251
|
Wang J. Preclinical and clinical research on inflammation after intracerebral hemorrhage. Prog Neurobiol 2010; 92:463-77. [PMID: 20713126 DOI: 10.1016/j.pneurobio.2010.08.001] [Citation(s) in RCA: 472] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2010] [Revised: 07/24/2010] [Accepted: 08/09/2010] [Indexed: 12/15/2022]
Abstract
Intracerebral hemorrhage (ICH) is one of the most lethal stroke subtypes. Despite the high morbidity and mortality associated with ICH, its pathophysiology has not been investigated as well as that of ischemic stroke. Available evidence from preclinical and clinical studies suggests that inflammatory mechanisms are involved in the progression of ICH-induced secondary brain injury. For example, in preclinical ICH models, microglial activation has been shown to occur within 1h, much earlier than neutrophil infiltration. Recent advances in our understanding of neuroinflammatory pathways have revealed several new molecular targets, and related therapeutic strategies have been tested in preclinical ICH models. This review summarizes recent progress made in preclinical models of ICH, surveys preclinical and clinical studies of inflammatory cells (leukocytes, macrophages, microglia, and astrocytes) and inflammatory mediators (matrix metalloproteinases, nuclear factor erythroid 2-related factor 2, heme oxygenase, and iron), and highlights the emerging areas of therapeutic promise.
Collapse
Affiliation(s)
- Jian Wang
- Department of Anesthesiology/Critical Care Medicine, The Johns Hopkins University, School of Medicine, 720 Rutland Avenue, Traylor Building 809, Baltimore, MD 21205, USA.
| |
Collapse
|
252
|
Effect of Tianhuang Granule on intracranial pressure and serum matrix metalloproteinase-9 in patients with acute cerebral hemorrhage. Chin J Integr Med 2010; 16:304-8. [PMID: 20697940 DOI: 10.1007/s11655-010-0514-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Indexed: 10/19/2022]
Abstract
OBJECTIVE To study the effect and mechanism of Tianhuang Granule (, THG) on: hydrocephalus in the patients with acute cerebral hemorrhage (ACH) through intracranial pressure (ICP) monitoring, serum matrix metalloproteinase-9 (MMP-9) level observation, and National Institutes of Health Stroke Scale (NIHSS) scoring (for nerve function de ficit). METHODS Sixty patients with ACH were equally randomized: into two groups by lottery, the control group and the THG group; all were treated with conventional therapy, but to the patients in the THG group, THG was given orally in addition for 28 days. Changes of ICP, MMP-9 expression, and NIHSS scores, as well as the degree of cerebral hematoma and hydrocephalus (by cranial CT scanning) in the patients, were estimated and compared. RESULTS (1) ICP was lowered more significantly in the: THG group, showing a significant difference between groups on day 7 (P<0.05). (2) MMP-9 expression was down-regulated in the THG group more significantly and earlier than that in the control group. (3) The degrees of cerebral hematoma and hydrocephalus in the THG group on day 7 were reduced significantly as compared with those on day 3 (P<0.05), but in the control group, the day of significant reduction was delayed to day 14, and the degrees on day 7 and day 14 in the two groups were significantly different (P<0.05 and P<0.01). (4) NIHSS score was significantly lower in the THG group than that in the control group on day 14 and day 28 (P<0.05 and P<0.01). CONCLUSION THG can effectively lower ICP, down-regulate MMP-9 expression, promote the absorption: of cerebral hematoma and hydrocephalus, and improve the nerve function, showing a clinical effectiveness than conventional therapy.
Collapse
|
253
|
Csiba L, Farkas S, Kollár J, Berényi E, Nagy K, Bereczki D. Visualization of the ischemic core on native human brain slices by potassium staining method. J Neurosci Methods 2010; 192:17-21. [PMID: 20624426 DOI: 10.1016/j.jneumeth.2010.07.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 07/01/2010] [Accepted: 07/02/2010] [Indexed: 10/19/2022]
Abstract
The potassium staining method is based on the formation of potassium cobaltnitrite crystals after the treatment by sodium cobaltnitrite of brain tissue. The degree of staining correlates with the distinct potassium content of infracted and non-infarcted brain areas. The aim of the present study was to prove that potassium staining technique is a reliable method for localization of ischemic core on native whole hemisphere cryosections of stroke patients. Furthermore, potassium stained sections have been compared with appropriate postmortem MRI images of respective brains. Brains of stroke patients were removed within 24h after death and postmortem MRI scanning was performed. Horizontal cryosections of frozen brains were taken and potassium staining was performed. Using the stained whole hemisphere sections as "map" tissue sampling has been made in order to determine water and potassium content. Potassium content of infarcted samples was significantly decreased in comparison with intact regions (0.7346+/-0.2142 mg/L and 1.928+/-0.447 mg/L, respectively, p<0.01) (mean values+/-SD). Water content of affected areas (expressed in percents) has been found to be above non-infarcted regions (81.657%+/-4.07 and 72.96%+/-6.37, respectively, p<0.01). According to our results the potassium staining method of human whole hemisphere brain sections reliably differentiates focal ischemic areas from intact brain regions. In conclusion, the postmortem examination of ischemic brain could be started with making the potassium map of infarcted whole hemisphere cryosections providing guidance for targeted tissue sampling and base of comparison for further examinations.
Collapse
Affiliation(s)
- László Csiba
- Department of Neurology, University of Debrecen Medical and Health Science Center, Móricz Zsigmond Street 22, Debrecen, H-4032, Hungary.
| | | | | | | | | | | |
Collapse
|
254
|
Denes A, Thornton P, Rothwell NJ, Allan SM. Inflammation and brain injury: acute cerebral ischaemia, peripheral and central inflammation. Brain Behav Immun 2010; 24:708-23. [PMID: 19770034 DOI: 10.1016/j.bbi.2009.09.010] [Citation(s) in RCA: 218] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2009] [Revised: 09/15/2009] [Accepted: 09/15/2009] [Indexed: 12/18/2022] Open
Abstract
Inflammation is a classical host defence response to infection and injury that has many beneficial effects. However, inappropriate (in time, place and magnitude) inflammation is increasingly implicated in diverse disease states, now including cancer, diabetes, obesity, atherosclerosis, heart disease and, most relevant here, CNS disease. A growing literature shows strong correlations between inflammatory status and the risk of cerebral ischaemia (CI, most commonly stroke), as well as with outcome from an ischaemic event. Intervention studies to demonstrate a causal link between inflammation and CI (or its consequences) are limited but are beginning to emerge, while experimental studies of CI have provided direct evidence that key inflammatory mediators (cytokines, chemokines and inflammatory cells) contribute directly to ischaemic brain injury. However, it remains to be determined what the relative importance of systemic (largely peripheral) versus CNS inflammation is in CI. Animal models in which CI is driven by a CNS intervention may not accurately reflect the clinical condition; stroke being typically induced by atherosclerosis or cardiac dysfunction, and hence current experimental paradigms may underestimate the contribution of peripheral inflammation. Experimental studies have already identified a number of potential anti-inflammatory therapeutic interventions that may limit ischaemic brain damage, some of which have been tested in early clinical trials with potentially promising results. However, a greater understanding of the contribution of inflammation to CI is still required, and this review highlights some of the key mechanism that may offer future therapeutic targets.
Collapse
Affiliation(s)
- A Denes
- Faculty of Life Sciences, The University of Manchester, Manchester, UK
| | | | | | | |
Collapse
|
255
|
Stamatovic SM, Keep RF, Andjelkovic AV. Brain endothelial cell-cell junctions: how to "open" the blood brain barrier. Curr Neuropharmacol 2010; 6:179-92. [PMID: 19506719 PMCID: PMC2687937 DOI: 10.2174/157015908785777210] [Citation(s) in RCA: 367] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Revised: 03/10/2008] [Accepted: 04/02/2008] [Indexed: 01/19/2023] Open
Abstract
The blood-brain barrier (BBB) is a highly specialized structural and biochemical barrier that regulates the entry of blood-borne molecules into brain, and preserves ionic homeostasis within the brain microenvironment. BBB properties are primarily determined by junctional complexes between the cerebral endothelial cells. These complexes are comprised of tight and adherens junctions. Such restrictive angioarchitecture at the BBB reduces paracellular diffusion, while minimal vesicle transport activity in brain endothelial cells limits transcellular transport. Under normal conditions, this largely prevents the extravasation of large and small solutes (unless specific transporters are present) and prevents migration of any type of blood-borne cell. However, this is changed in many pathological conditions. There, BBB disruption (“opening”) can lead to increased paracellular permeability, allowing entry of leukocytes into brain tissue, but also contributing to edema formation. In parallel, there are changes in the endothelial pinocytotic vesicular system resulting in the uptake and transfer of fluid and macromolecules into brain parenchyma. This review highlights the route and possible factors involved in BBB disruption in a variety of neuropathological disorders (e.g. CNS inflammation, Alzheimer’s disease, Parkinson’s disease, epilepsy). It also summarizes proposed signal transduction pathways that may be involved in BBB “opening”.
Collapse
|
256
|
Wu H, Zhang Z, Li Y, Zhao R, Li H, Song Y, Qi J, Wang J. Time course of upregulation of inflammatory mediators in the hemorrhagic brain in rats: correlation with brain edema. Neurochem Int 2010; 57:248-53. [PMID: 20541575 DOI: 10.1016/j.neuint.2010.06.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 05/24/2010] [Accepted: 06/02/2010] [Indexed: 11/29/2022]
Abstract
Intracerebral hemorrhage (ICH) can cause secondary brain damage through inflammation-related pathways. Thrombin and one of its receptors, protease activated receptor-1 (PAR-1); matrix metalloproteinase (MMP)-9; and aquaporin (AQP)-4 are stroke-related inflammatory mediators that have been implicated in ICH pathology. To further characterize the inflammatory response after ICH, we studied the temporal profile of the expression of these inflammatory mediators and assessed their potential correlation with brain edema formation after brain hemorrhage in rats. ICH was modeled by infusing autologous blood into the striatum. Then mRNA and protein expression was assessed over the course of 5 days. We found that the mRNA and/or protein expression of thrombin, PAR-1, AQP-4, and MMP-9 was upregulated between 2h and 5 days after ICH. Each reached a maximal level at day 2, except for AQP-4 protein, which peaked at day 5. Brain water content after ICH presented a similar trend; it was increased at 2h, peaked at day 2, and then decreased but remained elevated at day 5. Our data provide novel evidence that upregulation of these selected inflammatory mediators occurs very early and persists for several days after ICH, and that temporal patterns of expression of thrombin and AQP-4 are associated with brain edema formation. These findings have important implications for efforts to reduce secondary brain damage after ICH.
Collapse
Affiliation(s)
- He Wu
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin 150001, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
257
|
Adler I, Batton D, Betz B, Bezinque S, Ecklund K, Junewick J, McCauley R, Miller C, Seibert J, Specter B, Westra S, Leviton A. Mechanisms of injury to white matter adjacent to a large intraventricular hemorrhage in the preterm brain. JOURNAL OF CLINICAL ULTRASOUND : JCU 2010; 38:254-258. [PMID: 20232402 PMCID: PMC2989674 DOI: 10.1002/jcu.20683] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The purpose of this article is to investigate the hyperechoic lesion seen adjacent to a lateral ventricle that contains blood but is not distended. The literature on ependymal barrier dysfunction was reviewed in search of mechanisms of injury to the white matter adjacent to an intraventricular hemorrhage. The clinical literature on the clinical diagnosis of periventricular hemorrhagic infarction was also reviewed to find out how frequently this diagnosis was made. Support was found for the possibility that the ventricular wall does not always function as an efficient barrier, allowing ventricular contents to gain access to the white matter where they cause damage. Hemorrhagic infarction may not be the only or the most frequent mechanism of white matter damage adjacent to a large intraventricular hemorrhage.
Collapse
Affiliation(s)
- Ira Adler
- Eastern Radiologists, Greenville, NC 27834, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
258
|
Cardoso FL, Brites D, Brito MA. Looking at the blood-brain barrier: molecular anatomy and possible investigation approaches. ACTA ACUST UNITED AC 2010; 64:328-63. [PMID: 20685221 DOI: 10.1016/j.brainresrev.2010.05.003] [Citation(s) in RCA: 409] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2009] [Revised: 05/18/2010] [Accepted: 05/19/2010] [Indexed: 12/17/2022]
Abstract
The blood-brain barrier (BBB) is a dynamic and complex interface between blood and the central nervous system that strictly controls the exchanges between the blood and brain compartments, therefore playing a key role in brain homeostasis and providing protection against many toxic compounds and pathogens. In this review, the unique properties of brain microvascular endothelial cells and intercellular junctions are examined. The specific interactions between endothelial cells and basement membrane as well as neighboring perivascular pericytes, glial cells and neurons, which altogether constitute the neurovascular unit and play an essential role in both health and function of the central nervous system, are also explored. Some relevant pathways across the endothelium, as well as mechanisms involved in the regulation of BBB permeability, and the emerging role of the BBB as a signaling interface are addressed as well. Furthermore, we summarize some of the experimental approaches that can be used to monitor BBB properties and function in a variety of conditions and have allowed recent advances in BBB knowledge. Elucidation of the molecular anatomy and dynamics of the BBB is an essential step for the development of new strategies directed to maintain or restore BBB integrity and barrier function and ultimately preserve the delicate interstitial brain environment.
Collapse
Affiliation(s)
- Filipa Lourenço Cardoso
- Research Institute for Medicines and Pharmaceutical Sciences (iMed.UL), Faculty of Pharmacy, University of Lisbon, Lisbon, Portugal
| | | | | |
Collapse
|
259
|
Wu H, Zhang Z, Hu X, Zhao R, Song Y, Ban X, Qi J, Wang J. Dynamic changes of inflammatory markers in brain after hemorrhagic stroke in humans: a postmortem study. Brain Res 2010; 1342:111-7. [PMID: 20420814 DOI: 10.1016/j.brainres.2010.04.033] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 04/12/2010] [Accepted: 04/15/2010] [Indexed: 12/15/2022]
Abstract
This histopathologic case-control study was designed to characterize the dynamic changes in protein expression of nuclear factor-kappa B (NF-kappaB)/p65 subunit, macrophage inflammatory protein-2 (MIP-2), and matrix metalloproteinase-9 (MMP-9) in postmortem brains of patients with and without intracerebral hemorrhage (ICH). Thirty-six human brains from patients with ICH and six control brains were included in this study. We found that expression levels of NF-kappaB/p65, MIP-2, and MMP-9 were each upregulated on the injured side of the hippocampus at times ranging from 2h to 5days post-ICH. Interestingly, the expression of all three markers was also upregulated on the uninjured side of the hippocampus and in the cerebellum, although to a lesser extent. These data suggest that inflammation occurs early and persists for several days after ICH in humans and could be involved in the progression of ICH-induced secondary brain damage.
Collapse
Affiliation(s)
- He Wu
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin 150001, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
260
|
Lindsberg PJ, Strbian D, Karjalainen-Lindsberg ML. Mast cells as early responders in the regulation of acute blood-brain barrier changes after cerebral ischemia and hemorrhage. J Cereb Blood Flow Metab 2010; 30:689-702. [PMID: 20087366 PMCID: PMC2949160 DOI: 10.1038/jcbfm.2009.282] [Citation(s) in RCA: 137] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The inflammatory response triggered by stroke has been viewed as harmful, focusing on the influx and migration of blood-borne leukocytes, neutrophils, and macrophages. This review hypothesizes that the brain and meninges have their own resident cells that are capable of fast host response, which are well known to mediate immediate reactions such as anaphylaxis, known as mast cells (MCs). We discuss novel research suggesting that by acting rapidly on the cerebral vessels, this cell type has a potentially deleterious role in the very early phase of acute cerebral ischemia and hemorrhage. Mast cells should be recognized as a potent inflammatory cell that, already at the outset of ischemia, is resident within the cerebral microvasculature. By releasing their cytoplasmic granules, which contain a host of vasoactive mediators such as tumor necrosis factor-alpha, histamine, heparin, and proteases, MCs act on the basal membrane, thus promoting blood-brain barrier (BBB) damage, brain edema, prolonged extravasation, and hemorrhage. This makes them a candidate for a new pharmacological target in attempts to even out the inflammatory responses of the neurovascular unit, and to stabilize the BBB after acute stroke.
Collapse
Affiliation(s)
- Perttu Johannes Lindsberg
- Department of Neurology, Helsinki University Central Hospital, Haartmaninkatu 8, 00290 Helsinki, Finland.
| | | | | |
Collapse
|
261
|
Molecular insights and therapeutic targets for blood-brain barrier disruption in ischemic stroke: critical role of matrix metalloproteinases and tissue-type plasminogen activator. Neurobiol Dis 2010; 38:376-85. [PMID: 20302940 DOI: 10.1016/j.nbd.2010.03.008] [Citation(s) in RCA: 198] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Revised: 03/06/2010] [Accepted: 03/10/2010] [Indexed: 01/09/2023] Open
Abstract
Blood-brain barrier (BBB) disruption, mediated through matrix metalloproteinases (MMPs) and other mechanisms, is a critical event during ischemic stroke. Tissue plasminogen activator (tPA) is the only FDA-approved thrombolytic therapy for acute ischemic stroke, but the efficacy and safety of its therapeutic application are limited by narrow treatment time windows and side effects. Thus, there is a pressing need to develop combinational therapy that could offset tPA side effects and improve efficacy in clinical practice. Recent experimental studies indicate that tPA has previously unidentified functions in the brain beyond its well-established thrombolytic activity, which might contribute to tPA-related side effects through MMPs (mainly MMP-9) and several signaling pathways involved in LDL receptor-related protein (LRP), activated protein C (APC) and protease-activated receptor 1 (PAR-1), platelet-derived growth factor C (PDGF-C), and N-methyl-d-aspartate (NMDA) receptor. Therapeutic targeting of MMPs and/or tPA-related signaling pathways might offer promising new approaches to combination therapies for ischemic stroke. This review provides an overview of the relationship between structural components and function of the BBB/neurovascular unit with respect to ischemic stroke. We discuss how MMPs and tPA contribute to BBB disruption during ischemic stroke and highlight recent findings of molecular signaling pathways involved in neurotoxicity of tPA therapy.
Collapse
|
262
|
Jin R, Yang G, Li G. Inflammatory mechanisms in ischemic stroke: role of inflammatory cells. J Leukoc Biol 2010; 87:779-89. [PMID: 20130219 DOI: 10.1189/jlb.1109766] [Citation(s) in RCA: 1186] [Impact Index Per Article: 79.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Inflammation plays an important role in the pathogenesis of ischemic stroke and other forms of ischemic brain injury. Experimentally and clinically, the brain responds to ischemic injury with an acute and prolonged inflammatory process, characterized by rapid activation of resident cells (mainly microglia), production of proinflammatory mediators, and infiltration of various types of inflammatory cells (including neutrophils, different subtypes of T cells, monocyte/macrophages, and other cells) into the ischemic brain tissue. These cellular events collaboratively contribute to ischemic brain injury. Despite intense investigation, there are still numerous controversies concerning the time course of the recruitment of inflammatory cells in the brain and their pathogenic roles in ischemic brain injury. In this review, we provide an overview of the time-dependent recruitment of different inflammatory cells following focal cerebral I/R. We discuss how these cells contribute to ischemic brain injury and highlight certain recent findings and currently unanswered questions about inflammatory cells in the pathophysiology of ischemic stroke.
Collapse
Affiliation(s)
- Rong Jin
- Department of Neurosurgery, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, USA
| | | | | |
Collapse
|
263
|
Tejima E, Guo S, Murata Y, Arai K, Lok J, van Leyen K, Rosell A, Wang X, Lo EH. Neuroprotective effects of overexpressing tissue inhibitor of metalloproteinase TIMP-1. J Neurotrauma 2010. [PMID: 19469687 DOI: 10.1089/neu.2009-0959] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Accumulating data suggest that matrix metalloproteinases (MMPs) may be important mediators in the pathophysiology of acute brain injury after trauma or stroke. Here, we test the hypothesis that the endogenous tissue inhibitor of metalloproteinase (TIMP-1) is neuroprotective in vitro and in vivo. For in vitro studies, primary cortical neuronal cultures were subjected to hypoxia and reoxygenation. Treatment with recombinant TIMP-1 protein significantly decreased neuronal death. In vivo studies in models of brain trauma and stroke supported these cell culture results. After controlled cortical impact, 24-h MMP-9 levels were significantly reduced in transgenic mice overexpressing TIMP-1 compared to wild-type mice. And at 7 days post-trauma, brain lesion volumes were also significantly decreased by TIMP-1 overexpression as well. In a model of transient 2-h focal cerebral ischemia, MMP-9 levels were lower in TIMP-1 transgenic mice compared with wild-types. Correspondingly, blood-brain barrier leakage was ameliorated by TIMP-1 overexpression, and 24-h infarction volumes were also reduced. Taken together, these cell culture and in vivo data provide initial proof-of-principle that TIMP-1 is neuroprotective against traumatic and ischemic brain injury in mice.
Collapse
Affiliation(s)
- Emiri Tejima
- Program in Neuroscience, Harvard Medical School, Charlestown, Massachusetts, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
264
|
Tejima E, Guo S, Murata Y, Arai K, Lok J, van Leyen K, Rosell A, Wang X, Lo EH. Neuroprotective effects of overexpressing tissue inhibitor of metalloproteinase TIMP-1. J Neurotrauma 2010; 26:1935-41. [PMID: 19469687 DOI: 10.1089/neu.2009.0959] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Accumulating data suggest that matrix metalloproteinases (MMPs) may be important mediators in the pathophysiology of acute brain injury after trauma or stroke. Here, we test the hypothesis that the endogenous tissue inhibitor of metalloproteinase (TIMP-1) is neuroprotective in vitro and in vivo. For in vitro studies, primary cortical neuronal cultures were subjected to hypoxia and reoxygenation. Treatment with recombinant TIMP-1 protein significantly decreased neuronal death. In vivo studies in models of brain trauma and stroke supported these cell culture results. After controlled cortical impact, 24-h MMP-9 levels were significantly reduced in transgenic mice overexpressing TIMP-1 compared to wild-type mice. And at 7 days post-trauma, brain lesion volumes were also significantly decreased by TIMP-1 overexpression as well. In a model of transient 2-h focal cerebral ischemia, MMP-9 levels were lower in TIMP-1 transgenic mice compared with wild-types. Correspondingly, blood-brain barrier leakage was ameliorated by TIMP-1 overexpression, and 24-h infarction volumes were also reduced. Taken together, these cell culture and in vivo data provide initial proof-of-principle that TIMP-1 is neuroprotective against traumatic and ischemic brain injury in mice.
Collapse
Affiliation(s)
- Emiri Tejima
- Program in Neuroscience, Harvard Medical School, Charlestown, Massachusetts, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
265
|
Starke RM, Komotar RJ, Hwang BY, Hahn DK, Otten ML, Hickman ZL, Garrett MC, Sisti MB, Lavine SD, Meyers PM, Solomon RA, Connolly ES. Systemic Expression of Matrix Metalloproteinase-9 in Patients With Cerebral Arteriovenous Malformations. Neurosurgery 2010; 66:343-8; discussion 348. [DOI: 10.1227/01.neu.0000363599.72318.ba] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Abstract
OBJECTIVE
Increased expression angiogenic factors, such as matrix metalloproteinases (MMPs), are associated with the formation of cerebral arteriovenous malformations (AVMs). The objective of this study was to determine plasma levels of MMP-9 of patients with AVMs.
METHODS
Blood samples were drawn from 15 patients with AVMs before treatment, 24 hours postembolization, 24 hours postresection, and 30 days postresection. Blood samples were also obtained from 30 healthy controls. Plasma MMP-9 concentrations were measured via enzyme-linked immunosorbent assay.
RESULTS
The mean plasma MMP-9 level in AVM patients at baseline was significantly higher than in control patients: 108.04 ± 16.11 versus 41.44 ± 2.44 ng/mL, respectively. The mean plasma MMP-9 level 1 day after embolization increased to 172.35 ± 53.76 ng/mL, which was not significantly elevated over pretreatment levels. One day after resection, plasma MMP-9 levels increased significantly over pretreatment levels to 230.97 ± 51.00 ng/mL. Mean plasma MMP-9 concentrations 30 days after resection decreased to 92.8 ± 18.7 ng/mL, which was not different from pretreatment levels but was still significantly elevated over control levels. MMP-9 levels did not correlate with patient sex, age, presentation, or AVM size.
CONCLUSION
Plasma MMP-9 levels are significantly elevated over controls at baseline, increase significantly immediately after surgery, and decrease to pretreatment levels during follow-up.
Collapse
Affiliation(s)
- Robert M. Starke
- Department of Neurosurgery, University of Virginia, Charlottesville, Virginia (Starke)
| | - Ricardo J. Komotar
- Department of Neurosurgery, Columbia University, New York, New York (Komotar) (Hwang) (Otten) (Hickman) (Garrett) (Sisti) (Lavine) (Meyers) (Solomon) (Connolly)
| | - Brian Y. Hwang
- Department of Neurosurgery, Columbia University, New York, New York (Komotar) (Hwang) (Otten) (Hickman) (Garrett) (Sisti) (Lavine) (Meyers) (Solomon) (Connolly)
| | - David K. Hahn
- Department of Neurosurgery, Northwestern University, Chicago, Illinois (Hahn)
| | - Marc L. Otten
- Department of Neurosurgery, Columbia University, New York, New York (Komotar) (Hwang) (Otten) (Hickman) (Garrett) (Sisti) (Lavine) (Meyers) (Solomon) (Connolly)
| | - Zachary L. Hickman
- Department of Neurosurgery, Columbia University, New York, New York (Komotar) (Hwang) (Otten) (Hickman) (Garrett) (Sisti) (Lavine) (Meyers) (Solomon) (Connolly)
| | - Matthew C. Garrett
- Department of Neurosurgery, Columbia University, New York, New York (Komotar) (Hwang) (Otten) (Hickman) (Garrett) (Sisti) (Lavine) (Meyers) (Solomon) (Connolly)
| | - Michael B. Sisti
- Department of Neurosurgery, Columbia University, New York, New York (Komotar) (Hwang) (Otten) (Hickman) (Garrett) (Sisti) (Lavine) (Meyers) (Solomon) (Connolly)
| | - Sean D. Lavine
- Department of Neurosurgery, Columbia University, New York, New York (Komotar) (Hwang) (Otten) (Hickman) (Garrett) (Sisti) (Lavine) (Meyers) (Solomon) (Connolly)
| | - Philip M. Meyers
- Department of Neurosurgery, Columbia University, New York, New York (Komotar) (Hwang) (Otten) (Hickman) (Garrett) (Sisti) (Lavine) (Meyers) (Solomon) (Connolly)
| | - Robert A. Solomon
- Department of Neurosurgery, Columbia University, New York, New York (Komotar) (Hwang) (Otten) (Hickman) (Garrett) (Sisti) (Lavine) (Meyers) (Solomon) (Connolly)
| | - E. Sander Connolly
- Department of Neurosurgery, Columbia University, New York, New York (Komotar) (Hwang) (Otten) (Hickman) (Garrett) (Sisti) (Lavine) (Meyers) (Solomon) (Connolly)
| |
Collapse
|
266
|
Abstract
Microvessels and neurons respond rapidly and simultaneously in focal regions of ischaemic injury in such a way as to suggest that the responses could be coordinated. The ability of neurons to modulate cerebral blood flow in regions of activation results from neurovascular coupling. But little is known about the microvessel-to-neuron direction of the relationship. The presence and participation of intervening glial cells implies the association of microvessels, glia, and neurons in a 'neurovascular unit'. The interdependent functions of the cellular and matrix components of this theoretical unit have not been rigorously explored, except under conditions of injury where, for the most part, only single components or tissue samples have been studied. Whereas maintenance or timely re-establishment of flow reduces tissue and neuron injury in both humans and animal models, protection of neuron function in humans has not prevented the evolution of injury despite the inherent mechanisms of neurovascular coupling. However, occlusion of flow to the brain rapidly identifies regions of neuron-vascular vulnerability within the vascular territory-at-risk. These coalesce to become the mature ischaemic lesion. The failure, so far, of clinical trials of neuron protectant agents to achieve detectable tissue salvage could be explained by the vulnerability (and lack of protection) of essential components of the 'unit'. This presentation summarizes evidence and thoughts on this topic. These support the need to understand component interactions within the neurovascular unit.
Collapse
Affiliation(s)
- G J del Zoppo
- Department of Medicine, University of Washington, Seattle, WA 98104, USA.
| |
Collapse
|
267
|
Grossetete M, Phelps J, Arko L, Yonas H, Rosenberg GA. Elevation of matrix metalloproteinases 3 and 9 in cerebrospinal fluid and blood in patients with severe traumatic brain injury. Neurosurgery 2010; 65:702-8. [PMID: 19834375 DOI: 10.1227/01.neu.0000351768.11363.48] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Traumatic brain injury (TBI) causes an increase in matrix metalloproteinases (MMPs), which are associated with neuroinflammation, blood-brain barrier disruption, hemorrhage, and cell death. We hypothesized that patients with TBI have an increase in MMPs in ventricular cerebrospinal fluid (CSF) and plasma. METHODS Patients with TBI and a ventricular catheter were entered into the study. Samples of CSF and plasma were collected at the time of catheter placement and at 24 and 72 hours after admission. Seven TBI patients were entered into the study, with 6 having complete data for analysis. Only patients who had a known time of insult that fell within a 6-hour window from initial insult to ventriculostomy were accepted into the study. Control CSF came from ventricular fluid in patients undergoing shunt placement for normal pressure hydrocephalus. Both MMP-2 and MMP-9 were measured with gelatin zymography and MMP-3 with Western immunoblot. RESULTS We found a significant elevation in the levels of the latent form of MMP-9 (92-kD) in the CSF obtained at the time of arrival (P < 0.05). Elevated levels of MMP-2 were detected in plasma at 72 hours, but not in the CSF. Using albumin from both CSF and blood, we calculated the MMP-9 index, which was significantly increased in the CSF, indicating endogenous MMP production. Western immunoblot showed elevated levels of MMP-3 in CSF at all times measured, whereas MMP-3 was not detected in the CSF of normal pressure hydrocephalus. CONCLUSION We show that MMPs are increased in the CSF of TBI patients. Although the number of patients was small, the results were robust and clearly demonstrated increases in MMP-3 and MMP-9 in ventricular CSF in TBI patients compared with controls. Although these preliminary results will need to be replicated, we propose that MMPs may be important in blood-brain barrier opening and hemorrhage secondary to brain injury in patients.
Collapse
Affiliation(s)
- Mark Grossetete
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico 87131-0001, USA
| | | | | | | | | |
Collapse
|
268
|
Timing of serum active MMP-9 and MMP-2 levels in acute and subacute phases after spontaneous intracerebral hemorrhage. ACTA NEUROCHIRURGICA. SUPPLEMENT 2010; 106:137-40. [PMID: 19812936 DOI: 10.1007/978-3-211-98811-4_24] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Serum active matrix metalloproteinase (MMP)-9 and -2 levels and their tissue inhibitors TIMP-1 and -2 were measured in 28 patients with spontaneous intracerebral hemorrhage (SICH) at 24 h, 48 h and 7 days after bleeding. Perihematomal edema volume was calculated on non-enhanced computed tomography scans by using the formula AxBxC/2 at the same time points. Mean levels of serum active MMP-9 and MMP-2, as well as perihematomal edema volume, were significantly different over time (p < 0.0001). In comparison to values observed at 24 h, serum active MMP-9 mean concentrations increased at 48 h and reached their peak at 7 days, serum active MMP-2 mean levels progressively declined at 48 h and at 7 days, whereas perihematomal edema volume increased at 48 h and at 7 days. Perihematomal edema volume was positively correlated with active MMP-9 and MMP-2 at 24 h (p < 0.02 and p < 0.05, respectively) and with active MMP-9 at 48 h (p < 0.05), but was inversely correlated with active MMP-2 at 7 days (p < 0.02). These findings suggest a different involvement of active MMP-9 and MMP-2 in perihematomal-associated inflammatory response occurring in the transition from acute to subacute phases after SICH.
Collapse
|
269
|
Barr TL, Latour LL, Lee KY, Schaewe TJ, Luby M, Chang GS, El-Zammar Z, Alam S, Hallenbeck JM, Kidwell CS, Warach S. Blood-brain barrier disruption in humans is independently associated with increased matrix metalloproteinase-9. Stroke 2009; 41:e123-8. [PMID: 20035078 DOI: 10.1161/strokeaha.109.570515] [Citation(s) in RCA: 156] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Matrix metalloproteinases (MMP) may play a role in blood-brain barrier (BBB) disruption after ischemic stroke. We hypothesized that plasma concentrations of MMP-9 are associated with a marker of BBB disruption in patients evaluated for acute stroke. METHODS Patients underwent MRI on presentation and approximately 24 hours later. The MRI marker, termed hyperintense acute reperfusion injury marker (HARM), is gadolinium enhancement of cerebrospinal fluid on fluid-attenuated inversion recovery MRI. Plasma MMP-9 and tissue inhibitor of matrix metalloproteinase-1 were measured by enzyme-linked immunosorbent assay. Logistic regression models tested for predictors of HARM on 24-hour follow-up scans separately for MMP-9 and the ratio of MMP-9 to TIMP-1. RESULTS For the 41 patients enrolled, diagnoses were: acute ischemic cerebrovascular syndrome, 33 (80.6%); intracerebral hemorrhage, 6 (14.6%); stroke mimic, 1 (2.4%); and no stroke, 1 (2.4%). HARM was present in 17 (41.5%) patients. In model 1, HARM was associated with baseline plasma MMP-9 concentration (odds ratio [OR], 1.01; 95% confidence interval [CI], 1.001-1.019; P=0.033). In model 2, HARM was associated with the ratio of MMP-9 to tissue inhibitor of matrix metalloproteinase-1 (OR, 4.94; 95% CI, 1.27-19.14; P=0.021). CONCLUSIONS Baseline MMP-9 was a significant predictor of HARM at 24-hour follow-up, supporting the hypothesis that MMP-9 is associated with BBB disruption. If the association between MMP-9 and BBB disruption is confirmed in future studies, HARM may be a useful imaging marker to evaluate MMP-9 inhibition in ischemic stroke and other populations with BBB disruption.
Collapse
Affiliation(s)
- Taura L Barr
- National Institute of Nursing Research, Bethesda, Md, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
270
|
Dong X, Song YN, Liu WG, Guo XL. Mmp-9, a potential target for cerebral ischemic treatment. Curr Neuropharmacol 2009; 7:269-75. [PMID: 20514206 PMCID: PMC2811860 DOI: 10.2174/157015909790031157] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2009] [Revised: 09/22/2009] [Accepted: 10/05/2009] [Indexed: 11/27/2022] Open
Abstract
Matrix metalloproteinase-9 (MMP-9) which is a member of matrix metalloproteinases family that normally remodel the extracellular matrix, has been shown to play an important role in both animal models of cerebral ischemia and human stroke. The expression of MMP-9 is elevated after cerebral ischemia which is involved in accelerating matrix degradation, disrupting the blood-brain barrier, increasing the infarct size and relating to hemorrhagic transformation. Recently, many drugs, such as tetracycline derivatives, cyclooxygenase inhibitors, ACEI inhibitors and AT1 receptor blockers, etc., have been found to attenuate the elevated expression levels of MMP-9 after ischemia and to reduce the damage of cerebral ischemic. This article reviews the physiological features of MMP-9 and its important role in the genesis, propagation, and therapeutics of cerebral ischemic diseases.
Collapse
Affiliation(s)
- Xue Dong
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, No. 44 WenHuaXi Road, Jinan 250012, P.R. China
| | - Yu-Ning Song
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, No. 44 WenHuaXi Road, Jinan 250012, P.R. China
| | - Wei-Guo Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, No. 44 WenHuaXi Road, Jinan 250012, P.R. China
- Qianfoshan Hospital, Jinan 250014, P.R. China
| | - Xiu-Li Guo
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, No. 44 WenHuaXi Road, Jinan 250012, P.R. China
| |
Collapse
|
271
|
Liu R, Liu Q, He S, Simpkins JW, Yang SH. Combination therapy of 17beta-estradiol and recombinant tissue plasminogen activator for experimental ischemic stroke. J Pharmacol Exp Ther 2009; 332:1006-12. [PMID: 19952306 DOI: 10.1124/jpet.109.160937] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Thrombolysis with recombinant tissue plasminogen activator (rtPA) in ischemic stroke is limited by the increased risk of hemorrhage transformation due to blood-brain barrier breakdown. We determined the interaction of 17beta-estradiol (E2) and rtPA on activation of plasminogen system and matrix metalloproteinases (MMPs) in a transient middle cerebral artery occlusion (MCAO) model. Ovariectomized female rats were subjected to 1-h transient focal cerebral ischemia using a suture MCAO model. Ischemic lesion volume was significantly reduced with acute treatment of E2 despite of exogenous administration of rtPA. The expression and activation of urokinase (uPA), MMP2, and MMP9 were significantly increased in ischemic hemisphere after transient cerebral ischemia. Exogenous rtPA administration further enhanced expression and activation of uPA, MMP2, and MMP9, which was blocked by E2 treatment. We further determined the effect of combination therapy of E2 and rtPA in an embolic MCAO model. Although no protection was indicated upon acute treatment of E2 alone, combination treatment of E2 and rtPA provided protective action at 3 h after embolism. Collectively, the present study suggests that estrogen could be a candidate for combination therapy with rtPA to attenuate its side effect and hence expand its short therapeutic window for treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ran Liu
- Department of Pharmacology and Neuroscience, University of North Texas Health Science Center at Fort Worth, 3500 Camp Bowie Blvd., Fort Worth, TX 76107, USA
| | | | | | | | | |
Collapse
|
272
|
Lin HC, Song TY, Hu ML. S-Adenosylhomocysteine promotes the invasion of C6 glioma cells via increased secretion of matrix metalloproteinase-2 in murine microglial BV2 cells. Toxicol Sci 2009; 112:322-30. [PMID: 19770485 DOI: 10.1093/toxsci/kfp218] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
S-Adenosylhomocysteine (SAH) is a risk factor for many diseases, including tumor progression and neurodegenerative disease. In this study, we examined the hypothesis that SAH may indirectly enhance the invasion of C6 glioma cells by induction of matrix metalloproteinase-2 (MMP-2) secreted from the murine microglia BV2 cells. We obtained conditioned medium (CM) by incubating BV2 cells with SAH (1-50nM) for 24 h. We found that the SAH-containing CM (SAH-BV2-CM) strongly enhanced the invasiveness of C6 glioma cells and that this effect increased with increasing concentrations of SAH in the SAH-BV2-CM. The effect of CM could be attributed to its MMP-2 activity, as a result of increased protein and messenger RNA expression of MMP-2 in BV2 cells induced by SAH. In BV2 cells treated with SAH, the binding abilities of nuclear factor-kappa B (NF-kappaB) and stimulatory protein-1 (Sp1) to the MMP-2 promoter were increased, whereas the level of NF-kappaB inhibitor was decreased. In addition, SAH significantly increased the phosphorylation of extracellular signal-regulated kinase (ERK) and phosphatidylinositol-3-kinase/serine/threonine protein kinase (or protein kinase B) (PI3K/Akt) proteins but did not affect that of c-Jun NH2-terminal kinase or p38. Pretreatment of BV2 cells with an inhibitor specific for ERK (U0126) markedly abated the expression of ERK and MMP-2. Furthermore, SAH significantly and dose dependently decreased tissue inhibitor of metalloproteinase-2 (TIMP-2) in BV2 cells. Thus, SAH may induce the invasiveness of C6 glioma cells by decreased TIMP-2 expression and increased MMP-2 expression in BV2 cells. The latter effect is likely mediated through the ERK and PI3K/Akt pathways, with increased binding activities of NF-kappaB and Sp1 to the MMP-2 gene promoter.
Collapse
Affiliation(s)
- Hung-Chi Lin
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan, Republic of China
| | | | | |
Collapse
|
273
|
Cuadrado E, Rosell A, Penalba A, Slevin M, Alvarez-Sabín J, Ortega-Aznar A, Montaner J. Vascular MMP-9/TIMP-2 and neuronal MMP-10 up-regulation in human brain after stroke: a combined laser microdissection and protein array study. J Proteome Res 2009; 8:3191-7. [PMID: 19317417 DOI: 10.1021/pr801012x] [Citation(s) in RCA: 84] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Matrix Metalloproteinases (MMPs) play an important role in brain injury after ischemic stroke. In the present study, we aimed to assess the global expression of MMP-Family proteins in the human brain after stroke by using a combination of Searchlight Protein Array and Laser Microdissection to determine their cellular origin. This study demonstrated that MMP-1, MMP-2, MMP-3, MMP-8, MMP-9, MMP-10, MMP-13, and TIMP-1 were upregulated in the infarcted tissue compared to healthy control areas. Using laser microdissection we obtained specific neuronal and vascular populations from both infarcted and control areas. From these fractions, we showed that MMP-9 and TIMP-2 were highly produced in brain microvessels while MMP-10 was notably increased in neurons of the ischemic brain but not in healthy areas. These findings demonstrate a selective cell-dependent MMP secretion, opening the possibility of selectively targeting specific MMPs for neuroprotection or vasculoprotection following stroke.
Collapse
Affiliation(s)
- Eloy Cuadrado
- Neurovascular Research Laboratory, Neurovascular Unit, Department of Neurology, Universitat Autonoma de Barcelona, Institut de Recerca, Hospital Vall d'Hebron, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
274
|
Sunagawa S, Ichiyama T, Honda R, Fukunaga S, Maeba S, Furukawa S. Matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 in perinatal asphyxia. Brain Dev 2009; 31:588-93. [PMID: 18849127 DOI: 10.1016/j.braindev.2008.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2008] [Revised: 09/01/2008] [Accepted: 09/02/2008] [Indexed: 11/25/2022]
Abstract
Matrix metalloproteinase-9 (MMP-9) and tissue inhibitor of metalloproteinase-1 (TIMP-1) play important roles in the function of the blood-brain-barrier (BBB). We investigated the roles of MMP-9 and TIMP-1 in the pathogenesis of hypoxic-ischemic encephalopathy following perinatal asphyxia. Serum concentrations of MMP-9 and TIMP-1 were determined by ELISA in 12 neonates with perinatal asphyxia and 15 controls on the birth day and the next day. Serum MMP-9 concentrations in asphyxiated neonates with neurological sequelae (n=5) were significantly higher than concentration in asphyxiated neonates without sequelae (n=7) and controls on birth day (p=0.003 and p<0.001, respectively). The ratios of serum MMP-9/TIMP-1 on birth day in asphyxiated neonates with neurological sequelae were significantly higher than those in asphyxiated neonates without sequelae (p=0.048). There were no significant differences in the serum MMP-9 concentrations or the ratios of MMP-9/TIMP-1 between asphyxiated neonates with and without neurological sequelae on the day after birth. Our preliminary study suggests that serum MMP-9 levels on birth day are important for predicting neurological prognosis of neonates with asphyxia.
Collapse
Affiliation(s)
- Shinpei Sunagawa
- Department of Pediatrics, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | | | | | | | | | | |
Collapse
|
275
|
Hernández-Guillamon M, Delgado P, Ortega L, Pares M, Rosell A, García-Bonilla L, Fernández-Cadenas I, Borrell-Pagès M, Boada M, Montaner J. Neuronal TIMP-1 release accompanies astrocytic MMP-9 secretion and enhances astrocyte proliferation induced by beta-amyloid 25-35 fragment. J Neurosci Res 2009; 87:2115-25. [PMID: 19235898 DOI: 10.1002/jnr.22034] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The neuropathology of Alzheimer's disease (AD) is accompanied by an inflammatory response that includes neurodegeneration and glial reactivity. Tissue remodeling proteins, such as matrix metalloproteinases (MMPs) and their endogenous tissue inhibitors (TIMPs), are inflammatory mediators that might play a dual role in the AD brain. We aimed to investigate the effects of beta-amyloid (Abeta) on the MMP-9/TIMP-1 balance and its involvement in Abeta toxicity in neurons and glial cells. Our results demonstrate that the neurotoxic 25-35 Abeta fragment induces the activation of MMP-9 and the increase of proMMP-2/9 secretion and promotes the release of TIMP-1 in a mixed cortical neuroglial culture. The same treatments performed in pure neuronal or astrocytic cultures confirm that astroglial cells are the major source of MMP-9, whereas increased TIMP-1 levels have a neuronal origin. Moreover, 25-35 Abeta fragment not only induced a release of these molecules but also caused expressional changes in MMP-9 and TIMP-1, correlated with the neurotoxicity process. We also show that TIMP-1 promoted cell proliferation in a mixed neuroglial culture, and we confirm this effect in primary cultured astrocytes induced by rTIMP-1 and 25-35 Abeta. Because the proliferative effect caused by Abeta 25-35 was enhanced by the presence of TIMP-1, we suggest that the astroglial reactivity induced by chronic exposure of the peptide might be mediated in part by TIMP-1, which is secreted mainly by injured neurons. In conclusion, our data suggest that the Abeta 25-35 fragment stimulates the MMP-9-TIMP-1 pathway, promoting gliosis, in a self-defensive attempt to eliminate amyloid deposition from AD brains.
Collapse
Affiliation(s)
- Mar Hernández-Guillamon
- Neurovascular Research Laboratory, Institut de Recerca, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
276
|
Kassner A, Roberts TPL, Moran B, Silver FL, Mikulis DJ. Recombinant tissue plasminogen activator increases blood-brain barrier disruption in acute ischemic stroke: an MR imaging permeability study. AJNR Am J Neuroradiol 2009; 30:1864-9. [PMID: 19661169 DOI: 10.3174/ajnr.a1774] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Although thrombolytic therapy (recombinant tissue plasminogen activator [rtPA]) represents an important step forward in acute ischemic stroke (AIS) management, there is a clear need to identify high-risk patients. The purpose of this study was to investigate the role of quantitative permeability (KPS) MR imaging in patients with AIS treated with and without rtPA. We hypothesized that rtPA would increase KPS and that KPS MR imaging can be used to predict the risk of hemorrhagic transformation (HT). MATERIALS AND METHODS Thirty-six patients with AIS were examined within a mean of 3.6 hours of documented symptom onset. KPS MR imaging was performed as part of our AIS protocol. KPS coefficients in the stroke lesion were estimated for all patients, and the relationship between KPS and both HT and rtPA was investigated by using Student t tests. Receiver operating characteristic (ROC) curves were computed for predicting HT from KPS. RESULTS The occurrence rate of HT for patients who received rtPA and those who did not was 43% and 37%, respectively. Assessment of KPS in the lesion revealed significant differences between those who hemorrhaged and those who did not (P < .0001) as well as between rtPA-treated and untreated patients (P = .008). ROC analysis indicated a KPS threshold of 0.67 mL/100 g/min, with a sensitivity of 92% and a specificity of 78%. CONCLUSIONS The results of this study indicate that KPS is able to identify patients at higher risk of HT and may allow use of physiologic imaging rather than time from onset of symptoms to guide treatment decision.
Collapse
Affiliation(s)
- A Kassner
- Department of Diagnostic Imaging, Hospital for Sick Children, Toronto, Ontario, Canada.
| | | | | | | | | |
Collapse
|
277
|
Abstract
Serum biomarkers related to the cascade of inflammatory, hemostatic, glial and neuronal perturbations have been identifed to diagnose and characterize intracerebral hemorrhage and cerebral ischemia. Interpretation of most markers is confounded by their latent rise, blood-brain barrier effects, the heterogeneity of etiologies and the wide range of normal values, limiting their application for early diagnosis, lesion size estimation and long-term outcome prediction. Certain hemostatic and inflammatory constituents have been found to predict response to thrombolysis and worsening due to infarct progression and secondary hemorrhage, offering a potential role for improved treatment selection and individualization of therapy. Biomarkers will become increasingly relevant for developing targets for neuroprotective therapies, monitoring response to treatment and as surrogate end points for treatment trials.
Collapse
Affiliation(s)
- Matthew B Maas
- 175 Cambridge Street, Suite 300, Boston, MA 02114, USA, Tel.: +1 617 643 2713; ;
| | | |
Collapse
|
278
|
Klohs J, Baeva N, Steinbrink J, Bourayou R, Boettcher C, Royl G, Megow D, Dirnagl U, Priller J, Wunder A. In vivo near-infrared fluorescence imaging of matrix metalloproteinase activity after cerebral ischemia. J Cereb Blood Flow Metab 2009; 29:1284-92. [PMID: 19417756 DOI: 10.1038/jcbfm.2009.51] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Matrix metalloproteinases (MMPs) have been implicated in the pathophysiology of cerebral ischemia. In this study, we explored whether MMP activity can be visualized by noninvasive near-infrared fluorescence (NIRF) imaging using an MMP-activatable probe in a mouse model of stroke. C57Bl6 mice were subjected to transient middle cerebral artery occlusion (MCAO) or sham operation. Noninvasive NIRF imaging was performed 24 h after probe injection, and target-to-background ratios (TBRs) between the two hemispheres were determined. TBRs were significantly higher in MCAO mice injected with the MMP-activatable probe than in sham-operated mice and in MCAO mice that were injected with the nonactivatable probe as controls. Treatment with an MMP inhibitor resulted in significantly lower TBRs and lesion volumes compared to injection of vehicle. To test the contribution of MMP-9 to the fluorescence signal, MMP9-deficient (MMP9(-/-)) mice and wild-type controls were subjected to MCAO of different durations to attain comparable lesion volumes. TBRs were significantly lower in MMP9(-/-) mice, suggesting a substantial contribution of MMP-9 activity to the signal. Our study shows that MMP activity after cerebral ischemia can be imaged noninvasively with NIRF using an MMP-activatable probe, which might be a useful tool to study MMP activity in the pathophysiology of the disease.
Collapse
Affiliation(s)
- Jan Klohs
- Department of Experimental Neurology, Center for Stroke Research Berlin, Charité University Medicine Berlin, Charitéplatz 1, Berlin 10117, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
279
|
TYAGI NEETU, GILLESPIE WILLIAM, VACEK JONATHANC, SEN UTPAL, TYAGI SURESHC, LOMINADZE DAVID. Activation of GABA-A receptor ameliorates homocysteine-induced MMP-9 activation by ERK pathway. J Cell Physiol 2009; 220:257-66. [PMID: 19308943 PMCID: PMC2811271 DOI: 10.1002/jcp.21757] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hyperhomocysteinemia (HHcy) is a risk factor for neuroinflammatory and neurodegenerative diseases. Homocysteine (Hcy) induces redox stress, in part, by activating matrix metalloproteinase-9 (MMP-9), which degrades the matrix and leads to blood-brain barrier dysfunction. Hcy competitively binds to gamma-aminbutyric acid (GABA) receptors, which are excitatory neurotransmitter receptors. However, the role of GABA-A receptor in Hcy-induced cerebrovascular remodeling is not clear. We hypothesized that Hcy causes cerebrovascular remodeling by increasing redox stress and MMP-9 activity via the extracellular signal-regulated kinase (ERK) signaling pathway and by inhibition of GABA-A receptors, thus behaving as an inhibitory neurotransmitter. Hcy-induced reactive oxygen species production was detected using the fluorescent probe, 2'-7'-dichlorodihydrofluorescein diacetate. Hcy increased nicotinamide adenine dinucleotide phosphate-oxidase-4 concomitantly suppressing thioredoxin. Hcy caused activation of MMP-9, measured by gelatin zymography. The GABA-A receptor agonist, muscimol ameliorated the Hcy-mediated MMP-9 activation. In parallel, Hcy caused phosphorylation of ERK and selectively decreased levels of tissue inhibitors of metalloproteinase-4 (TIMP-4). Treatment of the endothelial cell with muscimol restored the levels of TIMP-4 to the levels in control group. Hcy induced expression of iNOS and decreased eNOS expression, which lead to a decreased NO bioavailability. Furthermore muscimol attenuated Hcy-induced MMP-9 via ERK signaling pathway. These results suggest that Hcy competes with GABA-A receptors, inducing the oxidative stress transduction pathway and leading to ERK activation.
Collapse
Affiliation(s)
- NEETU TYAGI
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - WILLIAM GILLESPIE
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - JONATHAN C. VACEK
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - UTPAL SEN
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - SURESH C. TYAGI
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - DAVID LOMINADZE
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
280
|
Maddahi A, Chen Q, Edvinsson L. Enhanced cerebrovascular expression of matrix metalloproteinase-9 and tissue inhibitor of metalloproteinase-1 via the MEK/ERK pathway during cerebral ischemia in the rat. BMC Neurosci 2009; 10:56. [PMID: 19497125 PMCID: PMC2700114 DOI: 10.1186/1471-2202-10-56] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Accepted: 06/04/2009] [Indexed: 12/30/2022] Open
Abstract
Background Cerebral ischemia is usually characterized by a reduction in local blood flow and metabolism and by disruption of the blood-brain barrier in the infarct region. The formation of oedema and opening of the blood-brain barrier in stroke is associated with enhanced expression of metalloproteinase-9 (MMP-9) and tissue inhibitor of metalloproteinase-1 (TIMP-1). Results Here, we found an infarct volume of 24.8 ± 2% and a reduced neurological function after two hours of middle cerebral artery occlusion (MCAO), followed by 48 hours of recirculation in rat. Immunocytochemistry and confocal microscopy revealed enhanced expression of MMP-9, TIMP-1, and phosphorylated ERK1/2 in the smooth muscle cells of the ischemic MCA and associated intracerebral microvessels. The specific MEK1/2 inhibitor U0126, given intraperitoneal zero or 6 hours after the ischemic event, reduced the infarct volume significantly (11.8 ± 2% and 14.6 ± 3%, respectively; P < 0.05), improved neurological function, normalized expression of phosphorylated ERK1/2, and reduced expression of MMP-9 and TIMP-1 in the vessel walls. Administration of U0126 12 hours after MCAO did not alter the expression of MMP-9. Immunocytochemistry showed no overlap in expression between MMP-9/TIMP-1 and the astrocyte/glial cell marker GFAP in the vessel walls. Conclusion These data are the first to show that the elevated vascular expression of MMP-9 and TIMP-1, associated with breakdown of the blood-brain barrier following focal ischemia, are transcriptionally regulated via the MEK/ERK pathway.
Collapse
Affiliation(s)
- Aida Maddahi
- Department of Clinical Sciences, Division of Experimental Vascular Research, Lund University, Lund, Sweden.
| | | | | |
Collapse
|
281
|
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; 87:668-76. [PMID: 18831008 DOI: 10.1002/jnr.21877] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Microvascular integrity is lost during cerebral ischemia. Detachment of the microvascular basement membrane (BM) from the astrocyte, as well as degradation of the BM, is responsible for the loss of microvascular integrity. However, their ultrastructural and temporal changes during cerebral ischemia are not well known. Male Sprague-Dawley rats were subjected to permanent middle cerebral artery occlusion (MCAO) for 1, 4, 8, 12, 16, 20, and 48 hr. By using transmission electron microscopy, the proportion of intact BM-astrocyte contacts and electron densities of the BM were measured from five randomly selected microvessels in the ischemic basal ganglia. Their temporal changes and associations with activities of the matrix metalloproteinases (MMPs) were investigated. The intact portion of the BM-astrocyte contacts was decreased significantly within 4 hr and was rarely observed at 48 hr after MCAO. Decreases in the electron density and degradation of the BM were significant 12 hr after MCAO. The intact BM-astrocyte contacts and the mean BM density showed a significant positive correlation (r = 0.784, P < 0.001). MMP-9 activity was correlated negatively with the intact BM-astrocyte contacts (r = -0.711, P < 0.001) and with the BM density (r = -0.538, P = 0.0016). The increase in MMP-9 coincided temporally with the loss of the BM-astrocyte contacts and a decrease in the BM density. Ultrastructural alterations occurring in the microvascular BM and its contacts with astrocyte endfeet were temporally associated in cerebral ischemia. Time courses of their alterations should be considered in the treatment targeted to the microvascular BM and its contact with astrocytes.
Collapse
Affiliation(s)
- Il Kwon
- Department of Neurology, National Core Research Center for Nanomedical Technology, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | |
Collapse
|
282
|
Sapienza P, Borrelli V, di Marzo L, Cavallaro A. MMP and TIMP alterations in asymptomatic and symptomatic severe recurrent carotid artery stenosis. Eur J Vasc Endovasc Surg 2009; 37:525-30. [PMID: 19297218 DOI: 10.1016/j.ejvs.2009.01.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Accepted: 01/22/2009] [Indexed: 10/21/2022]
Abstract
OBJECTIVES This study aimed to determine whether the plasma levels of matrix metalloproteinases (MMPs)-2 and -9 and their specific inhibitors (tissue inhibitors of metalloproteinases (TIMPs-1 and -2)) were altered in patients with symptomatic and asymptomatic, severe, recurrent carotid artery stenosis. PATIENTS Fifty-two patients (out of a total of 621) who had undergone successful carotid artery endarterectomy (CEA) between 1999 and 2003 and developed recurrent carotid artery stenosis (>/=70%) were included in the study. Restenosis was symptomatic in 23 patients and asymptomatic in 29 patients. METHODS Recurrent carotid artery stenosis was classified based on presentation, and as early-intermediate (6 months to 3 years) or late (>3 years). A detailed clinical history was taken and two blood samples were drawn from each patient to determine plasma levels of MMPs and TIMPs along with other biological parameters. Recurrent stenosis was confirmed with computed tomographic angiography. RESULTS Patients with symptomatic restenosis had significantly (p<0.001) higher active MMP-2 and -9 plasma values and significantly (p<0.001) lower TIMP-1 and -2 plasma values when compared to patients with asymptomatic restenosis. Plasma concentrations of active MMPs were higher and TIMPs lower in patients affected with late recurrent stenosis as compared to early-intermediate restenosis (p<0.001). No differences were recorded in latent MMP plasma values. Multivariate analysis showed that active MMP-2 and -9 were independent predictors of late recurrent carotid artery stenosis (p<0.03 and p<0.001, respectively). CONCLUSIONS Higher plasma concentrations of active MMP-2 and -9 were associated with an increased risk of carotid restenosis with plaque recurrence.
Collapse
Affiliation(s)
- P Sapienza
- Department of Surgery "Pietro Valdoni", University of Rome "La Sapienza", Policlinico Umberto I degrees , Viale del Policlinico 155, 00161 Rome, Italy
| | | | | | | |
Collapse
|
283
|
Abstract
Nitric oxide (NO) is an important messenger molecule in a variety of physiological systems. NO, a gas, is produced from L-arginine by different isoforms of nitric oxide synthase (NOS) and serves many normal physiologic purposes, such as promoting vasodilation of blood vessels and mediating communication between nervous system cells. In addition to its physiologic actions, free radical activity of NO can cause cellular damage through a phenomenon known as nitrosative stress. Here, we review the role of NO in health and disease, focusing on its role in function and dysfunction of the nervous system. Substantial evidence indicates that NO plays a key role in most common neurodegenerative diseases, and, although the mechanism of NO-mediated neurodegeneration remains uncertain, studies suggest several possibilities. NO has been shown to modify protein function by nitrosylation and nitrotyrosination, contribute to glutamate excitotoxicity, inhibit mitochondrial respiratory complexes, participate in organelle fragmentation, and mobilize zinc from internal stores. In this review, we discuss and analyze the evidence for each of these mechanisms in different neurodegenerative diseases and propose future directions for research of the role of NO in neurodegeneration.
Collapse
Affiliation(s)
- Andrew B Knott
- Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, 4000 Central Florida Blvd., Orlando, Florida, USA
| | | |
Collapse
|
284
|
Mechanisms and markers for hemorrhagic transformation after stroke. ACTA NEUROCHIRURGICA. SUPPLEMENT 2009; 105:173-8. [PMID: 19066105 DOI: 10.1007/978-3-211-09469-3_34] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Intracerebral hemorrhagic transformation is a multifactorial phenomenon in which ischemic brain tissue converts into a hemorrhagic lesion with blood vessel leakage. Hemorrhagic transformation can significantly contribute to additional brain injury after stroke. Especially threatening are the thrombolytic-induced hemorrhages after reperfusion therapy with tissue plasminogen activator (tPA), the only treatment available for ischemic stroke. In this context, it is important to understand its underlying mechanisms and identify early markers of hemorrhagic transformation, so that we can both search for new treatments as well as predict clinical outcomes in patients. In this review, we discuss the emerging mechanisms for hemorrhagic transformation after stroke, and briefly survey potential molecular, genetic, and neuroimaging markers that might be used for early detection of this challenging clinical problem.
Collapse
|
285
|
Immunohistochemical analysis of MMP-9, MMP-2 and TIMP-1, TIMP-2 expression in the central nervous system following infection with viral and bacterial meningitis. Folia Histochem Cytobiol 2009; 46:437-42. [PMID: 19141395 DOI: 10.2478/v10042-008-0058-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are capable of degrading components of the basal lamina of cerebral vessels, thereby disrupting the blood-brain barrier and inducing leukocyte recruitment. This study provides comprehensive information regarding the cell specificity of matrix metalloproteinases (MMP-2, MMP-9) and their binding tissue inhibitors (TIMP-1, TIMP-2) in the central nervous system during viral and bacterial meningitis. Specifically, we evaluated the immunoreactivity of MMPs and TIMPs in various cell types in brain parenchyma and meninges obtained from autopsy tissues. We found that a higher proportion of endothelial cells were positive for MMP-9 during meningitis when compared to controls. In addition, the immunoreactivity of MMP-9 decreased and the immunoreactivity of TIMP-1 increased in astrocytes upon infection. Furthermore, the results of this study revealed that mononuclear cells were highly immunoreactive for TIMP-1, TIMP-2 and MMP-9 during viral meningitis and that the expression of TIMPs in polymorphonuclear cells was even higher during bacterial meningitis. Taken together the results of this study indicated that the central nervous system resident cells and inflammatory infiltrates contribute to MMPs activity and that the expression patterns vary between cell types and in response to viral and bacterial meningitis.
Collapse
|
286
|
Amantea D, Nappi G, Bernardi G, Bagetta G, Corasaniti MT. Post-ischemic brain damage: pathophysiology and role of inflammatory mediators. FEBS J 2009; 276:13-26. [PMID: 19087196 DOI: 10.1111/j.1742-4658.2008.06766.x] [Citation(s) in RCA: 324] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neuroinflammatory mediators play a crucial role in the pathophysiology of brain ischemia, exerting either deleterious effects on the progression of tissue damage or beneficial roles during recovery and repair. Within hours after the ischemic insult, increased levels of cytokines and chemokines enhance the expression of adhesion molecules on cerebral endothelial cells, facilitating the adhesion and transendothelial migration of circulating neutrophils and monocytes. These cells may accumulate in the capillaries, further impairing cerebral blood flow, or extravasate into the brain parenchyma. Infiltrating leukocytes, as well as resident brain cells, including neurons and glia, may release pro-inflammatory mediators, such as cytokines, chemokines and oxygen/nitrogen free radicals that contribute to the evolution of tissue damage. Moreover, recent studies have highlighted the involvement of matrix metalloproteinases in the propagation and regulation of neuroinflammatory responses to ischemic brain injury. These enzymes cleave protein components of the extracellular matrix such as collagen, proteoglycan and laminin, but also process a number of cell-surface and soluble proteins, including receptors and cytokines such as interleukin-1beta. The present work reviewed the role of neuroinflammatory mediators in the pathophysiology of ischemic brain damage and their potential exploitation as drug targets for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Diana Amantea
- Department of Pharmacobiology, University of Calabria, Rende (CS), Italy.
| | | | | | | | | |
Collapse
|
287
|
Lee SR, Kim HY, Hong JS, Baek WK, Park JW. PPARγ agonist pioglitazone reduces matrix metalloproteinase-9 activity and neuronal damage after focal cerebral ischemia. Biochem Biophys Res Commun 2009; 380:17-21. [DOI: 10.1016/j.bbrc.2008.12.181] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2008] [Accepted: 12/28/2008] [Indexed: 11/16/2022]
|
288
|
Cuadrado E, Rosell A, Borrell-Pagès M, García-Bonilla L, Hernández-Guillamon M, Ortega-Aznar A, Montaner J. Matrix metalloproteinase-13 is activated and is found in the nucleus of neural cells after cerebral ischemia. J Cereb Blood Flow Metab 2009; 29:398-410. [PMID: 18985055 DOI: 10.1038/jcbfm.2008.130] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Matrix metalloproteinases (MMPs) have been implicated in the pathophysiology of ischemic stroke. In this study, we investigated the time course of gelatinolytic activation in a rat model of permanent ischemia. We observed an activation of MMPs as early as 30 mins after the ischemic insult, mainly in the nuclei of brain cells. Besides, we explored MMP-13 expression in brain samples of the animal model and stroke deceased patients. We observed an upregulation of active MMP-13 in rat brains (P<0.05) after 90 mins of cerebral ischemia. Human infarct/periinfarct samples also showed higher levels of active MMP-13 (P<0.05) compared with contralateral ones. Interestingly, we found that MMP-13 colocalized with 46-diamidino-2-phenyl indole signal by immunohistochemistry in both humans and rats, suggesting an intranuclear localization for MMP-13. Immunohistochemistry also revealed that MMP-13 was mainly produced by neurons, in both species, but also by oligodendrocytes in rats, and by astrocytes in humans. Finally we subjected a rat primary neuronal culture to oxygen and glucose deprivation (OGD) and we reproduced the nuclear translocation of MMP-13 in vitro. Nuclear extracts from cells confirmed upregulation of active MMP-13 after OGD (P<0.05). These results suggest that MMP-13 activation and its nuclear translocation is an early consequence of an ischemic stimulus.
Collapse
Affiliation(s)
- Eloy Cuadrado
- Department of Neurology, Institut de Recerca, Hospital Vall d'Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
289
|
Copin JC, Gasche Y. Effect of the duration of middle cerebral artery occlusion on the risk of hemorrhagic transformation after tissue plasminogen activator injection in rats. Brain Res 2008; 1243:161-6. [DOI: 10.1016/j.brainres.2008.09.025] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2008] [Revised: 09/12/2008] [Accepted: 09/12/2008] [Indexed: 11/29/2022]
|
290
|
Abstract
PURPOSE The objective of this study is to review the role of matrix metalloproteinases in intracerebral hemorrhage, which is associated with hypertension, head trauma and premature birth. MATERIALS AND METHODS A PubMed search of literature pertaining to this study was conducted in April 2008 using specific keyword search terms pertaining to intracerebral hemorrhage and matrix metalloproteinases. Some papers are not cited here as it is not possible to be all inclusive or due to the space limit from the journal. DISCUSSION The prognosis following ICH is more detrimental than that of ischemic strokes. Matrix metalloproteases have been implicated in the pathogenesis of brain damage following ICH. The goal of this review is to bring together recent diverse data concerning the roles of matrix metalloproteinases after intracerebral hemorrhage, which includes the role of matrix metalloproteinases in central nervous system, matrix metalloproteinases in animal models and humans of intracerebral hemorrhage, the relationship between matrix metalloproteinases and neuroinflammation, neuronal death, blood-brain barrier disruption and interaction with other molecules, as well as treatment of intracerebral hemorrhage with anti-matrix metalloproteinases agents. Besides deleterious roles in the acute period of intracerebral hemorrhage, some matrix metalloproteinases function in the later stages following intracerebral hemorrhage may have beneficial remodeling activity. CONCLUSION At present, the experimental data support the use of pharmacologic anti-matrix metalloproteinases strategies in the acute periods following intracerebral hemorrhage to alleviate injury.
Collapse
Affiliation(s)
- Mengzhou Xue
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alta, Canada
| | | |
Collapse
|
291
|
The role of p53 in brain edema after 24 h of experimental subarachnoid hemorrhage in a rat model. Exp Neurol 2008; 214:37-46. [DOI: 10.1016/j.expneurol.2008.07.006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2007] [Revised: 06/05/2008] [Accepted: 07/06/2008] [Indexed: 01/18/2023]
|
292
|
Abstract
The involvement of matrix metalloproteinase (MMP) 9 in methamphetamine-induced neurotoxicity was evaluated. Injection of mice with stimulant or toxic doses of methamphetamine upregulated MMP9 gene expression in the brain within 5 min. By 24 h, MMP9 gene expression returned to control levels in the stimulant-treated mice, but remained elevated in animals exposed to toxic doses of methamphetamine. Reductions in striatal dopamine levels, a marker of methamphetamine neurotoxicity, developed 1-7 days after methamphetamine exposure, but were not accompanied by concomitant changes in MMP9 gene expression. In MMP9 knockout mice, methamphetamine retained its ability to elicit neurotoxicity. The data suggest that MMP9 expression does not contribute to methamphetamine-induced neurotoxicity, and may instead be involved in remodeling of the nervous system.
Collapse
|
293
|
Wang X, Qin ZH, Shi H, Savitz SI, Qin AP, Jiang Y, Zhang HL. Protective effect of Ginkgolids (A+B) is associated with inhibition of NIK/IKK/IkappaB/NF-kappaB signaling pathway in a rat model of permanent focal cerebral ischemia. Brain Res 2008; 1234:8-15. [PMID: 18722355 DOI: 10.1016/j.brainres.2008.07.102] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2007] [Revised: 04/01/2008] [Accepted: 07/18/2008] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND PURPOSE We have previously reported that Ginkgolids which contain Ginkgolids A and B (Ginkgolids (A+B), GKAB) reduce infarct size in a rat model of focal ischemia. NF-kappaB-inducing kinase (NIK)-IkappaBalpha kinase (IKK) pathway plays an important role in activation of nuclear factor kappaB (NF-kappaB). A previous study demonstrated that Ginkgolid B inhibited lipopolysaccharide (LPS)- and platelet activating factor (PAF)-induced NF-kappaB activation in rat pleural polymorphonuclear granulocytes. However, little is known about the inhibitory mechanisms of Ginkgolids on the activation of NF-kappaB. The present study evaluated the effects of GKAB on NIK/IKK/IkappaB/NF-kappaB signaling pathway in a rat model of permanent focal cerebral ischemia. METHODS Rats were subjected to permanent middle cerebral artery occlusion (pMCAO) by intraluminal suture blockade. GKAB was injected intravenously (iv) immediately after ischemic onset. Western blot analysis was employed to determine alterations in IkappaBalpha, phosphorylated NIK (p-NIK) and phosphorylated IKKalpha (p-IKKalpha). Immunohistochemistry was used to confirm the nuclear translocation of NF-kappaB p65. RT-PCR was used to detect induction of NF-kappaB target gene c-Myc mRNA. RESULTS The results showed a brief increase in p-NIK levels after ischemia. GKAB blocked ischemia-induced increases in p-NIK and p-IKKalpha levels, and reversed the decline in IkappaBalpha levels. Ischemia-induced nuclear translocation of NF-kappaB p65 was attenuated by GKAB(.) GKAB also repressed the ischemia-induced increase in expression of NF-kappaB target gene c-Myc mRNA. CONCLUSIONS These findings suggest that GKAB-mediated neuroprotective effect against ischemia appears to be associated with blocking NF-kappaB activation by suppressing the NIK-IKK pathway.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Soochow University School of Medicine, Suzhou, China
| | | | | | | | | | | | | |
Collapse
|
294
|
Xue M, Fan Y, Liu S, Zygun DA, Demchuk A, Yong VW. Contributions of multiple proteases to neurotoxicity in a mouse model of intracerebral haemorrhage. ACTA ACUST UNITED AC 2008; 132:26-36. [PMID: 18772219 DOI: 10.1093/brain/awn215] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Proteases such as matrix metalloproteinases (MMPs) and thrombin are implicated in intracerebral haemorrhage (ICH) but their interactions amongst one another and interdependency remain to be defined. The latter is important since proteases acting through different mechanisms to inflict neurotoxicity would require separate targeting compared with proteases acting through the same cascade. We reported recently that MMP-9 and thrombin combined to promote neurotoxicity in ICH; however, as there was still substantial injury when both MMP-9 and thrombin were inhibited, we sought other factors that also contribute to ICH pathology. MMP-3, another member of the MMP family, has been correlated with poor prognosis in ICH in humans and it has been shown to increase rapidly after ICH in animals. Moreover, MMP-3 can convert the MMP-9 zymogen to its active form. Thus, we have examined whether MMP-3 is neurotoxic and addressed whether its potential effect in ICH is dependent on, or additional to, damage inflicted by MMP-9 and thrombin. We report that cultured neurons are killed by MMP-3 and that neuronal death is most marked when all three proteases, MMP-3, MMP-9 and thrombin, are combined. In vivo, the injection of autologous blood into the right striatum to produce ICH injury resulted in MMP-3 expression within 3 h. The blood-induced lesion and neuronal death was significantly reduced in MMP-3 or MMP-9 null mice compared with wild-type counterparts, and MMP-3 and -9 double null mice had even less brain damage. Significantly, pathological destruction after ICH was least in MMP-3 and -9 double null mice treated with a thrombin antagonist, hirudin. These results provide insights into molecules that inflict neurotoxicity in ICH and demonstrate that multiple proteases would need to be targeted simultaneously to successfully reduce ICH neurotoxicity.
Collapse
Affiliation(s)
- Mengzhou Xue
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | | | | | | | | | | |
Collapse
|
295
|
Diverse roles of matrix metalloproteinases and tissue inhibitors of metalloproteinases in neuroinflammation and cerebral ischemia. Neuroscience 2008; 158:983-94. [PMID: 18621108 DOI: 10.1016/j.neuroscience.2008.06.025] [Citation(s) in RCA: 412] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2008] [Revised: 06/04/2008] [Accepted: 06/08/2008] [Indexed: 12/15/2022]
Abstract
Regulation of the extracellular matrix by proteases and protease inhibitors is a fundamental biological process for normal growth, development and repair in the CNS. Matrix metalloproteinases (MMPs) and the tissue inhibitors of metalloproteinases (TIMPs) are the major extracellular-degrading enzymes. Two other enzyme families, a disintegrin and metalloproteinase (ADAM), and the serine proteases, plasminogen/plasminogen activator (P/PA) system, are also involved in extracellular matrix degradation. Normally, the highly integrated action of these enzyme families remodels all of the components of the matrix and performs essential functions at the cell surface involved in signaling, cell survival, and cell death. During the inflammatory response induced in infection, autoimmune reactions and hypoxia/ischemia, abnormal expression and activation of these proteases lead to breakdown of the extracellular matrix, resulting in the opening of the blood-brain barrier (BBB), preventing normal cell signaling, and eventually leading to cell death. There are several key MMPs and ADAMs that have been implicated in neuroinflammation: gelatinases A and B (MMP-2 and -9), stromelysin-1 (MMP-3), membrane-type MMP (MT1-MMP or MMP-14), and tumor necrosis factor-alpha converting enzyme (TACE). In addition, TIMP-3, which is bound to the cell surface, promotes cell death and impedes angiogenesis. Inhibitors of metalloproteinases are available, but balancing the beneficial and detrimental effects of these agents remains a challenge.
Collapse
|
296
|
Delayed matrix metalloproteinase inhibition reduces intracerebral hemorrhage after embolic stroke in rats. Exp Neurol 2008; 213:196-201. [PMID: 18590727 DOI: 10.1016/j.expneurol.2008.05.022] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2008] [Revised: 05/02/2008] [Accepted: 05/29/2008] [Indexed: 11/21/2022]
Abstract
Hemorrhagic transformation (HT) and brain edema are life-threatening complications of recombinant tissue plasminogen activator (rt-PA)-induced reperfusion after ischemic stroke. The risk of HT limits the therapeutic window for reperfusion to 3 h after stroke onset. Pre-treatment with matrix metalloproteinase (MMP) inhibitors reduces HT and cerebral edema in experimental stroke. However, whether a delayed therapeutic intervention would be beneficial is unknown. In this study, 215 male Sprague-Dawley rats were subjected to embolic stroke and 75 rats were included in the final analysis. The animals were treated with the MMP inhibitor p-aminobenzoyl-gly-pro-D-leu-D-ala-hydroxamate before or after 3 or 6 h of ischemia. Animals were monitored for reperfusion and received rt-PA 6 h after ischemia onset. The results at 24 h showed that MMP inhibition 3 h after ischemia significantly decreased the degree of brain edema (17% of hemispheric enlargement in the treated group versus 24% in controls, P=0.018), reduced the risk (OR=0.163; 95% CI: 0.029 to 0.953) and gravity (0.09 versus 0.19 mg of parenchymal hemoglobin, P=0.02) of intracerebral hemorrhage, and improved neurological outcome (20% of the treated animals had a slight deficit; all of the controls had a bad outcome, P<0.05). Delaying MMP inhibition to 6 h after ischemia restricted the beneficial role of the treatment to a reduction in the risk of parenchymal hemorrhage (OR=0.242; 95% CI: 0.060 to 0.989). Our results confirm the involvement of MMPs in HT and support the possibility of extending the therapeutic window for thrombolysis in stroke by administering a broad-spectrum MMP inhibitor after the onset of ischemia.
Collapse
|
297
|
Delgado P, Cuadrado E, Rosell A, Álvarez-Sabín J, Ortega-Aznar A, Hernández-Guillamón M, Penalba A, Molina CA, Montaner J. Fas System Activation in Perihematomal Areas After Spontaneous Intracerebral Hemorrhage. Stroke 2008; 39:1730-4. [DOI: 10.1161/strokeaha.107.500876] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Pilar Delgado
- From the Neurovascular Research Laboratory and Stroke Unit, Departments of Neurology (P.D., E.C., A.R., J.A.-S., M.H.-G., A.P., C.A.M., J.M.) and Pathology (A.O.-A.), Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Eloy Cuadrado
- From the Neurovascular Research Laboratory and Stroke Unit, Departments of Neurology (P.D., E.C., A.R., J.A.-S., M.H.-G., A.P., C.A.M., J.M.) and Pathology (A.O.-A.), Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Rosell
- From the Neurovascular Research Laboratory and Stroke Unit, Departments of Neurology (P.D., E.C., A.R., J.A.-S., M.H.-G., A.P., C.A.M., J.M.) and Pathology (A.O.-A.), Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - José Álvarez-Sabín
- From the Neurovascular Research Laboratory and Stroke Unit, Departments of Neurology (P.D., E.C., A.R., J.A.-S., M.H.-G., A.P., C.A.M., J.M.) and Pathology (A.O.-A.), Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Arantxa Ortega-Aznar
- From the Neurovascular Research Laboratory and Stroke Unit, Departments of Neurology (P.D., E.C., A.R., J.A.-S., M.H.-G., A.P., C.A.M., J.M.) and Pathology (A.O.-A.), Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mar Hernández-Guillamón
- From the Neurovascular Research Laboratory and Stroke Unit, Departments of Neurology (P.D., E.C., A.R., J.A.-S., M.H.-G., A.P., C.A.M., J.M.) and Pathology (A.O.-A.), Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Penalba
- From the Neurovascular Research Laboratory and Stroke Unit, Departments of Neurology (P.D., E.C., A.R., J.A.-S., M.H.-G., A.P., C.A.M., J.M.) and Pathology (A.O.-A.), Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Carlos A. Molina
- From the Neurovascular Research Laboratory and Stroke Unit, Departments of Neurology (P.D., E.C., A.R., J.A.-S., M.H.-G., A.P., C.A.M., J.M.) and Pathology (A.O.-A.), Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Joan Montaner
- From the Neurovascular Research Laboratory and Stroke Unit, Departments of Neurology (P.D., E.C., A.R., J.A.-S., M.H.-G., A.P., C.A.M., J.M.) and Pathology (A.O.-A.), Hospital Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|
298
|
Henning EC, Latour LL, Warach S. Verification of enhancement of the CSF space, not parenchyma, in acute stroke patients with early blood-brain barrier disruption. J Cereb Blood Flow Metab 2008; 28:882-6. [PMID: 18091756 PMCID: PMC5257002 DOI: 10.1038/sj.jcbfm.9600598] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Enhancement on post-contrast fluid-attenuated inversion recovery (FLAIR) images after acute stroke has been attributed to early blood-brain barrier disruption. Using an estimate of parenchymal volume fraction and the apparent diffusion coefficient (ADC), we investigated the relative contributions of cerebral spinal fluid (CSF) and parenchyma to enhancement seen on postcontrast FLAIR. Enhancing regions were found to have low parenchymal volume fractions and high ADC values, approaching that of pure CSF. These findings suggest that contrast enhancement on FLAIR occurs predominately in the CSF space, not parenchyma.
Collapse
Affiliation(s)
- Erica C Henning
- Section on Stroke Diagnostics and Therapeutics, Stroke Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | |
Collapse
|
299
|
Expression of cell adhesion proteins and proteins related to angiogenesis and fatty acid metabolism in benign, atypical, and anaplastic meningiomas. J Neurooncol 2008; 89:73-87. [PMID: 18418552 DOI: 10.1007/s11060-008-9588-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2007] [Accepted: 03/28/2008] [Indexed: 02/05/2023]
Abstract
Most meningiomas are benign tumours of arachnoidal origin, although a small number have high proliferative rates and invasive properties which complicate complete surgical resection and are associated with increased recurrence rates. Few prognostic indicators exist for meningiomas and further research is necessary to identify factors that influence tumour invasion, oedema and recurrence. Paraffin sections from 25 intracranial meningiomas were analysed for expression of the proteins vascular endothelial growth factor (VEGF), VEGF receptors Flt1 and Flk1, E-cadherin, metalloproteinases 2 and 9 (MMP2, MMP9), CD44, receptor for hyaluronic acid-mediated motility (RHAMM), hyaluronic acid (HA), CD45, cyclooxygenase 2 (COX2), brain fatty acid binding protein (BFABP), Ki67, and proliferating cell nuclear antigen (PCNA). Correlations among protein expression were found for several markers of proliferation (Ki67, PCNA, MI) and microvessel density (MVD). COX2 expression increased with increasing with tumour grade and correlated with Ki67, PCNA, MI, MVD, and BFABP. BFABP expression also correlated with Ki67 and PCNA expression. Relationships were also identified among angiogenic factors (VEGF, Flt1, Flk1) and proliferation markers. Oedema was found to correlate with MMP9 expression and MMP9 also correlated with proliferation markers. No correlations were found for MMP2, E-cadherin, or CD44 in meningiomas. In conclusion Ki67, PCNA, MI, MVD, BFABP, and COX2 were significantly correlated with meningioma tumour grade and with each other. These findings, by correlating both intracellular fatty acid transport and eicosanoid metabolism with tumour proliferation, as determined by Ki67 labelling and mitotic index, suggest fatty acids are involved in the progression of meningiomas.
Collapse
|
300
|
Haddad M, Beray-Berthat V, Coqueran B, Palmier B, Szabo C, Plotkine M, Margaill I. Reduction of hemorrhagic transformation by PJ34, a poly(ADP-ribose)polymerase inhibitor, after permanent focal cerebral ischemia in mice. Eur J Pharmacol 2008; 588:52-7. [PMID: 18468597 DOI: 10.1016/j.ejphar.2008.04.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 03/28/2008] [Accepted: 04/03/2008] [Indexed: 11/26/2022]
Abstract
Hemorrhagic transformation is an aggravating event that occurs in 15 to 43% of patients suffering from ischemic stroke. This phenomenon due to blood-brain barrier breakdown appears to be mediated in part by matrix metalloproteinases (MMPs) among which MMP-2 and MMP-9 could be particularly involved. Recent experimental studies demonstrated that post-ischemic MMP-9 overexpression is regulated by poly(ADP-ribose)polymerase (PARP). In this context, our study aimed to evaluate the effect of PJ34 (N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-2-(N,N-dimethylamino)acetamide), a potent PARP inhibitor, on MMP-2 and MMP-9 levels and on hemorrhagic transformations in a model of permanent focal cerebral ischemia in mice. PJ34 (6.25-12.5 mg/kg, i.p.) was given at the time of ischemia onset and 4 h later. Hemorrhagic transformations, divided into microscopic and macroscopic hemorrhages, were counted 48 h after ischemia on 12 coronal brain slices. Microscopic and macroscopic hemorrhages were respectively reduced by 38% and 69% with 6.25 mg/kg PJ34. The anti-hemorrhagic effect of PJ34 was associated with a 57% decrease in MMP-9 overexpression assessed by gelatin zymography. No increase in MMP-2 activity was observed after ischemia in our model. The vascular protection achieved by PJ34 was associated with a reduction in the motor deficit (P<0.05) and in infarct volume (-31%, P<0.01). In conclusion, our study demonstrates for the first time that PJ34 reduces hemorrhagic transformations after cerebral ischemia. Thus this PARP inhibitor exhibits both anti-hemorrhagic and neuroprotective effects that may be of valuable interest for the treatment of stroke.
Collapse
Affiliation(s)
- Marianne Haddad
- Equipe de recherche "Pharmacologie de la Circulation Cérébrale" (EA 2510), Université Paris Descartes, Faculté des Sciences Pharmaceutiques et Biologiques, Paris, France
| | | | | | | | | | | | | |
Collapse
|