Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

General Research Article

Inhibition of microRNA-155 Alleviates Neurological Dysfunction Following Transient Global Ischemia and Contribution of Neuroinflammation and Oxidative Stress in the Hippocampus

Author(s): Lichao Sun, Shouqin Ji and Jihong Xing*

Volume 25, Issue 40, 2019

Page: [4310 - 4317] Pages: 8

DOI: 10.2174/1381612825666190926162229

Price: $65

Abstract

Background/Aims: Central pro-inflammatory cytokine (PIC) signal is involved in neurological deficits after transient global ischemia induced by cardiac arrest (CA). The present study was to examine the role of microRNA- 155 (miR-155) in regulating IL-1β, IL-6 and TNF-α in the hippocampus of rats with induction of CA. We further examined the levels of products of oxidative stress 8-isoprostaglandin F2α (8-iso PGF2α, indication of oxidative stress); and 8-hydroxy-2’-deoxyguanosine (8-OHdG, indication of protein oxidation) after cerebral inhibition of miR-155.

Methods: CA was induced by asphyxia and followed by cardiopulmonary resuscitation in rats. ELISA and western blot analysis were used to determine the levels of PICs and products of oxidative stress; and the protein expression of NADPH oxidase (NOXs) in the hippocampus. In addition, neurological severity score and brain edema were examined to assess neurological functions.

Results: We observed amplification of IL-1β, IL-6 and TNF-α along with 8-iso PGF2α and 8-OHdG in the hippocampus of CA rats. Cerebral administration of miR-155 inhibitor diminished upregulation of PICs in the hippocampus. This also attenuated products of oxidative stress and upregulation of NOX4. Notably, inhibition of miR-155 improved neurological severity score and brain edema and this was linked to signal pathways of PIC and oxidative stress.

Conclusion: We showed the significant role of blocking miR-155 signal in improving the neurological function in CA rats likely via inhibition of signal pathways of neuroinflammation and oxidative stress, suggesting that miR-155 may be a target in preventing and/or alleviating development of the impaired neurological functions during CA-evoked global cerebral ischemia.

Keywords: microRNA-155, cardiac arrest, cardiopulmonary resuscitation, hippocampus, protein oxidation, asphyxia.

« Previous
[1]
Nolan JP, Neumar RW, Adrie C, et al. Post-cardiac arrest syndrome: epidemiology, pathophysiology, treatment, and prognostication: a scientific statement from the international liaison committee on resuscitation; the american heart association emergency cardiovascular care committee; the council on cardiovascular surgery and anesthesia; the council on cardiopulmonary, perioperative, and critical care; the council on clinical cardiology; the council on stroke. Resuscitation 2008; 79(3): 350-79.
[http://dx.doi.org/10.1016/j.resuscitation.2008.09.017] [PMID: 18963350]
[2]
Wang GN, Chen XF, Zhang G, et al. A case of thyroid emergency with cardiac arrest supported by extracorporeal membrane oxygenation. World J Emerg Med 2018; 9(4): 288-90.
[http://dx.doi.org/10.5847/wjem.j.1920-8642.2018.04.009] [PMID: 30181798]
[3]
Peng Z-R, Yang AL, Yang Q-D. The effect of hyperbaric oxygen on intracephalic angiogenesis in rats with intracerebral hemorrhage. J Neurol Sci 2014; 342(1-2): 114-23.
[http://dx.doi.org/10.1016/j.jns.2014.04.037] [PMID: 24836574]
[4]
Ojaghihaghighi S, Vahdati SS, Mikaeilpour A, Ramouz A. Comparison of neurological clinical manifestation in patients with hemorrhagic and ischemic stroke. World J Emerg Med 2017; 8(1): 34-8.
[http://dx.doi.org/10.5847/wjem.j.1920-8642.2017.01.006] [PMID: 28123618]
[5]
Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009; 136(2): 215-33.
[http://dx.doi.org/10.1016/j.cell.2009.01.002] [PMID: 19167326]
[6]
Sayed D, Abdellatif M. MicroRNAs in development and disease. Physiol Rev 2011; 91(3): 827-87.
[http://dx.doi.org/10.1152/physrev.00006.2010] [PMID: 21742789]
[7]
Eacker SM, Dawson TM, Dawson VL. Understanding microRNAs in neurodegeneration. Nat Rev Neurosci 2009; 10(12): 837-41.
[http://dx.doi.org/10.1038/nrn2726] [PMID: 19904280]
[8]
Farazi TA, Spitzer JI, Morozov P, Tuschl T. miRNAs in human cancer. J Pathol 2011; 223(2): 102-15.
[http://dx.doi.org/10.1002/path.2806] [PMID: 21125669]
[9]
Han M, Toli J, Abdellatif M. MicroRNAs in the cardiovascular system. Curr Opin Cardiol 2011; 26(3): 181-9.
[http://dx.doi.org/10.1097/HCO.0b013e328345983d] [PMID: 21464712]
[10]
Gambari R, Fabbri E, Borgatti M, et al. Targeting microRNAs involved in human diseases: a novel approach for modification of gene expression and drug development. Biochem Pharmacol 2011; 82(10): 1416-29.
[http://dx.doi.org/10.1016/j.bcp.2011.08.007] [PMID: 21864506]
[11]
Calame K. MicroRNA-155 function in B Cells. Immunity 2007; 27(6): 825-7.
[http://dx.doi.org/10.1016/j.immuni.2007.11.010] [PMID: 18093533]
[12]
Elton TS, Selemon H, Elton SM, Parinandi NL. Regulation of the MIR155 host gene in physiological and pathological processes. Gene 2013; 532(1): 1-12.
[http://dx.doi.org/10.1016/j.gene.2012.12.009] [PMID: 23246696]
[13]
Faraoni I, Antonetti FR, Cardone J, Bonmassar E. MiR-155 gene: a typical multifunctional microRNA. Biochim Biophys Acta 2009; 1792(6): 497-505.
[http://dx.doi.org/10.1016/j.bbadis.2009.02.013] [PMID: 19268705]
[14]
O’Connell RM, Rao DS, Baltimore D. MicroRNA regulation of inflammatory responses. Annu Rev Immunol 2012; 30: 295-312.
[http://dx.doi.org/10.1146/annurev-immunol-020711-075013] [PMID: 22224773]
[15]
Sonkoly E, Janson P, Majuri ML, et al. MiR-155 is overexpressed in patients with atopic dermatitis and modulates T-cell proliferative responses by targeting cytotoxic T lymphocyte-associated antigen 4 Allergy Clin Immunol 2010; 126(3): 581-9.e1-20.
[16]
Moore CS, Rao VT, Durafourt BA, et al. miR-155 as a multiple sclerosis-relevant regulator of myeloid cell polarization. Ann Neurol 2013; 74(5): 709-20.
[http://dx.doi.org/10.1002/ana.23967] [PMID: 23818336]
[17]
Marttinen M, Takalo M, Natunen T, et al. Molecular mechanisms of synaptotoxicity and neuroinflammation in alzheimer’s disease. Front Neurosci 2018; 12: 963.
[http://dx.doi.org/10.3389/fnins.2018.00963] [PMID: 30618585]
[18]
Sharma P, Srivastava P, Seth A, Tripathi PN, Banerjee AG, Shrivastava SK. Comprehensive review of mechanisms of pathogenesis involved in alzheimer’s disease and potential therapeutic strategies. Prog Neurobiol 2019; 174: 53-89.
[http://dx.doi.org/10.1016/j.pneurobio.2018.12.006] [PMID: 30599179]
[19]
Wang CX, Shuaib A. Involvement of inflammatory cytokines in central nervous system injury. Prog Neurobiol 2002; 67(2): 161-72.
[http://dx.doi.org/10.1016/S0301-0082(02)00010-2] [PMID: 12126659]
[20]
Fu CY, He XY, Li XF, et al. Nefiracetam attenuates pro-inflammatory cytokines and gaba transporter in specific brain regions of rats with post-ischemic seizures. Cell Physiol Biochem 2015; 37(5): 2023-31.
[http://dx.doi.org/10.1159/000438562] [PMID: 26584300]
[21]
Saito K, Suyama K, Nishida K, Sei Y, Basile AS. Early increases in TNF-alpha, IL-6 and IL-1 beta levels following transient cerebral ischemia in gerbil brain. Neurosci Lett 1996; 206(2-3): 149-52.
[http://dx.doi.org/10.1016/S0304-3940(96)12460-5] [PMID: 8710173]
[22]
Oppenheim JJ. Cytokines: past, present, and future. Int J Hematol 2001; 74(1): 3-8.
[http://dx.doi.org/10.1007/BF02982543] [PMID: 11530802]
[23]
Xing J, Lu J. HIF-1alpha activation attenuates IL-6 and TNF-alpha pathways in hippocampus of rats following transient global ischemia. Cell Physiol Biochem 2016; 39(2): 511-20.
[http://dx.doi.org/10.1159/000445643] [PMID: 27383646]
[24]
Liu XL, Lu J, Xing J. Stabilization of HIF-1α modulates VEGF and Caspase-3 in the hippocampus of rats following transient global ischemia induced by asphyxial cardiac arrest. Life Sci 2016; 151: 243-9.
[http://dx.doi.org/10.1016/j.lfs.2016.03.005] [PMID: 26987747]
[25]
Sochocka M, Koutsouraki ES, Gasiorowski K, Leszek J. Vascular oxidative stress and mitochondrial failure in the pathobiology of alzheimer’s disease: a new approach to therapy. CNS Neurol Disord Drug Targets 2013; 12(6): 870-81.
[http://dx.doi.org/10.2174/18715273113129990072] [PMID: 23469836]
[26]
Yan MH, Wang X, Zhu X. Mitochondrial defects and oxidative stress in alzheimer disease and Parkinson disease. Free Radic Biol Med 2013; 62: 90-101.
[http://dx.doi.org/10.1016/j.freeradbiomed.2012.11.014] [PMID: 23200807]
[27]
Butterfield DA, Swomley AM, Sultana R. Amyloid β-peptide (1-42)-induced oxidative stress in alzheimer disease: importance in disease pathogenesis and progression. Antioxid Redox Signal 2013; 19(8): 823-35.
[http://dx.doi.org/10.1089/ars.2012.5027] [PMID: 23249141]
[28]
Zhang W, Wang L, Pang X, Zhang J, Guan Y. Role of microRNA-155 in modifying neuroinflammation and γ-aminobutyric acid transporters in specific central regions after post-ischaemic seizures. J Cell Mol Med 2019; 23(8): 5017-24.
[http://dx.doi.org/10.1111/jcmm.14358] [PMID: 31144434]
[29]
Xu S, Wu Q, Guo G, Ding X. The protective effects of urocortin1 against intracerebral hemorrhage by activating JNK1/2 and p38 phosphorylation and further increasing VEGF via corticotropin-releasing factor receptor 2. Neurosci Lett 2015; 589: 31-6.
[http://dx.doi.org/10.1016/j.neulet.2015.01.015] [PMID: 25576701]
[30]
Zhang Y, Yi B, Ma J, et al. Quercetin promotes neuronal and behavioral recovery by suppressing inflammatory response and apoptosis in a rat model of intracerebral hemorrhage. Neurochem Res 2015; 40(1): 195-203.
[http://dx.doi.org/10.1007/s11064-014-1457-1] [PMID: 25543848]
[31]
Karthikeyan A, Patnala R, Jadhav SP, Eng-Ang L, Dheen ST. MicroRNAs: key players in microglia and astrocyte mediated inflammation in CNS pathologies. Curr Med Chem 2016; 23(30): 3528-46.
[http://dx.doi.org/10.2174/0929867323666160814001040] [PMID: 27528056]
[32]
Khoshnam SE, Winlow W, Farzaneh M. The interplay of MicroRNAs in the inflammatory mechanisms following ischemic stroke. J Neuropathol Exp Neurol 2017; 76(7): 548-61.
[http://dx.doi.org/10.1093/jnen/nlx036] [PMID: 28535304]
[33]
Roitbak T. Silencing a multifunctional microrna is beneficial for stroke recovery. Front Mol Neurosci 2018; 11: 58.
[http://dx.doi.org/10.3389/fnmol.2018.00058] [PMID: 29527155]
[34]
Cardoso AL, Guedes JR, Pereira de Almeida L, Pedroso de Lima MC. miR-155 modulates microglia-mediated immune response by down-regulating SOCS-1 and promoting cytokine and nitric oxide production. Immunology 2012; 135(1): 73-88.
[http://dx.doi.org/10.1111/j.1365-2567.2011.03514.x] [PMID: 22043967]
[35]
Yasukawa H, Sasaki A, Yoshimura A. Negative regulation of cytokine signaling pathways. Annu Rev Immunol 2000; 18: 143-64.
[http://dx.doi.org/10.1146/annurev.immunol.18.1.143] [PMID: 10837055]
[36]
Chinen T, Kobayashi T, Ogata H, et al. Suppressor of cytokine signaling-1 regulates inflammatory bowel disease in which both IFNgamma and IL-4 are involved. Gastroenterology 2006; 130(2): 373-88.
[http://dx.doi.org/10.1053/j.gastro.2005.10.051] [PMID: 16472593]
[37]
Hanada T, Yoshida H, Kato S, et al. Suppressor of cytokine signaling-1 is essential for suppressing dendritic cell activation and systemic autoimmunity. Immunity 2003; 19(3): 437-50.
[http://dx.doi.org/10.1016/S1074-7613(03)00240-1] [PMID: 14499118]
[38]
Eisenhardt SU, Weiss JB, Smolka C, et al. MicroRNA-155 aggravates ischemia-reperfusion injury by modulation of inflammatory cell recruitment and the respiratory oxidative burst. Basic Res Cardiol 2015; 110(3): 32.
[http://dx.doi.org/10.1007/s00395-015-0490-9] [PMID: 25916938]
[39]
Wen Y, Zhang X, Dong L, Zhao J, Zhang C, Zhu C. Acetylbritannilactone modulates MicroRNA-155-mediated inflammatory response in ischemic cerebral tissues. Mol Med 2015; 21: 197-209.
[http://dx.doi.org/10.2119/molmed.2014.00199] [PMID: 25811992]
[40]
Altenhöfer S, Kleikers PW, Radermacher KA, et al. The NOX toolbox: validating the role of NADPH oxidases in physiology and disease. Cell Mol Life Sci 2012; 69(14): 2327-43.
[http://dx.doi.org/10.1007/s00018-012-1010-9] [PMID: 22648375]
[41]
Salvemini D, Little JW, Doyle T, Neumann WL. Roles of reactive oxygen and nitrogen species in pain. Free Radic Biol Med 2011; 51(5): 951-66.
[http://dx.doi.org/10.1016/j.freeradbiomed.2011.01.026] [PMID: 21277369]
[42]
Lam GY, Huang J, Brumell JH. The many roles of NOX2 NADPH oxidase-derived ROS in immunity. Semin Immunopathol 2010; 32(4): 415-30.
[http://dx.doi.org/10.1007/s00281-010-0221-0] [PMID: 20803017]
[43]
Gavazzi G, Banfi B, Deffert C, et al. Decreased blood pressure in NOX1-deficient mice. FEBS Lett 2006; 580(2): 497-504.
[http://dx.doi.org/10.1016/j.febslet.2005.12.049] [PMID: 16386251]
[44]
Suzuki Y, Hattori K, Hamanaka J, et al. Pharmacological inhibition of TLR4-NOX4 signal protects against neuronal death in transient focal ischemia. Sci Rep 2012; 2: 896.
[http://dx.doi.org/10.1038/srep00896] [PMID: 23193438]
[45]
Kallenborn-Gerhardt W, Schröder K, Del Turco D, et al. NADPH oxidase-4 maintains neuropathic pain after peripheral nerve injury. J Neurosci 2012; 32(30): 10136-45.
[http://dx.doi.org/10.1523/JNEUROSCI.6227-11.2012] [PMID: 22836249]
[46]
Lu J, Shen Y, Qian HY, et al. Effects of mild hypothermia on the ROS and expression of caspase-3 mRNA and LC3 of hippocampus nerve cells in rats after cardiopulmonary resuscitation. World J Emerg Med 2014; 5(4): 298-305.
[http://dx.doi.org/10.5847/wjem.j.issn.1920-8642.2014.04.010] [PMID: 25548605]
[47]
Fei M, Cai WW, Zhou SA. Characteristics and outcomes of out-of-hospital cardiac arrest in Zhejiang Province. World J Emerg Med 2018; 9(2): 141-3.
[http://dx.doi.org/10.5847/wjem.j.1920-8642.2018.02.010] [PMID: 29576828]
[48]
Pei L, Meng S, Yu W, Wang Q, Song F, Ma L. Inhibition of MicroRNA-383 ameliorates injury after focal cerebral ischemia via targeting PPARgamma. Cell Physiol Biochem 2016; 39(4): 1339-46.
[http://dx.doi.org/10.1159/000447838] [PMID: 27607022]
[49]
Wang J, Xu Z, Chen X, et al. MicroRNA-182-5p attenuates cerebral ischemia-reperfusion injury by targeting Toll-like receptor 4. Biochem Biophys Res Commun 2018; 505(3): 677-84.
[http://dx.doi.org/10.1016/j.bbrc.2018.09.165] [PMID: 30292407]
[50]
Yu Y, Zhang X, Han Z, Zhao W, Zhang L. Expression and regulation of miR-449a and AREG in cerebral ischemic injury. Metab Brain Dis 2019; 34(3): 821-32.
[http://dx.doi.org/10.1007/s11011-019-0393-9] [PMID: 30773606]
[51]
Zuo X, Lu J, Manaenko A, et al. MicroRNA-132 attenuates cerebral injury by protecting blood-brain-barrier in MCAO mice. Exp Neurol 2019; 316: 12-9.
[http://dx.doi.org/10.1016/j.expneurol.2019.03.017] [PMID: 30930097]
[52]
Kim J, Seo M, Kim SK, Bae YS. Flagellin-induced NADPH oxidase 4 activation is involved in atherosclerosis. Sci Rep 2016; 6: 25437.
[http://dx.doi.org/10.1038/srep25437] [PMID: 27146088]
[53]
Liu T, Zhang L, Joo D, Sun SC. NF-κB signaling in inflammation. Signal Transduct Target Ther 2017; 2: 17023.
[http://dx.doi.org/10.1038/sigtrans.2017.23] [PMID: 29158945]
[54]
Pahwa R, Jialal I. Hyperglycemia induces toll-like receptor activity through increased oxidative stress. Metab Syndr Relat Disord 2016; 14(5): 239-41.
[http://dx.doi.org/10.1089/met.2016.29006.pah] [PMID: 27105077]

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