Advances in the study of S100A9 in cardiovascular diseases

Fengling Chen , Ziyu He , Chengming Wang , Jiajia Si , Zhu Chen , Yuan Guo

Cell Proliferation ›› 2024, Vol. 57 ›› Issue (8) : e13636

PDF
Cell Proliferation ›› 2024, Vol. 57 ›› Issue (8) : e13636 DOI: 10.1002/cpr.13636
REVIEW

Advances in the study of S100A9 in cardiovascular diseases

Author information +
History +
PDF

Abstract

Cardiovascular disease (CVD) is a group of diseases that primarily affect the heart or blood vessels, with high disability and mortality rates, posing a serious threat to human health. The causative factors, pathogenesis, and characteristics of common CVD differ, but they all involve common pathological processes such as inflammation, oxidative stress, and fibrosis. S100A9 belongs to the S100 family of calcium-binding proteins, which are mainly secreted by myeloid cells and bind to the Toll-like receptor 4 and receptor for advanced glycation end products and is involved in regulating pathological processes such as inflammatory response, fibrosis, vascular calcification, and endothelial barrier function in CVD. The latest research has found that S100A9 is a key biomarker for diagnosing and predicting various CVD. Therefore, this article reviews the latest research progress on the diagnostic and predictive, and therapeutic value of S100A9 in inflammatory-related CVD such as atherosclerosis, myocardial infarction, and arterial aneurysm and summarizes its molecular mechanisms in the progression of CVD, aiming to explore new predictive methods and to identify potential intervention targets for CVD in clinical practice.

Cite this article

Download citation ▾
Fengling Chen, Ziyu He, Chengming Wang, Jiajia Si, Zhu Chen, Yuan Guo. Advances in the study of S100A9 in cardiovascular diseases. Cell Proliferation, 2024, 57(8): e13636 DOI:10.1002/cpr.13636

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

JosephP, LeongD, McKeeM, et al. Reducing the global burden of cardiovascular disease, part 1: the epidemiology and risk factors. Circ Res. 2017;121(6):677-694.

[2]

SchultzWM, KelliHM, LiskoJC, et al. Socioeconomic status and cardiovascular outcomes: challenges and interventions. Circulation. 2018;137(20):2166-2178.

[3]

ImigJD, Cervenka L, NeckarJ. Epoxylipids and soluble epoxide hydrolase in heart diseases. Biochem Pharmacol. 2022;195:114866.

[4]

GoliaE, Limongelli G, NataleF, et al. Inflammation and cardiovascular disease: from pathogenesis to therapeutic target. Curr Atheroscler Rep. 2014;16(9):435.

[5]

SpeerT, Dimmeler S, SchunkSJ, FliserD, RidkerPM. Targeting innate immunity-driven inflammation in CKD and cardiovascular disease. Nat Rev Nephrol. 2022;18(12):762-778.

[6]

YiX, ZhuQX, WuXL, TanTT, JiangXJ. Histone methylation and oxidative stress in cardiovascular diseases. Oxid Med Cell Longev. 2022;2022:6023710.

[7]

SorrientoD, Iaccarino G. Inflammation and cardiovascular diseases: the Most recent findings. Int J Mol Sci. 2019;20(16):3879.

[8]

MohebiR, McCarthy CP, GagginHK, van KimmenadeRRJ, Januzzi JL. Inflammatory biomarkers and risk of cardiovascular events in patients undergoing coronary angiography. Am Heart J. 2022;252:51-59.

[9]

TraversJG, TharpCA, RubinoM, McKinsey TA. Therapeutic targets for cardiac fibrosis: from old school to next-gen. J Clin Invest. 2022;132(5):e148554.

[10]

GuoY, ChenJ, QiuH. Novel mechanisms of exercise-induced cardioprotective factors in myocardial infarction. Front Physiol. 2020;11:199.

[11]

SenonerT, DichtlW. Oxidative stress in cardiovascular diseases: still a therapeutic target? Nutrients. 2019;11(9):2090.

[12]

Medina-LeyteDJ, Zepeda-García O, Domínguez-Pérez M, González-GarridoA, Villarreal-MolinaT, Jacobo-AlbaveraL. Endothelial dysfunction, inflammation and coronary artery disease: potential biomarkers and promising therapeutical approaches. Int J Mol Sci. 2021;22(8):3850.

[13]

JinZ, NiuJ, KapoorN, Liang J, BecerraE, KolattukudyPE. Essential role of endothelial MCPIP in vascular integrity and post-ischemic remodeling. Int J Mol Sci. 2019;20(1):172.

[14]

XuS, IlyasI, LittlePJ, et al. Endothelial dysfunction in atherosclerotic cardiovascular diseases and beyond: from mechanism to pharmacotherapies. Pharmacol Rev. 2021;73(3):924-967.

[15]

ImmanuelJ, YunS. Vascular inflammatory diseases and endothelial phenotypes. Cells. 2023;12(12):1640.

[16]

ZhengY, HuangS, ZhangJ, et al. Melatonin alleviates vascular endothelial cell damage by regulating an autophagy-apoptosis axis in Kawasaki disease. Cell Prolif. 2022;55(6):e13251.

[17]

Silvestre-RoigC, Braster Q, Ortega-GomezA, SoehnleinO. Neutrophils as regulators of cardiovascular inflammation. Nat Rev Cardiol. 2020;17(6):327-340.

[18]

WangS, SongR, WangZ, Jing Z, WangS, MaJ. S100A8/A9 in inflammation. Front Immunol. 2018;9:1298.

[19]

SchiopuA, CotoiOS. S100A8 and S100A9: DAMPs at the crossroads between innate immunity, traditional risk factors, and cardiovascular disease. Mediators Inflamm. 2013;2013:828354.

[20]

ChenX, HeJ, XieY, et al. Tetrahedral framework nucleic acid nanomaterials reduce the inflammatory damage in sepsis by inhibiting pyroptosis. Cell Prolif. 2023;56(8):e13424.

[21]

BhardwajRS, ZotzC, RothJ, et al. The calcium-binding proteins MRP8 and MRP14 form a membrane-associated heterodimer in a subset of monocytes/macrophages present in acute but absent in chronic inflammatory lesions. Eur J Immunol. 1992;22(7):1891-1897.

[22]

HunterMJ, ChazinWJ. High level expression and dimer characterization of the S100 EF-hand proteins, migration inhibitory factor-related proteins 8 and 14. J Biol Chem. 1998;273(20):12427-12435.

[23]

BjörkP, Björk A, VoglT, et al. Identification of human S100A9 as a novel target for treatment of autoimmune disease via binding to quinoline-3-carboxamides. PLoS Biol. 2009;7(4):e1000097.

[24]

WuY, LiY, ZhangC, et al. S100a8/a9 released by CD11b+Gr1+ neutrophils activates cardiac fibroblasts to initiate angiotensin II-induced cardiac inflammation and injury. Hypertension. 2014;63(6):1241-1250.

[25]

SunY, WangZ, HouJ, et al. Shuangxinfang prevents S100A9-induced macrophage/microglial inflammation to improve cardiac function and depression-like behavior in rats after acute myocardial infarction. Front Pharmacol. 2022;13:832590.

[26]

SimardJC, CesaroA, Chapeton-MontesJ, et al. S100A8 and S100A9 induce cytokine expression and regulate the NLRP3 inflammasome via ROS-dependent activation of NF-κB(1.). PloS One. 2013;8(8):e72138.

[27]

ZhangW, LavineKJ, EpelmanS, et al. Necrotic myocardial cells release damage-associated molecular patterns that provoke fibroblast activation in vitro and trigger myocardial inflammation and fibrosis in vivo. J Am Heart Assoc. 2015;4(6):e001993.

[28]

XiaC, Braunstein Z, ToomeyAC, ZhongJ, RaoX. S100 proteins As an important regulator of macrophage inflammation. Front Immunol. 2017;8:1908.

[29]

FanS, ZhaoH, LiuY, et al. Isoproterenol triggers ROS/P53/S100-A9 positive feedback to aggravate myocardial damage associated with complement activation. Chem Res Toxicol. 2020;33(10):2675-2685.

[30]

WangL, LuoH, ChenX, Jiang Y, HuangQ. Functional characterization of S100A8 and S100A9 in altering monolayer permeability of human umbilical endothelial cells. PloS One. 2014;9(3):e90472.

[31]

DahlemC, KadoSY, HeY, et al. AHR signaling interacting with nutritional factors regulating the expression of markers in vascular inflammation and atherogenesis. Int J Mol Sci. 2020;21(21):E8287.

[32]

ZhaoB, YuJ, LuoY, et al. Deficiency of S100 calcium binding protein A9 attenuates vascular dysfunction in aged mice. Redox Biol. 2023;63:102721.

[33]

NakanishiT, IidaS, MaruyamaJ, et al. Arteriosclerosis derived from cutaneous inflammation is ameliorated by the deletion of IL-17A and IL-17F. Int J Mol Sci. 2023;24(6):5434.

[34]

LiuY, LuoG, HeD. Clinical importance of S100A9 in osteosarcoma development and as a diagnostic marker and therapeutic target. Bioengineered. 2019;10(1):133-141.

[35]

ItouH, YaoM, FujitaI, et al. The crystal structure of human MRP14 (S100A9), a Ca(2+)-dependent regulator protein in inflammatory process. J Mol Biol. 2002;316(2):265-276.

[36]

SalminenA, Vlachopoulou E, HavulinnaAS, et al. Genetic variants contributing to circulating matrix metalloproteinase 8 levels and their association with cardiovascular diseases: a genome-wide analysis. Circ Cardiovasc Genet. 2017;10(6):e001731.

[37]

ChenH, LunneyJK, ChengL, et al. Porcine S100A8 and S100A9: molecular characterizations and crucial functions in response to Haemophilus parasuis infection. Dev Comp Immunol. 2011;35(4):490-500.

[38]

TamulytėR, Jankaitytė E, ToleikisZ, SmirnovasV, Jankunec M. Pro-inflammatory protein S100A9 alters membrane organization by dispersing ordered domains. Biochim Biophys Acta Biomembr. 2023;1865(3):184113.

[39]

PruensterM, ImmlerR, RothJ, et al. E-selectin-mediated rapid NLRP3 inflammasome activation regulates S100A8/S100A9 release from neutrophils via transient gasdermin D pore formation. Nat Immunol. 2023;30:2021-2031.

[40]

FanZP, PengML, ChenYY, et al. S100A9 activates the immunosuppressive switch through the PI3K/Akt pathway to maintain the immune suppression function of testicular macrophages. Front Immunol. 2021;12:743354.

[41]

SrikrishnaG. S100A8 and S100A9: New insights into their roles in malignancy. J Innate Immun. 2011;4(1):31-40.

[42]

AverillMM, Barnhart S, BeckerL, et al. S100A9 differentially modifies phenotypic states of neutrophils, macrophages, and dendritic cells: implications for atherosclerosis and adipose tissue inflammation. Circulation. 2011;123(11):1216-1226.

[43]

MonteiroC, MiarkaL, Perea-GarcíaM, et al. Stratification of radiosensitive brain metastases based on an actionable S100A9/RAGE resistance mechanism. Nat Med. 2022;28(4):752-765. doi:10.1038/s41591-022-01749-8

[44]

XiaoX, YangC, QuSL, et al. S100 proteins in atherosclerosis. Clin Chim Acta. 2020;502:293-304.

[45]

BertoliniI, PeregoM, NefedovaY, et al. Intercellular hif1α reprograms mammary progenitors and myeloid immune evasion to drive high-risk breast lesions. J Clin Invest. 2023;133(8):e164348.

[46]

HuangX, ShenW, VeizadesS, Liang G, SayedN, NguyenPK. Single-cell transcriptional profiling reveals sex and age diversity of gene expression in mouse endothelial cells. Front Genet. 2021;12:590377.

[47]

BoteanuRM, SuicaVI, UyyE, et al. Short-term blockade of pro-inflammatory alarmin S100A9 favorably modulates left ventricle proteome and related signaling pathways involved in post-myocardial infarction recovery. Int J Mol Sci. 2022;23(9):5289.

[48]

MihailaAC, Ciortan L, MacarieRD, et al. Transcriptional profiling and functional analysis of N1/N2 neutrophils reveal an immunomodulatory effect of S100A9-blockade on the pro-inflammatory N1 subpopulation. Front Immunol. 2021;12:708770.

[49]

UrsinoG, Lucibello G, TeixeiraPDS, et al. S100A9 exerts insulin-independent antidiabetic and anti-inflammatory effects. Sci Adv. 2024;10(1):eadj4686.

[50]

ChenTJ, YehYT, PengFS, Li AH, WuSC. S100A8/A9 enhances immunomodulatory and tissue-repairing properties of human amniotic mesenchymal stem cells in myocardial ischemia-reperfusion injury. Int J Mol Sci. 2021;22(20):11175.

[51]

VoglT, Stratis A, WixlerV, et al. Autoinhibitory regulation of S100A8/S100A9 alarmin activity locally restricts sterile inflammation. J Clin Invest. 2018;128(5):1852-1866.

[52]

ZhanX, WuR, KongXH, et al. Elevated neutrophil extracellular traps by HBV-mediated S100A9-TLR4/RAGE-ROS cascade facilitate the growth and metastasis of hepatocellular carcinoma. Cancer Commun. 2023;43(2):225-245.

[53]

ZhangX, WeiL, WangJ, et al. Suppression colitis and colitis-associated colon cancer by anti-S100a9 antibody in mice. Front Immunol. 2017;8:1774.

[54]

LiC, ChenH, DingF, et al. A novel p53 target gene, S100A9, induces p53-dependent cellular apoptosis and mediates the p53 apoptosis pathway. Biochem J. 2009;422(2):363-372.

[55]

ZhangY, ZhaZ, ShenW, et al. Anemoside B4 ameliorates TNBS-induced colitis through S100A9/MAPK/NF-κB signaling pathway. Chin Med. 2021;16(1):11.

[56]

GuoS, SuQ, WenJ, et al. S100A9 induces nucleus pulposus cell degeneration through activation of the NF-κB signaling pathway. J Cell Mol Med. 2021;25(10):4709-4720.

[57]

NagareddyPR, Kraakman M, MastersSL, et al. Adipose tissue macrophages promote myelopoiesis and monocytosis in obesity. Cell Metab. 2014;19(5):821-835.

[58]

UrsinoG, Ramadori G, HöflerA, et al. Hepatic non-parenchymal S100A9-TLR4-mTORC1 axis normalizes diabetic ketogenesis. Nat Commun. 2022;13(1):4107.

[59]

WangA, GuoB, JiaQ, ChenYU, GaoX, XuS. S100A9-containing serum exosomes of burn injury patients promote permeability of pulmonary microvascular endothelial cells. J Biosci. 2021;46:33.

[60]

LeeNR, ParkBS, KimSY, et al. Cytokine secreted by S100A9 via TLR4 in monocytes delays neutrophil apoptosis by inhibition of caspase 9/3 pathway. Cytokine. 2016;86:53-63.

[61]

YiW, ZhuR, HouX, WuF, FengR. Integrated analysis reveals S100a8/a9 regulates autophagy and apoptosis through the MAPK and PI3K-AKT signaling pathway in the early stage of myocardial infarction. Cells. 2022;11(12):1911.

[62]

MüllerI, VoglT, PappritzK, et al. Pathogenic role of the damage-associated molecular patterns S100A8 and S100A9 in coxsackievirus B3-induced myocarditis. Circ Heart Fail. 2017;10(11):e004125.

[63]

KawakamiR, Katsuki S, TraversR, et al. S100A9-RAGE Axis accelerates formation of macrophage-mediated extracellular vesicle microcalcification in diabetes mellitus. Arterioscler Thromb Vasc Biol. 2020;40(8):1838-1853.

[64]

BiswasAK, HanS, TaiY, et al. Targeting S100A9-ALDH1A1-retinoic acid signaling to suppress brain relapse in EGFR-mutant lung cancer. Cancer Discov. 2022;12(4):1002-1021.

[65]

ZhaH, LiX, SunH, et al. S100A9 promotes the proliferation and migration of cervical cancer cells by inducing epithelial-mesenchymal transition and activating the Wnt/β-catenin pathway. Int J Oncol. 2019;55(1):35-44. doi:10.3892/ijo.2019.4793

[66]

TumurkhuuG, Shimada K, DagvadorjJ, et al. Ogg1-dependent DNA repair regulates NLRP3 inflammasome and prevents atherosclerosis. Circ Res. 2016;119(6):e76-e90.

[67]

KongP, CuiZY, HuangXF, Zhang DD, GuoRJ, HanM. Inflammation and atherosclerosis: signaling pathways and therapeutic intervention. Signal Transduct Target Ther. 2022;7(1):131.

[68]

ZhaolinZ, GuohuaL, ShiyuanW, Zuo W. Role of pyroptosis in cardiovascular disease. Cell Prolif. 2019;52(2):e12563.

[69]

HanssonGK, Hermansson A. The immune system in atherosclerosis. Nat Immunol. 2011;12(3):204-212.

[70]

LibbyP. The changing landscape of atherosclerosis. Nature. 2021;592(7855):524-533.

[71]

NewSEP, Goettsch C, AikawaM, et al. Macrophage-derived matrix vesicles: an alternative novel mechanism for microcalcification in atherosclerotic plaques. Circ Res. 2013;113(1):72-77.

[72]

IonitaMG, VinkA, DijkeIE, et al. High levels of myeloid-related protein 14 in human atherosclerotic plaques correlate with the characteristics of rupture-prone lesions. Arterioscler Thromb Vasc Biol. 2009;29(8):1220-1227.

[73]

LangleySR, Willeit K, DidangelosA, et al. Extracellular matrix proteomics identifies molecular signature of symptomatic carotid plaques. J Clin Invest. 2017;127(4):1546-1560.

[74]

FlynnMC, Kraakman MJ, TikellisC, et al. Transient intermittent hyperglycemia accelerates atherosclerosis by promoting myelopoiesis. Circ Res. 2020;127(7):877-892.

[75]

HanssenNMJ, Tikellis C, PickeringRJ, et al. Pyridoxamine prevents increased atherosclerosis by intermittent methylglyoxal spikes in the aortic arches of ApoE−/− mice. Biomed Pharmacother. 2023;158:114211.

[76]

KraakmanMJ, LeeMK, Al-ShareaA, et al. Neutrophil-derived S100 calcium-binding proteins A8/A9 promote reticulated thrombocytosis and atherogenesis in diabetes. J Clin Invest. 2017;127(6):2133-2147.

[77]

ThygesenK, AlpertJS, JaffeAS, et al. Fourth universal definition of myocardial infarction. J Am Coll Cardiol. 2018;72(18):2231-2264.

[78]

MarinkovićG, Koenis DS, de CampL, et al. S100A9 links inflammation and repair in myocardial infarction. Circ Res. 2020;127(5):664-676.

[79]

MichaudK, BassoC, d’AmatiG, et al. Diagnosis of myocardial infarction at autopsy: AECVP reappraisal in the light of the current clinical classification. Virchows Arch. 2020;476(2):179-194.

[80]

MarinkovićG, Grauen Larsen H, YndigegnT, et al. Inhibition of pro-inflammatory myeloid cell responses by short-term S100A9 blockade improves cardiac function after myocardial infarction. Eur Heart J. 2019;40(32):2713-2723.

[81]

SreejitG, NootiSK, JaggersRM, et al. Retention of the NLRP3 inflammasome-primed neutrophils in the bone marrow is essential for myocardial infarction-induced granulopoiesis. Circulation. 2022;145(1):31-44.

[82]

PanW, ZhangJ, ZhangL, et al. Comprehensive view of macrophage autophagy and its application in cardiovascular diseases. Cell Prolif. 2023;57:e13525.

[83]

GuoY, LuoF, LiuQ, XuD. Regulatory non-coding RNAs in acute myocardial infarction. J Cell Mol Med. 2017;21(5):1013-1023.

[84]

AydinS, UgurK, AydinS, Sahin İ, YardimM. Biomarkers in acute myocardial infarction: current perspectives. Vasc Health Risk Manag. 2019;15:1-10.

[85]

HealyAM, Pickard MD, PradhanAD, et al. Platelet expression profiling and clinical validation of myeloid-related protein-14 as a novel determinant of cardiovascular events. Circulation. 2006;113(19):2278-2284.

[86]

AltweggLA, Neidhart M, HersbergerM, et al. Myeloid-related protein 8/14 complex is released by monocytes and granulocytes at the site of coronary occlusion: a novel, early, and sensitive marker of acute coronary syndromes. Eur Heart J. 2007;28(8):941-948.

[87]

FraccarolloD, NeuserJ, MöllerJ, RiehleC, Galuppo P, BauersachsJ. Expansion of CD10neg neutrophils and CD14+HLA-DRneg/low monocytes driving proinflammatory responses in patients with acute myocardial infarction. Elife. 2021;10:e66808.

[88]

JoshiA, Schmidt LE, BurnapSA, et al. Neutrophil-derived protein S100A8/A9 alters the platelet proteome in acute myocardial infarction and is associated with changes in platelet reactivity. Arterioscler Thromb Vasc Biol. 2022;42(1):49-62.

[89]

LinZL, LiuYC, GaoYL, et al. S100A9 and SOCS3 as diagnostic biomarkers of acute myocardial infarction and their association with immune infiltration. Genes Genet Syst. 2022;97(2):67-79.

[90]

SreejitG, Abdel-Latif A, AthmanathanB, et al. Neutrophil-derived S100A8/A9 amplify granulopoiesis after myocardial infarction. Circulation. 2020;141(13):1080-1094.

[91]

LiY, ChenB, YangX, et al. S100a8/a9 signaling causes mitochondrial dysfunction and cardiomyocyte death in response to ischemic/reperfusion injury. Circulation. 2019;140(9):751-764.

[92]

ChaliseU, Becirovic-Agic M, DasekeMJ, et al. S100A9 is a functional effector of infarct wall thinning after myocardial infarction. Am J Physiol Heart Circ Physiol. 2022;322(2):H145-H155.

[93]

SimonF, Oberhuber A, FlorosN, et al. Acute limb ischemia-much more than just a lack of oxygen. Int J Mol Sci. 2018;19(2):374.

[94]

RichardsGHC, HongKL, HeneinMY, Hanratty C, BolesU. Coronary artery ectasia: review of the non-atherosclerotic molecular and pathophysiologic concepts. Int J Mol Sci. 2022;23(9):5195.

[95]

HuY, ChiL, KueblerWM, Goldenberg NM. Perivascular inflammation in pulmonary arterial hypertension. Cells. 2020;9(11):2338.

[96]

CuiH, ChenY, LiK, et al. Untargeted metabolomics identifies succinate as a biomarker and therapeutic target in aortic aneurysm and dissection. Eur Heart J. 2021;42(42):4373-4385.

[97]

AdayAW, Matsushita K. Epidemiology of peripheral artery disease and polyvascular disease. Circ Res. 2021;128(12):1818-1832.

[98]

ChanNC, XuK, de VriesTAC, Eikelboom JW, HirshJ. Inflammation as a mechanism and therapeutic target in peripheral artery disease. Can J Cardiol. 2022;38(5):588-600.

[99]

ZhangL, WangY, WuG, et al. Blockade of JAK2 protects mice against hypoxia-induced pulmonary arterial hypertension by repressing pulmonary arterial smooth muscle cell proliferation. Cell Prolif. 2020;53(2):e12742.

[100]

ZhangQ, CaoY, LuoQ, et al. The transient receptor potential vanilloid-3 regulates hypoxia-mediated pulmonary artery smooth muscle cells proliferation via PI3K/AKT signaling pathway. Cell Prolif. 2018;51(3):e12436.

[101]

QiuH, ZhangY, LiZ, et al. Donepezil ameliorates pulmonary arterial hypertension by inhibiting M2-macrophage activation. Front Cardiovasc Med. 2021;8:639541.

[102]

RabinovitchM, Guignabert C, HumbertM, NicollsMR. Inflammation and immunity in the pathogenesis of pulmonary arterial hypertension. Circ Res. 2014;115(1):165-175.

[103]

ZhangL, ZengXX, LiYM, et al. Keratin 1 attenuates hypoxic pulmonary artery hypertension by suppressing pulmonary artery media smooth muscle expansion. Acta Physiol (Oxf). 2021;231(2):e13558.

[104]

NakamuraK, Sakaguchi M, MatsubaraH, et al. Crucial role of RAGE in inappropriate increase of smooth muscle cells from patients with pulmonary arterial hypertension. PloS One. 2018;13(9):e0203046.

[105]

ThenappanT, Ormiston ML, RyanJJ, ArcherSL. Pulmonary arterial hypertension: pathogenesis and clinical management. BMJ. 2018;360:j5492.

[106]

YakuA, Inagaki T, AsanoR, et al. Regnase-1 prevents pulmonary arterial hypertension through mRNA degradation of interleukin-6 and platelet-derived growth factor in alveolar macrophages. Circulation. 2022;146(13):1006-1022.

[107]

GuoY, HeZ, ChenZ, et al. Inhibition of Th17 cells by donepezil ameliorates experimental lung fibrosis and pulmonary hypertension. Theranostics. 2023;13(6):1826-1842.

[108]

ZengH, LiuX, ZhangY. Identification of potential biomarkers and immune infiltration characteristics in idiopathic pulmonary arterial hypertension using bioinformatics analysis. Front Cardiovasc Med. 2021;8:624714.

[109]

TazTA, AhmedK, PaulBK, Al-Zahrani FA, MahmudSMH, MoniMA. Identification of biomarkers and pathways for the SARS-CoV-2 infections that make complexities in pulmonary arterial hypertension patients. Brief Bioinform. 2021;22(2):1451-1465.

[110]

DiekmannF, Chouvarine P, SallmonH, et al. Soluble receptor for advanced glycation end products (sRAGE) is a sensitive biomarker in human pulmonary arterial hypertension. Int J Mol Sci. 2021;22(16):8591.

[111]

HeM, ShenJ, ZhangC, Chen Y, WangW, TaoK. Long-chain non-coding RNA metastasis-related lung adenocarcinoma transcript 1 (MALAT1) promotes the proliferation and migration of human pulmonary artery smooth muscle cells (hPASMCs) by regulating the MicroRNA-503 (miR-503)/toll-like receptor 4 (TLR4) signal axis. Med Sci Monit. 2020;26:e923123.

[112]

ZuoZT, MaY, SunY, BaiCQ, ZhouHY, Chen BH. Role of TLR4/NF-κB signalling pathway in pulmonary arterial hypertension in patients with chronic obstructive pulmonary disease. J Coll Physicians Surg Pak. 2020;30(6):568-573.

[113]

JungE, RomeroR, YeoL, et al. The etiology of preeclampsia. Am J Obstet Gynecol. 2022;226(2S):S844-S866.

[114]

OzekiA, OogakiY, HenmiY, et al. Elevated S100A9 in preeclampsia induces soluble endoglin and IL-1β secretion and hypertension via the NLRP3 inflammasome. J Hypertens. 2022;40(1):84-93.

[115]

NakanoK, WadaK, NomuraR, et al. Characterization of aortic aneurysms in cardiovascular disease patients harboring Porphyromonas gingivalis. Oral Dis. 2011;17(4):370-378.

[116]

NakaokaH, TajimaA, YoneyamaT, et al. Gene expression profiling reveals distinct molecular signatures associated with the rupture of intracranial aneurysm. Stroke. 2014;45(8):2239-2245.

[117]

de KorteAM, Aquarius R, VoglT, et al. Elevation of inflammatory S100A8/S100A9 complexes in intracranial aneurysms. J Neurointerv Surg. 2020;12(11):1117-1121.

[118]

LechM, GuessJ, DuffnerJ, et al. Circulating markers of inflammation persist in children and adults with giant aneurysms after Kawasaki disease. Circ Genom Precis Med. 2019;12(4):e002433.

[119]

WangC, KouY, HanY, LiX. Early serum calprotectin (S100A8/A9) predicts delayed cerebral ischemia and outcomes after aneurysmal subarachnoid hemorrhage. J Stroke Cerebrovasc Dis. 2020;29(5):104770.

[120]

ZhangB, ZengK, GuanRC, et al. Single-cell RNA-seq analysis reveals macrophages are involved in the pathogenesis of human sporadic acute type A aortic dissection. Biomolecules. 2023;13(2):399.

[121]

SalyersZR, Mariani V, BalestrieriN, et al. S100A8 and S100A9 are elevated in chronically threatened ischemic limb muscle and induce ischemic mitochondrial pathology in mice. JVS Vasc Sci. 2022;3:232-245.

[122]

Saenz-PipaonG, San Martín P, PlanellN, et al. Functional and transcriptomic analysis of extracellular vesicles identifies calprotectin as a new prognostic marker in peripheral arterial disease (PAD). J Extracell Vesicles. 2020;9(1):1729646.

[123]

Saenz-PipaonG, Ravassa S, LarsenKL, et al. Lipocalin-2 and calprotectin potential prognosis biomarkers in peripheral arterial disease. Eur J Vasc Endovasc Surg. 2022;63(4):648-656.

[124]

LiJ, YaoY, WangY, et al. Modulation of the crosstalk between Schwann cells and macrophages for nerve regeneration: a therapeutic strategy based on a multifunctional tetrahedral framework nucleic acids system. Adv Mater. 2022;34(46):e2202513.

[125]

GantaVC, ChoiM, FarberCR, Annex BH. Antiangiogenic VEGF165b regulates macrophage polarization via S100A8/S100A9 in peripheral artery disease. Circulation. 2019;139(2):226-242.

[126]

MaronBJ. Clinical course and Management of Hypertrophic Cardiomyopathy. N Engl J Med. 2018;379(7):655-668.

[127]

GargL, GuptaM, SabzwariSRA, et al. Atrial fibrillation in hypertrophic cardiomyopathy: prevalence, clinical impact, and management. Heart Fail Rev. 2019;24(2):189-197.

[128]

LekawanvijitS. Cardiotoxicity of uremic toxins: a driver of cardiorenal syndrome. Toxins. 2018;10(9):352.

[129]

BeckerRC, OwensAP, SadayappanS. Tissue-level inflammation and ventricular remodeling in hypertrophic cardiomyopathy. J Thromb Thrombolysis. 2020;49(2):177-183.

[130]

CaiX, HongL, LiuY, HuangX, LaiH, ShaoL. Salmonella pathogenicity island 1 knockdown confers protection against myocardial fibrosis and inflammation in uremic cardiomyopathy via down-regulation of S100 calcium binding protein A8/A9 transcription. Ren Fail. 2022;44(1):1819-1832.

[131]

AjoolabadyA, NattelS, LipGYH, Ren J. Inflammasome signaling in atrial fibrillation: JACC state-of-the-art review. J Am Coll Cardiol. 2022;79(23):2349-2366.

[132]

ZhaoW, WuT, ZhanJ, Dong Z. Identification of the immune status of hypertrophic cardiomyopathy by integrated analysis of bulk-and single-cell RNA sequencing data. Comput Math Methods Med. 2022;2022:7153491.

[133]

LiuL, YuY, HuLL, et al. Potential target genes in the development of atrial fibrillation: a comprehensive bioinformatics analysis. Med Sci Monit. 2021;27:e928366.

[134]

ChuY, YuF, WuY, et al. Identification of genes and key pathways underlying the pathophysiological association between nonalcoholic fatty liver disease and atrial fibrillation. BMC Med Genomics. 2022;15(1):150.

[135]

IharaK, SasanoT. Role of inflammation in the pathogenesis of atrial fibrillation. Front Physiol. 2022;13:862164.

[136]

MüllerI, VoglT, KühlU, et al. Serum alarmin S100A8/S100A9 levels and its potential role as biomarker in myocarditis. ESC Heart Fail. 2020;7(4):1442-1451.

[137]

MüllerI, JansonL, SauterM, et al. Myeloid-derived suppressor cells restrain natural killer cell activity in acute coxsackievirus B3-induced myocarditis. Viruses. 2021;13(5):889.

[138]

XiaoSJ, ZhouYF, JiaH, WuQ, PanDF. Identification of the pivotal differentially expressed genes and pathways involved in Staphylococcus aureus-induced infective endocarditis by using bioinformatics analysis. Eur Rev Med Pharmacol Sci. 2021;25(1):487-497. doi:10.26355/eurrev_202101_24420

[139]

SchiopuA, Marinkovic G, De CampL, et al. Short-term blockade of the S100A8/A9 alarmin in the immediate post-myocardial infarction period inhibits acute myocardial inflammation and preserves myocardial repair. Eur Heart J. 2017;38(suppl_1):ehx504.P4026.

[140]

HesselstrandR, Distler JHW, RiemekastenG, et al. An open-label study to evaluate biomarkers and safety in systemic sclerosis patients treated with paquinimod. Arthritis Res Ther. 2021;23(1):204.

[141]

BrückW, Pförtner R, PhamT, et al. Reduced astrocytic NF-κB activation by laquinimod protects from cuprizone-induced demyelination. Acta Neuropathol. 2012;124(3):411-424.

[142]

Schulze-TopphoffU, Shetty A, Varrin-DoyerM, et al. Laquinimod, a quinoline-3-carboxamide, induces type II myeloid cells that modulate central nervous system autoimmunity. PloS One. 2012;7(3):e33797.

[143]

ComiG, DadonY, SassonN, et al. CONCERTO: a randomized, placebo-controlled trial of oral laquinimod in relapsing-remitting multiple sclerosis. Mult Scler. 2022;28(4):608-619.

[144]

D’HaensG, Sandborn WJ, ColombelJF, et al. A phase II study of laquinimod in Crohn’s disease. Gut. 2015;64(8):1227-1235.

[145]

DuY, CaiY, LvY, et al. Single-cell RNA sequencing unveils the communications between malignant T and myeloid cells contributing to tumor growth and immunosuppression in cutaneous T-cell lymphoma. Cancer Lett. 2022;551:215972.

[146]

ArmstrongAJ, AnandA, EdenbrandtL, et al. Phase 3 assessment of the automated bone scan index as a prognostic imaging biomarker of overall survival in men with metastatic castration-resistant prostate cancer: a secondary analysis of a randomized clinical trial. JAMA Oncol. 2018;4(7):944-951.

[147]

EscudierB, FaivreS, Van CutsemE, et al. A phase II multicentre, open-label, proof-of-concept study of Tasquinimod in hepatocellular, ovarian, renal cell, and gastric cancers. Target Oncol. 2017;12(5):655-661.

[148]

SunY, WangZ, WangC, Tang Z, ZhaoH. Psycho-cardiology therapeutic effects of Shuangxinfang in rats with depression-behavior post acute myocardial infarction: focus on protein S100A9 from proteomics. Biomed Pharmacother. 2021;144:112303.

[149]

ZhouL, MiaoK, YinB, et al. Cardioprotective role of myeloid-derived suppressor cells in heart failure. Circulation. 2018;138(2):181-197.

[150]

FengL, LiG, AnJ, et al. Exercise training protects against heart failure via expansion of myeloid-derived suppressor cells through regulating IL-10/STAT3/S100A9 pathway. Circ Heart Fail. 2022;15(3):e008550.

[151]

ParkJ, GaoH, WangY, Hu H, SimonDI, SteinmetzNF. S100A9-targeted tobacco mosaic virus nanoparticles exhibit high specificity toward atherosclerotic lesions in ApoE-/-mice. J Mater Chem B. 2019;7(11):1842-1846.

[152]

KawanoT, Shimamura M, NakagamiH, et al. Therapeutic vaccine against S100A9 (S100 calcium-binding protein A9) inhibits thrombosis without increasing the risk of bleeding in ischemic stroke in mice. Hypertension. 2018;72(6):1355-1364.

[153]

ShimamuraM, Kaikita K, NakagamiH, et al. Development of anti-thrombotic vaccine against human S100A9 in rhesus monkey. Sci Rep. 2021;11(1):11472.

[154]

Ortega-RiveraOA, ShinMD, Moreno-GonzalezMA, PokorskiJK, Steinmetz NF. A single-dose Qβ VLP vaccine against S100A9 protein reduces atherosclerosis in a preclinical model. Adv Ther. 2022;5(10):2200092.

[155]

GaoY, ChenX, TianT, et al. A lysosome-activated tetrahedral nanobox for encapsulated siRNA delivery. Adv Mater. 2022;34(46):e2201731.

[156]

TianT, LiY, LinY. Prospects and challenges of dynamic DNA nanostructures in biomedical applications. Bone Res. 2022;10(1):40.

RIGHTS & PERMISSIONS

2024 The Authors. Cell Proliferation published by Beijing Institute for Stem Cell and Regenerative Medicine and John Wiley & Sons Ltd.

AI Summary AI Mindmap
PDF

128

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/