Expression of Nik-related kinase in smooth muscle cells attenuates vascular inflammation and intimal hyperplasia

Yi-Jhu Lu, Yee-Jee Jan, Bor-Sheng Ko, Shu-Man Liang, Lujen Chen, Chih-Cheng Wu, Chih-Hui Chin, Cheng-Chin Kuo, Shaw-Fang Yet, Jun-Yang Liou, Yi-Jhu Lu, Yee-Jee Jan, Bor-Sheng Ko, Shu-Man Liang, Lujen Chen, Chih-Cheng Wu, Chih-Hui Chin, Cheng-Chin Kuo, Shaw-Fang Yet, Jun-Yang Liou

Abstract

Inflammation of the vascular microenvironment modulates distinct types of vascular cells, and plays important roles in promoting atherosclerosis, stenosis/restenosis, and vascular-related diseases. Nik-related kinase (Nrk), a member of the Ste20-type kinase family, has been reported to be selectively expressed in embryonic skeletal muscle. However, whether Nrk is expressed in adult vascular smooth muscle, and if it influences intimal hyperplasia is unclear. Here, we found that Nrk is abundantly expressed in cultured vascular smooth muscle cells (VSMC) and mouse arterial intima. Treatment of mouse VSMCs with lipopolysaccharide (LPS) or platelet-derived growth factor significantly reduced Nrk expression. In addition, expression of Nrk was significantly reduced in regions of neointimal formation caused by guide-wire carotid artery injuries in mice, as well as in human atherosclerotic tissues, when compared to normal vessels. We identified that expression of matrix metalloproteinases (MMP3, MMP8 and MMP12) and inflammatory cytokines/chemokines (CCL6, CCL8, CCL11, CXCL1, CXCL3, CXCL5 and CXCL9) are synergistically induced by Nrk siRNA in LPS-treated mouse VSMCs. Moreover, we found that resveratrol significantly impaired LPS- and Nrk siRNA-induced expression of MMP3, CCL8, CCL11, CXCL3 and CXCL5. These results suggested that Nrk may play important roles in regulating pathological progression of atherosclerosis or neointimal- hyperplasia-related vascular diseases.

Keywords: Nik-related kinase; inflammation; intimal hyperplasia; resveratrol; smooth muscle cell.

Conflict of interest statement

CONFLICTS OF INTEREST: The authors have no conflicts of interest to declare.

Figures

Figure 1
Figure 1
Expression of Nrk in VSMCs. (A) Expression of Nrk protein was determined by western blotting analysis in mVSMCs, rVSMCs (A10), hVSMCs, HUVECs, HCAECs, HPAECs, C2C12 and A549 cells. Primary antibodies against mNrk (upper panel) and hNrk (middle panel) were employed for the detection of Nrk. Actin was used as a loading control (lower panel). (B) Expression of mNrk in normal carotid artery of wild-type C57BL/6 mice was examined by immunohistochemical staining with primary antibodies against mNrk, CD31, αSMA, and elastic stain. Bar= 50 μM. (C) Expression and localization of αSMA (green) and mNrk (red) on mouse carotid artery was examined by double staining of immunofluorescence confocal microscopy.
Figure 2
Figure 2
Expression of Nrk was suppressed by PDGF and LPS in mVSMCs. mVSMCs were serum starved (0.5% FBS in DMEM) for 24 h, followed by stimulation with PDGF (10 ng/ml) or LPS (100 ng/ml) for an additional 24 h. Expression of mNrk was determined by (A) western blotting (n=6) and (B) qPCR analysis (n=4). Gene expression of qPCR analysis results were normalized to both control cells as well as to GAPDH. Tubulin was used as a loading control for western blotting analysis. Scale bars: means ± SD. *, p <0.05.
Figure 3
Figure 3
Expression of mNrk, αSMA, and elastic staining in carotid artery of wild-type C57BL/6 mice subjected to guide wire injury for 4 weeks. The expression of mNrk, αSMA, and elastic staining in three set of sections of (A) non-injured and (B) injured carotid arteries at 150-μm intervals was examined by immunohistochemical staining. Bar = 50 μM. (C) Left panel: Quantitation of intima/media (I/M) ratio (left panel, p = 0.00056) and Nrk expression (right panel, p = 1.184 × 10-5) in normal and injured carotid arteries. n = 9 for each group.
Figure 4
Figure 4
Representative immunohistochemical staining of hNrk and calponin 1 in human normal (left panels) and atherosclerotic (right panels) vessels. Bar = 100 μm.
Figure 5
Figure 5
Expression of MMPs and chemokines in LPS- and Nrk-siRNA treated mVSMCs. mVSMCs were serum starved (0.5% FBS in DMEM) for 24 h and then treated with LPS (100 ng/mL) for 24 h. Cells were further transfected with 20 nM of negative control or mNrk siRNA for an additional 48 h. Expression of (A) MMP3 (n=13), MMP8 (n=13) and MMP12 (n=14); (B) CCL6 (n=14), CCL8 (n=14) and CCL11 (n=14); (C) CXCL1 (n=14), CXCL3 (n=11), CXCL5 (n=10) and CXCL9 (n=13) was determined by qPCR. Gene expression results of qPCR analysis were normalized to both control cells as well as GAPDH. Scale bars: means ± SD. *, p < 0.05, **, p < 0.01, ***, p < 0.001.
Figure 6
Figure 6
Effect of resveratrol on LPS- and Nrk siRNA-stimulated MMPs and chemokines. (A) mVSMCs were serum starved (0.5% FBS in DMEM) for 24 h and then treated with LPS (100 ng/mL) and/or resveratrol (50 μM) for 24 h. Cells were further transfected with 20 nM of negative control or mNrk siRNA for an additional 48 h. Expression of (A) MMP3 (n=8), CCL8 (n=8), CCL11 (n=8), CXCL3 (n=6) and CXCL5 (n=6) was determined by qPCR. Gene expression results of qPCR analysis were normalized to both control cells as well as GAPDH. (B) Protein levels of MMP3 (n=7), CCL8 (n=6) and CCL11 (n=7) in cultured conditioned media were determined by ELISA (normalized to total protein concentration). Scale bars: means ± SD. *, p < 0.05, **, p < 0.01, ***, p<0.001.

References

    1. Hansson GK. Inflammation, atherosclerosis, and coronary artery disease. N Engl J Med. 2005; 352:1685–95. 10.1056/NEJMra043430
    1. Libby P, Hansson GK. Inflammation and immunity in diseases of the arterial tree: players and layers. Circ Res. 2015; 116:307–11. 10.1161/CIRCRESAHA.116.301313
    1. Klingenberg R, Hansson GK. Treating inflammation in atherosclerotic cardiovascular disease: emerging therapies. Eur Heart J. 2009; 30:2838–44. 10.1093/eurheartj/ehp477
    1. Libby P, Ridker PM, Hansson GK. Progress and challenges in translating the biology of atherosclerosis. Nature. 2011; 473:317–25. 10.1038/nature10146
    1. Ramji DP, Davies TS. Cytokines in atherosclerosis: key players in all stages of disease and promising therapeutic targets. Cytokine Growth Factor Rev. 2015; 26:673–85. 10.1016/j.cytogfr.2015.04.003
    1. Johnson JL. Metalloproteinases in atherosclerosis. Eur J Pharmacol. 2017; 816:93–106. 10.1016/j.ejphar.2017.09.007
    1. Myasoedova VA, Chistiakov DA, Grechko AV, Orekhov AN. Matrix metalloproteinases in pro-atherosclerotic arterial remodeling. J Mol Cell Cardiol. 2018; 123:159–67. 10.1016/j.yjmcc.2018.08.026
    1. Galis ZS, Sukhova GK, Lark MW, Libby P. Increased expression of matrix metalloproteinases and matrix degrading activity in vulnerable regions of human atherosclerotic plaques. J Clin Invest. 1994; 94:2493–503. 10.1172/JCI117619
    1. Galis ZS, Muszynski M, Sukhova GK, Simon-Morrissey E, Libby P. Enhanced expression of vascular matrix metalloproteinases induced in vitro by cytokines and in regions of human atherosclerotic lesions. Ann N Y Acad Sci. 1995; 748:501–07. 10.1111/j.1749-6632.1994.tb17348.x
    1. Galis ZS, Muszynski M, Sukhova GK, Simon-Morrissey E, Unemori EN, Lark MW, Amento E, Libby P. Cytokine-stimulated human vascular smooth muscle cells synthesize a complement of enzymes required for extracellular matrix digestion. Circ Res. 1994; 75:181–89. 10.1161/01.RES.75.1.181
    1. Kanai-Azuma M, Kanai Y, Okamoto M, Hayashi Y, Yonekawa H, Yazaki K. Nrk: a murine X-linked NIK (Nck-interacting kinase)-related kinase gene expressed in skeletal muscle. Mech Dev. 1999; 89:155–59. 10.1016/S0925-4773(99)00193-8
    1. Nakano K, Yamauchi J, Nakagawa K, Itoh H, Kitamura N. NESK, a member of the germinal center kinase family that activates the c-Jun N-terminal kinase pathway and is expressed during the late stages of embryogenesis. J Biol Chem. 2000; 275:20533–39. 10.1074/jbc.M001009200
    1. Nakano K, Kanai-Azuma M, Kanai Y, Moriyama K, Yazaki K, Hayashi Y, Kitamura N. Cofilin phosphorylation and actin polymerization by NRK/NESK, a member of the germinal center kinase family. Exp Cell Res. 2003; 287:219–27. 10.1016/S0014-4827(03)00136-8
    1. Denda K, Nakao-Wakabayashi K, Okamoto N, Kitamura N, Ryu JY, Tagawa Y, Ichisaka T, Yamanaka S, Komada M. Nrk, an X-linked protein kinase in the germinal center kinase family, is required for placental development and fetoplacental induction of labor. J Biol Chem. 2011; 286:28802–10. 10.1074/jbc.M111.258160
    1. Morioka Y, Nam JM, Ohashi T. Nik-related kinase regulates trophoblast proliferation and placental development by modulating AKT phosphorylation. PLoS One. 2017; 12:e0171503. 10.1371/journal.pone.0171503
    1. Ernst C, Sequeira A, Klempan T, Ernst N, Ffrench-Mullen J, Turecki G. Confirmation of region-specific patterns of gene expression in the human brain. Neurogenetics. 2007; 8:219–24. 10.1007/s10048-007-0084-2
    1. Yanagawa T, Denda K, Inatani T, Fukushima T, Tanaka T, Kumaki N, Inagaki Y, Komada M. Deficiency of X-Linked Protein Kinase Nrk during Pregnancy Triggers Breast Tumor in Mice. Am J Pathol. 2016; 186:2751–60. 10.1016/j.ajpath.2016.06.005
    1. Dong P, Yu B, Pan L, Tian X, Liu F. Identification of Key Genes and Pathways in Triple-Negative Breast Cancer by Integrated Bioinformatics Analysis. Biomed Res Int. 2018; 2018:2760918. 10.1155/2018/2760918
    1. Zhao Y, Biswas SK, McNulty PH, Kozak M, Jun JY, Segar L. PDGF-induced vascular smooth muscle cell proliferation is associated with dysregulation of insulin receptor substrates. Am J Physiol Cell Physiol. 2011; 300:C1375–85. 10.1152/ajpcell.00670.2008
    1. Li L, Blumenthal DK, Terry CM, He Y, Carlson ML, Cheung AK. PDGF-induced proliferation in human arterial and venous smooth muscle cells: molecular basis for differential effects of PDGF isoforms. J Cell Biochem. 2011; 112:289–98. 10.1002/jcb.22924
    1. Jiang D, Yang Y, Li D. Lipopolysaccharide induced vascular smooth muscle cells proliferation: A new potential therapeutic target for proliferative vascular diseases. Cell Prolif. 2017; 50:50. 10.1111/cpr.12332
    1. Engelmann MG, Redl CV, Nikol S. Recurrent perivascular inflammation induced by lipopolysaccharide (endotoxin) results in the formation of atheromatous lesions in vivo. Lab Invest. 2004; 84:425–32. 10.1038/labinvest.3700065
    1. Li H, Xia N, Hasselwander S, Daiber A. Resveratrol and Vascular Function. Int J Mol Sci. 2019; 20:20. 10.3390/ijms20092155
    1. Pervaiz S. Resveratrol: from grapevines to mammalian biology. FASEB J. 2003; 17:1975–85. 10.1096/fj.03-0168rev
    1. Ardigo D, Assimes TL, Fortmann SP, Go AS, Hlatky M, Hytopoulos E, Iribarren C, Tsao PS, Tabibiazar R, Quertermous T, and ADVANCE Investigators. Circulating chemokines accurately identify individuals with clinically significant atherosclerotic heart disease. Physiol Genomics. 2007; 31:402–09. 10.1152/physiolgenomics.00104.2007
    1. Huma ZE, Sanchez J, Lim HD, Bridgford JL, Huang C, Parker BJ, Pazhamalil JG, Porebski BT, Pfleger KD, Lane JR, Canals M, Stone MJ. Key determinants of selective binding and activation by the monocyte chemoattractant proteins at the chemokine receptor CCR2. Sci Signal. 2017; 10:10. 10.1126/scisignal.aai8529
    1. Ma S, Tian XY, Zhang Y, Mu C, Shen H, Bismuth J, Pownall HJ, Huang Y, Wong WT. E-selectin-targeting delivery of microRNAs by microparticles ameliorates endothelial inflammation and atherosclerosis. Sci Rep. 2016; 6:22910. 10.1038/srep22910
    1. Haley KJ, Lilly CM, Yang JH, Feng Y, Kennedy SP, Turi TG, Thompson JF, Sukhova GH, Libby P, Lee RT. Overexpression of eotaxin and the CCR3 receptor in human atherosclerosis: using genomic technology to identify a potential novel pathway of vascular inflammation. Circulation. 2000; 102:2185–89. 10.1161/01.CIR.102.18.2185
    1. Wang L, Shah PK, Wang W, Song L, Yang M, Sharifi BG. Tenascin-C deficiency in apo E-/- mouse increases eotaxin levels: implications for atherosclerosis. Atherosclerosis. 2013; 227:267–74. 10.1016/j.atherosclerosis.2013.01.039
    1. Ye S, Watts GF, Mandalia S, Humphries SE, Henney AM. Preliminary report: genetic variation in the human stromelysin promoter is associated with progression of coronary atherosclerosis. Br Heart J. 1995; 73:209–15. 10.1136/hrt.73.3.209
    1. Ye S, Eriksson P, Hamsten A, Kurkinen M, Humphries SE, Henney AM. Progression of coronary atherosclerosis is associated with a common genetic variant of the human stromelysin-1 promoter which results in reduced gene expression. J Biol Chem. 1996; 271:13055–60. 10.1074/jbc.271.22.13055
    1. Terashima M, Akita H, Kanazawa K, Inoue N, Yamada S, Ito K, Matsuda Y, Takai E, Iwai C, Kurogane H, Yoshida Y, Yokoyama M. Stromelysin promoter 5A/6A polymorphism is associated with acute myocardial infarction. Circulation. 1999; 99:2717–19. 10.1161/01.CIR.99.21.2717
    1. Inoue T, Kato T, Takayanagi K, Uchida T, Yaguchi I, Kamishirado H, Morooka S, Yoshimoto N. Circulating matrix metalloproteinase-1 and -3 in patients with an acute coronary syndrome. Am J Cardiol. 2003; 92:1461–64. 10.1016/j.amjcard.2003.08.061
    1. Samnegård A, Silveira A, Lundman P, Boquist S, Odeberg J, Hulthe J, McPheat W, Tornvall P, Bergstrand L, Ericsson CG, Hamsten A, Eriksson P. Serum matrix metalloproteinase-3 concentration is influenced by MMP-3 -1612 5A/6A promoter genotype and associated with myocardial infarction. J Intern Med. 2005; 258:411–19. 10.1111/j.1365-2796.2005.01561.x
    1. Wu TC, Leu HB, Lin WT, Lin CP, Lin SJ, Chen JW. Plasma matrix metalloproteinase-3 level is an independent prognostic factor in stable coronary artery disease. Eur J Clin Invest. 2005; 35:537–45. 10.1111/j.1365-2362.2005.01548.x
    1. Kaplan RC, Smith NL, Zucker S, Heckbert SR, Rice K, Psaty BM. Matrix metalloproteinase-3 (MMP3) and MMP9 genes and risk of myocardial infarction, ischemic stroke, and hemorrhagic stroke. Atherosclerosis. 2008; 201:130–37. 10.1016/j.atherosclerosis.2008.01.003
    1. Kelly D, Khan S, Cockerill G, Ng LL, Thompson M, Samani NJ, Squire IB. Circulating stromelysin-1 (MMP-3): a novel predictor of LV dysfunction, remodelling and all-cause mortality after acute myocardial infarction. Eur J Heart Fail. 2008; 10:133–39. 10.1016/j.ejheart.2007.12.009
    1. Halim SA, Neely ML, Pieper KS, Shah SH, Kraus WE, Hauser ER, Califf RM, Granger CB, Newby LK. Simultaneous consideration of multiple candidate protein biomarkers for long-term risk for cardiovascular events. Circ Cardiovasc Genet. 2015; 8:168–77. 10.1161/CIRCGENETICS.113.000490
    1. Cavusoglu E, Marmur JD, Kassotis JT, Yanamadala S, Chopra V, Eng C. Usefulness of Plasma Matrix Metalloproteinase-3 Levels to Predict Myocardial Infarction in Men With and Without Acute Coronary Syndrome. Am J Cardiol. 2016; 117:881–86. 10.1016/j.amjcard.2015.12.022
    1. Raghuraman G, Hsiung J, Zuniga MC, Baughman BD, Hitchner E, Guzman RJ, Zhou W. Eotaxin Augments Calcification in Vascular Smooth Muscle Cells. J Cell Biochem. 2017; 118:647–54. 10.1002/jcb.25752
    1. Economou E, Tousoulis D, Katinioti A, Stefanadis C, Trikas A, Pitsavos C, Tentolouris C, Toutouza MG, Toutouzas P. Chemokines in patients with ischaemic heart disease and the effect of coronary angioplasty. Int J Cardiol. 2001; 80:55–60. 10.1016/S0167-5273(01)00454-5
    1. Kodali RB, Kim WJ, Galaria II, Miller C, Schecter AD, Lira SA, Taubman MB. CCL11 (Eotaxin) induces CCR3-dependent smooth muscle cell migration. Arterioscler Thromb Vasc Biol. 2004; 24:1211–16. 10.1161/01.ATV.0000131654.90788.f5
    1. Soleas GJ, Diamandis EP, Goldberg DM. Resveratrol: a molecule whose time has come? And gone? Clin Biochem. 1997; 30:91–113. 10.1016/S0009-9120(96)00155-5
    1. Almajdoob S, Hossain E, Anand-Srivastava MB. Resveratrol attenuates hyperproliferation of vascular smooth muscle cells from spontaneously hypertensive rats: role of ROS and ROS-mediated cell signaling. Vascul Pharmacol. 2018; 101:48–56. 10.1016/j.vph.2017.12.064
    1. Seo Y, Park J, Choi W, Ju Son D, Sung Kim Y, Kim MK, Yoon BE, Pyee J, Tae Hong J, Go YM, Park H. Antiatherogenic Effect of Resveratrol Attributed to Decreased Expression of ICAM-1 (Intercellular Adhesion Molecule-1). Arterioscler Thromb Vasc Biol. 2019; 39:675–84. 10.1161/ATVBAHA.118.312201
    1. Zhu Y, Takayama T, Wang B, Kent A, Zhang M, Binder BY, Urabe G, Shi Y, DiRenzo D, Goel SA, Zhou Y, Little C, Roenneburg DA, et al.. Restenosis Inhibition and Re-differentiation of TGFβ/Smad3-activated Smooth Muscle Cells by Resveratrol. Sci Rep. 2017; 7:41916. 10.1038/srep41916
    1. Ray JL, Leach R, Herbert JM, Benson M. Isolation of vascular smooth muscle cells from a single murine aorta. Methods Cell Sci. 2001; 23:185–88. 10.1023/A:1016357510143
    1. Wu ML, Chen CH, Lin YT, Jheng YJ, Ho YC, Yang LT, Chen L, Layne MD, Yet SF. Divergent signaling pathways cooperatively regulate TGFβ induction of cysteine-rich protein 2 in vascular smooth muscle cells. Cell Commun Signal. 2014; 12:22. 10.1186/1478-811X-12-22
    1. Lin YT, Liang SM, Wu YJ, Wu YJ, Lu YJ, Jan YJ, Ko BS, Chuang YJ, Shyue SK, Kuo CC, Liou JY. Cordycepin Suppresses Endothelial Cell Proliferation, Migration, Angiogenesis, and Tumor Growth by Regulating Focal Adhesion Kinase and p53. Cancers (Basel). 2019; 11:11. 10.3390/cancers11020168
    1. Tzeng BH, Chen YH, Huang CH, Lin SS, Lee KR, Chen CC. The Ca(v)3.1 T-type calcium channel is required for neointimal formation in response to vascular injury in mice. Cardiovasc Res. 2012; 96:533–42. 10.1093/cvr/cvs257
    1. Holt AW, Tulis DA. Experimental Rat and Mouse Carotid Artery Surgery: Injury & Remodeling Studies. ISRN Minim Invasive Surg. 2013; 2013:167407. 10.1155/2013/167407
    1. Manka DR, Gilson W, Sarembock I, Ley K, Berr SS. Noninvasive in vivo magnetic resonance imaging of injury-induced neointima formation in the carotid artery of the apolipoprotein-E null mouse. J Magn Reson Imaging. 2000; 12:790–94. 10.1002/1522-2586(200011)12:5<790::AID-JMRI19>;2-6
    1. Liu TA, Jan YJ, Ko BS, Liang SM, Chen SC, Wang J, Hsu C, Wu YM, Liou JY. 14-3-3ε overexpression contributes to epithelial-mesenchymal transition of hepatocellular carcinoma. PLoS One. 2013; 8:e57968. 10.1371/journal.pone.0057968
    1. Liu TA, Jan YJ, Ko BS, Chen SC, Liang SM, Hung YL, Hsu C, Shen TL, Lee YM, Chen PF, Wang J, Shyue SK, Liou JY. Increased expression of 14-3-3β promotes tumor progression and predicts extrahepatic metastasis and worse survival in hepatocellular carcinoma. Am J Pathol. 2011; 179:2698–708. 10.1016/j.ajpath.2011.08.010
    1. Ko BS, Chang TC, Hsu C, Chen YC, Shen TL, Chen SC, Wang J, Wu KK, Jan YJ, Liou JY. Overexpression of 14-3-3ε predicts tumour metastasis and poor survival in hepatocellular carcinoma. Histopathology. 2011; 58:705–11. 10.1111/j.1365-2559.2011.03789.x
    1. Barnes DM, Harris WH, Smith P, Millis RR, Rubens RD. Immunohistochemical determination of oestrogen receptor: comparison of different methods of assessment of staining and correlation with clinical outcome of breast cancer patients. Br J Cancer. 1996; 74:1445–51. 10.1038/bjc.1996.563
    1. Chang GC, Liu KJ, Hsieh CL, Hu TS, Charoenfuprasert S, Liu HK, Luh KT, Hsu LH, Wu CW, Ting CC, Chen CY, Chen KC, Yang TY, et al.. Identification of alpha-enolase as an autoantigen in lung cancer: its overexpression is associated with clinical outcomes. Clin Cancer Res. 2006; 12:5746–54. 10.1158/1078-0432.CCR-06-0324
    1. Jan YJ, Ko BS, Hsu C, Chang TC, Chen SC, Wang J, Liou JY. Overexpressed focal adhesion kinase predicts a higher incidence of extrahepatic metastasis and worse survival in hepatocellular carcinoma. Hum Pathol. 2009; 40:1384–90. 10.1016/j.humpath.2009.03.006

Source: PubMed

3
订阅