Review Article

LncRNA介导的内皮细胞调控:冠状动脉粥样硬化发病机制的新进展

卷 31, 期 10, 2024

发表于: 22 March, 2023

页: [1251 - 1264] 页: 14

弟呕挨: 10.2174/0929867330666230213100732

价格: $65

Open Access Journals Promotions 2
摘要

冠状动脉粥样硬化性疾病(CAD)是一种常见的心血管疾病,也是导致死亡的重要原因。此外,内皮细胞(EC)损伤是CAD的早期病理生理特征,并且长链非编码RNA(lncRNA)可以调节基因表达。最近的研究表明lncRNA参与CAD的发病机制,尤其是通过调节内皮细胞。在这篇综述中,我们总结了lncRNA调节的内皮细胞在CAD发病机制中的新进展,包括内皮细胞增殖、迁移、粘附、血管生成、炎症、细胞凋亡、自噬和pyroptosis。因此,由于lncRNA调节CAD中的内皮细胞,lncRNA将为CAD的诊断和药物治疗提供理想和新的靶点。

关键词: 长链非编码RNA,内皮细胞,冠状动脉粥样硬化性心脏病,诊断,药物治疗,平滑肌细胞。

[1]
Sun, S.; Cao, W.; Ge, Y.; Ran, J.; Sun, F.; Zeng, Q.; Guo, M.; Huang, J.; Lee, R.S.Y.; Tian, L.; Wellenius, G.A. Outdoor light at night and risk of coronary heart disease among older adults: a prospective cohort study. Eur. Heart J., 2021, 42(8), 822-830.
[http://dx.doi.org/10.1093/eurheartj/ehaa846] [PMID: 33205210]
[2]
Peters, S.A.E.; Colantonio, L.D.; Dai, Y.; Zhao, H.; Bittner, V.; Farkouh, M.E.; Dluzniewski, P.; Poudel, B.; Muntner, P.; Woodward, M. Trends in recurrent coronary heart disease after myocardial infarction among us women and men between 2008 and 2017. Circulation, 2021, 143(7), 650-660.
[http://dx.doi.org/10.1161/CIRCULATIONAHA.120.047065] [PMID: 32951451]
[3]
Zeitouni, M.; Nanna, M.G.; Sun, J.L.; Chiswell, K.; Peterson, E.D.; Navar, A.M. Performance of guideline recommendations for prevention of myocardial infarction in young adults. J. Am. Coll. Cardiol., 2020, 76(6), 653-664.
[http://dx.doi.org/10.1016/j.jacc.2020.06.030] [PMID: 32762899]
[4]
Deal, B.J.; Huffman, M.D.; Binns, H.; Stone, N.J. Perspective: childhood obesity requires new strategies for prevention. Adv. Nutr., 2020, 11(5), 1071-1078.
[http://dx.doi.org/10.1093/advances/nmaa040] [PMID: 32361757]
[5]
Li, J.J.; Li, S.; Zhu, C.G.; Wu, N.Q.; Zhang, Y.; Guo, Y.L.; Gao, Y.; Li, X.L.; Qing, P.; Cui, C.J.; Xu, R.X.; Jiang, Z.W.; Sun, J.; Liu, G.; Dong, Q. Familial hypercholesterolemia phenotype in chinese patients undergoing coronary angiography. Arterioscler. Thromb. Vasc. Biol., 2017, 37(3), 570-579.
[http://dx.doi.org/10.1161/ATVBAHA.116.308456] [PMID: 27932355]
[6]
Mulders, T.A.; Sivapalaratnam, S.; Stroes, E.S.G.; Kastelein, J.J.P.; Guerci, A.D.; Pinto-Sietsma, S.J. Asymptomatic individuals with a positive family history for premature coronary artery disease and elevated coronary calcium scores benefit from statin treatment: a post hoc analysis from the St. Francis Heart Study. JACC Cardiovasc. Imaging, 2012, 5(3), 252-260.
[http://dx.doi.org/10.1016/j.jcmg.2011.11.014] [PMID: 22421169]
[7]
Bai, Y.; Zhang, Q.; Su, Y.; Pu, Z.; Li, K. Modulation of the proliferation/apoptosis balance of vascular smooth muscle cells in atherosclerosis by lncRNA-MEG3 via regulation of miR-26a/Smad1 Axis. Int. Heart J., 2019, 60(2), 444-450.
[http://dx.doi.org/10.1536/ihj.18-195] [PMID: 30745534]
[8]
Taheri, M.; Shoorei, H.; Dinger, M.E.; Ghafouri-Fard, S. Perspectives on the role of non-coding RNAs in the regulation of expression and function of the estrogen receptor. Cancers (Basel), 2020, 12(8), 2162.
[http://dx.doi.org/10.3390/cancers12082162] [PMID: 32759784]
[9]
Jackson, C.L.; Keeton, J.Z.; Eason, S.J.; Ahmad, Z.A.; Ayers, C.R.; Gore, M.O.; McGuire, D.K.; Sayers, M.H.; Khera, A. Identifying familial hypercholesterolemia using a blood donor screening program with more than 1 million volunteer donors. JAMA Cardiol., 2019, 4(7), 685-689.
[http://dx.doi.org/10.1001/jamacardio.2019.1518] [PMID: 31116347]
[10]
Sun, H.; Wang, J.; Que, J.; Peng, Y.; Yu, Y.; Wang, L.; Ye, H.; Huang, K.; Xue, Y.; Zhou, Y.; Ji, K. RNA sequencing revealing the role of AMP-activated protein kinase signaling in mice myocardial ischemia reperfusion injury. Gene, 2019, 703, 91-101.
[http://dx.doi.org/10.1016/j.gene.2019.04.010] [PMID: 30974198]
[11]
Forini, F.; Nicolini, G.; Kusmic, C.; D’Aurizio, R.; Mercatanti, A.; Iervasi, G.; Pitto, L. T3 critically affects the Mhrt/Brg1 axis to regulate the cardiac MHC switch: role of an epigenetic cross-talk. Cells, 2020, 9(10), 2155.
[http://dx.doi.org/10.3390/cells9102155] [PMID: 32987653]
[12]
Xu, S.; Xu, Y.; Liu, P.; Zhang, S.; Liu, H.; Slavin, S.; Kumar, S.; Koroleva, M.; Luo, J.; Wu, X.; Rahman, A.; Pelisek, J.; Jo, H.; Si, S.; Miller, C.L.; Jin, Z.G. The novel coronary artery disease risk gene JCAD/KIAA1462 promotes endothelial dysfunction and atherosclerosis. Eur. Heart J., 2019, 40(29), 2398-2408.
[http://dx.doi.org/10.1093/eurheartj/ehz303] [PMID: 31539914]
[13]
Park, S.H.; Belcastro, E.; Hasan, H.; Matsushita, K.; Marchandot, B.; Abbas, M.; Toti, F.; Auger, C.; Jesel, L.; Ohlmann, P.; Morel, O.; Schini-Kerth, V.B. Angiotensin II-induced upregulation of SGLT1 and 2 contributes to human microparticle-stimulated endothelial senescence and dysfunction: protective effect of gliflozins. Cardiovasc. Diabetol., 2021, 20(1), 65.
[http://dx.doi.org/10.1186/s12933-021-01252-3] [PMID: 33726768]
[14]
Boulberdaa, M.; Scott, E.; Ballantyne, M.; Garcia, R.; Descamps, B.; Angelini, G.D.; Brittan, M.; Hunter, A.; McBride, M.; McClure, J.; Miano, J.M.; Emanueli, C.; Mills, N.L.; Mountford, J.C.; Baker, A.H. A role for the long noncoding RNA sencr in commitment and function of endothelial cells. Mol. Ther., 2016, 24(5), 978-990.
[http://dx.doi.org/10.1038/mt.2016.41] [PMID: 26898221]
[15]
Thomas, A.A.; Biswas, S.; Feng, B.; Chen, S.; Gonder, J.; Chakrabarti, S. lncRNA H19 prevents endothelial–mesenchymal transition in diabetic retinopathy. Diabetologia, 2019, 62(3), 517-530.
[http://dx.doi.org/10.1007/s00125-018-4797-6] [PMID: 30612136]
[16]
Dieci, G.; Fiorino, G.; Castelnuovo, M.; Teichmann, M.; Pagano, A. The expanding RNA polymerase III transcriptome. Trends Genet., 2007, 23(12), 614-622.
[http://dx.doi.org/10.1016/j.tig.2007.09.001] [PMID: 17977614]
[17]
Holoch, D.; Moazed, D. RNA-mediated epigenetic regulation of gene expression. Nat. Rev. Genet., 2015, 16(2), 71-84.
[http://dx.doi.org/10.1038/nrg3863] [PMID: 25554358]
[18]
St Laurent, G.; Wahlestedt, C.; Kapranov, P. The Landscape of long noncoding RNA classification. Trends Genet., 2015, 31(5), 239-251.
[http://dx.doi.org/10.1016/j.tig.2015.03.007] [PMID: 25869999]
[19]
Jiang, S.; Cheng, S.J.; Ren, L.C.; Wang, Q.; Kang, Y.J.; Ding, Y.; Hou, M.; Yang, X.X.; Lin, Y.; Liang, N.; Gao, G. An expanded landscape of human long noncoding RNA. Nucleic Acids Res., 2019, 47(15), 7842-7856.
[http://dx.doi.org/10.1093/nar/gkz621] [PMID: 31350901]
[20]
Wang, C.; Wang, L.; Ding, Y.; Lu, X.; Zhang, G.; Yang, J.; Zheng, H.; Wang, H.; Jiang, Y.; Xu, L. LncRNA structural characteristics in epigenetic regulation. Int. J. Mol. Sci., 2017, 18(12), 2659.
[http://dx.doi.org/10.3390/ijms18122659] [PMID: 29292750]
[21]
Liu, J.; Li, Y.; Tong, J.; Gao, J.; Guo, Q.; Zhang, L.; Wang, B.; Zhao, H.; Wang, H.; Jiang, E.; Kurita, R.; Nakamura, Y.; Tanabe, O.; Engel, J.D.; Bresnick, E.H.; Zhou, J.; Shi, L. Long non-coding RNA-dependent mechanism to regulate heme biosynthesis and erythrocyte development. Nat. Commun., 2018, 9(1), 4386.
[http://dx.doi.org/10.1038/s41467-018-06883-x] [PMID: 30349036]
[22]
Sun, Q.; Hao, Q.; Prasanth, K.V. Nuclear long noncoding RNAs: Key regulators of gene expression. Trends Genet., 2018, 34(2), 142-157.
[http://dx.doi.org/10.1016/j.tig.2017.11.005] [PMID: 29249332]
[23]
Yubero-Serrano, E.M.; Fernandez-Gandara, C.; Garcia-Rios, A.; Rangel-Zuñiga, O.A.; Gutierrez-Mariscal, F.M.; Torres-Peña, J.D.; Marin, C.; Lopez-Moreno, J.; Castaño, J.P.; Delgado-Lista, J.; Ordovas, J.M.; Perez-Martinez, P.; Lopez-Miranda, J. Mediterranean diet and endothelial function in patients with coronary heart disease: An analysis of the CORDIOPREV randomized controlled trial. PLoS Med., 2020, 17(9), e1003282.
[http://dx.doi.org/10.1371/journal.pmed.1003282] [PMID: 32903262]
[24]
Latorre, E.; Pilling, L.C.; Lee, B.P.; Bandinelli, S.; Melzer, D.; Ferrucci, L.; Harries, L.W. The VEGFA156b isoform is dysregulated in senescent endothelial cells and may be associated with prevalent and incident coronary heart disease. Clin. Sci. (Lond.), 2018, 132(3), 313-325.
[http://dx.doi.org/10.1042/CS20171556] [PMID: 29330351]
[25]
Zhou, H.; Simion, V.; Pierce, J.B.; Haemmig, S.; Chen, A.F.; Feinberg, M.W. LncRNA-MAP3K4 regulates vascular inflammation through the p38 MAPK signaling pathway and cis -modulation of MAP3K4. FASEB J., 2021, 35(1), e21133.
[http://dx.doi.org/10.1096/fj.202001654RR] [PMID: 33184917]
[26]
Radhakrishnan, R.; Kowluru, R.A.; Long Noncoding, R.N.A. Long noncoding RNA MALAT1 and regulation of the antioxidant defense system in diabetic retinopathy. Diabetes, 2021, 70(1), 227-239.
[http://dx.doi.org/10.2337/db20-0375] [PMID: 33051272]
[27]
Liao, B.; Chen, R.; Lin, F.; Mai, A.; Chen, J.; Li, H.; Xu, Z.; Dong, S. Long noncoding RNA HOTTIP promotes endothelial cell proliferation and migration via activation of the Wnt/β-catenin pathway. J. Cell. Biochem., 2018, 119(3), 2797-2805.
[http://dx.doi.org/10.1002/jcb.26448] [PMID: 29058802]
[28]
Wu, Z.; He, Y.; Li, D.; Fang, X.; Shang, T.; Zhang, H.; Zheng, X. Long noncoding RNA MEG3 suppressed endothelial cell proliferation and migration through regulating miR-21. Am. J. Transl. Res., 2017, 9(7), 3326-3335.
[PMID: 28804550]
[29]
Li, P.; Li, Y.; Chen, L.; Ma, X.; Yan, X.; Yan, M.; Qian, B.; Wang, F.; Xu, J.; Yin, J.; Xu, G.; Sun, K. Long noncoding RNA uc003pxg.1 regulates endothelial cell proliferation and migration via miR-25-5p in coronary artery disease. Int. J. Mol. Med., 2021, 48(2), 160.
[http://dx.doi.org/10.3892/ijmm.2021.4993] [PMID: 34212983]
[30]
Shang, J.; Li, Q.Z.; Zhang, J.Y.; Yuan, H.J. FAL1 regulates endothelial cell proliferation in diabetic arteriosclerosis through PTEN/AKT pathway. Eur. Rev. Med. Pharmacol. Sci., 2018, 22(19), 6492-6499.
[PMID: 30338819]
[31]
Wang, X.; Zhao, Z.; Zhang, W.; Wang, Y. Long noncoding RNA LINC00968 promotes endothelial cell proliferation and migration via regulating miR-9-3p expression. J. Cell. Biochem., 2018.
[PMID: 30485507]
[32]
Du, H.; Yang, L.; Zhang, H.; Zhang, X.; Shao, H. LncRNA TUG1 silencing enhances proliferation and migration of ox-LDL-treated human umbilical vein endothelial cells and promotes atherosclerotic vascular injury repairing via the Runx2/ANPEP axis. Int. J. Cardiol., 2021, 338, 204-214.
[http://dx.doi.org/10.1016/j.ijcard.2021.05.014] [PMID: 33971184]
[33]
Zhang, S.; Xie, B.; Wang, L.; Yang, H.; Zhang, H.; Chen, Y.; Wang, F.; Liu, C.; He, H. Macrophage-mediated vascular permeability via VLA4/VCAM1 pathway dictates ascites development in ovarian cancer. J. Clin. Invest., 2021, 131(3), e140315.
[http://dx.doi.org/10.1172/JCI140315] [PMID: 33295887]
[34]
Simion, V.; Zhou, H.; Pierce, J.B.; Yang, D.; Haemmig, S.; Tesmenitsky, Y.; Sukhova, G.; Stone, P.H.; Libby, P.; Feinberg, M.W. LncRNA VINAS regulates atherosclerosis by modulating NF-κB and MAPK signaling. JCI Insight, 2020, 5(21), e140627.
[http://dx.doi.org/10.1172/jci.insight.140627] [PMID: 33021969]
[35]
Li, Y.; Cho, H.; Wang, F.; Canela-Xandri, O.; Luo, C.; Rawlik, K.; Archacki, S.; Xu, C.; Tenesa, A.; Chen, Q.; Wang, Q.K. Statistical and functional studies identify epistasis of cardiovascular risk genomic variants from genome-wide association studies. J. Am. Heart Assoc., 2020, 9(7), e014146.
[http://dx.doi.org/10.1161/JAHA.119.014146] [PMID: 32237974]
[36]
Arnold, L.; Weberbauer, M.; Herkel, M.; Fink, K.; Busch, H.J.; Diehl, P.; Grundmann, S.; Bode, C.; Elsässer, A.; Moser, M.; Helbing, T. Endothelial BMP4 promotes leukocyte rolling and adhesion and is elevated in patients after survived out-of-hospital cardiac arrest. Inflammation, 2020, 43(6), 2379-2391.
[http://dx.doi.org/10.1007/s10753-020-01307-9] [PMID: 32803667]
[37]
Cho, H.; Shen, G.Q.; Wang, X.; Wang, F.; Archacki, S.; Li, Y.; Yu, G.; Chakrabarti, S.; Chen, Q.; Wang, Q.K. Long noncoding RNA ANRIL regulates endothelial cell activities associated with coronary artery disease by up-regulating CLIP1, EZR, and LYVE1 genes. J. Biol. Chem., 2019, 294(11), 3881-3898.
[http://dx.doi.org/10.1074/jbc.RA118.005050] [PMID: 30655286]
[38]
Cho, H.; Li, Y.; Archacki, S.; Wang, F.; Yu, G.; Chakrabarti, S.; Guo, Y.; Chen, Q.; Wang, Q.K. Splice variants of lncRNA RNA ANRIL exert opposing effects on endothelial cell activities associated with coronary artery disease. RNA Biol., 2020, 17(10), 1391-1401.
[http://dx.doi.org/10.1080/15476286.2020.1771519] [PMID: 32602777]
[39]
Leisegang, M.S.; Bibli, S.I.; Günther, S.; Pflüger-Müller, B.; Oo, J.A.; Höper, C.; Seredinski, S.; Yekelchyk, M.; Schmitz-Rixen, T.; Schürmann, C.; Hu, J.; Looso, M.; Sigala, F.; Boon, R.A.; Fleming, I.; Brandes, R.P. Pleiotropic effects of laminar flow and statins depend on the Krüppel-like factor-induced lncRNA MANTIS. Eur. Heart J., 2019, 40(30), 2523-2533.
[http://dx.doi.org/10.1093/eurheartj/ehz393] [PMID: 31222221]
[40]
Lyu, Q.; Xu, S.; Lyu, Y.; Choi, M.; Christie, C.K.; Slivano, O.J.; Rahman, A.; Jin, Z.G.; Long, X.; Xu, Y.; Miano, J.M. SENCR stabilizes vascular endothelial cell adherens junctions through interaction with CKAP4. Proc. Natl. Acad. Sci. USA, 2019, 116(2), 546-555.
[http://dx.doi.org/10.1073/pnas.1810729116] [PMID: 30584103]
[41]
Petre, A.; Balta, C.; Herman, H.; Gharbia, S.; Codreanu, A.; Onita-Mladin, B.; Anghel-Zurbau, N.; Hermenean, A.G.; Ignat, S.R.; Dinescu, S.; Urzica, I.; Drafta, S.; Oancea, L.; Hermenean, A. A novel experimental approach to evaluate guided bone regeneration (GBR) in the rat femur using a 3D-printed CAD/CAM zirconia space-maintaining barrier. J. Adv. Res., 2021, 28, 221-229.
[http://dx.doi.org/10.1016/j.jare.2020.07.012] [PMID: 33364058]
[42]
Zhu, Q.M.; MacDonald, B.T.; Mizoguchi, T.; Chaffin, M.; Leed, A.; Arduini, A.; Malolepsza, E.; Lage, K.; Kaushik, V.K.; Kathiresan, S.; Ellinor, P.T. Endothelial ARHGEF26 is an angiogenic factor promoting VEGF signaling. Cardiovasc. Res., 2021.
[43]
Tsai, W.C.; Chiang, W.H.; Wu, C.H.; Li, Y.C.; Campbell, M.; Huang, P.H.; Lin, M.W.; Lin, C.H.; Cheng, S.M.; Chang, P.C.; Cheng, C.C. miR-548aq-3p is a novel target of Far infrared radiation which predicts coronary artery disease endothelial colony forming cell responsiveness. Sci. Rep., 2020, 10(1), 6805.
[http://dx.doi.org/10.1038/s41598-020-63311-1] [PMID: 32322002]
[44]
Ouyang, S.; Li, Y.; Wu, X.; Wang, Y.; Liu, F.; Zhang, J.; Qiu, Y.; Zhou, Z.; Wang, Z.; Xia, W.; Lin, X. GPR4 signaling is essential for the promotion of acid-mediated angiogenic capacity of endothelial progenitor cells by activating STAT3/VEGFA pathway in patients with coronary artery disease. Stem Cell Res. Ther., 2021, 12(1), 149.
[http://dx.doi.org/10.1186/s13287-021-02221-z] [PMID: 33632325]
[45]
Mushimiyimana, I.; Tomas Bosch, V.; Niskanen, H.; Downes, N.L.; Moreau, P.R.; Hartigan, K.; Ylä-Herttuala, S.; Laham-Karam, N.; Kaikkonen, M.U. Genomic landscapes of noncoding RNAs regulating VEGFA and VEGFC expression in endothelial cells. Mol. Cell. Biol., 2021, 41(7), e00594-20.
[http://dx.doi.org/10.1128/MCB.00594-20] [PMID: 33875575]
[46]
Zhang, M.; Wang, X.; Yao, J.; Qiu, Z. Long non-coding RNA NEAT1 inhibits oxidative stress-induced vascular endothelial cell injury by activating the miR-181d-5p/CDKN3 axis. Artif. Cells Nanomed. Biotechnol., 2019, 47(1), 3129-3137.
[http://dx.doi.org/10.1080/21691401.2019.1646264] [PMID: 31352814]
[47]
Huang, P.; Wang, L.; Li, Q.; Tian, X.; Xu, J.; Xu, J.; Xiong, Y.; Chen, G.; Qian, H.; Jin, C.; Yu, Y.; Cheng, K.; Qian, L.; Yang, Y. Atorvastatin enhances the therapeutic efficacy of mesenchymal stem cells-derived exosomes in acute myocardial infarction via up-regulating long non-coding RNA H19. Cardiovasc. Res., 2020, 116(2), 353-367.
[http://dx.doi.org/10.1093/cvr/cvz139] [PMID: 31119268]
[48]
Kong, C.; Lyu, D.; He, C.; Li, R.; Lu, Q. Dioscin elevates lncRNA MANTIS in therapeutic angiogenesis for heart diseases. Aging Cell, 2021, 20(7), e13392.
[http://dx.doi.org/10.1111/acel.13392] [PMID: 34081836]
[49]
Hosen, M.R.; Li, Q.; Liu, Y.; Zietzer, A.; Maus, K.; Goody, P.; Uchida, S.; Latz, E.; Werner, N.; Nickenig, G.; Jansen, F. CAD increases the long noncoding RNA PUNISHER in small extracellular vesicles and regulates endothelial cell function via vesicular shuttling. Mol. Ther. Nucleic Acids, 2021, 25, 388-405.
[http://dx.doi.org/10.1016/j.omtn.2021.05.023] [PMID: 34484864]
[50]
Kai, H.; Wu, Q.; Yin, R.; Tang, X.; Shi, H.; Wang, T.; Zhang, M.; Pan, C. LncRNA NORAD Promotes vascular endothelial cell injury and atherosclerosis through suppressing VEGF gene transcription via enhancing H3K9 deacetylation by recruiting HDAC6. Front. Cell Dev. Biol., 2021, 9, 701628.
[http://dx.doi.org/10.3389/fcell.2021.701628] [PMID: 34307380]
[51]
Noerman, S.; Kokla, M.; Koistinen, V.M.; Lehtonen, M.; Tuomainen, T.P.; Brunius, C.; Virtanen, J.K.; Hanhineva, K. Associations of the serum metabolite profile with a healthy Nordic diet and risk of coronary artery disease. Clin. Nutr., 2021, 40(5), 3250-3262.
[http://dx.doi.org/10.1016/j.clnu.2020.10.051] [PMID: 33190988]
[52]
Lin, A.; Kolossváry, M.; Yuvaraj, J.; Cadet, S.; McElhinney, P.A.; Jiang, C.; Nerlekar, N.; Nicholls, S.J.; Slomka, P.J.; Maurovich-Horvat, P.; Wong, D.T.L.; Dey, D. Myocardial infarction associates with a distinct pericoronary adipose tissue radiomic phenotype. JACC Cardiovasc. Imaging, 2020, 13(11), 2371-2383.
[http://dx.doi.org/10.1016/j.jcmg.2020.06.033] [PMID: 32861654]
[53]
Shirai, T.; Nazarewicz, R.R.; Wallis, B.B.; Yanes, R.E.; Watanabe, R.; Hilhorst, M.; Tian, L.; Harrison, D.G.; Giacomini, J.C.; Assimes, T.L.; Goronzy, J.J.; Weyand, C.M. The glycolytic enzyme PKM2 bridges metabolic and inflammatory dysfunction in coronary artery disease. J. Exp. Med., 2016, 213(3), 337-354.
[http://dx.doi.org/10.1084/jem.20150900] [PMID: 26926996]
[54]
Molina, E.; Chew, G.S.; Myers, S.A.; Clarence, E.M.; Eales, J.M.; Tomaszewski, M.; Charchar, F.J. A novel Y-specific long non-coding RNA associated with cellular lipid accumulation in HepG2 cells and atherosclerosis-related genes. Sci. Rep., 2017, 7(1), 16710.
[http://dx.doi.org/10.1038/s41598-017-17165-9] [PMID: 29196750]
[55]
Lei, D.; Lv, L.; Yang, L.; Wu, W.; Liu, Y.; Tu, Y.; Xu, D.; Jin, Y.; Nong, X.; He, L. Genome-wide analysis of mRNA and long noncoding RNA profiles in chronic actinic dermatitis. BioMed Res. Int., 2017, 2017, 7479523.
[http://dx.doi.org/10.1155/2017/7479523] [PMID: 29359156]
[56]
Wang, Q.C.; Wang, Z.Y.; Xu, Q.; Chen, X.L.; Shi, R.Z. lncRNA expression profiles and associated ceRNA network analyses in epicardial adipose tissue of patients with coronary artery disease. Sci. Rep., 2021, 11(1), 1567.
[http://dx.doi.org/10.1038/s41598-021-81038-5] [PMID: 33452392]
[57]
Li, P.; Xing, J.; Zhang, J.; Jiang, J.; Liu, X.; Zhao, D.; Zhang, Y. Inhibition of long noncoding RNA HIF1A-AS2 confers protection against atherosclerosis via ATF2 downregulation. J. Adv. Res., 2020, 26, 123-135.
[http://dx.doi.org/10.1016/j.jare.2020.07.015] [PMID: 33133688]
[58]
Guo, F.; Tang, C.; Li, Y.; Liu, Y.; Lv, P.; Wang, W.; Mu, Y. The interplay of Lnc RNA ANRIL and miR-181b on the inflammation-relevant coronary artery disease through mediating NF -κB signalling pathway. J. Cell. Mol. Med., 2018, 22(10), 5062-5075.
[http://dx.doi.org/10.1111/jcmm.13790] [PMID: 30079603]
[59]
Bai, J.; Liu, J.; Fu, Z.; Feng, Y.; Wang, B.; Wu, W.; Zhang, R. Silencing lncRNA AK136714 reduces endothelial cell damage and inhibits atherosclerosis. Aging (Albany NY), 2021, 13(10), 14159-14169.
[http://dx.doi.org/10.18632/aging.203031] [PMID: 34015766]
[60]
Wang, Y.; Yang, X.; Jiang, A.; Wang, W.; Li, J.; Wen, J. Methylation-dependent transcriptional repression of RUNX3 by KCNQ1OT1 regulates mouse cardiac microvascular endothelial cell viability and inflammatory response following myocardial infarction. FASEB J., 2019, 33(12), 13145-13160.
[http://dx.doi.org/10.1096/fj.201900310R] [PMID: 31625414]
[61]
Jaeschke, H.; Adelusi, O.B.; Akakpo, J.Y.; Nguyen, N.T.; Sanchez-Guerrero, G.; Umbaugh, D.S.; Ding, W.X.; Ramachandran, A. Recommendations for the use of the acetaminophen hepatotoxicity model for mechanistic studies and how to avoid common pitfalls. Acta Pharm. Sin. B, 2021, 11(12), 3740-3755.
[http://dx.doi.org/10.1016/j.apsb.2021.09.023] [PMID: 35024303]
[62]
Sun, J.; Singh, P.; Österlund, J.; Orho-Melander, M.; Melander, O.; Engström, G.; Edsfeldt, A. Hyperglycaemia-associated Caspase-3 predicts diabetes and coronary artery disease events. J. Intern. Med., 2021, 290(4), 855-865.
[http://dx.doi.org/10.1111/joim.13327] [PMID: 34309093]
[63]
Jayasuriya, R.; Ganesan, K.; Xu, B.; Ramkumar, K.M. Emerging role of long non-coding RNAs in endothelial dysfunction and their molecular mechanisms. Biomed. Pharmacother., 2022, 145, 112421.
[http://dx.doi.org/10.1016/j.biopha.2021.112421] [PMID: 34798473]
[64]
Ni, J.; Huang, Z.; Wang, D. LncRNA TP73-AS1 promotes oxidized low-density lipoprotein-induced apoptosis of endothelial cells in atherosclerosis by targeting the miR-654-3p/AKT3 axis. Cell. Mol. Biol. Lett., 2021, 26(1), 27.
[http://dx.doi.org/10.1186/s11658-021-00264-x] [PMID: 34103010]
[65]
Zhang, H.; Ji, N.; Gong, X.; Ni, S.; Wang, Y. NEAT1/miR-140-3p/MAPK1 mediates the viability and survival of coronary endothelial cells and affects coronary atherosclerotic heart disease. Acta Biochim. Biophys. Sin. (Shanghai), 2020, 52(9), 967-974.
[http://dx.doi.org/10.1093/abbs/gmaa087] [PMID: 32844995]
[66]
Wu, L.; Tan, G.; Li, X.; Jiang, X.; Run, B.; Zhou, W.; Liao, H. LncRNA TONSL-AS1 participates in coronary artery disease by interacting with miR-197. Microvasc. Res., 2021, 136, 104152.
[http://dx.doi.org/10.1016/j.mvr.2021.104152] [PMID: 33662410]
[67]
You, G.; Long, X.; Song, F.; Huang, J.; Tian, M.; Xiao, Y.; Deng, S.; Wu, Q.; Long Noncoding, R.N.A. Long Noncoding, R.N.A. Long noncoding RNA EZR-AS1 regulates the proliferation, migration, and apoptosis of human venous endothelial cells via SMYD3. BioMed Res. Int., 2020, 2020, 1-11.
[http://dx.doi.org/10.1155/2020/6840234] [PMID: 32596350]
[68]
Sun, Y.; Huang, S.; Wan, C.; Ruan, Q.; Xie, X.; Wei, D.; Li, G.; Lin, S.; Li, H.; Wu, S. Knockdown of lncRNA ENST00000609755.1 confers protection against early oxLDL-induced coronary heart disease. Front. Cardiovasc. Med., 2021, 8, 650212.
[http://dx.doi.org/10.3389/fcvm.2021.650212] [PMID: 34095248]
[69]
Li, W.; He, P.; Huang, Y.; Li, Y.F.; Lu, J.; Li, M.; Kurihara, H.; Luo, Z.; Meng, T.; Onishi, M.; Ma, C.; Jiang, L.; Hu, Y.; Gong, Q.; Zhu, D.; Xu, Y.; Liu, R.; Liu, L.; Yi, C.; Zhu, Y.; Ma, N.; Okamoto, K.; Xie, Z.; Liu, J.; He, R.R.; Feng, D. Selective autophagy of intracellular organelles: Recent research advances. Theranostics, 2021, 11(1), 222-256.
[http://dx.doi.org/10.7150/thno.49860] [PMID: 33391472]
[70]
Wang, L.; Xu, C.; Johansen, T.; Berger, S.L.; Dou, Z. SIRT1 – a new mammalian substrate of nuclear autophagy. Autophagy, 2021, 17(2), 593-595.
[http://dx.doi.org/10.1080/15548627.2020.1860541] [PMID: 33292048]
[71]
Nnah, I.C.; Wang, B.; Saqcena, C.; Weber, G.F.; Bonder, E.M.; Bagley, D.; De Cegli, R.; Napolitano, G.; Medina, D.L.; Ballabio, A.; Dobrowolski, R. TFEB-driven endocytosis coordinates MTORC1 signaling and autophagy. Autophagy, 2019, 15(1), 151-164.
[http://dx.doi.org/10.1080/15548627.2018.1511504] [PMID: 30145926]
[72]
Chao, T.; Shih, H.T.; Hsu, S.C.; Chen, P.J.; Fan, Y.S.; Jeng, Y.M.; Shen, Z.Q.; Tsai, T.F.; Chang, Z.F. Autophagy restricts mitochondrial DNA damage-induced release of ENDOG (endonuclease G) to regulate genome stability. Autophagy, 2021, 17(11), 3444-3460.
[http://dx.doi.org/10.1080/15548627.2021.1874209] [PMID: 33465003]
[73]
Hwang, H.Y.; Shim, J.S.; Kim, D.; Kwon, H.J. Antidepressant drug sertraline modulates AMPK-MTOR signaling-mediated autophagy via targeting mitochondrial VDAC1 protein. Autophagy, 2021, 17(10), 2783-2799.
[http://dx.doi.org/10.1080/15548627.2020.1841953] [PMID: 33124469]
[74]
Meng, Q.; Li, Y.; Ji, T.; Chao, Y.; Li, J.; Fu, Y.; Wang, S.; Chen, Q.; Chen, W.; Huang, F.; Wang, Y.; Zhang, Q.; Wang, X.; Bian, H. Estrogen prevent atherosclerosis by attenuating endothelial cell pyroptosis via activation of estrogen receptor α-mediated autophagy. J. Adv. Res., 2021, 28, 149-164.
[http://dx.doi.org/10.1016/j.jare.2020.08.010] [PMID: 33364052]
[75]
He, L.; Chen, Y.; Hao, S.; Qian, J. Uncovering novel landscape of cardiovascular diseases and therapeutic targets for cardioprotection via long noncoding RNA–miRNA–mRNA axes. Epigenomics, 2018, 10(5), 661-671.
[http://dx.doi.org/10.2217/epi-2017-0176] [PMID: 29692219]
[76]
Liang, W.; Fan, T.; Liu, L.; Zhang, L. Knockdown of growth-arrest specific transcript 5 restores oxidized low-density lipoprotein-induced impaired autophagy flux via upregulating miR-26a in human endothelial cells. Eur. J. Pharmacol., 2019, 843, 154-161.
[http://dx.doi.org/10.1016/j.ejphar.2018.11.005] [PMID: 30468731]
[77]
Wang, K.; Yang, C.; Shi, J.; Gao, T. Ox-LDL-induced lncRNA MALAT1 promotes autophagy in human umbilical vein endothelial cells by sponging miR-216a-5p and regulating Beclin-1 expression. Eur. J. Pharmacol., 2019, 858, 172338.
[http://dx.doi.org/10.1016/j.ejphar.2019.04.019] [PMID: 31029709]
[78]
Zhu, Y.; Yang, T.; Duan, J.; Mu, N.; Zhang, T. MALAT1/miR-15b-5p/MAPK1 mediates endothelial progenitor cells autophagy and affects coronary atherosclerotic heart disease via mTOR signaling pathway. Aging (Albany NY), 2019, 11(4), 1089-1109.
[http://dx.doi.org/10.18632/aging.101766] [PMID: 30787203]
[79]
Xiao, J.; Lu, Y.; Yang, X. THRIL mediates endothelial progenitor cells autophagy via AKT pathway and FUS. Mol. Med., 2020, 26(1), 86.
[http://dx.doi.org/10.1186/s10020-020-00201-2] [PMID: 32907536]
[80]
You, G.; Long, X.; Song, F.; Huang, J.; Tian, M.; Xiao, Y.; Deng, S.; Wu, Q. Metformin activates the AMPK-mTOR pathway by modulating lncRNA TUG1 to induce autophagy and inhibit atherosclerosis. Drug Des. Devel. Ther., 2020, 14, 457-468.
[http://dx.doi.org/10.2147/DDDT.S233932] [PMID: 32099330]
[81]
Jin, H.; Zhu, Y.; Wang, X.; Luo, E.; Li, Y.; Wang, B.; Chen, Y. BDNF corrects NLRP3 inflammasome-induced pyroptosis and glucose metabolism reprogramming through KLF2/HK1 pathway in vascular endothelial cells. Cell. Signal., 2021, 78, 109843.
[http://dx.doi.org/10.1016/j.cellsig.2020.109843] [PMID: 33253911]
[82]
McCarty, M.F.; Iloki Assanga, S.B.; Lewis Luján, L.; O’Keefe, J.H.; DiNicolantonio, J.J. Nutraceutical strategies for suppressing NLRP3 inflammasome activation: pertinence to the management of COVID-19 and beyond. Nutrients, 2020, 13(1), 47.
[http://dx.doi.org/10.3390/nu13010047] [PMID: 33375692]
[83]
Wu, P.; Chen, J.; Chen, J.; Tao, J.; Wu, S.; Xu, G.; Wang, Z.; Wei, D.; Yin, W. Trimethylamine N-oxide promotes apoE −/− mice atherosclerosis by inducing vascular endothelial cell pyroptosis via the SDHB/ROS pathway. J. Cell. Physiol., 2020, 235(10), 6582-6591.
[http://dx.doi.org/10.1002/jcp.29518] [PMID: 32012263]
[84]
Yang, M.; Lv, H.; Liu, Q.; Zhang, L.; Zhang, R.; Huang, X.; Wang, X.; Han, B.; Hou, S.; Liu, D.; Wang, G.; Hou, J.; Yu, B. Colchicine alleviates cholesterol crystal-induced endothelial cell pyroptosis through activating AMPK/SIRT1 pathway. Oxid. Med. Cell. Longev., 2020, 2020, 9173530.
[http://dx.doi.org/10.1155/2020/9173530] [PMID: 32733639]
[85]
Zhang, Y.; Liu, X.; Bai, X.; Lin, Y.; Li, Z.; Fu, J.; Li, M.; Zhao, T.; Yang, H.; Xu, R.; Li, J.; Ju, J.; Cai, B.; Xu, C.; Yang, B. Melatonin prevents endothelial cell pyroptosis via regulation of long noncoding RNA MEG3/ miR-223/NLRP3 axis. J. Pineal Res., 2018, 64(2), e12449.
[http://dx.doi.org/10.1111/jpi.12449] [PMID: 29024030]
[86]
Song, B.; Dang, H.; Dong, R. Differential expression of LOXL1-AS1 in coronary heart disease and its regulatory mechanism in ox-LDL-induced human coronary artery endothelial cell pyroptosis. Cardiovasc. Drugs Ther., 2021. [Online ahead of print].
[http://dx.doi.org/10.1007/s10557-021-07274-z] [PMID: 34633594]
[87]
Wu, L.M.; Wu, S.G.; Chen, F.; Wu, Q.; Wu, C.M.; Kang, C.M.; He, X.; Zhang, R.Y.; Lu, Z.F.; Li, X.H.; Xu, Y.J.; Li, L.M.; Ding, L.; Bai, H.L.; Liu, X.H.; Hu, Y.W.; Zheng, L. Atorvastatin inhibits pyroptosis through the lncRNA NEXN-AS1/NEXN pathway in human vascular endothelial cells. Atherosclerosis, 2020, 293, 26-34.
[http://dx.doi.org/10.1016/j.atherosclerosis.2019.11.033] [PMID: 31830726]
[88]
Song, Y.; Yang, L.; Guo, R.; Lu, N.; Shi, Y.; Wang, X. Long noncoding RNA MALAT1 promotes high glucose-induced human endothelial cells pyroptosis by affecting NLRP3 expression through competitively binding miR-22. Biochem. Biophys. Res. Commun., 2019, 509(2), 359-366.
[http://dx.doi.org/10.1016/j.bbrc.2018.12.139] [PMID: 30591217]
[89]
Zhang, X.; Chen, Z.; Zang, J.; Yao, C.; Shi, J.; Nie, R.; Wu, G. LncRNA-mRNA co-expression analysis discovered the diagnostic and prognostic biomarkers and potential therapeutic agents for myocardial infarction. Aging (Albany NY), 2021, 13(6), 8944-8959.
[http://dx.doi.org/10.18632/aging.202713] [PMID: 33668039]
[90]
Chen, Z.; Zhou, D.; Zhang, X.; Wu, Q.; Wu, G. Diagnostic biomarkers and potential drug targets for coronary artery disease as revealed by systematic analysis of lncRNA characteristics. Ann. Transl. Med., 2021, 9(15), 1243.
[http://dx.doi.org/10.21037/atm-21-3276] [PMID: 34532380]
[91]
Zuo, J.; Xu, M.; Wang, D.; Bai, W.; Li, G. Role of competitive endogenous RNA networks in the pathogenesis of coronary artery disease. Ann. Transl. Med., 2021, 9(15), 1234.
[http://dx.doi.org/10.21037/atm-21-2737] [PMID: 34532371]
[92]
Zhao, Z.; Sun, W.; Guo, Z.; Liu, B.; Yu, H.; Zhang, J. Long noncoding RNAs in myocardial ischemia-reperfusion injury. Oxid. Med. Cell. Longev., 2021, 2021, 8889123.
[http://dx.doi.org/10.1155/2021/8889123] [PMID: 33884101]
[93]
Boon, R.A.; Jaé, N.; Holdt, L.; Dimmeler, S. Long noncoding RNAs. J. Am. Coll. Cardiol., 2016, 67(10), 1214-1226.
[http://dx.doi.org/10.1016/j.jacc.2015.12.051] [PMID: 26965544]
[94]
Qian, X.; Zhao, J.; Yeung, P.Y.; Zhang, Q.C.; Kwok, C.K. Revealing lncRNA structures and interactions by sequencing-based approaches. Trends Biochem. Sci., 2019, 44(1), 33-52.
[http://dx.doi.org/10.1016/j.tibs.2018.09.012] [PMID: 30459069]
[95]
Ali, T.; Grote, P. Beyond the RNA-dependent function of LncRNA genes. eLife, 2020, 9, e60583.
[http://dx.doi.org/10.7554/eLife.60583] [PMID: 33095159]

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