Src Kinase Inhibition Improves Acute Outcomes After Experimental Intracerebral Hemorrhage

Share Embed


Descripción

Src Kinase Inhibition Improves Acute Outcomes After Experimental Intracerebral Hemorrhage Timothy D. Ardizzone, PhD; Xinhua Zhan, MD, PhD; Brad P. Ander, PhD; Frank R. Sharp, MD Background and Purpose—The mechanisms by which intracerebral hemorrhages produce changes of blood flow and metabolism, cell death, and behavioral abnormalities are complex. In this study, we begin to test the hypothesis that intracerebral hemorrhage activates Src kinases that phosphorylate other molecules to produce cell injury and behavioral deficits after intracerebral hemorrhage (ICH). Methods—ICH was produced in adult Sprague Dawley rats by direct injection of autologous blood (50 ␮L) into striatum. Src kinase activity, glucose hypermetabolic areas around the ICH, TUNEL-stained cells, and apomorphine-induced rotational behaviors were assessed in animals with ICH pretreated with the Src kinase inhibitor, PP1, or with vehicle. Results—PP1 (3 mg/kg) blocked increases of Src kinase activity (5-fold) at 3 hours after ICH. PP1 also blocked the areas of glucose hypermetabolism and decreased the numbers of TUNEL-stained cells surrounding the ICH at 24 hours. Finally, apomorphine-induced (1 mg/kg) rotation at 24 hours after ICH was markedly attenuated by previous treatment with PP1 (3 mg/kg intraperitoneal). Conclusions—PP1 decreases Src kinase activation, glucose metabolic activation, cell death, and behavioral abnormalities after ICH in striatum of adult rats. It is hypothesized that intracerebral hemorrhage, possibly via thrombin activation of protease-activated receptors, activates Src that phosphorylates NMDA receptors, matrix metalloproteinases, and other proteins that mediate injury after ICH. (Stroke. 2007;38:1621-1625.) Key Words: apoptosis 䡲 behavior 䡲 intracerebral hemorrhage 䡲 Src-kinase 䡲 striatum

R

ecent studies of experimental intracerebral hemorrhage (ICH) showed decreased expression of mRNA for some molecules, including Akt and PI3K.1,2 In contrast, expression of genes related to nitric oxide increased including GTP cyclohydrolase (4.1-fold), arginase (4.2-fold), and inducible nitric oxide synthase (NOS2, 15-fold). Importantly for this study, one of the most upregulated genes was a nonreceptor kinase, a Src-family kinase Lyn, that increased expression 21-fold.1,2 Src family kinases may be relevant for the injury that occurs after ICH for a number of reasons. Src potentiates NMDA receptor function through direct phosphorylation of tyrosine residues in the NR2A subunit of the NMDA receptor.3 Thrombin activates Src.4 Finally, Src phosphorylates and regulates a number of molecules including matrix metalloproteinases, HIF, and HIF target genes including vascular endothelial growth factor (VEGF), occluding, and other blood– brain barrier proteins that may be important in producing brain edema and brain injury after ICH.5–17 Clot formation appears to damage the perihematoma brain in part via edema and apoptosis.18,19 These could be mediated by thrombin activation of Src, because thrombin mediates the acute brain edema after ICH.20,21 Src could mediate thrombininduced edema and injury by Src increases of HIF target genes

including VEGF,5,22–24 by Src activation of matrix metalloproteinases,4 and by Src modulation of proteins like occludin in tight junctions of the blood– brain barrier.12,15 The increases of HIF in brain after ICH and thrombin injection25 appear to be mediated by Src,26,27 and HIF induction of VEGF can increase vascular permeability.4 Src activation of matrix metalloproteinases can also increase vascular permeability.6,7,28,29 Based on these data we hypothesize that Src is important for the development of injury, glucose hypermetabolism, edema formation, and apoptosis in peri-hematoma brain.30 –33 Therefore, the current study examined the effect of ICH on Src kinase activity; the effect of the Src antagonist PP1 on ICH-induced glucose hypermetabolism; the effect of NMDA antagonist and Src antagonist on TUNEL-stained in perihematoma brain; and the effects of the Src antagonist PP1 on behavioral function after acute experimental ICH in rats.

Materials and Methods Animals Sprague-Dawley male rats (250 to 300 grams; Charles River Labs, Wilmington, Mass) were used. Generally, there were 6 animals per group for each of the studies described, with 32 animals used for the behavioral studies.

Received November 28, 2006; accepted December 14, 2006. From Department of Neurology and the Medical Investigation of Neurodevelopmental Diseases Institute, University of California at Davis, Sacramento, Calif. Correspondence to Dr Frank R. Sharp, Medical Investigation of Neurodevelopmental Diseases Institute and Department of Neurology, University of California at Davis Medical Center, 2805 50th St, Rm 2416, Sacramento, CA 95817. E-mail [email protected] or [email protected] © 2007 American Heart Association, Inc. Stroke is available at http://www.strokeaha.org

DOI: 10.1161/STROKEAHA.106.478966

1621 Downloaded from http://stroke.ahajournals.org/ by guest on July 2, 2015

1622

Stroke

May 2007

Figure 1. Src kinase activity. Adult Sprague Dawley rats were anesthetized and a 50-␮L volume of blood or saline was injected into the left striatum. Animals were euthanized 3 hours later. The striatum with the hemorrhage (hemorrhage, 3 hours), the contralateral striatum, and saline-injected striatum were dissected (n⫽6 separate animals for each group). Some animals had the Src kinase inhibitor PP1 injected intraperitoneally before the hemorrhage (PP1, hemorrhage, 3 hours). Src kinase activity was measured as described in Materials and Methods. The y axis shows Src kinase activity expressed as a fold change from control (1⫽control levels; 5⫽5-fold increase in activity). Hemorrhage (hemorrhage, 3 hours) increased Src kinase activity (*P⬍0.05) compared with the contralateral striatum and the saline-injected striatum; and PP1 blocked this effect (**P⬍0.05). Hem indicates hemorrhage.

Surgery: ICH Model ICH was produced by local injection of 50 ␮L of autologous blood into the striatum of anesthetized adult male rats as previously described.30 The methods are similar to other experimental studies of brain hemorrhage in rodents.34 –38

Src Kinase Activity

Figure 2. The Src inhibitor PP1 attenuates hypermetabolism in perihematoma brain after ICH. Rats were either pretreated with vehicle (24 hours, hemorrhage; n⫽6) or pretreated with PP1 (24 hours, hemorrhage⫹PP1, 3.0 mg/kg; n⫽6) 15 minutes before injection of blood into the striatum. Twenty-four hours after ICH, the rats were injected with [14C] 2-deoxyglucose and euthanized 30 minutes later by decapitation. The brains were rapidly frozen and sectioned in a cryostat. The sections were dried on a hot plate and the sections were exposed to film for 5 to 7 days. The autoradiographs were scanned and analyzed with MCID elite software. There was a significant reduction in the % hypermetabolic area at all 3 locations from Bregma (1.6, 0.6, and ⫺0.4) for PP1 (24 hours, hemorrhage⫹PP1) vs saline (24 hours, hemorrhage) and control (*P⬍0.05 for PP1). Hem indicates hemorrhage.

Apomorphine-Induced Turning Behavior Animals had ICH, and had vehicle (0.9% saline), an intermediate dose of PP1 (1.5 mg/kg), or higher dose PP1 (3 mg/kg) administered intraperitoneally 30 minutes before the ICH. At 24 hours after the ICH surgery, rats were administered apomorphine (1 mg/kg, intraperitoneally). Apomorphine induced significant turning toward the side of the ICH. The numbers of turns per 5-minute interval were counted for each animal over a period of 1 hour (Figure 4). Differences were assessed using a nonparametric Kruskal–Wallis test.

Three hours after the injection of blood or saline, rats were euthanized and the ipsilateral and contralateral striatum were dissected, homogenized, and protein-isolated. Src protein was immunoprecipitated with a c-Src antibody (Delta Biolabs), which produced a single band on Western blots (not shown). The immunoprecipitated c-Src kinase was assayed for activity using the Tyrosine Kinase Activity Assay Kit (Chemicon). Differences between the 4 groups were assessed with an ANOVA and post hoc Schefe (Figure 1).

Autoradiography and Data Analysis Local cerebral glucose utilization was measured using [14C]-2 deoxyglucose (1.3 mCi/kg; 55 mCi/mmol; American Radiolabeled Chemicals) 24 hours after ICH as previously described (Figure 2).30

TUNEL Staining To assess cell death, animals were injected either with vehicle (0.9% saline, intraperitoneal), MK801 (1 mg/kg, intraperitoneal), or with PP1 (3 mg/kg, intraperitoneal) 30 minutes before producing the ICH. After 24 hours animals were euthanized and TUNEL staining was performed on brain sections using methods published from our laboratory39,40 and others.19,31,41– 43 For quantification, the TUNELstained cells around a hemorrhage were counted using the optical dissector method. Hemorrhage is manifested by hemoglobin/pink/ red staining of the striatum. The average number of TUNEL-stained cells were calculated for each animal and differences between the groups assessed with an ANOVA and post hoc Schefe (Figure 3).

Figure 3. Decrease in TUNEL positive cells after ICH by pretreatment with the glutamate receptor antagonist, MK801, or the Src kinase inhibitor, PP1. Rats were either pretreated with vehicle (24 hours, hemorrhage) or injected with MK801 (1 mg/kg; 24 hours, hemorrhage⫹MK801) or PP1 (1.5 mg/kg; 24 hours, hemorrhage⫹PP1) before injection of blood into the striatum (n⫽6 per group). The rats were euthanized 24 hours later and the brains were rapidly removed and frozen. The brains were sectioned on a cryostat and the slices were TUNEL-stained. The data represent the averages from 6 animals in each group (*P⬍0.05 for MK801 or PP1 treatment compared with 24-hour hemorrhage/vehicle). Hem indicates hemorrhage.

Downloaded from http://stroke.ahajournals.org/ by guest on July 2, 2015

Ardizzone et al

Src Kinase Inhibition Improves ICH

1623

turning at most times after apomorphine administration (P⬍0.05; PP1 3 mg/kg versus vehicle; no drug at time points 2 to 7; Kruskal–Wallis followed by post hoc Tukey).

Discussion

Figure 4. Inhibition of Src kinase activity blocks apomorphineinduced rotational behavior after ICH. Blood was injected into the left striatum of rats. One group of rats was pretreated with saline vehicle (no drug, n⫽14) and the 2 other groups were pretreated with the Src antagonist PP1 (1.5 and 3.0 mg/kg intraperitoneal, n⫽9 for each group) 15 minutes before surgery. One day (24 hours) after the surgery, the rats were injected with apomorphine (1 mg/kg) and rotational behavior was measured for 1 hour. Location of the hemorrhage was confirmed by examining brain sections through the striatum after the experiment.

Results Src kinase activity assays showed similar activity in the striatum injected with saline and in striatum contralateral to the blood injection (Figure 1). Injection of unclotted blood into striatum increased Src kinase activity ⬎5-fold at 3 hours after the blood injection (*P⬍0.05; hemorrhage 3 hours compared with contralateral and to saline; Figure 1). Injection of PP1 30 minutes before the ICH completely blocked the increase of Src kinase activity produced by hemorrhage (**P⬍0.05; PP1 hemorrhage 3 hours versus hemorrhage 3 hours). At 1 day (24 hours) after the ICH, local cerebral glucose metabolism is increased in the regions surrounding the ICH. Quantification of these areas showed glucose hypermetabolic areas at 1.6, 0.6, and ⫺0.4 mm anterior and posterior to bregma at 24 hours after hemorrhage (Figure 2). Treatment of animals with PP1 (3 mg/kg) 30 minutes before the ICH almost completely blocks these glucose hypermetabolic areas at 24 hours after hemorrhage (*P⬍0.05, 24-hour hemorrhage⫹PP1 at each level of bregma versus 24-hour hemorrhage; ANOVA with post hoc Schefe; Figure 2). One day after ICH there were significant numbers of TUNEL-stained cells that surrounded the hemorrhage (Figure 3). Administration of either MK801 (1 mg/kg, intraperitoneal) or of PP1 (3 mg/kg, intraperitoneal) 30 minutes before the ICH markedly decreased the numbers of TUNEL-stained cells (*P⬍0.05, 24-hour hemorrhage⫹MK801 versus 24-hour Hem; *P⬍0.05, 24-hour hemorrhage⫹PP1 versus 24-hour Hem; Figure 3). There were not significant differences in the numbers of TUNEL-stained cells in ICH animals treated with MK801 compared with PP1 (Figure 3). After ICH and the administration of apomorphine, animals turned repeatedly in the direction of the ICH, with the turning being maximal at ⬇10 to 20 minutes after the apomorphine, and decreasing to baseline by 1 hour (Figure 4). Administration of PP1 produced a dose-related reduction in the turning, with the larger dose (PP1, 3 mg/kg) markedly decreasing the

The major finding of this study is that Src kinase inhibition improves several acute outcome measures after experimental ICH. Specifically, ICH markedly activates Src kinase activity, and inhibiting Src kinases decreases areas of glucose hypermetabolism, decreases TUNEL-positive cells around the hemorrhage, and decreases apomorphine-induced turning after ICH. The data are consistent with the hypothesis that Src kinases mediate, at least in part, glucose metabolic changes, cell death, and behavioral deficits after ICH. Thrombin mediates acute edema in brain after ICH because thrombin inhibitors prevent ICH-induced acute brain edema.37,44 Although thrombin is a serine protease that is essential in the blood coagulation cascade, it also acts on thrombin receptors, called protease activated receptors (PAR), PAR-1, PAR-3, and PAR-4.45 Activation of the PARs leads to activation of specific intracellular kinases, including Src kinases, with several studies showing that thrombin activates Src via PAR thrombin receptors.4,21 Thus, ICH activation of thrombin at PAR receptors could activate Src kinases. Src kinases have previously been shown to be of importance in several cerebrovascular diseases. In a mouse model of ischemic stroke, Src inhibition decreased edema and infarct size by blocking the vascular permeability effect of VEGF.5 The Src kinase inhibitor PP2 decreased infarct volumes and improved neurological outcomes after middle cerebral artery occlusion.46 In a subarachnoid hemorrhage model, PP1, the same Src family kinase inhibitor used in this study, reduced blood– brain barrier permeability, brain edema, and mortality, and decreased the phosphorylation of VEGF and MAPKs.47 This study concluded that Src contributed to injury after subarachnoid hemorrhage via VEGF and MAPK pathways.47 Our previous study showed increased cerebral glucose utilization at 3 hours and 8 hours after ICH.30 Blocking NMDA or AMPA glutamate receptors blocked increased glucose metabolism.30 These data coupled with studies showing that pp60-Src (Src) potentiates the function of NMDA receptors through direct phosphorylation of the NR2A subunit,13 anchoring NMDA receptors48 and phosphorylation of the scaffolding protein PSD95 at the postsynaptic density of NMDA receptors,49 suggest that blocking Src might block NMDA receptor activation and, in turn, block the ICHinduced increases of glucose metabolism in this study.30 Alternatively, because Src phosphorylates many targets, Src might modulate glucose metabolism after ICH by phosphorylating other molecules, perhaps even on non-neuronal cells. Previous studies have demonstrated TUNEL-positive cells around ICH.19,31,32,42,50 –54 Similar TUNEL-positive cells occur in subarachnoid hemorrhage models.55 The TUNELpositive cells around ICH can be decreased by caspase inhibitors,19 minocycline,33 memantine,56 FK506,57 knockdown of tumor necrosis factor,58 adenosine A2A activator CGS 21680,59 tauroursodeoxycholic acid,60 15d-prostaglandin J2 that activates PPAR␥,54 and erythropoietin.61 The current study adds to this list by showing that glutamate

Downloaded from http://stroke.ahajournals.org/ by guest on July 2, 2015

1624

Stroke

May 2007

receptor blockade with MK801 and Src kinase blocker can also decrease the TUNEL-positive cells around an ICH. NMDA receptors are potentiated by thrombin receptor activation that causes Src-mediated phosphorylation of NR2A subunits.13 NMDA receptors are directly linked to Src via an adapter protein to produce NMDA (NR2a)-NADH dehydrogenase subunit 2-Src complexes.48 Src kinases regulate glutamate release from synaptic vesicles because PP2 can block glutamate release.62 Thus, it is not surprising that blocking either Src or NMDA receptors decreases cell death around an ICH. Taken together, these data indicate that Src, glutamate, and inflammatory mediators contribute to cell death around ICH. In these studies, any given compound reduces, but does not eliminate, cell death. It would be of interest to determine whether a combination of agents might eliminate the cell death. A previous study demonstrated that intrastriatal hemorrhages in adult rats were associated with ipsilateral rotational behavior at 1 to 70 days after high-dose apomorphine.63 Similarly, administration of high-dose amphetamine also induces rotational behavior with striatal ICH.64 Both of these studies are consistent with higher dopamine levels and dopamine activation in the intact striatum that produces turning toward the ICH injected striatum. As noted, many neurotransmitter receptors are downregulated in the ICH striatum, including the mRNA for the dopamine D2 receptor that decreased by ⬎25-fold in the ipsilateral striatum at 24 hours after ICH.1,2 Thus, one potential mechanism for the improvement of behavioral function after ICH with the Src kinase inhibitor would be blocking Src downregulation of D2 dopamine receptors. An alternative mechanism for improving the rotational behavior after striatal ICH might be related to the edema produced. Although not measured in these studies, experimental ICH results in reproducible acute edema that is maximal at 1 to 3 days in rat models.37 Thrombin activation of Src could mediate this edema, because Src kinases phosphorylate HIF, and HIF target proteins like VEGF, metalloproteinases, and blood– brain barrier proteins including occludin. Src-mediated changes of function of VEGF, matrix metalloproteinases, occludin, and other blood– brain barrier proteins could increase vascular permeability at the site of the ICH and impair function of the ipsilateral striatum. Blocking these permeability changes with the Src kinase antagonist might improve striatal function and decrease rotational behavior after ICH. This initial study does not directly address the mechanism by which ICH activates Src kinases. We postulate that ICH-related clotting leads to thrombin activation, which in turn cleaves PAR receptors to activate Src kinases. The thrombin/PAR receptor-mediated Src kinase activation would produce the changes observed in this study. Future studies will be necessary to support the hypothesis that the ICHmediated changes of Src activation are mediated by thrombin activation of PAR receptors.

Sources of Funding This work was supported by grants from the National Institutes of Neurological Diseases and Stroke branch of the National Institutes of Health (NS054652, NS028167, and NS054652).

Disclosures None.

References 1. Tang Y, Lu A, Aronow BJ, Wagner KR, Sharp FR. Genomic responses of the brain to ischemic stroke, intracerebral haemorrhage, kainate seizures, hypoglycemia, and hypoxia. Eur J Neurosci. 2002;15:1937–1952. 2. Lu A, Tang Y, Ran R, Ardizzone TL, Wagner KR, Sharp FR. Brain genomics of intracerebral hemorrhage. J Cereb Blood Flow Metab. 2006; 26:230 –252. 3. Manzerra P, Behrens MM, Canzoniero LM, Wang XQ, Heidinger V, Ichinose T, Yu SP, Choi DW. Zinc induces a src family kinase-mediated up-regulation of NMDA receptor activity and excitotoxicity. Proc Natl Acad Sci U S A. 2001;98:11055–11061. 4. Thomas SM, Brugge JS. Cellular functions regulated by src family kinases. Annu Rev Cell Dev Biol. 1997;13:513– 609. 5. Paul R, Zhang ZG, Eliceiri BP, Jiang Q, Boccia AD, Zhang RL, Chopp M, Cheresh DA, Schwartzberg PL, Hood JD, Leng J. Src deficiency or blockade of src activity in mice provides cerebral protection following stroke selective requirement for src kinases during VEGF-induced angiogenesis and vascular permeability. Nat Med. 2001;7:222–227. 6. Sato H, Kita M, Seiki M. V-Src activates the expression of 92-kda type iv collagenase gene through the ap-1 site and the gt box homologous to retinoblastoma control elements: a mechanism regulating gene expression independent of that by inflammatory cytokines. J Biol Chem. 1993;268: 23460–23468. 7. Onodera S, Nishihira J, Iwabuchi K, Koyama Y, Yoshida K, Tanaka S, Minami A. Macrophage migration inhibitory factor up-regulates matrix metalloproteinase-9 and -13 in rat osteoblasts: relevance to intracellular signaling pathways. J Biol Chem. 2002;277:7865–7874. 8. Zhang Q, Thomas SM, Xi S, Smithgall TE, Siegfried JM, Kamens J, Gooding WE, Grandis JR. Src family kinases mediate epidermal growth factor receptor ligand cleavage, proliferation, and invasion of head and neck cancer cells. Cancer Res. 2004;64:6166 – 6173. 9. Shin EY, Ma EK, Kim CK, Kwak SJ, Kim EG. Src/erk but not phospholipase d is involved in keratinocyte growth factor-stimulated secretion of matrix metalloprotease-9 and urokinase-type plasminogen activator in snu-16 human stomach cancer cell. J Cancer Res Clin Oncol. 2002;128: 596 – 602. 10. Ernst M, Inglese M, Scholz GM, Harder KW, Clay FJ, Bozinovski S, Waring P, Darwiche R, Kay T, Sly P, Collins R, Turner D, Hibbs ML, Anderson GP, Dunn AR. Constitutive activation of the Src family kinase Hck results in spontaneous pulmonary inflammation and an enhanced innate immune response. J Exp Med. 2002;196:589 – 604. 11. Guerrero J, Santibanez JF, Gonzalez A, Martinez J. EGF receptor transactivation by urokinase receptor stimulus through a mechanism involving src and matrix metalloproteinases. Exp Cell Res. 2004;292:201–208. 12. Kale G, Naren AP, Sheth P, Rao RK, Basuroy S, Kuppuswamy D, Balasubramanian S, Ray RM. Tyrosine phosphorylation of occludin attenuates its interactions with zo-1, zo-2, and zo-3 expression of kinaseinactive c-src delays oxidative stress-induced disassembly and accelerates calcium-mediated reassembly of tight junctions in the caco-2 cell monolayer. Biochem Biophys Res Commun. 2003;302:324 –329. 13. Liu Y, Zhang G, Gao C, Hou X. NMDA receptor activation results in tyrosine phosphorylation of nmda receptor subunit 2a(nr2a) and interaction of pyk2 and Src with nr2a after transient cerebral ischemia and reperfusion. Brain Res. 2001;909:51–58. 14. Pedram A, Razandi M, Levin ER. Deciphering vascular endothelial cell growth factor/vascular permeability factor signaling to vascular permeability: inhibition by atrial natriuretic peptide. J Biol Chem. 2002;277:44385–44398. 15. Meyer TN, Schwesinger C, Ye J, Denker BM, Nigam SK. Reassembly of the tight junction after oxidative stress depends on tyrosine kinase activity. J Biol Chem. 2001;276:22048 –22055. 16. Gomez S, del Mont Llosas M, Verdu J, Roura S, Lloreta J, Fabre M, Garcia de Herreros A. Independent regulation of adherens and tight junctions by tyrosine phosphorylation in caco-2 cells. Biochim Biophys Acta. 1999;1452:121–132. 17. Lee SW, Kim WJ, Choi YK, Song HS, Son MJ, Gelman IH, Kim YJ, Kim KW, Achen MG, Clauss M, Schnurch H, Risau W. SSECKS regulates angiogenesis and tight junction formation in blood-brain barrier the nonreceptor tyrosine kinase Lyn is localised in the developing murine blood-brain barrier. Nat Med. 2003;9:900 –906. 18. Xi G, Wagner KR, Keep RF, Hua Y, de Courten-Myers GM, Broderick JP, Brott TG, Hoff JT, Muizelaar JP. Role of blood clot formation on

Downloaded from http://stroke.ahajournals.org/ by guest on July 2, 2015

Ardizzone et al

19.

20.

21. 22. 23.

24.

25.

26.

27.

28.

29.

30.

31.

32.

33.

34.

35.

36. 37. 38. 39.

40.

41. 42.

43.

early edema development after experimental intracerebral hemorrhage. Stroke. 1998;29:2580 –2586. Matsushita K, Meng W, Wang X, Asahi M, Asahi K, Moskowitz MA, Lo EH. Evidence for apoptosis after intercerebral hemorrhage in rat striatum. J Cereb Blood Flow Metab. 2000;20:396 – 404. Xi G, Keep RF, Hua Y, Xiang J, Hoff JT. Attenuation of thrombininduced brain edema by cerebral thrombin preconditioning. Stroke. 1999; 30:1247–1255. Ferrell JE Jr, Martin GS. Platelet tyrosine-specific protein phosphorylation is regulated by thrombin. Mol Cell Biol. 1988;8:3603–3610. Sharp FR, Bernaudin M. HIF1 and oxygen sensing in the brain. Nat Rev Neurosci. 2004;5:437– 448. Eliceiri BP, Paul R, Schwartzberg PL, Hood JD, Leng J, Cheresh DA. Selective requirement for src kinases during vegf-induced angiogenesis and vascular permeability. Mol Cell. 1999;4:915–924. Liu Y, Cox SR, Morita T, Kourembanas S, Kern FG, McLeskey SW, Zhang L, Kurebayashi J, Ding IY, Kharbanda S, Chen D, Miller D, Cullen K. Hypoxia regulates vascular endothelial growth factor gene expression in endothelial cells: identification of a 5⬘ enhancer transfected mcf-7 cells as a model for breast-cancer progression. Circ Res. 1995;77:638–643. Jiang Y, Wu J, Keep RF, Hua Y, Hoff JT, Xi G. Hypoxia-inducible factor-1alpha accumulation in the brain after experimental intracerebral hemorrhage. J Cereb Blood Flow Metab. 2002;22:689 – 696. Jiang BH, Agani F, Passaniti A, Semenza GL. V-Src induces expression of hypoxia-inducible factor 1 (HIF-1) and transcription of genes encoding vascular endothelial growth factor and enolase 1: involvement of HIF-1 in tumor progression. Cancer Res. 1997;57:5328 –5335. Karni R, Dor Y, Keshet E, Meyuhas O, Levitzki A. Activated pp60c-src leads to elevated hypoxia-inducible factor (HIF)-1alpha expression under normoxia. J Biol Chem. 2002;277:42919 – 42925. Vincenti MP, Coon CI, White LA, Barchowsky A, Brinckerhoff CE. Src-related tyrosine kinases regulate transcriptional activation of the interstitial collagenase gene, MMP-1, in interleukin-1-stimulated synovial fibroblasts. Arthritis Rheum. 1996;39:574 –582. Rosenberg GA, Estrada EY, Dencoff JE. Matrix metalloproteinases and timps are associated with blood-brain barrier opening after reperfusion in rat brain. Stroke. 1998;29:2189 –2195. Ardizzone TD, Lu A, Wagner KR, Tang Y, Ran R, Sharp FR. Glutamate receptor blockade attenuates glucose hypermetabolism in perihematomal brain after experimental intracerebral hemorrhage in rat. Stroke. 2004;35: 2587–2591. Nakashima K, Yamashita K, Uesugi S, Ito H. Temporal and spatial profile of apoptotic cell death in transient intracerebral mass lesion of the rat. J Neurotr. 1999;16:143–151. Qureshi AI, Suri MF, Ostrow PT, Kim SH, Ali Z, Shatla AA, Guterman LR, Hopkins LN. Apoptosis as a form of cell death in intracerebral hemorrhage. Neurosurgery. 2003;52:1041–1048. Power C, Henry S, Del Bigio MR, Larsen PH, Corbett D, Imai Y, Yong VW, Peeling J. Intracerebral hemorrhage induces macrophage activation and matrix metalloproteinases. Ann Neurol. 2003;53:731–742. Deinsberger W, Vogel J, Fuchs C, Auer LM, Kuschinsky W, Boker DK. Fibrinolysis and aspiration of experimental intracerebral hematoma reduces the volume of ischemic brain in rats. Neurol Res. 1999;21:517–523. Del Bigio MR, Yan HJ, Buist R, Peeling J. Experimental intracerebral hemorrhage in rats: magnetic resonance imaging and histopathological correlates. Stroke. 1996;27:2312–2319; discussion 2319 –2320. Hoff JT, Xi G. Brain edema from intracerebral hemorrhage. Acta Neurochir Suppl. 2003;86:11–15. Xi G, Keep RF, Hoff JT. Pathophysiology of brain edema formation. Neurosurg Clin North Am. 2002;13:371–383. Andaluz N, Zuccarello M, Wagner KR. Experimental animal models of intracerebral hemorrhage. Neurosurg Clin North Am. 2002;13:385–393. Honkaniemi J, Massa SM, Breckinridge M, Sharp FR. Global ischemia induces apoptosis-associated genes in hippocampus. Brain Res Mol Brain Res. 1996;42:79 – 88. Lu A, Ran R, Parmentier-Batteur S, Nee A, Sharp FR. Geldanamycin induces heat shock proteins in brain and protects against focal cerebral ischemia. J Neurochem. 2002;81:355–364. Gong C, Boulis N, Qian J, Turner DE, Hoff JT, Keep RF. Intracerebral hemorrhage-induced neuronal death. Neurosurgery. 2001;48:875– 882. Hickenbottom SL, Grotta JC, Strong R, Denner LA, Aronowski J. Nuclear factor-kappab and cell death after experimental intracerebral hemorrhage in rats. Stroke. 1999;30:2477–2478. Xue M, Del Bigio MR. Acute tissue damage after injections of thrombin and plasmin into rat striatum. Stroke. 2001;32:2164 –2169.

Src Kinase Inhibition Improves ICH

1625

44. Xi G, Reiser G, Keep RF. The role of thrombin and thrombin receptors in ischemic, hemorrhagic and traumatic brain injury:Deleterious or protective? J Neurochem. 2003;84:3–9. 45. Coughlin SR. Thrombin signalling and protease-activated receptors. Nature. 2000;407:258 –264. 46. Lennmyr F, Ericsson A, Gerwins P, Akterin S, Ahlstrom H, Terent A. Src family kinase-inhibitor PP2 reduces focal ischemic brain injury. Acta Neurol Scand. 2004;110:175–179. 47. Kusaka G, Ishikawa M, Nanda A, Granger DN, Zhang JH. Signaling pathways for early brain injury after subarachnoid hemorrhage. J Cereb Blood Flow Metab. 2004;24:916 –925. 48. Gingrich JR, Pelkey KA, Fam SR, Huang Y, Petralia RS, Wenthold RJ, Salter MW. Unique domain anchoring of src to synaptic nmda receptors via the mitochondrial protein NADH dehydrogenase subunit 2. Proc Natl Acad Sci U S A. 2004;101:6237– 6242. 49. Kalia LV, Salter MW. Interactions between Src family protein tyrosine kinases and PSD-95. Neuropharmacology. 2003;45:720 –728. 50. Qureshi AI, Ling GS, Khan J, Suri MF, Miskolczi L, Guterman LR, Hopkins LN. Quantitative analysis of injured, necrotic, and apoptotic cells in a new experimental model of intracerebral hemorrhage. Crit Care Med. 2001;29:152–157. 51. Karwacki Z, Kowianski P, Dziewatkowski J, Domaradzka-Pytel B, Ludkiewcz B, Wojcik S, Narkiewicz O, Morys J. Apoptosis in the course of experimetal intracerebral haemorrhage in the rat. Folia Morphol (Warsz). 2005;64:248 –252. 52. Xue M, Del Bigio MR. Injections of blood, thrombin, and plasminogen more severely damage neonatal mouse brain than mature mouse brain. Brain Pathol. 2005;15:273–280. 53. Wagner KR, Dean C, Beiler S, Bryan DW, Packard BA, Smulian AG, Linke MJ, de Courten-Myers GM. Plasma infusions into porcine cerebral white matter induce early edema, oxidative stress, pro-inflammatory cytokine gene expression and DNA fragmentation: implications for white matter injury with increased blood-brain-barrier permeability. Curr Neurovasc Res. 2005;2:149 –155. 54. Zhao X, Zhang Y, Strong R, Grotta JC, Aronowski J. 15d-prostaglandin J2 activates peroxisome proliferator-activated receptor-gamma, promotes expression of catalase, and reduces inflammation, behavioral dysfunction, and neuronal loss after intracerebral hemorrhage in rats. J Cereb Blood Flow Metab. 2006;26:811– 820. 55. Matz PG, Fujimura M, Chan PH. Subarachnoid hemolysate produces DNA fragmentation in a pattern similar to apoptosis in mouse brain. Brain Res. 2000;858:312–319. 56. Lee ST, Chu K, Jung KH, Kim J, Kim EH, Kim SJ, Sinn DI, Ko SY, Kim M, Roh JK. Memantine reduces hematoma expansion in experimental intracerebral hemorrhage, resulting in functional improvement. J Cereb Blood Flow Metab. 2006;26:536 –544. 57. Peeling J, Yan HJ, Corbett D, Xue M, Del Bigio MR. Effect of fk-506 on inflammation and behavioral outcome following intracerebral hemorrhage in rat. Exp Neurol. 2001;167:341–347. 58. Mayne M, Ni W, Yan HJ, Xue M, Johnston JB, Del Bigio MR, Peeling J, Power C. Antisense oligodeoxynucleotide inhibition of tumor necrosis factor-alpha expression is neuroprotective after intracerebral hemorrhage. Stroke. 2001;32:240 –248. 59. Mayne M, Fotheringham J, Yan HJ, Power C, Del Bigio MR, Peeling J, Geiger JD. Adenosine a2a receptor activation reduces proinflammatory events and decreases cell death following intracerebral hemorrhage. Ann Neurol. 2001;49:727–735. 60. Rodrigues CM, Sola S, Nan Z, Castro RE, Ribeiro PS, Low WC, Steer CJ. Tauroursodeoxycholic acid reduces apoptosis and protects against neurological injury after acute hemorrhagic stroke in rats. Proc Natl Acad Sci U S A. 2003;100:6087– 6092. 61. Lee ST, Chu K, Sinn DI, Jung KH, Kim EH, Kim SJ, Kim JM, Ko SY, Kim M, Roh JK. Erythropoietin reduces perihematomal inflammation and cell death with enos and stat3 activations in experimental intracerebral hemorrhage. J Neurochem. 2006;96:1728 –1739. 62. Wang SJ. A role for src kinase in the regulation of glutamate release from rat cerebrocortical nerve terminals. Neuroreport. 2003;14:1519 –1522. 63. Chesney JA, Kondoh T, Conrad JA, Low WC. Collagenase-induced intrastriatal hemorrhage in rats results in long-term locomotor deficits. Stroke. 1995;26:317. 64. Imamura N, Hida H, Aihara N, Ishida K, Kanda Y, Nishino H, Yamada K. Neurodeg. Neurodegeneration of substantia nigra accompanied with macrophage/microglia infiltration after intrastriatal hemorrhage. Neurosci Res. 2003;46:289 –298.

Downloaded from http://stroke.ahajournals.org/ by guest on July 2, 2015

Src Kinase Inhibition Improves Acute Outcomes After Experimental Intracerebral Hemorrhage Timothy D. Ardizzone, Xinhua Zhan, Brad P. Ander and Frank R. Sharp Stroke. 2007;38:1621-1625; originally published online March 29, 2007; doi: 10.1161/STROKEAHA.106.478966 Stroke is published by the American Heart Association, 7272 Greenville Avenue, Dallas, TX 75231 Copyright © 2007 American Heart Association, Inc. All rights reserved. Print ISSN: 0039-2499. Online ISSN: 1524-4628

The online version of this article, along with updated information and services, is located on the World Wide Web at: http://stroke.ahajournals.org/content/38/5/1621

Permissions: Requests for permissions to reproduce figures, tables, or portions of articles originally published in Stroke can be obtained via RightsLink, a service of the Copyright Clearance Center, not the Editorial Office. Once the online version of the published article for which permission is being requested is located, click Request Permissions in the middle column of the Web page under Services. Further information about this process is available in the Permissions and Rights Question and Answer document. Reprints: Information about reprints can be found online at: http://www.lww.com/reprints Subscriptions: Information about subscribing to Stroke is online at: http://stroke.ahajournals.org//subscriptions/

Downloaded from http://stroke.ahajournals.org/ by guest on July 2, 2015

Lihat lebih banyak...

Comentarios

Copyright © 2017 DATOSPDF Inc.