Generic placeholder image

Current Neurovascular Research

Editor-in-Chief

ISSN (Print): 1567-2026
ISSN (Online): 1875-5739

Research Article

Propofol Rescued Astrocytes from LPS-induced Inflammatory Response via Blocking LncRNA-MEG3/NF-κB Axis

Author(s): Fan Zhang, Zhihua Wang, Bei Sun, Yan Huang, Cheng Chen, Jie Hu, Longyan Li, Pingping Xia* and Zhi Ye*

Volume 19, Issue 1, 2022

Published on: 23 May, 2022

Page: [5 - 18] Pages: 14

DOI: 10.2174/1567202619666220316112509

open access plus

Abstract

Objective: Evidences demonstrate that propofol attenuates neuro-inflammation following brain ischemia. Moreover, LncRNA-MEG3 has been identified as an independent prognostic marker for ischemic stroke patients, and found to correlate to cerebral ischemia in animal models. Therefore, the current study explored the role of propofol in lipopolysaccharide (LPS)-mediated inflammation in cultured astrocytes, along with the molecular mechanism involved in LncRNAMEG3/ NF-κB axis.

Methods: The primary cultured astrocytes isolated from rats were used to establish an inflammatory model, which were treated with LPS. Propofol was administrated to the primary cultured astrocytes during LPS treatment. The effects of propofol on pro-inflammatory cytokines and the LncRNAMEG3/ NF-κB pathway were detected by ELISA, qRT-PCR and Western Blot assay, respectively. Then, dual-luciferase assay, chromatin immunoprecipitation and RNA immunoprecipitation were used to determine the interaction between LncRNA-MEG3 and NF-κB.

Results: Our study found propofol to significantly reduce LncRNA-MEG3 expression, which was elevated in LPS-stimulated astrocytes. Moreover, both propofol and LncRNA-MEG3 knockdown remarkably alleviated LPS-induced cytotoxicity by suppressing expressions and release of proinflammatory cytokines. Loss of LncRNA-MEG3 notably suppressed the NF-κB activity and its phosphorylated activation. Additionally, it was also observed that LncRNA-MEG3 could bind nuclear p65/p50, and promote the binding of NF-κB to IL-6 and TNF-α promoters in the nucleus, subsequently stimulating the production of inflammatory cytokines in LPS-treated astrocytes. Furthermore, a specific inhibitor of NF-κB, PDTC, rescued astrocytes from LPS exposure without affecting the LncRNA-MEG3 expression.

Conclusion: These findings demonstrate that LncRNA-MEG3 acts as a positive regulator of NF-κB, mediating the neuroprotection of propofol in LPS-triggered astrocytes injury.

Keywords: Propofol, cerebral ischemic injury, inflammation, MEG3, NF-kB, astrocytes.

[1]
Cipolla MJ, Liebeskind DS, Chan SL. The importance of comorbidities in ischemic stroke: Impact of hypertension on the cerebral circula-tion. J Cereb Blood Flow Metab 2018; 38(12): 2129-49.
[http://dx.doi.org/10.1177/0271678X18800589] [PMID: 30198826]
[2]
Winship IR. Cerebral collaterals and collateral therapeutics for acute ischemic stroke. Microcirculation 2015; 22(3): 228-36.
[http://dx.doi.org/10.1111/micc.12177] [PMID: 25351102]
[3]
Grysiewicz RA, Thomas K, Pandey DK. Epidemiology of ischemic and hemorrhagic stroke: Incidence, prevalence, mortality, and risk factors. Neurol Clin 2008; 26(4): 871-95. [vii.
[http://dx.doi.org/10.1016/j.ncl.2008.07.003] [PMID: 19026895]
[4]
Jiang Q, Stone CR, Elkin K, Geng X, Ding Y. Immunosuppression and neuroinflammation in stroke pathobiology. Exp Neurobiol 2021; 30(2): 101-12.
[http://dx.doi.org/10.5607/en20033] [PMID: 33972464]
[5]
Anrather J, Iadecola C. Inflammation and stroke: An overview. Neurotherapeutics 2016; 13(4): 661-70.
[http://dx.doi.org/10.1007/s13311-016-0483-x] [PMID: 27730544]
[6]
Weber RZ, Perron P, Rust R. Astrocytes for brain repair: More than just a neuron’s sidekick. Brain Pathol 2021; 31(5), e12999.
[http://dx.doi.org/10.1111/bpa.12999] [PMID: 34196052]
[7]
Liu Z, Chopp M. Astrocytes, therapeutic targets for neuroprotection and neurorestoration in ischemic stroke. Prog Neurobiol 2016; 144: 103-20.
[http://dx.doi.org/10.1016/j.pneurobio.2015.09.008] [PMID: 26455456]
[8]
Sims NR, Yew WP. Reactive astrogliosis in stroke: Contributions of astrocytes to recovery of neurological function. Neurochem Int 2017; 107: 88-103.
[http://dx.doi.org/10.1016/j.neuint.2016.12.016] [PMID: 28057555]
[9]
Yu H, Wang X, Kang F, Chen Z, Meng Y, Dai M. Propofol attenuates inflammatory damage on neurons following cerebral infarction by inhibiting excessive activation of microglia. Int J Mol Med 2019; 43(1): 452-60.
[http://dx.doi.org/10.3892/ijmm.2018.3974] [PMID: 30431058]
[10]
Shi SS, Yang WZ, Chen Y, Chen JP, Tu XK. Propofol reduces inflammatory reaction and ischemic brain damage in cerebral ischemia in rats. Neurochem Res 2014; 39(5): 793-9.
[http://dx.doi.org/10.1007/s11064-014-1272-8] [PMID: 24610527]
[11]
Ma G, Chen J, Meng X, Deng L, Gao Y, Meng J. High-dose propofol reduces S-100β protein and neuron-specific enolase levels in patients undergoing cardiac surgery. J Cardiothorac Vasc Anesth 2013; 27(3): 510-5.
[http://dx.doi.org/10.1053/j.jvca.2012.10.013] [PMID: 23561840]
[12]
Sun B, Ou H, Ren F, et al. Propofol inhibited autophagy through Ca2+/CaMKKβ/AMPK/mTOR pathway in OGD/R-induced neuron injury. Mol Med 2018; 24(1): 58.
[http://dx.doi.org/10.1186/s10020-018-0054-1] [PMID: 30470173]
[13]
Wu Z, Wu P, Zuo X, et al. LncRNA-N1LR enhances neuroprotection against ischemic stroke probably by inhibiting p53 phosphorylation. Mol Neurobiol 2017; 54(10): 7670-85.
[http://dx.doi.org/10.1007/s12035-016-0246-z] [PMID: 27844279]
[14]
Cao DW, Liu MM, Duan R, et al. The lncRNA Malat1 functions as a ceRNA to contribute to berberine-mediated inhibition of HMGB1 by sponging miR-181c-5p in poststroke inflammation. Acta Pharmacol Sin 2020; 41(1): 22-33.
[http://dx.doi.org/10.1038/s41401-019-0284-y] [PMID: 31431734]
[15]
Ghafouri-Fard S, Shirvani-Farsani Z, Hussen BM, Taheri M, Arefian N. Emerging impact of non-coding RNAs in the pathology of stroke. Front Aging Neurosci 2021; 13, 780489.
[http://dx.doi.org/10.3389/fnagi.2021.780489] [PMID: 34867304]
[16]
Chen C, Huang Y, Xia P, et al. Long noncoding RNA Meg3 mediates ferroptosis induced by oxygen and glucose deprivation combined with hyperglycemia in rat brain microvascular endothelial cells, through modulating the p53/GPX4 axis. Eur J Histochem 2021; 65(3): 3224.
[http://dx.doi.org/10.4081/ejh.2021.3224] [PMID: 34587716]
[17]
Liu J, Li Q, Zhang KS, et al. Downregulation of the long non-coding RNA Meg3 promotes angiogenesis after ischemic brain injury by activating Notch signaling. Mol Neurobiol 2017; 54(10): 8179-90.
[http://dx.doi.org/10.1007/s12035-016-0270-z] [PMID: 27900677]
[18]
Yan H, Rao J, Yuan J, et al. Long non-coding RNA MEG3 functions as a competing endogenous RNA to regulate ischemic neuronal death by targeting miR-21/PDCD4 signaling pathway. Cell Death Dis 2017; 8(12): 3211.
[http://dx.doi.org/10.1038/s41419-017-0047-y] [PMID: 29238035]
[19]
Zha F, Qu X, Tang B, et al. Long non-coding RNA MEG3 promotes fibrosis and inflammatory response in diabetic nephropathy via miR-181a/Egr-1/TLR4 axis. Aging (Albany NY) 2019; 11(11): 3716-30.
[http://dx.doi.org/10.18632/aging.102011] [PMID: 31195367]
[20]
Yan L, Liu Z, Yin H, Guo Z, Luo Q. Silencing of MEG3 inhibited ox-LDL-induced inflammation and apoptosis in macrophages via mod-ulation of the MEG3/miR-204/CDKN2A regulatory axis. Cell Biol Int 2019; 43(4): 409-20.
[http://dx.doi.org/10.1002/cbin.11105] [PMID: 30672051]
[21]
Fann DY, Lim YA, Cheng YL, et al. Evidence that NF-kappa B and MAPK signaling promotes NLRP inflammasome activation in neurons following ischemic stroke. Mol Neurobiol 2018; 55(2): 1082-96.
[http://dx.doi.org/10.1007/s12035-017-0394-9] [PMID: 28092085]
[22]
Howell JA, Bidwell GL III. Targeting the NF-κB pathway for therapy of ischemic stroke. Ther Deliv 2020; 11(2): 113-23.
[http://dx.doi.org/10.4155/tde-2019-0075] [PMID: 31928138]
[23]
Mettang M, Reichel SN, Lattke M, et al. IKK2/NF-κB signaling protects neurons after traumatic brain injury. FASEB J 2018; 32(4): 1916-32.
[http://dx.doi.org/10.1096/fj.201700826R] [PMID: 29187362]
[24]
Deng YL, Ma YL, Zhang ZL, et al. Astrocytic N-Myc downstream-regulated Gene-2 is involved in nuclear transcription factor kappa B-mediated inflammation induced by global cerebral ischemia. Anesthesiology 2018; 128(3): 574-86.
[http://dx.doi.org/10.1097/ALN.0000000000002044] [PMID: 29252510]
[25]
Tan L, Li J, Wang Y, Tan R. Anti-neuroinflammatory effect of alantolactone through the suppression of the NF-kappa B and MAPK sig-naling pathways. Cells 2019; 8(7): 739.
[http://dx.doi.org/10.3390/cells8070739] [PMID: 31323885]
[26]
Cui DR, Wang L, Jiang W, Qi AH, Zhou QH, Zhang XL. Propofol prevents cerebral ischemia-triggered autophagy activation and cell death in the rat hippocampus through the NF-κB/p53 signaling pathway. Neuroscience 2013; 246: 117-32.
[http://dx.doi.org/10.1016/j.neuroscience.2013.04.054] [PMID: 23644056]
[27]
Zhang Y, Wu J, Jing H, Huang G, Sun Z, Xu S. Long noncoding RNA MEG3 inhibits breast cancer growth via upregulating endoplasmic reticulum stress and activating NF-κB and p53. J Cell Biochem 2019; 120(4): 6789-97.
[http://dx.doi.org/10.1002/jcb.27982] [PMID: 30556250]
[28]
Bao D, Yuan RX, Zhang Y. Effects of lncRNA MEG3 on proliferation and apoptosis of gallbladder cancer cells through regulating NF-κB signaling pathway. Eur Rev Med Pharmacol Sci 2020; 24(12): 6632-8.
[http://dx.doi.org/10.26355/eurrev_202006_21649] [PMID: 32633352]
[29]
Xia P, Pan Y, Zhang F, et al. Pioglitazone confers neuroprotection against ischemia-induced pyroptosis due to its inhibitory effects on HMGB-1/RAGE and Rac1/ROS pathway by activating PPAR-γ. Cell Physiol Biochem 2018; 45(6): 2351-68.
[http://dx.doi.org/10.1159/000488183] [PMID: 29554649]
[30]
Pan Y, Wang N, Xia P, Wang E, Guo Q, Ye Z. Inhibition of Rac1 ameliorates neuronal oxidative stress damage via reducing Bcl-2/Rac1 complex formation in mitochondria through PI3K/Akt/mTOR pathway. Exp Neurol 2018; 300: 149-66.
[http://dx.doi.org/10.1016/j.expneurol.2017.10.030] [PMID: 29129468]
[31]
Dabrowska S, Andrzejewska A, Kozlowska H, Strzemecki D, Janowski M, Lukomska B. Neuroinflammation evoked by brain injury in a rat model of lacunar infarct. Exp Neurol 2021; 336, 113531.
[http://dx.doi.org/10.1016/j.expneurol.2020.113531] [PMID: 33221395]
[32]
Shaheryar ZA, Khan MA, Adnan CS, Zaidi AA, Hänggi D, Muhammad S. Neuroinflammatory triangle presenting novel pharmacological targets for ischemic brain injury. Front Immunol 2021; 12, 748663.
[http://dx.doi.org/10.3389/fimmu.2021.748663] [PMID: 34691061]
[33]
Chen CX, Huang J, Tu GQ, et al. NAMPT inhibitor protects ischemic neuronal injury in rat brain via anti-neuroinflammation. Neuroscience 2017; 356: 193-206.
[http://dx.doi.org/10.1016/j.neuroscience.2017.05.022] [PMID: 28528966]
[34]
Revuelta M, Elicegui A, Moreno-Cugnon L, Bührer C, Matheu A, Schmitz T. Ischemic stroke in neonatal and adult astrocytes. Mech Ageing Dev 2019; 183, 111147.
[http://dx.doi.org/10.1016/j.mad.2019.111147] [PMID: 31493435]
[35]
Liu H, Wu X, Luo J, et al. Pterostilbene attenuates astrocytic inflammation and neuronal oxidative injury after ischemia-reperfusion by inhibiting NF-kappa B phosphorylation. Front Immunol 2019; 10: 2408.
[http://dx.doi.org/10.3389/fimmu.2019.02408] [PMID: 31681297]
[36]
Zhang M, Ma Y, Chai L, Mao H, Zhang J, Fan X. Storax protected oxygen-glucose deprivation/reoxygenation induced primary astrocyte injury by inhibiting NF-kappa B activation in vitro. Front Pharmacol 2019; 9: 1527.
[http://dx.doi.org/10.3389/fphar.2018.01527] [PMID: 30687092]
[37]
Sommerkamp P, Renders S, Ladel L, et al. The long non-coding RNA Meg3 is dispensable for hematopoietic stem cells. Sci Rep 2019; 9(1): 2110.
[http://dx.doi.org/10.1038/s41598-019-38605-8] [PMID: 30765776]
[38]
Zhao H, Wang X, Feng X, et al. Long non-coding RNA MEG3 regulates proliferation, apoptosis, and autophagy and is associated with prognosis in glioma. J Neurooncol 2018; 140(2): 281-8.
[http://dx.doi.org/10.1007/s11060-018-2874-9] [PMID: 30284203]
[39]
Zhan R, Xu K, Pan J, Xu Q, Xu S, Shen J. Long noncoding RNA MEG3 mediated angiogenesis after cerebral infarction through regulating p53/NOX4 axis. Biochem Biophys Res Commun 2017; 490(3): 700-6.
[http://dx.doi.org/10.1016/j.bbrc.2017.06.104] [PMID: 28634073]
[40]
Liang J, Wang Q, Li JQ, Guo T, Yu D. Long non-coding RNA MEG3 promotes cerebral ischemia-reperfusion injury through increasing pyroptosis by targeting miR-485/AIM2 axis. Exp Neurol 2020; 325, 113139.
[http://dx.doi.org/10.1016/j.expneurol.2019.113139] [PMID: 31794744]
[41]
Schwaninger M, Sallmann S, Petersen N, et al. Bradykinin induces interleukin-6 expression in astrocytes through activation of nuclear factor-kappaB. J Neurochem 1999; 73(4): 1461-6.
[http://dx.doi.org/10.1046/j.1471-4159.1999.0731461.x] [PMID: 10501190]
[42]
Shu Q, Fan H, Li SJ, et al. Protective effects of Progranulin against focal cerebral ischemia-reperfusion injury in rats by suppressing endo-plasmic reticulum stress and NF-κB activation in reactive astrocytes. J Cell Biochem 2018; 119(8): 6584-97.
[http://dx.doi.org/10.1002/jcb.26790] [PMID: 29665049]
[43]
Kuprash DV, Udalova IA, Turetskaya RL, Kwiatkowski D, Rice NR, Nedospasov SA. Similarities and differences between human and murine TNF promoters in their response to lipopolysaccharide. J Immunol 1999; 162(7): 4045-52.
[PMID: 10201927]
[44]
Dendorfer U, Oettgen P, Libermann TA. Multiple regulatory elements in the interleukin-6 gene mediate induction by prostaglandins, cyclic AMP, and lipopolysaccharide. Mol Cell Biol 1994; 14(7): 4443-54.
[http://dx.doi.org/10.1128/mcb.14.7.4443-4454.1994] [PMID: 8007951]
[45]
IIott NE, Heward JA, Roux B, et al.. Long non-coding RNAs and enhancer RNAs regulate the lipopolysaccharide-induced inflammatory response in human monocytes. Nat Commun 2014; 5: 3979.
[http://dx.doi.org/10.1038/ncomms4979] [PMID: 24909122]
[46]
Tao XW, Zeng LK, Wang HZ, Liu HC. LncRNA MEG3 ameliorates respiratory syncytial virus infection by suppressing TLR4 signaling. Mol Med Rep 2018; 17(3): 4138-44.
[http://dx.doi.org/10.3892/mmr.2017.8303] [PMID: 29257348]

© 2024 Bentham Science Publishers | Privacy Policy