Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-tj2md Total loading time: 0 Render date: 2024-04-23T17:18:11.877Z Has data issue: false hasContentIssue false

Section 1 - Etiology, Pathophysiology, and Imaging

Published online by Cambridge University Press:  16 May 2019

Michael Brainin
Affiliation:
Donau-Universität Krems, Austria
Wolf-Dieter Heiss
Affiliation:
Universität zu Köln
Get access
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2019

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

References

Shakir, R, Norrving, B. Stroke in ICD-11: the end of a long exile. Lancet 2017; 389(10087): 2373.Google Scholar
Sanz, J, Moreno, PR, Fuster, V. The year in atherothrombosis. J Am Coll Cardiol 2012; 60(10): 932–42.Google Scholar
Rajamani, K, Fisher, M, Fisher, M. Atherosclerosis – pathogenesis and pathophysiology. In: Ginsberg, MD, Bogousslavsky, J, eds. Cerebrovascular Disease: Pathophysiology, Diagnosis and Management. II. London: Blackwell Science; 1998: 308–18.Google Scholar
Willeit, J, Kiechl, S. Biology of arterial atheroma. Cerebrovasc Dis 2000; 10(Suppl 5): 18.CrossRefGoogle ScholarPubMed
Ross, R. Atherosclerosis – an inflammatory disease. N Engl J Med 1999; 340(2): 115–26.CrossRefGoogle ScholarPubMed
Faxon, DP, Fuster, V, Libby, P, et al. Atherosclerotic Vascular Disease Conference: Writing Group III: Pathophysiology. Circulation 2004; 109(21): 2617–25.CrossRefGoogle ScholarPubMed
Aikawa, M, Libby, P. The vulnerable atherosclerotic plaque: pathogenesis and therapeutic approach. Cardiovasc Pathol 2004; 13(3): 125–38.CrossRefGoogle ScholarPubMed
Koga, J, Aikawa, M. Crosstalk between macrophages and smooth muscle cells in atherosclerotic vascular diseases. Vascular Pharmacology 2012; 57(1): 24–8.CrossRefGoogle ScholarPubMed
Dzau, VJ, Braun-Dullaeus, RC, Sedding, DG. Vascular proliferation and atherosclerosis: new perspectives and therapeutic strategies. Nat Med 2002; 8(11): 1249–56.Google Scholar
Madden, JA. Role of the vascular endothelium and plaque in acute ischemic stroke. Neurology 2012; 79(13 Suppl 1): S58–62.CrossRefGoogle ScholarPubMed
Glagov, S, Weisenberg, E, Zarins, CK, Stankunavicius, R, Kolettis, GJ. Compensatory enlargement of human atherosclerotic coronary arteries. N Engl J Med 1987; 316(22): 1371–5.Google Scholar
Rauch, U, Osende, JI, Fuster, V, Badimon, JJ, Fayad, Z, Chesebro, JH. Thrombus formation on atherosclerotic plaques: pathogenesis and clinical consequences. Ann Intern Med 2001; 134(3): 224–38.CrossRefGoogle ScholarPubMed
Lubos, E, Handy, DE, Loscalzo, J. Role of oxidative stress and nitric oxide in atherothrombosis. Front Biosci 2008; 13: 5323–44.Google ScholarPubMed
Fisher, CM. Cerebral miliary aneurysms in hypertension. Am J Pathol 1972; 66(2): 313–30.Google ScholarPubMed
Zülch, K-J. The Cerebral Infarct. Pathology, Pathogenesis, and Computed Tomography. Berlin, Heidelberg, New York, Tokyo: Springer-Verlag; 1985.Google Scholar
Russell, RWR. Observations on intracerebral aneurysms. Brain 1963; 86: 425–42.CrossRefGoogle Scholar
Zülch, K-J. Über die Entstehung und Lokalisation der Hirninfarkte Zbl für. Neurochirurgie 1961; 21: 158–78.Google Scholar
Dichgans, M. Genetics of ischaemic stroke. Lancet Neurol 2007; 6(2): 149–61.Google Scholar
Mohr, JP, Choi, DW, Grotta, JC, Weir, B, Wolf, PA, eds. Stroke – Pathophysiology, Diagnosis, and Management. 4th edn. Philadelphia: Churchill Livingstone; 2004.Google Scholar
Pantoni, L. Cerebral small vessel disease: from pathogenesis and clinical characteristics to therapeutic challenges. Lancet Neurol 2010; 9(7): 689701.Google Scholar
Ringelstein, EB, Nabavi, DG. Cerebral small vessel diseases: cerebral microangiopathies. Curr Opin Neurol 2005; 18(2): 179–88.Google Scholar
Wolf, PA. Epidemiology of stroke. In: Mohr, JP, Choi, DW, Grotta, JC, Weir, B, Wolf, PA, eds. Stroke – Pathophysiology, Diagnosis, and Management. Philadelphia: Churchill Livingstone; 2004: 1334.Google Scholar
Benjamin, EJ, Blaha, MJ, Chiuve, SE, et al. Heart Disease and Stroke Statistics – 2017 Update: A Report From the American Heart Association. Circulation 2017; 135(10): e146–603.CrossRefGoogle ScholarPubMed
Stochdorph, O. Der Mythos der letzten Wiese. Zbl allg Pathol path Anat 1977; 121: 554.Google Scholar
Ringelstein, EB, Zunker, P. Low-flow infarction. In: Ginsberg, MD, Bogousslavsky, J, eds. Cerebrovascular Disease: Pathophysiology, Diagnosis, and Management. II. London: Blackwell Science; 1998: 1075–89.Google Scholar
Fisher, CM. Lacunes: small, deep cerebral infarcts. Neurology 1965; 15: 774–84.CrossRefGoogle ScholarPubMed
Amarenco, P, Bogousslavsky, J, Caplan, LR, Donnan, GA, Hennerici, MG. New approach to stroke subtyping: the A-S-C-O (phenotypic) classification of stroke. Cerebrovasc Dis 2009; 27(5): 502–8.CrossRefGoogle ScholarPubMed
Beghi, E, Bogliun, G, Cavaletti, G, et al. Hemorrhagic infarction: risk factors, clinical and tomographic features, and outcome. A case-control study. Acta Neurol Scand 1989; 80(3): 226–31.Google Scholar
Lodder, J, Krijne-Kubat, B, Broekman, J. Cerebral hemorrhagic infarction at autopsy: cardiac embolic cause and the relationship to the cause of death. Stroke 1986; 17(4): 626–9.Google Scholar
Fisher, M, Adams, RD. Observations on brain embolism with special reference to the mechanism of hemorrhagic infarction. J Neuropathol Exp Neurol 1951; 10(1): 92–4.Google Scholar
Mohr, JP, Caplan, LR, Melski, JW, et al. The Harvard Cooperative Stroke Registry: a prospective registry. Neurology 1978; 28: 754–62.Google Scholar
Sacco, RL, Wolf, PA, Bharucha, NE, et al. Subarachnoid and intracerebral hemorrhage: natural history, prognosis, and precursive factors in the Framingham Study. Neurology 1984; 34: 847–54.Google Scholar
Feldman, E. Intracerebral Hemorrhage. Armonk, NY: Futura; 1994.Google Scholar
Schütz, H. Spontane intrazerebrale Hämatome. Pathophysiologie, Klinik und Therapie. Berlin, Heidelberg, New York: Springer-Verlag; 1988.CrossRefGoogle Scholar
Qureshi, AI, Tuhrim, S, Broderick, JP, Batjer, HH, Hondo, H, Hanley, DF. Spontaneous intracerebral hemorrhage. N Engl J Med 2001; 344(19): 1450–60.Google Scholar
Kase, CS, Mohr, JP, Caplan, LR. Intracerebral hemorrhage. In: Mohr, JP, Choi, DW, Grotta, JC, Weir, B, Wolf, PA, eds. Stroke – Pathophysiology, Diagnosis, and Management. Philadelphia: Churchill Livingstone; 2004: 327–76.Google Scholar
Fisher, CM. Pathological observations in hypertensive cerebral hemorrhage. J Neuropathol Exp Neurol 1971; 30(3): 536–50.CrossRefGoogle ScholarPubMed
Brott, T, Broderick, J, Kothari, R, et al. Early hemorrhage growth in patients with intracerebral hemorrhage. Stroke 1997; 28(1): 15.Google Scholar
Gonzalez-Duarte, A, Cantu, C, Ruiz-Sandoval, JL, Barinagarrementeria, F. Recurrent primary cerebral hemorrhage: frequency, mechanisms, and prognosis. Stroke 1998; 29(9): 1802–5.CrossRefGoogle ScholarPubMed
Bousser, MG, Barnett, HJM. Cerebral venous thrombosis. In: Mohr, JP, Choi, DW, Grotta, JC, Weir, B, Wolf, PA, eds. Stroke – Pathophysiology, Diagnosis, and Management. 4th edn. Philadelphia: Churchill Livingstone; 2004: 301–25.Google Scholar
Petito, CK. The Neuropathology of Focal Brain Ischemia. Basel: ISN Neuropath; 2005.Google Scholar
Sakadzic, S, Lee, J, Boas, DA, Ayata, C. High-resolution in vivo optical imaging of stroke injury and repair. Brain Res 2015; 1623: 174–92.CrossRefGoogle ScholarPubMed
Zille, M, Farr, TD, Przesdzing, I, et al. Visualizing cell death in experimental focal cerebral ischemia: promises, problems, and perspectives. J Cereb Blood Flow Metab 2012; 32(2): 213–31.Google Scholar
Brown, AW, Brierley, JB. Anoxic-ischaemic cell change in rat brain light microscopic and fine-structural observations. J Neurol Sci 1972; 16(1): 5984.CrossRefGoogle ScholarPubMed
Jortner, BS. The return of the dark neuron. A histological artifact complicating contemporary neurotoxicologic evaluation. Neurotoxicology 2006; 27(4): 628–34.Google Scholar
Kirino, T, Sano, K. Selective vulnerability in the gerbil hippocampus following transient ischemia. Acta Neuropathol 1984; 62: 201–8.Google Scholar
Martin, LJ. Neuronal cell death in nervous system development, disease, and injury (Review). Int J Mol Med 2001; 7(5): 455–78.Google Scholar
Dixon, SJ, Lemberg, KM, Lamprecht, MR, et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 2012; 149(5): 1060–72.Google Scholar
Charriaut-Marlangue, C, Ben-Ari, Y. A cautionary note on the use of the TUNEL stain to determine apoptosis. Neuroreport 1995; 7(1): 61–4.Google Scholar
Hossmann, KA. Disturbances of cerebral protein synthesis and ischemic cell death. ProgBrain Res 1993; 96: 161–77.Google Scholar
DeGracia, DJ, Jamison, JT, Szymanski, JJ, Lewis, MK. Translation arrest and ribonomics in post-ischemic brain: layers and layers of players. J Neurochem 2008; 106(6): 2288–301.Google Scholar
Stanimirovic, DB, Friedman, A. Pathophysiology of the neurovascular unit: disease cause or consequence? J Cereb Blood Flow Metab 2012; 32(7): 1207–21.Google Scholar
Dalkara, T, Alarcon-Martinez, L. Cerebral microvascular pericytes and neurogliovascular signaling in health and disease. Brain Res 2015; 1623: 317.Google Scholar
Wiltrout, C, Lang, B, Yan, Y, Dempsey, RJ, Vemuganti, R. Repairing brain after stroke: a review on post-ischemic neurogenesis. Neurochem Int 2007; 50(7–8): 1028–41.Google Scholar
Kamouchi, M, Ago, T, Kuroda, J, Kitazono, T. The possible roles of brain pericytes in brain ischemia and stroke. Cell Mol Neurobiol 2012; 32(2): 159–65.CrossRefGoogle ScholarPubMed
Huo, W, Liu, X, Tan, C, et al. Stem cell transplantation for treating stroke: status, trends and development. Neural Regen Res 2014; 9(17): 1643–8.Google Scholar
Unsworth, DJ, Mathias, JL. Traumatic brain injury and alcohol/substance abuse: a Bayesian meta-analysis comparing the outcomes of people with and without a history of abuse. J Clin Exp Neuropsychol 2017; 39(6): 547–62.Google Scholar
Kumar, A, Aakriti, Gupta V. A review on animal models of stroke: an update. Brain Res Bull 2016; 122: 3544.CrossRefGoogle ScholarPubMed
Hudgins, WR, Garcia, JH. Transorbital approach to the middle cerebral artery of the squirrel monkey: a technique for experimental cerebral infarction applicable to ultrastructural studies. Stroke 1970; 1: 107–11.CrossRefGoogle Scholar
Tamura, A, Graham, DI, McCulloch, J, Teasdale, GM. Focal cerebral ischaemia in the rat: 1. Description of technique and early neuropathological consequences following middle cerebral artery occlusion. J Cereb Blood Flow Metab 1981; 1: 5360.Google Scholar
Knapp, L, Gellert, L, Heredi, J, et al. A simple novel technique to induce short-lasting local brain ischaemia in the rat. Neuropathol Appl Neurobiol 2014; 40(5): 603–9.Google Scholar
Koizumi, J, Yoshida, Y, Nakazawa, T, Ooneda, G. Experimental studies of ischemic brain edema. 1. A new experimental model of cerebral embolism in rats in which recirculation can be introduced in the ischemic area. Japanese Journal of Stroke 1986; 8: 18.Google Scholar
Rogers, DC, Campbell, CA, Stretton, JL, Mackay, KB. Correlation between motor impairment and infarct volume after permanent and transient middle cerebral artery occlusion in the rat. Stroke 1997; 28(10): 2060–5.Google Scholar
Sommer, CJ. Ischemic stroke: experimental models and reality. Acta Neuropathol 2017; 133(2): 245–61.Google Scholar
Dinapoli, VA, Rosen, CL, Nagamine, T, Crocco, T. Selective MCA occlusion: a precise embolic stroke model. J Neurosci Methods 2006; 154(1–2): 233–8.Google Scholar
Chen, Y, Zhu, W, Zhang, W, et al. A novel mouse model of thromboembolic stroke. J Neurosci Methods 2015; 256: 203–11.Google Scholar
Chen, F, Suzuki, Y, Nagai, N, et al. Rodent stroke induced by photochemical occlusion of proximal middle cerebral artery: evolution monitored with MR imaging and histopathology. Eur J Radiol 2007; 63(1): 6875.Google Scholar
Lees, KR, Bluhmki, E, von Kummer, R, et al. Time to treatment with intravenous alteplase and outcome in stroke: an updated pooled analysis of ECASS, ATLANTIS, NINDS, and EPITHET trials. Lancet 2010; 375(9727): 1695–703.Google Scholar
Goyal, M, Menon, BK, van Zwam, WH, et al. Endovascular thrombectomy after large-vessel ischaemic stroke: a meta-analysis of individual patient data from five randomised trials. Lancet 2016; 387(10029): 1723–31.Google Scholar
Vidale, S, Agostoni, E. Endovascular treatment of ischemic stroke: an updated meta-analysis of efficacy and safety. Vasc Endovascular Surg 2017; 51(4): 215–19.CrossRefGoogle ScholarPubMed
Mordasini, P, Frabetti, N, Gralla, J, et al. In vivo evaluation of the first dedicated combined flow-restoration and mechanical thrombectomy device in a swine model of acute vessel occlusion. AJNR Am J Neuroradiol 2011; 32(2): 294300.Google Scholar
Sutherland, BA, Neuhaus, AA, Couch, Y, et al. The transient intraluminal filament middle cerebral artery occlusion model as a model of endovascular thrombectomy in stroke. J Cereb Blood Flow Metab 2016; 36(2): 363–9.Google Scholar
Sharma, P, Yadav, S, Meschia, JF. Genetics of ischaemic stroke. J Neurol Neurosurg Psychiatry 2013; 84(12): 1302–8.Google Scholar
Traylor, M, Farrall, M, Holliday, EG, et al. Genetic risk factors for ischaemic stroke and its subtypes (the METASTROKE collaboration): a meta-analysis of genome-wide association studies. Lancet Neurol 2012; 11(11): 951–62.Google Scholar
Biffi, A, Sonni, A, Anderson, CD, et al. Variants at APOE influence risk of deep and lobar intracerebral hemorrhage. Ann Neurol 2010; 68(6): 934–43.Google Scholar
Lindgren, A. Stroke genetics: a review and update. J Stroke 2014; 16(3): 114–23.Google Scholar
Koller, A, Toth, P. Contribution of flow-dependent vasomotor mechanisms to the autoregulation of cerebral blood flow. Journal of Vascular Research 2012; 49(5): 375–89.CrossRefGoogle Scholar
Feuerstein, D, Takagaki, M, Gramer, M, et al. Detecting tissue deterioration after brain injury: regional blood flow level versus capacity to raise blood flow. J Cereb Blood Flow Metab 2014; 34(7): 1117–27.Google Scholar
Hata, R, Maeda, K, Hermann, D, Mies, G, Hossmann, KA. Evolution of brain infarction after transient focal cerebral ischemia in mice. J Cereb Blood Flow Metab 2000; 20(6): 937–46.Google Scholar
del Zoppo, GJ. The neurovascular unit in the setting of stroke. J Intern Med 2010; 267(2): 156–71.Google Scholar
Fernandez-Klett, F, Priller, J. Diverse functions of pericytes in cerebral blood flow regulation and ischemia. J Cereb Blood Flow Metab 2015; 35(6): 883–7.Google Scholar
Ito, U, Hakamata, Y, Kawakami, E, Oyanagi, K. Temporary cerebral ischemia results in swollen astrocytic end-feet that compress microvessels and lead to delayed focal cortical infarction. J Cereb Blood Flow Metab 2011; 31(1): 328–38.CrossRefGoogle ScholarPubMed
Gursoy-Ozdemir, Y, Yemisci, M, Dalkara, T. Microvascular protection is essential for successful neuroprotection in stroke. J Neurochem 2012; 123(Suppl 2): 211.Google Scholar
Liebeskind, DS. Collateral circulation. Stroke 2003; 34(9): 2279–84.Google Scholar
Nishijima, Y, Akamatsu, Y, Weinstein, PR, Liu, J. Collaterals: implications in cerebral ischemic diseases and therapeutic interventions. Brain Res 2015; 1623: 1829.Google Scholar
Toole, JF, McGraw, CP. The steal syndromes. Annual Review of Medicine 1975; 26: 321–9.Google Scholar
Opitz, E, Schneider, M. Über die Sauerstoffversorgung des Gehirns und den Mechanismus der Mangelwirkungen. Ergebn Physiol 1950; 46: 126260.Google Scholar
Symon, L, Branston, NM, Strong, AJ, Hope, TD. The concepts of thresholds of ischaemia in relation to brain structure and function. J Clin Pathol Suppl (R Coll Pathol) 1977; 11: 149–54.Google Scholar
Astrup, J, Siesjo, BK, Symon, L. Thresholds in cerebral ischemia – the ischemic penumbra. Stroke 1981; 12(6): 723–5.Google Scholar
Pakkenberg, B, Gundersen, HJ. Neocortical neuron number in humans: effect of sex and age. Journal of Comparative Neurology 1997; 384(2): 312–20.Google ScholarPubMed
Clarke, DD, Sokoloff, L. Circulation and energy metabolism of the brain. In: Siegel, G, Agranoff, B, Albers, RW, Fisher, S, eds. Basic Neurochemistry: Molecular, Cellular, and Medical Aspects. 6th edn. Philadelphia: Lippincott-Raven; 1999: 637–69.Google Scholar
Sokoloff, L. Energetics of functional activation in neural tissues. Neurochem Res 1999; 24(2): 321–9.Google Scholar
Magistretti, PJ, Pellerin, L. Astrocytes couple synaptic activity to glucose utilization in the brain. News Physiol Sci 1999; 14: 177–82.Google Scholar
Laughlin, SB, Attwell, D. The metabolic cost of neural information: from fly eye to mammalian cortex. In: Frackowiak, RSJ, Magistretti, PJ, Shulman, RG, Altman, JS, Adams, M, eds. Neuroenergetics: Relevance for Functional Brain Imaging. Strasbourg: HFSP Workshop XI; 2001: 5464.Google Scholar
Frackowiak, RSJ, Magistretti, PJ, Shulman, RG, Altman, JS, Adams, M. Neuroenergetics: Relevance for Functional Brain Imaging. Strasbourg: HFSP Workshop XI; 2001.Google Scholar
Heiss, WD, Hayakawa, T, Waltz, AG. Cortical neuronal function during ischemia. Effects of occlusion of one middle cerebral artery on single-unit activity in cats. ArchNeurol 1976; 33(12): 813–20.Google Scholar
Hossmann, KA. Cerebral ischemia: models, methods and outcomes. Neuropharmacology 2008; 55(3): 257–70.Google Scholar
Hata, R, Maeda, K, Hermann, D, Mies, G, Hossmann, KA. Dynamics of regional brain metabolism and gene expression after middle cerebral artery occlusion in mice. J Cereb Blood Flow Metab 2000; 20(2): 306–15.Google Scholar
Dani, KA, Warach, S. Metabolic imaging of ischemic stroke: the present and future. AJNR Am J Neuroradiol 2014; 35(6 Suppl): S37–43.Google Scholar
Heiss, WD. Ischemic penumbra: evidence from functional imaging in man. J Cereb Blood Flow Metab 2000; 20(9): 1276–93.CrossRefGoogle ScholarPubMed
Takasawa, M, Beech, JS, Fryer, TD, et al. Imaging of brain hypoxia in permanent and temporary middle cerebral artery occlusion in the rat using 18F-fluoromisonidazole and positron emission tomography: a pilot study. J Cereb Blood Flow Metab 2007; 27(4): 679–89.Google Scholar
Kane, I, Sandercock, P, Wardlaw, J. Magnetic resonance perfusion diffusion mismatch and thrombolysis in acute ischaemic stroke: a systematic review of the evidence to date. J Neurol Neurosurg Psychiatry 2007; 78(5): 485–91.Google Scholar
Hoehn-Berlage, M, Norris, DG, Kohno, K, et al. Evolution of regional changes in apparent diffusion-coefficient during focal ischemia of rat brain: the relationship of quantitative diffusion NMR imaging to reduction in cerebral blood flow and metabolic disturbances. J CerebBlood Flow Metab 1995; 15(6): 1002–11.Google Scholar
Sun, PZ, Zhou, J, Sun, W, Huang, J, van Zijl, PC. Detection of the ischemic penumbra using pH-weighted MRI. J Cereb Blood Flow Metab 2007; 27(6): 1129–36.Google Scholar
Weber, RA, Hui, ES, Jensen, JH, et al. Diffusional kurtosis and diffusion tensor imaging reveal different time-sensitive stroke-induced microstructural changes. Stroke 2015; 46(2): 545–50.Google Scholar
Vaas, M, Klohs, J. Noninvasive optical imaging in rodent models of stroke. In: Dirnagl, U, ed. Rodent Models of Stroke. 2nd edn. New York: Humana Press; 2016: 187–98.Google Scholar
Baskerville, TA, Macrae, IM, Holmes, WM, McCabe, C. The influence of gender on “tissue at risk” in acute stroke: a diffusion-weighted magnetic resonance imaging study in a rat model of focal cerebral ischaemia. J Cereb Blood Flow Metab 2016; 36(2): 381–6.Google Scholar
del Zoppo, GJ, Sharp, FR, Heiss, WD, Albers, GW. Heterogeneity in the penumbra. J Cereb Blood Flow Metab 2011; 31(9): 1836–51.Google Scholar
Dreier, JP. The role of spreading depression, spreading depolarization and spreading ischemia in neurological disease. Nat Med 2011; 17(4): 439–47.CrossRefGoogle ScholarPubMed
Feuerstein, D, Backes, H, Gramer, M, et al. Regulation of cerebral metabolism during cortical spreading depression. J Cereb Blood Flow Metab 2016; 36(11): 1965–77.Google Scholar
Strong, AJ, Anderson, PJ, Watts, HR, et al. Peri-infarct depolarizations lead to loss of perfusion in ischaemic gyrencephalic cerebral cortex. Brain 2007; 130(4): 9951008.Google Scholar
Mies, G, Iijima, T, Hossmann, K-A. Correlation between periinfarct DC shifts and ischemic neuronal damage in rat. NeuroReport 1993; 4(6): 709–11.Google Scholar
Moskowitz, MA, Lo, EH, Iadecola, C. The science of stroke: mechanisms in search of treatments. Neuron 2010; 67(2): 181–98.Google Scholar
Karsy, M, Brock, A, Guan, J, Taussky, P, Kalani, MY, Park, MS. Neuroprotective strategies and the underlying molecular basis of cerebrovascular stroke. Neurosurg Focus 2017; 42(4): E3.Google Scholar
Simon, R, Xiong, Z. Acidotoxicity in brain ischaemia. Biochem Soc Trans 2006; 34(6): 1356–61.Google Scholar
Choi, DW. Excitotoxicity, apoptosis and ischemic stroke. Journal of Biochemistry and Molecular Biology 2001; 34: 814.Google Scholar
Szydlowska, K, Tymianski, M. Calcium, ischemia and excitotoxicity. Cell Calcium 2010; 47(2): 122–9.Google Scholar
MacDonald, JF, Xiong, ZG, Jackson, MF. Paradox of Ca2+ signaling, cell death and stroke. Trends in Neurosciences 2006; 29(2): 7581.Google Scholar
Chan, PH. Reactive oxygen radicals in signaling and damage in the ischemic brain. J Cereb Blood Flow Metab 2001; 21(1): 214.Google Scholar
Granger, DN, Kvietys, PR. Reperfusion injury and reactive oxygen species: the evolution of a concept. Redox Biol 2015; 6: 524–51.Google Scholar
Dalkara, T, Moskowitz, MA. The complex role of nitric oxide in the pathophysiology of focal cerebral ischemia. Brain Pathol 1994; 4: 4957.Google Scholar
Shuttleworth, CW, Weiss, JH. Zinc: new clues to diverse roles in brain ischemia. Trends Pharmacol Sci 2011; 32(8): 480–6.Google Scholar
Sensi, SL, Jeng, JM. Rethinking the excitotoxic ionic milieu: the emerging role of Zn(2+) in ischemic neuronal injury. Current Molecular Medicine 2004; 4(2): 87111.Google Scholar
Yang, W, Paschen, W. Unfolded protein response in brain ischemia: a timely update. J Cereb Blood Flow Metab 2016; 36(12): 2044–50.Google Scholar
Youle, RJ, van der Bliek, AM. Mitochondrial fission, fusion, and stress. Science 2012; 337(6098): 1062–5.Google Scholar
Gouriou, Y, Demaurex, N, Bijlenga, P, De Marchi, U. Mitochondrial calcium handling during ischemia-induced cell death in neurons. Biochimie 2011; 93(12): 2060–7.Google Scholar
Norenberg, MD, Rao, KV. The mitochondrial permeability transition in neurologic disease. Neurochem Int 2007; 50(7–8): 983–97.Google Scholar
Kerr, JF, Wyllie, AH, Currie, AR. Apoptosis: a basic biological phenomenon with wide-ranging implications in tissue kinetics. Br J Cancer 1972; 26(4): 239–57.Google Scholar
Niizuma, K, Yoshioka, H, Chen, H, et al. Mitochondrial and apoptotic neuronal death signaling pathways in cerebral ischemia. Biochim Biophys Acta 2010; 1802(1): 92–9.Google Scholar
Ferrer, I, Planas, AM. Signaling of cell death and cell survival following focal cerebral ischemia: life and death struggle in the penumbra. J Neuropathol Exp Neurol 2003; 62(4): 329–39.Google Scholar
MacManus, JP, Buchan, AM. Apoptosis after experimental stroke: fact or fashion? J Neurotrauma 2000; 17(10): 899914.Google Scholar
Thushara Vijayakumar, N, Sangwan, A, Sharma, B, Majid, A, Rajanikant, GK. Cerebral ischemic preconditioning: the road so far. Mol Neurobiol 2016; 53(4): 2579–93.Google Scholar
Dirnagl, U, Simon, RP, Hallenbeck, JM. Ischemic tolerance and endogenous neuroprotection. Trends in Neurosciences 2003; 26(5): 248–54.Google Scholar
Zhao, H, Sapolsky, RM, Steinberg, GK. Interrupting reperfusion as a stroke therapy: ischemic postconditioning reduces infarct size after focal ischemia in rats. J Cereb Blood Flow Metab 2006; 26(9): 1114–21.Google Scholar
Wang, CX, Shuaib, A. Critical role of microvasculature basal lamina in ischemic brain injury. Prog Neurobiol 2007; 83(3): 140–8.Google Scholar
Walz, B, Zimmermann, C, Bottger, S, Haberl, RL. Prognosis of patients after hemicraniectomy in malignant middle cerebral artery infarction. J Neurol 2002; 249(9): 1183–90.Google Scholar
Neumann-Haefelin, C, Brinker, G, Uhlenkuken, U, Pillekamp, F, Hossmann, KA, Hoehn, M. Prediction of hemorrhagic transformation after thrombolytic therapy of clot embolism: an MRI investigation in rat brain. Stroke 2002; 33(5): 1392–8.Google Scholar
Lansberg, MG, Thijs, VN, O'Brien, MW, et al. Evolution of apparent diffusion coefficient, diffusion-weighted, and T2-weighted signal intensity of acute stroke. AJNR Am J Neuroradiol 2001; 22(4): 637–44.Google Scholar
Badaut, J, Lasbennes, F, Magistretti, PJ, Regli, L. Aquaporins in brain: distribution, physiology, and pathophysiology. J Cereb Blood Flow Metab 2002; 22(4): 367–78.Google Scholar
Rothwell, NJ, Luheshi, GN. Interleukin 1 in the brain: biology, pathology and therapeutic target. Trends in Neurosciences 2000; 23(12): 618–25.Google Scholar
Planas, AM, Gorina, R, Chamorro, A. Signalling pathways mediating inflammatory responses in brain ischaemia. Biochem Soc Trans 2006; 34(6): 1267–70.Google Scholar
Chamorro, A, Dirnagl, U, Urra, X, Planas, AM. Neuroprotection in acute stroke: targeting excitotoxicity, oxidative and nitrosative stress, and inflammation. Lancet Neurol 2016; 15(8): 869–81.Google Scholar
Saver, JL, Smith, EE, Fonarow, GC, et al. The “golden hour” and acute brain ischemia: presenting features and lytic therapy in >30,000 patients arriving within 60 minutes of stroke onset. Stroke 2010; 41(7): 1431–9.Google Scholar
Hossmann, KA. The two pathophysiologies of focal brain ischemia: implications for translational stroke research. J Cereb Blood Flow Metab 2012; 32(7): 1310–16.Google Scholar
Kuhl, DE, Phelps, ME, Kowell, AP, et al. Effects of stroke on local cerebral metabolism and perfusion: mapping by emission computed tomography of 18 FDG and 13 NH 3. Ann Neurol 1980; 8: 4760.Google Scholar
Baron, JC, Frackowiak, RS, Herholz, K, et al. Use of PET methods for measurement of cerebral energy metabolism and hemodynamics in cerebrovascular disease. J Cereb Blood Flow Metab 1989; 9(6): 723–42.Google Scholar
Ackerman, RH, Correia, JA, Alpert, NM, et al. Positron imaging in ischemic stroke disease using compounds labeled with oxygen 15. Initial results of clinicophysiologic correlations. Arch Neurol 1981; 38(9): 537–43.Google Scholar
Baron, JC, Bousser, MG, Comar, D, Soussaline, F, Castaigne, P. Noninvasive tomographic study of cerebral blood flow and oxygen metabolism in vivo. Potentials, limitations, and clinical applications in cerebral ischemic disorders. Eur Neurol 1981; 20(3): 273–84.Google Scholar
Lenzi, GL, Frackowiak, RSJ, Jones, T. Cerebral oxygen metabolism and blood flow in human cerebral ischemic infarction. J Cereb Blood Flow Metab 1982; 2: 321–35.Google Scholar
Powers, WJ, Grubb, RL, Jr., Darriet, D, Raichle, ME. Cerebral blood flow and cerebral metabolic rate of oxygen requirements for cerebral function and viability in humans. J Cereb Blood Flow Metab 1985; 5: 600–8.Google Scholar
Baron, JC, Bousser, MG, Rey, A, Guillard, A, Comar, D, Castaigne, P. Reversal of focal “misery-perfusion syndrome” by extra-intracranial arterial bypass in hemodynamic cerebral ischemia. A case study with 15O positron emission tomography. Stroke 1981; 12(4): 454–9.CrossRefGoogle ScholarPubMed
Heiss, W-D, Graf, R, Löttgen, J, et al. 11C-flumazenil PET predicts extent of cortical infarction in reversible middle cerebral artery occlusion of cats. J Cereb Blood Flow Metab 1997; 17(Suppl 1): S4.Google Scholar
Heiss, WD. Experimental evidence of ischemic thresholds and functional recovery. Stroke 1992; 23(11): 1668–72.Google Scholar
Heiss, WD, Grond, M, Thiel, A, et al. Permanent cortical damage detected by flumazenil positron emission tomography in acute stroke. Stroke 1998; 29(2): 454–61.Google Scholar
Baird, AE, Benfield, A, Schlaug, G, et al. Enlargement of human cerebral ischemic lesion volumes measured by diffusion-weighted magnetic resonance imaging. Ann Neurol 1997; 41(5): 581–9.Google Scholar
Kidwell, CS, Alger, JR, Saver, JL. Beyond mismatch: evolving paradigms in imaging the ischemic penumbra with multimodal magnetic resonance imaging. Stroke 2003; 34(11): 2729–35.Google Scholar
Calamante, F, Gadian, DG, Connelly, A. Quantification of perfusion using bolus tracking magnetic resonance imaging in stroke: assumptions, limitations, and potential implications for clinical use. Stroke 2002; 33(4): 1146–51.Google Scholar
Heiss, WD, Zaro Weber, O. Validation of MRI determination of the penumbra by PET measurements in ischemic stroke. J Nucl Med 2017; 58(2): 187–93.Google Scholar
Heiss, WD, Sobesky, J, Smekal, U, et al. Probability of cortical infarction predicted by flumazenil binding and diffusion-weighted imaging signal intensity: a comparative positron emission tomography/magnetic resonance imaging study in early ischemic stroke. Stroke 2004; 35(8): 1892–8.Google Scholar
Kane, I, Carpenter, T, Chappell, F, et al. Comparison of 10 different magnetic resonance perfusion imaging processing methods in acute ischemic stroke: effect on lesion size, proportion of patients with diffusion/perfusion mismatch, clinical scores, and radiologic outcomes. Stroke 2007; 38(12): 3158–64.Google Scholar
Sobesky, J, Weber, OZ, Lehnhardt, FG, et al. Does the mismatch match the penumbra? Magnetic resonance imaging and positron emission tomography in early ischemic stroke. Stroke 2005; 36(5): 980–5.Google Scholar
Olivot, JM, Albers, GW. Diffusion-perfusion MRI for triaging transient ischemic attack and acute cerebrovascular syndromes. Curr Opin Neurol 2011; 24(1): 44–9.Google Scholar
Heiss, WD, Thiel, A, Grond, M, Graf, R. Which targets are relevant for therapy of acute ischemic stroke? Stroke 1999; 30(7): 1486–9.Google Scholar
Donnan, GA, Baron, JC, Ma, H, Davis, SM. Penumbral selection of patients for trials of acute stroke therapy. Lancet Neurol 2009; 8(3): 261–9.Google Scholar
Marks, MP, Tong, DC, Beaulieu, C, et al. Evaluation of early reperfusion and IV tPA therapy using diffusion- and perfusion-weighted MRI. Neurology 1999; 52(9): 1792–8.Google Scholar
Campbell, BC, Christensen, S, Foster, SJ, et al. Visual assessment of perfusion-diffusion mismatch is inadequate to select patients for thrombolysis. Cerebrovasc Dis 2010; 29(6): 592–6.Google Scholar
Mishra, NK, Albers, GW, Davis, SM, et al. Mismatch-based delayed thrombolysis: a meta-analysis. Stroke 2010; 41(1): e25–33.Google Scholar
Garcia, JH, Liu, KF, Ho, KL. Neuronal necrosis after middle cerebral artery occlusion in Wistar rats progresses at different time intervals in the caudoputamen and the cortex. Stroke 1995; 26: 636–42.Google Scholar
Magistretti, PJ. Coupling synaptic activity to glucose metabolism. In: Frackowiak, RSJ, Magistretti, PJ, Shulman, RG, Altman, JS, Adams, M, eds. Neuroenergetics: Relevance for Functional Brain Imaging. Strasbourg: HFSP Workshop XI; 2001: 133–42.Google Scholar
Heiss, WD, Rosner, G. Functional recovery of cortical neurons as related to degree and duration of ischemia. Ann Neurol 1983; 14(3): 294301.Google Scholar
Hossmann, K-A, Mies, G. Multimodal mapping of the ischemic penumbra in animal models. In: Donnan, G, Baron, JC, Sharp, FR, eds. The Ischaemic Penumbra. New York: Marcel Dekker; 2007: 7792.Google Scholar
Jacobs, AH, Tavitian, B, consortium, IN. Noninvasive molecular imaging of neuroinflammation. J Cereb Blood Flow Metab 2012; 32(7): 1393–415.Google Scholar

References

Adams, HP, Bendixen, BH, Kappelle, LJ, et al. Classification of subtype of acute ischemicstroke. Definitions for use in a multicenter clinical trial. Stroke 1993; 24: 3541.Google Scholar
Ay, H, Benner, T, Arsava, EM, et al. A computerized algorithm for etiologic classification of ischemic stroke: the Causative Classification of Stroke System. Stroke 2007; 38: 2979–84.Google Scholar
Arsava, EM, Ballabio, E, Benner, T, et al. The Causative Classification of Stroke system: an international reliability and optimization study. Neurology 2010; 75: 1277–84.Google Scholar
Adams, RJ, Chimowitz, MI, Alpert, JS, et al. Coronary risk evaluation in patients with transient ischemic attack and ischemic stroke: a scientific statement for healthcare professionals from the Stroke Council and the Council on Clinical Cardiology of the American Heart Association/American Stroke Association. Stroke 2003; 34: 2310–22.Google Scholar
Gongora-Rivera, F, Labreuche, J, Jaramillo, A, et al. Autopsy prevalence of coronary atherosclerosis in patients with fatal stroke. Stroke 2007; 38: 1203–10.Google Scholar
Gorelick, PB, Wong, KS, Bae, H-J, et al. Large artery intracranial occlusive disease. A large worldwide burden but a relatively neglected frontier. Stroke 2008; 39: 2396–9.Google Scholar
Pu, Y, Liu, L, Wang, Y, et al. Geographic and sex difference in the distribution of intracranial atherosclerosis in China. Stroke 2013; 44: 2109–14.Google Scholar
Amarenco, P, Cohen, A, Tzuorio, C, et al. Atherosclerotic disease of the aortic arch and the risk of ischemic stroke. N Engl J Med 1994; 331: 1474–9.Google Scholar
Rothwell, PM. Atherothrombosis and ischaemic stroke. BMJ 2007; 334: 379–80.Google Scholar
Giles, MF, Rothwell, PM. Risk of stroke early after transient ischaemic attack: a systematic review and meta-analysis. Lancet Neurol 2007; 6: 1063–72.Google Scholar
Wu, CM, McLaughlin, K, Lorenzetti, DL, et al. Early risk of stroke after transient ischemic attack. A systematic review and meta-analysis. Arch Intern Med 2007; 167: 2417–22.Google Scholar
Caplan, LR, Wong, KS, Gao, S, et al. Is hypoperfusion an important cause of strokes? If so, how? Cerebrovasc Dis 2006; 21: 145–53.Google Scholar
Ogata, J, Yutani, C, Otsubo, R, et al. Heart and vessel pathology underlying brain infarction in 142 stroke patients. Ann Neurol 2008; 63: 770–81.Google Scholar
Buchwald, F, Norrving, B, Petersson, J. Atrial fibrillation in transient ischemic attack versus ischemic stroke: a Swedish Stroke Register Study. Stroke 2016; 47: 2456–61.Google Scholar
Sposato, LA, Cipriano, LE, Saposnik, G, et al. Diagnosis of atrial fibrillation after ischemic stroke and transient ischemic attack: a systematic review and meta-analysis. Lancet Neurol 2015; 14: 377-87.Google Scholar
Mas, JL, Arquizan, C, Lamy, C, et al. Recurrent cerebrovascular events associated with patent foramen ovale, atrial septal aneurysm, or both. N Engl J Med 2001; 345: 1740–6.Google Scholar
Lamy, C, Giannesini, C, Zuber, M, et al. Clinical and imaging findings in cryptogenetic stroke patients with and without patent foramen ovale: the PFO-ASA Study. Stroke 2002; 33: 706–11.Google Scholar
Handke, M, Harloff, A, Olschewski, M, et al. Patent foramen ovale and cryptogenic stroke in older patients. N Engl J Med 2007; 357: 2262–8.Google Scholar
Ntaios, G, Papavasileiou, V, Sagris, D, et al. Closure of patent foramen ovale versus medical therapy in patients with cryptogenic stroke or transient ischemic attack: updated systematic review and meta-analysis. Stroke 2018; 49: 412–18.Google Scholar
Palacio, S, Hart, RG. Neurologic manifestations of cardiogenic embolism: an update. Neurol Clin 2002; 20: 179–93.Google Scholar
Ferro, JM. Cardioembolic stroke: an update. Lancet Neurol 2003; 2: 177–88.Google Scholar
Kang, DW, Latour, LL, Chalela, JA, et al. Early ischemic lesion recurrence within a week after acute ischemic stroke. Ann Neurol 2003; 54: 6674.Google Scholar
Paciaroni, M, Agnelli, G, Corea, F, et al. Early hemorrhagic transformation of brain infarction: rate, predictive factors, and influence on clinical outcome: results of a prospective multicenter study. Stroke 2008; 39: 2249–56.Google Scholar
Molina, CA, Montaner, J, Abilleira, S, et al. Timing of spontaneous recanalization and risk of hemorrhagic transformation in acute cardioembolic stroke. Stroke 2001; 32: 1079–84.Google Scholar
Minematsu, K, Yamaguchi, T, Omae, T. Spectacular shrinking deficit: rapid recovery from a major hemispheric syndrome by migration of an embolus. Neurology 1992; 42: 1762.Google Scholar
Gass, A, Szabo, K, Behrens, S, et al. A diffusion-weighted MRI study of acute ischemic distal arm paresis. Neurology 2001; 57: 1589–94.Google Scholar
Caplan, LR. Posterior Circulation Disease. Clinical Findings, Diagnosis and Management. Cambridge MA: Blackwell Science; 1996.Google Scholar
Kang, DW, Chalela, JA, Ezzeddine, MA, et al. Association of ischaemic lesion patterns on early diffusion-weighted imaging with TOAST stroke subtypes. Arch Neurol 2003; 60: 1730–4.Google Scholar
Wardlaw, JM, Smith, C, Dichgans, M. Mechanisms of sporadic cerebral small vessel disease: insights from neuroimaging. Lancet Neurol 2013; 12: 483–97.Google Scholar
Jackson, C, Sudlow, C. Are lacunar strokes really different? A systematic review of differences in risk factor profiles beween lacuna and nonlacunar infarcts. Stroke 2005; 36: 891901.Google Scholar
Steinke, W, Ley, SC. Lacunar stroke is the major cause of progressive motor deficits. Stroke 2002; 33: 1510–16.Google Scholar
Thömke, F, Gutmann, L, Stoeter, P, et al. Cerebrovascular brainstem diseases with isolated cranial nerve palsies. Cerebrovasc Dis 2002; 13: 147–55.Google Scholar
Vermeer, S, Longstreth, WT, Jr., Koudstaal, PJ. Silent brain infarcts: a systematic review. Lancet Neurol 2007; 6: 611–19.Google Scholar
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.Google Scholar
Rolfs, A, Fazekas, F, Grittner, U, et al. Acute cerebrovascular disease in the young: the Stroke in Young Fabry Patients Study. Stroke 2013; 44: 340–9.Google Scholar
Thaler, DE, Ruthazer, R, Weimar, C, et al. Recurrent stroke predictors differ in medically treated patients with pathogenic vs. other PFOs. Neurology 2014; 83: 221–6.Google Scholar
Hart, RG, Diener, HC, Coutts, SB, et al. Embolic strokes of undetermined source: the case for a new clinical construct. Lancet Neurol 2014; 13: 429–38.Google Scholar

References

Bill, O, Faouzi, M, Meuli, R, et al. Added value of multimodal computed tomography imaging: analysis of 1994 acute ischaemic strokes. Eur J Neurol 2017; 24: 167–74.Google Scholar
Michel, P, Odier, C, Rutgers, M, et al. The Acute STroke Registry and Analysis of Lausanne (ASTRAL): design and baseline analysis of an ischemic stroke registry including acute multimodal imaging. Stroke 2010; 41: 2491–8.Google Scholar
Na, DG, Kim, EY, Ryoo, JW, et al. CT sign of brain swelling without concomitant parenchymal hypoattenuation: comparison with diffusion- and perfusion-weighted MR imaging. Radiology 2005; 235: 986–92.Google Scholar
Barber, PA, Demchuk, AM, Zhang, J, Buchan, AM. Validity and reliability of a quantitative computed tomography score in predicting outcome of hyperacute stroke before thrombolytic therapy. ASPECTS Study Group. Alberta Stroke Programme Early CT Score. Lancet Lond Engl 2000; 355: 1670–4.Google Scholar
Puetz, V, Dzialowski, I, Hill, MD, Demchuk, AM. The Alberta Stroke Program Early CT Score in clinical practice: what have we learned? Int J Stroke Off J Int Stroke Soc 2009; 4: 354–64.Google Scholar
Hill, MD, Rowley, HA, Adler, F, et al. Selection of acute ischemic stroke patients for intra-arterial thrombolysis with pro-urokinase by using ASPECTS. Stroke 2003; 34: 1925–31.Google Scholar
Demchuk, AM, Hill, MD, Barber, PA, et al. Importance of early ischemic computed tomography changes using ASPECTS in NINDS rtPA Stroke Study. Stroke 2005; 36: 2110–15.Google Scholar
Goyal, M, Menon, BK, van Zwam, WH, et al. Endovascular thrombectomy after large-vessel ischaemic stroke: a meta-analysis of individual patient data from five randomised trials. Lancet Lond Engl 2016; 387: 1723–31.Google Scholar
Strbian, D, Engelter, S, Michel, P, et al. Symptomatic intracranial hemorrhage after stroke thrombolysis: the SEDAN score. Ann Neurol 2012; 71: 634–41.Google Scholar
Nezu, T, Koga, M, Nakagawara, J, et al. Early ischemic change on CT versus diffusion-weighted imaging for patients with stroke receiving intravenous recombinant tissue-type plasminogen activator therapy: stroke acute management with urgent risk-factor assessment and improvement (SAMURAI) rt-PA registry. Stroke 2011; 42: 2196–200.Google Scholar
Latchaw, RE, Yonas, H, Hunter, GJ, et al. Guidelines and recommendations for perfusion imaging in cerebral ischemia: a scientific statement for healthcare professionals by the writing group on perfusion imaging, from the Council on Cardiovascular Radiology of the American Heart Association. Stroke 2003; 34: 1084–104.Google Scholar
Wintermark, M, Reichhart, M, Thiran, J-P, et al. Prognostic accuracy of cerebral blood flow measurement by perfusion computed tomography, at the time of emergency room admission, in acute stroke patients. Ann Neurol 2002; 51: 417–32.Google Scholar
Koenig, M, Kraus, M, Theek, C, et al. Quantitative assessment of the ischemic brain by means of perfusion-related parameters derived from perfusion CT. Stroke 2001; 32: 431–7.Google Scholar
Eastwood, JD, Lev, MH, Azhari, T, et al. CT perfusion scanning with deconvolution analysis: pilot study in patients with acute middle cerebral artery stroke. Radiology 2002; 222: 227–36.Google Scholar
Murphy, BD, Fox, AJ, Lee, DH, et al. White matter thresholds for ischemic penumbra and infarct core in patients with acute stroke: CT perfusion study. Radiology 2008; 247: 818–25.Google Scholar
Wintermark, M, Fischbein, NJ, Smith, WS, et al. Accuracy of dynamic perfusion CT with deconvolution in detecting acute hemispheric stroke. AJNR Am J Neuroradiol 2005; 26: 104–12.Google Scholar
Youn, SW, Kim, JH, Weon, Y-C, et al. Perfusion CT of the brain using 40-mm-wide detector and toggling table technique for initial imaging of acute stroke. AJR Am J Roentgenol 2008; 191: W120–6.Google Scholar
Wintermark, M, Flanders, AE, Velthuis, B, et al. Perfusion-CT assessment of infarct core and penumbra: receiver operating characteristic curve analysis in 130 patients suspected of acute hemispheric stroke. Stroke 2006; 37: 979–85.Google Scholar
Bivard, A, Spratt, N, Levi, C, Parsons, M. Perfusion computer tomography: imaging and clinical validation in acute ischaemic stroke. Brain J Neurol 2011; 134: 3408–16.Google Scholar
Campbell, BCV, Christensen, S, Levi, CR, et al. Cerebral blood flow is the optimal CT perfusion parameter for assessing infarct core. Stroke 2011; 42: 3435–40.Google Scholar
Bivard, A, Kleinig, T, Miteff, F, et al. Ischemic core thresholds change with time to reperfusion: a case control study. Ann Neurol 2017; 82: 9951003.Google Scholar
Dehkharghani, S, Bammer, R, Straka, M, et al. Performance and predictive value of a user-independent platform for CT perfusion analysis: threshold-derived automated systems outperform examiner-driven approaches in outcome prediction of acute ischemic stroke. AJNR Am J Neuroradiol 2015; 36: 1419–25.Google Scholar
Austein, F, Riedel, C, Kerby, T, et al. Comparison of perfusion CT software to predict the final infarct volume after thrombectomy. Stroke 2016; 47: 2311–17.Google Scholar
Demeestere, J, Garcia-Esperon, C, Garcia-Bermejo, P, et al. Evaluation of hyperacute infarct volume using ASPECTS and brain CT perfusion core volume. Neurology 2017; 88: 2248–53.Google Scholar
Meyer, IA, Cereda, CW, Correia, PN, et al. Factors associated with focal computed tomographic perfusion abnormalities in supratentorial transient ischemic attacks. Stroke 2018; 49: 6875.Google Scholar
Gelfand, JM, Wintermark, M, Josephson, SA. Cerebral perfusion-CT patterns following seizure. Eur J Neurol 2010; 17: 594601.Google Scholar
Koome, M, Churilov, L, Chen, Z, et al. Computed tomography perfusion as a diagnostic tool for seizures after ischemic stroke. Neuroradiology 2016; 58: 577–84.Google Scholar
Gonzalez-Delgado, M, Michel, P, Reichhart, M, et al. The significance of focal hypoperfusion during migraine with aura (abstract). Stroke 2005; 36: 444.Google Scholar
Furtado, AD, Smith, WS, Koroshetz, W, et al. Perfusion CT imaging follows clinical severity in left hemispheric strokes. Eur Neurol 2008; 60: 244–52.Google Scholar
Pallesen, L-P, Lambrou, D, Eskandari, A, et al. Perfusion computed tomography in posterior circulation stroke: predictors and prognostic implications of focal hypoperfusion. Eur J Neurol 2018; 25: 725–31.CrossRefGoogle ScholarPubMed
Zhu, G, Michel, P, Aghaebrahim, A, et al. Computed tomography workup of patients suspected of acute ischemic stroke: perfusion computed tomography adds value compared with clinical evaluation, noncontrast computed tomography, and computed tomography angiogram in terms of predicting outcome. Stroke 2013; 44: 1049–55.Google Scholar
Zhu, G, Michel, P, Aghaebrahim, A, et al. Prediction of recanalization trumps prediction of tissue fate: the penumbra: a dual-edged sword. Stroke 2013; 44: 1014–19.Google Scholar
Silvennoinen, HM, Hamberg, LM, Lindsberg, PJ, Valanne, L, Hunter, GJ. CT perfusion identifies increased salvage of tissue in patients receiving intravenous recombinant tissue plasminogen activator within 3 hours of stroke onset. AJNR Am J Neuroradiol 2008; 29: 1118–23.Google Scholar
Psychogios, M-N, Schramm, P, Frölich, AM, et al. Alberta Stroke Program Early CT Scale evaluation of multimodal computed tomography in predicting clinical outcomes of stroke patients treated with aspiration thrombectomy. Stroke 2013; 44: 2188–93.Google Scholar
Lansberg, MG, Christensen, S, Kemp, S, et al. Computed tomographic perfusion to predict response to recanalization in ischemic stroke. Ann Neurol 2017; 81: 849–56.Google Scholar
Borst, J, Berkhemer, OA, Roos, YBWEM, et al. Value of computed tomographic perfusion-based patient selection for intra-arterial acute ischemic stroke treatment. Stroke 2015; 46: 3375–82.Google Scholar
Campbell, BCV, Mitchell, PJ, Kleinig, TJ, et al. Endovascular therapy for ischemic stroke with perfusion-imaging selection. N Engl J Med 2015; 372: 1009–18.Google Scholar
Nogueira, RG, Jadhav, AP, Haussen, DC, et al. Thrombectomy 6 to 24 hours after stroke with a mismatch between deficit and infarct. N Engl J Med 2018; 378: 1121.Google Scholar
Albers, GW, Marks, MP, Kemp, S, et al. Thrombectomy for stroke at 6 to 16 hours with selection by perfusion imaging. N Engl J Med 2018; 378: 708–18.Google Scholar
Tsetsou, S, Amiguet, M, Eskandari, A, et al. Severe cerebral hypovolemia on perfusion CT and lower body weight are associated with parenchymal haemorrhage after thrombolysis. Neuroradiology 2017; 59: 23–9.Google Scholar
Hermitte, L, Cho, T-H, Ozenne, B, et al. Very low cerebral blood volume predicts parenchymal hematoma in acute ischemic stroke. Stroke 2013; 44: 2318–20.Google Scholar
Minnerup, J, Wersching, H, Ringelstein, EB, et al. Prediction of malignant middle cerebral artery infarction using computed tomography-based intracranial volume reserve measurements. Stroke 2011; 42: 3403–9.Google Scholar
Bektas, H, Wu, T-C, Kasam, M, et al. Increased blood–brain barrier permeability on perfusion CT might predict malignant middle cerebral artery infarction. Stroke 2010; 41: 2539–44.Google Scholar
Rotzinger, DC, Mosimann, PJ, Meuli, RA, Maeder, P, Michel, P. Site and rate of occlusive disease in cervicocerebral arteries: a CT angiography study of 2209 patients with acute ischemic stroke. AJNR Am J Neuroradiol 2017; 38: 868–74.Google Scholar
Bash, S, Villablanca, JP, Jahan, R, et al. Intracranial vascular stenosis and occlusive disease: evaluation with CT angiography, MR angiography, and digital subtraction angiography. AJNR Am J Neuroradiol 2005; 26: 1012–21.Google Scholar
Knauth, M, von Kummer, R, Jansen, O, et al. Potential of CT angiography in acute ischemic stroke. AJNR Am J Neuroradiol 1997; 18: 1001–10.Google Scholar
Riedel, CH, Zimmermann, P, Jensen-Kondering, U, et al. The importance of size: successful recanalization by intravenous thrombolysis in acute anterior stroke depends on thrombus length. Stroke 2011; 42: 1775–7.Google Scholar
Riedel, CH, Yoo, AJ. Clot characterization by noncontrast CT to predict IV tPA failure. AJNR Am J Neuroradiol 2012; 33: E63; author reply E64.Google Scholar
Puetz, V, Dzialowski, I, Hill, MD, et al. Intracranial thrombus extent predicts clinical outcome, final infarct size and hemorrhagic transformation in ischemic stroke: the clot burden score. Int J Stroke Off J Int Stroke Soc 2008; 3: 230–6.Google Scholar
Medlin, F, Amiguet, M, Vanacker, P, Michel, P. Influence of arterial occlusion on outcome after intravenous thrombolysis for acute ischemic stroke. Stroke 2015; 46: 126–31.Google Scholar
Smith, WS, Schwab, S. Advances in stroke: critical care and emergency medicine. Stroke 2012; 43: 308–9.Google Scholar
González, RG, Lev, MH, Goldmacher, GV, et al. Improved outcome prediction using CT angiography in addition to standard ischemic stroke assessment: results from the STOPStroke study. PloS One 2012; 7: e30352.Google Scholar
Tan, IYL, Demchuk, AM, Hopyan, J, et al. CT angiography clot burden score and collateral score: correlation with clinical and radiologic outcomes in acute middle cerebral artery infarct. AJNR Am J Neuroradiol 2009; 30: 525–31.Google Scholar
Saarinen, JT, Sillanpää, N, Rusanen, H, et al. The mid-M1 segment of the middle cerebral artery is a cutoff clot location for good outcome in intravenous thrombolysis. Eur J Neurol 2012; 19: 1121–7.Google Scholar
Menon, BK, Smith, EE, Modi, J, et al. Regional leptomeningeal score on CT angiography predicts clinical and imaging outcomes in patients with acute anterior circulation occlusions. AJNR Am J Neuroradiol 2011; 32: 1640–5.Google Scholar
Tan, JC, Dillon, WP, Liu, S, et al. Systematic comparison of perfusion-CT and CT-angiography in acute stroke patients. Ann Neurol 2007; 61: 533–43.Google Scholar
Miteff, F, Levi, CR, Bateman, GA, et al. The independent predictive utility of computed tomography angiographic collateral status in acute ischaemic stroke. Brain J Neurol 2009; 132: 2231–8.Google Scholar
Angermaier, A, Langner, S, Kirsch, M, et al. CT-angiographic collateralization predicts final infarct volume after intra-arterial thrombolysis for acute anterior circulation ischemic stroke. Cerebrovasc Dis Basel Switz 2011; 31: 177–84.Google Scholar
Goyal, M, Demchuk, AM, Menon, BK, et al. Randomized assessment of rapid endovascular treatment of ischemic stroke. N Engl J Med 2015; 372: 1019–30.Google Scholar
Schramm, P, Schellinger, PD, Fiebach, JB, et al. Comparison of CT and CT angiography source images with diffusion-weighted imaging in patients with acute stroke within 6 hours after onset. Stroke 2002; 33: 2426–32.Google Scholar
Lev, MH, Segal, AZ, Farkas, J, et al. Utility of perfusion-weighted CT imaging in acute middle cerebral artery stroke treated with intra-arterial thrombolysis: prediction of final infarct volume and clinical outcome. Stroke 2001; 32: 2021–8.Google Scholar
Puetz, V, Sylaja, PN, Hill, MD, et al. CT angiography source images predict final infarct extent in patients with basilar artery occlusion. AJNR Am J Neuroradiol 2009; 30: 1877–83.Google Scholar
Demchuk, AM, Dowlatshahi, D, Rodriguez-Luna, D, et al. Prediction of haematoma growth and outcome in patients with intracerebral haemorrhage using the CT-angiography spot sign (PREDICT): a prospective observational study. Lancet Neurol 2012; 11: 307–14.CrossRefGoogle ScholarPubMed
Fainardi, E, Borrelli, M, Saletti, A, et al. CT perfusion mapping of hemodynamic disturbances associated to acute spontaneous intracerebral hemorrhage. Neuroradiology 2008; 50: 729–40.Google Scholar
Fiebach, JB, Schellinger, PD, Jansen, O, et al. CT and diffusion-weighted MR imaging in randomized order: diffusion-weighted imaging results in higher accuracy and lower interrater variability in the diagnosis of hyperacute ischemic stroke. Stroke 2002; 33: 2206–10.Google Scholar
Chalela, JA, Kidwell, CS, Nentwich, LM, et al. Magnetic resonance imaging and computed tomography in emergency assessment of patients with suspected acute stroke: a prospective comparison. Lancet Lond Engl 2007; 369: 293–8.Google Scholar
von Kummer, R, Bourquain, H, Bastianello, S, et al. Early prediction of irreversible brain damage after ischemic stroke at CT. Radiology 2001; 219: 95100.Google Scholar
Sedlaczek, O, Hirsch, JG, Grips, E, et al. Detection of delayed focal MR changes in the lateral hippocampus in transient global amnesia. Neurology 2004; 62: 2165–70.Google Scholar
Chatzikonstantinou, A, Gass, A, Förster, A, Hennerici, MG, Szabo, K. Features of acute DWI abnormalities related to status epilepticus. Epilepsy Res 2011; 97: 4551.Google Scholar
Eisele, P, Szabo, K, Griebe, M, et al. Reduced diffusion in a subset of acute MS lesions: a serial multiparametric MRI study. AJNR Am J Neuroradiol 2012; 33: 1369–73.Google Scholar
Ma, L, Gao, P-Y, Lin, Y, et al. Can baseline magnetic resonance angiography (MRA) status become a foremost factor in selecting optimal acute stroke patients for recombinant tissue plasminogen activator (rt-PA) thrombolysis beyond 3 hours? Neurol Res 2009; 31: 355–61.Google Scholar
Chemmanam, T, Campbell, BCV, Christensen, S, et al. Ischemic diffusion lesion reversal is uncommon and rarely alters perfusion-diffusion mismatch. Neurology 2010; 75: 1040–7.Google Scholar
Fiehler, J, Knudsen, K, Kucinski, T, et al. Predictors of apparent diffusion coefficient normalization in stroke patients. Stroke 2004; 35: 514–19.Google Scholar
Yoo, AJ, Barak, ER, Copen, WA, et al. Combining acute diffusion-weighted imaging and mean transmit time lesion volumes with National Institutes of Health Stroke Scale Score improves the prediction of acute stroke outcome. Stroke 2010; 41: 1728–35.Google Scholar
Parsons, MW, Christensen, S, McElduff, P, et al. Pretreatment diffusion- and perfusion-MR lesion volumes have a crucial influence on clinical response to stroke thrombolysis. J Cereb Blood Flow Metab 2010; 30: 1214–25.Google Scholar
Mlynash, M, Lansberg, MG, De Silva, DA, et al. Refining the definition of the malignant profile: insights from the DEFUSE-EPITHET pooled data set. Stroke 2011; 42: 1270–5.Google Scholar
Thomalla, G, Cheng, B, Ebinger, M, et al. DWI-FLAIR mismatch for the identification of patients with acute ischaemic stroke within 4·5 h of symptom onset (PRE-FLAIR): a multicentre observational study. Lancet Neurol 2011; 10: 978–86.Google Scholar
Mackey, J, Kleindorfer, D, Sucharew, H, et al. Population-based study of wake-up strokes. Neurology 2011; 76: 1662–7.Google Scholar
Thomalla, G, Simonsen, CZ, Boutitie, F, et al. MRI-guided thrombolysis for stroke with unknown time of onset. N Engl J Med 2018; 379: 611–22.Google Scholar
Fiebach, JB, Al-Rawi, Y, Wintermark, M, et al. Vascular occlusion enables selecting acute ischemic stroke patients for treatment with desmoteplase. Stroke 2012; 43: 1561–6.Google Scholar
Fiehler, J, Knudsen, K, Thomalla, G, et al. Vascular occlusion sites determine differences in lesion growth from early apparent diffusion coefficient lesion to final infarct. AJNR Am J Neuroradiol 2005; 26: 1056–61.Google Scholar
Schellinger, PD, Chalela, JA, Kang, D-W, Latour, LL, Warach, S. Diagnostic and prognostic value of early MR imaging vessel signs in hyperacute stroke patients imaged <3 hours and treated with recombinant tissue plasminogen activator. AJNR Am J Neuroradiol 2005; 26: 618–24.Google Scholar
Galinovic, I, Ostwaldt, A-C, Soemmer, C, et al. Search for a map and threshold in perfusion MRI to accurately predict tissue fate: a protocol for assessing lesion growth in patients with persistent vessel occlusion. Cerebrovasc Dis Basel Switz 2011; 32: 186–93.Google Scholar
Rovira, A, Orellana, P, Alvarez-Sabín, J, et al. Hyperacute ischemic stroke: middle cerebral artery susceptibility sign at echo-planar gradient-echo MR imaging. Radiology 2004; 232: 466–73.Google Scholar
Thomalla, G, Schwark, C, Sobesky, J, et al. Outcome and symptomatic bleeding complications of intravenous thrombolysis within 6 hours in MRI-selected stroke patients: comparison of a German multicenter study with the pooled data of ATLANTIS, ECASS, and NINDS tPA trials. Stroke 2006; 37: 852–8.Google Scholar
Hacke, W, Albers, G, Al-Rawi, Y, et al. The Desmoteplase in Acute Ischemic Stroke Trial (DIAS): a phase II MRI-based 9-hour window acute stroke thrombolysis trial with intravenous desmoteplase. Stroke 2005; 36: 6673.Google Scholar
Butcher, K, Parsons, M, Allport, L, et al. Rapid assessment of perfusion-diffusion mismatch. Stroke 2008; 39: 7581.Google Scholar
Albers, GW, Thijs, VN, Wechsler, L, et al. Magnetic resonance imaging profiles predict clinical response to early reperfusion: the diffusion and perfusion imaging evaluation for understanding stroke evolution (DEFUSE) study. Ann Neurol 2006; 60: 508–17.Google Scholar
Davis, SM, Donnan, GA, Parsons, MW, et al. Effects of alteplase beyond 3 h after stroke in the Echoplanar Imaging Thrombolytic Evaluation Trial (EPITHET): a placebo-controlled randomised trial. Lancet Neurol 2008; 7: 299309.Google Scholar
Warach, SJ, Luby, M, Albers, GW, et al. Acute stroke imaging research roadmap III imaging selection and outcomes in acute stroke reperfusion clinical trials: consensus recommendations and further research priorities. Stroke 2016; 47: 1389–98.Google Scholar
Nagakane, Y, Christensen, S, Brekenfeld, C, et al. EPITHET: positive result after reanalysis using baseline diffusion-weighted imaging/perfusion-weighted imaging co-registration. Stroke 2011; 42: 5964.Google Scholar
Lansberg, MG, Straka, M, Kemp, S, et al. MRI profile and response to endovascular reperfusion after stroke (DEFUSE 2): a prospective cohort study. Lancet Neurol 2012; 11: 860–7.Google Scholar
Kidwell, CS, Alger, JR, Saver, JL. Beyond mismatch: evolving paradigms in imaging the ischemic penumbra with multimodal magnetic resonance imaging. Stroke 2003; 34: 2729–35.Google Scholar
Kucinski, T, Naumann, D, Knab, R, et al. Tissue at risk is overestimated in perfusion-weighted imaging: MR imaging in acute stroke patients without vessel recanalization. AJNR Am J Neuroradiol 2005; 26: 815–19.Google Scholar
Zaharchuk, G, El Mogy, IS, Fischbein, NJ, Albers, GW. Comparison of arterial spin labeling and bolus perfusion-weighted imaging for detecting mismatch in acute stroke. Stroke 2012; 43: 1843–8.Google Scholar
Bokkers, RPH, Hernandez, DA, Merino, JG, et al. Whole-brain arterial spin labeling perfusion MRI in patients with acute stroke. Stroke 2012; 43: 1290–4.Google Scholar
Niibo, T, Ohta, H, Yonenaga, K, et al. Arterial spin-labeled perfusion imaging to predict mismatch in acute ischemic stroke. Stroke 2013; 44: 2601–3.Google Scholar
Kidwell, CS, Chalela, JA, Saver, JL, et al. Comparison of MRI and CT for detection of acute intracerebral hemorrhage. JAMA 2004; 292: 1823–30.Google Scholar
Fiehler, J. Cerebral microbleeds: old leaks and new haemorrhages. Int J Stroke 2006; 1: 122–30.Google Scholar
Fiehler, J, Albers, GW, Boulanger, J-M, et al. Bleeding risk analysis in stroke imaging before thrombolysis (BRASIL): pooled analysis of T2*-weighted magnetic resonance imaging data from 570 patients. Stroke 2007; 38: 2738–44.Google Scholar
Tanikawa, M, Mase, M, Yamada, K, et al. Surgical treatment of chronic subdural hematoma based on intrahematomal membrane structure on MRI. Acta Neurochir (Wien) 2001; 143: 613–18; discussion 618–19.Google Scholar
Wiesmann, M, Mayer, TE, Yousry, I, et al. Detection of hyperacute subarachnoid hemorrhage of the brain by using magnetic resonance imaging. J Neurosurg 2002; 96: 684–9.Google Scholar
Baird, AE, Benfield, A, Schlaug, G, et al. Enlargement of human cerebral ischemic lesion volumes measured by diffusion-weighted magnetic resonance imaging. Ann Neurol 1997; 41: 581–9.Google Scholar
Schaefer, PW, Barak, ER, Kamalian, S, et al. Quantitative assessment of core/penumbra mismatch in acute stroke: CT and MR perfusion imaging are strongly correlated when sufficient brain volume is imaged. Stroke 2008; 39: 2986–92.Google Scholar
Wintermark, M, Reichhart, M, Cuisenaire, O, et al. Comparison of admission perfusion computed tomography and qualitative diffusion- and perfusion-weighted magnetic resonance imaging in acute stroke patients. Stroke 2002; 33: 2025–31.Google Scholar
Wintermark, M, Meuli, R, Browaeys, P, et al. Comparison of CT perfusion and angiography and MRI in selecting stroke patients for acute treatment. Neurology 2007; 68: 694–7.Google Scholar
Dani, KA, Thomas, RGR, Chappell, FM, et al. Computed tomography and magnetic resonance perfusion imaging in ischemic stroke: definitions and thresholds. Ann Neurol 2011; 70: 384401.Google Scholar
Wintermark, M, Maeder, P, Thiran, JP, Schnyder, P, Meuli, R. Quantitative assessment of regional cerebral blood flows by perfusion CT studies at low injection rates: a critical review of the underlying theoretical models. Eur Radiol 2001; 11: 1220–30.Google Scholar
Liu, Y, Karonen, JO, Vanninen, RL, et al. Acute ischemic stroke: predictive value of 2D phase-contrast MR angiography – serial study with combined diffusion and perfusion MR imaging. Radiology 2004; 231: 517–27.Google Scholar
Schievink, WI. Spontaneous dissection of the carotid and vertebral arteries. N Engl J Med 2001; 344: 898906.Google Scholar
Lev, MH, Koroshetz, WJ, Schwamm, LH, Gonzalez, RG. CT or MRI for imaging patients with acute stroke: visualization of “tissue at risk”? Stroke 2002; 33: 2736–7.Google Scholar
Mnyusiwalla, A, Aviv, RI, Symons, SP. Radiation dose from multidetector row CT imaging for acute stroke. Neuroradiology 2009; 51: 635–40.Google Scholar
Smith-Bindman, R, Lipson, J, Marcus, R, et al. Radiation dose associated with common computed tomography examinations and the associated lifetime attributable risk of cancer. Arch Intern Med 2009; 169: 2078–86.Google Scholar
Aulicky, P, Mikulík, R, Goldemund, D, et al. Safety of performing CT angiography in stroke patients treated with intravenous thrombolysis. J Neurol Neurosurg Psychiatry 2010; 81: 783–7.Google Scholar
Thomsen, HS, European Society of Urogenital Radiology (ESUR). ESUR guideline: gadolinium-based contrast media and nephrogenic systemic fibrosis. Eur Radiol 2007; 17: 2692–6.Google Scholar
Mortimer, AM, Simpson, E, Bradley, MD, Renowden, SA. Computed tomography angiography in hyperacute ischemic stroke: prognostic implications and role in decision-making. Stroke 2013; 44: 1480–8.Google Scholar
Kidwell, CS, Jahan, R, Gornbein, J, et al. A trial of imaging selection and endovascular treatment for ischemic stroke. N Engl J Med 2013; 368: 914–23.Google Scholar
Ogata, T, Christensen, S, Nagakane, Y, et al. The effects of alteplase 3 to 6 hours after stroke in the EPITHET-DEFUSE combined dataset: post hoc case-control study. Stroke 2013; 44: 8793.Google Scholar
Sztriha, LK, Manawadu, D, Jarosz, J, Keep, J, Kalra, L. Safety and clinical outcome of thrombolysis in ischaemic stroke using a perfusion CT mismatch between 3 and 6 hours. PloS One 2011; 6: e25796.Google Scholar
Obach, V, Oleaga, L, Urra, X, et al. Multimodal CT-assisted thrombolysis in patients with acute stroke: a cohort study. Stroke 2011; 42: 1129–31.Google Scholar
Hassan, AE, Zacharatos, H, Rodriguez, GJ, et al. A comparison of computed tomography perfusion-guided and time-guided endovascular treatments for patients with acute ischemic stroke. Stroke 2010; 41: 1673–8.Google Scholar
Warach, S, Al-Rawi, Y, Furlan, AJ, et al. Refinement of the magnetic resonance diffusion-perfusion mismatch concept for thrombolytic patient selection: insights from the desmoteplase in acute stroke trials. Stroke 2012; 43: 2313–18.Google Scholar
Saver, JL, Goyal, M, van der Lugt, A, et al. Time to treatment with endovascular thrombectomy and outcomes from ischemic stroke: a meta-analysis. JAMA 2016; 316: 1279–88.Google Scholar

References

WHO. Shaping the Future. Geneva: World Health Organization; 2003.Google Scholar
Feigin, VL, Krishnamurthi, RV, Parmar, P, et al. Update on the global burden of ischemic and hemorrhagic stroke in 1990–2013: the GBD 2013 Study. Neuroepidemiology 2015; 45(3): 161–76.Google Scholar
Miller, EL, Murray, L, Richards, L, et al. Comprehensive overview of nursing and interdisciplinary rehabilitation care of the stroke patient: a scientific statement from the American Heart Association. Stroke 2010; 41(10): 2402–48.Google Scholar
Gordon, NF, Gulanick, M, Costa, F, et al. Physical activity and exercise recommendations for stroke survivors: an American Heart Association scientific statement from the Council on Clinical Cardiology, Subcommittee on Exercise, Cardiac Rehabilitation, and Prevention; the Council on Cardiovascular Nursing; the Council on Nutrition, Physical Activity, and Metabolism; and the Stroke Council. Stroke 2004; 35(5): 1230–40.Google Scholar
Kwakkel, G, Veerbeek, JM, Harmeling-van der Wel, BC, van Wegen, E, Kollen, BJ. Early prediction of functional outcome after stroke I. Diagnostic accuracy of the Barthel Index for measuring activities of daily living outcome after ischemic hemispheric stroke: does early poststroke timing of assessment matter? Stroke 2011; 42(2): 342–6.Google Scholar
Veerbeek, JM, Kwakkel, G, van Wegen, EE, Ket, JC, Heymans, MW. Early prediction of outcome of activities of daily living after stroke: a systematic review. Stroke 2011; 42(5): 1482–8.Google Scholar
Weimar, C, Konig, IR, Kraywinkel, K, Ziegler, A, Diener, HC. German Stroke Study C. Age and National Institutes of Health Stroke Scale Score within 6 hours after onset are accurate predictors of outcome after cerebral ischemia: development and external validation of prognostic models. Stroke 2004; 35(1): 158–62.Google Scholar
Kwakkel, G, Veerbeek, JM, van Wegen, EE, et al. Predictive value of the NIHSS for ADL outcome after ischemic hemispheric stroke: does timing of early assessment matter? J Neurol Sci 2010; 294(1–2): 5761.Google Scholar
Konig, IR, Ziegler, A, Bluhmki, E, et al. Predicting long-term outcome after acute ischemic stroke: a simple index works in patients from controlled clinical trials. Stroke 2008; 39(6): 1821–6.Google Scholar
Granger, CV, Dewis, LS, Peters, NC, Sherwood, CC, Barrett, JE. Stroke rehabilitation: analysis of repeated Barthel Index measures. Arch Phys Med Rehabil 1979; 60(1): 1417.Google Scholar
Alexander, MP. Stroke rehabilitation outcome. A potential use of predictive variables to establish levels of care. Stroke 1994; 25(1): 128–34.Google Scholar
Muscari, A, Puddu, GM, Santoro, N, Zoli, M. A simple scoring system for outcome prediction of ischemic stroke. Acta Neurol Scand 2011; 124(5): 334–42.Google Scholar
Reid, JM, Gubitz, GJ, Dai, D, et al. Predicting functional outcome after stroke by modelling baseline clinical and CT variables. Age Ageing 2010; 39(3): 360–6.Google Scholar
Tilling, K, Sterne, JA, Rudd, AG, et al. A new method for predicting recovery after stroke. Stroke 2001; 32(12): 2867–73.Google Scholar
Ntaios, G, Faouzi, M, Ferrari, J, et al. An integer-based score to predict functional outcome in acute ischemic stroke: the ASTRAL score. Neurology 2012; 78(24): 1916–22.Google Scholar
Johnston, KC, Connors, AF, Jr., Wagner, DP, et al. A predictive risk model for outcomes of ischemic stroke. Stroke 2000; 31(2): 448–55.Google Scholar
Ntaios, G, Gioulekas, F, Papavasileiou, V, Strbian, D, Michel, P. ASTRAL, DRAGON and SEDAN scores predict stroke outcome more accurately than physicians. Eur J Neurol 2016; 23(11): 1651–7.Google Scholar
Cao, KG, Fu, CH, Li, HQ, Xin, XY, Gao, Y. A new prognostic scale for the early prediction of ischemic stroke recovery mainly based on traditional Chinese medicine symptoms and NIHSS score: a retrospective cohort study. BMC Complement Altern Med 2015; 15: 407.Google Scholar
Haselbach, D, Renggli, A, Carda, S, Croquelois, A. Determinants of neurological functional recovery potential after stroke in young adults. Cerebrovasc Dis Extra 2014; 4(1): 7783.Google Scholar
Lamola, G, Bertolucci, F, Rossi, B, Chisari, C. Clinical assessments for predicting functional recovery after stroke. Int J Neurorehab 2015; 2(174); doi: 10.4172/2376-0281.1000174.Google Scholar
Kwah, LK, Herbert, RD. Prediction of walking and arm recovery after stroke: a critical review Brain Sci 2016; 6(4): 53.Google Scholar
Glize, B, Villain, M, Richert, L, et al. Language features in the acute phase of post-stroke severe aphasia could predict the outcome. Eur J Phys Rehabil Med 2016; 53(2): 249–55.Google Scholar
Lee, SY, Lim, JY, Kang, EK, et al. Prediction of good functional recovery after stroke based on combined motor and somatosensory evoked potential findings. J Rehabil Med 2010; 42(1): 1620.Google Scholar
Arsava, EM, Helenius, J, Avery, R, et al. Assessment of the predictive validity of etiologic stroke classification. JAMA Neurology 2017; 74(4): 419–26.Google Scholar
Vogt, G, Laage, R, Shuaib, A, Schneider, A, Collaboration, V. Initial lesion volume is an independent predictor of clinical stroke outcome at day 90: an analysis of the Virtual International Stroke Trials Archive (VISTA) database. Stroke 2012; 43(5): 1266–72.Google Scholar
Borsody, M, Warner Gargano, J, Reeves, M, Jacobs, B. Group MI-SS. Infarction involving the insula and risk of mortality after stroke. Cerebrovasc Dis Basel Switz 2009; 27(6): 564–71.Google Scholar
Ois, A, Cuadrado-Godia, E, Solano, A, Perich-Alsina, X, Roquer, J. Acute ischemic stroke in anterior choroidal artery territory. J Neurol Sci 2009; 281(1–2): 80–4.Google Scholar
Schiemanck, SK, Kwakkel, G, Post, MW, Kappelle, LJ, Prevo, AJ. Impact of internal capsule lesions on outcome of motor hand function at one year post-stroke. J Rehabil Med 2008; 40(2): 96101.Google Scholar
Battey, TW, Karki, M, Singhal, AB, et al. Brain edema predicts outcome after nonlacunar ischemic stroke. Stroke 2014; 45(12): 3643–8.Google Scholar
Barber, PA, Demchuk, AM, Zhang, J, Buchan, AM, Group, AS. Validity and reliability of a quantitative computed tomography score in predicting outcome of hyperacute stroke before thrombolytic therapy. Lancet 2000; 355(9216): 1670–4.Google Scholar
Hill, MD, Rowley, HA, Adler, F, et al. Selection of acute ischemic stroke patients for intra-arterial thrombolysis with pro-urokinase by using ASPECTS. Stroke 2003; 34(8): 1925–31.Google Scholar
Alexander, LD, Pettersen, JA, Hopyan, JJ, Sahlas, DJ, Black, SE. Long-term prediction of functional outcome after stroke using the Alberta stroke program early computed tomography score in the subacute stage. J Stroke Cerebrovasc Dis 2012; 21(8): 737–44.Google Scholar
Branco, JP, Costa, JS, Sargento-Freitas, J, et al. Neuroimaging and blood biomarkers in functional prognosis after stroke. Acta Med Port 2016; 29(11): 749–54.Google Scholar
Strbian, D, Meretoja, A, Ahlhelm, FJ, et al. Predicting outcome of IV thrombolysis-treated ischemic stroke patients: the DRAGON score. Neurology 2012; 78(6): 427–32.Google Scholar
Schiemanck, SK, Kwakkel, G, Post, MW, Prevo, AJ. Predictive value of ischemic lesion volume assessed with magnetic resonance imaging for neurological deficits and functional outcome poststroke: a critical review of the literature. Neurorehabil Neural Repair 2006; 20(4): 492502.Google Scholar
Lindenberg, R, Renga, V, Zhu, LL, et al. Structural integrity of corticospinal motor fibers predicts motor impairment in chronic stroke. Neurology 2010; 74(4): 280–7.Google Scholar
Kissela, B, Lindsell, CJ, Kleindorfer, D, et al. Clinical prediction of functional outcome after ischemic stroke: the surprising importance of periventricular white matter disease and race. Stroke 2009; 40(2): 530–6.Google Scholar
Kidwell, CS, Chalela, JA, Saver, JL, et al. Comparison of MRI and CT for detection of acute intracerebral hemorrhage. JAMA 2004; 292(15): 1823–30.Google Scholar
Strbian, D, Sairanen, T, Meretoja, A, et al. Patient outcomes from symptomatic intracerebral hemorrhage after stroke thrombolysis. Neurology 2011; 77(4): 341–8.Google Scholar
Park, JH, Ko, Y, Kim, WJ, et al. Is asymptomatic hemorrhagic transformation really innocuous? Neurology 2012; 78(6): 421–6.Google Scholar
Thijs, VN, Lansberg, MG, Beaulieu, C, et al. Is early ischemic lesion volume on diffusion-weighted imaging an independent predictor of stroke outcome? A multivariable analysis. Stroke 2000; 31(11): 2597–602.Google Scholar
Johnston, KC, Wagner, DP, Wang, XQ, et al. Validation of an acute ischemic stroke model: does diffusion-weighted imaging lesion volume offer a clinically significant improvement in prediction of outcome? Stroke 2007; 38(6): 1820–5.Google Scholar
Barrett, KM, Ding, YH, Wagner, DP, et al. Change in diffusion-weighted imaging infarct volume predicts neurologic outcome at 90 days: results of the Acute Stroke Accurate Prediction (ASAP) trial serial imaging substudy. Stroke 2009; 40(7): 2422–7.Google Scholar
Jang, SH. Prediction of motor outcome for hemiparetic stroke patients using diffusion tensor imaging: a review. NeuroRehabilitation 2010; 27(4): 367–72.Google Scholar
Bigourdan, A, Munsch, F, Coupe, P, et al. Early fiber number ratio is a surrogate of corticospinal tract integrity and predicts motor recovery after stroke. Stroke 2016; 47(4): 1053–9.Google Scholar
Kim, KH, Kim, YH, Kim, MS, et al. Prediction of motor recovery using diffusion tensor tractography in supratentorial stroke patients with severe motor involvement. Ann Rehabil Med 2015; 39(4): 570–6.Google Scholar
Cho, SH, Kim, DG, Kim, DS, et al. Motor outcome according to the integrity of the corticospinal tract determined by diffusion tensor tractography in the early stage of corona radiata infarct. Neurosci Lett 2007; 426(2): 123–7.Google Scholar
Radlinska, B, Ghinani, S, Leppert, IR, et al. Diffusion tensor imaging, permanent pyramidal tract damage, and outcome in subcortical stroke. Neurology 2010; 75(12): 1048–54.Google Scholar
Liang, Z, Zeng, J, Zhang, C, et al. Longitudinal investigations on the anterograde and retrograde degeneration in the pyramidal tract following pontine infarction with diffusion tensor imaging. Cerebrovasc Dis Basel Switz 2008; 25(3): 209–16.Google Scholar
Puig, J, Pedraza, S, Blasco, G, et al. Acute damage to the posterior limb of the internal capsule on diffusion tensor tractography as an early imaging predictor of motor outcome after stroke. AJNR Am J Neuroradiol 2011; 32(5): 857–63.Google Scholar
Kumar, P, Kathuria, P, Nair, P, Prasad, K. Prediction of upper limb motor recovery after subacute ischemic stroke using diffusion tensor imaging: a systematic review and meta-analysis. J Stroke 2016; 18(1): 50–9.Google Scholar
Lindenberg, R, Zhu, LL, Ruber, T, Schlaug, G. Predicting functional motor potential in chronic stroke patients using diffusion tensor imaging. Hum Brain Mapp 2012; 33(5): 1040–51.Google Scholar
Riley, JD, Le, V, Der-Yeghiaian, L, et al. Anatomy of stroke injury predicts gains from therapy. Stroke 2011; 42(2): 421–6.Google Scholar
Kim, SH, Lee, DG, You, H, et al. The clinical application of the arcuate fasciculus for stroke patients with aphasia: a diffusion tensor tractography study. NeuroRehabilitation 2011; 29(3): 305–10.Google Scholar
Breier, JI, Hasan, KM, Zhang, W, Men, D, Papanicolaou, AC. Language dysfunction after stroke and damage to white matter tracts evaluated using diffusion tensor imaging. AJNR Am J Neuroradiol 2008; 29(3): 483–7.Google Scholar
Kim, SH, Jang, SH. Prediction of aphasia outcome using diffusion tensor tractography for arcuate fasciculus in stroke. AJNR Am J Neuroradiol 2013; 34(4): 785–90.Google Scholar
Stinear, CM, Ward, NS. How useful is imaging in predicting outcomes in stroke rehabilitation? Int J Stroke 2013; 8(1): 33–7.Google Scholar
Kim, B, Winstein, C. Can neurological biomarkers of brain impairment be used to predict poststroke motor recovery? A systematic review. Neurorehabil Neural Repair 2017; 31(1): 324.Google Scholar
Fischer, U, Arnold, M, Nedeltchev, K, et al. NIHSS score and arteriographic findings in acute ischemic stroke. Stroke 2005; 36(10): 2121–5.Google Scholar
Smith, WS, Lev, MH, English, JD, et al. Significance of large vessel intracranial occlusion causing acute ischemic stroke and TIA. Stroke 2009; 40(12): 3834–40.Google Scholar
Gonzalez, RG, Lev, MH, Goldmacher, GV, et al. Improved outcome prediction using CT angiography in addition to standard ischemic stroke assessment: results from the STOP Stroke study. PLoS One 2012; 7(1): e30352.Google Scholar
Mortimer, AM, Simpson, E, Bradley, MD, Renowden, SA. Computed tomography angiography in hyperacute ischemic stroke: prognostic implications and role in decision-making. Stroke 2013; 44(5): 1480–8.Google Scholar
Kucinski, T, Koch, C, Eckert, B, et al. Collateral circulation is an independent radiological predictor of outcome after thrombolysis in acute ischaemic stroke. Neuroradiology 2003; 45(1): 1118.Google Scholar
Bang, OY, Saver, JL, Kim, SJ, et al. Collateral flow predicts response to endovascular therapy for acute ischemic stroke. Stroke 2011; 42(3): 693–9.Google Scholar
Maas, MB, Lev, MH, Ay, H, et al. Collateral vessels on CT angiography predict outcome in acute ischemic stroke. Stroke 2009; 40(9): 3001–5.Google Scholar
Lima, FO, Furie, KL, Silva, GS, et al. The pattern of leptomeningeal collaterals on CT angiography is a strong predictor of long-term functional outcome in stroke patients with large vessel intracranial occlusion. Stroke 2010; 41(10): 2316–22.Google Scholar
Tan, BY, Kong, WY, Ngiam, JN, et al. The role of topographic collaterals in predicting functional outcome after thrombolysis in anterior circulation ischemic stroke. J Neuroimaging 2017; 27(2): 217–20.Google Scholar
Kane, I, Carpenter, T, Chappell, F, et al. Comparison of 10 different magnetic resonance perfusion imaging processing methods in acute ischemic stroke: effect on lesion size, proportion of patients with diffusion/perfusion mismatch, clinical scores, and radiologic outcomes. Stroke 2007; 38(12): 3158–64.Google Scholar
Bivard, A, Spratt, N, Levi, C, Parsons, M. Perfusion computer tomography: imaging and clinical validation in acute ischaemic stroke. Brain 2011; 134(11): 3408–16.Google Scholar
Heiss, WD, Zeiler, K, Havelec, L. Hirndurchblutung und soziale Prognose nach ischämischem zerebralem Insult. DtschMedWschr 1978; 103(70): 597602.Google Scholar
Giubilei, F, Lenzi, GL, Di Piero, V, et al. Predictive value of brain perfusion single-photon emission computed tomography in acute ischemic stroke. Stroke 1990; 21(6): 895900.Google Scholar
Parsons, MW, Pepper, EM, Chan, V, et al. Perfusion computed tomography: prediction of final infarct extent and stroke outcome. Ann Neurol 2005; 58(5): 672–9.Google Scholar
Psychogios, MN, Schramm, P, Frolich, AM, et al. Alberta Stroke Program Early CT Scale evaluation of multimodal computed tomography in predicting clinical outcomes of stroke patients treated with aspiration thrombectomy. Stroke 2013; 44(8): 2188–93.Google Scholar
Payabvash, S, Souza, LC, Kamalian, S, et al. Location-weighted CTP analysis predicts early motor improvement in stroke: a preliminary study. Neurology 2012; 78(23): 1853–9.Google Scholar
Payabvash, S, Kamalian, S, Fung, S, et al. Predicting language improvement in acute stroke patients presenting with aphasia: a multivariate logistic model using location-weighted atlas-based analysis of admission CT perfusion scans. AJNR Am J Neuroradiol 2010; 31(9): 1661–8.Google Scholar
Yoo, AJ, Barak, ER, Copen, WA, et al. Combining acute diffusion-weighted imaging and mean transmit time lesion volumes with National Institutes of Health Stroke Scale Score improves the prediction of acute stroke outcome. Stroke 2010; 41(8): 1728–35.Google Scholar
Schaefer, PW, Pulli, B, Copen, WA, et al. Combining MRI with NIHSS thresholds to predict outcome in acute ischemic stroke: value for patient selection. AJNR Am J Neuroradiol 2015; 36(2): 259–64.Google Scholar
Padroni, M, Bernardoni, A, Tamborino, C, et al. Cerebral blood volume ASPECTS is the best predictor of clinical outcome in acute ischemic stroke: a retrospective, combined semi-quantitative and quantitative assessment. PLoS One 2016; 11(1): e0147910.Google Scholar
Tsogkas, I, Knauth, M, Schregel, K, et al. Added value of CT perfusion compared to CT angiography in predicting clinical outcomes of stroke patients treated with mechanical thrombectomy. Eur Radiol 2016; 26(11): 4213–19.Google Scholar
Heiss, W-D, Weber, OZ. Uncertainties in the assessment of cortical flow by perfusion-weighted MRI in acute stroke. Cerebrovascular Diseases 2011; 32(2): 194–5.Google Scholar
Wahlgren, N, Ahmed, N, Eriksson, N, et al. Multivariable analysis of outcome predictors and adjustment of main outcome results to baseline data profile in randomized controlled trials: Safe Implementation of Thrombolysis in Stroke-MOnitoring STudy (SITS-MOST). Stroke 2008; 39(12): 3316–22.Google Scholar
Strbian, D, Seiffge, DJ, Breuer, L, et al. Validation of the DRAGON score in 12 stroke centers in anterior and posterior circulation. Stroke 2013; 44(10): 2718–21.Google Scholar
Turc, G, Apoil, M, Naggara, O, et al. Magnetic resonance imaging-DRAGON score: 3-month outcome prediction after intravenous thrombolysis for anterior circulation stroke. Stroke 2013; 44(5): 1323–8.Google Scholar
Vora, NA, Shook, SJ, Schumacher, HC, et al. A 5-item scale to predict stroke outcome after cortical middle cerebral artery territory infarction: validation from results of the Diffusion and Perfusion Imaging Evaluation for Understanding Stroke Evolution (DEFUSE) Study. Stroke 2011; 42(3): 645–9.Google Scholar
Davis, SM, Donnan, GA, Parsons, MW, et al. Effects of alteplase beyond 3 h after stroke in the Echoplanar Imaging Thrombolytic Evaluation Trial (EPITHET): a placebo-controlled randomised trial. Lancet Neurol 2008; 7(4): 299309.Google Scholar
Albers, GW, Thijs, VN, Wechsler, L, et al. Magnetic resonance imaging profiles predict clinical response to early reperfusion: the Diffusion and Perfusion Imaging Evaluation for Understanding Stroke Evolution (DEFUSE) study. Ann Neurol 2006; 60(5): 508–17.Google Scholar
Lansberg, MG, Straka, M, Kemp, S, et al. MRI profile and response to endovascular reperfusion after stroke (DEFUSE 2): a prospective cohort study. Lancet Neurol 2012; 11(10): 860–7.Google Scholar
Parsons, M, Spratt, N, Bivard, A, et al. A randomized trial of tenecteplase versus alteplase for acute ischemic stroke. N Engl J Med 2012; 366(12): 1099–107.Google Scholar
Kidwell, CS, Jahan, R, Gornbein, J, et al. A trial of imaging selection and endovascular treatment for ischemic stroke. N Engl J Med 2013; 368(10): 914–23.Google Scholar
Sarraj, A, Albright, K, Barreto, AD, et al. Optimizing prediction scores for poor outcome after intra-arterial therapy in anterior circulation acute ischemic stroke. Stroke 2013; 44(12): 3324–30.Google Scholar
Rijntjes, M, Weiller, C. Recovery of motor and language abilities after stroke: the contribution of functional imaging. Progress in Neurobiology 2002; 66(2): 109–22.Google Scholar
Thirumala, P, Hier, DB, Patel, P. Motor recovery after stroke: lessons from functional brain imaging. Neurol Res 2002; 24(5): 453–8.Google Scholar
Herholz, K, Heiss, WD. Functional imaging correlates of recovery after stroke in humans. J Cereb Blood Flow Metab 2000; 20(12): 1619–31.Google Scholar
Ward, NS. Future perspectives in functional neuroimaging in stroke recovery. Europa Medicophysica 2007; 43(2): 285–94.Google Scholar
Cramer, SC. Repairing the human brain after stroke: I. Mechanisms of spontaneous recovery. Ann Neurol 2008; 63(3): 272–87.Google Scholar
Rossini, PM, Calautti, C, Pauri, F, Baron, JC. Post-stroke plastic reorganisation in the adult brain. Lancet Neurol 2003; 2(8): 493502.Google Scholar
Eliassen, JC, Boespflug, EL, Lamy, M, et al. Brain-mapping techniques for evaluating poststroke recovery and rehabilitation: a review. Top Stroke Rehabil 2008; 15(5): 427–50.Google Scholar
Sokoloff, L, Reivich, M, Kennedy, C, et al. The (14 C)-deoxyglucose method for the measurement of local cerebral glucose utilization: theory, procedure, and normal values in the conscious and anesthetized albino rat. J Neurochem 1977; 28(5): 897916.Google Scholar
Reivich, M, Kuhl, D, Wolf, A, et al. The [18 F]fluorodeoxyglucose method for the measurement of local cerebral glucose utilization in man. Circulation Research 1979; 44(1): 127–37.Google Scholar
Wienhard, K, Pawlik, G, Herholz, K, Wagner, R, Heiss, WD. Estimation of local cerebral glucose utilization by positron emission tomography of [18F]2-fluoro-2-deoxy-D-glucose: a critical appraisal of optimization procedures. J Cereb Blood Flow Metab 1985; 5(1): 115–25.Google Scholar
Heiss, WD, Emunds, HG, Herholz, K. Cerebral glucose metabolism as a predictor of rehabilitation after ischemic stroke. Stroke 1993; 24(12): 1784–8.Google Scholar
Herscovitch, P, Martin, WRW, Raichle, ME. The autoradiographic measurement of regional cerebral blood flow (CBF) with positron emission tomography: validation studies. J Nucl Med 1983; 24(9): P62–3.Google Scholar
Frackowiak, RSJ, Lenzi, GL, Jones, T, Heather, JD. Quantitative measurement of regional cerebral blood flow and oxygen metabolism in man using 15 O and positron emission tomography: theory, procedure, and normal values. J Comput Assist Tomogr 1980; 4(6): 727–36.Google Scholar
Mintun, MA, Raichle, ME, Martin, WRW, Herscovitch, P. Brain oxygen utilization measured with O-15 radiotracers and positron emission tomography. J Nucl Med 1984; 25(2): 177–87.Google Scholar
Mintun, MA, Lundstrom, BN, Snyder, AZ, et al. Blood flow and oxygen delivery to human brain during functional activity: theoretical modeling and experimental data. Proceedings of the National Academy of Sciences of the United States of America 2001; 98(12): 6859–64.Google Scholar
Ogawa, S, Lee, TM, Kay, AR, Tank, DW. Brain magnetic resonance imaging with contrast dependent on blood oxygenation. Proceedings of the National Academy of Sciences of the United States of America 1990; 87(24): 9868–72.Google Scholar
Turner, R, Howseman, A, Rees, G, Josephs, O. Functional imaging with magnetic resonance. In: Frackowiak, RSJ, Friston, KJ, Frith, CD, Dolan, RJ, Mazziotta, JC, eds. Human Brain Function. San Diego: Academic Press; 1997: 467–86.Google Scholar
Stinear, CM, Byblow, WD. Predicting and accelerating motor recovery after stroke. Curr Opin Neurol 2014; 27(6): 624–30.Google Scholar
Stinear, CM, Barber, PA, Petoe, M, Anwar, S, Byblow, WD. The PREP algorithm predicts potential for upper limb recovery after stroke. Brain 2012; 135(8): 2527–35.Google Scholar
Weiller, C. Recovery from motor stroke: human positron emission tomography studies. Cerebrovasc Dis 1995; 5: 282–91.Google Scholar
Heiss, WD, Thiel, A, Winhuisen, L, et al. Functional imaging in the assessment of capability for recovery after stroke. J Rehab Med 2003; 41: 2733.Google Scholar
Horn, U, Grothe, M, Lotze, M. MRI biomarkers for hand-motor outcome prediction and therapy monitoring following stroke. Neural Plasticity 2016.Google Scholar
Rizzolatti, G, Luppino, G, Matelli, M. The organization of the cortical motor system: new concepts. Electroencephalography and Clinical Neurophysiology 1998; 106(4): 283–96.Google Scholar
Loubinoux, I, Dechaumont-Palacin, S, Castel-Lacanal, E, et al. Prognostic value of FMRI in recovery of hand function in subcortical stroke patients. Cereb Cortex 2007; 17(12): 2980–7.Google Scholar
Nelles, G, Jentzen, W, Jueptner, M, Mller, S, Diener, HC. Arm training induced brain plasticity in stroke studied with serial positron emission tomography. Neuroimage 2001; 13(6): 1146–54.Google Scholar
Marshall, RS, Perera, GM, Lazar, RM, et al. Evolution of cortical activation during recovery from corticospinal tract infarction. Stroke 2000; 31(3): 656–61.Google Scholar
Calautti, C, Leroy, F, Guincestre, JY, Baron, JC. Dynamics of motor network overactivation after striatocapsular stroke: a longitudinal PET study using a fixed-performance paradigm. Stroke 2001; 32(11): 2534–42.Google Scholar
Murase, N, Duque, J, Mazzocchio, R, Cohen, LG. Influence of interhemispheric interactions on motor function in chronic stroke. Ann Neurol 2004; 55(3): 400–9.Google Scholar
Shimizu, T, Hosaki, A, Hino, T, et al. Motor cortical disinhibition in the unaffected hemisphere after unilateral cortical stroke. Brain 2002; 125(8): 1896–907.Google Scholar
Hsu, WY, Cheng, CH, Liao, KK, Lee, IH, Lin, YY. Effects of repetitive transcranial magnetic stimulation on motor functions in patients with stroke: a meta-analysis. Stroke 2012; 43(7): 1849–57.Google Scholar
Thiel, A, Aleksic, B, Klein, JC, Rudolf, J, Heiss, W-D. Changes in proprioceptive systems activity during recovery from post-stroke hemiparesis. J Rehabilitation Medicine 2007; 39(7): 520–5.Google Scholar
Heiss, WD, Kessler, J, Thiel, A, Ghaemi, M, Karbe, H. Differential capacity of left and right hemispheric areas for compensation of poststroke aphasia. Ann Neurol 1999; 45(4): 430–8.Google Scholar
Cao, Y, Vikingstad, EM, George, KP, Johnson, AF, Welch, KMA. Cortical language activation in stroke patients recovering from aphasia with functional MRI. Stroke 1999; 30(11): 2331–40.Google Scholar
Warburton, E, Price, CJ, Swinburn, K, Wise, RJS. Mechanisms of recovery from aphasia: evidence from positron emission tomography studies. J Neurol Neurosurg Psychiatry 1999; 66(2): 155–61.Google Scholar
Saur, D, Lange, R, Baumgaertner, A, et al. Dynamics of language reorganization after stroke. Brain 2006; 129(6): 1371–84.Google Scholar
Kobayashi, M, Pascual-Leone, A. Transcranial magnetic stimulation in neurology. Lancet Neurol 2003; 2(3): 145–56.Google Scholar
Naeser, MA, Martin, PI, Nicholas, M, et al. Improved picture naming in chronic aphasia after TMS to part of right Broca's area: an open-protocol study. Brain Lang 2005; 93(1): 95105.Google Scholar
Thiel, A, Schumacher, B, Wienhard, K, et al. Direct demonstration of transcallosal disinhibition in language networks. J Cereb Blood Flow Metab 2006; 26(9): 1122–7.Google Scholar
Thiel, A, Hartmann, A, Rubi-Fessen, I, et al. Effects of non-invasive brain stimulation on language networks and recovery in early poststroke aphasia. Stroke 2013; 44(8): 2240–6.Google Scholar
Heiss, W-D, Thiel, A. Basic principles of rTMS in aphasia. In: Platz, T, ed. Therapeutic rTMS in Neurology: Principles, Evidence, and Practice Recommendations. Springer International Publishing Switzerland; 2016.Google Scholar
Umarova, RM, Nitschke, K, Kaller, CP, et al. Predictors and signatures of recovery from neglect in acute stroke. Ann Neurol 2016; 79(4): 673–86.Google Scholar

References

Brinjikji, W, Rabinstein, AA, Lanzino, G, et al. Ultrasound characteristics of symptomatic carotid plaques: a systematic review and meta-analysis. Cerebrovasc Dis 2015; 40(3–4): 165–74.Google Scholar
Silver, B. Carotid ultrasound. http://emedicine.medscape.com/article/1155193-overview. Updated: 15 December 2008.Google Scholar
Liu, CH, Chang, CH, Chang, TY, et al. Carotid artery stenting improves cerebral hemodynamics regardless of the flow direction of ophthalmic artery. Angiology 2015; 66(2): 180–6.Google Scholar
Grant, EG, Benson, CB, Moneta, GL, et al. Carotid artery stenosis: grayscale and Doppler ultrasound diagnosis – Society of Radiologists in Ultrasound consensus conference. Ultrasound Q 2003; 19(4): 190–8.Google Scholar
Nadalo, LA, Walters, MC. Carotid artery, stenosis: imaging. http://emedicine.medscape.com/article/417524-overview.Google Scholar
von Reutern, GM, Goertler, MW, Bornstein, NM, et al. Grading carotid stenosis using ultrasonic methods. Stroke 2012; 43(3): 916–21.Google Scholar
Jauch, EC, Saver, JL, Adams, HP Jr., et al. Guidelines for the early management of patients with acute ischemic stroke: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke 2013; 44(3): 870947.Google Scholar
Masdeu, JC, Irimiaa, P, Asenbaumb, S, et al. EFNS guideline on neuroimaging in acute stroke. Report of an EFNS task force. Eur J Neurol 2006; 13: 1271–83.Google Scholar
Afsharfard, A, Gholizadeh, B, Abdolalian, Y, Hosseini, SM. Carotid free-floating thrombus causing stroke in a young woman with lupus anticoagulant: a case report and review of the literature. Int J Surg Case Rep 2017; 33: 35–7.Google Scholar
Alexandrov, AV, Sloan, MA, Wong, LK, et al. American Society of Neuroimaging Practice Guidelines Committee. Practice standards for transcranial Doppler ultrasound: part I – test performance. J Neuroimaging 2007; 17(1): 1118.Google Scholar
Alexandrov, AV, Sloan, MA, Tegeler, CH, et al. Practice standards for transcranial Doppler (TCD) ultrasound. Part II. Clinical indications and expected outcomes. J Neuroimaging 2012; 22(3): 215–24.Google Scholar
Zipper, SG, Stolz, E. Clinical application of transcranial colour-coded duplex sonography – a review. Eur J Neurol 2002; 9(1): 18.Google Scholar
Baumgartner, RW. Transcranial color-coded duplex sonography. J Neurol 1999; 246(8): 637–47.Google Scholar
Valdueza, JM, Schreiber, SJ, Roehl, JE, Klingebiel, R. Neurosonology and Neuroimaging of Stroke. 2nd edn. Stuttgart, New York: Thieme; 2017.Google Scholar
Gerriets, T, Goertler, M, Stolz, E, et al. Feasibility and validity of transcranial duplex sonography in patients with acute stroke. J Neurol Neurosurg Psychiatry 2002; 73: 1720.Google Scholar
Chernyshev, OY, Garami, Z, Calleja, S, et al. Yield and accuracy of urgent combined carotid-transcranial ultrasound testing in acute cerebral ischemia. Stroke 2005; 36: 32–7.Google Scholar
Sharma, VK, Venketasubramanian, N, Khurana, DK, Tsivgoulis, G, Alexandrov, AV. Role of transcranial Doppler ultrasonography in acute stroke. Ann Indian Acad Neurol 2008; 11: 3951.Google Scholar
Azarpazhooh, MR, Chambers, BR. Clinical application of transcranial Doppler monitoring for embolic signals. J Clin Neurosci 2006; 13(8): 799810.Google Scholar
Segura, T, Serena, J, Castellanos, M, et al. Embolism in acute middle cerebral artery stenosis. Neurology 2001; 56: 497501.Google Scholar
Tegos, TJ, Sabetai, MM, Nicolaides, AN, et al. Correlates of embolic events detected by means of transcranial Doppler in patients with carotid atheroma. J Vasc Surg 2001; 33: 131–8.Google Scholar
Del Sette, M, Angeli, S, Stara, I, Finocchi, C, Gandolfo, C. Microembolic signals with serial transcranial Doppler monitoring in acute focal ischemic deficit. A local phenomenon? Stroke 1997; 28: 1311–13.Google Scholar
Markus, HS, Droste, DW, Kaps, M, et al. Dual antiplatelet therapy with clopidogrel and aspirin in symptomatic carotid stenosis evaluated using Doppler embolic signal detection: the Clopidogrel and Aspirin for Reduction of Emboli in Symptomatic Carotid Stenosis (CARESS) trial. Circulation 2005; 111(17): 2233–40.Google Scholar
Ritter, MA, Dittrich, R, Thoenissen, N, Ringelstein, EB, Nabavi, DG. Prevalence and prognostic impact of microembolic signals in arterial sources of embolism. A systematic review of the literature. J Neurol 2008; 255(7): 953–61.Google Scholar
Best, LM, Webb, AC, Gurusamy, KS, Cheng, SF, Richards, T. Transcranial Doppler ultrasound detection of microemboli as a predictor of cerebral events in patients with symptomatic and asymptomatic carotid disease: a systematic review and meta-analysis. Eur J Vasc Endovasc Surg 2016; 52(5): 565–80.Google Scholar
Markus, HS, King, A, Shipley, M, et al. Asymptomatic embolisation for prediction of stroke in the Asymptomatic Carotid Emboli Study (ACES): a prospective observational study. Lancet Neurol 2010; 9(7): 663–71.Google Scholar
Artemis, D, Alonso, A, Hennerici, MG, Meairs, S, Kern, R. Real-time ultrasound perfusion imaging in acute stroke: assessment of cerebral perfusion deficits related to arterial recanalization. Ultrasound Med Biol 2013; 39(5): 745–52.Google Scholar
Della Martina, A, Meyer-Wiethe, K, Allemann, E, Seidel, G. Ultrasound contrast agents for brain perfusion imaging and ischemic stroke therapy. J Neuroimaging 2005; 15(3): 217–32.Google Scholar
Seidel, G, Meyer-Wiethe, K. Acute stroke: perfusion imaging. Front Neurol Neurosci 2006; 21: 127–39.Google Scholar
Meairs, S, Kern, R. Intracranial perfusion imaging with ultrasound. Front Neurol Neurosci 2015; 36: 5770.Google Scholar
Seidel, G, Meyer-Wiethe, K, Berdien, G, et al. Ultrasound perfusion imaging in acute middle cerebral artery infarction predicts outcome. Stroke 2004; 35: 1107–11.Google Scholar
Vinke, EJ, Kortenbout, AJ, Eyding, J, et al. Potential of contrast-enhanced ultrasound as a bedside monitoring technique in cerebral perfusion: a systematic review. Ultrasound Med Biol 2017; 43(12): 2751–7.Google Scholar
Allendoerfer, J, Goertler, M, Reutern, GM. Prognostic relevance of ultra-early Doppler sonography in acute ischaemic stroke: a prospective multicentre study. Lancet Neurol 2006; 5(10): 835–40.Google Scholar
Demchuk, AM, Burgin, WS, Christou, I, et al. Thrombolysis in brain ischemia (TIBI) transcranial Doppler flow grades predict clinical severity, early recovery, and mortality in patients treated with intravenous tissue plasminogen activator. Stroke 2001; 32(1): 8993.Google Scholar
Baracchini, C, Manara, R, Ermani, M, Meneghetti, G. The quest for early predictors of stroke evolution: can TCD be a guiding light? Stroke 2000; 31: 2942–7.Google Scholar
Perren, F, Loulidi, J, Graves, R, et al. Combined IV–intraarterial thrombolysis: a color-coded duplex pilot study. Neurology 2006; 67: 324–6.Google Scholar
Saqqur, M, Uchino, K, Demchuk, AM, et al. Site of arterial occlusion identified by transcranial Doppler predicts the response to intravenous thrombolysis for stroke. Stroke 2007; 38(3): 948–54.Google Scholar
Alexandrov, AV, Burgin, SW, Demchuk, AM, El-Mitwalli, A, Grotta, JC. Speed of intracranial clot lysis with intravenous tissue plasminogen activator therapy: sonographic classification and short-term improvement. Circulation 2001; 103: 2897–902.Google Scholar
Rubiera, M, Ribo, M, Delgado-Mederos, R, et al. Tandem internal carotid artery/middle cerebral artery occlusion: an independent predictor of poor outcome after systemic thrombolysis. Stroke 2006; 37: 2301–5.Google Scholar
Nedelmann, M et al. and the TCCS Consensus Group. Consensus recommendations for transcranial color-coded duplex sonography for the assessment of intracranial arteries in clinical trials on acute stroke. Stroke 2009; 40(10): 3238–44.Google Scholar
Alexandrov, AV, Molina, CA, Grotta, JC, et al. For the CLOTBUST investigators: ultrasound-enhanced thrombolysis for acute ischemic stroke. N Engl J Med 2004; 351: 2170–8.Google Scholar
Barlinn, K, Tsivgoulis, G, Barreto, AD, et al. Outcomes following sonothrombolysis in severe acute ischemic stroke: subgroup analysis of the CLOTBUST trial. Int J Stroke 2014; 9(8): 1006–10.Google Scholar
Schellinger, PD, Alexandrov, AV, Barreto, AD, et al. CLOTBUSTER investigators. Combined lysis of thrombus with ultrasound and systemic tissue plasminogen activator for emergent revascularization in acute ischemic stroke (CLOTBUST-ER): design and methodology of a multinational phase 3 trial. Int J Stroke 2015; 10(7): 1141–8.Google Scholar
Ricci, S, Dinia, L, Del Sette, M, et al. Sonothrombolysis for acute ischaemic stroke (review). The Cochrane Library 2012, Issue 10, 135.Google Scholar
Droste, DW, Silling, K, Stypmann, J, et al. Contrast transcranial Doppler ultrasound in the detection of right-to-left shunts: time window and threshold in microbubble numbers. Stroke 2000; 31(7): 1640–5.Google Scholar
Belvis, R, Leta, RG, Marti-Fabregas, J, et al. Almost perfect concordance between simultaneous transcranial Doppler and transesophageal echocardiography in the quantification of right-to-left shunts. J Neuroimaging 2006; 16(2): 133–8.Google Scholar
Katsanos, AH, Psaltopoulou, T, Sergentanis, TN, et al. Transcranial Doppler versus transthoracic echocardiography for the detection of patent foramen ovale in patients with cryptogenic cerebral ischemia: a systematic review and diagnostic test accuracy meta-analysis. Ann Neurol 2016; 79(4): 625–35.Google Scholar
Seidel, G, Gerriets, T, Kaps, M, Missler, U. Dislocation of the third ventricle due to space-occupying stroke evaluated by transcranial duplex sonography. J Neuroimaging 1996; 6(4): 227–30.Google Scholar
Bellner, J, Romner, B, Reinstrup, P, et al. Transcranial Doppler sonography pulsatility index (PI) reflects intracranial pressure (ICP). Surg Neurol 2004; 62(1): 4551.Google Scholar
Lau, VI, Arntfield, RT. Point-of-care transcranial Doppler by intensivists. Crit Ultrasound J 2017; 9(1): 21.Google Scholar
Siebler, M. Neuroorbital ultrasound. In Csiba, L and Baracchini, C, eds. Manual of Neurosonology. Cambridge University Press; 2016: 300–5.Google Scholar
Ohle, R, McIsaac, SM, Woo, MY, Perry, JJ. Sonography of the optic nerve sheath diameter for detection of raised intracranial pressure compared to computed tomography: a systematic review and meta-analysis. J Ultrasound Med 2015; 34(7): 1285–94.Google Scholar
Khan, MN, Shallwani, H, Khan, MU, Shamim, MS. Noninvasive monitoring intracranial pressure – a review of available modalities. Surgical Neurology International 2017; 8: 51.Google Scholar
Rajajee, V, Vanaman, M, Fletcher, JJ, Jacobs, TL. Optic nerve ultrasound for the detection of raised intracranial pressure. Neurocrit Care 2011; 15(3): 506–15.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×