The role of the microcirculation in delayed cerebral ischemia and chronic degenerative changes after subarachnoid hemorrhage

Leif Østergaard, Rasmus Aamand, Sanja Karabegovic, Anna Tietze, Jakob Udby Blicher, Irene Klaerke Mikkelsen, Nina Kerting Iversen, Niels Secher, Thorbjørn Søndergaard Engedal, Mariam Anzabi, Eugenio Gutierrez Jimenez, Changsi Cai, Klaus Ulrik Koch, Erhard Trillingsgaard Naess-Schmidt, Annette Obel, Niels Juul, Mads Rasmussen, Jens Christian Hedemann Sørensen, Leif Østergaard, Rasmus Aamand, Sanja Karabegovic, Anna Tietze, Jakob Udby Blicher, Irene Klaerke Mikkelsen, Nina Kerting Iversen, Niels Secher, Thorbjørn Søndergaard Engedal, Mariam Anzabi, Eugenio Gutierrez Jimenez, Changsi Cai, Klaus Ulrik Koch, Erhard Trillingsgaard Naess-Schmidt, Annette Obel, Niels Juul, Mads Rasmussen, Jens Christian Hedemann Sørensen

Abstract

The mortality after aneurysmal subarachnoid hemorrhage (SAH) is 50%, and most survivors suffer severe functional and cognitive deficits. Half of SAH patients deteriorate 5 to 14 days after the initial bleeding, so-called delayed cerebral ischemia (DCI). Although often attributed to vasospasms, DCI may develop in the absence of angiographic vasospasms, and therapeutic reversal of angiographic vasospasms fails to improve patient outcome. The etiology of chronic neurodegenerative changes after SAH remains poorly understood. Brain oxygenation depends on both cerebral blood flow (CBF) and its microscopic distribution, the so-called capillary transit time heterogeneity (CTH). In theory, increased CTH can therefore lead to tissue hypoxia in the absence of severe CBF reductions, whereas reductions in CBF, paradoxically, improve brain oxygenation if CTH is critically elevated. We review potential sources of elevated CTH after SAH. Pericyte constrictions in relation to the initial ischemic episode and subsequent oxidative stress, nitric oxide depletion during the pericapillary clearance of oxyhemoglobin, vasogenic edema, leukocytosis, and astrocytic endfeet swelling are identified as potential sources of elevated CTH, and hence of metabolic derangement, after SAH. Irreversible changes in capillary morphology and function are predicted to contribute to long-term relative tissue hypoxia, inflammation, and neurodegeneration. We discuss diagnostic and therapeutic implications of these predictions.

Figures

Figure 1
Figure 1
Changes in the capillary morphology after subarachnoid hemorrhage (SAH), ischemia, and hypotonic hyponatremia. Panels A and B show swelling of astrocytic endfeet (*) and endothelial protrusions (arrows) after SAH. Scale bars indicate 5 μm in panel A and 2 μm in panel B, respectively. Reproduced from ref. with permission from the publisher. Panel C shows segmental narrowing of capillaries due to capillary constrictions after ischemia and reperfusion (bottom) compared with slender, thread-like, horseradish peroxidase-filled capillaries in the normal hemisphere (top). Reproduced from ref. with permission from the publisher. Panels D and E show tangential sections through capillaries in sham-operated brain (D) and in a hypotonic hyponatremic edema model (E) in rabbit. The distended astrocytic endfeet (OL), the membranes of which are marked by arrows, clearly compress the capillary lumen, as shown in the transverse section (F). Other astrocytic membranes are labeled ‘A', and axons ‘AX'. The magnifications were × 6500 (D and E) and × 5000 (F), respectively. Reproduced from ref. with permission from the publisher.
Figure 2
Figure 2
The relation between blood flow and net oxygen extraction in single capillaries. The curve in plot C shows the so-called flow–diffusion equation for oxygen, that is, shows the maximum amount of oxygen that can diffuse from a single capillary into tissue, for a given perfusion rate. The curve shape predicts three important properties of parallel-coupled capillaries: First, the curve slope decreases towards high-perfusion values, making vasodilation increasingly inefficient as a means of improving tissue oxygenation towards high-perfusion rates. Second, if erythrocyte flows differ among capillary paths (case B) instead of being equal (case A), then net tissue oxygen availability declines. This can be observed by using the curve to determine the net tissue oxygen availability resulting from the individual flows in case B. The resulting net tissue oxygen availability is the weighted average of the oxygen availabilities for the two flows, labeled b in the plot. Note that the resulting tissue oxygen availability will always be less than that of the homogenous case, labeled a. Conversely, homogenization of capillary flows during hyperemia has the opposite effect, and serves to compensate for the first property. By a similar argument, insert D disproves the traditional assumption that increased perfusion always results in improved tissue oxygenation: By increasing tissue perfusion from Fhom to Fhet, and again subdividing capillary flows in the latter case into f1 and f2, tissue oxygen availability in fact decreases in response to a flow increase, as indicated by the double asterisk. Note that, if erythrocyte flows are hindered (rather than continuously redistributed) along single capillary paths (as indicated by slow-passing white blood cell (WBC) and/or rugged capillary walls), upstream vasodilation is likely to amplify the redistribution losses, as erythrocytes are forced through other branches at very high speeds, with negligible net oxygenation gains. Reproduced from refs. and .
Figure 3
Figure 3
Metabolic thresholds. The green iso-contour surface corresponds to the metabolic rate of contralateral tissue in patients with focal ischemia. The red plane marks the boundary, left of which vasodilation fails to increase tissue oxygen availability (malignant capillary transit time heterogeneity (CTH)). The maximum value that CTH can attain at a tissue oxygen tension (PtO2) of 25 mm Hg, if oxygen availability is to remain above that of resting tissue, is indicated by the label A. As CTH increases further, a critical limit is reached as PtO2 approaches 0—label B. At this stage, the metabolic needs of tissue cannot be supported unless mean transit time (MTT) is prolonged to a threshold of approximately 4 seconds, corresponding to cerebral blood flow (CBF)=21 mL/100 mL/minute. Modified from ref. .
Figure 4
Figure 4
The subarachnoid hemorrhage (SAH) stages. The yellow arrows indicate flow with different velocities through the capillary system. The color within the vessels indicates oxygen saturation, and the background color outside the tissue oxygen tension. (A) The normal state, (B) the hyperemic state, where slight capillary transit time heterogeneity (CTH) increases lead to increased cerebral blood flow (CBF). (C) The flow suppression state. (D) Tissue hypoxia. OEF, oxygen extraction fraction.
Figure 5
Figure 5
The figure summarizes the pathways from the acute subarachnoid hemorrhage to early brain injury (EBI), delayed cerebral ischemia (DCI), and the long-term degenerative changes that are hypothesized to be the result of irreversible increases in capillary transit time heterogeneity (CTH). Nitric oxide synthetase (NOS) dysfunction refers to the loss of neuronal nitric oxide synthetase (nNOS) from nerve fibers in the arterial adventitia, and the inhibition of endothelial NOS (eNOS) by asymmetric dimethyl arginine (ADMA)—see text. The blue arrows indicate ‘vicious cycles' through which low oxygen tension exacerbates blood–brain barrier (BBB) disruption and capillary flow disturbances. The green text boxes indicate possible sites of therapeutic intervention discussed in the text. HgbO, oxyhemoglobin; NO, nitric oxide; ROS, reactive oxygen species; SD, spreading depolarizations.
Figure 6
Figure 6
Panel A summarizes the sources of capillary flow disturbances as identified in this review, and their estimated duration. They include the oxidative stress and nitric oxide (NO) depletion caused by pericapillary oxyhemoglobin, structural damage to the capillary wall (including endothelial cells, basement membranes, pericytes, and astrocytic endfeet), leukocytosis, and compression by pericapillary edema. The extent to which the structural damage to the capillary wall is permanent remains crucial in that permanent changes in capillary transit time heterogeneity (CTH) are potential sources of long-term, neurodegenerative changes. Panel B indicates the added effect of the changes in panel A in terms of the resulting change in CTH over time. The time of the peak CTH change is likely to coincide with the peak pericapillary oxyhemoglobin HgbO concentration, whereas the height of the peak is likely to correlate with the size of the hematoma. The extent to which one observes flow increases by transcranial Doppler sonography (TCD), angiographic vasospasm, or tissue lesions by computed tomography (CT) or magnetic resonance imaging (MRI), is predicted to depend on the individual sizes of the factors in panel A. Note that large hematomas are likely to generate large net CTH increases owing to their larger, net capillary NO depletion, and thus to result in both angiographic vasospasm and hypoxic lesions.

References

    1. van Gijn J, Kerr RS, Rinkel GJ. Subarachnoid haemorrhage. Lancet. 2007;369:306–318.
    1. Linn FH, Rinkel GJ, Algra A, van Gijn J. Incidence of subarachnoid hemorrhage: role of region, year, and rate of computed tomography: a meta-analysis. Stroke. 1996;27:625–629.
    1. Hop JW, Rinkel GJ, Algra A, van Gijn J. Case-fatality rates and functional outcome after subarachnoid hemorrhage: a systematic review. Stroke. 1997;28:660–664.
    1. Al-Khindi T, Macdonald RL, Schweizer TA. Cognitive and functional outcome after aneurysmal subarachnoid hemorrhage. Stroke. 2010;41:e519–e536.
    1. Broderick JP, Brott TG, Duldner JE, Tomsick T, Leach A. Initial and recurrent bleeding are the major causes of death following subarachnoid hemorrhage. Stroke. 1994;25:1342–1347.
    1. Grote E, Hassler W. The critical first minutes after subarachnoid hemorrhage. Neurosurgery. 1988;22:654–661.
    1. Voldby B, Enevoldsen EM. Intracranial pressure changes following aneurysm rupture. Part 1: clinical and angiographic correlations. J Neurosurg. 1982;56:186–196.
    1. Cahill J, Calvert JW, Zhang JH. Mechanisms of early brain injury after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2006;26:1341–1353.
    1. Doczi T. The pathogenetic and prognostic significance of blood-brain barrier damage at the acute stage of aneurysmal subarachnoid haemorrhage. Clinical and experimental studies. Acta Neurochir (Wien) 1985;77:110–132.
    1. Germano A, d'Avella D, Imperatore C, Caruso G, Tomasello F.Time-course of blood-brain barrier permeability changes after experimental subarachnoid haemorrhage Acta Neurochir (Wien) 2000142575–580.discussion 580-1.
    1. Winkler MK, Chassidim Y, Lublinsky S, Revankar GS, Major S, Kang EJ, et al. Impaired neurovascular coupling to ictal epileptic activity and spreading depolarization in a patient with subarachnoid hemorrhage: possible link to blood-brain barrier dysfunction. Epilepsia. 2012;53 (Suppl 6:22–30.
    1. Claassen J, Carhuapoma JR, Kreiter KT, Du EY, Connolly ES, Mayer SA. Global cerebral edema after subarachnoid hemorrhage: frequency, predictors, and impact on outcome. Stroke. 2002;33:1225–1232.
    1. Orakcioglu B, Fiebach JB, Steiner T, Kollmar R, Juttler E, Becker K, et al. Evolution of early perihemorrhagic changes—ischemia vs. edema: an MRI study in rats. Exp Neurol. 2005;193:369–376.
    1. Liu Y, Soppi V, Mustonen T, Kononen M, Koivisto T, Koskela A, et al. Subarachnoid hemorrhage in the subacute stage: elevated apparent diffusion coefficient in normal-appearing brain tissue after treatment. Radiology. 2007;242:518–525.
    1. Kreiter KT, Copeland D, Bernardini GL, Bates JE, Peery S, Claassen J, et al. Predictors of cognitive dysfunction after subarachnoid hemorrhage. Stroke. 2002;33:200–208.
    1. Hijdra A, Van Gijn J, Stefanko S, Van Dongen KJ, Vermeulen M, Van Crevel H. Delayed cerebral ischemia after aneurysmal subarachnoid hemorrhage: clinicoanatomic correlations. Neurology. 1986;36:329–333.
    1. Crowley RW, Medel R, Dumont AS, Ilodigwe D, Kassell NF, Mayer SA, et al. Angiographic vasospasm is strongly correlated with cerebral infarction after subarachnoid hemorrhage. Stroke. 2011;42:919–923.
    1. Macdonald RL, Weir BK. A review of hemoglobin and the pathogenesis of cerebral vasospasm. Stroke. 1991;22:971–982.
    1. Takeuchi K, Miyata N, Renic M, Harder DR, Roman RJ. Hemoglobin, NO, and 20-HETE interactions in mediating cerebral vasoconstriction following SAH. Am J Physiol Regul Integr Comp Physiol. 2006;290:R84–R89.
    1. Zimmermann M, Seifert V.Endothelin and subarachnoid hemorrhage: an overview Neurosurgery 199843863–875.discussion 875-6.
    1. Petzold GC, Einhaupl KM, Dirnagl U, Dreier JP. Ischemia triggered by spreading neuronal activation is induced by endothelin-1 and hemoglobin in the subarachnoid space. Ann Neurol. 2003;54:591–598.
    1. Gryglewski RJ, Palmer RM, Moncada S. Superoxide anion is involved in the breakdown of endothelium-derived vascular relaxing factor. Nature. 1986;320:454–456.
    1. Gursoy-Ozdemir Y, Qiu J, Matsuoka N, Bolay H, Bermpohl D, Jin H, et al. Cortical spreading depression activates and upregulates MMP-9. J Clin Invest. 2004;113:1447–1455.
    1. Takano T, Tian GF, Peng W, Lou N, Lovatt D, Hansen AJ, et al. Cortical spreading depression causes and coincides with tissue hypoxia. Nat Neurosci. 2007;10:754–762.
    1. Pluta RM, Afshar JK, Boock RJ, Oldfield EH. Temporal changes in perivascular concentrations of oxyhemoglobin, deoxyhemoglobin, and methemoglobin after subarachnoid hemorrhage. J Neurosurg. 1998;88:557–561.
    1. Pluta RM. Delayed cerebral vasospasm and nitric oxide: review, new hypothesis, and proposed treatment. Pharmacol Ther. 2005;105:23–56.
    1. Rey FE, Li XC, Carretero OA, Garvin JL, Pagano PJ. Perivascular superoxide anion contributes to impairment of endothelium-dependent relaxation: role of gp91(phox) Circulation. 2002;106:2497–2502.
    1. Sasaki T, Wakai S, Asano T, Watanabe T, Kirino T, Sano K. The effect of a lipid hydroperoxide of arachidonic acid on the canine basilar artery. An experimental study on cerebral vasospasm. J Neurosurg. 1981;54:357–365.
    1. Pyne-Geithman GJ, Nair SG, Stamper DN, Clark JF. Role of bilirubin oxidation products in the pathophysiology of DIND following SAH. Acta Neurochir Suppl. 2013;115:267–273.
    1. Pyne-Geithman GJ, Morgan CJ, Wagner K, Dulaney EM, Carrozzella J, Kanter DS, et al. Bilirubin production and oxidation in CSF of patients with cerebral vasospasm after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2005;25:1070–1077.
    1. Pluta RM, Thompson BG, Dawson TM, Snyder SH, Boock RJ, Oldfield EH. Loss of nitric oxide synthase immunoreactivity in cerebral vasospasm. J Neurosurg. 1996;84:648–654.
    1. Jung CS, Iuliano BA, Harvey-White J, Espey MG, Oldfield EH, Pluta RM. Association between cerebrospinal fluid levels of asymmetric dimethyl-L-arginine, an endogenous inhibitor of endothelial nitric oxide synthase, and cerebral vasospasm in a primate model of subarachnoid hemorrhage. J Neurosurg. 2004;101:836–842.
    1. Jung CS, Lange B, Zimmermann M, Seifert V. The CSF concentration of ADMA, but not of ET-1, is correlated with the occurrence and severity of cerebral vasospasm after subarachnoid hemorrhage. Neurosci Lett. 2012;524:20–24.
    1. Rabinstein AA, Friedman JA, Weigand SD, McClelland RL, Fulgham JR, Manno EM, et al. Predictors of cerebral infarction in aneurysmal subarachnoid hemorrhage. Stroke. 2004;35:1862–1866.
    1. Woitzik J, Dreier JP, Hecht N, Fiss I, Sandow N, Major S, et al. Delayed cerebral ischemia and spreading depolarization in absence of angiographic vasospasm after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2012;32:203–212.
    1. Macdonald RL, Higashida RT, Keller E, Mayer SA, Molyneux A, Raabe A, et al. Clazosentan, an endothelin receptor antagonist, in patients with aneurysmal subarachnoid haemorrhage undergoing surgical clipping: a randomised, double-blind, placebo-controlled phase 3 trial (CONSCIOUS-2) Lancet Neurol. 2011;10:618–625.
    1. Macdonald RL, Higashida RT, Keller E, Mayer SA, Molyneux A, Raabe A, et al. Randomized trial of clazosentan in patients with aneurysmal subarachnoid hemorrhage undergoing endovascular coiling. Stroke. 2012;43:1463–1469.
    1. Macdonald RL, Higashida RT, Keller E, Mayer SA, Molyneux A, Raabe A, et al. Randomised trial of clazosentan, an endothelin receptor antagonist, in patients with aneurysmal subarachnoid hemorrhage undergoing surgical clipping (CONSCIOUS-2) Acta Neurochir Suppl. 2013;115:27–31.
    1. Pluta RM, Hansen-Schwartz J, Dreier J, Vajkoczy P, Macdonald RL, Nishizawa S, et al. Cerebral vasospasm following subarachnoid hemorrhage: time for a new world of thought. Neurol Res. 2009;31:151–158.
    1. Dreier JP, Ebert N, Priller J, Megow D, Lindauer U, Klee R, et al. Products of hemolysis in the subarachnoid space inducing spreading ischemia in the cortex and focal necrosis in rats: a model for delayed ischemic neurological deficits after subarachnoid hemorrhage. J Neurosurg. 2000;93:658–666.
    1. Dreier JP, Woitzik J, Fabricius M, Bhatia R, Major S, Drenckhahn C, et al. Delayed ischaemic neurological deficits after subarachnoid haemorrhage are associated with clusters of spreading depolarizations. Brain. 2006;129:3224–3237.
    1. Koide M, Bonev AD, Nelson MT, Wellman GC. Inversion of neurovascular coupling by subarachnoid blood depends on large-conductance Ca2+-activated K+ (BK) channels. Proc Natl Acad Sci USA. 2012;109:E1387–E1395.
    1. Martinaud O, Perin B, Gerardin E, Proust F, Bioux S, Gars DL, et al. Anatomy of executive deficit following ruptured anterior communicating artery aneurysm. Eur J Neurol. 2009;16:595–601.
    1. Bendel P, Koivisto T, Aikia M, Niskanen E, Kononen M, Hanninen T, et al. Atrophic enlargement of CSF volume after subarachnoid hemorrhage: correlation with neuropsychological outcome. AJNR Am J Neuroradiol. 2010;31:370–376.
    1. Tam AK, Ilodigwe D, Li Z, Schweizer TA, Macdonald RL. Global cerebral atrophy after subarachnoid hemorrhage: a possible marker of acute brain injury and assessment of its impact on outcome. Acta Neurochir Suppl. 2013;115:17–21.
    1. Bendel P, Koivisto T, Niskanen E, Kononen M, Aikia M, Hanninen T, et al. Brain atrophy and neuropsychological outcome after treatment of ruptured anterior cerebral artery aneurysms: a voxel-based morphometric study. Neuroradiology. 2009;51:711–722.
    1. Bendel P, Koivisto T, Hanninen T, Kolehmainen A, Kononen M, Hurskainen H, et al. Subarachnoid hemorrhage is followed by temporomesial volume loss: MRI volumetric study. Neurology. 2006;67:575–582.
    1. Barth M, Capelle HH, Munch E, Thome C, Fiedler F, Schmiedek P, et al. Effects of the selective endothelin A (ET(A)) receptor antagonist Clazosentan on cerebral perfusion and cerebral oxygenation following severe subarachnoid hemorrhage—preliminary results from a randomized clinical series Acta Neurochir (Wien) 2007149911–918.discussion 918.
    1. Jespersen SN, Østergaard L. The roles of cerebral blood flow, capillary transit time heterogeneity and oxygen tension in brain oxygenation and metabolism. J Cereb Blood Flow Metab. 2012;32:264–277.
    1. Østergaard L, Jespersen SN, Mouridsen K, Mikkelsen IK, Jonsdottir KY, Tietze A, et al. The role of the cerebral capillaries in acute ischemic stroke: the extended penumbra model. J Cereb Blood Flow Metab. 2013;33:635–648.
    1. Østergaard L, Aamand R, Gutierrez-Jimenez E, Ho Y-L, Blicher JU, Madsen SM, et al. The capillary dysfunction hypothesis of Alzheimer's disease. Neurobiol Aging. 2013;34:1018–1031.
    1. Sehba FA, Friedrich V. Cerebral microvasculature is an early target of subarachnoid hemorrhage. Acta Neurochir Suppl. 2013;115:199–205.
    1. Yemisci M, Gursoy-Ozdemir Y, Vural A, Can A, Topalkara K, Dalkara T. Pericyte contraction induced by oxidative-nitrative stress impairs capillary reflow despite successful opening of an occluded cerebral artery. Nat Med. 2009;15:1031–1037.
    1. Peppiatt CM, Howarth C, Mobbs P, Attwell D. Bidirectional control of CNS capillary diameter by pericytes. Nature. 2006;443:700–704.
    1. Scholler K, Trinkl A, Klopotowski M, Thal SC, Plesnila N, Trabold R, et al. Characterization of microvascular basal lamina damage and blood-brain barrier dysfunction following subarachnoid hemorrhage in rats. Brain Res. 2007;1142:237–246.
    1. Kwon I, Kim EH, del Zoppo GJ, Heo JH. Ultrastructural and temporal changes of the microvascular basement membrane and astrocyte interface following focal cerebral ischemia. J Neurosci Res. 2009;87:668–676.
    1. Yee AH, Burns JD, Wijdicks EF. Cerebral salt wasting: pathophysiology, diagnosis, and treatment. Neurosurg Clin N Am. 2010;21:339–352.
    1. Olson JE, Banks M, Dimlich RV, Evers J. Blood-brain barrier water permeability and brain osmolyte content during edema development. Acad Emerg Med. 1997;4:662–673.
    1. LUSE SA, HARRIS B. Electron microscopy of the brain in experimental edema. J Neurosurg. 1960;17:439–446.
    1. Carare RO, Bernardes-Silva M, Newman TA, Page AM, Nicoll JA, Perry VH, et al. Solutes, but not cells, drain from the brain parenchyma along basement membranes of capillaries and arteries: significance for cerebral amyloid angiopathy and neuroimmunology. Neuropathol Appl Neurobiol. 2008;34:131–144.
    1. Iliff JJ, Wang M, Liao Y, Plogg BA, Peng W, Gundersen GA, et al. A paravascular pathway facilitates CSF flow through the brain parenchyma and the clearance of interstitial solutes, including amyloid beta. Sci Transl Med. 2012;4:147ra111.
    1. Rennels ML, Blaumanis OR, Grady PA. Rapid solute transport throughout the brain via paravascular fluid pathways. Adv Neurol. 1990;52:431–439.
    1. Ichimura T, Fraser PA, Cserr HF. Distribution of extracellular tracers in perivascular spaces of the rat brain. Brain Res. 1991;545:103–113.
    1. Sun BL, Xia ZL, Wang JR, Yuan H, Li WX, Chen YS, et al. Effects of blockade of cerebral lymphatic drainage on regional cerebral blood flow and brain edema after subarachnoid hemorrhage. Clin Hemorheol Microcirc. 2006;34:227–232.
    1. Sun BL, Xie FM, Yang MF, Cao MZ, Yuan H, Wang HT, et al. Blocking cerebral lymphatic drainage deteriorates cerebral oxidative injury in rats with subarachnoid hemorrhage. Acta Neurochir Suppl. 2011;110:49–53.
    1. Attwell D, Buchan AM, Charpak S, Lauritzen M, Macvicar BA, Newman EA. Glial and neuronal control of brain blood flow. Nature. 2010;468:232–243.
    1. Haefliger IO, Zschauer A, Anderson DR. Relaxation of retinal pericyte contractile tone through the nitric oxide-cyclic guanosine monophosphate pathway. Invest Ophthalmol Vis Sci. 1994;35:991–997.
    1. Haefliger IO, Anderson DR. Oxygen modulation of guanylate cyclase-mediated retinal pericyte relaxations with 3-morpholino-sydnonimine and atrial natriuretic peptide. Invest Ophthalmol Vis Sci. 1997;38:1563–1568.
    1. Dehouck MP, Vigne P, Torpier G, Breittmayer JP, Cecchelli R, Frelin C. Endothelin-1 as a mediator of endothelial cell-pericyte interactions in bovine brain capillaries. J Cereb Blood Flow Metab. 1997;17:464–469.
    1. Schonfelder U, Hofer A, Paul M, Funk RH. In situ observation of living pericytes in rat retinal capillaries. Microvasc Res. 1998;56:22–29.
    1. Lam HC, Lee JK, Lu CC, Chu CH, Chuang MJ, Wang MC. Role of endothelin in diabetic retinopathy. Curr Vasc Pharmacol. 2003;1:243–250.
    1. Renkin EMBW. Zweifach Award lecture. Regulation of the microcirculation. Microvasc Res. 1985;30:251–263.
    1. Provencio JJ. Inflammation in subarachnoid hemorrhage and delayed deterioration associated with vasospasm: a review. Acta Neurochir Suppl. 2013;115:233–238.
    1. Mazzoni MC, Schmid-Schonbein GW. Mechanisms and consequences of cell activation in the microcirculation. Cardiovasc Res. 1996;32:709–719.
    1. Tomita Y, Tomita M, Schiszler I, Amano T, Tanahashi N, Kobari M, et al. Moment analysis of microflow histogram in focal ischemic lesion to evaluate microvascular derangement after small pial arterial occlusion in rats. J Cereb Blood Flow Metab. 2002;22:663–669.
    1. Hudetz AG, Feher G, Weigle CG, Knuese DE, Kampine JP. Video microscopy of cerebrocortical capillary flow: response to hypotension and intracranial hypertension. Am J Physiol. 1995;268:H2202–H2210.
    1. Østergaard L, Sorensen AG, Chesler DA, Weisskoff RM, Koroshetz WJ, Wu O, et al. Combined diffusion-weighted and perfusion-weighted flow heterogeneity magnetic resonance imaging in acute stroke. Stroke. 2000;31:1097–1103.
    1. Simonsen CZ, Rohl L, Vestergaard-Poulsen P, Gyldensted C, Andersen G, Østergaard L. Final infarct size after acute stroke: prediction with flow heterogeneity. Radiology. 2002;225:269–275.
    1. Perkio J, Soinne L, Østergaard L, Helenius J, Kangasmaki A, Martinkauppi S, et al. Abnormal intravoxel cerebral blood flow heterogeneity in human ischemic stroke determined by dynamic susceptibility contrast magnetic resonance imaging. Stroke. 2005;36:44–49.
    1. Sette G, Baron JC, Mazoyer B, Levasseur M, Pappata S, Crouzel C.Local brain haemodynamics and oxygen metabolism in cerebrovascular disease. Positron emission tomography Brain 1989112Pt 4931–951.
    1. Stewart GN. Researches on the circulation time in organs and on the influences which affect it. Parts I.-III. J Physiol. 1894;15:1–89.
    1. Ndubuizu O, LaManna JC. Brain tissue oxygen concentration measurements. Antioxid Redox Signal. 2007;9:1207–1219.
    1. Moustafa RS, Baron JC.Perfusion Thresholds in Cerebral IschemiaIn: Donnan GA, Baron JC, Davis SM, Sharp FR (eds). Informa Healthcare USA, Inc.: New York; 200731–36.
    1. Pawlik G, Rackl A, Bing RJ. Quantitative capillary topography and blood flow in the cerebral cortex of cats: an in vivo microscopic study. Brain Res. 1981;208:35–58.
    1. Villringer A, Them A, Lindauer U, Einhaupl K, Dirnagl U. Capillary perfusion of the rat brain cortex. An in vivo confocal microscopy study. Circ Res. 1994;75:55–62.
    1. Kleinfeld D, Mitra PP, Helmchen F, Denk W. Fluctuations and stimulus-induced changes in blood flow observed in individual capillaries in layers 2 through 4 of rat neocortex. Proc Natl Acad Sci USA. 1998;95:15741–15746.
    1. Vogel J, Abounader R, Schrock H, Zeller K, Duelli R, Kuschinsky W. Parallel changes of blood flow and heterogeneity of capillary plasma perfusion in rat brains during hypocapnia. Am J Physiol. 1996;270:H1441–H1445.
    1. Abounader R, Vogel J, Kuschinsky W. Patterns of capillary plasma perfusion in brains in conscious rats during normocapnia and hypercapnia. Circ Res. 1995;76:120–126.
    1. Krolo I, Hudetz AG. Hypoxemia alters erythrocyte perfusion pattern in the cerebral capillary network. Microvasc Res. 2000;59:72–79.
    1. Nelson RJ, Perry S, Hames TK, Pickard JD. Transcranial Doppler ultrasound studies of cerebral autoregulation and subarachnoid hemorrhage in the rabbit. J Neurosurg. 1990;73:601–610.
    1. Grosset DG, Straiton J, McDonald I, Cockburn M, Bullock R. Use of transcranial Doppler sonography to predict development of a delayed ischemic deficit after subarachnoid hemorrhage. J Neurosurg. 1993;78:183–187.
    1. Aaslid R, Huber P, Nornes H. Evaluation of cerebrovascular spasm with transcranial Doppler ultrasound. J Neurosurg. 1984;60:37–41.
    1. Oertel MF, Scharbrodt W, Wachter D, Stein M, Schmidinger A, Boker DK. Arteriovenous differences of oxygen and transcranial Doppler sonography in the management of aneurysmatic subarachnoid hemorrhage. J Clin Neurosci. 2008;15:630–636.
    1. Voldby B, Enevoldsen EM, Jensen FT. Regional CBF, intraventricular pressure, and cerebral metabolism in patients with ruptured intracranial aneurysms. J Neurosurg. 1985;62:48–58.
    1. Cho HG, Shin HK, Shin YW, Lee JH, Hong KW. Role of nitric oxide in the CBF autoregulation during acute stage after subarachnoid haemorrhage in rat pial artery. Fundam Clin Pharmacol. 2003;17:563–573.
    1. Lassen NA. The luxury-perfusion syndrome and its possible relation to acute metabolic acidosis localised within the brain. Lancet. 1966;2:1113–1115.
    1. Voldby B, Enevoldsen EM, Jensen FT. Cerebrovascular reactivity in patients with ruptured intracranial aneurysms. J Neurosurg. 1985;62:59–67.
    1. Wang Z, Meng CJ, Shen XM, Shu Z, Ma C, Zhu GQ, et al. Potential contribution of hypoxia-inducible factor-1alpha, aquaporin-4, and matrix metalloproteinase-9 to blood-brain barrier disruption and brain edema after experimental subarachnoid hemorrhage. J Mol Neurosci. 2012;48:273–280.
    1. Yuan G, Khan SA, Luo W, Nanduri J, Semenza GL, Prabhakar NR. Hypoxia-inducible factor 1 mediates increased expression of NADPH oxidase-2 in response to intermittent hypoxia. J Cell Physiol. 2011;226:2925–2933.
    1. Shin HK, Lee JH, Kim KY, Kim CD, Lee WS, Rhim BY, et al. Impairment of autoregulatory vasodilation by NAD(P)H oxidase-dependent superoxide generation during acute stage of subarachnoid hemorrhage in rat pial artery. J Cereb Blood Flow Metab. 2002;22:869–877.
    1. Dawson VL, Dawson TM. Nitric oxide neurotoxicity. J Chem Neuroanat. 1996;10:179–190.
    1. Pacher P, Beckman JS, Liaudet L. Nitric oxide and peroxynitrite in health and disease. Physiol Rev. 2007;87:315–424.
    1. Iadecola C, Davisson RL. Hypertension and cerebrovascular dysfunction. Cell Metab. 2008;7:476–484.
    1. Sabri M, Ai J, Lakovic K, Macdonald RL. Mechanisms of microthrombosis and microcirculatory constriction after experimental subarachnoid hemorrhage. Acta Neurochir Suppl. 2013;115:185–192.
    1. Friedrich B, Muller F, Feiler S, Scholler K, Plesnila N. Experimental subarachnoid hemorrhage causes early and long-lasting microarterial constriction and microthrombosis: an in vivo microscopy study. J Cereb Blood Flow Metab. 2012;32:447–455.
    1. Powers WJ, Grubb RL, Jr, Baker RP, Mintun MA, Raichle ME. Regional cerebral blood flow and metabolism in reversible ischemia due to vasospasm. Determination by positron emission tomography. J Neurosurg. 1985;62:539–546.
    1. Kawamura S, Sayama I, Yasui N, Uemura K. Sequential changes in cerebral blood flow and metabolism in patients with subarachnoid haemorrhage. Acta Neurochir (Wien) 1992;114:12–15.
    1. Minhas PS, Menon DK, Smielewski P, Czosnyka M, Kirkpatrick PJ, Clark JC, et al. Positron emission tomographic cerebral perfusion disturbances and transcranial Doppler findings among patients with neurological deterioration after subarachnoid hemorrhage Neurosurgery 2003521017–1022.discussion 1022-4.
    1. Al-Rawi PG, Zygun D, Tseng MY, Hutchinson PJ, Matta BF, Kirkpatrick PJ. Cerebral blood flow augmentation in patients with severe subarachnoid haemorrhage. Acta Neurochir Suppl. 2005;95:123–127.
    1. Eltzschig HK, Carmeliet P. Hypoxia and inflammation. N Engl J Med. 2011;364:656–665.
    1. Reinprecht A, Czech T, Asenbaum S, Podreka I, Schmidbauer M.Low cerebrovascular reserve capacity in long-term follow-up after subarachnoid hemorrhage Surg Neurol 200564116–120.discussion 121.
    1. Szabo S, Sheth RN, Novak L, Rozsa L, Ficzere A. Cerebrovascular reserve capacity many years after vasospasm due to aneurysmal subarachnoid hemorrhage. A transcranial Doppler study with acetazolamide test. Stroke. 1997;28:2479–2482.
    1. Jeon H, Ai J, Sabri M, Tariq A, Shang X, Chen G, et al. Neurological and neurobehavioral assessment of experimental subarachnoid hemorrhage. BMC Neurosci. 2009;10:103–2202-10-103.
    1. Neil-Dwyer G, Cruickshank J. The blood leucocyte count and its prognostic significance in subarachnoid haemorrhage. Brain. 1974;97:79–86.
    1. Parkinson D, Stephensen S. Leukocytosis and subarachnoid hemorrhage. Surg Neurol. 1984;21:132–134.
    1. McGirt MJ, Mavropoulos JC, McGirt LY, Alexander MJ, Friedman AH, Laskowitz DT, et al. Leukocytosis as an independent risk factor for cerebral vasospasm following aneurysmal subarachnoid hemorrhage. J Neurosurg. 2003;98:1222–1226.
    1. Frontera JA, Fernandez A, Schmidt JM, Claassen J, Wartenberg KE, Badjatia N, et al. Defining vasospasm after subarachnoid hemorrhage: what is the most clinically relevant definition. Stroke. 2009;40:1963–1968.
    1. Mouridsen K, Friston K, Hjort N, Gyldensted L, Østergaard L, Kiebel S. Bayesian estimation of cerebral perfusion using a physiological model of microvasculature. Neuroimage. 2006;33:570–579.
    1. Mouridsen K, Østergaard L, Christensen S, Jespersen SN. Reliable estimation of capillary transit time distributions at voxel-level using DSC-MRI. Proc Int Soc Magn Reson Med 19th Annual Meeting and Exhibition. 2011. p. 3915.
    1. Kate M, Hansen MB, Mouridsen K, Østergaard L, Choi V, Gould B, et al. Blood pressure reduction does not reduce perihematoma oxygenation J Cereb Blood Flow Metabadvance online publication, 18 September 2013; doi:10.1038/jcbfm.2013.164
    1. Ayer RE, Zhang JH. Oxidative stress in subarachnoid haemorrhage: significance in acute brain injury and vasospasm. Acta Neurochir Suppl. 2008;104:33–41.
    1. Sen J, Belli A, Albon H, Morgan L, Petzold A, Kitchen N. Triple-H therapy in the management of aneurysmal subarachnoid haemorrhage. Lancet Neurol. 2003;2:614–621.
    1. Dankbaar JW, Slooter AJ, Rinkel GJ, Schaaf IC. Effect of different components of triple-H therapy on cerebral perfusion in patients with aneurysmal subarachnoid haemorrhage: a systematic review. Crit Care. 2010;14:R23.
    1. Connolly ES, Jr, Rabinstein AA, Carhuapoma JR, Derdeyn CP, Dion J, Higashida RT, et al. Guidelines for the management of aneurysmal subarachnoid hemorrhage: a guideline for healthcare professionals from the American Heart Association/American Stroke Association. Stroke. 2012;43:1711–1737.
    1. Borges N, Shi F, Azevedo I, Audus KL. Changes in brain microvessel endothelial cell monolayer permeability induced by adrenergic drugs. Eur J Pharmacol. 1994;269:243–248.
    1. Stocchetti N, Participants in the International Multi-Disciplinary Consensus Conference on the Critical Care Management of Subarachnoid Hemorrhage Triggers for aggressive interventions in subarachnoid hemorrhage. Neurocrit Care. 2011;15:324–328.
    1. Muench E, Horn P, Bauhuf C, Roth H, Philipps M, Hermann P, et al. Effects of hypervolemia and hypertension on regional cerebral blood flow, intracranial pressure, and brain tissue oxygenation after subarachnoid hemorrhage Crit Care Med 2007351844–1851.quiz 1852.
    1. Grande PO. The "Lund Concept" for the treatment of severe head trauma—physiological principles and clinical application. Intensive Care Med. 2006;32:1475–1484.
    1. Dizdarevic K, Hamdan A, Omerhodzic I, Kominlija-Smajic E. Modified Lund concept versus cerebral perfusion pressure-targeted therapy: a randomised controlled study in patients with secondary brain ischaemia. Clin Neurol Neurosurg. 2012;114:142–148.
    1. Kamel H, Navi BB, Nakagawa K, Hemphill JC, 3rd, Ko NU. Hypertonic saline versus mannitol for the treatment of elevated intracranial pressure: a meta-analysis of randomized clinical trials. Crit Care Med. 2011;39:554–559.
    1. Mortazavi MM, Romeo AK, Deep A, Griessenauer CJ, Shoja MM, Tubbs RS, et al. Hypertonic saline for treating raised intracranial pressure: literature review with meta-analysis. J Neurosurg. 2012;116:210–221.
    1. Torre-Healy A, Marko NF, Weil RJ. Hyperosmolar therapy for intracranial hypertension. Neurocrit Care. 2012;17:117–130.
    1. Bermueller C, Thal SC, Plesnila N, Schmid-Elsaesser R, Kreimeier U, Zausinger S. Hypertonic fluid resuscitation from subarachnoid hemorrhage in rats: a comparison between small volume resuscitation and mannitol. J Neurol Sci. 2006;241:73–82.
    1. Pascual JL, Khwaja KA, Chaudhury P, Christou NV. Hypertonic saline and the microcirculation. J Trauma. 2003;54:S133–S140.
    1. Al-Rawi PG, Tseng MY, Richards HK, Nortje J, Timofeev I, Matta BF, et al. Hypertonic saline in patients with poor-grade subarachnoid hemorrhage improves cerebral blood flow, brain tissue oxygen, and pH. Stroke. 2010;41:122–128.
    1. Nakagawa I, Hironaka Y, Nishimura F, Takeshima Y, Matsuda R, Yamada S, et al. Early inhibition of natriuresis suppresses symptomatic cerebral vasospasm in patients with aneurysmal subarachnoid hemorrhage. Cerebrovasc Dis. 2013;35:131–137.
    1. Kasprowicz M, Czosnyka M, Soehle M, Smielewski P, Kirkpatrick PJ, Pickard JD, et al. Vasospasm shortens cerebral arterial time constant. Neurocrit Care. 2012;16:213–218.
    1. Tseng MY, Al-Rawi PG, Pickard JD, Rasulo FA, Kirkpatrick PJ. Effect of hypertonic saline on cerebral blood flow in poor-grade patients with subarachnoid hemorrhage. Stroke. 2003;34:1389–1396.
    1. Tseng MY, Al-Rawi PG, Czosnyka M, Hutchinson PJ, Richards H, Pickard JD, et al. Enhancement of cerebral blood flow using systemic hypertonic saline therapy improves outcome in patients with poor-grade spontaneous subarachnoid hemorrhage. J Neurosurg. 2007;107:274–282.
    1. Hauer EM, Stark D, Staykov D, Steigleder T, Schwab S, Bardutzky J. Early continuous hypertonic saline infusion in patients with severe cerebrovascular disease. Crit Care Med. 2011;39:1766–1772.
    1. Froelich M, Ni Q, Wess C, Ougorets I, Hartl R. Continuous hypertonic saline therapy and the occurrence of complications in neurocritically ill patients. Crit Care Med. 2009;37:1433–1441.
    1. Bentsen G, Breivik H, Lundar T, Stubhaug A. Hypertonic saline (7.2%) in 6% hydroxyethyl starch reduces intracranial pressure and improves hemodynamics in a placebo-controlled study involving stable patients with subarachnoid hemorrhage. Crit Care Med. 2006;34:2912–2917.
    1. van Faassen EE, Bahrami S, Feelisch M, Hogg N, Kelm M, Kim-Shapiro DB, et al. Nitrite as regulator of hypoxic signaling in mammalian physiology. Med Res Rev. 2009;29:683–741.
    1. Montenegro MF, Amaral JH, Pinheiro LC, Sakamoto EK, Ferreira GC, Reis RI, et al. Sodium nitrite downregulates vascular NADPH oxidase and exerts antihypertensive effects in hypertension. Free Radic Biol Med. 2011;51:144–152.
    1. Shiva S. Mitochondria as metabolizers and targets of nitrite. Nitric Oxide. 2010;22:64–74.
    1. Shiva S. Nitrite: a physiological store of nitric oxide and modulator of mitochondrial function. Redox Biol. 2013;1:40–44.
    1. Larsen FJ, Schiffer TA, Borniquel S, Sahlin K, Ekblom B, Lundberg JO, et al. Dietary inorganic nitrate improves mitochondrial efficiency in humans. Cell Metab. 2011;13:149–159.
    1. Park JW, Piknova B, Huang PL, Noguchi CT, Schechter AN. Effect of blood nitrite and nitrate levels on murine platelet function. PLoS One. 2013;8:e55699.
    1. Pluta RM, Dejam A, Grimes G, Gladwin MT, Oldfield EH. Nitrite infusions to prevent delayed cerebral vasospasm in a primate model of subarachnoid hemorrhage. JAMA. 2005;293:1477–1484.
    1. Fathi AR, Pluta RM, Bakhtian KD, Qi M, Lonser RR. Reversal of cerebral vasospasm via intravenous sodium nitrite after subarachnoid hemorrhage in primates. J Neurosurg. 2011;115:1213–1220.
    1. Jung KH, Chu K, Ko SY, Lee ST, Sinn DI, Park DK, et al. Early intravenous infusion of sodium nitrite protects brain against in vivo ischemia-reperfusion injury. Stroke. 2006;37:2744–2750.
    1. Pluta RM. Prolonged intravenous infusion of sodium nitrite delivers nitric oxide (NO) in humans. Acta Neurochir Suppl. 2013;115:49–51.
    1. Oldfield EH, Loomba JJ, Monteith SJ, Crowley RW, Medel R, Gress DR, et al. Safety and pharmacokinetics of sodium nitrite in patients with subarachnoid hemorrhage: a phase IIA study. J Neurosurg. 2013;119:634–641.
    1. Yarnitsky D, Lorian A, Shalev A, Zhang ZD, Takahashi M, Agbaje-Williams M, et al. Reversal of cerebral vasospasm by sphenopalatine ganglion stimulation in a dog model of subarachnoid hemorrhage Surg Neurol 2005645–11.discussion 11.
    1. Takahashi M, Zhang ZD, Macdonald RL. Sphenopalatine ganglion stimulation for vasospasm after experimental subarachnoid hemorrhage. J Neurosurg. 2011;114:1104–1109.
    1. Nozaki K, Moskowitz MA, Maynard KI, Koketsu N, Dawson TM, Bredt DS, et al. Possible origins and distribution of immunoreactive nitric oxide synthase-containing nerve fibers in cerebral arteries. J Cereb Blood Flow Metab. 1993;13:70–79.
    1. Toda N, Ayajiki K, Yoshida K, Kimura H, Okamura T. Impairment by damage of the pterygopalatine ganglion of nitroxidergic vasodilator nerve function in canine cerebral and retinal arteries. Circ Res. 1993;72:206–213.
    1. Edvinsson L, Hamel E.Perivascular nerves in brain vesselsIn: Edvinsson L, Krause DN (eds). Cerebral Blood Flow and Metabolism Lippincott Williams & Wilkins: Philadelphia; 200243–67.
    1. Iadecola C, Beitz AJ, Renno W, Xu X, Mayer B, Zhang F. Nitric oxide synthase-containing neural processes on large cerebral arteries and cerebral microvessels. Brain Res. 1993;606:148–155.
    1. Ishikawa M, Kusaka G, Yamaguchi N, Sekizuka E, Nakadate H, Minamitani H, et al. Platelet and leukocyte adhesion in the microvasculature at the cerebral surface immediately after subarachnoid hemorrhage Neurosurgery 200964546–553.discussion 553-4.
    1. Gallia GL, Tamargo RJ. Leukocyte-endothelial cell interactions in chronic vasospasm after subarachnoid hemorrhage. Neurol Res. 2006;28:750–758.
    1. Katayama Y, Haraoka J, Hirabayashi H, Kawamata T, Kawamoto K, Kitahara T, et al. A randomized controlled trial of hydrocortisone against hyponatremia in patients with aneurysmal subarachnoid hemorrhage. Stroke. 2007;38:2373–2375.
    1. Feigin VL, Anderson N, Rinkel GJ, Algra A, van Gijn J, Bennett DA. Corticosteroids for aneurysmal subarachnoid haemorrhage and primary intracerebral haemorrhage. Cochrane Database Syst Rev. 2005. p. CD004583.
    1. McGirt MJ, Lynch JR, Parra A, Sheng H, Pearlstein RD, Laskowitz DT, et al. Simvastatin increases endothelial nitric oxide synthase and ameliorates cerebral vasospasm resulting from subarachnoid hemorrhage. Stroke. 2002;33:2950–2956.
    1. Parra A, Kreiter KT, Williams S, Sciacca R, Mack WJ, Naidech AM, et al. Effect of prior statin use on functional outcome and delayed vasospasm after acute aneurysmal subarachnoid hemorrhage: a matched controlled cohort study Neurosurgery 200556476–484.discussion 476-84.
    1. Tseng MY, Czosnyka M, Richards H, Pickard JD, Kirkpatrick PJ. Effects of acute treatment with pravastatin on cerebral vasospasm, autoregulation, and delayed ischemic deficits after aneurysmal subarachnoid hemorrhage: a phase II randomized placebo-controlled trial. Stroke. 2005;36:1627–1632.
    1. Kramer AH. Statins in the management of aneurysmal subarachnoid hemorrhage: an overview of animal research, observational studies, randomized controlled trials and meta-analyses. Acta Neurochir Suppl. 2011;110:193–201.
    1. Tseng MY, Participants in the International Multidisciplinary Consensus Conference on the Critical Care Management of Subarachnoid Hemorrhage Summary of evidence on immediate statins therapy following aneurysmal subarachnoid hemorrhage. Neurocrit Care. 2011;15:298–301.
    1. Laufs U, Wassmann S, Hilgers S, Ribaudo N, Bohm M, Nickenig G. Rapid effects on vascular function after initiation and withdrawal of atorvastatin in healthy, normocholesterolemic men. Am J Cardiol. 2001;88:1306–1307.
    1. Zschauer AO, Davis EB, Anderson DR. Glaucoma, capillaries and pericytes. 4. Beta-adrenergic activation of cultured retinal pericytes. Ophthalmologica. 1996;210:276–279.
    1. Kawamura H, Kobayashi M, Li Q, Yamanishi S, Katsumura K, Minami M, et al. Effects of angiotensin II on the pericyte-containing microvasculature of the rat retina. J Physiol. 2004;561:671–683.
    1. Matsugi T, Chen Q, Anderson DR. Contractile responses of cultured bovine retinal pericytes to angiotensin II. Arch Ophthalmol. 1997;115:1281–1285.
    1. Rasmussen G, Bergholdt B, Dahl B, Sunde N, Cold G, Voldby B. Effect of nimodipine on cerebral blood flow and cerebrovascular reactivity after subarachnoid haemorrhage. Acta Neurol Scand. 1999;99:182–186.

Source: PubMed

3
Se inscrever