. Neurobiol Aging. 2009;30:1737–48. doi: 10.1016/j.neurobiolaging.2008.01.012.
    1. Filippini N, MacIntosh BJ, Hough MG, et al. Distinct patterns of brain activity in young carriers of the APOE-epsilon4 allele. Proc Natl Acad Sci U S A. 2009;106:7209–14. doi: 10.1073/pnas.0811879106.
    1. Thambisetty M, Beason-Held L, An Y, Kraut MA, Resnick SM. APOE epsilon4 genotype and longitudinal changes in cerebral blood flow in normal aging. Arch Neurol. 2010;67:93–8.
    1. Girouard H, Park L, Anrather J, Zhou P, Iadecola C. Cerebrovascular nitrosative stress mediates neurovascular and endothelial dysfunction induced by angiotensin II. Arterioscler Thromb Vasc Biol. 2007;27:303–9. doi: 10.1161/01.ATV.0000253885.41509.25.
    1. Niwa K, Porter VA, Kazama K, Cornfield D, Carlson GA, Iadecola C. A beta-peptides enhance vasoconstriction in cerebral circulation. Am J Physiol Heart Circ Physiol. 2001;281:H2417–24.
    1. Touyz RM, Schiffrin EL. Reactive oxygen species in vascular biology: implications in hypertension. Histochem Cell Biol. 2004;122:339–52. doi: 10.1007/s00418-004-0696-7.
    1. Marin JM, Carrizo SJ, Vicente E, Agusti AG. Long-term cardiovascular outcomes in men with obstructive sleep apnoea-hypopnoea with or without treatment with continuous positive airway pressure: an observational study. Lancet. 2005;365:1046–53. doi: 10.1016/S0140-6736(05)74229-X.
    1. Pantoni L. Cerebral small vessel disease: from pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol. 2010;9:689–701. doi: 10.1016/S1474-4422(10)70104-6.
    1. Moore DF, Altarescu G, Barker WC, Patronas NJ, Herscovitch P, Schiffmann R. White matter lesions in Fabry disease occur in ‘prior’ selectively hypometabolic and hyperperfused brain regions. Brain Res Bull. 2003;62:231–40. doi: 10.1016/j.brainresbull.2003.09.021.
    1. Takahashi S, Tohgi H, Yonezawa H, Obara S, Nagane Y. Cerebral blood flow and oxygen metabolism before and after a stroke-like episode in patients with mitochondrial myopathy, encephalopathy, lactic acidosis and stroke-like episodes (MELAS) J Neurol Sci. 1998;158:58–64. doi: 10.1016/S0022-510X(98)00105-1.
    1. Lavallee P, Perchaud V, Gautier-Bertrand M, Grabli D, Amarenco P. Association between influenza vaccination and reduced risk of brain infarction. Stroke. 2002;33:513–8. doi: 10.1161/hs0202.102328.
    1. Nichol KL, Nordin J, Mullooly J, Lask R, Fillbrandt K, Iwane M. Influenza vaccination and reduction in hospitalizations for cardiac disease and stroke among the elderly. N Engl J Med. 2003;348:1322–32. doi: 10.1056/NEJMoa025028.
    1. Grau AJ, Fischer B, Barth C, Ling P, Lichy C, Buggle F. Influenza vaccination is associated with a reduced risk of stroke. Stroke. 2005;36:1501–6. doi: 10.1161/01.STR.0000170674.45136.80.
    1. James P, Ellis CJ, Whitlock RM, McNeil AR, Henley J, Anderson NE. Relation between troponin T concentration and mortality in patients presenting with an acute stroke: observational study. BMJ. 2000;320:1502–4. doi: 10.1136/bmj.320.7248.1502.
    1. Jensen JK, Kristensen SR, Bak S, Atar D, Hoilund-Carlsen PF, Mickley H. Frequency and significance of troponin T elevation in acute ischemic stroke. Am J Cardiol. 2007;99:108–12. doi: 10.1016/j.amjcard.2006.07.071.
    1. Østergaard L, Sorensen AG, Kwong KK, Weisskoff RM, Gyldensted C, Rosen BR. High resolution measurement of cerebral blood flow using intravascular tracer bolus passages. Part II: experimental comparison and preliminary results. Magn Reson Med. 1996;36:726–36. doi: 10.1002/mrm.1910360511.
    1. Østergaard L, Weisskoff RM, Chesler DA, Gyldensted C, Rosen BR. High resolution measurement of cerebral blood flow using intravascular tracer bolus passages. Part I: mathematical approach and statistical analysis. Magn Reson Med. 1996;36:715–25. doi: 10.1002/mrm.1910360510.
    1. Takasawa M, Jones PS, Guadagno JV, et al. How reliable is perfusion MR in acute stroke? Validation and determination of the penumbra threshold against quantitative PET. Stroke. 2008;39:870–7. doi: 10.1161/STROKEAHA.107.500090.
    1. Olivot JM, Mlynash M, Thijs VN, et al. Optimal Tmax threshold for predicting penumbral tissue in acute stroke. Stroke. 2009;40:469–75. doi: 10.1161/STROKEAHA.108.526954.
    1. Mouridsen K, Christensen S, Gyldensted L, Østergaard L. Automatic selection of arterial input function using cluster analysis. Magn Reson Med. 2006;55:524–31. doi: 10.1002/mrm.20759.
    1. Mouridsen K, Friston K, Hjort N, Gyldensted L, Østergaard L, Kiebel S. Bayesian estimation of cerebral perfusion using a physiological model of microvasculature. Neuroimage. 2006;33:570–9. doi: 10.1016/j.neuroimage.2006.06.015.
    1. Mouridsen K, Hansen MB, Østergaard L, Jespersen SN. Reliable estimation of capillary transit time distributions using DSC-MRI. J Cereb Blood Flow Metab. 2014;34:1511–21. doi: 10.1038/jcbfm.2014.111.
    1. Vogel J, Waschke KF, Kuschinsky W. Flow-independent heterogeneity of brain capillary plasma perfusion after blood exchange with a Newtonian fluid. Am J Physiol. 1997;272:H1833–7.
    1. Hutchinson EB, Stefanovic B, Koretsky AP, Silva AC. Spatial flow-volume dissociation of the cerebral microcirculatory response to mild hypercapnia. Neuroimage. 2006;32:520–30. doi: 10.1016/j.neuroimage.2006.03.033.
    1. Ashkanian M, Gjedde A, Mouridsen K, et al. Carbogen inhalation increases oxygen transport to hypoperfused brain tissue in patients with occlusive carotid artery disease: increased oxygen transport to hypoperfused brain. Brain Res. 2009;1304:90–5. doi: 10.1016/j.brainres.2009.09.076.
    1. Sakadzic S, Roussakis E, Yaseen MA, et al. Two-photon high-resolution measurement of partial pressure of oxygen in cerebral vasculature and tissue. Nat Methods. 2010;7:755–9. doi: 10.1038/nmeth.1490.
    1. Sakadzic S, Mandeville ET, Gagnon L, et al. Large arteriolar component of oxygen delivery implies a safe margin of oxygen supply to cerebral tissue. Nat Commun. 2014;5:5734. doi: 10.1038/ncomms6734.
    1. Lee J, Wu W, Lesage F, Boas DA. Multiple-capillary measurement of RBC speed, flux, and density with optical coherence tomography. J Cereb Blood Flow Metab. 2013;33:1707–10. doi: 10.1038/jcbfm.2013.158.
    1. Eltzschig HK, Carmeliet P. Hypoxia and inflammation. N Engl J Med. 2011;364:656–65. doi: 10.1056/NEJMra0910283.
    1. Zhang X, Le W. Pathological role of hypoxia in Alzheimer’s disease. Exp Neurol. 2010;223:299–303. doi: 10.1016/j.expneurol.2009.07.033.
    1. Bell RD, Deane R, Chow N, et al. SRF and myocardin regulate LRP-mediated amyloid-beta clearance in brain vascular cells. Nat Cell Biol. 2009;11:143–53. doi: 10.1038/ncb1819.
    1. Wardlaw JM, Smith EE, Biessels GJ, et al. Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration. Lancet Neurol. 2013;12:822–38. doi: 10.1016/S1474-4422(13)70124-8.
    1. Iadecola C, Davisson RL. Hypertension and cerebrovascular dysfunction. Cell Metab. 2008;7:476–84. doi: 10.1016/j.cmet.2008.03.010.
    1. Kittaka M, Wang L, Sun N, et al. Brain capillary tissue plasminogen activator in a diabetes stroke model. Stroke. 1996;27:712–9. doi: 10.1161/01.STR.27.4.712.
    1. Yemisci M, Gursoy-Ozdemir Y, Vural A, Can A, Topalkara K, Dalkara T. Pericyte contraction induced by oxidative-nitrative stress impairs capillary reflow despite successful opening of an occluded cerebral artery. Nat Med. 2009;15:1031–7. doi: 10.1038/nm.2022.
    1. Østergaard L, Aamand R, Karabegovic S, et al. The role of the microcirculation in delayed cerebral ischemia and chronic degenerative changes after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2013;33:1825–37. doi: 10.1038/jcbfm.2013.173.
    1. Perkio J, Soinne L, Østergaard L, et al. Abnormal intravoxel cerebral blood flow heterogeneity in human ischemic stroke determined by dynamic susceptibility contrast magnetic resonance imaging. Stroke. 2005;36:44–9. doi: 10.1161/01.STR.0000150495.96471.95.
    1. Simonsen CZ, Rohl L, Vestergaard-Poulsen P, Gyldensted C, Andersen G, Østergaard L. Final infarct size after acute stroke: prediction with flow heterogeneity. Radiology. 2002;225:269–75. doi: 10.1148/radiol.2251011249.
    1. Powers WJ, Clarke WR, Grubb RL, Jr, et al. Extracranial-intracranial bypass surgery for stroke prevention in hemodynamic cerebral ischemia: the Carotid Occlusion Surgery Study randomized trial. JAMA. 2011;306:1983–92. doi: 10.1001/jama.2011.1610.

Source: PubMed

3
Subskrybuj