J Clin Neurol. 2006 Sep;2(3):163-170. English.
Published online Sep 20, 2006.
Copyright © 2006 Korean Neurological Association
Review

Matrix Metalloproteinases in Cerebral Ischemia

Hahn Young Kim, M.D. and Seol-Heui Han, M.D.
    • Department of Neurology, Konkuk University Hospital, Center for Geriatric Neuroscience Research, Institute of Biomedical Science and Technology, Konkuk University, Seoul, Korea.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Matrix metalloproteinases (MMPs) are involved in the pathophysiology of several central nervous system diseases that share common pathogeneses, such as disruption of the blood-brain barrier (BBB), neuroinflammation, oxidative stress, and remodeling of the extracellular matrix (ECM). In early ischemic injury, MMPs participate in disruption of the BBB by digesting the basal lamina of capillaries and ECM, leading to vasogenic edema and hemorrhagic transformation. However, ECM degradation and remodeling are essential for tissue recovery, with MMPs having a key role as modulators of homeostasis between neuronal death and tissue regeneration. Thus, MMPs may be a double-edged sword that has a deleterious or beneficial role depending on the stage of brain injury.

Keywords
Matrix metalloproteinase; Extracellular matrix; Cerebral ischemia; Stroke

INTRODUCTION

Extracellular proteases comprise serine proteases, tissue-type and urokinase-type plasminogen activator (PA), and the families of matrix metalloproteinases (MMPs). MMPs are families of enzymes that require a zinc ion for catalytic activity. The role of MMPs has classically been described as strict regulators of extracellular matrix (ECM) remodeling in tissue morphogenesis and wound healing. MMPs regulate cellular activity in various ways, including ECM degradation, cell adhesion, proteolytic release of ECM-sequestrated molecules, and shedding of cell surface proteins that transduce signals from the extracellular environment (Table 1).1, 2 Recently, a deleterious role of MMPs in several diseases of the nervous system has been widely studied. However, some beneficial functions of MMPs during the recovery phase after brain injury have also been suggested. This review focuses on the role of MMPs in the acute and recovery phases of cerebral ischemia.

Table 1
Effects of MMPs in the central nervous system

STRUCTURE AND EXPRESSION OF MMPs

According to the basic protein domain and substrate preference, there are five major classes of MMPs: matrilysins, membrane-type MMPs (MT-MMPs), stromelysins, gelatinases, and other recently discovered MMP families.3 The protein structures of MMPs share a basic common pattern (Fig. 1) of propeptide and catalytic domains. However, they differ in more specific domains. The matrilysin MMP-7 has the simplest domain structure, with the propeptide domain and zinc containing catalytic domain, but it degrades most ECM components. The MT-MMPs (MMP-14, -15, and -16) have an additional transmembrane domain, whereas the stromelysins (MMP-1, -3, and -10) have a hemopexin domain. The gelatinases (MMP-2 and -9) have a unique fibronectin-binding domain, allowing them to bind to basement membranes. Thus, gelatinases specifically attack type IV collagen, laminin, and fibronectin. The common propeptide domain maintains the latency state of the enzyme, with removal of the signal peptide of this domain activating enzymes. Activation can also occur through a change in configuration. Proteolytic removal or reconfiguration of the propeptide domain activates the enzymes. Exposure of the catalytic site is referred to as the "cysteine switch".4 The various MMPs exist as secreted or membrane-bound enzymes that require conversion from zymogen to active forms through proteolytic processing, thus ensuring that MMP expression is tightly regulated at the transcriptional and posttranslational levels. Additionally, MMPs are also regulated extracellularly by proteins known as tissue inhibitors of metalloproteinases (TIMPs).5 The strict control of the activity of MMPs at multiple levels, such as transcription, posttranslational modification, and interaction with TIMPs is important, because MMPs play sophisticated roles and are extremely deleterious if they are erroneously activated.

INCREASED MMPs IN EXPERIMENTAL CEREBRAL ISCHEMIA

Recent studies have revealed the involvement of MMPs with clinical vascular diseases (Table 2). Ischemic stroke is often associated with disruption of the blood-brain barrier (BBB), leading to vasogenic edema and hemorrhage. Numerous animal studies suggest that disruption of the BBB and hemorrhagic transformation in ischemic stroke results from MMP activation in pathologic conditions.6-9 A deleterious role of MMPs associated with BBB injury has been demonstrated by a reduced infarct size and less BBB damage after focal ischemia in MMP-9 knockout mice,7, 9 and by both MMP activation and BBB injury being reduced in mice with superoxide dismutase overexpression.10 In nonhuman primates, early opening of the BBB after ischemic stroke correlates with elevated MMP-2, whereas hemorrhagic transformation is associated with elevated MMP-9.11, 12

Table 2
Clinical vascular diseases, target MMPs, and specimens in various studies

CLINICAL IMPLICATION OF MMPs IN ISCHEMIC STROKE

Gelatin zymography has been used to compare MMP activity between infarcted and matched noninfarcted cerebral tissue of acute or chronic ischemic stroke patients.13 MMP-9 activity was markedly elevated in the acute ischemic stroke tissue, whereas increases in MMP-2 activity were subtle at 2~5 days after ischemic stroke.13 An association between elevated MMPs and BBB disruption is also supported by the significant correlation between the level of MMP-9 and stroke severity being observed in stroke patients.14, 15 The MMP-9 levels measured by ELISA after acute cardioembolic stroke and the final NIHSS score are closely related,15 and MMP-9 appears also to be especially associated with hemorrhagic transformation.16-18 One shortcoming of analyzing the correlation between biomarkers and the clinicoradiologic outcome is the uncertainty of the cause-and-effect relationship. An increase in plasma MMP-9 levels may result from an acute-phase reaction or prior systemic causes. However, some studies have found that high plasma MMP-9 levels were related to hemorrhagic transformation, because increased MMP-9 levels were detected on admission in patients who subsequently developed hemorrhagic transformation in whom the initial stroke severity and final infarct volume were similar to those without hemorrhagic transformation.16, 19 Interestingly, a correlation between the baseline MMP-9 level and hemorrhagic complications after thrombolysis treatment has been observed, with the baseline MMP-9 level being a possible predictor of hemorrhagic complications after tPA thrombolysis.17 Measurement of the pretreatment level of MMP-9 was suggested as a possible safety measure when selecting candidates for thrombolysis. These data suggest a role for MMP-2 and MMP-9 in both the acute and chronic states of cerebral ischemia, BBB disruption, and efficacy and complication rate of tPA treatment. Future studies should focus on the temporal profiles of activation, selective activation among MMPs families, and the source of released MMPs.

tPA TREATMENT AND THE PA AXIS

PA and MMPs are the major proteases that modulate the ECM in the brain. Emerging data show important linkages between tPA, MMPs, vasogenic edema, and hemorrhage after stroke. Thrombolysis using tPA is the only FDA-approved treatment modality in acute stroke patients. The beneficial effect of tPA is intravascular thrombolysis restoring the blood flow to the ischemic parenchymal tissue. However, when tPA gains access to the extravascular neural parenchyma through a compromised BBB, any interaction between the NMDA receptors and exogenous tPA is a potential threat to the neural parenchyma. In addition to the direct neurotoxicity of tPA,20, 21 vasogenic edema or parenchymal hemorrhage induced by BBB disruption is another deleterious complication. MMP-9 may attack components of the basal lamina of cerebral vessels, and its activation can cause neurovascular damage of the BBB, leading to edema and hemorrhagic complications.6, 22, 23 The increased risk of edema and hemorrhagic complications in stroke patients following tPA therapy may therefore be due to tPA-induced upregulation of MMP-9. Plasmin is a potent protease that cleaves blood fibrin and activates other proteases, and thus is implicated in neurovascular and ECM degradation. Plasmin has an important role in the mechanism of tPA-induced hemorrhage by activating MMP-9 and MMP-2.24, 25 Whilst tPA can resolve intravascular blood clots, it can also produce abnormally high levels of plasmin that can trigger excessive activation of MMPs, leading to uncontrollable BBB disruption and, finally, intracranial hemorrhagic complications. MMP-9 levels are increased by tPA after experimentally induced focal ischemic stroke,26, 27 and MMP inhibitors reduce tPA-induced hemorrhagic transformation.27, 28 These data suggest that the activation of MMP-9 by administered tPA induces ischemic injury in the neurovascular unit. Therefore, modulating tPA and MMP-9 activity may provide a new approach for reducing complications of tPA thrombolysis. Once the clinical effects of MMPs are determined, clinical trials to modulate their activity could be started. Coadministration of MMP inhibitor or TIMPs with thrombolytic agents could avoid BBB disruption or ECM degradation. However, in clinical settings, such a drug should be highly tissue specific and avoid disturbing the physiologic ECM-rebuilding properties. Simple downregulation of MMP levels could evoke other side effects. Many MMP inhibitors have been tested for their efficacy in preventing cancer and rheumatoid arthritis,29, 30 but their side effects have so far precluded clinical trials. Thus, balanced modulation of MMP represents the major challenge for future stroke therapies.

MMPs IN STROKE RECOVERY

MMPs are known to be rapidly upregulated at 24~48 hours after ischemia,12, 25, 31 but little is known about their role during the subsequent recovery phase. The mechanism of stroke recovery mainly involves neuronal plasticity, neurogenesis, and angiogenesis.32, 33 MMPs may be critical for this recovery process because they can remodel components of the ECM. MMPs participate in dendritic and axonal extension as well as blood vessel formation.34 A recent study showed a beneficial role of MMP-9 in the recovery phase of experimental stroke,35 with surrogate markers of neuronal plasticity and vascular remodeling being correlated with MMP-9 signals in neurons and astrocytes in the peri-infarction area. Delayed inhibition of MMPs at 7 days poststroke worsened outcomes at 14 days by decreasing neurovascular remodeling, increasing infarction volumes, and blunting behavioral recovery.35 MMPs may in fact play beneficial roles during delayed stroke recovery, despite their deleterious effects in the acute phase of stroke. MMPs, and in particular MMP-9, have emerged as a target for stroke therapy in recent years.36 Pathophysiologic data from cell and animal models have been increasingly validated by the correlations between MMP-9 biomarkers and clinical outcomes in patients.16, 17 Therefore, there is growing momentum toward the development of MMP inhibitors for stroke therapy. In contrast to the therapeutic potential of MMP inhibition for acute stroke therapy, approaches to modulating or even enhancing MMPs in the late stages of stroke may represent novel techniques for improving stroke recovery and enhancing the long-term functional outcome. However, future studies should focus on how to modulate such a delicate and orchestrated role of MMPs according to the different phases of stroke.

MMPs IN SUBCORTICAL ISCHEMIC VASCULAR DEMENTIA

The significance of the white matter lesions that appear in various neurodegenerative diseases is still debated. The pathophysiology of subcortical ischemic vascular dementia or CADASIL (cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy) is significantly associated with white matter lesions.37-40 Demyelination around affected subcortical blood vessels, activated microglia, and macrophages can also be found in white matter lesions.38, 41 Brain tissue in patients with vascular dementia show fibrosis of small blood vessels with adjacent demyelination demyelination. Increased levels of several MMPs have also been observed in some patients with vascular dementia, with MMP-3 observed in reactive astrocytes and microglial cells.42 After controlling for the variation of MMP expression with age,43 the level of active MMP-9 in the cerebrospinal fluid or plasma of patients with vascular dementia still varies from that in Alzheimer's disease and other neurodegenerative conditions.44-46 MMPs may play an important role in nearly all kinds of dementia, especially in vascular dementia, but whether they are more important for the disease progress or are only an epiphenomenon of tissue adjustment is not clear. The profile of MMPs may in the future be a helpful marker for differentiating between subtypes of dementia.

CONCLUSIONS

MMPs are implicated in all cerebrovascular diseases and play a deleterious role in the pathophysiology of BBB disruption, neuroinflammation, oxidative stress, and delayed neuronal cell death. MMPs also have a role in the subacute or recovery phase through glial scarring, neuronal cell migration, and brain tissue recovery. Therapeutic strategies designed to modulate the activity of MMPs should consider these pleiotropic roles, and hence oversimplified experimental approaches with MMP inhibitors are likely to produce disappointing results. Carefully considering the various effects of MMPs is a challenge for future studies.

Notes

This study was supported by a grant of the Korea Health 21 R&D Project, Ministry of Health & Welfare, Republic of Korea (no. A050079).

References

    1. Cunningham LA, Wetzel M, Rosenberg GA. Multiple roles for MMPs and TIMPs in cerebral ischemia. Glia 2005;50:329–339.
    1. Yong VW, Power C, Forsyth P, Edwards DR. Metalloproteinases in biology and pathology of the nervous system. Nat Rev Neurosci 2001;2:502–511.
    1. Rosenberg GA. Matrix metalloproteinases in neuroinflammation. Glia 2002;39:279–291.
    1. Van Wart HE, Birkedal-Hansen H. The cysteine switch: a principle of regulation of metalloproteinase activity with potential applicability to the entire matrix metallopro teinase gene family. Proc Natl Acad Sci U S A 1990;87:5578–5582.
    1. Brew K, Dinakarpandian D, Nagase H. Tissue inhibitors of metalloproteinases: evolution, structure and function. Biochim Biophys Acta 2000;1477:267–283.
    1. Rosenberg GA, Estrada EY, Dencoff JE. Matrix metalloproteinases and TIMPs are associated with blood-brain barrier opening after reperfusion in rat brain. Stroke 1998;29:2189–2195.
    1. Asahi M, Asahi K, Jung JC, del Zoppo GJ, Fini ME, Lo EH. Role for matrix metalloproteinase 9 after focal cerebral ischemia: effects of gene knockout and enzyme inhibition with BB-94. J Cereb Blood Flow Metab 2000;20:1681–1689.
    1. Asahi M, Sumii T, Fini ME, Itohara S, Lo EH. Matrix metalloproteinase 2 gene knockout has no effect on acute brain injury after focal ischemia. Neuroreport 2001;12:3003–3007.
    1. Asahi M, Wang X, Mori T, Sumii T, Jung JC, Moskowitz MA, et al. Effects of matrix metalloproteinase-9 gene knock-out on the proteolysis of blood-brain barrier and white matter components after cerebral ischemia. J Neurosci 2001;21:7724–7732.
    1. Gasche Y, Copin JC, Sugawara T, Fujimura M, Chan PH. Matrix metalloproteinase inhibition prevents oxidative stress-associated blood-brain barrier disruption after transient focal cerebral ischemia. J Cereb Blood Flow Metab 2001;21:1393–1400.
    1. Chang DI, Hosomi N, Lucero J, Heo JH, Abumiya T, Mazar AP, et al. Activation systems for latent matrix metalloproteinase-2 are upregulated immediately after focal cerebral ischemia. J Cereb Blood Flow Metab 2003;23:1408–1419.
    1. Heo JH, Lucero J, Abumiya T, Koziol JA, Copeland BR, del Zoppo GJ. Matrix metalloproteinases increase very early during experimental focal cerebral ischemia. J Cereb Blood Flow Metab 1999;19:624–633.
    1. Clark AW, Krekoski CA, Bou SS, Chapman KR, Edwards DR. Increased gelatinase A (MMP-2) and gelatinase B (MMP-9) activities in human brain after focal ischemia. Neurosci Lett 1997;238:53–56.
    1. Horstmann S, Kalb P, Koziol J, Gardner H, Wagner S. Profiles of matrix metalloproteinases, their inhibitors, and laminin in stroke patients: influence of different therapies. Stroke 2003;34:2165–2170.
    1. Montaner J, Alvarez-Sabin J, Molina C, Angles A, Abilleira S, Arenillas J, et al. Matrix metalloproteinase expression after human cardioembolic stroke: temporal profile and relation to neurological impairment. Stroke 2001;32:1759–1766.
    1. Montaner J, Alvarez-Sabin J, Molina CA, Angles A, Abilleira S, Arenillas J, et al. Matrix metalloproteinase expression is related to hemorrhagic transformation after cardioembolic stroke. Stroke 2001;32:2762–2767.
    1. Montaner J, Molina CA, Monasterio J, Abilleira S, Arenillas JF, Ribo M, et al. Matrix metalloproteinase-9 pretreatment level predicts intracranial hemorrhagic complications after thrombolysis in human stroke. Circulation 2003;107:598–603.
    1. Rosell A, Ortega-Aznar A, Alvarez-Sabin J, Fernandez-Cadenas I, Ribo M, Molina CA, et al. Increased brain expression of matrix metalloproteinase-9 after ischemic and hemorrhagic human stroke. Stroke 2006;37:1399–1406.
    1. Castellanos M, Leira R, Serena J, Pumar JM, Lizasoain I, Castillo J, et al. Plasma metalloproteinase-9 concentration predicts hemorrhagic transformation in acute ischemic stroke. Stroke 2003;34:40–46.
    1. Nicole O, Docagne F, Ali C, Margaill I, Carmeliet P, MacKenzie ET, et al. The proteolytic activity of tissue-plasminogen activator enhances NMDA receptor-mediated signaling. Nat Med 2001;7:59–64.
    1. Matys T, Strickland S. Tissue plasminogen activator and NMDA receptor cleavage. Nat Med 2003;9:371–372.
    1. Lo EH, Wang X, Cuzner ML. Extracellular proteolysis in brain injury and inflammation: role for plasminogen activators and matrix metalloproteinases. J Neurosci Res 2002;69:1–9.
    1. Hamann GF, Okada Y, Fitridge R, del Zoppo GJ. Microvascular basal lamina antigens disappear during cerebral ischemia and reperfusion. Stroke 1995;26:2120–2126.
    1. Murphy G, Atkinson S, Ward R, Gavrilovic J, Reynolds JJ. The role of plasminogen activators in the regulation of connective tissue metalloproteinases. Ann N Y Acad Sci 1992;667:1–12.
    1. Mun-Bryce S, Rosenberg GA. Matrix metalloproteinases in cerebrovascular disease. J Cereb Blood Flow Metab 1998;18:1163–1172.
    1. Aoki T, Sumii T, Mori T, Wang X, Lo EH. Blood-brain barrier disruption and matrix metalloproteinase-9 expression during reperfusion injury: mechanical versus embolic focal ischemia in spontaneously hypertensive rats. Stroke 2002;33:2711–2717.
    1. Sumii T, Lo EH. Involvement of matrix metalloproteinase in thrombolysis-associated hemorrhagic transformation after embolic focal ischemia in rats. Stroke 2002;33:831–836.
    1. Lapchak PA, Chapman DF, Zivin JA. Metalloproteinase inhibition reduces thrombolytic (tissue plasminogen activator)-induced hemorrhage after thromboembolic stroke. Stroke 2000;31:3034–3040.
    1. Sternlicht MD, Werb Z. How matrix metalloproteinases regulate cell behavior. Annu Rev Cell Dev Biol 2001;17:463–516.
    1. Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases: structure, function, and biochemistry. Circ Res 2003;92:827–839.
    1. Gasche Y, Fujimura M, Morita-Fujimura Y, Copin JC, Kawase M, Massengale J, et al. Early appearance of activated matrix metalloproteinase-9 after focal cerebral ischemia in mice: a possible role in blood-brain barrier dysfunction. J Cereb Blood Flow Metab 1999;19:1020–1028.
    1. Cramer SC, Chopp M. Recovery recapitulates ontogeny. Trends Neurosci 2000;23:265–271.
    1. Chmielnicki E, Goldman SA. Induced neurogenesis by endogenous progenitor cells in the adult mammalian brain. Prog Brain Res 2002;138:451–464.
    1. Kaczmarek L, Lapinska-Dzwonek J, Szymczak S. Matrix metalloproteinases in the adult brain physiology: a link between c-Fos, AP-1 and remodeling of neuronal connections? EMBO J 2002;21:6643–6648.
    1. Zhao BQ, Wang S, Kim HY, Storrie H, Rosen BR, Mooney DJ, et al. Role of matrix metalloproteinases in delayed cortical responses after stroke. Nat Med 2006;12:441–445.
    1. Zlokovic BV. Remodeling after stroke. Nat Med 2006;12:390–391.
    1. Pantoni L, Garcia JH. Pathogenesis of leukoaraiosis: a review. Stroke 1997;28:652–659.
    1. Esiri MM, Wilcock GK, Morris JH. Neuropathological assessment of the lesions of significance in vascular dementia. J Neurol Neurosurg Psychiatry 1997;63:749–753.
    1. Singhal S, Rich P, Markus HS. The spatial distribution of MR imaging abnormalities in cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy and their relationship to age and clinical features. AJNR Am J Neuroradiol 2005;26:2481–2487.
    1. van Gijn J. Leukoaraiosis and vascular dementia. Neurology 1998;51:S3–S8.
    1. Akiguchi I, Tomimoto H, Suenaga T, Wakita H, Budka H. Alterations in glia and axons in the brains of Binswanger's disease patients. Stroke 1997;28:1423–1429.
    1. Rosenberg GA, Sullivan N, Esiri MM. White matter damage is associated with matrix metalloproteinases in vascular dementia. Stroke 2001;32:1162–1168.
    1. D'Armiento FP, Bianchi A, de Nigris F, Capuzzi DM, D'Armiento MR, Crimi G, et al. Age-related effects on atherogenesis and scavenger enzymes of intracranial and extracranial arteries in men without classic risk factors for atherosclerosis. Stroke 2001;32:2472–2479.
    1. Adair JC, Charlie J, Dencoff JE, Kaye JA, Quinn JF, Camicioli RM, et al. Measurement of gelatinase B (MMP-9) in the cerebrospinal fluid of patients with vascular dementia and Alzheimer disease. Stroke 2004;35:e159–e162.
    1. Lorenzl S, Albers DS, LeWitt PA, Chirichigno JW, Hilgenberg SL, Cudkowicz ME, et al. Tissue inhibitors of matrix metalloproteinases are elevated in cerebrospinal fluid of neurodegenerative diseases. J Neurol Sci 2003;207:71–76.
    1. Lorenzl S, Albers DS, Relkin N, Ngyuen T, Hilgenberg SL, Chirichigno J, et al. Increased plasma levels of matrix metalloproteinase-9 in patients with Alzheimer's disease. Neurochem Int 2003;43:191–196.
    1. Galis ZS, Sukhova GK, Lark MW, Libby P. Increased expression of matrix metalloproteinases and matrix degrading activity in vulnerable regions of human atherosclerotic plaques. J Clin Invest 1994;94:2493–2503.
    1. Nikkari ST, O'Brien KD, Ferguson M, Hatsukami T, Welgus HG, Alpers CE, et al. Interstitial collagenase (MMP-1) expression in human carotid atherosclerosis. Circulation 1995;92:1393–1398.
    1. Tziakas DN, Lazarides MK, Tentes IK, Georgiadis GS, Eleftheriadou E, Chalikias GK, et al. Gelatinases [matrix metalloproteinase-2 (MMP-2) and MMP-9] induce carotid plaque instability but their systemic levels are not predictive of local events. Ann Vasc Surg 2005;19:529–533.
    1. Turu MM, Krupinski J, Catena E, Rosell A, Montaner J, Rubio F, et al. Intraplaque MMP-8 levels are increased in asymptomatic patients with carotid plaque progression on ultrasound. Atherosclerosis 2006;187:161–169.
    1. Pawlak K, Pawlak D, Mysliwiec M. Serum matrix metalloproteinase-2 and increased oxidative stress are associated with carotid atherosclerosis in hemodialyzed patients. Atherosclerosis 2007;190:199–204.
    1. Montero I, Orbe J, Varo N, Beloqui O, Monreal JI, Rodriguez JA, et al. C-reactive protein induces matrix metalloproteinase-1 and -10 in human endothelial cells: implications for clinical and subclinical atherosclerosis. J Am Coll Cardiol 2006;47:1369–1378.
    1. Sigala F, Georgopoulos S, Papalambros E, Chasiotis D, Vourliotakis G, Niforou A, et al. Heregulin, cysteine rich-61 and matrix metalloproteinase 9 expression in human carotid atherosclerotic plaques: relationship with clinical data. Eur J Vasc Endovasc Surg 2006;32:238–245.
    1. Renko J, Kalela A, Jaakkola O, Laine S, Hoyhtya M, Alho H, et al. Serum matrix metalloproteinase-9 is elevated in men with a history of myocardial infarction. Scand J Clin Lab Invest 2004;64:255–261.
    1. Wu TC, Leu HB, Lin WT, Lin CP, Lin SJ, Chen JW. Plasma matrix metalloproteinase-3 level is an independent prognostic factor in stable coronary artery disease. Eur J Clin Invest 2005;35:537–545.
    1. Zeng B, Prasan A, Fung KC, Solanki V, Bruce D, Freedman SB, et al. Elevated circulating levels of matrix metalloproteinase-9 and -2 in patients with symptomatic coronary artery disease. Intern Med J 2005;35:331–335.
    1. Samnegard A, Silveira A, Tornvall P, Hamsten A, Ericsson CG, Eriksson P. Lower serum concentration of matrix metalloproteinase-3 in the acute stage of myocardial infarction. J Intern Med 2006;259:530–536.
    1. Alvarez-Sabin J, Delgado P, Abilleira S, Molina CA, Arenillas J, Ribo M, et al. Temporal profile of matrix metalloproteinases and their inhibitors after spontaneous intracerebral hemorrhage: relationship to clinical and radiological outcome. Stroke 2004;35:1316–1322.
    1. Kim SC, Singh M, Huang J, Prestigiacomo CJ, Winfree CJ, Solomon RA, et al. Matrix metalloproteinase-9 in cerebral aneurysms. Neurosurgery 1997;41:642–666.
    1. Todor DR, Lewis I, Bruno G, Chyatte D. Identification of a serum gelatinase associated with the occurrence of cerebral aneurysms as pro-matrix metalloproteinase-2. Stroke 1998;29:1580–1583.
    1. McGirt MJ, Lynch JR, Blessing R, Warner DS, Friedman AH, Laskowitz DT. Serum von Willebrand factor, matrix metalloproteinase-9, and vascular endothelial growth factor levels predict the onset of cerebral vasospasm after aneurysmal subarachnoid hemorrhage. Neurosurgery 2002;51:1128–1134.
    1. Asahina M, Yoshiyama Y, Hattori T. Expression of matrix metalloproteinase-9 and urinary-type plasminogen activator in Alzheimer's disease brain. Clin Neuropathol 2001;20:60–63.
    1. Leake A, Morris CM, Whateley J. Brain matrix metalloproteinase 1 levels are elevated in Alzheimer's disease. Neurosci Lett 2000;291:201–203.
    1. Yoshiyama Y, Asahina M, Hattori T. Selective distribution of matrix metalloproteinase-3 (MMP-3) in Alzheimer's disease brain. Acta Neuropathol (Berl) 2000;99:91–95.
    1. Inspector M, Aharon-Perez J, Glass-Marmor L, Miller A. Matrix metalloproteinase-9, its tissue inhibitor (TIMP)-1 and CRP in Alzheimer's disease. Eur Neurol 2005;53:155–157.

Metrics
Share
Figures

1 / 1

Tables

1 / 2

PERMALINK