Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2018

Open Access 01-12-2018 | Research

Cell permeable HMGB1-binding heptamer peptide ameliorates neurovascular complications associated with thrombolytic therapy in rats with transient ischemic stroke

Authors: Miaodan Li, Shumin Chen, Xue Shi, Chenfei Lyu, Yongfang Zhang, Miaoqin Tan, Chen Wang, Nailiang Zang, Xiaoxi Liu, Yafang Hu, Jiangang Shen, Liang Zhou, Yong Gu

Published in: Journal of Neuroinflammation | Issue 1/2018

Login to get access

Abstract

Background

Blood–brain barrier (BBB) breakdown and inflammatory responses are the major causes of tissue-type plasminogen activator (tPA)-induced hemorrhagic transformation (HT), while high-mobility group box 1 (HMGB1) exacerbates inflammatory damage to BBB during the process of brain ischemia/reperfusion. This study aimed to investigate the change of HMGB1 after thrombolytic therapy and whether blocking HMGB1 could ameliorate the neurovasculature complications secondary to tPA treatment in stroke rats.

Methods

Sera from acute stroke patients and rats with thrombolytic therapy were collected to investigate HMGB1 secretion. Male Sprague-Dawley rats with 2 h or 4.5 h middle cerebral artery occlusion were continuously infused with tPA followed by administration of membrane permeable HMGB1-binding heptamer peptide (HBHP). The mortality rate, neurological score, HT, brain swelling, BBB permeability, and inflammatory factors were determined.

Results

The results revealed that HMGB1 levels were elevated in both stroke patients and rats after tPA treatment. Blocking HMGB1 signaling by HBHP in the rat model of 4.5 h brain ischemia significantly attenuated tPA-related complications, including mortality rate, the degree of hemorrhage, brain swelling, neurological deficits, BBB impairment, microglia activation, and the expressions of inflammatory cytokines.

Conclusions

tPA treatment might induce HMGB1 secretion while blocking HMGB1 with HBHP could markedly reduce the risk of thrombolysis-associated brain hemorrhage and mortality through attenuating BBB damage and inflammatory reactions. These results indicate that HMGB1 may potentiate the risk of HT in tPA administration and that blocking HMGB1 signaling would be helpful in preventing complications brought by thrombolysis in ischemic stroke.

Trial registration

http://​www.​chictr.​org.​cn. Unique identifier: ChiCTR-OOC-16010052. Registered 30 November 2016.
Appendix
Available only for authorised users
Literature
1.
2.
go back to reference Group TNI of ND and S rt-PSS. Tissue plasminogen activator for acute ischemic stroke. N Engl J Med. 1995;333:1581–7.CrossRef Group TNI of ND and S rt-PSS. Tissue plasminogen activator for acute ischemic stroke. N Engl J Med. 1995;333:1581–7.CrossRef
3.
go back to reference Fonarow GC, Smith EE, Saver JL, Reeves MJ, Bhatt DL, Grau-Sepulveda MV, et al. Timeliness of tissue-type plasminogen activator therapy in acute ischemic stroke: patient characteristics, hospital factors, and outcomes associated with door-to-needle times within 60 minutes. Circulation. 2011;123:750–8.CrossRefPubMed Fonarow GC, Smith EE, Saver JL, Reeves MJ, Bhatt DL, Grau-Sepulveda MV, et al. Timeliness of tissue-type plasminogen activator therapy in acute ischemic stroke: patient characteristics, hospital factors, and outcomes associated with door-to-needle times within 60 minutes. Circulation. 2011;123:750–8.CrossRefPubMed
4.
go back to reference Wang X, Tsuji K, Lee SR, Ning M, Furie KL, Buchan AM, et al. Mechanisms of hemorrhagic transformation after tissue plasminogen activator reperfusion therapy for ischemic stroke. Stroke. 2004;35:2726–30.CrossRefPubMed Wang X, Tsuji K, Lee SR, Ning M, Furie KL, Buchan AM, et al. Mechanisms of hemorrhagic transformation after tissue plasminogen activator reperfusion therapy for ischemic stroke. Stroke. 2004;35:2726–30.CrossRefPubMed
5.
go back to reference Eltzschig HK, Eckle T. Ischemia and reperfusion--from mechanism to translation. Nat Med. 2011;17:1391–401.CrossRefPubMed Eltzschig HK, Eckle T. Ischemia and reperfusion--from mechanism to translation. Nat Med. 2011;17:1391–401.CrossRefPubMed
7.
go back to reference Lotze MT, Tracey KJ. High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol. 2005;5:331–42.CrossRefPubMed Lotze MT, Tracey KJ. High-mobility group box 1 protein (HMGB1): nuclear weapon in the immune arsenal. Nat Rev Immunol. 2005;5:331–42.CrossRefPubMed
8.
go back to reference Lu B, Wang C, Wang M, Li W, Chen F, Tracey KJ, et al. Molecular mechanism and therapeutic modulation of high mobility group box 1 release and action: an updated review. Expert Rev Clin Immunol. 2014;10:713–27.CrossRefPubMedPubMedCentral Lu B, Wang C, Wang M, Li W, Chen F, Tracey KJ, et al. Molecular mechanism and therapeutic modulation of high mobility group box 1 release and action: an updated review. Expert Rev Clin Immunol. 2014;10:713–27.CrossRefPubMedPubMedCentral
9.
go back to reference Liesz A, Dalpke A, Mracsko E, Antoine DJ, Roth S, Zhou W, et al. DAMP signaling is a key pathway inducing immune modulation after brain injury. J Neurosci. 2015;35:583–98.CrossRefPubMedPubMedCentral Liesz A, Dalpke A, Mracsko E, Antoine DJ, Roth S, Zhou W, et al. DAMP signaling is a key pathway inducing immune modulation after brain injury. J Neurosci. 2015;35:583–98.CrossRefPubMedPubMedCentral
10.
go back to reference Liu XX, Wang C, Huang SF, Chen Q, Hu YF, Zhou L, et al. Regnase-1 in microglia negatively regulates high mobility group box 1-mediated inflammation and neuronal injury. Sci Rep. 2016;6:24073.CrossRefPubMedPubMedCentral Liu XX, Wang C, Huang SF, Chen Q, Hu YF, Zhou L, et al. Regnase-1 in microglia negatively regulates high mobility group box 1-mediated inflammation and neuronal injury. Sci Rep. 2016;6:24073.CrossRefPubMedPubMedCentral
11.
go back to reference Goldstein RS, Gallowitsch-Puerta M, Yang L, Rosas-Ballina M, Huston JM, Czura CJ, et al. Elevated high-mobility group box 1 levels in patients with cerebral and myocardial ischemia. Shock. 2006;25:571–4.CrossRefPubMed Goldstein RS, Gallowitsch-Puerta M, Yang L, Rosas-Ballina M, Huston JM, Czura CJ, et al. Elevated high-mobility group box 1 levels in patients with cerebral and myocardial ischemia. Shock. 2006;25:571–4.CrossRefPubMed
12.
go back to reference Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol. 2010;28:367–88.CrossRefPubMed Sims GP, Rowe DC, Rietdijk ST, Herbst R, Coyle AJ. HMGB1 and RAGE in inflammation and cancer. Annu Rev Immunol. 2010;28:367–88.CrossRefPubMed
13.
go back to reference Liu K, Mori S, Takahashi HK, Tomono Y, Wake H, Kanke T, et al. Anti-high mobility group box 1 monoclonal antibody ameliorates brain infarction induced by transient ischemia in rats. FASEB J. 2007;21:3904–16.CrossRefPubMed Liu K, Mori S, Takahashi HK, Tomono Y, Wake H, Kanke T, et al. Anti-high mobility group box 1 monoclonal antibody ameliorates brain infarction induced by transient ischemia in rats. FASEB J. 2007;21:3904–16.CrossRefPubMed
14.
go back to reference Zhang J, Takahashi HK, Liu K, Wake H, Liu R, Maruo T, et al. Anti-high mobility group box-1 monoclonal antibody protects the blood-brain barrier from ischemia-induced disruption in rats. Stroke. 2011;42:1420–8.CrossRefPubMed Zhang J, Takahashi HK, Liu K, Wake H, Liu R, Maruo T, et al. Anti-high mobility group box-1 monoclonal antibody protects the blood-brain barrier from ischemia-induced disruption in rats. Stroke. 2011;42:1420–8.CrossRefPubMed
15.
go back to reference Kim ID, Shin JH, Lee HK, Jin YC, Lee JK. Intranasal delivery of HMGB1-binding heptamer peptide confers a robust neuroprotection in the postischemic brain. Neurosci Lett. 2012;525:179–83.CrossRefPubMed Kim ID, Shin JH, Lee HK, Jin YC, Lee JK. Intranasal delivery of HMGB1-binding heptamer peptide confers a robust neuroprotection in the postischemic brain. Neurosci Lett. 2012;525:179–83.CrossRefPubMed
16.
go back to reference Gu Y, Zheng G, Xu M, Li Y, Chen X, Zhu W, et al. Caveolin-1 regulates nitric oxide-mediated matrix metalloproteinases activity and blood-brain barrier permeability in focal cerebral ischemia and reperfusion injury. J Neurochem. 2012;120:147–56.CrossRefPubMed Gu Y, Zheng G, Xu M, Li Y, Chen X, Zhu W, et al. Caveolin-1 regulates nitric oxide-mediated matrix metalloproteinases activity and blood-brain barrier permeability in focal cerebral ischemia and reperfusion injury. J Neurochem. 2012;120:147–56.CrossRefPubMed
17.
go back to reference Kilkenny C, Browne W, Cuthill IC, Emerson M, Altman DG, Group NCRGW. Animal research: reporting in vivo experiments: the ARRIVE guidelines. Br J Pharmacol. 2010;160:1577–9.CrossRefPubMedPubMedCentral Kilkenny C, Browne W, Cuthill IC, Emerson M, Altman DG, Group NCRGW. Animal research: reporting in vivo experiments: the ARRIVE guidelines. Br J Pharmacol. 2010;160:1577–9.CrossRefPubMedPubMedCentral
18.
go back to reference Liu W, Hendren J, Qin XJ, Liu KJ. Normobaric hyperoxia reduces the neurovascular complications associated with delayed tissue plasminogen activator treatment in a rat model of focal cerebral ischemia. Stroke. 2009;40:2526–31.CrossRefPubMedPubMedCentral Liu W, Hendren J, Qin XJ, Liu KJ. Normobaric hyperoxia reduces the neurovascular complications associated with delayed tissue plasminogen activator treatment in a rat model of focal cerebral ischemia. Stroke. 2009;40:2526–31.CrossRefPubMedPubMedCentral
20.
go back to reference Chen J, Sanberg PR, Li Y, Wang L, Lu M, Willing AE, et al. Intravenous administration of human umbilical cord blood reduces behavioral deficits after stroke in rats. Stroke. 2001;32:2682–8.CrossRefPubMed Chen J, Sanberg PR, Li Y, Wang L, Lu M, Willing AE, et al. Intravenous administration of human umbilical cord blood reduces behavioral deficits after stroke in rats. Stroke. 2001;32:2682–8.CrossRefPubMed
22.
go back to reference Fagan SC, Garcia JH. Hemorrhagic transformation in focal cerebral ischemia: influence of time to artery reopening and tissue plasminogen activator. Pharmacotherapy. 1999;19:139–42.CrossRefPubMed Fagan SC, Garcia JH. Hemorrhagic transformation in focal cerebral ischemia: influence of time to artery reopening and tissue plasminogen activator. Pharmacotherapy. 1999;19:139–42.CrossRefPubMed
23.
go back to reference Jeong H, Ji K, Kim J, Jou I, Joe E. Repair of astrocytes , blood vessels , and myelin in the injured brain: possible roles of blood monocytes. Mol. Brain. 2013;6:1.CrossRef Jeong H, Ji K, Kim J, Jou I, Joe E. Repair of astrocytes , blood vessels , and myelin in the injured brain: possible roles of blood monocytes. Mol. Brain. 2013;6:1.CrossRef
24.
go back to reference Ozbek D, Ozturk O, Ekinci G, Midi I. Risk of hemorrhage in ischemic stroke and its relationship with cerebral microbleeds. Clin Neurol Neurosurg Elsevier. 2018;168:112–7.CrossRef Ozbek D, Ozturk O, Ekinci G, Midi I. Risk of hemorrhage in ischemic stroke and its relationship with cerebral microbleeds. Clin Neurol Neurosurg Elsevier. 2018;168:112–7.CrossRef
25.
go back to reference Antoine D, Harris H. A systematic nomenclature for the redox states of high mobility group box (HMGB) proteins. Mol Med. 2014;20:1.CrossRef Antoine D, Harris H. A systematic nomenclature for the redox states of high mobility group box (HMGB) proteins. Mol Med. 2014;20:1.CrossRef
26.
go back to reference Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, et al. HMGB1 in health and disease. Mol Aspects Med Elsevier Ltd. 2014;40:1–116.CrossRef Kang R, Chen R, Zhang Q, Hou W, Wu S, Cao L, et al. HMGB1 in health and disease. Mol Aspects Med Elsevier Ltd. 2014;40:1–116.CrossRef
27.
go back to reference Musumeci D, Roviello GN, Montesarchio D. An overview on HMGB1 inhibitors as potential therapeutic agents in HMGB1-related pathologies. Pharmacol Ther. 2014;141:347–57.CrossRefPubMed Musumeci D, Roviello GN, Montesarchio D. An overview on HMGB1 inhibitors as potential therapeutic agents in HMGB1-related pathologies. Pharmacol Ther. 2014;141:347–57.CrossRefPubMed
28.
go back to reference Hayakawa K, Pham LDD, Katusic ZS, Arai K, Lo EH. Astrocytic high-mobility group box 1 promotes endothelial progenitor cell-mediated neurovascular remodeling during stroke recovery. Proc Natl Acad Sci U S A. 2012;109:7505–10.CrossRefPubMedPubMedCentral Hayakawa K, Pham LDD, Katusic ZS, Arai K, Lo EH. Astrocytic high-mobility group box 1 promotes endothelial progenitor cell-mediated neurovascular remodeling during stroke recovery. Proc Natl Acad Sci U S A. 2012;109:7505–10.CrossRefPubMedPubMedCentral
29.
go back to reference Jickling GC, Liu D, Stamova B, Ander BP, Zhan X, Lu A, et al. Hemorrhagic transformation after ischemic stroke in animals and humans. J Cereb Blood Flow Metab. 2014;34:185–99.CrossRefPubMed Jickling GC, Liu D, Stamova B, Ander BP, Zhan X, Lu A, et al. Hemorrhagic transformation after ischemic stroke in animals and humans. J Cereb Blood Flow Metab. 2014;34:185–99.CrossRefPubMed
30.
go back to reference Wang X, Lee SR, Arai K, Lee SR, Tsuji K, Rebeck GW, et al. Lipoprotein receptor-mediated induction of matrix metalloproteinase by tissue plasminogen activator. Nat Med. 2003;9:1313–7.CrossRefPubMed Wang X, Lee SR, Arai K, Lee SR, Tsuji K, Rebeck GW, et al. Lipoprotein receptor-mediated induction of matrix metalloproteinase by tissue plasminogen activator. Nat Med. 2003;9:1313–7.CrossRefPubMed
31.
go back to reference Arumugam T, Ramachandran V, Gomez SB, Schmidt AM, Logsdon CD. S100P-derived RAGE antagonistic peptide reduces tumor growth and metastasis. Clin Cancer Res. 2012;18:4356–64.CrossRefPubMed Arumugam T, Ramachandran V, Gomez SB, Schmidt AM, Logsdon CD. S100P-derived RAGE antagonistic peptide reduces tumor growth and metastasis. Clin Cancer Res. 2012;18:4356–64.CrossRefPubMed
32.
go back to reference Mollica L, De Marchis F, Spitaleri A, Dallacosta C, Pennacchini D, Zamai M, et al. Glycyrrhizin binds to high-mobility group box 1 protein and inhibits its cytokine activities. Chem Biol. 2007;14:431–41.CrossRefPubMed Mollica L, De Marchis F, Spitaleri A, Dallacosta C, Pennacchini D, Zamai M, et al. Glycyrrhizin binds to high-mobility group box 1 protein and inhibits its cytokine activities. Chem Biol. 2007;14:431–41.CrossRefPubMed
33.
go back to reference Kim ID, Luo L, Lee HB, Lee HK, Lee JK. HMGB1-binding heptamer suppresses the synergistic effect of HMGB1 and LPS by interacting directly with HMGB1. Neurosci Lett. 2015;593:40–4.CrossRefPubMed Kim ID, Luo L, Lee HB, Lee HK, Lee JK. HMGB1-binding heptamer suppresses the synergistic effect of HMGB1 and LPS by interacting directly with HMGB1. Neurosci Lett. 2015;593:40–4.CrossRefPubMed
34.
go back to reference Shi X, Li M, Huang K, Zhou S, Hu Y, Pan S, et al. HMGB1 binding heptamer peptide improves survival and ameliorates brain injury in rats after cardiac arrest and cardiopulmonary resuscitation. Neurosci IBRO. 2017;360:128–38.CrossRef Shi X, Li M, Huang K, Zhou S, Hu Y, Pan S, et al. HMGB1 binding heptamer peptide improves survival and ameliorates brain injury in rats after cardiac arrest and cardiopulmonary resuscitation. Neurosci IBRO. 2017;360:128–38.CrossRef
35.
go back to reference Latour LL, Kang DW, Ezzeddine MA, Chalela JA, Warach S. Early blood-brain barrier disruption in human focal brain ischemia. Ann Neurol. 2004;56:468–77.CrossRefPubMed Latour LL, Kang DW, Ezzeddine MA, Chalela JA, Warach S. Early blood-brain barrier disruption in human focal brain ischemia. Ann Neurol. 2004;56:468–77.CrossRefPubMed
36.
go back to reference Leigh R, Jen SS, Hillis AE, Krakauer JW, Barker PB, Stir, et al. Pretreatment blood-brain barrier damage and post-treatment intracranial hemorrhage in patients receiving intravenous tissue-type plasminogen activator. Stroke. 2014;45:2030–5.CrossRefPubMedPubMedCentral Leigh R, Jen SS, Hillis AE, Krakauer JW, Barker PB, Stir, et al. Pretreatment blood-brain barrier damage and post-treatment intracranial hemorrhage in patients receiving intravenous tissue-type plasminogen activator. Stroke. 2014;45:2030–5.CrossRefPubMedPubMedCentral
37.
go back to reference Leigh R, Christensen S, Campbell BC, Marks MP, Albers GW, Lansberg MG, et al. Pretreatment blood-brain barrier disruption and post-endovascular intracranial hemorrhage. Neurology. 2016;87:263–9.CrossRefPubMedPubMedCentral Leigh R, Christensen S, Campbell BC, Marks MP, Albers GW, Lansberg MG, et al. Pretreatment blood-brain barrier disruption and post-endovascular intracranial hemorrhage. Neurology. 2016;87:263–9.CrossRefPubMedPubMedCentral
38.
go back to reference Roussel BD, Mysiorek C, Rouhiainen A, Jullienne A, Parcq J, Hommet Y, et al. HMGB-1 promotes fibrinolysis and reduces neurotoxicity mediated by tissue plasminogen activator. J Cell Sci. 2011;124:2070–6.CrossRefPubMed Roussel BD, Mysiorek C, Rouhiainen A, Jullienne A, Parcq J, Hommet Y, et al. HMGB-1 promotes fibrinolysis and reduces neurotoxicity mediated by tissue plasminogen activator. J Cell Sci. 2011;124:2070–6.CrossRefPubMed
40.
go back to reference Iadecola C, Niwa K, Nogawa S, Zhao X, Nagayama M, Araki E, et al. Reduced susceptibility to ischemic brain injury and N-methyl-D-aspartate-mediated neurotoxicity in cyclooxygenase-2-deficient mice. Proc Natl Acad Sci U S A. 2001;98:1294–9.CrossRefPubMedPubMedCentral Iadecola C, Niwa K, Nogawa S, Zhao X, Nagayama M, Araki E, et al. Reduced susceptibility to ischemic brain injury and N-methyl-D-aspartate-mediated neurotoxicity in cyclooxygenase-2-deficient mice. Proc Natl Acad Sci U S A. 2001;98:1294–9.CrossRefPubMedPubMedCentral
Metadata
Title
Cell permeable HMGB1-binding heptamer peptide ameliorates neurovascular complications associated with thrombolytic therapy in rats with transient ischemic stroke
Authors
Miaodan Li
Shumin Chen
Xue Shi
Chenfei Lyu
Yongfang Zhang
Miaoqin Tan
Chen Wang
Nailiang Zang
Xiaoxi Liu
Yafang Hu
Jiangang Shen
Liang Zhou
Yong Gu
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2018
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-018-1267-5

Other articles of this Issue 1/2018

Journal of Neuroinflammation 1/2018 Go to the issue