Generic placeholder image

CNS & Neurological Disorders - Drug Targets

Editor-in-Chief

ISSN (Print): 1871-5273
ISSN (Online): 1996-3181

Research Article

Combined Ischemic Preconditioning and Resveratrol Improved Bloodbrain Barrier Breakdown via Hippo/YAP/TAZ Signaling Pathway

Author(s): Ganji Hong, Ying Yan, Yali Zhong, Jianer Chen, Fei Tong* and Qilin Ma*

Volume 18, Issue 9, 2019

Page: [713 - 722] Pages: 10

DOI: 10.2174/1871527318666191021144126

Price: $65

Abstract

Background: Transient Ischemia/Reperfusion (I/R) is the main reason for brain injury and results in disruption of the Blood-Brain Barrier (BBB). It had been reported that BBB injury is one of the main risk factors for early death in patients with cerebral ischemia. Numerous investigations focus on the study of BBB injury which have been carried out.

Objective: The objective of this study was to investigate the treatment function of the activation of the Hippo/Yes-Associated Protein (YAP) signaling pathway by combined Ischemic Preconditioning (IPC) and resveratrol (RES) before brain Ischemia/Reperfusion (BI/R) improves Blood-Brain Barrier (BBB) disruption in rats.

Methods: Sprague-Dawley (SD) rats were pretreated with 20 mg/kg RES and IPC and then subjected to 2 h of ischemia and 22 h of reperfusion. The cerebral tissues were collected; the cerebral infarct volume was determined; the Evans Blue (EB) level, the brain Water Content (BWC), and apoptosis were assessed; and the expressions of YAP and TAZ were investigated in cerebral tissues.

Results: Both IPC and RES preconditioning reduced the cerebral infarct size, improved BBB permeability, lessened apoptosis, and upregulated expressions of YAP and transcriptional co-activator with PDZ-binding motif (TAZ) compared to the Ischemia/Reperfusion (I/R) group, while combined IPC and RES significantly enhanced this action.

Conclusion: combined ischemic preconditioning and resveratrol improved blood-brain barrier breakdown via Hippo/YAP/TAZ signaling pathway.

Keywords: Ischemic Preconditioning (IPC), Resveratrol (RES), Hippo/YAP/TAZ, Brain Ischemia/Reperfusion Injury (BI/RI), Blood-Brain Barrier (BBB), Brain Water Content (BWC).

Graphical Abstract
[1]
Zhao B, Tumaneng K, Guan KL. The Hippo pathway in organ size control, tissue regeneration and stem cell self-renewal. Nat Cell Biol 2011; 13(8): 877-83.
[http://dx.doi.org/10.1038/ncb2303] [PMID: 21808241]
[2]
Johnson R, Halder G. The two faces of Hippo: Targeting the Hippo pathway for regenerative medicine and cancer treatment. Nat Rev Drug Discov 2014; 13(1): 63-79.
[http://dx.doi.org/10.1038/nrd4161] [PMID: 24336504]
[3]
Yu FX, Zhao B, Guan KL. Hippo pathway in organ size control, tissue homeostasis, and cancer. Cell 2015; 163(4): 811-28.
[http://dx.doi.org/10.1016/j.cell.2015.10.044] [PMID: 26544935]
[4]
Gong P, Zhang Z, Zou C, et al. Hippo/YAP signaling pathway mitigates blood-brain barrier disruption after cerebral ischemia/ reperfusion injury. Behav Brain Res 2018; pii: S0166-4328(18): 30816..
[http://dx.doi.org/10.1016/j.bbr.2018.08.003]
[5]
Choi HJ, Kwon YG. Roles of YAP in mediating endothelial cell junctional stability and vascular remodeling. BMB Rep 2015; 48(8): 429-30.
[http://dx.doi.org/10.5483/BMBRep.2015.48.8.146] [PMID: 26169195]
[6]
Choi HJ, Zhang H, Park H, et al. Yes-associated protein regulates endothelial cell contact-mediated expression of angiopoietin-2. Nat Commun 2015; 6: 6943.
[http://dx.doi.org/10.1038/ncomms7943] [PMID: 25962877]
[7]
Wang L, Luo J, Li B, et al. Integrin-YAP/TAZJNK cascade mediates atheroprotective effect of unidirectional shear flow. Nature 2016; 540(7634): 579-82.
[http://dx.doi.org/10.1038/nature20602]
[8]
Ramalho-Santos M, Yoon S, Matsuzaki Y, Mulligan RC, Melton DA. “Stemness”: Transcriptional profiling of embryonic and adult stem cells. Science 2002; 298(5593): 597-600.
[http://dx.doi.org/10.1126/science.1072530] [PMID: 12228720]
[9]
Gu Y, Zheng G, Xu M, 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(1): 147-56.
[http://dx.doi.org/10.1111/j.1471-4159.2011.07542.x] [PMID: 22007835]
[10]
Durukan A, Tatlisumak T. Acute ischemic stroke: Overview of major experimental rodent models, pathophysiology, and therapy of focal cerebral ischemia. Pharmacol Biochem Behav 2007; 87(1): 179-97.
[http://dx.doi.org/10.1016/j.pbb.2007.04.015] [PMID: 17521716]
[11]
Li Q, Han X, Lan X, et al. Inhibition of tPA-induced hemorrhagic transformation involves adenosine A2b receptor activation after cerebral ischemia. Neurobiol Dis 2017; 108: 173-82.
[http://dx.doi.org/10.1016/j.nbd.2017.08.011] [PMID: 28830843]
[12]
Jiang C, Wang J, Yu L, et al. Comparison of the therapeutic effects of bone marrow mononuclear cells and microglia for permanent cerebral ischemia. Behav Brain Res 2013; 250: 222-9.
[http://dx.doi.org/10.1016/j.bbr.2013.05.011] [PMID: 23685323]
[13]
Wang J, Liu X, Lu H, et al. CXCR4(+)CD45(-) BMMNC subpopulation is superior to unfractionated BMMNCs for protection after ischemic stroke in mice. Brain Behav Immun 2015; 45: 98-108.
[http://dx.doi.org/10.1016/j.bbi.2014.12.015] [PMID: 25526817]
[14]
Jiang C, Zuo F, Wang Y, et al. Progesterone exerts neuroprotective effects and improves long-term neurologic outcome after intracerebral hemorrhage in middle-aged mice. Neurobiol Aging 2016; 42: 13-24.
[http://dx.doi.org/10.1016/j.neurobiolaging.2016.02.029] [PMID: 27143417]
[15]
Jiang X, Andjelkovic A, Zhu L, et al. Blood-brain barrier dysfunction and recovery after ischemic stroke. Prog Neurobiol 2017.
[PMID: 28987927]
[16]
Zhang Z, Song Y, Zhang Z, et al. Distinct role of heme oxygenase-1 in early- and late-stage intracerebral hemorrhage in 12-month-old mice. J Cereb Blood Flow Metab 2017; 37(1): 25-38.
[http://dx.doi.org/10.1177/0271678X16655814] [PMID: 27317654]
[17]
Yang J, Li Q, Wang Z, et al. Multimodality MRI assessment of grey and white matter injury and blood-brain barrier disruption after intracerebral haemorrhage in mice. Sci Rep 2017; 7: 40358.
[http://dx.doi.org/10.1038/srep40358] [PMID: 28084426]
[18]
He P, Zhang H, Hang D, et al. Research on the protective effect of diazoxide pretreatment on the blood-brain barrier of rats after cerebral ischemia/reperfusion injury. J Chin Physician (Chin) 2016; 18: 1309-12.
[19]
Li L, Wang P, Zhao H, Luo Y. Noncoding RNAs and intracerebral hemorrhage. CNS Neurol Disord Drug Targets 2019; 18(3): 205-11.
[http://dx.doi.org/10.2174/1871527318666190204102604] [PMID: 30714535]
[20]
Forouzanfar F, Shojapour M, Asgharzade S, Amini E. Causes and consequences of microRNA dysregulation following cerebral ischemia-reperfusion injury. CNS Neurol Disord Drug Targets 2019; 18(3): 212-21.
[http://dx.doi.org/10.2174/1871527318666190204104629] [PMID: 30714533]
[21]
Li F, Zhao H, Han Z, et al. Xuesaitong may protect against ischemic stroke by modulating microglial phenotypes and inhibiting neuronal cell apoptosis via the STAT3 signaling pathway. CNS Neurol Disord Drug Targets 2019; 18(2): 115-23.
[http://dx.doi.org/10.2174/1871527317666181114140340] [PMID: 30426907]
[22]
Mohammadi F, Nezafat N, Negahdaripour M, et al. Neuroprotective effects of heat shock protein70. CNS Neurol Disord Drug Targets 2018; 17(10): 736-42.
[http://dx.doi.org/10.2174/1871527317666180827111152] [PMID: 30147017]
[23]
Wang S, Ma F, Huang L, et al. Dl-3-n-butylphthalide (NBP): A promising therapeutic agent for ischemic stroke. CNS Neurol Disord Drug Targets 2018; 17(5): 338-47.
[http://dx.doi.org/10.2174/1871527317666180612125843] [PMID: 29895257]
[24]
Zhang X, Shu B, Zhang D, Huang L, Fu Q, Du G. The efficacy and safety of pharmacological treatments for post-stroke aphasia. CNS Neurol Disord Drug Targets 2018; 17(7): 509-21.
[http://dx.doi.org/10.2174/1871527317666180706143051] [PMID: 29984673]
[25]
Catalin B, Rogoveanu OC, Pirici I, et al. Cerebrolysin and aquaporin 4 inhibition improve pathological and motor recovery after ischemic stroke. CNS Neurol Disord Drug Targets 2018; 17(4): 299-308.
[http://dx.doi.org/10.2174/1871527317666180425124340] [PMID: 29692268]
[26]
Vasudeva K, Chaurasia P, Singh S, Munshi A. Genetic signatures in ischemic stroke: Focus on aspirin resistance. CNS Neurol Disord Drug Targets 2017; 16(9): 974-82.
[PMID: 28969559]
[27]
Crowley MG, Liska MG, Lippert T, Corey S, Borlongan CV. Utilizing delta opioid receptors and peptides for cytoprotection: Implications in stroke and other neurological disorders. CNS Neurol Disord Drug Targets 2017; 16(4): 414-24.
[http://dx.doi.org/10.2174/1871527316666170320150659] [PMID: 28322170]
[28]
Geng J, Wang L, Qu M, et al. Endothelial progenitor cells transplantation attenuated blood-brain barrier damage after ischemia in diabetic mice via HIF-1α. Stem Cell Res Ther 2017; 11(8): 163.
[29]
Imai T, Iwata S, Hirayama T, et al. Intracellular Fe2+ accumulation in endothelial cells and pericytes induces blood-brain barrier dysfunction in secondary brain injury after brain hemorrhage. Sci Rep 2019; 9(1): 6228.
[http://dx.doi.org/10.1038/s41598-019-42370-z] [PMID: 30996325]
[30]
Michinaga S, Koyama Y. Protection of the blood-brain barrier as a therapeutic strategy for brain damage. Biol Pharm Bull 2017; 40(5): 569-75.
[http://dx.doi.org/10.1248/bpb.b16-00991] [PMID: 28458343]
[31]
Wei X, Xu Y, Jin Y, Feng H, Xiao Y, Dong S. Granulocyte colony-stimulating factor attenuates blood-brain barrier damage and improves cognitive function in spontaneously hypertensive rats. CNS Neurol Disord Drug Targets 2017; 16(7): 781-8.
[http://dx.doi.org/10.2174/1871527316666170207155730] [PMID: 28176642]
[32]
Herrera AS, Ashraf GM, Del Carmen Arias Esparza M, et al. cerebrospinal fluid, brain electrolytes balance, the unsuspected intrinsic property of melanin to dissociate the water molecule. CNS Neurol Disord Drug Targets 2018; 17(10): 743-56.
[http://dx.doi.org/10.2174/1871527317666180904093430] [PMID: 30179148]
[33]
Alhadidi Q, Bin Sayeed MS, Shah ZA. The interplay between cofilin and phospho-cofilin: Its role in maintaining blood brain barrier integrity. CNS Neurol Disord Drug Targets 2017; 16(3): 279-90.
[http://dx.doi.org/10.2174/1871527316666170117115040] [PMID: 28124604]
[34]
Blaneo M, Lizasoain I, Sobrino T. Ischemic Preconditioning: A Novel Target for Neuroprotective Therapy. Cerebrovasc Dis 2006; 21: 38-47.
[http://dx.doi.org/10.1159/000091702]
[35]
Masada T, Hua Y, Xi G. Attenuation of ischemic brain edema and cerebrovascular injury after ischemic preconditioning in the rat. J Cereb Blood FIow Metab 2001; 2122
[36]
Zhang FY, Chen XC, Ren HM, Bao WM. Effects of ischemic preconditioning on blood-brain barrier permeability and MMP-9 expression of ischemic brain. Neurol Res 2006; 28(1): 21-4.
[http://dx.doi.org/10.1179/016164106X91825] [PMID: 16464358]
[37]
Kizmazoglu C, Aydin HE, Sevin IE, Kalemci O, Yüceer N, Atasoy MA. Neuroprotective effect of resveratrol on acute brain ischemia reperfusion injury by measuring annexin V, p53, Bcl-2 Levels in Rats. J Korean Neurosurg Soc 2015; 58(6): 508-12.
[http://dx.doi.org/10.3340/jkns.2015.58.6.508] [PMID: 26819684]
[38]
Fang L, Gao H, Zhang W, Zhang W, Wang Y. Resveratrol alleviates nerve injury after cerebral ischemia and reperfusion in mice by inhibiting inflammation and apoptosis. Int J Clin Exp Med 2015; 8(3): 3219-26.
[PMID: 26064211]
[39]
Jia JY, Tan ZG, Liu M, Jiang YG. Apurinic/apyrimidinic endonuclease 1 (APE1) contributes to resveratrol-induced neuroprotection against oxygen-glucose deprivation and re-oxygenation injury in HT22 cells: Involvement in reducing oxidative DNA damage. Mol Med Rep 2017; 16(6): 9786-94.
[http://dx.doi.org/10.3892/mmr.2017.7799] [PMID: 29039534]
[40]
He Q, Li Z, Wang Y, Hou Y, Li L, Zhao J. Resveratrol alleviates cerebral ischemia/reperfusion injury in rats by inhibiting NLRP3 inflammasome activation through Sirt1-dependent autophagy induction. Int Immunopharmacol 2017; 50: 208-15.
[http://dx.doi.org/10.1016/j.intimp.2017.06.029] [PMID: 28683365]
[41]
Jiang X, Andjelkovic A, Zhu L, et al. Blood-brain barrier dysfunction and recovery after ischemic stroke. Prog Neurobiol 2017.
[PMID: 28987927]
[42]
Leach JP, Heallen T, Zhang M, et al. Hippo pathway deficiency reverses systolic heart failure after infarction. Nature 2017; 550(7675): 260-4.
[http://dx.doi.org/10.1038/nature24045] [PMID: 28976966]
[43]
Matsuda T, Zhai P, Sciarretta S, et al. NF2 activates hippo signaling and promotes ischemia/reperfusion injury in the heart. Circ Res 2016; 119(5): 596-606.
[http://dx.doi.org/10.1161/CIRCRESAHA.116.308586] [PMID: 27402866]
[44]
Poon CL, Mitchell KA, Kondo S, Cheng LY, Harvey KF. The Hippo pathway regulates neuroblasts and brain size in Drosophila melanogaster. Curr Biol 2016; 26(8): 1034-42.
[http://dx.doi.org/10.1016/j.cub.2016.02.009] [PMID: 26996505]
[45]
Mindos T, Dun XP, North K, et al. Merlin controls the repair capacity of Schwann cells after injury by regulating Hippo/YAP activity. J Cell Biol 2017; 216(2): 495-510.
[http://dx.doi.org/10.1083/jcb.201606052] [PMID: 28137778]
[46]
Phng LK, Potente M, Leslie JD, et al. Nrarp coordinates endothelial Notch and WNT signaling to control vessel density in angiogenesis. Dev Cell 2009; 16(1): 70-82.
[http://dx.doi.org/10.1016/j.devcel.2008.12.009] [PMID: 19154719]
[47]
Zhang J, Fukuhara S, Sako K, et al. Angiopoietin-1/Tie2 signal augments basal Notch signal controlling vascular quiescence by inducing delta-like 4 expression through AKT-mediated activation of beta-catenin. J Biol Chem 2011; 286(10): 8055-66.
[http://dx.doi.org/10.1074/jbc.M110.192641] [PMID: 21212269]
[48]
Wang W, Li M, Wang Y, et al. GSK-3β as a target for protection against transient cerebral ischemia. Int J Med Sci 2017; 14(4): 333-9.
[http://dx.doi.org/10.7150/ijms.17514] [PMID: 28553165]
[49]
Wang Y, Rattner A, Zhou Y, Williams J, Smallwood PM, Nathans J. Norrin/Frizzled4 signaling in retinal vascular development and blood brain barrier plasticity. Cell 2012; 151(6): 1332-44.
[http://dx.doi.org/10.1016/j.cell.2012.10.042] [PMID: 23217714]
[50]
Yang B, Li W, Satani N, et al. Protective effects of autologous bone marrow mononuclear cells after administering t-PA in an embolic stroke model. Transl Stroke Res 2018; 9(2): 135-45.
[http://dx.doi.org/10.1007/s12975-017-0563-1] [PMID: 28836238]
[51]
Stamatovic SM, Martinez-Revollar G, Hu A, Choi J, Keep RF, Andjelkovic AV. Decline in Sirtuin-1 expression and activity plays a critical role in blood-brain barrier permeability in aging. Neurobiol Dis 2019; 126: 105-16.
[http://dx.doi.org/10.1016/j.nbd.2018.09.006] [PMID: 30196051]
[52]
Chen T, Dai SH, Li X, et al. Sirt1-Sirt3 axis regulates human blood-brain barrier permeability in response to ischemia. Redox Biol 2018; 14: 229-36.
[http://dx.doi.org/10.1016/j.redox.2017.09.016] [PMID: 28965081]
[53]
Zhang W, Zhang Y, Guo X, et al. Sirt1 Protects endothelial cells against LPS-induced barrier dysfunction. Oxid Med Cell Longev 2017; 2017 4082102
[http://dx.doi.org/10.1155/2017/4082102] [PMID: 29209448]
[54]
Jian C, Zou C, Xu N, Chen G, Zou D. Sirt1 protects neural stem cells from apoptosis by decreasing acetylation of histone 3K9. Stem Cells Cloning 2018; 11: 39-41.
[http://dx.doi.org/10.2147/SCCAA.S173852] [PMID: 30233218]
[55]
Rodriguez RM, Fernandez AF, Fraga MF. Role of sirtuins in stem cell differentiation. Genes Cancer 2013; 4(3-4): 105-11.
[http://dx.doi.org/10.1177/1947601913479798] [PMID: 24020001]
[56]
Hata S, Hirayama J, Kajiho H, et al. A novel acetylation cycle of transcription co-activator Yes-associated protein that is downstream of Hippo pathway is triggered in response to SN2 alkylating agents. J Biol Chem 2012; 287(26): 22089-98.
[http://dx.doi.org/10.1074/jbc.M111.334714] [PMID: 22544757]
[57]
Martins IJ. Anti-aging genes improve appetite regulation and reverse cell senescence and apoptosis in global populations. Adv Aging Res 2016; 5: 9-26.
[http://dx.doi.org/10.4236/aar.2016.51002]

Rights & Permissions Print Export Cite as
© 2024 Bentham Science Publishers | Privacy Policy