Skip to main content

Advertisement

Log in

Glycyrrhizin Prevents Hemorrhagic Transformation and Improves Neurological Outcome in Ischemic Stroke with Delayed Thrombolysis Through Targeting Peroxynitrite-Mediated HMGB1 Signaling

  • Original Article
  • Published:
Translational Stroke Research Aims and scope Submit manuscript

Abstract

Peroxynitrite (ONOO) and high mobility group box 1 protein (HMGB1) are important cytotoxic factors contributing to cerebral ischemia-reperfusion injury. However, the roles of ONOO in mediating HMGB1 expression and its impacts on hemorrhagic transformation (HT) in ischemic brain injury with delayed t-PA treatment remain unclear. In the present study, we tested the hypothesis that ONOO could directly mediate the activation and release of HMGB1 in ischemic brains with delayed t-PA treatment. With clinical studies, we found that plasma nitrotyrosine (NT, a surrogate marker of ONOO) was positively correlated with HMGB1 level in acute ischemic stroke patients. Hemorrhagic transformation and t-PA-treated ischemic stroke patients had increased levels of nitrotyrosine and HMGB1 in plasma. In animal experiments, we found that FeTmPyP, a representative ONOO decomposition catalyst (PDC), significantly reduced the expression of HMGB1 and its receptor TLR2, and inhibited MMP-9 activation, preserved collagen IV and tight junction claudin-5 in ischemic rat brains with delayed t-PA treatment. ONOO donor SIN-1 directly induced expression of HMGB1 and its receptor TLR2 in naive rat brains in vivo and induced HMGB1 in brain microvascular endothelial b.End3 cells in vitro. Those results suggest that ONOO could activate HMGB1/TLR2/MMP-9 signaling. We then addressed whether glycyrrhizin, a natural HMGB1 inhibitor, could inhibit ONOO production and the antioxidant properties of glycyrrhizin contribute to the inhibition of HMGB1 and the neuroprotective effects on attenuating hemorrhagic transformation in ischemic stroke with delayed t-PA treatment. Glycyrrhizin treatment downregulated the expressions of NADPH oxidase p47 phox and p67 phox and iNOS, inhibited superoxide and ONOO production, reduced the expression of HMGB1, TLR2, MMP-9, preserved type IV collagen and claudin-5 in ischemic brains. Furthermore, glycyrrhizin significantly decreased the mortality rate, attenuated hemorrhagic transformation, brain swelling, blood-brain barrier damage, neuronal apoptosis, and improved neurological outcomes in the ischemic stroke rat model with delayed t-PA treatment. In conclusion, peroxynitrite-mediated HMGB1/TLR2 signaling contributes to hemorrhagic transformation, and glycyrrhizin could be a potential adjuvant therapy to attenuate hemorrhagic transformation, possibly through inhibiting the ONOO/HMGB1/TLR2 signaling cascades.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

References

  1. Fugate JE, Giraldo EA, Rabinstein AA. Thrombolysis for cerebral ischemia. Front Neurol. 2010;1:139. https://doi.org/10.3389/fneur.2010.00139.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Emberson J, Lees KR, Lyden P, Blackwell L, Albers G, Bluhmki E, et al. Effect of treatment delay, age, and stroke severity on the effects of intravenous thrombolysis with alteplase for acute ischaemic stroke: a meta-analysis of individual patient data from randomised trials. Lancet. 2014;384(9958):1929–35. https://doi.org/10.1016/S0140-6736(14)60584-5.

  3. Ho WM, Reis C, Akyol O, Akyol GY, Applegate R, Stier G, et al. Pharmacological management options to prevent and reduce ischemic hemorrhagic transformation. Curr Drug Targets. 2017;18(12):1441–59. https://doi.org/10.2174/1389450117666160818115850.

    Article  PubMed  CAS  Google Scholar 

  4. Knecht T, Story J, Liu J, Davis W, Borlongan CV, Dela Pena IC. Adjunctive therapy approaches for ischemic stroke: innovations to expand time window of treatment. Int J Mol Sci. 2017;18(12). https://doi.org/10.3390/ijms18122756.

  5. Chen HS, Qi SH, Shen JG. One-compound-multi-target: combination prospect of natural compounds with thrombolytic therapy in acute ischemic stroke. Curr Neuropharmacol. 2017;15(1):134–56.

    Article  CAS  Google Scholar 

  6. Zhang L, Zhang ZG, Chopp M. The neurovascular unit and combination treatment strategies for stroke. Trends Pharmacol Sci. 2012;33(8):415–22. https://doi.org/10.1016/j.tips.2012.04.006.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  7. Sumii T, Lo EH. Involvement of matrix metalloproteinase in thrombolysis-associated hemorrhagic transformation after embolic focal ischemia in rats. Stroke. 2002;33(3):831–6.

    Article  CAS  Google Scholar 

  8. Tsuji K, Aoki T, Tejima E, Arai K, Lee SR, Atochin DN, et al. Tissue plasminogen activator promotes matrix metalloproteinase-9 upregulation after focal cerebral ischemia. Stroke. 2005;36(9):1954–9. https://doi.org/10.1161/01.STR.0000177517.01203.eb.

  9. Lapchak PA, Chapman DF, Zivin JA. Metalloproteinase inhibition reduces thrombolytic (tissue plasminogen activator)-induced hemorrhage after thromboembolic stroke. Stroke. 2000;31(12):3034–40.

    Article  CAS  Google Scholar 

  10. Pfefferkorn T, Rosenberg GA. Closure of the blood-brain barrier by matrix metalloproteinase inhibition reduces rtPA-mediated mortality in cerebral ischemia with delayed reperfusion. Stroke. 2003;34(8):2025–30. https://doi.org/10.1161/01.STR.0000083051.93319.28.

    Article  PubMed  Google Scholar 

  11. Qiu J, Nishimura M, Wang Y, Sims JR, Qiu S, Savitz SI, et al. Early release of HMGB-1 from neurons after the onset of brain ischemia. J Cereb Blood Flow Metab. 2008;28(5):927–38. https://doi.org/10.1038/sj.jcbfm.9600582.

  12. Qiu J, Xu J, Zheng Y, Wei Y, Zhu X, Lo EH, et al. High-mobility group box 1 promotes metalloproteinase-9 upregulation through Toll-like receptor 4 after cerebral ischemia. Stroke. 2010;41(9):2077–82.

    Article  CAS  Google Scholar 

  13. Kim JB, Lim CM, Yu YM, Lee JK. Induction and subcellular localization of high-mobility group box-1 (HMGB1) in the postischemic rat brain. J Neurosci Res. 2008;86(5):1125–31. https://doi.org/10.1002/jnr.21555.

    Article  PubMed  CAS  Google Scholar 

  14. 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(5):1420–8. https://doi.org/10.1161/STROKEAHA.110.598334.

  15. 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(14):3904–16. https://doi.org/10.1096/fj.07-8770com.

  16. Li M, Chen S, Shi X, Lyu C, Zhang Y, Tan M, et al. Cell permeable HMGB1-binding heptamer peptide ameliorates neurovascular complications associated with thrombolytic therapy in rats with transient ischemic stroke. J Neuroinflammation. 2018;15(1):237.

  17. Sun MS, Jin H, Sun X, Huang S, Zhang FL, Guo ZN, et al. Free radical damage in ischemia-reperfusion injury: an obstacle in acute ischemic stroke after revascularization therapy. Oxidative Med Cell Longev. 2018;2018:3804979. https://doi.org/10.1155/2018/3804979.

  18. Chen HS, Chen X, Li WT, Shen JG. Targeting RNS/caveolin-1/MMP signaling cascades to protect against cerebral ischemia-reperfusion injuries: potential application for drug discovery. Acta Pharmacol Sin. 2018;39(5):669–82. https://doi.org/10.1038/aps.2018.27.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Gu Y, Dee CM, Shen J. Interaction of free radicals, matrix metalloproteinases and caveolin-1 impacts blood-brain barrier permeability. Front Biosci (Schol Ed). 2011;3:1216–31.

    Article  Google Scholar 

  20. Gasche Y, Copin JC, Sugawara T, Fujimura M, Chan PH. Matrix metalloproteinase inhibition prevents oxidative stress-associated blood-brain barrier disruption after transient focal cerebral ischemia. J Cereb Blood Flow Metab. 2001;21(12):1393–400. https://doi.org/10.1097/00004647-200112000-00003.

    Article  PubMed  CAS  Google Scholar 

  21. Jian Liu K, Rosenberg GA. Matrix metalloproteinases and free radicals in cerebral ischemia. Free Radic Biol Med. 2005;39(1):71–80. https://doi.org/10.1016/j.freeradbiomed.2005.03.033.

    Article  PubMed  CAS  Google Scholar 

  22. Suzuki M, Tabuchi M, Ikeda M, Tomita T. Concurrent formation of peroxynitrite with the expression of inducible nitric oxide synthase in the brain during middle cerebral artery occlusion and reperfusion in rats. Brain Res. 2002;951(1):113–20.

    Article  CAS  Google Scholar 

  23. Virag L, Szabo E, Gergely P, Szabo C. Peroxynitrite-induced cytotoxicity: mechanism and opportunities for intervention. Toxicol Lett. 2003;140–141:113–24. https://doi.org/10.1016/s0378-4274(02)00508-8.

    Article  PubMed  Google Scholar 

  24. Gursoy-Ozdemir Y, Can A, Dalkara T. Reperfusion-induced oxidative/nitrative injury to neurovascular unit after focal cerebral ischemia. Stroke. 2004;35(6):1449–53. https://doi.org/10.1161/01.STR.0000126044.83777.f4.

    Article  PubMed  CAS  Google Scholar 

  25. Chen HS, Chen XM, Feng JH, Liu KJ, Qi SH, Shen JG. Peroxynitrite decomposition catalyst reduces delayed thrombolysis-induced hemorrhagic transformation in ischemia-reperfused rat brains. CNS Neurosci Ther. 2015;21(7):585–90. https://doi.org/10.1111/cns.12406.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Chen H, Guan B, Chen X, Chen X, Li C, Qiu J, et al. Baicalin attenuates blood-brain barrier disruption and hemorrhagic transformation and improves neurological outcome in ischemic stroke rats with delayed t-PA treatment: involvement of ONOO(−)-MMP-9 pathway. Transl Stroke Res. 2018;9(5):515–29. https://doi.org/10.1007/s12975-017-0598-3.

  27. Loukili N, Rosenblatt-Velin N, Li J, Clerc S, Pacher P, Feihl F, et al. Peroxynitrite induces HMGB1 release by cardiac cells in vitro and HMGB1 upregulation in the infarcted myocardium in vivo. Cardiovasc Res. 2011;89(3):586–94. https://doi.org/10.1093/cvr/cvq373.

  28. Chandrashekaran V, Seth RK, Dattaroy D, Alhasson F, Ziolenka J, Carson J, et al. HMGB1-RAGE pathway drives peroxynitrite signaling-induced IBD-like inflammation in murine nonalcoholic fatty liver disease. Redox Biol. 2017;13:8–19. https://doi.org/10.1016/j.redox.2017.05.005.

  29. 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(4):431–41. https://doi.org/10.1016/j.chembiol.2007.03.007.

    Article  PubMed  CAS  Google Scholar 

  30. Girard JP. A direct inhibitor of HMGB1 cytokine. Chem Biol. 2007;14(4):345–7. https://doi.org/10.1016/j.chembiol.2007.04.001.

    Article  PubMed  CAS  Google Scholar 

  31. Kim SW, Jin Y, Shin JH, Kim ID, Lee HK, Park S, et al. Glycyrrhizic acid affords robust neuroprotection in the postischemic brain via anti-inflammatory effect by inhibiting HMGB1 phosphorylation and secretion. Neurobiol Dis. 2012;46(1):147–56. https://doi.org/10.1016/j.nbd.2011.12.056.

  32. Gong G, Xiang L, Yuan L, Hu L, Wu W, Cai L, et al. Protective effect of glycyrrhizin, a direct HMGB1 inhibitor, on focal cerebral ischemia/reperfusion-induced inflammation, oxidative stress, and apoptosis in rats. PLoS One. 2014;9(3):e89450. https://doi.org/10.1371/journal.pone.0089450.

  33. Zhang J, Wu Y, Weng Z, Zhou T, Feng T, Lin Y. Glycyrrhizin protects brain against ischemia-reperfusion injury in mice through HMGB1-TLR4-IL-17A signaling pathway. Brain Res. 2014;1582:176–86. https://doi.org/10.1016/j.brainres.2014.07.002.

    Article  PubMed  CAS  Google Scholar 

  34. 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(7):2526–31. https://doi.org/10.1161/STROKEAHA.108.545483.

    Article  PubMed  PubMed Central  Google Scholar 

  35. 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(11):2682–8.

  36. Liu W, Sood R, Chen Q, Sakoglu U, Hendren J, Cetin O, et al. Normobaric hyperoxia inhibits NADPH oxidase-mediated matrix metalloproteinase-9 induction in cerebral microvessels in experimental stroke. J Neurochem. 2008;107(5):1196–205. https://doi.org/10.1111/j.1471-4159.2008.05664.x.

  37. Fagan SC, Lapchak PA, Liebeskind DS, Ishrat T, Ergul A. Recommendations for preclinical research in hemorrhagic transformation. Transl Stroke Res. 2013;4(3):322–7. https://doi.org/10.1007/s12975-012-0222-5.

    Article  PubMed  Google Scholar 

  38. Peng T, Chen X, Gao L, Zhang T, Wang W, Shen J, et al. A rationally designed rhodamine-based fluorescent probe for molecular imaging of peroxynitrite in live cells and tissues. Chem Sci. 2016;7(8):5407–13. https://doi.org/10.1039/c6sc00012f.

  39. Hu JJ, Wong NK, Ye S, Chen X, Lu MY, Zhao AQ, et al. Fluorescent probe HKSOX-1 for imaging and detection of endogenous superoxide in live cells and in vivo. J Am Chem Soc. 2015;137(21):6837–43. https://doi.org/10.1021/jacs.5b01881.

  40. 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(2):185–99. https://doi.org/10.1038/jcbfm.2013.203.

  41. Kanazawa M, Takahashi T, Nishizawa M, Shimohata T. Therapeutic strategies to attenuate hemorrhagic transformation after tissue plasminogen activator treatment for acute ischemic stroke. J Atheroscler Thromb. 2017;24(3):240–53. https://doi.org/10.5551/jat.RV16006.

  42. Chen H, Chen X, Luo Y, Shen J. Potential molecular targets of peroxynitrite in mediating blood-brain barrier damage and haemorrhagic transformation in acute ischaemic stroke with delayed tissue plasminogen activator treatment. Free Radic Res. 2018:1–20. https://doi.org/10.1080/10715762.2018.1521519.

  43. Kim JB, Sig Choi J, Yu YM, Nam K, Piao CS, Kim SW, et al. HMGB1, a novel cytokine-like mediator linking acute neuronal death and delayed neuroinflammation in the postischemic brain. J Neurosci. 2006;26(24):6413–21. https://doi.org/10.1523/JNEUROSCI.3815-05.2006.

  44. Muhammad S, Barakat W, Stoyanov S, Murikinati S, Yang H, Tracey KJ, et al. The HMGB1 receptor RAGE mediates ischemic brain damage. J Neurosci. 2008;28(46):12023–31. https://doi.org/10.1523/JNEUROSCI.2435-08.2008.

  45. Zhu H, Dai R, Fu H, Meng Q. MMP-9 Upregulation is attenuated by the monoclonal TLR2 antagonist T2. 5 after oxygen–glucose deprivation and reoxygenation in rat brain microvascular endothelial cells. J Stroke Cerebrovasc Dis. 2019;28(1):97–106.

    Article  Google Scholar 

  46. Musumeci D, Roviello GN, Montesarchio D. An overview on HMGB1 inhibitors as potential therapeutic agents in HMGB1-related pathologies. Pharmacol Ther. 2014;141(3):347–57.

    Article  CAS  Google Scholar 

  47. Wang C, Jiang J, Zhang X, Song L, Sun K, Xu R. Inhibiting HMGB1 reduces cerebral ischemia reperfusion injury in diabetic mice. Inflammation. 2016;39(6):1862–70. https://doi.org/10.1007/s10753-016-0418-z.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. 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(2):179–83. https://doi.org/10.1016/j.neulet.2012.07.040.

    Article  PubMed  CAS  Google Scholar 

  49. Xiong X, Gu L, Wang Y, Luo Y, Zhang H, Lee J, et al. Glycyrrhizin protects against focal cerebral ischemia via inhibition of T cell activity and HMGB1-mediated mechanisms. J Neuroinflammation. 2016;13(1):241. https://doi.org/10.1186/s12974-016-0705-5.

Download references

Funding

This work is supported by Hong Kong General Research Fund (GRF No. 17102915, GRF No. 17118717), Research Grant Council, Hong Kong SAR and Health and Medical Research Fund, Hong Kong SAR (NO. 13142901), AoE/P-705/16 Areas of Excellence Scheme, RGC, Hong Kong SAR; SIRI/04/04/2015/06 Shenzhen Basic Research Plan Project. National Natural Science Foundation of China (No. 81671164).

Author information

Authors and Affiliations

Authors

Contributions

Hansen Chen performed in vivo and in vitro experiments, analyzed data, and wrote the manuscript; Binghe Guan, Bin Wang, and Haiwei Pu performed clinical investigations and analyzed the data; Caiming Li, Jihong Liu, and Jinhua Qiu recruited the ischemic stroke patients, made clinical diagnosis, collected blood samples, and performed clinical studies; Xiaoyu Bai and Xi Chen performed in vitro study and conducted data analysis; Dan Yang synthesized the probes for ONOO detection and data analysis; Kejian Liu and Qi Wang contributed to the study design and discussion; Suhua Qi contributed to the study design in vivo study, clinical investigation and manuscript preparation; Jiangang Shen received the funding, guided and designed all experiments, interpreted the results, and prepared and revised the manuscripts.

Corresponding authors

Correspondence to Suhua Qi or Jiangang Shen.

Ethics declarations

Conflict of Interest

The authors declare that they have no conflict of interest.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic Supplementary Material

ESM 1

(DOCX 1125 kb).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chen, H., Guan, B., Wang, B. et al. Glycyrrhizin Prevents Hemorrhagic Transformation and Improves Neurological Outcome in Ischemic Stroke with Delayed Thrombolysis Through Targeting Peroxynitrite-Mediated HMGB1 Signaling. Transl. Stroke Res. 11, 967–982 (2020). https://doi.org/10.1007/s12975-019-00772-1

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12975-019-00772-1

Keywords

Navigation