Information

  • Author Services

Initiatives

You are accessing a machine-readable page. In order to be human-readable, please install an RSS reader.

All articles published by MDPI are made immediately available worldwide under an open access license. No special permission is required to reuse all or part of the article published by MDPI, including figures and tables. For articles published under an open access Creative Common CC BY license, any part of the article may be reused without permission provided that the original article is clearly cited. For more information, please refer to https://www.mdpi.com/openaccess .

Feature papers represent the most advanced research with significant potential for high impact in the field. A Feature Paper should be a substantial original Article that involves several techniques or approaches, provides an outlook for future research directions and describes possible research applications.

Feature papers are submitted upon individual invitation or recommendation by the scientific editors and must receive positive feedback from the reviewers.

Editor’s Choice articles are based on recommendations by the scientific editors of MDPI journals from around the world. Editors select a small number of articles recently published in the journal that they believe will be particularly interesting to readers, or important in the respective research area. The aim is to provide a snapshot of some of the most exciting work published in the various research areas of the journal.

Original Submission Date Received: .

  • Active Journals
  • Find a Journal
  • Proceedings Series
  • For Authors
  • For Reviewers
  • For Editors
  • For Librarians
  • For Publishers
  • For Societies
  • For Conference Organizers
  • Open Access Policy
  • Institutional Open Access Program
  • Special Issues Guidelines
  • Editorial Process
  • Research and Publication Ethics
  • Article Processing Charges
  • Testimonials
  • Preprints.org
  • SciProfiles
  • Encyclopedia

ijms-logo

Article Menu

atherosclerosis case study pdf

  • Subscribe SciFeed
  • Recommended Articles
  • PubMed/Medline
  • Google Scholar
  • on Google Scholar
  • Table of Contents

Find support for a specific problem in the support section of our website.

Please let us know what you think of our products and services.

Visit our dedicated information section to learn more about MDPI.

JSmol Viewer

Pathophysiology of atherosclerosis.

atherosclerosis case study pdf

1. Introduction

2. endothelium, 3. atherosclerosis initiation and fatty streak formation, 3.1. endothelial dysfunction in atherosclerosis development, 3.1.1. hemodynamic forces and endothelial dysfunction, 3.1.2. the role of nitric oxide in endothelial dysfunction, 3.2. ldl infiltration, ldl modifications in the intima, 3.3. endothelial activation, 3.4. monocyte recruitment and foam cell formation, 3.5. contribution of vsmcs to foam cell population, 4. fibrous plaque development, 4.1. fibrous cap, 4.2. necrotic core, 4.3. plaque calcification, 5. plaque stability and rupture, 5.1. vulnerable plaque, 5.2. plaque rupture and thrombus formation, 5.3. clinical complications, 6. inflammation in atherosclerosis, 7. post-transcriptional regulation of atherosclerotic plaques, 7.1. mirnas, 7.1.1. mirnas in atherosclerotic plaque initiation and progression, 7.1.2. mirnas and atherosclerotic plaque rupture, 7.1.3. distinct mirna regulation of vsmc and ec function, 7.2. lncrnas, 8. microbiota, 9. sex as an important risk factor in atherosclerosis, 9.1. impact of plaque size and morphology between sexes, 9.2. clinical implications, 10. cigarette-smoking-induced atherosclerosis, molecular mechanisms underlying clinical smoking-induced atherosclerosis, 11. conclusions, author contributions, institutional review board statement, informed consent statement, conflicts of interest.

  • Reitsma, S.; Slaaf, D.W.; Vink, H.; van Zandvoort, M.A.M.J.; Oude Egbrink, M.G. The endothelial glycocalyx: Composition, functions, and visualization. Pflug. Arch. Eur. J. Physiol. 2007 , 454 , 345–359. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Rhodin, J.A. Ultrastructure of mammalian venous capillaries, venules, and small collecting veins. J. Ultrastruct. Res. 1968 , 25 , 452–500. [ Google Scholar ] [ CrossRef ]
  • Zhang, B.; Gu, J.; Qian, M.; Niu, L.; Zhou, H.; Ghista, D. Correlation between quantitative analysis of wall shear stress and intima-media thickness in atherosclerosis development in carotid arteries. Biomed. Eng. Online 2017 , 16 , 137. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Roger, V.L.; Weston, S.A.; Killian, J.M.; Pfeifer, E.A.; Belau, P.G.; Kottke, T.E.; Frye, R.L.; Bailey, K.R.; Jacobsen, S.J. Time trends in the prevalence of atherosclerosis: A population-based autopsy study. Am. J. Med. 2001 , 110 , 267–273. [ Google Scholar ] [ CrossRef ]
  • Targonski, P. Referral to Autopsy: Effect of Antemortem Cardiovascular Disease A Population-Based Study in Olmsted County, Minnesota. Ann. Epidemiol. 2001 , 11 , 264–270. [ Google Scholar ] [ CrossRef ]
  • Böyum, A. Isolation of mononuclear cells and granulocytes from human blood. Isolation of monuclear cells by one centrifugation, and of granulocytes by combining centrifugation and sedimentation at 1 g. Scand. J. Clin. Lab. Investig. Suppl. 1968 , 97 , 77–89. [ Google Scholar ]
  • Hays, A.G.; Kelle, S.; Hirsch, G.A.; Soleimanifard, S.; Yu, J.; Agarwal, H.K.; Gerstenblith, G.; Schär, M.; Stuber, M.; Weiss, R.G. Regional Coronary Endothelial Function Is Closely Related to Local Early Coronary Atherosclerosis in Patients With Mild Coronary Artery Disease. Circ. Cardiovasc. Imaging 2012 , 5 , 341–348. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Senior, R.M.; Campbell, E.J.; Landis, J.A.; Cox, F.R.; Kuhn, C.; Koren, H.S. Elastase of U-937 Monocytelike Cells. J. Clin. Investig. 1982 , 69 , 384–393. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Favero, G.; Paganelli, C.; Buffoli, B.; Rodella, L.F.; Rezzani, R. Endothelium and Its Alterations in Cardiovascular Diseases: Life Style Intervention. BioMed Res. Int. 2014 , 2014 , 801896. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Campinho, P.; Vilfan, A.; Vermot, J. Blood Flow Forces in Shaping the Vascular System: A Focus on Endothelial Cell Behavior. Front. Physiol. 2020 , 11 , 552. [ Google Scholar ] [ CrossRef ]
  • Davies, P.F. Flow-mediated endothelial mechanotransduction. Physiol. Rev. 1995 , 75 , 519–560. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Esper, R.J.; Nordaby, R.A.; Vilariño, J.O.; Paragano, A.; Cacharrón, J.L.; Machado, R.A. Endothelial Dysfunction: A Comprehensive Appraisal. Cardiovasc. Diabetol. 2006 , 18 , 1–18. [ Google Scholar ]
  • Gimbrone, M.A., Jr.; García-Cardeña, G. Vascular Endothelium, Hemodynamics, and the Pathobiology of Atherosclerosis. Cardiovasc. Pathol. 2013 , 22 , 9–15. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Natural history of aortic and coronary atherosclerotic lesions in youth. Findings from the PDAY Study. Pathobiological Determinants of Atherosclerosis in Youth (PDAY) Research Group. Arter. Thromb. A J. Vasc. Biol. 1993 , 13 , 1291–1298. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Rubanyi, G.M. The Role of Endothelium in Cardiovascular Homeostasis and Diseases. J. Cardiovasc. Pharmacol. 1993 , 22 , S1–S14. [ Google Scholar ] [ CrossRef ]
  • Vanhoutte, P.M. How to assess endothelial function in human blood vessels. J. Hypertens. 1999 , 17 , 1047–1058. [ Google Scholar ] [ CrossRef ]
  • Hermida, N.; Balligand, J.-L. Low-Density Lipoprotein-Cholesterol-Induced Endothelial Dysfunction and Oxidative Stress: The Role of Statins. Antioxid. Redox Signal. 2014 , 20 , 1216–1237. [ Google Scholar ] [ CrossRef ]
  • Mundi, S.; Massaro, M.; Scoditti, E.; Carluccio, M.A.; van Hinsbergh, V.W.M.; Iruela-Arispe, M.L.; De Caterina, R. Endothelial permeability, LDL deposition, and cardiovascular risk factors—A review. Cardiovasc. Res. 2018 , 114 , 35–52. [ Google Scholar ] [ CrossRef ]
  • Yu, X.-H.; Fu, Y.-C.; Zhang, D.-W.; Yin, K.; Tang, C.-K. Foam cells in atherosclerosis. Clin. Chim. Acta 2013 , 424 , 245–252. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Allahverdian, S.; Chehroudi, A.C.; McManus, B.M.; Abraham, T.; Francis, G.A. Contribution of Intimal Smooth Muscle Cells to Cholesterol Accumulation and Macrophage-Like Cells in Human Atherosclerosis. Circulation 2014 , 129 , 1551–1559. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Stary, H.C.; Chandler, A.B.; Glagov, S.; Guyton, J.R.; Insull, W.; Rosenfeld, M.E.; Schaffer, S.A.; Schwartz, C.J.; Wagner, W.D.; Wissler, R.W. A definition of initial, fatty streak, and intermediate lesions of atherosclerosis. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation 1994 , 89 , 2462–2478. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Sun, H.-J.; Wu, Z.-Y.; Nie, X.-W.; Bian, J.-S. Role of Endothelial Dysfunction in Cardiovascular Diseases: The Link between Inflammation and Hydrogen Sulfide. Front. Pharmacol. 2020 , 10 , 1568. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Verma, S.; Anderson, T.J. Fundamentals of Endothelial Function for the Clinical Cardiologist. Circulation 2002 , 105 , 546–549. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Verma, S.; Buchanan, M.R.; Anderson, T.J. Endothelial Function Testing as a Biomarker of Vascular Disease. Circulation 2003 , 108 , 2054–2059. [ Google Scholar ] [ CrossRef ]
  • Ardestani, S.B.; Eftedal, I.; Pedersen, M.; Jeppesen, P.B.; Nørregaard, R.; Matchkov, V.V. Endothelial dysfunction in small arteries and early signs of atherosclerosis in ApoE knockout rats. Sci. Rep. 2020 , 10 , 15296. [ Google Scholar ] [ CrossRef ]
  • Mudau, M.; Genis, A.; Lochner, A.; Strijdom, H. Endothelial dysfunction: The early predictor of atherosclerosis. Cardiovasc. J. Afr. 2012 , 23 , 222–231. [ Google Scholar ] [ CrossRef ]
  • Gordon, E.; Schimmel, L.; Frye, M. The Importance of Mechanical Forces for in vitro Endothelial Cell Biology. Front. Physiol. 2020 , 11 , 684. [ Google Scholar ] [ CrossRef ]
  • Gimbrone, M.A., Jr.; García-Cardeña, G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circ. Res. 2016 , 118 , 620–636. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Chiu, J.-J.; Chien, S. Effects of Disturbed Flow on Vascular Endothelium: Pathophysiological Basis and Clinical Perspectives. Physiol. Rev. 2011 , 91 , 327–387. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Ross, R.; Glomset, J.A. The Pathogenesis of Atherosclerosis. N. Engl. J. Med. 1976 , 295 , 369–377. [ Google Scholar ] [ CrossRef ]
  • Chistiakov, D.A.; Orekhov, A.N.; Bobryshev, Y.V. Effects of shear stress on endothelial cells: Go with the flow. Acta Physiol. 2017 , 219 , 382–408. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Kang, H.; Cancel, L.M.; Tarbell, J.M. Effect of shear stress on water and LDL transport through cultured endothelial cell monolayers. Atherosclerosis 2014 , 233 , 682–690. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Caro, C.G.; Fitz-Gerald, J.M.; Schroter, R.C. Atheroma and arterial wall shear—Observation, correlation and proposal of a shear dependent mass transfer mechanism for atherogenesis. Proc. R. Soc. London. Ser. B Boil. Sci. 1971 , 177 , 109–133. [ Google Scholar ] [ CrossRef ]
  • Zhou, J.; Li, Y.-S.; Chien, S. Shear Stress–Initiated Signaling and Its Regulation of Endothelial Function. Arterioscler. Thromb. Vasc. Biol. 2014 , 34 , 2191–2198. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Baeyens, N.; Bandyopadhyay, C.; Coon, B.G.; Yun, S.; Schwartz, M.A. Endothelial fluid shear stress sensing in vascular health and disease. J. Clin. Investig. 2016 , 126 , 821–828. [ Google Scholar ] [ CrossRef ]
  • Souilhol, C.; Serbanovic-Canic, J.; Fragiadaki, M.; Chico, T.J.; Ridger, V.; Roddie, H.; Evans, P.C. Endothelial responses to shear stress in atherosclerosis: A novel role for developmental genes. Nat. Rev. Cardiol. 2020 , 17 , 52–63. [ Google Scholar ] [ CrossRef ]
  • Chien, S.; Shyy, J.Y.J. Effects of hemodynamic forces on gene expression and signal transduction in endothelial cells. Biol. Bull. 1998 , 194 , 390–393. [ Google Scholar ] [ CrossRef ]
  • Gimbrone, M.A.; Topper, J.N.; Nagel, T.; Anderson, K.R.; Garcia-Cardeña, G. Endothelial Dysfunction, Hemodynamic Forces, and Atherogenesisa. Ann. N. Y. Acad. Sci. 2006 , 902 , 230–240. [ Google Scholar ] [ CrossRef ]
  • Resnick, N.; Yahav, H.; Shay-Salit, A.; Shushy, M.; Schubert, S.; Zilberman, L.C.M.; Wofovitz, E. Fluid shear stress and the vascular endothelium: For better and for worse. Prog. Biophys. Mol. Biol. 2003 , 81 , 177–199. [ Google Scholar ] [ CrossRef ]
  • Chiu, J.-J.; Usami, S.; Chien, S. Vascular endothelial responses to altered shear stress: Pathologic implications for atherosclerosis. Ann. Med. 2009 , 41 , 19–28. [ Google Scholar ] [ CrossRef ]
  • Chien, S. Role of shear stress direction in endothelial mechanotransduction. Mol. Cell. Biomech. 2008 , 5 , 1–8. [ Google Scholar ] [ PubMed ]
  • García-Cardeña, G.; Gimbrone, M.A. Biomechanical Modulation of Endothelial Phenotype: Implications for Health and Disease. In The Vascular Endothelium II ; Moncada, S., Higgs, A., Eds.; Handbook of Experimental Pharmacology; Springer: Berlin/Heidelberg, Germany, 2006; Volume 176/II, pp. 79–95. ISBN 978-3-540-36027-8. [ Google Scholar ]
  • Shyy, J.Y.; Lin, M.C.; Han, J.; Lu, Y.; Petrime, M.; Chien, S. The cis-acting phorbol ester “12-O-tetradecanoylphorbol 13-acetate”-responsive element is involved in shear stress-induced monocyte chemotactic protein 1 gene expression. Proc. Natl. Acad. Sci. USA 1995 , 92 , 8069–8073. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Hsiai, T.K.; Cho, S.K.; Reddy, S.; Hama, S.; Navab, M.; Demer, L.; Honda, H.M.; Ho, C.-M. Pulsatile Flow Regulates Monocyte Adhesion to Oxidized Lipid-Induced Endothelial Cells. Arterioscler. Thromb. Vasc. Biol. 2001 , 21 , 1770–1776. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Hsiai, T.K.; Cho, S.K.; Wong, P.K.; Ing, M.; Salazar, A.; Sevanian, A.; Navab, M.; Demer, L.; Ho, C.-M. Monocyte recruitment to endothelial cells in response to oscillatory shear stress. FASEB J. 2003 , 17 , 1648–1657. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kraiss, L.W.; Geary, R.L.; Mattsson, E.J.; Vergel, S.; Au, Y.T.; Clowes, A.W. Acute Reductions in Blood Flow and Shear Stress Induce Platelet-Derived Growth Factor-A Expression in Baboon Prosthetic Grafts. Circ. Res. 1996 , 79 , 45–53. [ Google Scholar ] [ CrossRef ]
  • Malek, A.M.; Gibbons, G.H.; Dzau, V.J.; Izumo, S. Fluid shear stress differentially modulates expression of genes encoding basic fibroblast growth factor and platelet-derived growth factor B chain in vascular endothelium. J. Clin. Investig. 1993 , 92 , 2013–2021. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Wilcox, J.N.; Smith, K.M.; Williams, L.T.; Schwartz, S.M.; Gordon, D. Platelet-derived growth factor mRNA detection in human atherosclerotic plaques by in situ hybridization. J. Clin. Investig. 1988 , 82 , 1134–1143. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Silberman, M.; Barac, Y.D.; Yahav, H.; Wolfovitz, E.; Einav, S.; Resnick, N.; Binah, O. Shear stress-induced transcriptional regulation via hybrid promoters as a potential tool for promoting angiogenesis. Angiogenesis 2009 , 12 , 231–242. [ Google Scholar ] [ CrossRef ]
  • Davis, M.E.; Grumbach, I.; Fukai, T.; Cutchins, A.; Harrison, D.G. Shear Stress Regulates Endothelial Nitric-oxide Synthase Promoter Activity through Nuclear Factor κB Binding. J. Biol. Chem. 2004 , 279 , 163–168. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Drummond, G.; Cai, H.; Davis, M.E.; Ramasamy, S.; Harrison, D.G. Transcriptional and Posttranscriptional Regulation of Endothelial Nitric Oxide Synthase Expression by Hydrogen Peroxide. Circ. Res. 2000 , 86 , 347–354. [ Google Scholar ] [ CrossRef ]
  • Almendro, N.; Bellón, T.; Rius, C.; Lastres, P.; Langa, C.; Corbí, A.; Bernabeu, C. Cloning of the human platelet endothelial cell adhesion molecule-1 promoter and its tissue-specific expression. Structural and functional characterization. J. Immunol. 1996 , 157 , 5411–5421. [ Google Scholar ] [ PubMed ]
  • Houston, P.; White, B.P.; Campbell, C.J.; Braddock, M. Delivery and Expression of Fluid Shear Stress-Inducible Promoters to the Vessel Wall: Applications for Cardiovascular Gene Therapy. Hum. Gene Ther. 1999 , 10 , 3031–3044. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Landmesser, U.; Hornig, B.; Drexler, H. Endothelial Dysfunction in Hypercholesterolemia: Mechanisms, Pathophysiological Importance, and Therapeutic Interventions. Semin. Thromb. Hemost. 2000 , 26 , 529–538. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Archer, S.L.; Huang, J.M.; Hampl, V.; Nelson, D.P.; Shultz, P.J.; Weir, E.K. Nitric oxide and cGMP cause vasorelaxation by activation of a charybdotoxin-sensitive K channel by cGMP-dependent protein kinase. Proc. Natl. Acad. Sci. USA 1994 , 91 , 7583–7587. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Aggio, A.; Grassi, D.; Onori, E.; D’Alessandro, A.; Masedu, F.; Valenti, M.; Ferri, C. Endothelium/nitric oxide mechanism mediates vasorelaxation and counteracts vasoconstriction induced by low concentration of flavanols. Eur. J. Nutr. 2012 , 52 , 263–272. [ Google Scholar ] [ CrossRef ]
  • Cooke, J.P.; Tsao, P.S. Is NO an endogenous antiatherogenic molecule? Arter. Thromb. A J. Vasc. Biol. 1994 , 14 , 653–655. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Libby, P. Current Concepts of the Pathogenesis of the Acute Coronary Syndromes. Circulation 2001 , 104 , 365–372. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Marx, N.; Sukhova, G.K.; Collins, T.; Libby, P.; Plutzky, J. PPARα Activators Inhibit Cytokine-Induced Vascular Cell Adhesion Molecule-1 Expression in Human Endothelial Cells. Circulation 1999 , 99 , 3125–3131. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Chen, J.-Y.; Ye, Z.-X.; Wang, X.-F.; Chang, J.; Yang, M.-W.; Zhong, H.; Hong, F.-F.; Yang, S.-L. Nitric oxide bioavailability dysfunction involves in atherosclerosis. Biomed. Pharmacother. 2018 , 97 , 423–428. [ Google Scholar ] [ CrossRef ]
  • Carlström, M. Nitric oxide signalling in kidney regulation and cardiometabolic health. Nat. Rev. Nephrol. 2021 , 17 , 575–590. [ Google Scholar ] [ CrossRef ]
  • Schulz, E.; Gori, T.; Münzel, T. Oxidative stress and endothelial dysfunction in hypertension. Hypertens. Res. 2011 , 34 , 665–673. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Förstermann, U.; Xia, N.; Li, H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circ. Res. 2017 , 120 , 713–735. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Casino, P.R.; Kilcoyne, C.M.; A Quyyumi, A.; Hoeg, J.M.; A Panza, J. The role of nitric oxide in endothelium-dependent vasodilation of hypercholesterolemic patients. Circulation 1993 , 88 , 2541–2547. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • A Panza, J.; Casino, P.R.; Kilcoyne, C.M.; A Quyyumi, A. Role of endothelium-derived nitric oxide in the abnormal endothelium-dependent vascular relaxation of patients with essential hypertension. Circulation 1993 , 87 , 1468–1474. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Alexander, R.W. Hypertension and the Pathogenesis of Atherosclerosis. Hypertension 1995 , 25 , 155–161. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Michel, C.C.; Curry, F.E. Microvascular Permeability. Physiol. Rev. 1999 , 79 , 703–761. [ Google Scholar ] [ CrossRef ]
  • Rippe, B.; Haraldsson, B. Transport of macromolecules across microvascular walls: The two-pore theory. Physiol. Rev. 1994 , 74 , 163–219. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Rippe, B.; Rosengren, B.-I.; Carlsson, O.; Venturoli, D. Transendothelial Transport: The Vesicle Controversy. J. Vasc. Res. 2002 , 39 , 375–390. [ Google Scholar ] [ CrossRef ]
  • Pappenheimer, J.R.; Renkin, E.M.; Borrero, L.M. Filtration, Diffusion and Molecular Sieving Through Peripheral Capillary Membranes. Am. J. Physiol. Content 1951 , 167 , 13–46. [ Google Scholar ] [ CrossRef ]
  • Jang, E.; Robert, J.; Rohrer, L.; von Eckardstein, A.; Lee, W.L. Transendothelial transport of lipoproteins. Atheroscler. 2020 , 315 , 111–125. [ Google Scholar ] [ CrossRef ]
  • Zhang, X.; Sessa, W.C.; Fernández-Hernando, C. Endothelial Transcytosis of Lipoproteins in Atherosclerosis. Front. Cardiovasc. Med. 2018 , 5 , 130. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Goldstein, J.L.; Brown, M.S. The LDL Receptor. Arterioscler. Thromb. Vasc. Biol. 2009 , 29 , 431–438. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Babitt, J.; Trigatti, B.; Rigotti, A.; Smart, E.J.; Anderson, R.G.W.; Xu, S.; Krieger, M. Murine SR-BI, a High Density Lipoprotein Receptor That Mediates Selective Lipid Uptake, Is N-Glycosylated and Fatty Acylated and Colocalizes with Plasma Membrane Caveolae. J. Biol. Chem. 1997 , 272 , 13242–13249. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Santibanez, J.F.; Blanco, F.J.; Garrido-Martin, E.M.; Sanz-Rodriguez, F.; del Pozo, M.A.; Bernabeu, C. Caveolin-1 interacts and cooperates with the transforming growth factor-β type I receptor ALK1 in endothelial caveolae. Cardiovasc. Res. 2008 , 77 , 791–799. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Rothberg, K.G.; Heuser, J.E.; Donzell, W.C.; Ying, Y.-S.; Glenney, J.R.; Anderson, R.G. Caveolin, a protein component of caveolae membrane coats. Cell 1992 , 68 , 673–682. [ Google Scholar ] [ CrossRef ]
  • Fernández-Hernando, C.; Yu, J.; Suárez, Y.; Rahner, C.; Dávalos, A.; Lasunción, M.A.; Sessa, W.C. Genetic Evidence Supporting a Critical Role of Endothelial Caveolin-1 during the Progression of Atherosclerosis. Cell Metab. 2009 , 10 , 48–54. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Frank, P.G.; Pavlides, S.; Lisanti, M.P. Caveolae and transcytosis in endothelial cells: Role in atherosclerosis. Cell Tissue Res. 2009 , 335 , 41–47. [ Google Scholar ] [ CrossRef ]
  • Wang, D.-X.; Pan, Y.-Q.; Liu, B.; Dai, L. Cav-1 promotes atherosclerosis by activating JNK-associated signaling. Biochem. Biophys. Res. Commun. 2018 , 503 , 513–520. [ Google Scholar ] [ CrossRef ]
  • Ramírez, C.M.; Zhang, X.; Bandyopadhyay, C.; Rotllan, N.; Sugiyama, M.G.; Aryal, B.; Liu, X.; He, S.; Kraehling, J.R.; Ulrich, V.; et al. Caveolin-1 Regulates Atherogenesis by Attenuating Low-Density Lipoprotein Transcytosis and Vascular Inflammation Independently of Endothelial Nitric Oxide Synthase Activation. Circulation 2019 , 140 , 225–239. [ Google Scholar ] [ CrossRef ]
  • Zhang, X.; Fernández-Hernando, C. Transport of LDLs into the arterial wall: Impact in atherosclerosis. Curr. Opin. Lipidol. 2020 , 31 , 279–285. [ Google Scholar ] [ CrossRef ]
  • Yang, Y.; Schmidt, E.P. The endothelial glycocalyx. Tissue Barriers 2013 , 1 , e23494. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Ziegler, T.; Abdel Rahman, F.; Jurisch, V.; Kupatt, C. Atherosclerosis and the Capillary Network; Pathophysiology and Potential Therapeutic Strategies. Cells 2019 , 9 , 50. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Skålén, K.; Gustafsson, M.; Rydberg, E.K.; Hultén, L.M.; Wiklund, O.; Innerarity, T.L.; Borén, J. Subendothelial retention of atherogenic lipoproteins in early atherosclerosis. Nature 2002 , 417 , 750–754. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Khalil, M.F.; Wagner, W.D.; Goldberg, I.J. Molecular Interactions Leading to Lipoprotein Retention and the Initiation of Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2004 , 24 , 2211–2218. [ Google Scholar ] [ CrossRef ]
  • Carmena, R.; Ascaso, J.F.; Camejo, G.; Varela, G.; Hurt-Camejo, E.; Ordovas, J.; Martinez-Valls, J.; Bergstöm, M.; Wallin, B. Effect of olive and sunflower oils on low density lipoprotein level, composition, size, oxidation and interaction with arterial proteoglycans. Atherosclerosis 1996 , 125 , 243–255. [ Google Scholar ] [ CrossRef ]
  • Levitan, I.; Volkov, S.; Subbaiah, P.V. Oxidized LDL: Diversity, Patterns of Recognition, and Pathophysiology. Antioxid. Redox Signal. 2010 , 13 , 39–75. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Sheedy, F.J.; Grebe, A.; Rayner, K.J.; Kalantari, P.; Ramkhelawon, B.; Carpenter, S.B.; E Becker, C.; Ediriweera, H.N.; E Mullick, A.; Golenbock, D.T.; et al. CD36 coordinates NLRP3 inflammasome activation by facilitating intracellular nucleation of soluble ligands into particulate ligands in sterile inflammation. Nat. Immunol. 2013 , 14 , 812–820. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Witztum, J.L.; Steinberg, D. The Oxidative Modification Hypothesis of Atherosclerosis Does It Hold for Humans? Trends Cardiovasc. Med. 2001 , 11 , 93–102. [ Google Scholar ] [ CrossRef ]
  • Steinbrecher, U.P.; Zhang, H.; Lougheed, M. Role of oxidatively modified LDL in atherosclerosis. Free Radic. Biol. Med. 1990 , 9 , 155–168. [ Google Scholar ] [ CrossRef ]
  • Carr, A.C.; McCall, M.R.; Frei, B. Oxidation of LDL by Myeloperoxidase and Reactive Nitrogen Species. Arterioscler. Thromb. Vasc. Biol. 2000 , 20 , 1716–1723. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Pitanga, T.N.; França, L.D.A.; Rocha, V.C.J.; Meirelles, T.; Borges, V.M.; Gonçalves, M.S.; Pontes-De-Carvalho, L.C.; Noronha-Dutra, A.A.; Dos-Santos, W.L.C. Neutrophil-derived microparticles induce myeloperoxidase-mediated damage of vascular endothelial cells. BMC Cell Biol. 2014 , 15 , 21. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Nakajima, H.; Ikeda, M.; Hirata, Y.; Matsumoto, M.; Seike, M.; Kodama, H. Low-density lipoprotein is oxidized by phospholipase A 2 and lipoxygenase in xanthoma lesions. Eur. J. Lipid Sci. Technol. 2007 , 109 , 1053–1059. [ Google Scholar ] [ CrossRef ]
  • Takahashi, Y.; Zhu, H.; Yoshimoto, T. Essential Roles of Lipoxygenases in LDL Oxidation and Development of Atherosclerosis. Antioxid. Redox Signal. 2005 , 7 , 425–431. [ Google Scholar ] [ CrossRef ]
  • Parthasarathy, S.; Wieland, E.; Steinberg, D. A role for endothelial cell lipoxygenase in the oxidative modification of low density lipoprotein. Proc. Natl. Acad. Sci. USA 1989 , 86 , 1046–1050. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Esterbauer, H.; Jürgens, G.; Quehenberger, O.; Koller, E. Autoxidation of human low density lipoprotein: Loss of polyunsaturated fatty acids and vitamin E and generation of aldehydes. J. Lipid Res. 1987 , 28 , 495–509. [ Google Scholar ] [ CrossRef ]
  • Jialal, I.; Norkus, E.P.; Cristol, L.; Grundy, S.M. β-Carotene inhibits the oxidative modification of low-density lipoprotein. Biochim. Et Biophys. Acta (BBA)-Lipids Lipid Metab. 1991 , 1086 , 134–138. [ Google Scholar ] [ CrossRef ]
  • Esterbauer, H.; Waeg, G.; Puhl, H.; Dieber-Rotheneder, M.; Tatzber, F. Inhibition of LDL oxidation by antioxidants. Free Radic. Aging 1992 , 62 , 145–157. [ Google Scholar ] [ CrossRef ]
  • Heinecke, J.W. Mechanisms of oxidative damage of low density lipoprotein in human atherosclerosis. Curr. Opin. Lipidol. 1997 , 8 , 268–274. [ Google Scholar ] [ CrossRef ]
  • Parthasarathy, S.; Raghavamenon, A.; Garelnabi, M.O.; Santanam, N. Oxidized Low-Density Lipoprotein. In Free Radicals and Antioxidant Protocols ; Uppu, R.M., Murthy, S.N., Pryor, W.A., Parinandi, N.L., Eds.; Methods in Molecular Biology; Humana Press: Totowa, NJ, USA, 2010; Volume 610, pp. 403–417. ISBN 978-1-58829-710-5. [ Google Scholar ]
  • Berliner, J.A.; Navab, M.; Fogelman, A.M.; Frank, J.S.; Demer, L.; Edwards, P.A.; Watson, A.D.; Lusis, A.J. Atherosclerosis: Basic Mechanisms. Circulation 1995 , 91 , 2488–2496. [ Google Scholar ] [ CrossRef ]
  • Park, J.S.; Bae, S.H. Interplay between Saturated Free Fatty Acids and mmLDL Induces Inflammation in LPS-stimulated Macrophages. Korean Circ. J. 2021 , 51 , 81–82. [ Google Scholar ] [ CrossRef ]
  • Ann, S.-J.; Kim, K.-K.; Cheon, E.J.; Noh, H.-M.; Hwang, I.; Yu, J.-W.; Park, S.; Kang, S.-M.; Manabe, I.; Miller, Y.I.; et al. Palmitate and minimally-modified low-density lipoprotein cooperatively promote inflammatory responses in macrophages. PLoS ONE 2018 , 13 , e0193649. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Kraaijenhof, J.M.; Hovingh, G.K.; Stroes, E.S.; Kroon, J. The iterative lipid impact on inflammation in atherosclerosis. Curr. Opin. Lipidol. 2021 , 32 , 286–292. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Chistiakov, D.A.; Bobryshev, Y.V.; Orekhov, A.N. Macrophage-mediated cholesterol handling in atherosclerosis. J. Cell. Mol. Med. 2016 , 20 , 17–28. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Moore, K.J.; Freeman, M.W. Scavenger Receptors in Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2006 , 26 , 1702–1711. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Younis, N.; Sharma, R.; Soran, H.; Charlton-Menys, V.; Elseweidy, M.; Durrington, P.N. Glycation as an atherogenic modification of LDL. Curr. Opin. Lipidol. 2008 , 19 , 378–384. [ Google Scholar ] [ CrossRef ]
  • Greaves, D.R.; Gordon, S. The macrophage scavenger receptor at 30 years of age: Current knowledge and future challenges. J. Lipid Res. 2009 , 50 , S282–S286. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Öörni, K.; Pentikäinen, M.O.; Annila, A.; Kovanen, P.T. Oxidation of Low Density Lipoprotein Particles Decreases Their Ability to Bind to Human Aortic Proteoglycans. J. Biol. Chem. 1997 , 272 , 21303–21311. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Ling, W.; Lougheed, M.; Suzuki, H.; Buchan, A.; Kodama, T.; Steinbrecher, U.P. Oxidized or acetylated low density lipoproteins are rapidly cleared by the liver in mice with disruption of the scavenger receptor class A type I/II gene. J. Clin. Investig. 1997 , 100 , 244–252. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Lu, M.; Gursky, O. Aggregation and fusion of low-density lipoproteins in vivo and in vitro. Biomol. Concepts 2013 , 4 , 501–518. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Libby, P. The changing landscape of atherosclerosis. Nature 2021 , 592 , 524–533. [ Google Scholar ] [ CrossRef ]
  • van den Boogert, M.A.; Larsen, L.E.; Ali, L.; Kuil, S.D.; Chong, P.L.; Loregger, A.; Kroon, J.; Schnitzler, J.G.; Schimmel, A.W.; Peter, J.; et al. N-Glycosylation Defects in Humans Lower Low-Density Lipoprotein Cholesterol Through Increased Low-Density Lipoprotein Receptor Expression. Circulation 2019 , 140 , 280–292. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Liao, J.K. Linking endothelial dysfunction with endothelial cell activation. J. Clin. Investig. 2013 , 123 , 540–541. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Pober, J.S.; Sessa, W.C. Evolving functions of endothelial cells in inflammation. Nat. Rev. Immunol. 2007 , 7 , 803–815. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Yu, X.-H.; Zheng, X.-L.; Tang, C.-K. Nuclear Factor-ΚB Activation as a Pathological Mechanism of Lipid Metabolism and Atherosclerosis. In Advances in Clinical Chemistry ; Elsevier: Amsterdam, The Netherlands, 2015; Volume 70, pp. 1–30. ISBN 978-0-12-803316-6. [ Google Scholar ]
  • Liu, T.; Zhang, L.; Joo, D.; Sun, S.-C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017 , 2 , 17023. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Ropraz, P.; Imhof, B.A.; Matthes, T.; Wehrle-Haller, B.; Sidibé, A. Simultaneous Study of the Recruitment of Monocyte Subpopulations Under Flow In Vitro. J. Vis. Exp. 2018 , 58509 , e58509. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Gerrity, R.G. The role of the monocyte in atherogenesis: I. Transition of blood-borne monocytes into foam cells in fatty lesions. Am. J. Pathol. 1981 , 103 , 181–190. [ Google Scholar ]
  • Gerhardt, T.; Ley, K. Monocyte trafficking across the vessel wall. Cardiovasc. Res. 2015 , 107 , 321–330. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Teh, Y.C.; Ding, J.L.; Ng, L.G.; Chong, S.Z. Capturing the Fantastic Voyage of Monocytes through Time and Space. Front. Immunol. 2019 , 10 , 834. [ Google Scholar ] [ CrossRef ]
  • Ramos, C.L.; Huo, Y.; Jung, U.; Ghosh, S.; Manka, D.R.; Sarembock, I.J.; Ley, K. Direct Demonstration of P-Selectin–and VCAM-1–Dependent Mononuclear Cell Rolling in Early Atherosclerotic Lesions of Apolipoprotein E–Deficient Mice. Circ. Res. 1999 , 84 , 1237–1244. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Ley, K.; Laudanna, C.; Cybulsky, M.I.; Nourshargh, S. Getting to the site of inflammation: The leukocyte adhesion cascade updated. Nat. Rev. Immunol. 2007 , 7 , 678–689. [ Google Scholar ] [ CrossRef ]
  • Galkina, E.; Ley, K. Immune and Inflammatory Mechanisms of Atherosclerosis. Annu. Rev. Immunol. 2009 , 27 , 165–197. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Campbell, J.; Qin, S.; Bacon, K.B.; Mackay, C.; Butcher, E.C. Biology of chemokine and classical chemoattractant receptors: Differential requirements for adhesion-triggering versus chemotactic responses in lymphoid cells. J. Cell Biol. 1996 , 134 , 255–266. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Campbell, J.J.; Hedrick, J.; Zlotnik, A.; Siani, M.A.; Thompson, D.A.; Butcher, E.C. Chemokines and the Arrest of Lymphocytes Rolling Under Flow Conditions. Science 1998 , 279 , 381–384. [ Google Scholar ] [ CrossRef ]
  • Ley, K.; Miller, Y.I.; Hedrick, C.C. Monocyte and Macrophage Dynamics during Atherogenesis. Arterioscler. Thromb. Vasc. Biol. 2011 , 31 , 1506–1516. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Springer, T.A. Traffic signals for lymphocyte recirculation and leukocyte emigration: The multistep paradigm. Cell 1994 , 76 , 301–314. [ Google Scholar ] [ CrossRef ]
  • Sozzani, S.; Molino, M.; Locati, M.; Luini, W.; Cerletti, C.; Vecchi, A.; Mantovani, A. Receptor-activated calcium influx in human monocytes exposed to monocyte chemotactic protein-1 and related cytokines. J. Immunol. 1993 , 150 , 1544–1553. [ Google Scholar ] [ PubMed ]
  • Uguccioni, M.; D’Apuzzo, M.; Loetscher, M.; Dewald, B.; Baggiolini, M. Actions of the chemotactic cytokines MCP-1, MCP-2, MCP-3, RANTES, MIP-1α and MIP-1β on human monocytes. Eur. J. Immunol. 1995 , 25 , 64–68. [ Google Scholar ] [ CrossRef ]
  • Kakkar, V.; Lu, X. Impact of MCP -1 in Atherosclerosis. Curr. Pharm. Des. 2014 , 20 , 4580–4588. [ Google Scholar ] [ CrossRef ]
  • Nourshargh, S.; Alon, R. Leukocyte Migration into Inflamed Tissues. Immunity 2014 , 41 , 694–707. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Allport, J.R.; Muller, W.A.; Luscinskas, F.W. Monocytes Induce Reversible Focal Changes in Vascular Endothelial Cadherin Complex during Transendothelial Migration under Flow. J. Cell Biol. 2000 , 148 , 203–216. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Shaw, S.K.; Bamba, P.S.; Perkins, B.N.; Luscinskas, F.W. Real-Time Imaging of Vascular Endothelial-Cadherin during Leukocyte Transmigration Across Endothelium. J. Immunol. 2001 , 167 , 2323–2330. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Vestweber, D. Regulation of endothelial cell contacts during leukocyte extravasation. Curr. Opin. Cell Biol. 2002 , 14 , 587–593. [ Google Scholar ] [ CrossRef ]
  • Muller, W.A. Leukocyte–endothelial-cell interactions in leukocyte transmigration and the inflammatory response. Trends Immunol. 2003 , 24 , 327–334. [ Google Scholar ] [ CrossRef ]
  • Carman, C.V.; Springer, T.A. A transmigratory cup in leukocyte diapedesis both through individual vascular endothelial cells and between them. J. Cell Biol. 2004 , 167 , 377–388. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Carman, C.V.; Sage, P.T.; Sciuto, T.E.; de la Fuente, M.; Geha, R.S.; Ochs, H.D.; Dvorak, H.F.; Dvorak, A.M.; Springer, T.A. Transcellular Diapedesis Is Initiated by Invasive Podosomes. Immunity 2007 , 26 , 784–797. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Angelovich, T.A.; Hearps, A.C.; Maisa, A.; Kelesidis, T.; Jaworowski, A. Quantification of Monocyte Transmigration and Foam Cell Formation from Individuals with Chronic Inflammatory Conditions. J. Vis. Exp. 2017 , 128 , e56293. [ Google Scholar ] [ CrossRef ]
  • Hallmann, R.; Horn, N.; Selg, M.; Wendler, O.; Pausch, F.; Sorokin, L.M. Expression and Function of Laminins in the Embryonic and Mature Vasculature. Physiol. Rev. 2005 , 85 , 979–1000. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Italiani, P.; Boraschi, D. From Monocytes to M1/M2 Macrophages: Phenotypical vs. Functional Differentiation. Front. Immunol. 2014 , 5 , 514. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Shapouri-Moghaddam, A.; Mohammadian, S.; Vazini, H.; Taghadosi, M.; Esmaeili, S.-A.; Mardani, F.; Seifi, B.; Mohammadi, A.; Afshari, J.T.; Sahebkar, A. Macrophage plasticity, polarization, and function in health and disease. J. Cell. Physiol. 2018 , 233 , 6425–6440. [ Google Scholar ] [ CrossRef ]
  • Ferrante, C.J.; Pinhal-Enfield, G.; Elson, G.; Cronstein, B.N.; Hasko, G.; Outram, S.; Leibovich, S.J. The Adenosine-Dependent Angiogenic Switch of Macrophages to an M2-Like Phenotype is Independent of Interleukin-4 Receptor Alpha (IL-4Rα) Signaling. Inflammation 2013 , 36 , 921–931. [ Google Scholar ] [ CrossRef ]
  • De Paoli, F.; Staels, B.; Chinetti-Gbaguidi, G. Macrophage Phenotypes and Their Modulation in Atherosclerosis. Circ. J. 2014 , 78 , 1775–1781. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Chen, J.; Li, M.; Yang, C.; Yin, X.; Duan, K.; Wang, J.; Feng, B. Macrophage phenotype switch by sequential action of immunomodulatory cytokines from hydrogel layers on titania nanotubes. Colloids Surfaces B Biointerfaces 2018 , 163 , 336–345. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Peled, M.; Fisher, E.A. Dynamic Aspects of Macrophage Polarization during Atherosclerosis Progression and Regression. Front. Immunol. 2014 , 5 , 579. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Bobryshev, Y.V. Monocyte recruitment and foam cell formation in atherosclerosis. Micron 2006 , 37 , 208–222. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Wilfling, F.; Wang, H.; Haas, J.T.; Krahmer, N.; Gould, T.J.; Uchida, A.; Cheng, J.-X.; Graham, M.; Christiano, R.; Fröhlich, F.; et al. Triacylglycerol Synthesis Enzymes Mediate Lipid Droplet Growth by Relocalizing from the ER to Lipid Droplets. Dev. Cell 2013 , 24 , 384–399. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Ouimet, M.; Barrett, T.J.; Fisher, E.A. HDL and Reverse Cholesterol Transport. Circ. Res. 2019 , 124 , 1505–1518. [ Google Scholar ] [ CrossRef ]
  • Chinetti, G.; Baron, M.; Bouhlel, M.A.; Vanhoutte, J.; Copin, C.; Sebti, Y.; Derudas, B.; Mayi, T.; Bories, G.; Tailleux, A.; et al. Human Atherosclerotic Plaque Alternative Macrophages Display Low Cholesterol Handling but High Phagocytosis Because of Distinct Activities of the PPARγ and LXRα Pathways. Circ. Res. 2011 , 108 , 985–995. [ Google Scholar ] [ CrossRef ]
  • Maitra, U.; Parks, J.S.; Li, L. An Innate Immunity Signaling Process Suppresses Macrophage ABCA1 Expression through IRAK-1-Mediated Downregulation of Retinoic Acid Receptor α and NFATc2. Mol. Cell. Biol. 2009 , 29 , 5989–5997. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kim, T.W.; Febbraio, M.; Robinet, P.; Dugar, B.; Greene, D.; Cerny, A.; Latz, E.; Gilmour, R.; Staschke, K.; Chisolm, G.; et al. The Critical Role of IL-1 Receptor-Associated Kinase 4-Mediated NF-κB Activation in Modified Low-Density Lipoprotein-Induced Inflammatory Gene Expression and Atherosclerosis. J. Immunol. 2011 , 186 , 2871–2880. [ Google Scholar ] [ CrossRef ]
  • Rekhter, M.; Staschke, K.; Estridge, T.; Rutherford, P.; Jackson, N.; Gifford-Moore, D.; Foxworthy, P.; Reidy, C.; Huang, X.-D.; Kalbfleisch, M.; et al. Genetic ablation of IRAK4 kinase activity inhibits vascular lesion formation. Biochem. Biophys. Res. Commun. 2008 , 367 , 642–648. [ Google Scholar ] [ CrossRef ]
  • Takahashi, T.; Huang, Y.; Yamamoto, K.; Hamano, G.; Kakino, A.; Kang, F.; Imaizumi, Y.; Takeshita, H.; Nozato, Y.; Nozato, S.; et al. The endocytosis of oxidized LDL via the activation of the angiotensin II type 1 receptor. iScience 2021 , 24 , 102076. [ Google Scholar ] [ CrossRef ]
  • Nasiri, M.; Janoudi, A.; Vanderberg, A.; Frame, M.; Flegler, C.; Flegler, S.; Abela, G.S. Role of cholesterol crystals in atherosclerosis is unmasked by altering tissue preparation methods. Microsc. Res. Tech. 2015 , 78 , 969–974. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Levene, C.I. The early lesions of atheroma in the coronary arteries. J. Pathol. Bacteriol. 1956 , 72 , 79–82. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Katz, S.S.; Shipley, G.G.; Small, D.M. Physical chemistry of the lipids of human atherosclerotic lesions. Demonstration of a lesion intermediate between fatty streaks and advanced plaques. J. Clin. Investig. 1976 , 58 , 200–211. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Bogren, H.; Larsson, K. An X-ray-diffraction study of crystalline cholesterol in some pathological deposits in man. Biochim. Et Biophys. Acta 1963 , 75 , 65–69. [ Google Scholar ] [ CrossRef ]
  • Suhalim, J.L.; Chung, C.-Y.; Lilledahl, M.B.; Lim, R.S.; Levi, M.; Tromberg, B.J.; Potma, E.O. Characterization of Cholesterol Crystals in Atherosclerotic Plaques Using Stimulated Raman Scattering and Second-Harmonic Generation Microscopy. Biophys. J. 2012 , 102 , 1988–1995. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Baumer, Y.; Mehta, N.N.; Dey, A.K.; Powell-Wiley, T.M.; A Boisvert, W. Cholesterol crystals and atherosclerosis. Eur. Heart J. 2020 , 41 , 2236–2239. [ Google Scholar ] [ CrossRef ]
  • Abela, G.S. Cholesterol crystals piercing the arterial plaque and intima trigger local and systemic inflammation. J. Clin. Lipidol. 2010 , 4 , 156–164. [ Google Scholar ] [ CrossRef ]
  • Guo, H.; Callaway, J.B.; Ting, J.P.-Y. Inflammasomes: Mechanism of action, role in disease, and therapeutics. Nat. Med. 2015 , 21 , 677–687. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Hornung, V.; Bauernfeind, F.; Halle, A.; O Samstad, E.; Kono, H.; Rock, K.L.; Fitzgerald, K.; Latz, E. Silica crystals and aluminum salts activate the NALP3 inflammasome through phagosomal destabilization. Nat. Immunol. 2008 , 9 , 847–856. [ Google Scholar ] [ CrossRef ]
  • Zhou, R.; Yazdi, A.S.; Menu, P.; Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011 , 469 , 221–225, Erratum in Nature 2011 , 475 , 122. [ Google Scholar ] [ CrossRef ]
  • Mietus-Snyder, M.; Gowri, M.S.; Pitas, R.E. Class A Scavenger Receptor Up-regulation in Smooth Muscle Cells by Oxidized Low Density Lipoprotein. J. Biol. Chem. 2000 , 275 , 17661–17670. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Yan, P.; Xia, C.; Duan, C.; Li, S.; Mei, Z. Biological Characteristics of Foam Cell Formation in Smooth Muscle Cells Derived from Bone Marrow Stem Cells. Int. J. Biol. Sci. 2011 , 7 , 937–946. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Beyea, M.M.; Reaume, S.; Sawyez, C.G.; Edwards, J.Y.; O’Neil, C.; Hegele, R.A.; Pickering, J.G.; Huff, M.W. The Oxysterol 24 (S), 25-Epoxycholesterol Attenuates Human Smooth Muscle–Derived Foam Cell Formation Via Reduced Low-Density Lipoprotein Uptake and Enhanced Cholesterol Efflux. J. Am. Heart Assoc. 2012 , 1 , e000810. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Ricciarelli, R.; Zingg, J.-M.; Azzi, A. Vitamin E Reduces the Uptake of Oxidized LDL by Inhibiting CD36 Scavenger Receptor Expression in Cultured Aortic Smooth Muscle Cells. Circulation 1999 , 102 , 82–87. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Aoyama, T.; Chen, M.; Fujiwara, H.; Masaki, T.; Sawamura, T. LOX-1 mediates lysophosphatidylcholine-induced oxidized LDL uptake in smooth muscle cells. FEBS Lett. 2000 , 467 , 217–220. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Choi, H.Y.; Rahmani, M.; Wong, B.W.; Allahverdian, S.; McManus, B.M.; Pickering, J.G.; Chan, T.; Francis, G.A. ATP-Binding Cassette Transporter A1 Expression and Apolipoprotein A-I Binding Are Impaired in Intima-Type Arterial Smooth Muscle Cells. Circulation 2009 , 119 , 3223–3231. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Lusis, A.J. Atherosclerosis. Nature 2000 , 407 , 233–241. [ Google Scholar ] [ CrossRef ]
  • Fisher, E.A.; Feig, J.E.; Hewing, B.; Hazen, S.L.; Smith, J.D. High-Density Lipoprotein Function, Dysfunction, and Reverse Cholesterol Transport. Arterioscler. Thromb. Vasc. Biol. 2012 , 32 , 2813–2820. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Puri, R.; Nissen, S.E.; Ballantyne, C.M.; Barter, P.J.; Chapman, M.J.; Erbel, R.; Libby, P.; Raichlen, J.S.; John, J.S.; Wolski, K.; et al. Factors underlying regression of coronary atheroma with potent statin therapy. Eur. Heart J. 2013 , 34 , 1818–1825. [ Google Scholar ] [ CrossRef ]
  • Chamié, D.; Wang, Z.; Bezerra, H.; Rollins, A.M.; Costa, M.A. Optical Coherence Tomography and Fibrous Cap Characterization. Curr. Cardiovasc. Imaging Rep. 2011 , 4 , 276–283. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Watson, M.G.; Byrne, H.M.; Macaskill, C.; Myerscough, M.R. A two-phase model of early fibrous cap formation in atherosclerosis. J. Theor. Biol. 2018 , 456 , 123–136. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Schaper, W.; Ito, W.D. Molecular Mechanisms of Coronary Collateral Vessel Growth. Circ. Res. 1996 , 79 , 911–919. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Wolf, C.; Cai, W.-J.; Vosschulte, R.; Koltai, S.; Mousavipour, D.; Scholz, D.; Afsah-Hedjri, A.; Schaper, W.; Schaper, J. Vascular Remodeling and Altered Protein Expression During Growth of Coronary Collateral Arteries. J. Mol. Cell. Cardiol. 1998 , 30 , 2291–2305. [ Google Scholar ] [ CrossRef ]
  • Zalewski, A.; Shi, Y.; Johnson, A.G. Diverse Origin of Intimal Cells. Circ. Res. 2002 , 91 , 652–655. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Louis, S.F.; Zahradka, P. Vascular smooth muscle cell motility: From migration to invasion. Exp. Clin. Cardiol. 2010 , 15 , e75–e85. [ Google Scholar ]
  • Bennett, M.R.; Sinha, S.; Owens, G.K. Vascular Smooth Muscle Cells in Atherosclerosis. Circ. Res. 2016 , 118 , 692–702. [ Google Scholar ] [ CrossRef ]
  • Stiko-Rahm, A.; Hultgårdh-Nilsson, A.; Regnström, J.; Hamsten, A.; Nilsson, J. Native and oxidized LDL enhances production of PDGF AA and the surface expression of PDGF receptors in cultured human smooth muscle cells. Arter. Thromb. A J. Vasc. Biol. 1992 , 12 , 1099–1109. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kohno, M.; Yokokawa, K.; Yasunari, K.; Minami, M.; Kano, H.; Hanehira, T.; Yoshikawa, J. Induction by lysophosphatidylcholine, a major phospholipid component of atherogenic lipoproteins, of human coronary artery smooth muscle cell migration. Circulation 1998 , 98 , 353–359. [ Google Scholar ] [ CrossRef ]
  • Ross, R. The Pathogenesis of Atherosclerosis—An Update. N. Engl. J. Med. 1986 , 314 , 488–500. [ Google Scholar ] [ CrossRef ]
  • Raines, E.W.; Dower, S.K.; Ross, R. Interleukin-1 Mitogenic Activity for Fibroblasts and Smooth Muscle Cells Is Due to PDGF-AA. Science 1989 , 243 , 393–396. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Forsyth, E.A.; Aly, H.M.; Neville, R.F.; Sidawy, A.N. Proliferation and extracellular matrix production by human infragenicular smooth muscle cells in response to interleukin-1β. J. Vasc. Surg. 1997 , 26 , 1002–1008. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Bentzon, J.F.; Otsuka, F.; Virmani, R.; Falk, E. Mechanisms of Plaque Formation and Rupture. Circ. Res. 2014 , 114 , 1852–1866. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Shen, C.-M.; Mao, S.J.; Huang, G.; Yang, P.-C.; Chu, R.-M. Stimulation of smooth muscle cell proliferation by ox-LDL- and acetyl LDL-induced macrophage-derived foam cells. Life Sci. 2001 , 70 , 443–452. [ Google Scholar ] [ CrossRef ]
  • Sorokin, V.; Vickneson, K.; Kofidis, T.; Woo, C.C.; Lin, X.Y.; Foo, R.; Shanahan, C.M. Role of Vascular Smooth Muscle Cell Plasticity and Interactions in Vessel Wall Inflammation. Front. Immunol. 2020 , 11 , 599415. [ Google Scholar ] [ CrossRef ]
  • Badimon, L.; Padro, T.; Vilahur, G. Atherosclerosis, platelets and thrombosis in acute ischaemic heart disease. Eur. Heart J. Acute Cardiovasc. Care 2012 , 1 , 60–74. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Zhang, Z.; Wang, M.; Fan, X.-H.; Chen, J.-H.; Guan, Y.-Y.; Tang, Y.-B. Upregulation of TRPM7 Channels by Angiotensin II Triggers Phenotypic Switching of Vascular Smooth Muscle Cells of Ascending Aorta. Circ. Res. 2012 , 111 , 1137–1146. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Mack, C.P. Signaling Mechanisms That Regulate Smooth Muscle Cell Differentiation. Arterioscler. Thromb. Vasc. Biol. 2011 , 31 , 1495–1505. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Richardson, P.; Davies, M.; Born, G. Influence of Plaque Configuration and Stress Distribution on Fissuring of Coronary Atherosclerotic Plaques. Lancet 1989 , 334 , 941–944. [ Google Scholar ] [ CrossRef ]
  • Li, Z.-Y.; Howarth, S.P.; Tang, T.; Gillard, J.H. How Critical Is Fibrous Cap Thickness to Carotid Plaque Stability? Stroke 2006 , 37 , 1195–1199. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Anlamlert, W.; Lenbury, Y.; Bell, J. Modeling fibrous cap formation in atherosclerotic plaque development: Stability and oscillatory behavior. Adv. Differ. Equations 2017 , 2017 , 195. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Motoyama, S.; Sarai, M.; Harigaya, H.; Anno, H.; Inoue, K.; Hara, T.; Naruse, H.; Ishii, J.; Hishida, H.; Wong, N.D.; et al. Computed Tomographic Angiography Characteristics of Atherosclerotic Plaques Subsequently Resulting in Acute Coronary Syndrome. J. Am. Coll. Cardiol. 2009 , 54 , 49–57. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Visscher, M.; Moerman, A.M.; Burgers, P.C.; Van Beusekom, H.M.M.; Luider, T.M.; Verhagen, H.J.M.; Van der Steen, A.F.W.; Van der Heiden, K.; Van Soest, G. Data Processing Pipeline for Lipid Profiling of Carotid Atherosclerotic Plaque with Mass Spectrometry Imaging. J. Am. Soc. Mass Spectrom. 2019 , 30 , 1790–1800. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Thim, T.; Hagensen, M.K.; Bentzon, J.F.; Falk, E. From vulnerable plaque to atherothrombosis. J. Intern. Med. 2008 , 263 , 506–516. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Linton, M.F.; Babaev, V.R.; Huang, J.; Linton, E.F.; Tao, H.; Yancey, P.G. Macrophage Apoptosis and Efferocytosis in the Pathogenesis of Atherosclerosis. Circ. J. 2016 , 80 , 2259–2268. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Cabrera, J.T.O.; Makino, A. Efferocytosis of vascular cells in cardiovascular disease. Pharmacol. Ther. 2021 , 107919. [ Google Scholar ] [ CrossRef ]
  • Gonzalez, L.; Trigatti, B.L. Macrophage Apoptosis and Necrotic Core Development in Atherosclerosis: A Rapidly Advancing Field with Clinical Relevance to Imaging and Therapy. Can. J. Cardiol. 2017 , 33 , 303–312. [ Google Scholar ] [ CrossRef ]
  • Coornaert, I.; Hofmans, S.; Devisscher, L.; Augustyns, K.; Van der Veken, P.; De Meyer, G.; Martinet, W. Novel drug discovery strategies for atherosclerosis that target necrosis and necroptosis. Expert Opin. Drug Discov. 2018 , 13 , 477–488. [ Google Scholar ] [ CrossRef ]
  • Wanke, F.; Gutbier, S.; Rümmelin, A.; Steinberg, M.; Hughes, L.D.; Koenen, M.; Komuczki, J.; Regan-Komito, D.; Wagage, S.; Hesselmann, J.; et al. Ligand-dependent kinase activity of MERTK drives efferocytosis in human iPSC-derived macrophages. Cell Death Dis. 2021 , 12 , 538. [ Google Scholar ] [ CrossRef ]
  • Quillard, T.; Araújo, H.A.; Franck, G.; Tesmenitsky, Y.; Libby, P. Matrix Metalloproteinase-13 Predominates Over Matrix Metalloproteinase-8 as the Functional Interstitial Collagenase in Mouse Atheromata. Arterioscler. Thromb. Vasc. Biol. 2014 , 34 , 1179–1186. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Galis, Z.S.; Sukhova, G.K.; Kranzhöfer, R.; Clark, S.; Libby, P. Macrophage foam cells from experimental atheroma constitutively produce matrix-degrading proteinases. Proc. Natl. Acad. Sci. USA 1995 , 92 , 402–406. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Schneider, F.; Sukhova, G.K.; Aikawa, M.; Canner, J.; Gerdes, N.; Tang, S.-M.T.; Shi, G.-P.; Apte, S.; Libby, P. Matrix Metalloproteinase-14 Deficiency in Bone Marrow–Derived Cells Promotes Collagen Accumulation in Mouse Atherosclerotic Plaques. Circulation 2008 , 117 , 931–939. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Mulay, S.R.; Anders, H.-J. Crystallopathies. N. Engl. J. Med. 2016 , 374 , 2465–2476. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Nidorf, S.M.; Fiolet, A.; Abela, G.S. Viewing atherosclerosis through a crystal lens: How the evolving structure of cholesterol crystals in atherosclerotic plaque alters its stability. J. Clin. Lipidol. 2020 , 14 , 619–630. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Warnatsch, A.; Ioannou, M.; Wang, Q.; Papayannopoulos, V. Neutrophil extracellular traps license macrophages for cytokine production in atherosclerosis. Science 2015 , 349 , 316–320. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Johnson, J.L.; Jenkins, N.P.; Huang, W.-C.; Di Gregoli, K.; Sala-Newby, G.B.; Scholtes, V.P.W.; Moll, F.L.; Pasterkamp, G.; Newby, A.C. Relationship of MMP-14 and TIMP-3 Expression with Macrophage Activation and Human Atherosclerotic Plaque Vulnerability. Mediat. Inflamm. 2014 , 2014 , 276457. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • García-García, H.M.; Klauss, V.; Gonzalo, N.; Garg, S.; Onuma, Y.; Hamm, C.W.; Wijns, W.; Shannon, J.; Serruys, P.W. Relationship between cardiovascular risk factors and biomarkers with necrotic core and atheroma size: A serial intravascular ultrasound radiofrequency data analysis. Int. J. Cardiovasc. Imaging 2012 , 28 , 695–703. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Martinet, W.; Schrijvers, D.; De Meyer, G. Necrotic cell death in atherosclerosis. Basic Res. Cardiol. 2011 , 106 , 749–760. [ Google Scholar ] [ CrossRef ]
  • Steffel, J.; Lüscher, T.F.; Tanner, F.C. Tissue Factor in Cardiovascular Diseases. Circulation 2006 , 113 , 722–731. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Nakahara, T.; Dweck, M.; Narula, N.; Pisapia, D.; Narula, J.; Strauss, H.W. Coronary Artery Calcification. JACC Cardiovasc. Imaging 2017 , 10 , 582–593. [ Google Scholar ] [ CrossRef ]
  • Shi, X.; Gao, J.; Lv, Q.; Cai, H.; Wang, F.; Ye, R.; Liu, X. Calcification in Atherosclerotic Plaque Vulnerability: Friend or Foe? Front. Physiol. 2020 , 11 , 56. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Bobryshev, Y.V.; Killingsworth, M.C.; Lord, R.S.A.; Grabs, A.J. Matrix vesicles in the fibrous cap of atherosclerotic plaque: Possible contribution to plaque rupture. J. Cell. Mol. Med. 2008 , 12 , 2073–2082. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Anderson, H.C. Mechanisms of Pathologic Calcification. Rheum. Dis. Clin. N. Am. 1988 , 14 , 303–319. [ Google Scholar ] [ CrossRef ]
  • Kapustin, A.N.; Chatrou, M.L.; Drozdov, I.; Zheng, Y.; Davidson, S.M.; Soong, D.; Furmanik, M.; Sanchis, P.; de Rosales, R.T.M.; Alvarez-Hernandez, D.; et al. Vascular Smooth Muscle Cell Calcification Is Mediated by Regulated Exosome Secretion. Circ. Res. 2015 , 116 , 1312–1323. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • New, S.E.P.; Goettsch, C.; Aikawa, M.; Marchini, J.; Shibasaki, M.; Yabusaki, K.; Libby, P.; Shanahan, C.; Croce, K.; Aikawa, E. Macrophage-Derived Matrix Vesicles. Circ. Res. 2013 , 113 , 72–77. [ Google Scholar ] [ CrossRef ]
  • Canfield, A.; Sutton, A.; Hoyland, J.; Schor, A. Association of thrombospondin-1 with osteogenic differentiation of retinal pericytes in vitro. J. Cell Sci. 1996 , 109 , 343–353. [ Google Scholar ] [ CrossRef ]
  • Lomashvili, K.A.; Cobbs, S.; Hennigar, R.A.; Hardcastle, K.I.; O’Neill, W.C. Phosphate-Induced Vascular Calcification: Role of Pyrophosphate and Osteopontin. J. Am. Soc. Nephrol. 2004 , 15 , 1392–1401. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Otsuka, F.; Sakakura, K.; Yahagi, K.; Joner, M.; Virmani, R. Has Our Understanding of Calcification in Human Coronary Atherosclerosis Progressed? Arterioscler. Thromb. Vasc. Biol. 2014 , 34 , 724–736. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Proudfoot, D.; Skepper, J.N.; Shanahan, C.M.; Weissberg, P.L. Calcification of Human Vascular Cells in Vitro Is Correlated With High Levels of Matrix Gla Protein and Low Levels of Osteopontin Expression. Arterioscler. Thromb. Vasc. Biol. 1998 , 18 , 379–388. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Demer, L.L.; Tintut, Y. Vascular Calcification: Pathobiology of a multifaceted disease. Circulation 2008 , 117 , 2938–2948. [ Google Scholar ] [ CrossRef ]
  • Reynolds, J.L.; Joannides, A.J.; Skepper, J.N.; McNair, R.; Schurgers, L.J.; Proudfoot, D.; Jahnen-Dechent, W.; Weissberg, P.L.; Shanahan, C.M. Human Vascular Smooth Muscle Cells Undergo Vesicle-Mediated Calcification in Response to Changes in Extracellular Calcium and Phosphate Concentrations: A Potential Mechanism for Accelerated Vascular Calcification in ESRD. J. Am. Soc. Nephrol. 2004 , 15 , 2857–2867. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Reith, S.; Milzi, A.; Dettori, R.; Marx, N.; Burgmaier, M. Predictors for target lesion microcalcifications in patients with stable coronary artery disease: An optical coherence tomography study. Clin. Res. Cardiol. 2018 , 107 , 763–771. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Pugliese, G.; Iacobini, C.; Fantauzzi, C.B.; Menini, S. The dark and bright side of atherosclerotic calcification. Atherosclerosis 2015 , 238 , 220–230. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Slager, C.J.; Wentzel, J.J.; Gijsen, F.J.H.; Schuurbiers, J.C.H.; van der Wal, A.; Van Der Steen, A.F.W.; Serruys, P.W. The role of shear stress in the generation of rupture-prone vulnerable plaques. Nat. Clin. Pract. Cardiovasc. Med. 2005 , 2 , 401–407. [ Google Scholar ] [ CrossRef ]
  • Stefanadis, C.; Antoniou, C.; Tsiachris, D.; Pietri, P. Coronary Atherosclerotic Vulnerable Plaque: Current Perspectives. J. Am. Heart Assoc. 2017 , 6 , e005543. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Cuadrado, I.; Saura, M.; Castejón, B.; Martin, A.M.; Herruzo, I.; Balatsos, N.; Zamorano, J.L.; Zaragoza, C. Preclinical models of atherosclerosis. The future of Hybrid PET/MR technology for the early detection of vulnerable plaque. Expert Rev. Mol. Med. 2016 , 18 , E6. [ Google Scholar ] [ CrossRef ]
  • Sodhi, N.; Brown, D.L. Pathophysiology of Acute Coronary Syndromes. In Cardiac Intensive Care ; Elsevier BV: Amsterdam, The Netherlands, 2019; pp. 68–80.e2. [ Google Scholar ]
  • Libby, P. Inflammation in atherosclerosis. Nature 2002 , 420 , 868–874. [ Google Scholar ] [ CrossRef ]
  • Kovanen, P.T.; Kaartinen, M.; Paavonen, T. Infiltrates of Activated Mast Cells at the Site of Coronary Atheromatous Erosion or Rupture in Myocardial Infarction. Circulation 1995 , 92 , 1084–1088. [ Google Scholar ] [ CrossRef ]
  • Sarén, P.; Welgus, H.G.; Kovanen, P.T. TNF-alpha and IL-1beta selectively induce expression of 92-kDa gelatinase by human macrophages. J. Immunol. 1996 , 157 , 4159–4165. [ Google Scholar ]
  • Pasterkamp, G.; Schoneveld, A.H.; van der Wal, A.C.; Hijnen, D.-J.; van Wolveren, W.J.A.; Plomp, S.; Teepen, H.L.J.M.; Borst, C. Inflammation of the Atherosclerotic Cap and Shoulder of the Plaque Is a Common and Locally Observed Feature in Unruptured Plaques of Femoral and Coronary Arteries. Arterioscler. Thromb. Vasc. Biol. 1999 , 19 , 54–58. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Groen, H.C.; Gijsen, F.J.; Van Der Lugt, A.; Ferguson, M.S.; Hatsukami, T.S.; Van Der Steen, A.F.; Yuan, C.; Wentzel, J.J. Plaque Rupture in the Carotid Artery Is Localized at the High Shear Stress Region. Stroke 2007 , 38 , 2379–2381. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Vergallo, R.; Crea, F. Atherosclerotic Plaque Healing. N. Engl. J. Med. 2020 , 383 , 846–857. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Yun, S.-H.; Sim, E.-H.; Goh, R.-Y.; Park, J.-I.; Han, J.-Y. Platelet Activation: The Mechanisms and Potential Biomarkers. BioMed Res. Int. 2016 , 2016 , 9060143. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Osaki, T.; Ichinose, A. [Current views of activating and regulatory mechanisms of blood coagulation]. Nihon rinsho. Jpn. J. Clin. Med. 2014 , 72 , 1206–1211. [ Google Scholar ]
  • Mackman, N.; Tilley, R.E.; Key, N.S. Role of the extrinsic pathway of blood coagulation in hemostasis and thrombosis. Arterioscler. Thromb. Vasc. Biol. 2007 , 27 , 1687–1693. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Grainger, D.J.; Wakefield, L.; Bethell, H.W.; Farndale, R.W.; Metcalfe, J.C. Release and activation of platelet latent TGF–β in blood clots during dissolution with plasmin. Nat. Med. 1995 , 1 , 932–937. [ Google Scholar ] [ CrossRef ]
  • Epstein, F.H.; Fuster, V.; Badimon, L.; Badimon, J.J.; Chesebro, J.H. The Pathogenesis of Coronary Artery Disease and the Acute Coronary Syndromes. N. Engl. J. Med. 1992 , 326 , 310–318. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Théroux, P.; Fuster, V. Acute Coronary Syndromes. Circulation 1998 , 97 , 1195–1206. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Thanvi, B.; Robinson, T. Complete occlusion of extracranial internal carotid artery: Clinical features, pathophysiology, diagnosis and management. Postgrad. Med. J. 2007 , 83 , 95–99. [ Google Scholar ] [ CrossRef ]
  • Lyaker, M.R.; Tulman, D.B.; Dimitrova, G.T.; Pin, R.H.; Papadimos, T.J. Arterial embolism. Int. J. Crit. Illn. Inj. Sci. 2013 , 3 , 77–87. [ Google Scholar ] [ CrossRef ]
  • Ridker, P.M.; Howard, C.P.; Walter, V.; Everett, B.; Libby, P.; Hensen, J.; Thuren, T. Effects of Interleukin-1β Inhibition With Canakinumab on Hemoglobin A1c, Lipids, C-Reactive Protein, Interleukin-6, and Fibrinogen. Circulation 2012 , 126 , 2739–2748. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Meiliana, A.; Dewi, N.M.; Wijaya, A. Advanced in Molecular Mechanisms of Atherosclerosis: From Lipids to Inflammation. Indones. Biomed. J. 2018 , 10 , 104–122. [ Google Scholar ] [ CrossRef ]
  • Cushing, S.D.; A Berliner, J.; Valente, A.J.; Territo, M.C.; Navab, M.; Parhami, F.; Gerrity, R.; Schwartz, C.J.; Fogelman, A.M. Minimally modified low density lipoprotein induces monocyte chemotactic protein 1 in human endothelial cells and smooth muscle cells. Proc. Natl. Acad. Sci. USA 1990 , 87 , 5134–5138. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Quinn, M.T.; Parthasarathy, S.; Fong, L.G.; Steinberg, D. Oxidatively modified low density lipoproteins: A potential role in recruitment and retention of monocyte/macrophages during atherogenesis. Proc. Natl. Acad. Sci. USA 1987 , 84 , 2995–2998. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Soehnlein, O.; Drechsler, M.; Döring, Y.; Lievens, D.; Hartwig, H.; Kemmerich, K.; Ortega-Gomez, A.; Mandl, M.; Vijayan, S.; Projahn, D.; et al. Distinct functions of chemokine receptor axes in the atherogenic mobilization and recruitment of classical monocytes. EMBO Mol. Med. 2013 , 5 , 471–481. [ Google Scholar ] [ CrossRef ]
  • Chávez-Sánchez, L.; Madrid-Miller, A.; Chávez-Rueda, K.; Legorreta-Haquet, M.; Tesoro-Cruz, E.; Blanco-Favela, F. Activation of TLR2 and TLR4 by minimally modified low-density lipoprotein in human macrophages and monocytes triggers the inflammatory response. Hum. Immunol. 2010 , 71 , 737–744. [ Google Scholar ] [ CrossRef ]
  • Miller, Y.I.; Viriyakosol, S.; Worrall, D.S.; Boullier, A.; Butler, S.; Witztum, J.L. Toll-Like Receptor 4–Dependent and –Independent Cytokine Secretion Induced by Minimally Oxidized Low-Density Lipoprotein in Macrophages. Arterioscler. Thromb. Vasc. Biol. 2005 , 25 , 1213–1219. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Schroder, K.; Tschopp, J. The Inflammasomes. Cell 2010 , 140 , 821–832. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Lopes-Virella, M.F.; Virella, G. Pathogenic Role of Modified LDL Antibodies and Immune Complexes in Atherosclerosis. J. Atheroscler. Thromb. 2013 , 20 , 743–754. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Rhoads, J.P.; Lukens, J.R.; Wilhelm, A.J.; Moore, J.L.; Mendez-Fernandez, Y.; Kanneganti, T.-D.; Major, A.S. Oxidized Low-Density Lipoprotein Immune Complex Priming of the Nlrp3 Inflammasome Involves TLR and FcγR Cooperation and Is Dependent on CARD9. J. Immunol. 2017 , 198 , 2105–2114. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Duewell, P.; Kono, H.; Rayner, K.J.; Sirois, C.M.; Vladimer, G.; Bauernfeind, F.G.; Abela, G.S.; Franchi, L.; Nuñez, G.; Schnurr, M.; et al. NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature 2010 , 464 , 1357–1361. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Mofidi, R.; Crotty, T.B.; McCarthy, P.; Sheehan, S.J.; Mehigan, D.; Keaveny, T.V. Association between plaque instability, angiogenesis and symptomatic carotid occlusive disease. Br. J. Surg. 2002 , 88 , 945–950. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Spagnoli, L.G.; Bonanno, E.; Sangiorgi, G.; Mauriello, A. Role of Inflammation in Atherosclerosis. J. Nucl. Med. 2007 , 48 , 1800–1815. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Akhmedov, A.; Sawamura, T.; Chen, C.-H.; Kraler, S.; Vdovenko, D.; Lüscher, T.F. Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1): A crucial driver of atherosclerotic cardiovascular disease. Eur. Heart J. 2021 , 42 , 1797–1807. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Chen, L.; Yang, G.; Monslow, J.; Todd, L.; Cormode, D.P.; Tang, J.; Grant, G.R.; DeLong, J.H.; Tang, S.Y.; Lawson, J.A.; et al. Myeloid cell microsomal prostaglandin E synthase-1 fosters atherogenesis in mice. Proc. Natl. Acad. Sci. USA 2014 , 111 , 6828–6833. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Yang, G.; Chen, L. An Update of Microsomal Prostaglandin E Synthase-1 and PGE2Receptors in Cardiovascular Health and Diseases. Oxidative Med. Cell. Longev. 2016 , 2016 , 5249086. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Haemmig, S.; Simion, V.; Yang, D.; Deng, Y.; Feinberg, M.W. Long noncoding RNAs in cardiovascular disease, diagnosis, and therapy. Curr. Opin. Cardiol. 2017 , 32 , 776–783. [ Google Scholar ] [ CrossRef ]
  • Freedman, J.E.; Miano, J.M. Challenges and Opportunities in Linking Long Noncoding RNAs to Cardiovascular, Lung, and Blood Diseases. Arterioscler. Thromb. Vasc. Biol. 2017 , 37 , 21–25. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Boettger, T.; Braun, T. A New Level of Complexity. Circ. Res. 2012 , 110 , 1000–1013. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Wang, R.; Dong, L.-D.; Meng, X.-B.; Shi, Q.; Sun, W.-Y. Unique MicroRNA signatures associated with early coronary atherosclerotic plaques. Biochem. Biophys. Res. Commun. 2015 , 464 , 574–579. [ Google Scholar ] [ CrossRef ]
  • Di Gregoli, K.; Jenkins, N.; Salter, R.; White, S.; Newby, A.C.; Johnson, J.L. MicroRNA-24 Regulates Macrophage Behavior and Retards Atherosclerosis. Arterioscler. Thromb. Vasc. Biol. 2014 , 34 , 1990–2000. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Di Gregoli, K.; Anuar, N.N.M.; Bianco, R.; White, S.J.; Newby, A.C.; George, S.J.; Johnson, J.L. MicroRNA-181b Controls Atherosclerosis and Aneurysms Through Regulation of TIMP-3 and Elastin. Circ. Res. 2017 , 120 , 49–65. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Raitoharju, E.; Lyytikäinen, L.-P.; Levula, M.; Oksala, N.; Mennander, A.; Tarkka, M.; Klopp, N.; Illig, T.; Kähönen, M.; Karhunen, P.J.; et al. miR-21, miR-210, miR-34a, and miR-146a/b are up-regulated in human atherosclerotic plaques in the Tampere Vascular Study. Atherosclerosis 2011 , 219 , 211–217. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Denecke, B.; Rostalsky, A.; Hristov, M.; Koeppel, T.A.; Bidzhekov, K.; Gan, L.; Zernecke, A.; Weber, C. microRNA expression signatures and parallels between monocyte subsets and atherosclerotic plaque in humans. Thromb. Haemost. 2012 , 107 , 619–625. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Cipollone, F.; Felicioni, L.; Sarzani, R.; Ucchino, S.; Spigonardo, F.; Mandolini, C.; Malatesta, S.; Bucci, M.; Mammarella, C.; Santovito, D.; et al. A Unique MicroRNA Signature Associated With Plaque Instability in Humans. Stroke 2011 , 42 , 2556–2563. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Zhu, N.; Zhang, D.; Chen, S.; Liu, X.; Lin, L.; Huang, X.; Guo, Z.; Liu, J.; Wang, Y.; Yuan, W.; et al. Endothelial enriched microRNAs regulate angiotensin II-induced endothelial inflammation and migration. Atherosclerosis 2011 , 215 , 286–293. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Lee, D.-Y.; Yang, T.-L.; Huang, Y.-H.; Lee, C.-I.; Chen, L.-J.; Shih, Y.-T.; Wei, S.-Y.; Wang, W.-L.; Wu, C.-C.; Chiu, J.-J. Induction of microRNA-10a using retinoic acid receptor-α and retinoid x receptor-α agonists inhibits atherosclerotic lesion formation. Atherosclerosis 2018 , 271 , 36–44. [ Google Scholar ] [ CrossRef ]
  • Suárez, Y.; Wang, C.; Manes, T.D.; Pober, J.S. Cutting Edge: TNF-Induced MicroRNAs Regulate TNF-Induced Expression of E-Selectin and Intercellular Adhesion Molecule-1 on Human Endothelial Cells: Feedback Control of Inflammation. J. Immunol. 2010 , 184 , 21–25. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Fish, J.E.; Santoro, M.M.; Morton, S.U.; Yu, S.; Yeh, R.-F.; Wythe, J.D.; Ivey, K.N.; Bruneau, B.G.; Stainier, D.Y.R.; Srivastava, D. miR-126 Regulates Angiogenic Signaling and Vascular Integrity. Dev. Cell 2008 , 15 , 272–284. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Sun, X.; Icli, B.; Wara, A.K.; Belkin, N.; He, S.; Kobzik, L.; Hunninghake, G.M.; Pinilla-Vera, M.; Blackwell, T.S.; Baron, R.M.; et al. MicroRNA-181b regulates NF-κB–mediated vascular inflammation. J. Clin. Investig. 2012 , 122 , 1973–1990. [ Google Scholar ] [ CrossRef ]
  • Yang, K.; He, Y.S.; Wang, X.Q.; Lu, L.; Chen, Q.J.; Liu, J.; Sun, Z.; Shen, W.F. MiR-146a inhibits oxidized low-density lipoprotein-induced lipid accumulation and inflammatory response via targeting toll-like receptor 4. FEBS Lett. 2011 , 585 , 854–860. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Chen, T.; Huang, Z.; Wang, L.; Wang, Y.; Wu, F.; Meng, S.; Wang, C. MicroRNA-125a-5p partly regulates the inflammatory response, lipid uptake, and ORP9 expression in oxLDL-stimulated monocyte/macrophages. Cardiovasc. Res. 2009 , 83 , 131–139. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Andreou, I.; Sun, X.; Stone, P.H.; Edelman, E.R.; Feinberg, M.W. miRNAs in atherosclerotic plaque initiation, progression, and rupture. Trends Mol. Med. 2015 , 21 , 307–318. [ Google Scholar ] [ CrossRef ]
  • Wu, W.; Xiao, H.; Laguna-Fernandez, A.; Villarreal, G., Jr.; Wang, K.-C.; Geary, G.G.; Zhang, Y.; Wang, W.-C.; Huang, H.-D.; Zhou, J.; et al. Flow-Dependent Regulation of Krüppel-Like Factor 2 Is Mediated by MicroRNA-92a. Circulation 2011 , 124 , 633–641. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Loyer, X.; Potteaux, S.; Vion, A.-C.; Guérin, C.L.; Boulkroun, S.; Rautou, P.-E.; Ramkhelawon, B.; Esposito, B.; Dalloz, M.; Paul, J.-L.; et al. Inhibition of MicroRNA-92a Prevents Endothelial Dysfunction and Atherosclerosis in Mice. Circ. Res. 2014 , 114 , 434–443. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Hsu, P.; Lin, W.; Lin, R.; Juo, S.H. MicroRNA let-7g inhibits angiotensin II-induced endothelial senescence via the LOX-1-independent mechanism. Int. J. Mol. Med. 2018 , 41 , 2243–2251. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Yang, S.; Mi, X.; Chen, Y.; Feng, C.; Hou, Z.; Hui, R.; Zhang, W. MicroRNA-216a induces endothelial senescence and inflammation via Smad3/IκBα pathway. J. Cell. Mol. Med. 2018 , 22 , 2739–2749. [ Google Scholar ] [ CrossRef ]
  • Chaudhuri, A.A.; So, A.Y.-L.; Sinha, N.; Gibson, W.S.J.; Taganov, K.D.; O’Connell, R.M.; Baltimore, D. MicroRNA-125b Potentiates Macrophage Activation. J. Immunol. 2011 , 187 , 5062–5068. [ Google Scholar ] [ CrossRef ]
  • Wei, Y.; Nazari-Jahantigh, M.; Chan, L.; Zhu, M.; Heyll, K.; Corbalán-Campos, J.; Hartmann, P.; Thiemann, A.; Weber, C.; Schober, A. The microRNA-342-5p Fosters Inflammatory Macrophage Activation Through an Akt1- and microRNA-155–Dependent Pathway during Atherosclerosis. Circulation 2013 , 127 , 1609–1619. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Price, N.L.; Goedeke, L.; Suárez, Y.; Fernández-Hernando, C. miR-33 in cardiometabolic diseases: Lessons learned from novel animal models and approaches. EMBO Mol. Med. 2021 , 13 , e12606. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Alshanwani, A.R.; Riches-Suman, K.; O’Regan, D.J.; Wood, I.C.; Turner, N.; Porter, K.E. MicroRNA-21 drives the switch to a synthetic phenotype in human saphenous vein smooth muscle cells. IUBMB Life 2018 , 70 , 649–657. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Liu, X.; Cheng, Y.; Zhang, S.; Lin, Y.; Yang, J.; Zhang, C. A Necessary Role of miR-221 and miR-222 in Vascular Smooth Muscle Cell Proliferation and Neointimal Hyperplasia. Circ. Res. 2009 , 104 , 476–487. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Leeper, N.J.; Raiesdana, A.; Kojima, Y.; Chun, H.; Azuma, J.; Maegdefessel, L.; Kundu, R.K.; Quertermous, T.; Tsao, P.S.; Spin, J.M. MicroRNA-26a is a novel regulator of vascular smooth muscle cell function. J. Cell. Physiol. 2011 , 226 , 1035–1043. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Torella, D.; Iaconetti, C.; Catalucci, D.; Ellison, G.; Leone, A.; Waring, C.D.; Bochicchio, A.; Vicinanza, C.; Aquila, I.; Curcio, A.; et al. MicroRNA-133 Controls Vascular Smooth Muscle Cell Phenotypic Switch In Vitro and Vascular Remodeling In Vivo. Circ. Res. 2011 , 109 , 880–893. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Talasila, A.; Yu, H.; Ackers-Johnson, M.; Bot, M.; Van Berkel, T.; Bennett, M.R.; Bot, I.; Sinha, S. Myocardin Regulates Vascular Response to Injury Through miR-24/-29a and Platelet-Derived Growth Factor Receptor-β. Arterioscler. Thromb. Vasc. Biol. 2013 , 33 , 2355–2365. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Li, P.; Zhu, N.; Yi, B.; Wang, N.; Chen, M.; You, X.; Zhao, X.; Solomides, C.C.; Qin, Y.; Sun, J. MicroRNA-663 Regulates Human Vascular Smooth Muscle Cell Phenotypic Switch and Vascular Neointimal Formation. Circ. Res. 2013 , 113 , 1117–1127. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Xu, F.; Ahmed, A.; Kang, X.; Hu, G.; Liu, F.; Zhang, W.; Zhou, J. MicroRNA-15b/16 Attenuates Vascular Neointima Formation by Promoting the Contractile Phenotype of Vascular Smooth Muscle Through Targeting YAP. Arterioscler. Thromb. Vasc. Biol. 2015 , 35 , 2145–2152. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Ouimet, M.; Ediriweera, H.N.; Gundra, U.M.; Sheedy, F.; Ramkhelawon, B.; Hutchison, S.B.; Rinehold, K.; Van Solingen, C.; Fullerton, M.D.; Cecchini, K.; et al. MicroRNA-33–dependent regulation of macrophage metabolism directs immune cell polarization in atherosclerosis. J. Clin. Investig. 2015 , 125 , 4334–4348. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Stary, H.C.; Blankenhorn, D.H.; Chandler, A.B.; Glagov, S.; Insull, W.; Richardson, M.; E Rosenfeld, M.; A Schaffer, S.; Schwartz, C.J.; Wagner, W.D. A definition of the intima of human arteries and of its atherosclerosis-prone regions. A report from the Committee on Vascular Lesions of the Council on Arteriosclerosis, American Heart Association. Circulation 1992 , 85 , 391–405. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Fuster, V.; Moreno, P.R.; Fayad, Z.A.; Corti, R.; Badimon, J.J. Atherothrombosis and High-Risk Plaque: Part I: Evolving Concepts. J. Am. Coll. Cardiol. 2005 , 46 , 937–954. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Chen, Y.-C.; Bui, A.V.; Diesch, J.; Manasseh, R.; Hausding, C.; Rivera, J.; Haviv, I.; Agrotis, A.; Htun, N.M.; Jowett, J.; et al. A Novel Mouse Model of Atherosclerotic Plaque Instability for Drug Testing and Mechanistic/Therapeutic Discoveries Using Gene and MicroRNA Expression Profiling. Circ. Res. 2013 , 113 , 252–265. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Son, D.J.; Kumar, S.; Takabe, W.; Kim, C.W.; Ni, C.-W.; Alberts-Grill, N.; Jang, I.-H.; Kim, S.; Kim, W.; Kang, S.W.; et al. The atypical mechanosensitive microRNA-712 derived from pre-ribosomal RNA induces endothelial inflammation and atherosclerosis. Nat. Commun. 2013 , 4 , 3000. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Shi, L.; Yu, C.; Tian, X.; Ma, C.; Wang, L.; Xia, D.; Cui, C.; Chen, X.; Jiang, T.; Gu, Y.; et al. Effect of microRNA-133a-3p/matrix metalloproteinase-9 axis on the growth of atherosclerotic vascular smooth muscle cells. Exp. Ther. Med. 2019 . [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Wezel, A.; Welten, S.M.J.; Razawy, W.; Lagraauw, H.M.; de Vries, M.R.; Goossens, E.A.C.; Boonstra, M.C.; Hamming, J.F.; Kandimalla, E.R.; Kuiper, J.; et al. Inhibition of MicroRNA-494 Reduces Carotid Artery Atherosclerotic Lesion Development and Increases Plaque Stability. Ann. Surg. 2015 , 262 , 841–848. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Ma, F.; Xu, S.; Liu, X.; Zhang, Q.; Xu, X.; Liu, M.; Hua, M.; Li, N.; Yao, H.; Cao, X. The microRNA miR-29 controls innate and adaptive immune responses to intracellular bacterial infection by targeting interferon-γ. Nat. Immunol. 2011 , 12 , 861–869. [ Google Scholar ] [ CrossRef ]
  • Boon, R.A.; Seeger, T.; Heydt, S.; Fischer, A.; Hergenreider, E.; Horrevoets, A.J.; Vinciguerra, M.; Rosenthal, N.; Sciacca, S.; Pilato, M.; et al. MicroRNA-29 in Aortic Dilation: Implications for Aneurysm Formation. Circ. Res. 2011 , 109 , 1115–1119. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Das, A.; Ganesh, K.; Khanna, S.; Sen, C.K.; Roy, S. Engulfment of Apoptotic Cells by Macrophages: A Role of MicroRNA-21 in the Resolution of Wound Inflammation. J. Immunol. 2014 , 192 , 1120–1129. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Haneklaus, M.; Gerlic, M.; Kurowska-Stolarska, M.; Rainey, A.-A.; Pich, D.; McInnes, I.B.; Hammerschmidt, W.; O’Neill, L.A.J.; Masters, S.L. Cutting Edge: miR-223 and EBV miR-BART15 Regulate the NLRP3 Inflammasome and IL-1β Production. J. Immunol. 2012 , 189 , 3795–3799. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Ghorpade, D.S.; Leyland, R.; Kurowska-Stolarska, M.; Patil, S.A.; Balaji, K.N. MicroRNA-155 Is Required for Mycobacterium bovis BCG-Mediated Apoptosis of Macrophages. Mol. Cell. Biol. 2012 , 32 , 2239–2253. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Gao, Y.; Peng, J.; Ren, Z.; He, N.-Y.; Li, Q.; Zhao, X.-S.; Wang, M.-M.; Wen, H.-Y.; Tang, Z.-H.; Jiang, Z.-S.; et al. Functional regulatory roles of microRNAs in atherosclerosis. Clin. Chim. Acta 2016 , 460 , 164–171. [ Google Scholar ] [ CrossRef ]
  • Santulli, G.; Wronska, A.; Uryu, K.; Diacovo, T.G.; Gao, M.; Marx, S.O.; Kitajewski, J.; Chilton, J.M.; Akat, K.M.; Tuschl, T.; et al. A selective microRNA-based strategy inhibits restenosis while preserving endothelial function. J. Clin. Investig. 2014 , 124 , 4102–4114. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Cordes, K.R.; Sheehy, N.T.; White, M.P.; Berry, E.C.; Morton, S.U.; Muth, A.N.; Lee, T.-H.; Miano, J.M.; Ivey, K.N.; Srivastava, D. miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature 2009 , 460 , 705–710. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Avgeris, M.; Mavridis, K.; Tokas, T.; Stravodimos, K.; Fragoulis, E.G.; Scorilas, A. Uncovering the clinical utility of miR-143, miR-145 and miR-224 for predicting the survival of bladder cancer patients following treatment. Carcinogenesis 2015 , 36 , 528–537. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Engelhardt, S.; Leierseder, S. Coinciding functions for miR-145 in vascular smooth muscle and cardiac fibroblasts. J. Mol. Cell. Cardiol. 2013 , 65 , 105–107. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Elia, L.; Quintavalle, M.; Zhang, J.; Contu, R.; Cossu, L.; Latronico, M.V.G.; Peterson, K.L.; Indolfi, C.; Catalucci, D.; Chen, J.; et al. The knockout of miR-143 and -145 alters smooth muscle cell maintenance and vascular homeostasis in mice: Correlates with human disease. Cell Death Differ. 2009 , 16 , 1590–1598. [ Google Scholar ] [ CrossRef ]
  • Climent, M.; Quintavalle, M.; Miragoli, M.; Chen, J.; Condorelli, G.; Elia, L. TGFβ Triggers miR-143/145 Transfer From Smooth Muscle Cells to Endothelial Cells, Thereby Modulating Vessel Stabilization. Circ. Res. 2015 , 116 , 1753–1764. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Wang, S.; Aurora, A.B.; Johnson, B.A.; Qi, X.; McAnally, J.; Hill, J.A.; Richardson, J.A.; Bassel-Duby, R.; Olson, E.N. The Endothelial-Specific MicroRNA miR-126 Governs Vascular Integrity and Angiogenesis. Dev. Cell 2008 , 15 , 261–271. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Feng, X.; Tan, W.; Cheng, S.; Wang, H.; Ye, S.; Yu, C.; He, Y.; Zeng, J.; Cen, J.; Hu, J.; et al. Upregulation of microRNA-126 in Hepatic Stellate Cells May Affect Pathogenesis of Liver Fibrosis Through the NF-κB Pathway. DNA Cell Biol. 2015 , 34 , 470–480. [ Google Scholar ] [ CrossRef ]
  • Rohde, J.-H.; Weigand, J.E.; Suess, B.; Dimmeler, S. A Universal Aptamer Chimera for the Delivery of Functional microRNA-126. Nucleic Acid Ther. 2015 , 25 , 141–151. [ Google Scholar ] [ CrossRef ]
  • Shibayama, Y.; Kondo, T.; Ohya, H.; Fujisawa, S.-I.; Teshima, T.; Iseki, K. Upregulation of microRNA-126-5p is associated with drug resistance to cytarabine and poor prognosis in AML patients. Oncol. Rep. 2015 , 33 , 2176–2182. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Hu, J.; Zeng, L.; Huang, J.; Wang, G.; Lu, H. miR-126 promotes angiogenesis and attenuates inflammation after contusion spinal cord injury in rats. Brain Res. 2015 , 1608 , 191–202. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Nakano, M.; Fukushima, Y.; Yokota, S.-I.; Fukami, T.; Takamiya, M.; Aoki, Y.; Yokoi, T.; Nakajima, M. CYP2A7 Pseudogene Transcript Affects CYP2A6 Expression in Human Liver by Acting as a Decoy for miR-126*. Drug Metab. Dispos. 2015 , 43 , 703–712. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Meng, Q.; Wang, W.; Yu, X.; Li, W.; Kong, L.; Qian, A.; Li, C.; Li, X. Upregulation of MicroRNA-126 Contributes to Endothelial Progenitor Cell Function in Deep Vein Thrombosis via Its Target PIK3R2. J. Cell. Biochem. 2015 , 116 , 1613–1623. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Mocharla, P.; Briand, S.; Giannotti, G.; Dörries, C.; Jakob, P.; Paneni, F.; Lüscher, T.; Landmesser, U. AngiomiR-126 expression and secretion from circulating CD34+ and CD14+ PBMCs: Role for proangiogenic effects and alterations in type 2 diabetics. Blood 2013 , 121 , 226–236. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Novák, J.; Vašků, J.B.; Souček, M. Long non-coding RNAs in the pathophysiology of atherosclerosis. Vnitřní Lékařství 2018 , 64 , 77–82. [ Google Scholar ] [ CrossRef ]
  • Sallam, T.; Jones, M.; Thomas, B.J.; Wu, X.; Gilliland, T.; Qian, K.; Eskin, A.; Casero, D.; Zhang, Z.; Sandhu, J.; et al. Transcriptional regulation of macrophage cholesterol efflux and atherogenesis by a long noncoding RNA. Nat. Med. 2018 , 24 , 304–312. [ Google Scholar ] [ CrossRef ]
  • Arslan, S.; Berkan, Ö.; Lalem, T.; Özbilüm, N.; Göksel, S.; Korkmaz, Ö; Çetin, N.; Devaux, Y. Long non-coding RNAs in the atherosclerotic plaque. Atherosclerosis 2017 , 266 , 176–181. [ Google Scholar ] [ CrossRef ]
  • Huangfu, N.; Xu, Z.; Zheng, W.; Wang, Y.; Cheng, J.; Chen, X. LncRNA MALAT1 regulates oxLDL-induced CD36 expression via activating β-catenin. Biochem. Biophys. Res. Commun. 2018 , 495 , 2111–2117. [ Google Scholar ] [ CrossRef ]
  • Hennessy, E.J. Cardiovascular Disease and Long Noncoding RNAs. Circ. Cardiovasc. Genet. 2017 , 10 , e001556. [ Google Scholar ] [ CrossRef ]
  • Hennessy, E.J.; Van Solingen, C.; Scacalossi, K.R.; Ouimet, M.; Afonso, M.; Prins, J.; Koelwyn, G.J.; Sharma, M.; Ramkhelawon, B.; Carpenter, S.; et al. The long noncoding RNA CHROME regulates cholesterol homeostasis in primates. Nat. Metab. 2019 , 1 , 98–110. [ Google Scholar ] [ CrossRef ]
  • Lu, W.; He, X.; Su, L.; Miao, J. Long Noncoding RNA-CERNA1 Stabilized Atherosclerotic Plaques in apolipoprotein E−/− Mice. J. Cardiovasc. Transl. Res. 2019 , 12 , 425–434. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Haemmig, S.; Yang, D.; Sun, X.; Das, D.; Ghaffari, S.; Molinaro, R.; Chen, L.; Deng, Y.; Freeman, D.; Moullan, N.; et al. Long noncoding RNA SNHG12 integrates a DNA-PK–mediated DNA damage response and vascular senescence. Sci. Transl. Med. 2020 , 12 , eaaw1868. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Hu, Y.-W.; Guo, F.-X.; Xu, Y.-J.; Li, P.; Lu, Z.-F.; McVey, D.G.; Zheng, L.; Wang, Q.; Ye, J.; Kang, C.-M.; et al. Long noncoding RNA NEXN-AS1 mitigates atherosclerosis by regulating the actin-binding protein NEXN. J. Clin. Investig. 2019 , 129 , 1115–1128. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Leisegang, M.S.; Bibli, S.-I.; Günther, S.; Pflüger-Müller, B.; A Oo, J.; Höper, C.; Seredinski, S.; Yekelchyk, M.; Schmitz-Rixen, T.; Schürmann, C.; et al. Pleiotropic effects of laminar flow and statins depend on the Krüppel-like factor-induced lncRNA MANTIS. Eur. Heart J. 2019 , 40 , 2523–2533. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Bell, R.D.; Long, X.; Lin, M.; Bergmann, J.H.; Nanda, V.; Cowan, S.L.; Zhou, Q.; Han, Y.; Spector, D.L.; Zheng, D.; et al. Identification and Initial Functional Characterization of a Human Vascular Cell–Enriched Long Noncoding RNA. Arterioscler. Thromb. Vasc. Biol. 2014 , 34 , 1249–1259. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Boulberdaa, M.; Scott, E.; Ballantyne, M.; Garcia, R.; Descamps, B.; Angelini, G.; Brittan, M.; Hunter, A.; McBride, M.; McClure, J.; et al. A Role for the Long Noncoding RNA SENCR in Commitment and Function of Endothelial Cells. Mol. Ther. 2016 , 24 , 978–990. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Zhang, Y.; Zheng, L.; Xu, B.-M.; Tang, W.-H.; Ye, Z.-D.; Huang, C.; Ma, X.; Zhao, J.-J.; Guo, F.-X.; Kang, C.-M.; et al. LncRNA-RP11-714G18.1 suppresses vascular cell migration via directly targeting LRP2BP. Immunol. Cell Biol. 2018 , 96 , 175–189. [ Google Scholar ] [ CrossRef ]
  • Ballantyne, M.D.; Pinel, K.; Dakin, R.; Vesey, A.T.; Diver, L.; MacKenzie, R.; Garcia, R.; Welsh, P.; Sattar, N.; Hamilton, G.; et al. Smooth Muscle Enriched Long Noncoding RNA (SMILR) Regulates Cell Proliferation. Circulation 2016 , 133 , 2050–2065. [ Google Scholar ] [ CrossRef ]
  • Lu, Y.-B.; Shi, C.; Yang, B.; Lu, Z.-F.; Wu, Y.-L.; Zhang, R.-Y.; He, X.; Li, L.-M.; Hu, B.; Hu, Y.-W.; et al. Long noncoding RNA ZNF800 suppresses proliferation and migration of vascular smooth muscle cells by upregulating PTEN and inhibiting AKT/mTOR/HIF-1α signaling. Atherosclerosis 2020 , 312 , 43–53. [ Google Scholar ] [ CrossRef ]
  • Shan, K.; Jiang, Q.; Wang, X.Q.; Wang, Y.N.Z.; Yang, H.; Yao, M.D.; Liu, C.; Li, X.M.; Yao, J.; Liu, B.; et al. Role of long non-coding RNA-RNCR3 in atherosclerosis-related vascular dysfunction. Cell Death Dis. 2016 , 7 , e2248. [ Google Scholar ] [ CrossRef ]
  • Wu, G.; Cai, J.; Han, Y.; Chen, J.; Huang, Z.-P.; Chen, C.; Cai, Y.; Huang, H.; Yang, Y.; Liu, Y.; et al. LincRNA-p21 Regulates Neointima Formation, Vascular Smooth Muscle Cell Proliferation, Apoptosis, and Atherosclerosis by Enhancing p53 Activity. Circulation 2014 , 130 , 1452–1465. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Khyzha, N.; Khor, M.; DiStefano, P.V.; Wang, L.; Matic, L.; Hedin, U.; Wilson, M.D.; Maegdefessel, L.; Fish, J.E. Regulation of CCL2 expression in human vascular endothelial cells by a neighboring divergently transcribed long noncoding RNA. Proc. Natl. Acad. Sci. USA 2019 , 116 , 16410–16419. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Weber, D.G.; Casjens, S.; Brik, A.; Raiko, I.; Lehnert, M.; Taeger, D.; Gleichenhagen, J.; Kollmeier, J.; Bauer, T.T.; Brüning, T.; et al. Circulating long non-coding RNA GAS5 (growth arrest-specific transcript 5) as a complement marker for the detection of malignant mesothelioma using liquid biopsies. Biomark. Res. 2020 , 8 , 15. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Ye, J.; Wang, C.; Wang, D.; Yuan, H. LncRBA GSA5, up-regulated by ox-LDL, aggravates inflammatory response and MMP expression in THP-1 macrophages by acting like a sponge for miR-221. Exp. Cell Res. 2018 , 369 , 348–355. [ Google Scholar ] [ CrossRef ]
  • Zhong, X.; Ma, X.; Zhang, L.; Li, Y.; Li, Y.; He, R. MIAT promotes proliferation and hinders apoptosis by modulating miR-181b/STAT3 axis in ox-LDL-induced atherosclerosis cell models. Biomed. Pharmacother. 2018 , 97 , 1078–1085. [ Google Scholar ] [ CrossRef ]
  • Li, H.; Liu, X.; Zhang, L.; Li, X. LncRNA BANCR facilitates vascular smooth muscle cell proliferation and migration through JNK pathway. Oncotarget 2017 , 8 , 114568–114575. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Holdt, L.M.; Beutner, F.; Scholz, M.; Gielen, S.; Gäbel, G.; Bergert, H.; Schuler, G.; Thiery, J.; Teupser, D. ANRIL Expression Is Associated With Atherosclerosis Risk at Chromosome 9p21. Arterioscler. Thromb. Vasc. Biol. 2010 , 30 , 620–627. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Holdt, L.M.; Teupser, D. Long Noncoding RNA ANRIL: Lnc-ing Genetic Variation at the Chromosome 9p21 Locus to Molecular Mechanisms of Atherosclerosis. Front. Cardiovasc. Med. 2018 , 5 , 145. [ Google Scholar ] [ CrossRef ]
  • Holdt, L.M.; Stahringer, A.; Sass, K.; Pichler, G.; Kulak, N.A.; Wilfert, W.; Kohlmaier, A.; Herbst, A.; Northoff, B.H.; Nicolaou, A.; et al. Circular non-coding RNA ANRIL modulates ribosomal RNA maturation and atherosclerosis in humans. Nat. Commun. 2016 , 7 , 12429. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Sonnenburg, J.L.; Bäckhed, F. Diet–microbiota interactions as moderators of human metabolism. Nature 2016 , 535 , 56–64. [ Google Scholar ] [ CrossRef ]
  • Gilbert, J.A.; Quinn, R.A.; Debelius, J.; Xu, Z.Z.; Morton, J.; Garg, N.; Jansson, J.K.; Dorrestein, P.C.; Knight, R. Microbiome-wide association studies link dynamic microbial consortia to disease. Nature 2016 , 535 , 94–103. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Karlsson, F.; Fåk, F.; Nookaew, I.; Tremaroli, V.; Fagerberg, B.; Petranovic, D.; Bäckhed, F.; Nielsen, J. Symptomatic atherosclerosis is associated with an altered gut metagenome. Nat. Commun. 2012 , 3 , 1245. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Jonsson, A.L.; Bäckhed, F. Role of gut microbiota in atherosclerosis. Nat. Rev. Cardiol. 2017 , 14 , 79–87. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Sharma, V.; Aggarwal, A. Helicobacter pylori: Does it add to risk of coronary artery disease. World J. Cardiol. 2015 , 7 , 19–25. [ Google Scholar ] [ CrossRef ]
  • Shen, X.; Li, L.; Sun, Z.; Zang, G.; Zhang, L.; Shao, C.; Wang, Z. Gut Microbiota and Atherosclerosis—Focusing on the Plaque Stability. Front. Cardiovasc. Med. 2021 , 8 , 668532. [ Google Scholar ] [ CrossRef ]
  • Clemente, J.C.; Ursell, L.K.; Parfrey, L.W.; Knight, R. The Impact of the Gut Microbiota on Human Health: An Integrative View. Cell 2012 , 148 , 1258–1270. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Li, J.; Jia, H.; Cai, X.; Zhong, H.; Feng, Q.; Sunagawa, S.; Arumugam, M.; Kultima, J.R.; Prifti, E.; Nielsen, T.; et al. An integrated catalog of reference genes in the human gut microbiome. Nat. Biotechnol. 2014 , 32 , 834–841. [ Google Scholar ] [ CrossRef ]
  • Tang, W.W.; Kitai, T.; Hazen, S.L. Gut Microbiota in Cardiovascular Health and Disease. Circ. Res. 2017 , 120 , 1183–1196. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Yamashiro, K.; Tanaka, R.; Urabe, T.; Ueno, Y.; Yamashiro, Y.; Nomoto, K.; Takahashi, T.; Tsuji, H.; Asahara, T.; Hattori, N. Gut dysbiosis is associated with metabolism and systemic inflammation in patients with ischemic stroke. PLoS ONE 2017 , 12 , e0171521, Correction in PLoS ONE 2017 , 12 , e0176062. [ Google Scholar ] [ CrossRef ]
  • Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in Gut Microbiota Control Metabolic Endotoxemia-Induced Inflammation in High-Fat Diet-Induced Obesity and Diabetes in Mice. Diabetes 2008 , 57 , 1470–1481. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Eshghjoo, S.; Jayaraman, A.; Sun, Y.; Alaniz, R.C. Microbiota-Mediated Immune Regulation in Atherosclerosis. Molecules 2021 , 26 , 179. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • He, Z.; Chen, Z.-Y. What Are Missing Parts in the Research Story of Trimethylamine-N-oxide (TMAO)? J. Agric. Food Chem. 2017 , 65 , 5227–5228. [ Google Scholar ] [ CrossRef ]
  • Sun, X.; Jiao, X.; Ma, Y.; Liu, Y.; Zhang, L.; He, Y.; Chen, Y. Trimethylamine N-oxide induces inflammation and endothelial dysfunction in human umbilical vein endothelial cells via activating ROS-TXNIP-NLRP3 inflammasome. Biochem. Biophys. Res. Commun. 2016 , 481 , 63–70. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Wang, Z.; Klipfell, E.; Bennett, B.J.; Koeth, R.; Levison, B.S.; DuGar, B.; Feldstein, A.E.; Britt, E.B.; Fu, X.; Chung, Y.-M.; et al. Gut Flora Metabolism of Phosphatidylcholine Promotes Cardiovascular Disease. Nature 2011 , 472 , 57–63. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Koren, O.; Spor, A.; Felin, J.; Fåk, F.; Stombaugh, J.; Tremaroli, V.; Behre, C.J.; Knight, R.; Fagerberg, B.; Ley, R.E.; et al. Human oral, gut, and plaque microbiota in patients with atherosclerosis. Proc. Natl. Acad. Sci. USA 2010 , 108 , 4592–4598. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • De Oliveira, C.; Watt, R.; Hamer, M. Toothbrushing, inflammation, and risk of cardiovascular disease: Results from Scottish Health Survey. BMJ 2010 , 340 , c2451. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Fåk, F.; Tremaroli, V.; Bergström, G.; Bäckhed, F. Oral microbiota in patients with atherosclerosis. Atherosclerosis 2015 , 243 , 573–578. [ Google Scholar ] [ CrossRef ]
  • Pietiäinen, M.; Liljestrand, J.M.; Kopra, E.; Pussinen, P.J. Mediators between oral dysbiosis and cardiovascular diseases. Eur. J. Oral Sci. 2018 , 126 , 26–36. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Yu, E.; Rimm, E.; Qi, L.; Rexrode, K.; Albert, C.; Sun, Q.; Willett, W.C.; Hu, F.B.; Manson, J.E. Diet, Lifestyle, Biomarkers, Genetic Factors, and Risk of Cardiovascular Disease in the Nurses’ Health Studies. Am. J. Public Heal. 2016 , 106 , 1616–1623. [ Google Scholar ] [ CrossRef ]
  • Berger, J.S.; Hochman, J.; Lobach, I.; Adelman, M.; Riles, T.S.; Rockman, C. Modifiable risk factor burden and the prevalence of peripheral artery disease in different vascular territories. J. Vasc. Surg. 2013 , 58 , 673–681.e1. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Perk, J.; De Backer, G.; Gohlke, H.; Graham, I.; Reiner, Z.; Verschuren, M.; Verschuren, M.; Albus, C.; Benlian, P.; Boysen, G.; et al. European Guidelines on cardiovascular disease prevention in clinical practice (version 2012). The Fifth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of nine societies and by invited experts). Eur. Heart J. 2012 , 33 , 1635–1701. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Galicia-Garcia, U.; Benito-Vicente, A.; Jebari, S.; Larrea-Sebal, A.; Siddiqi, H.; Uribe, K.B.; Ostolaza, H.; Martín, C. Pathophysiology of Type 2 Diabetes Mellitus. Int. J. Mol. Sci. 2020 , 21 , 6275. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Dawber, T.R.; Meadors, G.F.; Moore, F.E., Jr. Epidemiological Approaches to Heart Disease: The Framingham Study. Am. J. Public Health 1951 , 41 , 279–286. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Yusuf, S.; Hawken, S.; Ôunpuu, S.; Dans, T.; Avezum, A.; Lanas, F.; McQueen, M.; Budaj, A.; Pais, P.; Varigos, J.; et al. Effect of potentially modifiable risk factors associated with myocardial infarction in 52 countries (the INTERHEART study): Case-control study. Lancet 2004 , 364 , 937–952. [ Google Scholar ] [ CrossRef ]
  • Aggarwal, N.R.; Patel, H.N.; Mehta, L.S.; Sanghani, R.M.; Lundberg, G.P.; Lewis, S.J.; Mendelson, M.A.; Wood, M.J.; Volgman, A.S.; Mieres, J.H. Sex Differences in Ischemic Heart Disease. Circ. Cardiovasc. Qual. Outcomes 2018 , 11 , e004437. [ Google Scholar ] [ CrossRef ]
  • Okunrintemi, V.; Tibuakuu, M.; Virani, S.S.; Sperling, L.S.; Volgman, A.S.; Gulati, M.; Cho, L.; Leucker, T.M.; Blumenthal, R.S.; Michos, E.D. Sex Differences in the Age of Diagnosis for Cardiovascular Disease and Its Risk Factors Among US Adults: Trends From 2008 to 2017, the Medical Expenditure Panel Survey. J. Am. Heart Assoc. 2020 , 9 , e018764. [ Google Scholar ] [ CrossRef ]
  • Virani, S.S.; Alonso, A.; Benjamin, E.J.; Bittencourt, M.S.; Callaway, C.W.; Carson, A.P.; Chamberlain, A.M.; Chang, A.R.; Cheng, S.; Delling, F.N.; et al. Heart Disease and Stroke Statistics—2020 Update: A Report from the American Heart Association. Circulation 2020 , 141 , e139–e596. [ Google Scholar ] [ CrossRef ]
  • Man, J.J.; Beckman, J.A.; Jaffe, I.Z. Sex as a Biological Variable in Atherosclerosis. Circ. Res. 2020 , 126 , 1297–1319. [ Google Scholar ] [ CrossRef ]
  • World Health Organization Statistical Information System 2004. Available online: www.who.int/whosis (accessed on 21 November 2021).
  • Ramirez, F.D.; Hibbert, B. Letter by Ramirez and Hibbert Regarding Article, “Consideration of Sex Differences in Design and Reporting of Experimental Arterial Pathology Studies: A Statement from the Arteriosclerosis, Thrombosis, and Vascular Biology council. Arterioscler. Thromb. Vasc. Biol. 2018 , 38 , e99–e100. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Lu, H.S.; Schmidt, A.M.; Hegele, R.A.; Mackman, N.; Rader, D.J.; Weber, C.; Daugherty, A. Reporting Sex and Sex Differences in Preclinical Studies. Arterioscler. Thromb. Vasc. Biol. 2018 , 38 , e171–e184. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Lu, H.S.; Schmidt, A.M.; Hegele, R.A.; Mackman, N.; Rader, D.J.; Weber, C.; Daugherty, A. Annual Report on Sex in Preclinical Studies. Arterioscler. Thromb. Vasc. Biol. 2020 , 40 , e1–e9. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Libby, P.; Buring, J.E.; Badimon, L.; Hansson, G.K.; Deanfield, J.; Bittencourt, M.S.; Tokgözoğlu, L.; Lewis, E.F. Atherosclerosis. Nat. Rev. Dis. Primers 2019 , 5 , 56. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Sturlaugsdottir, R.; Aspelund, T.; Bjornsdottir, G.; Sigurdsson, S.; Thorsson, B.; Eiriksdottir, G.; Gudnason, V. Prevalence and determinants of carotid plaque in the cross-sectional REFINE-Reykjavik study. BMJ Open 2016 , 6 , e012457. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Lansky, A.J.; Ng, V.G.; Maehara, A.; Weisz, G.; Lerman, A.; Mintz, G.S.; De Bruyne, B.; Farhat, N.; Niess, G.; Jankovic, I.; et al. Gender and the Extent of Coronary Atherosclerosis, Plaque Composition, and Clinical Outcomes in Acute Coronary Syndromes. JACC Cardiovasc. Imaging 2012 , 5 , S62–S72. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Plank, F.; Beyer, C.; Friedrich, G.; Wildauer, M.; Feuchtner, G. Sex differences in coronary artery plaque composition detected by coronary computed tomography: Quantitative and qualitative analysis. Neth. Heart J. 2019 , 27 , 272–280. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kataoka, Y.; Puri, R.; Hammadah, M.; Duggal, B.; Uno, K.; Kapadia, S.R.; Tuzcu, E.M.; Nissen, S.E.; King, P.; Nicholls, S.J. Sex Differences in Nonculprit Coronary Plaque Microstructures on Frequency-Domain Optical Coherence Tomography in Acute Coronary Syndromes and Stable Coronary Artery Disease. Circ. Cardiovasc. Imaging 2016 , 9 , e004506. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Wendorff, C.; Wendorff, H.; Pelisek, J.; Tsantilas, P.; Zimmermann, A.; Zernecke, A.; Kuehnl, A.; Eckstein, H.-H. Carotid Plaque Morphology Is Significantly Associated With Sex, Age, and History of Neurological Symptoms. Stroke 2015 , 46 , 3213–3219. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Vrijenhoek, J.E.; Ruijter, H.M.D.; De Borst, G.J.; de Kleijn, D.P.; De Vries, J.-P.P.; Bots, M.L.; Van de Weg, S.M.; Vink, A.; Moll, F.L.; Pasterkamp, G. Sex Is Associated With the Presence of Atherosclerotic Plaque Hemorrhage and Modifies the Relation Between Plaque Hemorrhage and Cardiovascular Outcome. Stroke 2013 , 44 , 3318–3323. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Halvorsen, D.S.; Johnsen, S.H.; Mathiesen, E.B.; Njølstad, I. The Association between Inflammatory Markers and Carotid Atherosclerosis Is Sex Dependent: The Tromsø Study. Cerebrovasc. Dis. 2009 , 27 , 392–397. [ Google Scholar ] [ CrossRef ]
  • Jia, H.; Kubo, T.; Akasaka, T.; Yu, B. Optical Coherence Tomography Guidance in Management of Acute Coronary Syndrome Caused by Plaque Erosion. Circ. J. 2018 , 82 , 302–308. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Sato, T.; Minami, Y.; Asakura, K.; Katamine, M.; Kato, A.; Katsura, A.; Muramatsu, Y.; Kakizaki, R.; Nemoto, T.; Hashimoto, T.; et al. Age- and Gender-Related Differences in Coronary Lesion Plaque Composition on Optical Coherence Tomography. Circ. J. 2020 , 84 , 463–470. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Salami, J.A.; Warraich, H.; Valero-Elizondo, J.; Spatz, E.S.; Desai, N.R.; Rana, J.S.; Virani, S.S.; Blankstein, R.; Khera, A.; Blaha, M.J.; et al. National Trends in Statin Use and Expenditures in the US Adult Population From 2002 to 2013. JAMA Cardiol. 2017 , 2 , 56–65. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Peterson, E.D.; Delong, E.R.; Masoudi, F.A.; O’Brien, S.M.; Peterson, P.N.; Rumsfeld, J.S.; Shahian, D.M.; Shaw, R.E. ACCF/AHA 2010 Position Statement on Composite Measures for Healthcare Performance Assessment. Circulation 2010 , 121 , 1780–1791. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Nanna, M.G.; Wang, T.Y.; Xiang, Q.; Goldberg, A.C.; Robinson, J.G.; Roger, V.L.; Virani, S.S.; Wilson, P.W.; Louie, M.J.; Koren, A.; et al. Sex Differences in the Use of Statins in Community Practice. Circ. Cardiovasc. Qual. Outcomes 2019 , 12 , e005562. [ Google Scholar ] [ CrossRef ]
  • Tsujita, K.; Sugiyama, S.; Sumida, H.; Shimomura, H.; Yamashita, T.; Yamanaga, K.; Komura, N.; Sakamoto, K.; Oka, H.; Nakao, K.; et al. Impact of Dual Lipid-Lowering Strategy With Ezetimibe and Atorvastatin on Coronary Plaque Regression in Patients With Percutaneous Coronary Intervention. J. Am. Coll. Cardiol. 2015 , 66 , 495–507. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Dayspring, T.D.; Varvel, S.A.; Ghaedi, L.; Thiselton, D.L.; Bruton, J.; McConnell, J.P. Biomarkers of cholesterol homeostasis in a clinical laboratory database sample comprising 667,718 patients. J. Clin. Lipidol. 2015 , 9 , 807–816. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Fujino, A.; Mintz, G.S.; Matsumura, M.; Lee, T.; Kim, S.-Y.; Hoshino, M.; Usui, E.; Yonetsu, T.; Haag, E.S.; Shlofmitz, R.A.; et al. A new optical coherence tomography-based calcium scoring system to predict stent underexpansion. EuroIntervention 2018 , 13 , 2182–2189. [ Google Scholar ] [ CrossRef ]
  • Maejima, N.; Hibi, K.; Saka, K.; Akiyama, E.; Konishi, M.; Endo, M.; Iwahashi, N.; Tsukahara, K.; Kosuge, M.; Ebina, T.; et al. Relationship Between Thickness of Calcium on Optical Coherence Tomography and Crack Formation After Balloon Dilatation in Calcified Plaque Requiring Rotational Atherectomy. Circ. J. 2016 , 80 , 1413–1419. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kubo, T.; Shimamura, K.; Ino, Y.; Yamaguchi, T.; Matsuo, Y.; Shiono, Y.; Taruya, A.; Nishiguchi, T.; Shimokado, A.; Teraguchi, I.; et al. Superficial Calcium Fracture After PCI as Assessed by OCT. JACC Cardiovasc. Imaging 2015 , 8 , 1228–1229. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Kim, Y.; Wilkens, L.R.; Park, S.-Y.; Goodman, M.T.; Monroe, K.R.; Kolonel, L.N. Association between various sedentary behaviours and all-cause, cardiovascular disease and cancer mortality: The Multiethnic Cohort Study. Int. J. Epidemiol. 2013 , 42 , 1040–1056. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Meisinger, C.; Heier, M.; Löwel, H.; Schneider, A.; Döring, A. Sleep Duration and Sleep Complaints and Risk of Myocardial Infarction in Middle-aged Men and Women from the General Population: The MONICA/KORA Augsburg Cohort Study. Sleep 2007 , 30 , 1121–1127. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Pan, B.; Jin, X.; Jun, L.; Qiu, S.; Zheng, Q.; Pan, M. The relationship between smoking and stroke. Medicine 2019 , 98 , e14872. [ Google Scholar ] [ CrossRef ]
  • Larsson, S.C.; Burgess, S.; Michaëlsson, K. Smoking and stroke: A mendelian randomization study. Ann. Neurol. 2019 , 86 , 468–471. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • National Center for Chronic Disease Prevention and Health Promotion (US); Office on Smoking and Health. The Health Consequences of Smoking—50 Years of Progress: A Report of the Surgeon General ; Reports of the Surgeon General; Centers for Disease Control and Prevention (US): Atlanta, GA, USA, 2014.
  • Thun, M.J.; Carter, B.D.; Feskanich, D.; Freedman, N.D.; Prentice, R.; Lopez, A.D.; Hartge, P.; Gapstur, S.M. 50-Year Trends in Smoking-Related Mortality in the United States. N. Engl. J. Med. 2013 , 368 , 351–364. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Gambardella, J.; Sardu, C.; Sacra, C.; DEL Giudice, C.; Santulli, G. Quit smoking to outsmart atherogenesis: Molecular mechanisms underlying clinical evidence. Atherosclerosis 2017 , 257 , 242–245. [ Google Scholar ] [ CrossRef ]
  • Cheezum, M.K.; Kim, A.; Bittencourt, M.S.; Kassop, D.; Nissen, A.; Thomas, D.; Nguyen, B.; Glynn, R.J.; Shah, N.R.; Villines, T.C. Association of tobacco use and cessation with coronary atherosclerosis. Atherosclerosis 2017 , 257 , 201–207. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Stein, J.H.; Smith, S.S.; Hansen, K.M.; Korcarz, C.E.; Piper, M.E.; Fiore, M.C.; Baker, T.B. Longitudinal effects of smoking cessation on carotid artery atherosclerosis in contemporary smokers: The Wisconsin Smokers Health Study. Atherosclerosis 2020 , 315 , 62–67. [ Google Scholar ] [ CrossRef ]
  • Tollefson, A.K.; Oberley-Deegan, R.E.; Butterfield, K.T.; Nicks, M.E.; Weaver, M.R.; Remigio, L.K.; Decsesznak, J.; Chu, H.; Bratton, D.L.; Riches, D.W.; et al. Endogenous enzymes (NOX and ECSOD) regulate smoke-induced oxidative stress. Free Radic. Biol. Med. 2010 , 49 , 1937–1946. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Collins, T. Endothelial nuclear factor-kappa B and the initiation of the atherosclerotic lesion. Lab. Investig. 1993 , 68 , 499–508. [ Google Scholar ]
  • Barbieri, S.S.; Weksler, B.B. Tobacco smoke cooperates with interleukin-1β to alter β-catenin trafficking in vascular endothelium resulting in increased permeability and induction of cyclooxygenase-2 expression in vitro and in vivo. FASEB J. 2007 , 21 , 1831–1843. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Wang, C.-N.; Yang, G.-H.; Wang, Z.-Q.; Liu, C.-W.; Li, T.-J.; Lai, Z.-C.; Miao, S.-Y.; Wang, L.-F.; Liu, B. Role of perivascular adipose tissue in nicotine-induced endothelial cell inflammatory responses. Mol. Med. Rep. 2016 , 14 , 5713–5718. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Demirjian, L.; Abboud, R.T.; Li, H.; Duronio, V. Acute effect of cigarette smoke on TNF-α release by macrophages mediated through the erk1/2 pathway. Biochim. Et Biophys. Acta (BBA)-Mol. Basis Dis. 2006 , 1762 , 592–597. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Csordas, A.; Bernhard, D. The biology behind the atherothrombotic effects of cigarette smoke. Nat. Rev. Cardiol. 2013 , 10 , 219–230. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • Raveendran, M.; Senthil, D.; Utama, B.; Shen, Y.; Dudley, D.; Wang, J.; Zhang, Y.; Wang, X.L. Cigarette suppresses the expression of P4Hα and vascular collagen production. Biochem. Biophys. Res. Commun. 2004 , 323 , 592–598. [ Google Scholar ] [ CrossRef ] [ PubMed ]
  • O’Toole, T.E.; Zheng, Y.-T.; Hellmann, J.; Conklin, D.J.; Barski, O.; Bhatnagar, A. Acrolein activates matrix metalloproteinases by increasing reactive oxygen species in macrophages. Toxicol. Appl. Pharmacol. 2009 , 236 , 194–201. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Ljungberg, L.U.; Persson, K.; Eriksson, A.C.; Gréen, H.; Whiss, P.A. Effects of nicotine, its metabolites and tobacco extracts on human platelet function in vitro. Toxicol. Vitr. 2013 , 27 , 932–938. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Feinberg, M.W.; Moore, K.J. MicroRNA Regulation of Atherosclerosis. Circ. Res. 2016 , 118 , 703–720. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Yokoyama, Y.; Mise, N.; Suzuki, Y.; Tada-Oikawa, S.; Izuoka, K.; Zhang, L.; Zong, C.; Takai, A.; Yamada, Y.; Ichihara, S. MicroRNAs as Potential Mediators for Cigarette Smoking Induced Atherosclerosis. Int. J. Mol. Sci. 2018 , 19 , 1097. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Zhou, J.; Wang, K.C.; Wu, W.; Subramaniam, S.; Shyy, J.Y.; Chiu, J.J.; Li, J.Y.; Chien, S. MicroRNA-21 targets peroxisome proliferators-activated receptor-α in an autoregulatory loop to modulate flow-induced endothelial inflammation. Proc. Natl. Acad. Sci. USA 2011 , 108 , 10355–10360. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Bala, S.; Csak, T.; Saha, B.; Zatsiorsky, J.; Kodys, K.; Catalano, D.; Satishchandran, A.; Szabo, G. The pro-inflammatory effects of miR-155 promote liver fibrosis and alcohol-induced steatohepatitis. J. Hepatol. 2016 , 64 , 1378–1387. [ Google Scholar ] [ CrossRef ] [ Green Version ]
  • Gu, W.; Yao, L.; Li, L.; Zhang, J.; Place, A.T.; Minshall, R.D.; Liu, G. ICAM-1 regulates macrophage polarization by suppressing MCP-1 expression via miR-124 upregulation. Oncotarget 2017 , 8 , 111882–111901. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Cerutti, C.; Soblechero-Martin, P.; Wu, D.; Lopez-Ramirez, M.A.; De Vries, H.; Sharrack, B.; Male, D.K.; Romero, I.A. MicroRNA-155 contributes to shear-resistant leukocyte adhesion to human brain endothelium in vitro. Fluids Barriers CNS 2016 , 13 , 8. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Deshmane, S.L.; Kremlev, S.; Amini, S.; Sawaya, B.E. Monocyte Chemoattractant Protein-1 (MCP-1): An Overview. J. Interf. Cytokine Res. 2009 , 29 , 313–326. [ Google Scholar ] [ CrossRef ]
  • Weber, M.; Baker, M.B.; Moore, J.P.; Searles, C.D. MiR-21 is induced in endothelial cells by shear stress and modulates apoptosis and eNOS activity. Biochem. Biophys. Res. Commun. 2010 , 393 , 643–648. [ Google Scholar ] [ CrossRef ] [ PubMed ] [ Green Version ]
  • Liu, Y.; Pan, Q.; Zhao, Y.; He, C.; Bi, K.; Chen, Y.; Zhao, B.; Chen, Y.; Ma, X. MicroRNA-155 Regulates ROS Production, NO Generation, Apoptosis and Multiple Functions of Human Brain Microvessel Endothelial Cells Under Physiological and Pathological Conditions. J. Cell. Biochem. 2015 , 116 , 2870–2881. [ Google Scholar ] [ CrossRef ] [ PubMed ]

Click here to enlarge figure

LocationUpregulatedDownregulated
Coronary arteriesmiR-29, miR-100, miR-155, miR-199, miR-221, miR-363, miR-497, miR-508 and miR-181 [ , , ].miR-1273, miR-490, miR-24 and miR-1284 [ , , ].
Aorta, femoral, and carotid arteriesmiR-21, miR-34, miR-146 and miR-210 [ ].
Only in carotid plaques:
miR-15, miR-26, miR-30, miR-98, miR-125, miR-152, miR-181, miR-100, miR-127, miR-133, miR-145 and miR-422 [ , ].
Only in carotid plaques:
miR-520, miR-105 [ , ].
Cell LineAthero-ProtectivePro-Atherogenic
Endothelial cells miR-155 [ ]
miR-10a [ ], miR-31 and miR-17-3p [ ]
miR-146a [ ]
miR-181b [ ]
miR-92a [ , ]
miR-let-7g [ ]
miR-216a [ ]
Macrophages miR-146a [ ]
miR-125a [ ]
miR-223 [ ]
miR-125b [ ]
miR-342-p [ ]
miR-33 [ ]
VSMCs * miR-21 [ ]
miR-221 and miR-222 [ ]
miR-26a [ ]
miR-663 [ ]
miR-143, miR-145 and miR-1 [ ]
miR-29a and miR-24 [ ]
miR-133a [ ]
miR-15b and miR-16 [ ]
Anti-AtherogenicPro-Atherogenic
miR-24 [ ]miR-322 [ ]
miR-133a [ ]miR-712 [ ]
miR-29 [ , ] miR-494 [ ]
miR-21 [ ]miR-155 [ ]
miR-223 [ ]miR-365 [ ]
Athero-ProtectivePro-Atherogenic
MeXis [ ]
MALAT1 [ , ]
CERNA1 [ ]
SNHG12 [ ]
NEXN-AS1 [ ]
MANTIS [ ]
SENCR [ ]
RP11-714G18.1 [ ]
CHROME [ , ]
circANRIL [ ]
SMILR [ ]
CCL2 [ ]
GAS5 [ , ]
MIAT [ , ]
BANCR [ ]
ANRIL [ , ]
MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

Jebari-Benslaiman, S.; Galicia-García, U.; Larrea-Sebal, A.; Olaetxea, J.R.; Alloza, I.; Vandenbroeck, K.; Benito-Vicente, A.; Martín, C. Pathophysiology of Atherosclerosis. Int. J. Mol. Sci. 2022 , 23 , 3346. https://doi.org/10.3390/ijms23063346

Jebari-Benslaiman S, Galicia-García U, Larrea-Sebal A, Olaetxea JR, Alloza I, Vandenbroeck K, Benito-Vicente A, Martín C. Pathophysiology of Atherosclerosis. International Journal of Molecular Sciences . 2022; 23(6):3346. https://doi.org/10.3390/ijms23063346

Jebari-Benslaiman, Shifa, Unai Galicia-García, Asier Larrea-Sebal, Javier Rekondo Olaetxea, Iraide Alloza, Koen Vandenbroeck, Asier Benito-Vicente, and César Martín. 2022. "Pathophysiology of Atherosclerosis" International Journal of Molecular Sciences 23, no. 6: 3346. https://doi.org/10.3390/ijms23063346

Article Metrics

Article access statistics, further information, mdpi initiatives, follow mdpi.

MDPI

Subscribe to receive issue release notifications and newsletters from MDPI journals

CASE REPORT article

Case report: coronary atherosclerosis in a patient with long-standing very low ldl-c without lipid-lowering therapy.

\r\nGiorgio Mottola

  • 1 Department of Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, United States
  • 2 Division of Cardiology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
  • 3 Department of Radiology & Biomedical Imaging, Section of Vascular & Interventional Radiology, Yale School of Medicine, New Haven, CT, United States

Background: ApoB-containing lipoproteins including low-density lipoprotein cholesterol (LDL-C) are necessary for the development of atherosclerosis, and lifelong exposure to low serum levels of LDL-C have been associated with a substantial reduction of cardiovascular risk. Although plaque regression has been observed in patients with serum LDL-C less than 70–80 mg/dl on lipid-lowering therapy, an LDL-C level under which atherosclerosis cannot develop has not been established.

Case presentation: In this case we describe a 60-year-old man with well-controlled diabetes mellitus and hypertension who presented to the hospital after an acute stroke likely due to an atrial myxoma discovered on imaging. A coronary computed tomography angiography scan performed in preparation for the planned surgical myxoma resection revealed an anomalous origin of the right coronary artery as well as evidence of nonobstructive coronary atherosclerosis in the right coronary and non-anomalous left coronary system. Despite not having ever been on any lipid-lowering therapy, this patient was found to have low LDL-C levels (<40 mg/dl) during this admission and on routine laboratory data collected over the prior 16 years. His family history strongly suggested heterozygous familial hypobetalipoproteinemia as a possible diagnosis.

Conclusions: This case illustrates that even long-standing, very low levels of LDL-C may be insufficient to completely prevent atherosclerosis and emphasizes the importance of primordial prevention of all cardiovascular risk factors.

Introduction

While the biological mechanisms underlying the pathogenesis of atherosclerosis involve numerous factors, apolipoprotein B (ApoB)-containing lipoproteins inclusive of low-density lipoprotein cholesterol (LDL-C) are necessary for its development ( 1 ). Mendelian randomization studies and prospective studies of individuals with genetic variants leading to naturally low LDL-C levels have demonstrated that lifelong exposure to low LDL-C results in substantial reductions in risk of atherosclerotic cardiovascular disease (ASCVD) even in the presence of other risk factors ( 2 ). Atherosclerosis detected by coronary calcification can also occur in asymptomatic middle-aged adults without other risk factors and at serum LDL-C levels below 100 mg/dl ( 3 ). Reduction in atherogenic lipoproteins early in life is therefore central to primary prevention of ASCVD, though the serum levels of ApoB or LDL-C at which atherosclerosis fails to develop has not been firmly established and likely varies among individuals. It has been hypothesized that lifelong LDL-C of less than 30 mg/dl would be sufficient to completely prevent atherosclerosis ( 4 ), though studies of patients with established ASCVD have demonstrated plaque regression with serum LDL-C less than 70–80 mg/dl on lipid-lowering therapy ( 5 ).

Case description

A 60-year-old man presented to his local hospital with new-onset weakness, ataxia and paresthesia in the left upper extremity and was found to have an acute ischemic stroke based on MRI imaging showing multiple areas of acute cerebral and cerebellar infarction. A transthoracic echocardiogram was performed which showed a 4.3 × 2.0 cm mass in the left atrium ( Supplementary Video S1 ), consistent with an atrial myxoma which was thought to be the cause of his acute stroke. He was subsequently transferred to a tertiary medical center for further work-up and evaluation for cardiothoracic surgery. Prior to surgery for surgical myxoma resection, inpatient cardiology was consulted for preoperative evaluation. Aside from mild residual weakness, ataxia and paresthesia in his left upper extremity, the patient was asymptomatic and had been physically active. On physical exam, he was noted to have mild weakness of his left upper extremity (4+/5 strength in shoulder abduction) and trace dysmetria of the left hand. His cardiac exam was notable for a low-pitched sound heard early in diastole with no associated murmurs. His physical exam was otherwise unremarkable.

The patient reported being diagnosed with hypertension and type 2 diabetes mellitus approximately 7 years prior to admission. He was currently taking amlodipine and losartan and reported his systolic blood pressure was typically less than 130 mmHg at all recent clinic visits. At the time of diabetes diagnosis, his hemoglobin A1c (HbA1c) was 12.3 and he reported his diet contained high amounts of processed foods, saturated fat, and sugar-sweetened beverages. He was prescribed metformin and subsequently stopped drinking sugar-sweetened beverages, made other healthful changes to his diet and started exercising on a regular basis. He subsequently lost 20 pounds and his HbA1c after one year was markedly improved at 5.0. On admission his HbA1c was 6.2 with a body mass index of 27.9 Kg/m 2 . He was also noted to have hepatic steatosis on an outpatient liver ultrasound prior to admission. He denied ever using tobacco or drugs and rarely consumed alcohol.

On admission the patient's standard lipid panel showed a serum LDL-C of 32 mg/dl and a non-HDL-C of 46 mg/dl. The patient had never been on lipid-lowering therapy and reported being previously told he had naturally very low blood cholesterol levels, which was confirmed on outpatient labs over the prior 16 years ( Table 1 ). He recalled being told his mother and maternal grandmother had very low levels of LDL-C. He had no family history of premature ASCVD. Further testing during his hospitalization revealed an ApoB level of 35 mg/dl, directly measured LDL-C of 48 mg/dl, and lipoprotein(a) of 36 nmol/L.

www.frontiersin.org

Table 1 . Patient laboratory tests for Serum lipids and HbA1c.

As part of pre-operative planning for cardiac surgery, the patient underwent coronary computed tomography angiography (CCTA) which demonstrated an anomalous right coronary artery (RCA; Figures 1A,B ) at the origin of the left sinus with mild atherosclerotic plaques in the RCA. Mild atherosclerotic plaques were also noted in the left anterior descending artery (LAD) and left circumflex artery (LCx; Figures 1C,D ), with normal LAD and LCx origins. To assess for ischemia due to the anomalous RCA, pharmacologic myocardial perfusion imaging with positron emission tomography was performed which showed a small-sized, mild-intensity, reversible perfusion defect in the apical inferior wall which was thought to be clinically insignificant due to lack of any exertional symptoms.

www.frontiersin.org

Figure 1 . Patient's coronary computed tomographic angiography (CCTA). Anomalous origin of the right coronary artery (RCA; A ). Multiplanar reconstructions of the RCA ( B ), the left anterior descending artery (LAD; C ) and the left circumflex artery (LCx; D ).

The patient underwent successful robotic-assisted resection of the left atrial mass (confirmed to be a myxoma on pathology) with no intervention performed to the RCA. He was discharged on post-operative day 4 with rosuvastatin and metoprolol. He was also recommended for consideration of outpatient genetic screening for hypobetalipoproteinemia.

Despite chronic and presumably life-long levels of LDL-C typically less than 40 mg/dl, the patient presented here was found to have mild atherosclerosis in multiple coronary arteries by CCTA. Genetic testing was not performed in this patient, though his family history, long-standing low blood cholesterol levels and hepatic steatosis suggest his low LDL-C is likely due to heterozygous familial hypobetalipoproteinemia, a genetic cause of low LDL-C with a ∼1:1000 prevalence in most populations ( 6 ). Patients with familial hypobetalipoproteinemia typically have LDL-C levels between 20 and 50 mg/dl and though the APOB mutations often associated with this condition lead to substantially lower risk of ASCVD ( 7 ), prevalence of coronary atherosclerosis specifically in this population has not been well described.

Importantly, this case demonstrates that coronary atherosclerosis can occur even with long-standing LDL-C less than 40 mg/dl when other risk factors such as hypertension and diabetes are present and otherwise well controlled. Reports of atherosclerosis in individuals with lifelong very low LDL-C are extremely rare ( 8 ). The patient in this instance did have uncontrolled diabetes mellitus on initial diagnosis with a HbA1c of 12.3 in 2015, and due to lack of regular follow-up it is unclear how long that level of hyperglycemia may have occurred and contributed to plaque development. His serum ApoB was not assessed at that time and his LDL-C and non-HDL-C were higher than on all his other laboratory checks, though still at very low levels. Notably, his diabetes rapidly improved with only lifestyle changes and metformin. At the time of hospitalization, the patient's serum ApoB was concordantly low and his lipoprotein(a) level was also low, and therefore these factors are unlikely to explain development of atherosclerosis. There is little evidence available on whether anomalous coronary artery origin directly contributes to atherosclerosis; it has previously been shown that anomalous right-sided arteries with a retroaortic course may develop atherosclerosis earlier than non-anomalous arteries within the same patient. However, the case patient was also observed to have atherosclerosis in the non-anomalous left coronary system ( 9 ). To our knowledge, this is the first case report showing calcified coronary plaque on CCTA in a patient with long-standing LDL-C less than 40–50 mg/dl while not on lipid-lowering therapy.

This case indicates that even lifelong LDL-C at very low levels, as could theoretically be achieved with early initiation of statins and PCSK9 inhibition with medications or gene editing ( 10 – 12 ), may be insufficient to completely prevent atherosclerosis when other risk factors are present. Therefore, primordial prevention of risk factors, rather than only managing them after they have developed, remains critical to reducing prevalence of atherosclerosis and ASCVD across populations. However, atherosclerosis specifically in patients with familial hypobetalipoproteinemia is not well understood, and there may be unique factors that promote development in this population. Given that our observations were made from a single patient, definitive conclusions about coronary plaque development cannot be made from this report. Further study will be needed to determine what specific factors drive development of atherosclerosis in the setting of low LDL-C, including in patients with familial hypobetalipoproteinemia.

Data availability statement

The original contributions presented in the study are included in the article/ Supplementary Material , further inquiries can be directed to the corresponding author.

Ethics statement

Additional written informed consent was not required because we had obtained verbal informed consent from the patient to publish this case report prior to submission. Informed consent for publication was documented in the patient's medical record.

Author contributions

GM: Conceptualization, Data curation, Visualization, Writing – original draft, Writing – review & editing. FW: Conceptualization, Data–curation, Writing – review & editing. HM: Conceptualization, Visualization, Writing – review & editing. KF: Conceptualization, Supervision, Visualization, Writing – original draft, Writing – review editing, Funding acquisition.

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Conflict of interest

HM is a consultant for Inari Medical and receives an educational grant from the same company.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher's note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

Supplementary material

The Supplementary Material for this article can be found online at: https://www.frontiersin.org/articles/10.3389/fcvm.2023.1272944/full#supplementary-material

1. Ference BA, Ginsberg HN, Graham I, Ray KK, Packard CJ, Bruckert E, et al. Low-density lipoproteins cause atherosclerotic cardiovascular disease. 1. Evidence from genetic, epidemiologic, and clinical studies. A consensus statement from the European atherosclerosis society consensus panel. Eur Heart J . (2017) 38(32):2459–72. doi: 10.1093/eurheartj/ehx144

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Ference BA, Yoo W, Alesh I, Mahajan N, Mirowska KK, Mewada A, et al. Effect of long-term exposure to lower low-density lipoprotein cholesterol beginning early in life on the risk of coronary heart disease: a Mendelian randomization analysis. J Am Coll Cardiol . (2012) 60(25):2631–9. doi: 10.1016/j.jacc.2012.09.017

3. Fernández-Friera L, Fuster V, López-Melgar B, Oliva B, García-Ruiz JM, Mendiguren J, et al. Normal LDL-cholesterol levels are associated with subclinical atherosclerosis in the absence of risk factors. J Am Coll Cardiol . (2017) 70(24):2979–91. doi: 10.1016/j.jacc.2017.10.024

CrossRef Full Text | Google Scholar

4. Libby P, Buring JE, Badimon L, Hansson GK, Deanfield J, Bittencourt MS, et al. Atherosclerosis. Nature Reviews Disease Primers . (2019) 5(1):56. doi: 10.1038/s41572-019-0106-z

5. Ahmadi A, Argulian E, Leipsic J, Newby DE, Narula J. From subclinical atherosclerosis to plaque progression and acute coronary events: JACC state-of-the-art review. J Am Coll Cardiol . (2019) 74(12):1608–17. doi: 10.1016/j.jacc.2019.08.012

6. Olsson AG, Angelin B, Assmann G, Binder CJ, Björkhem I, Cedazo-Minguez A, et al. Can LDL cholesterol be too low? Possible risks of extremely low levels. J Intern Med . (2017) 281(6):534–53. doi: 10.1111/joim.12614

7. Welty FK. Hypobetalipoproteinemia and abetalipoproteinemia: liver disease and cardiovascular disease. Curr Opin Lipidol . (2020) 31(2):49–55. doi: 10.1097/MOL.0000000000000663

8. Welty FK, Ordovas J, Schaefer EJ, Wilson PW, Young SG. Identification and molecular analysis of two apoB gene mutations causing low plasma cholesterol levels. Circulation . (1995) 92(8):2036–40. doi: 10.1161/01.CIR.92.8.2036

9. Samarendra P, Kumari S, Hafeez M, Vasavada BC, Sacchi TJ. Anomalous circumflex coronary artery: benign or predisposed to selective atherosclerosis. Angiology . (2001) 52(8):521–6. doi: 10.1177/000331970105200803

10. Braunwald E. How to live to 100 before developing clinical coronary artery disease: a suggestion. Eur Heart J . (2022) 43(4):249–50. doi: 10.1093/eurheartj/ehab532

11. Lee RG, Mazzola AM, Braun MC, Platt C, Vafai SB, Kathiresan S, et al. Efficacy and safety of an investigational single-course CRISPR base-editing therapy targeting PCSK9 in nonhuman primate and mouse models. Circulation . (2023) 147(3):242–53. doi: 10.1161/CIRCULATIONAHA.122.062132

12. Oostveen RF, Khera AV, Kathiresan S, Stroes ESG, Fitzgerald K, Harms MJ, et al. New approaches for targeting PCSK9: small-interfering ribonucleic acid and genome editing. Arterioscler Thromb Vasc Biol . (2023) 43(7):1081–92. doi: 10.1161/ATVBAHA.122.317963

Keywords: LDL-C, atherosclerosis, familial hypobetalipoproteinemia (FHBL), primordial prevention of CVD, coronary artery disease

Citation: Mottola G, Welty FK, Mojibian HR and Faridi KF (2023) Case report: Coronary atherosclerosis in a patient with long-standing very low LDL-C without lipid-lowering therapy. Front. Cardiovasc. Med. 10:1272944. doi: 10.3389/fcvm.2023.1272944

Received: 4 August 2023; Accepted: 8 September 2023; Published: 19 September 2023.

Reviewed by:

© 2023 Mottola, Welty, Mojibian and Faridi. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY) . The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Kamil F. Faridi [email protected]

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.

Renew Your Membership

Create Free Account or

atherosclerosis case study pdf

  • Acute Coronary Syndromes
  • Anticoagulation Management
  • Arrhythmias and Clinical EP
  • Cardiac Surgery
  • Cardio-Oncology
  • Cardiovascular Care Team
  • Congenital Heart Disease and Pediatric Cardiology
  • COVID-19 Hub
  • Diabetes and Cardiometabolic Disease
  • Dyslipidemia
  • Geriatric Cardiology
  • Heart Failure and Cardiomyopathies
  • Invasive Cardiovascular Angiography and Intervention
  • Noninvasive Imaging
  • Pericardial Disease
  • Pulmonary Hypertension and Venous Thromboembolism
  • Sports and Exercise Cardiology
  • Stable Ischemic Heart Disease
  • Valvular Heart Disease
  • Vascular Medicine
  • Clinical Updates & Discoveries
  • Advocacy & Policy
  • Perspectives & Analysis
  • Meeting Coverage
  • ACC Member Publications
  • ACC Podcasts
  • View All Cardiology Updates
  • Earn Credit
  • View the Education Catalog
  • ACC Anywhere: The Cardiology Video Library
  • CardioSource Plus for Institutions and Practices
  • ECG Drill and Practice
  • Heart Songs
  • Nuclear Cardiology
  • Online Courses
  • Collaborative Maintenance Pathway (CMP)
  • Understanding MOC
  • Image and Slide Gallery
  • Annual Scientific Session and Related Events
  • Chapter Meetings
  • Live Meetings
  • Live Meetings - International
  • Webinars - Live
  • Webinars - OnDemand
  • Certificates and Certifications
  • ACC Accreditation Services
  • ACC Quality Improvement for Institutions Program
  • CardioSmart
  • National Cardiovascular Data Registry (NCDR)
  • Advocacy at the ACC
  • Cardiology as a Career Path
  • Cardiology Careers
  • Cardiovascular Buyers Guide
  • Clinical Solutions
  • Clinician Well-Being Portal
  • Diversity and Inclusion
  • Infographics
  • Innovation Program
  • Mobile and Web Apps

A 52-Year-Old Man With Atherosclerosis

A 52-year-old executive was referred to our clinic for risk factor management after undergoing coronary computed tomography angiography (CTA) as part of an Executive Physical. He has no history of coronary artery disease and exercises regularly without experiencing anginal symptoms.

His family history is notable for a myocardial infarction (MI) in his father at the age of 52 years. He is a lifelong non-smoker. He does not take medications.

His blood pressure was 110/75. His exam was notable for being overweight with a BMI of 27, but was otherwise unremarkable.

His total cholesterol is 206 mg/dL, HDL-C is 46 mg/dL, triglycerides are 178 mg/dL, calculated LDL-C is 124 mg/dL, and non HDL-C is 160 mg/dL. His fasting glucose is 86 mg/dL. His Hgb A1c is 5.6%.

His 10-year risk based on the 2013 ACC/AHA pooled ASCVD risk estimator is 3.7%.

His coronary artery calcium (CAC) score is 120, which places him in the 87th percentile for his age, gender, and ethnicity.

His coronary CTA shows the following in the proximal LAD:

In addition to maximizing therapeutic lifestyle changes (exercise, weight loss), what is the next step in this patient’s management?

  • A. There is nothing more to do since his estimated 10-year risk is low.
  • B. Maximize risk factor modification by starting a low-dose aspirin and statin therapy.
  • C. Pursue stress testing.
  • D. Pursue coronary catheterization.
  • E. B and C.

Show Answer

The correct answer is: B. Maximize risk factor modification by starting a low-dose aspirin and statin therapy.

Atherosclerosis is necessary for nearly all coronary events. The development of atherosclerosis is multifactorial. There is significant heterogeneity in the contribution of common traditional modifiable risk factors including apolipoprotein B (apoB)-containing lipoproteins, smoking, diabetes, hypertension, and sedentary lifestyle to atherogenesis. 1 In participants from the Multi-Ethnic Study of Atherosclerosis (MESA) without atherosclerosis as measured by coronary artery calcium (CAC), even the presence of multiple modifiable risk factors was associated with low event rates (3.1%). In contrast, in those with elevated CAC and no modifiable risk factors, the event rates were significantly higher at nearly 11%. 1

While risk estimators are improving in accuracy, 2,3 the presence of subclinical atherosclerosis (primarily by CAC) has consistently been an additive predictor of coronary events in individuals and further discriminates between those at higher and lower risk for events. 4-6 The case patient is at elevated risk because of the burden of subclinical atherosclerosis and, therefore, answer choice A is incorrect.

The recent 2013 ACC/AHA guidelines on cholesterol treatment take a risk-based approach to recommendations for statin therapy. 7 The patient in this case has a low estimated 10-year risk at 3.7%. The current guidelines suggest a risk discussion in this case based on the patient's family history of premature coronary heart disease (CHD). Subclinical atherosclerosis imaging by CAC scanning can help with this discussion. 8

Recently, eight-year follow-up from the Dallas Heart Study showed that among participants with a family history of MI, those without CAC experienced a significantly lower CHD event rate of 1.9% compared to 8.8% in those with any CAC. 9 The case patient has a CAC score >100 that places him above the 75th percentile for his age, gender, and ethnicity. We would recommend moderate- or high-intensity statin therapy during a risk discussion based on an estimated 10-year event rate that exceeds 7.5%. Furthermore, recent data support the use of aspirin in those with CAC >100. Therefore, answer choice B is the correct answer.

Subclinical atherosclerosis imaging with CAC scanning has been endorsed by several committees to assist with risk assessment. The 2010 ACC/AHA risk assessment guidelines gave CAC scanning a IIA recommendation in those deemed to be at intermediate risk for CHD events. 11 In the 2010 appropriate use criteria for cardiac CT endorsed by multiple societies, CAC scanning was deemed appropriate among low-risk asymptomatic patients with a family history of premature CHD in addition to those at intermediate risk. 12 Most recently, the 2013 ACC/AHA risk assessment guidelines gave CAC scoring a IIB recommendation. Specifically, the committee suggests that if there is uncertainty about whether to start pharmacotherapy after risk estimation, then CAC scoring could be considered. 2

An important consideration in this case is the use of CTA to identify subclinical atherosclerosis. While coronary CTA is a more specific and sensitive test for atherosclerosis, there is no evidence that CTA adds significantly to CAC scanning in an asymptomatic population. However, CTA may identify vulnerable features of plaque that are not picked up by CAC scanning, such as those present in the case example. 13 Motoyama et al. identified high-risk features for acute coronary syndromes (ACS) in asymptomatic subjects on CTA including positive remodeling and low-attenuation plaque, in addition to spotty calcification in those presenting with ACS. 14,15 A small preliminary study suggested a benefit of statin therapy on plaque volume and the amount of low-attenuation plaque. 16 The role of CTA in asymptomatic, primary prevention patients is actively under investigation. 17 The benefits of CTA should be weighed against exposure to contrast, expense, and the need to train readers. With rapidly advancing technology, CTAs can now be performed with the equivalent radiation exposure of two mammograms. 18

Currently, the use of CTA is useful in appropriate symptomatic patients, particularly in chest pain protocols in the emergency department. 19 Many of these patients have mild or moderate, non-obstructive atherosclerosis that is unlikely to be the cause of their presenting symptoms, but should be managed with aggressive preventive therapies. In symptomatic patients from the CONFIRM registry, there is an increased hazard of mortality in those with non-obstructive atherosclerosis on CTA compared to those without atherosclerosis on CTA. 20

Some have advocated for stress testing in those with CAC scores > 400; however, the patient in this case does not meet this criteria. 21,22 Therefore, answer choices C and D are incorrect as the patient is asymptomatic.

  • Silverman MG, Blaha MJ, Krumholz HM, et al. Impact of coronary artery calcium on coronary heart disease events in individuals at the extremes of traditional risk factor burden: the Multi-Ethnic Study of Atherosclerosis. Eur Heart J. 2013 Dec 13 [Epub ahead of print].
  • Goff DC, Jr., Lloyd-Jones DM, Bennett G, et al. 2013 ACC/AHA guideline on the assessment of cardiovascular risk: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol 2014;63:2935-59.
  • Muntner P, Colantonio LD, Cushman M, et al. Validation of the atherosclerotic cardiovascular disease Pooled Cohort risk equations. JAMA 2014;311:1406-15.
  • Polonsky TS, McClelland RL, Jorgensen NW, et al. Coronary artery calcium score and risk classification for coronary heart disease prediction. JAMA 2010;303:1610-6.
  • Erbel R, Möhlenkamp S, Moebus S, et al. Coronary risk stratification, discrimination, and reclassification improvement based on quantification of subclinical coronary atherosclerosis: the Heinz Nixdorf Recall study. J Am Coll Cardiol 2010;56:1397-406.
  • Yeboah J, McClelland RL, Polonsky TS, et al. Comparison of novel risk markers for improvement in cardiovascular risk assessment in intermediate-risk individuals. JAMA 2012;308:788-95.
  • Stone NJ, Robinson J, Lichtenstein AH, et al. 2013 ACC/AHA guideline on the treatment of blood cholesterol to reduce atherosclerotic cardiovascular risk in adults: a report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol 2014;63:2889–934.
  • Nasir K, Budoff MJ, Wong ND, et al. Family history of premature coronary heart disease and coronary artery calcification: Multi-Ethnic Study of Atherosclerosis (MESA). Circulation 2007;116:619-26.
  • Paixao AR, Berry JD, Neeland IJ, et al. Coronary artery calcification and family history of myocardial infarction in the Dallas Heart Study. JACC Cardiovasc Imaging . 2014 June [Epub ahead of print].
  • Miedema MD, Duprez DA, Misialek JR, et al. Use of coronary artery calcium testing to guide aspirin utilization for primary prevention: estimates from the multi-ethnic study of atherosclerosis. Circ Cardiovasc Qual Outcomes 2014;7:453-60.
  • Greenland P, Alpert JS, Beller GA, et al. 2010 ACCF/AHA guideline for assessment of cardiovascular risk in asymptomatic adults: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol 2010;56:e50-103.
  • Taylor AJ, Cerqueira M, Hodgson JM, et al. ACCF/SCCT/ACR/AHA/ASE/ASNC/NASCI/SCAI/SCMR 2010 appropriate use criteria for cardiac computed tomography. a report of the American College of Cardiology Foundation Appropriate Use Criteria Task Force, the Society of Cardiovascular Computed Tomography, the American College of Radiology, the American Heart Association, the American Society of Echocardiography, the American Society of Nuclear Cardiology, the North American Society for Cardiovascular Imaging, the Society for Cardiovascular Angiography and Interventions, and the Society for Cardiovascular Magnetic Resonance. J Am Coll Cardiol 2010;56:1864-94.
  • Voros S, Rinehart S, Qian Z, et al. Coronary atherosclerosis imaging by coronary CT angiography: current status, correlation with intravascular interrogation and meta-analysis. JACC Cardiovasc Imaging 2011;4:537-48.
  • Motoyama S, Sarai M, Harigaya H, et al. Computed tomographic angiography characteristics of atherosclerotic plaques subsequently resulting in acute coronary syndrome. J Am Coll Cardiol 2009;54:49-57.
  • Motoyama S, Kondo T, Sarai M et al. Multislice computed tomographic characteristics of coronary lesions in acute coronary syndromes. J Am Coll Cardiol 2007;50:319-26.
  • Inoue K, Motoyama S, Sarai M, et al. Serial coronary CT angiography-verified changes in plaque characteristics as an end point: evaluation of effect of statin intervention. JACC Cardiovasc Imaging 2010;3:691-8.
  • U.S. National Institutes of Health.Detection of Subclinical Atherosclerosis in Asymptomatic Individuals (Decide CTA). (ClinicalTrials.gov website). 2009-2014. Available at: http://clinicaltrials.gov/ct2/show/NCT00862056?term=DECIDE-CTA . Accessed June 22, 2014.
  • Achenbach S, Marwan M, Ropers, D et al. Coronary computed tomography angiography with a consistent dose below 1 mSv using prospectively electrocardiogram-triggered high-pitch spiral acquisition. Eur Heart J 2010;31:340-6.
  • Hoffmann U, Truong QA, Schoenfeld DA, et al. Coronary CT angiography versus standard evaluation in acute chest pain. N Engl J Med 2012;367:299-308.
  • Min JK, Dunning A, Lin FY, et al. Age- and sex-related differences in all-cause mortality risk based on coronary computed tomography angiography findings results from the International Multicenter CONFIRM (Coronary CT Angiography Evaluation for Clinical Outcomes: An International Multicenter Registry) of 23,854 patients without known coronary artery disease. J Am Coll Cardiol 2011;58:849-60.
  • Hendel RC, Berman DS, Di Carli MF, et al. ACCF/ASNC/ACR/AHA/ASE/SCCT/SCMR/SNM 2009 appropriate use criteria for cardiac radionuclide imaging: a report of the American College of Cardiology Foundation Appropriate Use Criteria Task Force, the American Society of Nuclear Cardiology, the American College of Radiology, the American Heart Association, the American Society of Echocardiography, the Society of Cardiovascular Computed Tomography, the Society for Cardiovascular Magnetic Resonance, and the Society of Nuclear Medicine. J Am Coll Cardiol 2009;53:2201-29.
  • Berman DS, Hachamovitch R, Shaw LJ, et al. Roles of nuclear cardiology, cardiac computed tomography, and cardiac magnetic resonance: Noninvasive risk stratification and a conceptual framework for the selection of noninvasive imaging tests in patients with known or suspected coronary artery disease. J Nucl Med 2006;47:1107-18.

You must be logged in to save to your library.

Jacc journals on acc.org.

  • JACC: Advances
  • JACC: Basic to Translational Science
  • JACC: CardioOncology
  • JACC: Cardiovascular Imaging
  • JACC: Cardiovascular Interventions
  • JACC: Case Reports
  • JACC: Clinical Electrophysiology
  • JACC: Heart Failure
  • Current Members
  • Campaign for the Future
  • Become a Member
  • Renew Your Membership
  • Member Benefits and Resources
  • Member Sections
  • ACC Member Directory
  • ACC Innovation Program
  • Our Strategic Direction
  • Our History
  • Our Bylaws and Code of Ethics
  • Leadership and Governance
  • Annual Report
  • Industry Relations
  • Support the ACC
  • Jobs at the ACC
  • Press Releases
  • Social Media
  • Book Our Conference Center

Clinical Topics

  • Chronic Angina
  • Congenital Heart Disease and     Pediatric Cardiology
  • Diabetes and Cardiometabolic     Disease
  • Hypertriglyceridemia
  • Invasive Cardiovascular Angiography    and Intervention
  • Pulmonary Hypertension and Venous     Thromboembolism

Latest in Cardiology

Education and meetings.

  • Online Learning Catalog
  • Products and Resources
  • Annual Scientific Session

Tools and Practice Support

  • Quality Improvement for Institutions
  • Accreditation Services
  • Practice Solutions

YouTube

Heart House

  • 2400 N St. NW
  • Washington , DC 20037
  • Contact Member Care
  • Phone: 1-202-375-6000
  • Toll Free: 1-800-253-4636
  • Fax: 1-202-375-6842
  • Media Center
  • Advertising & Sponsorship Policy
  • Clinical Content Disclaimer
  • Editorial Board
  • Privacy Policy
  • Registered User Agreement
  • Terms of Service
  • Cookie Policy

© 2024 American College of Cardiology Foundation. All rights reserved.

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • View all journals
  • Explore content
  • About the journal
  • Publish with us
  • Sign up for alerts
  • Open access
  • Published: 14 August 2024

Nonlinear dynamics of multi-omics profiles during human aging

  • Xiaotao Shen   ORCID: orcid.org/0000-0002-9608-9964 1 , 2 , 3   na1 ,
  • Chuchu Wang   ORCID: orcid.org/0000-0003-2015-7331 4 , 5   na1 ,
  • Xin Zhou   ORCID: orcid.org/0000-0001-8089-4507 1 , 6 ,
  • Wenyu Zhou 1 ,
  • Daniel Hornburg   ORCID: orcid.org/0000-0002-6618-7774 1 ,
  • Si Wu 1 &
  • Michael P. Snyder   ORCID: orcid.org/0000-0003-0784-7987 1 , 6  

Nature Aging ( 2024 ) Cite this article

110k Accesses

2637 Altmetric

Metrics details

  • Biochemistry
  • Systems biology

Aging is a complex process associated with nearly all diseases. Understanding the molecular changes underlying aging and identifying therapeutic targets for aging-related diseases are crucial for increasing healthspan. Although many studies have explored linear changes during aging, the prevalence of aging-related diseases and mortality risk accelerates after specific time points, indicating the importance of studying nonlinear molecular changes. In this study, we performed comprehensive multi-omics profiling on a longitudinal human cohort of 108 participants, aged between 25 years and 75 years. The participants resided in California, United States, and were tracked for a median period of 1.7 years, with a maximum follow-up duration of 6.8 years. The analysis revealed consistent nonlinear patterns in molecular markers of aging, with substantial dysregulation occurring at two major periods occurring at approximately 44 years and 60 years of chronological age. Distinct molecules and functional pathways associated with these periods were also identified, such as immune regulation and carbohydrate metabolism that shifted during the 60-year transition and cardiovascular disease, lipid and alcohol metabolism changes at the 40-year transition. Overall, this research demonstrates that functions and risks of aging-related diseases change nonlinearly across the human lifespan and provides insights into the molecular and biological pathways involved in these changes.

Similar content being viewed by others

atherosclerosis case study pdf

Personal aging markers and ageotypes revealed by deep longitudinal profiling

atherosclerosis case study pdf

Considerations for reproducible omics in aging research

atherosclerosis case study pdf

Principal component-based clinical aging clocks identify signatures of healthy aging and targets for clinical intervention

Aging is a complex and multifactorial process of physiological changes strongly associated with various human diseases, including cardiovascular diseases (CVDs), diabetes, neurodegeneration and cancer 1 . The alterations of molecules (including transcripts, proteins, metabolites and cytokines) are critically important to understand the underlying mechanism of aging and discover potential therapeutic targets for aging-related diseases. Recently, the development of high-throughput omics technologies has enabled researchers to study molecular changes at the system level 2 . A growing number of studies have comprehensively explored the molecular changes that occur during aging using omics profiling 3 , 4 , and most focus on linear changes 5 . It is widely recognized that the occurrence of aging-related diseases does not follow a proportional increase with age. Instead, the risk of these diseases accelerates at specific points throughout the human lifespan 6 . For example, in the United States, the prevalence of CVDs (encompassing atherosclerosis, stroke and myocardial infarction) is approximately 40% between the ages of 40 and 59, increases to about 75% between 60 and 79 and reaches approximately 86% in individuals older than 80 years 7 . Similarly, also in the United States, the prevalence of neurodegenerative diseases, such as Parkinson’s disease and Alzheimer’s disease, exhibits an upward trend as well as human aging progresses, with distinct turning points occurring around the ages of 40 and 65, respectively 8 , 9 , 10 . Some studies also found that brain aging followed an accelerated decline in flies 11 and chimpanzees 12 that lived past middle age and advanced age.

The observation of a nonlinear increase in the prevalence of aging-related diseases implies that the process of human aging is not a simple linear trend. Consequently, investigating the nonlinear changes in molecules will likely reveal previously unreported molecular signatures and mechanistic insights. Some studies examined the nonlinear alterations of molecules during human aging 13 . For instance, nonlinear changes in RNA and protein expression related to aging have been documented 14 , 15 , 16 . Moreover, certain DNA methylation sites have exhibited nonlinear changes in methylation intensity during aging, following a power law pattern 17 . Li et al. 18 identified the 30s and 50s as transitional periods during women’s aging. Although aging patterns are thought to reflect the underlying biological mechanisms, the comprehensive landscape of nonlinear changes of different types of molecules during aging remains largely unexplored. Remarkably, the global monitoring of nonlinear changing molecular profiles throughout human aging has yet to be fully used to extract basic insights into the biology of aging.

In the present study, we conducted a comprehensive deep multi-omics profiling on a longitudinal human cohort comprising 108 individuals aged from 25 years to 75 years. The cohort was followed over a span of several years (median, 1.7 years), with the longest monitoring period for a single participant reaching 6.8 years (2,471 days). Various types of omics data were collected from the participants’ biological samples, including transcriptomics, proteomics, metabolomics, cytokines, clinical laboratory tests, lipidomics, stool microbiome, skin microbiome, oral microbiome and nasal microbiome. The investigation explored the changes occurring across different omics profiles during human aging. Remarkably, many molecular markers and biological pathways exhibited a nonlinear pattern throughout the aging process, thereby providing valuable insight into periods of dramatic alterations during human aging.

Most of the molecules change nonlinearly during aging

We collected longitudinal biological samples from 108 participants over several years, with a median tracking period of 1.7 years and a maximum period of 6.8 years, and conducted multi-omics profiling on the samples. The participants were sampled every 3–6 months while healthy and had diverse ethnic backgrounds and ages ranging from 25 years to 75 years (median, 55.7 years). The participants’ body mass index (BMI) ranged from 19.1 kg m −2 to 40.8 kg m −2 (median, 28.2 kg m −2 ). Among the participants, 51.9% were female (Fig. 1a and Extended Data Fig. 1a–d ). For each visit, we collected blood, stool, skin swab, oral swab and nasal swab samples. In total, 5,405 biological samples (including 1,440 blood samples, 926 stool samples, 1,116 skin swab samples, 1,001 oral swab samples and 922 nasal swab samples) were collected. The biological samples were used for multi-omics data acquisition (including transcriptomics from peripheral blood mononuclear cells (PBMCs), proteomics from plasma, metabolomics from plasma, cytokines from plasma, clinical laboratory tests from plasma, lipidomics from plasma, stool microbiome, skin microbiome, oral microbiome and nasal microbiome; Methods ). In total, 135,239 biological features (including 10,346 transcripts, 302 proteins, 814 metabolites, 66 cytokines, 51 clinical laboratory tests, 846 lipids, 52,460 gut microbiome taxons, 8,947 skin microbiome taxons, 8,947 oral microbiome taxons and 52,460 nasal microbiome taxons) were acquired, resulting in 246,507,456,400 data points (Fig. 1b and Extended Data Fig. 1e,f ). The average sampling period and number of samples for each participant were 626 days and 47 samples, respectively. Notably, one participant was deeply monitored for 6.8 years (2,471 days), during which 367 samples were collected (Fig. 1c ). Overall, this extensive and longitudinal multi-omics dataset enables us to examine the molecular changes that occur during the human aging process. The detailed characteristics of all participants are provided in the Supplementary Data . For each participant, the omics data were aggregated and averaged across all healthy samples to represent the individual’s mean value, as detailed in the Methods section. Compared to cross-sectional cohorts, which have only a one-time point sample from each participant, our longitudinal dataset, which includes multiple time point samples from each participant, is more robust for detecting complex aging-related changes in molecules and functions. This is because analysis of multi-time point samples can detect participants’ baseline and robustly evaluate individuals’ longitudinal molecular changes.

figure 1

a , The demographics of the 108 participants in the study are presented. b , Sample collection and multi-omics data acquisition of the cohort. Four types of biological samples were collected, and 10 types of omics data were acquired. c , Collection time range and sample numbers for each participant. The top x axis represents the collection range for each participant (read line), and the bottom x axis represents the sample number for each participant (bar plot). Bars are color-coded by omics type. d , Significantly changed molecules and microbes during aging were detected using the Spearman correlation approach ( P  < 0.05). The P values were not adjusted ( Methods ). Dots are color-coded by omics type. e , Differential expressional molecules/microbes in different age ranges compared to baseline (25–40 years old, two-sided Wilcoxon test, P  < 0.05). The P values were not adjusted ( Methods ). f , The linear changing molecules comprised only a small part of dysregulated molecules in at least one age range. g , Heatmap depicting the nonlinear changing molecules and microbes during human aging.

We included samples only from healthy visits and adjusted for confounding factors (for example, BMI, sex, insulin resistance/insulin sensitivity (IRIS) and ethnicity; Extended Data Fig. 1a–d ), allowing us to discern the molecules and microbes genuinely associated with aging ( Methods ). Two common and traditional approaches, linear regression and Spearman correlation, were first used to identify the linear changing molecules during human aging 5 . The linear regression method is commonly used for linear changing molecules. As expected, both approaches have very high consistent results for each type of omics data (Supplementary Fig. 1a ). For convenience, the Spearman correlation approach was used in the analysis. Interestingly, only a small portion of all the molecules and microbes (749 out of 11,305, 6.6%; only genus level was used for microbiome data; Methods ) linearly changed during human aging (Fig. 1d and Supplementary Fig. 1b ), consistent with our previous studies 5 ( Methods ). Next, we examined nonlinear effects by categorizing all participants into distinct age stages according to their ages and investigated the dysregulated molecules within each age stage compared to the baseline (25–40 years old; Methods ). Interestingly, using this approach, 81.03% of molecules (9,106 out of 11,305) exhibited changes in at least one age stage compared to the baseline (Fig. 1e and Extended Data Fig. 2a ). Remarkably, the percentage of linear changing molecules was relatively small compared to the overall dysregulated molecules during aging (mean, 16.2%) (Fig. 1f and Extended Data Fig. 2b ). To corroborate our findings, we employed a permutation approach to calculate permutated P values, which yielded consistent results ( Methods ). The heatmap depicting all dysregulated molecules also clearly illustrates pronounced nonlinear changes (Fig. 1g ). Taken together, these findings strongly suggest that a substantial number of molecules and microbes undergo nonlinear changes throughout human aging.

Clustering reveals nonlinear multi-omics changes during aging

Next, we assessed whether the multi-omics data collected from the longitudinal cohort could serve as reliable indicators of the aging process. Our analysis revealed a substantial correlation between a significant proportion of the omics data and the ages of the participants (Fig. 2a ). Particularly noteworthy was the observation that, among all the omics data examined, metabolomics, cytokine and oral microbiome data displayed the strongest association with age (Fig. 2a and Extended Data Fig. 3a–c ). Partial least squares (PLS) regression was further used to compare the strength of the age effect across different omics data types. The results are consistent with the results presented above in Fig. 2a ( Methods ). These findings suggest the potential utility of these datasets as indicators of the aging process while acknowledging that further research is needed for validation 4 . As the omics data are not accurately matched across all the samples, we then smoothed the omics data using our previously published approach 19 ( Methods and Supplementary Fig. 2a–c ). Next, to reveal the specific patterns of molecules that change during human aging, we then grouped all the molecules with similar trajectories using an unsupervised fuzzy c-means clustering approach 19 ( Methods , Fig. 3b and Supplementary Fig. 2d,e ). We identified 11 clusters of molecular trajectories that changed during aging, which ranged in size from 638 to 1,580 molecules/microbes (Supplementary Fig. 2f and Supplementary Data ). We found that most molecular patterns exhibit nonlinear changes, indicating that aging is not a linear process (Fig. 2b ). Among the 11 identified clusters, three distinct clusters (2, 4 and 5) displayed compelling, straightforward and easily understandable patterns that spanned the entire lifespan (Fig. 2c ). Most molecules within these three clusters primarily consist of transcripts (Supplementary Fig. 2f ), which is expected because transcripts dominate the multi-omics data (8,556 out of 11,305, 75.7%). Cluster 4 exhibits a relatively stable pattern until approximately 60 years of age, after which it shows a rapid decrease (Fig. 2c ). Conversely, clusters 2 and 5 display fluctuations before 60 years of age, followed by a sharp increase and an upper inflection point at approximately 55–60 years of age (Fig. 2c ). We also attempted to observe this pattern of molecular change during aging individually. The participant with the longest follow-up period of 6.8 years (Fig. 1c ) approached the age of 60 years (range, 59.5–66.3 years; Extended Data Fig. 1g ), and it was not possible to identify obvious patterns in this short time window (Supplementary Fig. 2g ). Tracking individuals longitudinally over longer periods (decades) will be required to observe these trajectories at an individual level.

figure 2

a , Spearman correlation (cor) between the first principal component and ages for each type of omics data. The shaded area around the regression line represents the 95% confidence interval. b , The heatmap shows the molecular trajectories in 11 clusters during human aging. The right stacked bar plots show the percentages of different kinds of omics data, and the right box plots show the correlation distribution between features and ages ( n  = 108 participants). c , Three notable clusters of molecules that exhibit clear and straightforward nonlinear changes during human aging. The top stacked bar plots show the percentages of different kinds of omics data, and the top box plots show the correlation distribution between features and ages ( n  = 108 participants). The box plot shows the median (line), interquartile range (IQR) (box) and whiskers extending to 1.5 × IQR. Bars and lines are color-coded by omics type. Abs, absolute.

figure 3

a , Pathway enrichment and module analysis for each transcriptome cluster. The left panel is the heatmap for the pathways that undergo nonlinear changes across aging. The right panel is the pathway similarity network ( Methods ) ( n  = 108 participants). b , Pathway enrichment for metabolomics in each cluster. Enriched pathways and related metabolites are illustrated (Benjamini–Hochberg-adjusted P  < 0.05). c , Four clinical laboratory tests that change during human aging: blood urea nitrogen, serum/plasma glucose, mean corpuscular hemoglobin and red cell distribution width ( n  = 108 participants). The box plot shows the median (line), interquartile range (IQR) (box) and whiskers extending to 1.5 × IQR.

Although confounders, including sex, were corrected before analysis ( Methods ), we acknowledge that the age range for menopause in females is typically between 45 years and 55 years of age 20 , which is very close to the major transition points in all three clusters (Fig. 2c ). Therefore, we conducted further investigation into whether the menopausal status of females in the dataset contributed to the observed transition point at approximately 55 years of age (Fig. 2c ) by performing separate clustering analyses on the male and female datasets. Surprisingly, both the male and female datasets exhibited similar clusters, as illustrated in Extended Data Fig. 4a . This suggests that the transition point observed at approximately 55 years of age is not solely attributed to female menopause but, rather, represents a common phenomenon in the aging process of both sexes. This result is consistent with previous studies 14 , 15 , further supporting the notion that this transition point is a major characteristic feature of human aging. Moreover, to investigate the possibility that the transcriptomics data might skew the results toward transcriptomic changes as age-related factors, we conducted two additional clustering analyses—one focusing solely on transcriptomic data and another excluding it. Interestingly, both analyses yielded nearly identical three-cluster configurations, as observed using the complete omics dataset (Extended Data Fig. 4b ). This reinforces the robustness of the identified clusters and confirms that they are consistent across various omics platforms, not just driven by transcriptomic data.

Nonlinear changes in function and disease risk during aging

To gain further insight into the biological functions associated with the nonlinear changing molecules within the three identified clusters, we conducted separate functional analyses for transcriptomics, proteomics and metabolomics datasets for all three clusters. In brief, we constructed a similarity network using enriched pathways from various databases (Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and Reactome) and identified modules to eliminate redundant annotations. We then used all modules from different databases to reduce redundancy further using the same approach and define the final functional modules ( Methods , Extended Data Fig. 4c and Supplementary Data ). We identified some functional modules that were reported in previous studies, but we defined their more accurate patterns of change during human aging. Additionally, we also found previously unreported potential functional modules during human aging ( Supplementary Data ). For instance, in cluster 2, we identified a transcriptomic module associated with GTPase activity (adjusted P  = 1.64 × 10 −6 ) and histone modification (adjusted P   =  6.36 × 10 −7 ) (Fig. 3a ). Because we lack epigenomic data in this study, our findings should be validated through additional experiments in the future. GTPase activity is closely correlated with programmed cell death (apoptosis), and some previous studies showed that this activity increases during aging 21 . Additionally, histone modifications have been demonstrated to increase during human aging 22 . In cluster 4, we identified one transcriptomics module associated with oxidative stress; this module includes antioxidant activity, oxygen carrier activity, oxygen binding and peroxidase activity (adjusted P  = 0.029) (Fig. 3a ). Previous studies demonstrated that oxidative stress and many reactive oxygen species (ROS) are positively associated with increased inflammation in relation to aging 23 . In cluster 5, the first transcriptomics module is associated with mRNA stability, which includes mRNA destabilization (adjusted P   =  0.0032), mRNA processing (adjusted P   =  3.2 × 10 −4 ), positive regulation of the mRNA catabolic process (adjusted P   =  1.46 × 10 −4 ) and positive regulation of the mRNA metabolic process (adjusted P   =  0.00177) (Fig. 3a ). Previous studies showed that mRNA turnover is associated with aging 24 . The second module is associated with autophagy (Fig. 3a ), which increases during human aging, as demonstrated in previous studies 25 .

In addition, we also identified certain modules in the clusters that suggest a nonlinear increase in several disease risks during human aging. For instance, in cluster 2, where components increase gradually and then rapidly after age 60, the phenylalanine metabolism pathway (adjusted P   =  4.95 × 10 −4 ) was identified (Fig. 3b ). Previous studies showed that aging is associated with a progressive increase in plasma phenylalanine levels concomitant with cardiac dysfunction, and dysregulated phenylalanine catabolism is a factor that triggers deviations from healthy cardiac aging trajectories 26 . Additionally, C-X-C motif chemokine 5 (CXCL5 or ENA78) from proteomics data, which has higher concentrations in atherosclerosis 27 , is also detected in cluster 2 ( Supplementary Data ). The clinical laboratory test blood urea nitrogen, which provides important information about kidney function, is also detected in cluster 2 (Fig. 3c ). This indicates that kidney function nonlinearly decreases during aging. Furthermore, the clinical laboratory test for serum/plasma glucose, a marker of type 2 diabetes (T2D), falls within cluster 2. This is consistent with and supported by many previous studies demonstrating that aging is a major risk factor for T2D 28 . Collectively, these findings suggest a nonlinear escalation in the risk of cardiovascular and kidney diseases and T2D with advancing age, particularly after the age of 60 years (Fig. 2c ).

The identified modules in cluster 4 also indicate a nonlinear increase in disease risks. For instance, the unsaturated fatty acids biosynthesis pathway (adjusted P   =  4.71 × 10 −7 ) is decreased in cluster 4. Studies have shown that unsaturated fatty acids are helpful in reducing CVD risk and maintaining brain function 29 , 30 . The pathway of alpha-linolenic acid and linolenic acid metabolism (adjusted P   =  1.32 × 10 −4 ) can reduce aging-associated diseases, such as CVD 31 . We also detected the caffeine metabolism pathway (adjusted P   =  7.34 × 10 −5 ) in cluster 4, which suggests that the ability to metabolize caffeine decreases during aging. Additionally, the cytokine MCP1 (chemokine (C-C motif) ligand 2 (CCL2)), a member of the CC chemokine family, plays an important immune regulatory role and is also in cluster 4 ( Supplementary Data ). These findings further support previous observations and highlight the nonlinear increase in age-related disease risk as individuals age.

Cluster 5 comprises the clinical tests of mean corpuscular hemoglobin and red cell distribution width (Fig. 3c ). These tests assess the average hemoglobin content per red blood cell and the variability in the size and volume of red blood cells, respectively. These findings align with the aforementioned transcriptomic data, which suggest a nonlinear reduction in the oxygen-carrying capacity associated with the aging process.

Aside from these three distinct clusters (Fig. 2c ), we also conducted pathway enrichment analysis across all other eight clusters, which displayed highly nonlinear trajectories, employing the same method (Fig. 2b and Supplementary Data ). Notably, cluster 11 exhibited a consistent increase up until the age of 50, followed by a decline until the age of 56, after which no substantial changes were observed up to the age of 75. A particular transcriptomics module related to DNA repair was identified, encompassing three pathways: positive regulation of double-strand break repair (adjusted P   =  0.042), peptidyl−lysine acetylation (adjusted P   =  1.36 × 10 −5 ) and histone acetylation (adjusted P   =  3.45 × 10 −4 ) (Extended Data Fig. 4d ). These three pathways are critical in genomic stability, gene expression and metabolic balances, with their levels diminishing across the human lifespan 32 , 33 , 34 . Our findings reveal a nonlinear alteration across the human lifespan in these pathways, indicating an enhancement in DNA repair capabilities before the age of 50, a marked reduction between the ages of 50 and 56 and stabilization after that until the age of 75. The pathway enrichment results for all clusters are detailed in the Supplementary Data .

Altogether, the comprehensive functional analysis offers valuable insights into the nonlinear changes observed in molecular profiles and their correlations with biological functions and disease risks across the human lifespan. Our findings reveal that individuals aged 60 and older exhibit increased susceptibility to CVD, kidney issues and T2D. These results carry important implications for both the diagnosis and prevention of these diseases. Notably, many clinically actionable markers were identified, which have the potential for improved healthcare management and enhanced overall well-being of the aging population.

Uncovering waves of aging-related molecules during aging

Although the trajectory clustering approaches described above effectively identify nonlinear changing molecules and microbes that exhibit clear and compelling patterns throughout human aging, it may not be as effective in capturing substantial changes that occur at specific chronological aging periods. In such cases, alternative analytical approaches may be necessary to detect and characterize these dynamics. To gain a comprehensive understanding of changes in multi-omics profiling during human aging, we used a modified version of the DE-SWAN algorithm 14 , as described in the Methods section. This algorithm identifies dysregulated molecules and microbes throughout the human lifespan by analyzing molecule levels within 20-year windows and comparing two groups in 10-year parcels while sliding the window incrementally from young to old ages 14 . Using this approach and multiomics data, we detected changes at specific stages of lifespan and uncovered the sequential effects of aging. Our analysis revealed thousands of molecules exhibiting changing patterns throughout aging, forming distinct waves, as illustrated in Fig. 3a . Notably, we observed two prominent crests occurring around the ages of 45 and 65, respectively (Fig. 4a ). Notably, too, these crests were consistent with findings from a previous study that included only proteomics data 14 . Specifically, crest 2 aligns with our previous trajectory clustering result, indicating a turning point at approximately 60 years of age (Fig. 2c ).

figure 4

a , Number of molecules and microbes differentially expressed during aging. Two local crests at the ages of 44 years and 60 years were identified. b , c , The same waves were detected using different q value ( b ) and window ( c ) cutoffs. d , The number of molecules/microbes differentially expressed for different types of omics data during human aging.

To demonstrate the significance of the two crests, we employed different q value cutoffs and sliding window parameters, which consistently revealed the same detectable waves (Fig. 4b,c and Supplementary Fig. 4a,b ). Furthermore, when we permuted the ages of individuals, the crests disappeared (Supplementary Figs. 3a and 4c ) ( Methods ). These observations highlight the robustness of the two major waves of aging-related molecular changes across the human lifespan. Although we already accounted for confounders before our statistical analysis, we took additional steps to explore their impact. Specifically, we investigated whether confounders, such as insulin sensitivity, sex and ethnicity, differed between the two crests across various age ranges. As anticipated, these confounders did not show significant differences across other age brackets (Supplementary Fig. 4d ). This further supports our conclusion that the observed differences in the two crests are attributable to age rather than other confounding variables.

The identified crests represent notable milestones in the aging process and suggest specific age ranges where substantial molecular alterations occur. Therefore, we investigated the age-related waves for each type of omics data. Interestingly, most types of omics data exhibited two distinct crests that were highly robust (Fig. 3b and Supplementary Fig. 4 ). Notably, the proteomics data displayed two age-related crests at ages around 40 years and 60 years. Only a small overlap was observed between our dataset and the results from the previous study (1,305 proteins versus 302 proteins, with only 75 proteins overlapping). The observed pattern in our study was largely consistent with the previous findings 14 . However, our finding that many types of omics data, including transcriptomics, proteomics, metabolomics, cytokine, gut microbiome, skin microbiome and nasal microbiome, exhibit these waves, often with a similar pattern as the proteomics data (Fig. 4d ), supports the hypothesis that aging-related changes are not limited to a specific omics layer but, rather, involve a coordinated and systemic alteration across multiple molecular components. Identifying consistent crests across different omics data underscores the robustness and reliability of these molecular milestones in the aging process.

Next, we investigated the roles and functions of dysregulated molecules within two distinct crests. Notably, we found that the two crests related to aging predominantly consisted of the same molecules (Supplementary Fig. 6 ). To focus on the unique biological functions associated with each crest and eliminate commonly occurring molecules, we removed background molecules present in most stages. To explore the specific biological functions associated with each type of omics data (transcriptomics, proteomics and metabolomics) for both crests, we employed the function annotation approach described above ( Methods ). In brief, we constructed a similarity network of enriched pathways and identified modules to remove redundant annotations (Supplementary Fig. 6 and Extended Data Fig. 5a,b ). Furthermore, we applied the same approach to all modules to reduce redundancy and identify the final functional modules ( Methods and Extended Data Fig. 6a ). Our analysis revealed significant changes in multiple modules associated with the two crests (Extended Data Fig. 6b–d ). To present the results clearly, Fig. 5a displays the top 20 pathways (according to adjusted P value) for each type of omics data, and the Supplementary Data provides a comprehensive list of all identified functional modules.

figure 5

a , Pathway enrichment and biological functional module analysis for crests 1 and 2. Dots and lines are color-coded by omics type. b , The overlapping of molecules between two crests and three clusters.

Interestingly, the analysis identifies many dysregulated functional modules in crests 1 and 2, indicating a nonlinear risk for aging-related diseases. In particular, several modules associated with CVD were identified in both crest 1 and crest 2 (Fig. 5a ), which is consistent with the above results (Fig. 3b ). For instance, the dysregulation of platelet degranulation (crest 1: adjusted P   =  1.77 × 10 −30 ; crest 2: adjusted P   =  1.73 × 10 −26 ) 35 , 36 , complement cascade (crest 1: adjusted P   =  3.84 × 10 −30 ; crest 2: adjusted P   =  2.02 × 10 −28 ), complement and coagulation cascades (crest 1: adjusted P   =  1.78 × 10 −46 ; crest 2: adjusted P   =  2.02 × 10 −28 ) 37 , 38 , protein activation cascade (crest 1: adjusted P   =  1.56 × 10 −17 ; crest 2: adjusted P   =  1.61 × 10 −8 ) and protease binding (crest 1: adjusted P   =  2.7 × 10 −6 ; crest 2: adjusted P   =  0.0114) 39 have various effects on the cardiovascular system and can contribute to various CVDs. Furthermore, blood coagulation (crest 1: adjusted P   =  1.48 × 10 −28 ; crest 2: adjusted P   =  9.10 × 10 −17 ) and fibrinolysis (crest 1: adjusted P   =  2.11 × 10 −15 ; crest 2: adjusted P   =  1.64 × 10 −10 ) were also identified, which are essential processes for maintaining blood fluidity, and dysregulation in these modules can lead to thrombotic and cardiovascular events 40 , 41 . We also identified certain dysregulated metabolic modules associated with CVD. For example, aging has been linked to an incremental rise in plasma phenylalanine levels (crest 1: adjusted P   =  9.214 × 10 −4 ; crest 2: adjusted P   =  0.0453), which can contribute to the development of cardiac hypertrophy, fibrosis and dysfunction 26 . Branched-chain amino acids (BCAAs), including valine, leucine and isoleucine (crest 1: adjusted P : not significant (NS); crest 2: adjusted P   =  0.0173), have also been implicated in CVD development 42 , 43 and T2D, highlighting their relevance in CVD pathophysiology. Furthermore, research suggests that alpha-linolenic acid (ALA) and linoleic acid metabolism (crest 1: adjusted P : NS; crest 2: adjusted P   =  0.0217) may be protective against coronary heart disease 44 , 45 . Our investigation also identified lipid metabolism modules that are associated with CVD, including high-density lipoprotein (HDL) remodeling (crest 1: adjusted P   =  1.073 × 10 −8 ; crest 2: adjusted P   =  2.589 × 10 −9 ) and glycerophospholipid metabolism (crest 1: adjusted P : NS; crest 2: adjusted P   =  0.0033), which influence various CVDs 46 , 47 , 48 .

In addition, the dysregulation of skin and muscle stability was found to be increased at crest 1 and crest 2, as evidenced by the identification of numerous modules associated with these processes (Fig. 5a,b ). This suggests that the aging of skin and muscle is markedly accelerated at crest 1 and crest 2. The extracellular matrix (ECM) provides structural stability, mechanical strength, elasticity and hydration to the tissues and cells, and the ECM of the skin is mainly composed of collagen, elastin and glycosaminoglycans (GAGs) 49 . Phosphatidylinositols are a class of phospholipids that have various roles in cytoskeleton organization 50 . Notably, the dysregulation of ECM structural constituent (crest 1: adjusted P   =  3.32 × 10 −8 ; crest 2: adjusted P   =  1.61 × 10 −8 ), GAG binding (crest 1: adjusted P   =  1.805 × 10 −8 ; crest 2: adjusted P   =  4.093 × 10 −6 ) and phosphatidylinositol binding (crest 1: adjusted P   =  3.391 × 10 −6 ; crest 2: adjusted P   =  7.832 × 10 −6 ) were identified 51 , 52 . We also identified cytolysis (crest 1: adjusted P   =  2.973 × 10 −5 ; crest 2: adjusted P : NS), which can increase water loss 53 . The dysregulated actin binding (crest 1: adjusted P   =  3.536 × 10 −8 ; crest 2: adjusted P   =  3.435 × 10 −9 ), actin filament organization (crest 1: adjusted P   =  8.406 × 10 −9 ; crest 2: adjusted P   =  1.157 × 10 −9 ) and regulation of actin cytoskeleton (crest 1: adjusted P   =  0.00090242; crest 2: adjusted P   =  6.788 × 10 −4 ) were identified, which affect the structure and function of various tissues 54 , 55 , 56 , 57 , 58 . Additionally, cell adhesion is the attachment of a cell to another cell or to ECM via adhesion molecules 59 . We identified the positive regulation of cell adhesion (crest 1: adjusted P   =  3.618 × 10 −5 ; crest 2: adjusted P   =  8.272 × 10 −9 ) module, which can prevent or delay skin aging 60 , 61 . Threonine can affect sialic acid production, which is involved in cell adhesion 62 . We also identified the glycine, serine and threonine metabolism (crest 1: adjusted P : NS; crest 2: adjusted P   =  0.00506) 62 . Additionally, scavenging of heme from plasma was identified (crest 1: adjusted P   =  1.176 × 10 −11 ; crest 2: adjusted P   =  1.694 × 10 −8 ), which can modulate skin aging as excess-free heme can damage cellular components 63 , 64 . Rho GTPases regulate a wide range of cellular responses, including changes to the cytoskeleton and cell adhesion (RHO GTPase cycle, crest 1: adjusted P   =  9.956 × 10 −10 ; crest 2: adjusted P   =  1.546 × 10 −5 ) 65 . In relation to muscle, previous studies demonstrated that muscle mass decreases by approximately 3–8% per decade after the age of 30, with an even higher decline rate after the age of 60, which consistently coincides with the observed second crest 66 . Interestingly, we identified dysregulation in the module associated with the structural constituent of muscle (crest 1: adjusted P   =  0.00565; crest 2: adjusted P   =  0.0162), consistent with previous findings 66 . Furthermore, we identified the pathway associated with caffeine metabolism (crest 1: adjusted P   =  0.00378; crest 2: adjusted P   =  0.0162), which is consistent with our observations above (Fig. 2b ) and implies that the capacity to metabolize caffeine undergoes a notable alteration not only around 60 years of age but also around the age of 40 years.

In crest 1, we identified specific modules associated with lipid and alcohol metabolism. Previous studies established that lipid metabolism declines with human aging 67 . Our analysis revealed several modules related to lipid metabolism, including plasma lipoprotein remodeling (crest 1: adjusted P   =  2.269 × 10 −9 ), chylomicron assembly (crest 1: adjusted P   =  9.065 × 10 −7 ) and ATP-binding cassette (ABC) transporters (adjusted P   =  1.102 × 10 −4 ). Moreover, we discovered a module linked to alcohol metabolism (alcohol binding, adjusted P   =  8.485 × 10 −7 ), suggesting a decline in alcohol metabolization efficiency with advancing age, particularly around the age of 40, when it significantly diminishes. In crest 2, we observed prominent modules related to immune dysfunction, encompassing acute-phase response (adjusted P   =  2.851 × 10 −8 ), antimicrobial humoral response (adjusted P   =  2.181 × 10 −5 ), zymogen activation (adjusted P   =  4.367 × 10 −6 ), complement binding (adjusted P   =  0.002568), mononuclear cell differentiation (adjusted P   =  9.352 × 10 −8 ), viral process (adjusted P   =  5.124 × 10 −7 ) and regulation of hemopoiesis (adjusted P   =  3.522 × 10 −7 ) (Fig. 5a ). Age-related changes in the immune system, collectively known as immunosenescence, have been extensively documented 68 , 69 , 70 , and our results demonstrate a rapid decline at age 60. Furthermore, we also identified modules associated with kidney function (glomerular filtration, adjusted P   =  0.00869) and carbohydrate metabolism (carbohydrate binding, adjusted P   =  0.01045). Our previous findings indicated a decline in kidney function around the age of 60 years (Fig. 3c ), as did the present result of this observation. Previous studies indicated the influence of carbohydrates on aging, characterized by the progressive decline of physiological functions and increased susceptibility to diseases over time 71 , 72 .

In summary, our analysis identifies many dysregulated functional modules identified in both crest 1 and crest 2 that underlie the risk for various diseases and alterations of biological functions. Notably, we observed an overlap of dysregulated functional modules among clusters 2, 4 and 6 because they overlap at the molecular level, as depicted in Fig. 5b . This indicates that certain molecular components are shared among these clusters and the identified crests. However, it is important to note that numerous molecules are specific to each of the two approaches employed in our study. This suggests that these two approaches complement each other in identifying nonlinear changes in molecules and functions during human aging. By using both approaches, we were able to capture a more comprehensive understanding of the molecular alterations associated with aging and their potential implications for diseases.

Analyzing a longitudinal multi-omics dataset involving 108 participants, we successfully captured the dynamic and nonlinear molecular changes that occur during human aging. Our study’s strength lies in the comprehensive nature of the dataset, which includes multiple time point samples for each participant. This longitudinal design enhances the reliability and robustness of our findings compared to cross-sectional studies with only one time point sample for each participant. The first particularly intriguing finding from our analysis is that only a small fraction of molecules (6.6%) displayed linear changes throughout human aging (Fig. 1d ). This observation is consistent with previous research and underscores the limitations of relying solely on linear regression to understand the complexity of aging-related molecular changes 5 . Instead, our study revealed that a considerable number of molecules (81.0%) exhibited nonlinear patterns (Fig. 1e ). Notably, this nonlinear trend was observed across all types of omics data with remarkably high consistency (Fig. 1e,g ), highlighting the widespread functionally relevant nature of these dynamic changes. By unveiling the nonlinear molecular alterations associated with aging, our research contributes to a more comprehensive understanding of the aging process and its molecular underpinnings.

To further investigate the nonlinear changing molecules observed in our study, we employed a trajectory clustering approach to group molecules with similar temporal patterns. This analysis revealed the presence of three distinct clusters (Fig. 2c ) that exhibited clear and compelling patterns across the human lifespan. These clusters suggest that there are specific age ranges, such as around 60 years old, where distinct and extensive molecular changes occur (Fig. 2c ). Functional analysis revealed several modules that exhibited nonlinear changes during human aging. For example, we identified a module associated with oxidative stress, which is consistent with previous studies linking oxidative stress to the aging process 23 (Fig. 3a ). Our analysis indicates that this pathway increases significantly after the age of 60 years. In cluster 5, we identified a transcriptomics module associated with mRNA stabilization and autophagy (Fig. 3a ). Both of these processes have been implicated in the aging process and are involved in maintaining cellular homeostasis and removing damaged components. Furthermore, our analysis uncovered nonlinear changes in disease risk across aging. In cluster 2, we identified the phenylalanine metabolism pathway (Fig. 3b ), which has been associated with cardiac dysfunction during aging 26 . Additionally, we found that the clinical laboratory tests blood urea nitrogen and serum/plasma glucose increase significantly with age (cluster 2; Fig. 3c ), indicating a nonlinear decline in kidney function and an increased risk of T2D with age, with a critical threshold occurring approximately at the age of 60 years. In cluster 4, we identified pathways related to cardiovascular health, such as the biosynthesis of unsaturated fatty acids and caffeine metabolism (Fig. 3b ). Overall, our study provides compelling evidence for the existence of nonlinear changes in molecular profiles during human aging. By elucidating the specific functional modules and disease-related pathways that exhibit such nonlinear changes, we contribute to a better understanding of the complex molecular dynamics underlying the aging process and its implications for disease risk.

Although the trajectory clustering approach proves effective in identifying molecules that undergo nonlinear changes, it may not be as proficient in capturing substantial alterations that occur at specific time points without exhibiting a consistent pattern in other stages. We then employed a modified version of the DE-SWAN algorithm 14 to comprehensively investigate changes in multi-omics profiling throughout human aging. This approach enabled us to identify waves of dysregulated molecules and microbes across the human lifespan. Our analysis revealed two prominent crests occurring around the ages of 40 years and 60 years, which were consistent across various omics data types, suggesting their universal nature (Fig. 4a,e ). Notably, in the proteomics data, we observed crests around the ages of 40 years and 60 years, which aligns approximately with a previous study (which reported crests at ages 34 years, 60 years and 78 years) 14 . Due to the age range of our cohort being 25–75 years, we did not detect the third peak. Furthermore, the differences in proteomics data acquisition platforms (mass spectrometry versus SomaScan) 14 , 73 resulted in different identified proteins, with only a small overlap (1,305 proteins versus 302 proteins, of which only 75 were shared). This discrepancy may explain the age variation of the first crest identified in the two studies (approximately 10 years). However, despite the differences in the two proteomics datasets, the wave patterns observed in both studies were highly similar 14 (Fig. 4a ). Remarkably, by considering multiple omics data types, we consistently identified similar crests for each type, indicating the universality of these waves of change across plasma molecules and microbes from various body sites (Fig. 4e and Supplementary Fig. 3 ).

The analysis of molecular functionality in the two distinct crests revealed the presence of several modules, indicating a nonlinear increase in the risks of various diseases (Fig. 5a ). Both crest 1 and crest 2 exhibit the identification of multiple modules associated with CVD, which aligns with the aforementioned findings (Fig. 3b ). Moreover, we observed an escalated dysregulation in skin and muscle functioning in both crest 1 and crest 2. Additionally, we identified a pathway linked to caffeine metabolism, indicating a noticeable alteration in caffeine metabolization not only around the age of 60 but also around the age of 40. This shift may be due to either a metabolic shift or a change in caffeine consumption. In crest 1, we also identified specific modules associated with lipid and alcohol metabolism, whereas crest 2 demonstrated prominent modules related to immune dysfunction. Furthermore, we also detected modules associated with kidney function and carbohydrate metabolism, which is consistent with our above results. These findings reinforce our previous observations regarding a decline in kidney function around the age of 60 years (Fig. 3c ) while shedding light on the impact of dysregulated functional modules in both crest 1 and crest 2, suggesting nonlinear changes in disease risk and functional dysregulation. Notably, we identified an overlap of dysregulated functional modules among clusters 2, 4 and 6, indicating molecular-level similarities between these clusters and the identified crests (Fig. 5b ). This suggests the presence of shared molecular components among these clusters and crests. However, it is crucial to note that there are also numerous molecules specific to each of the two approaches employed in our study, indicating that these approaches complement each other in identifying nonlinear changes in molecules and functions during human aging.

The present research is subject to certain constraints. We accounted for many basic characteristics (confounders) of participants in the cohort; but because this study primarily reflects between-individual differences, there may be additional confounders due to the different age distributions of the participants. For example, we identified a notable decrease in oxygen carrier activity around age 60 (Figs. 2c and 3a ) and marked variations in alcohol and caffeine metabolism around ages 40 and 60 (Fig. 3a ). However, these findings might be shaped by participants’ lifestyle—that is, physical activity and their alcohol and caffeine intake. Regrettably, we do not have such detailed behavioral data for the entire group, necessitating validation in upcoming research. Although initial BMI and insulin sensitivity measurements were available at cohort entry, subsequent metrics during the observation span were absent, marking a study limitation.

A further constraint is our cohort’s modest size, encompassing merely 108 individuals (eight individuals between 25 years and 40 years of age), which hampers the full utilization of deep learning and may affect the robustness of the identification of nonlinear changing features in Fig. 1e . Although advanced computational techniques, including deep learning, are pivotal for probing nonlinear patterns, our sample size poses restrictions. Expanding the cohort size in subsequent research would be instrumental in harnessing the full potential of machine learning tools. Another limitation of our study is that the recruitment of participants was within the community around Stanford University, driven by rigorous sample collection procedures and the substantial expenses associated with setting up a longitudinal cohort. Although our participants exhibited a considerable degree of ethnic age and biological sex diversity (Fig. 1a and Supplementary Data ), it is important to acknowledge that our cohort may not fully represent the diversity of the broader population. The selectivity of our cohort limits the generalizability of our findings. Future studies should aim to include a more diverse cohort to enhance the external validity and applicability of the results.

In addition, the mean observation span for participants was 626 days, which is insufficient for detailed inflection point analyses. Our cohort’s age range of 25–70 years lacks individuals who lie outside of this range. The molecular nonlinearity detected might be subject to inherent variations or oscillations, a factor to consider during interpretation. Our analysis has not delved into the nuances of the dynamical systems theory, which provides a robust mathematical framework for understanding observed behaviors. Delving into this theory in future endeavors may yield enhanced clarity and interpretation of the data.

Moreover, it should be noted that, in our study, the observed nonlinear molecular changes occurred across individuals of varying ages rather than within the same individuals. This is attributed to the fact that, despite our longitudinal study, the follow-up period for our participants was relatively brief for following aging patterns (median, 1.7 years; Extended Data Fig. 1g ). Such a timeframe is inadequate for detecting nonlinear molecular changes that unfold over decades throughout the human lifespan. Addressing this limitation in future research is essential.

Lastly, our study’s molecular data are derived exclusively from blood samples, casting doubt on its direct relevance to specific tissues, such as the skin or muscles. We propose that blood gene expression variations might hint at overarching physiological alterations, potentially impacting the ECM in tissues, including skin and muscle. Notably, some blood-based biomarkers and transcripts have demonstrated correlations with tissue modifications, inflammation and other elements influencing the ECM across diverse tissues 74 , 75 .

In our future endeavors, the definitive confirmation of our findings hinges on determining if nonlinear molecular patterns align with nonlinear changes in functional capacities, disease occurrences and mortality hazards. For a holistic grasp of this, amalgamating multifaceted data from long-term cohort studies covering several decades becomes crucial. Such data should encompass molecular markers, comprehensive medical records, functional assessments and mortality data. Moreover, employing cutting-edge statistical techniques is vital to intricately decipher the ties between these nonlinear molecular paths and health-centric results.

In summary, the unique contribution of our study lies not merely in reaffirming the nonlinear nature of aging but also in the depth and breadth of the multi-omics data that we analyzed. Our study goes beyond stating that aging is nonlinear by identifying specific patterns, inflection points and potential waves in aging across multiple layers of biological data during human aging. Identifying specific clusters with distinct patterns, functional implications and disease risks enhances our understanding of the aging process. By considering the nonlinear dynamics of aging-related changes, we can gain insights into specific periods of significant changes (around age 40 and age 60) and the molecular mechanisms underlying age-related diseases, which could lead to the development of early diagnosis and prevention strategies. These comprehensive multi-omics data and the approach allow for a more nuanced understanding of the complexities involved in the aging process, which we think adds value to the existing body of research. However, further research is needed to validate and expand upon these findings, potentially incorporating larger cohorts to capture the full complexity of aging.

The participant recruitment, sample collection, data acquisition and data processing were documented in previous studies conducted by Zhou et al. 76 , Ahadi et al. 5 , Schüssler-Fiorenza Rose et al. 77 , Hornburg et al. 78 and Zhou et al. 79 .

Participant recruitment

Participants provided informed written consent for the study under research protocol 23602, which was approved by the Stanford University institutional review board. This study adheres to all relevant ethical regulations, ensuring informed consents were obtained from all participants. All participants consented to publication of potentially identifiable information. The cohort comprised 108 participants who underwent follow-up assessments. Exclusion criteria encompassed conditions such as anemia, kidney disease, a history of CVD, cancer, chronic inflammation or psychiatric illnesses as well as any prior bariatric surgery or liposuction. Each participant who met the eligibility criteria and provided informed consent underwent a one-time modified insulin suppression test to quantify insulin-mediated glucose uptake at the beginning of the enrollment 76 . The steady-state plasma glucose (SSPG) levels served as a direct indicator of each individual’s insulin sensitivity in processing a glucose load. We categorized individuals with SSPG levels below 150 mg dl −1 as insulin sensitive and those with levels of 150 mg dl −1 or higher as insulin resistant 80 , 81 . Thirty-eight participants were missing SSPG values, rendering their insulin resistance or sensitivity status undetermined. We also collected fasting plasma glucose (FPG) data for 69 participants at enrollment. Based on the FPG levels, two participants were identified as having diabetes at enrollment, with FPG levels exceeding 126 mg dl −1 ( Supplementary Data ). Additionally, we measured hemoglobin A1C (HbA1C) levels during each visit, using it as a marker for average glucose levels over the past 3 months: 6.5% or higher indicates diabetes. Accordingly, four participants developed diabetes during the study period. At the beginning of the enrollment, BMI was also measured for each participant. Participants received no compensation.

Comprehensive sample collection was conducted during the follow-up period, and multi-omics data were acquired (Fig. 1b ). For each visit, the participants self-reported as healthy or non-healthy 76 . To ensure accuracy and minimize the impact of confounding factors, only samples from individuals classified as healthy were selected for subsequent analysis.

Transcriptomics

Transcriptomic profiling was conducted on flash-frozen PBMCs. RNA isolation was performed using a QIAGEN All Prep kit. Subsequently, RNA libraries were assembled using an input of 500 ng of total RNA. In brief, ribosomal RNA (rRNA) was selectively eliminated from the total RNA pool, followed by purification and fragmentation. Reverse transcription was carried out using a random primer outfitted with an Illumina-specific adaptor to yield a cDNA library. A terminal tagging procedure was used to incorporate a second adaptor sequence. The final cDNA library underwent amplification. RNA sequencing libraries underwent sequencing on an Illumina HiSeq 2000 platform. Library quantification was performed via an Agilent Bioanalyzer and Qubit fluorometric quantification (Thermo Fisher Scientific) using a high-sensitivity dsDNA kit. After normalization, barcoded libraries were pooled at equimolar ratios into a multiplexed sequencing library. An average of 5–6 libraries were processed per HiSeq 2000 lane. Standard Illumina pipelines were employed for image analysis and base calling. Read alignment to the hg19 reference genome and personal exomes was achieved using the TopHat package, followed by transcript assembly and expression quantification via HTseq and DESeq2. In the realm of data pre-processing, genes with an average read count across all samples lower than 0.5 were excluded. Samples exhibiting an average read count lower than 0.5 across all remaining genes were likewise removed. For subsequent global variance and correlation assessments, genes with an average read count of less than 1 were eliminated.

Plasma sample tryptic peptides were fractionated using a NanoLC 425 System (SCIEX) operating at a flow rate of 5 μl min −1 under a trap-elute configuration with a 0.5 × 10 mm ChromXP column (SCIEX). The liquid chromatography gradient was programmed for a 43-min run, transitioning from 4% to 32% of mobile phase B, with an overall run time of 1 h. Mobile phase A consisted of water with 0.1% formic acid, and mobile phase B was formulated with 100% acetonitrile and 0.1% formic acid. An 8-μg aliquot of non-depleted plasma was loaded onto a 15-cm ChromXP column. Mass spectrometry analysis was executed employing SWATH acquisition on a TripleTOF 6600 system. A set of 100 variable Q1 window SWATH acquisition methods was designed in high-sensitivity tandem mass spectrometry (MS/MS) mode. Subsequent data analysis included statistical scoring of peak groups from individual runs via pyProphet 82 , followed by multi-run alignment through TRIC60, ultimately generating a finalized data matrix with a false discovery rate (FDR) of 1% at the peptide level and 10% at the protein level. Protein quantitation was based on the sum of the three most abundant peptide signals for each protein. Batch effect normalization was achieved by subtracting principal components that primarily exhibited batch-associated variation, using Perseus software v.1.4.2.40.

Untargeted metabolomics

A ternary solvent system of acetone, acetonitrile and methanol in a 1:1:1 ratio was used for metabolite extraction. The extracted metabolites were dried under a nitrogen atmosphere and reconstituted in a 1:1 methanol:water mixture before analysis. Metabolite profiles were generated using both hydrophilic interaction chromatography (HILIC) and reverse-phase liquid chromatography (RPLC) under positive and negative ion modes. Thermo Q Exactive Plus mass spectrometers were employed for HILIC and RPLC analyses, respectively, in full MS scan mode. MS/MS data were acquired using quality control (QC) samples. For the HILIC separations, a ZIC-HILIC column was used with mobile phase solutions of 10 mM ammonium acetate in 50:50 and 95:5 acetonitrile:water ratios. In the case of RPLC, a Zorbax SBaq column was used, and the mobile phase consisted of 0.06% acetic acid in water and methanol. Metabolic feature detection was performed using Progenesis QI software. Features from blanks and those lacking sufficient linearity upon dilution were excluded. Only features appearing in more than 33% of the samples were retained for subsequent analyses, and any missing values were imputed using the k -nearest neighbors approach. We employed locally estimated scatterplot smoothing (LOESS) normalization 83 to correct the metabolite-specific signal drift over time. The metid package 84 was used for metabolite annotation.

Cytokine data

A panel of 62 human cytokines, chemokines and growth factors was analyzed in EDTA-anticoagulated plasma samples using Luminex-based multiplex assays with conjugated antibodies (Affymetrix). Raw fluorescence measurements were standardized to median fluorescence intensity values and subsequently subjected to variance-stabilizing transformation to account for batch-related variations. As previously reported 76 , data points characterized by background noise, termed CHEX, that deviate beyond five standard deviations from the mean (mean ± 5 × s.d.) were excluded from the analyses.

Clinical laboratory test

The tests encompassed a comprehensive metabolic panel, a full blood count, glucose and HbA1C levels, insulin assays, high-sensitivity C-reactive protein (hsCRP), immunoglobulin M (IgM) and lipid, kidney and liver panels.

Lipid extraction and quantification procedures were executed in accordance with established protocols 78 . In summary, complex lipids were isolated from 40 μl of EDTA plasma using a solvent mixture comprising methyl tertiary-butyl ether, methanol and water, followed by a biphasic separation. Subsequent lipid analysis was conducted on the Lipidyzer platform, incorporating a differential mobility spectrometry device (SelexION Technology) and a QTRAP 5500 mass spectrometer (SCIEX).

Immediately after arrival, samples were stored at −80 °C. Stool and nasal samples were processed and sequenced in-house at the Jackson Laboratory for Genomic Medicine, whereas oral and skin samples were outsourced to uBiome for additional processing. Skin and oral samples underwent 30 min of beads-beating lysis, followed by a silica-guanidinium thiocyanate-based nucleic acid isolation protocol. The V4 region of the 16S rRNA gene was amplified using specific primers, after which the DNA was barcoded and sequenced on an Illumina NextSeq 500 platform via a 2 × 150-bp paired-end protocol. Similarly, stool and nasal samples were processed for 16S rRNA V1–V3 region amplification using a different set of primers and sequenced on an Illumina MiSeq platform. For data processing, the raw sequencing data were demultiplexed using BCL2FASTQ software and subsequently filtered for quality. Reads with a Q-score lower than 30 were excluded. The DADA2 R package was used for further sequence data processing, which included filtering out reads with ambiguous bases and errors, removing chimeras and aligning sequences against a validated 16S rRNA gene database. Relative abundance calculations for amplicon sequence variants (ASVs) were performed, and samples with inadequate sequencing depth (<1,000 reads) were excluded. Local outlier factor (LOF) was calculated for each point on a depth-richness plot, and samples with abnormal LOF were removed. In summary, rigorous procedures were followed in both the collection and processing stages, leveraging automated systems and specialized software to ensure the quality and integrity of the microbiome data across multiple body sites.

Statistics and reproducibility

For all data processing, statistical analysis and data visualization tasks, RStudio, along with R language (v.4.2.1), was employed. A comprehensive list of the packages used can be found in the Supplementary Note . The Benjamini–Hochberg method was employed to account for multiple comparisons. Spearman correlation coefficients were calculated using the R functions ‘cor’ and ‘cor.test’. Principal-component analysis (PCA) was conducted using the R function ‘princomp’. Before all the analyses, the confounders, such as BMI, sex, IRIS and ethnicity, were adjusted using the previously published method 19 . In brief, we used the intensity of each feature as the dependent variable (Y) and the confounding factors as the independent variables (X) to build a linear regression model. The residuals from this model were then used as the adjusted values for that specific feature.

All the omics data were acquired randomly. No statistical methods were used to predetermine the sample size, but our sample sizes are similar to those reported in previous publications 5 , 76 , 77 , 78 , 79 , and no data were excluded from the analyses. Additionally, the investigators were blinded to allocation during experiments and outcome assessment to the conditions of the experiments. Data distribution was assumed to be normal, but this was not formally tested.

The icons used in figures are from iconfont.cn, which can be used for non-commercial purposes under the MIT license ( https://pub.dev/packages/iconfont/license ).

Cross-sectional dataset generation

The ‘cross-sectional’ dataset was created by briefly extracting information from the longitudinal dataset. The mean value was calculated to represent each molecule’s intensity for each participant. Similarly, the age of each participant was determined by calculating the mean value of ages across all sample collection time points.

Linear changing molecule detection

We detected linear changing molecules during human aging using Spearman correlation and linear regression modeling. The confounders, such as BMI, sex, IRIS and ethnicity, were adjusted using the previously published method 19 . Our analysis revealed a high correlation between these two approaches in identifying such molecules. Based on these findings, we used the Spearman correlation approach to showcase the linear changing molecules during human aging. The permutation test was also used to get the permutated P values for each feature. In brief, each feature was subjected to sample label shuffling followed by a recalculation of the Spearman correlation. This process was reiterated 10,000 times, yielding 10,000 permuted Spearman correlations. The original Spearman correlation was then compared against these permuted values to obtain the permuted P values.

Dysregulated molecules compared to baseline during human aging

To depict the dysregulated molecules during human aging compared to the baseline, we categorized the participants into different age stages based on their ages. The baseline stage was defined as individuals aged 25–40 years. For each age stage group, we employed the Wilcoxon test to identify dysregulated molecules in comparison to the baseline, considering a significance threshold of P  < 0.05. Before the statistical analysis, all the confounders were corrected. Subsequently, we visualized the resulting dysregulated molecules at different age stages using a Sankey plot. The permutation test was also used to get the permutated P values for each feature. In brief, we shuffled the sample labels and recalculated the absolute mean difference between the two groups, against which the actual absolute mean difference was benchmarked to derive the permuted P values. To identify the molecules and microbes that exhibited significant changes at any given age stage, we adjusted the P values for each feature by multiplying them by 6. This adjustment adheres to the Bonferroni correction method, ensuring a rigorous evaluation of statistical significance.

Evaluation of the age reflected by different types of omics data

To assess whether each type of omics data accurately reflects the ages of individuals in our dataset, we conducted a PCA. Subsequently, we computed the Spearman correlation coefficient between the ages of participants and the first principal component (PC1). The absolute value of this coefficient was used to evaluate the degree to which the omics data reflect the ages (Fig. 2a ). PLS regression was also used to compare the strength of the age effect to the different omics data types. In brief, the ‘pls’ function from the R package mixOmics was used to construct the regression model between omics data and ages. Then, the ‘perf’ function was used to assess the performance of all the modules with sevenfold cross-validation. The R 2 was extracted to assess the strength of the age effect on the different omics data types.

To accommodate the varying time points of biological and omics data, we employed the LOESS approach. This approach allowed us to smooth and predict the multi-omics data at specific time points (that is, every half year) 14 , 85 . In brief, for each molecule, we fitted a LOESS regression model. During the fitting process, the LOESS argument ‘span’ was optimized through cross-validation. This ensured that the LOESS model provided an accurate and non-overfitting fit to the data (Supplementary Fig. 2a,b ). Once we obtained the LOESS prediction model, we applied it to predict the intensity of each molecule at every half-year time point.

Trajectory clustering analysis

To conduct trajectory clustering analysis, we employed the fuzzy c-means clustering approach available in the R package ‘Mfuzz.’ This approach was previously described in our publication 19 . The analysis proceeded in several steps. First, the omics data were auto-scaled to ensure comparable ranges. Next, we computed the minimum centroid distances for a range of cluster numbers, specifically from 2 to 22, in step 1. These minimum centroid distances served as a cluster validity index, helping us determine the optimal cluster number. Based on predefined rules, we selected the optimal cluster number. To refine the accuracy of this selection, we merged clusters with center expression data correlations greater than 0.8 into a single cluster. This step aimed to capture similar patterns within the data. The resulting optimal cluster number was then used for the fuzzy c-means clustering. Only molecules with memberships above 0.5 were retained within each cluster for further analysis. This threshold ensured that the molecules exhibited a strong association with their assigned cluster and contributed considerably to the cluster’s characteristics.

Pathway enrichment analysis and functional module identification

Transcriptomics and proteomics pathway enrichment.

Pathway enrichment analysis was conducted using the ‘clusterProfiler’ R package 86 . The GO, KEGG and Reactome databases were used. The P values were adjusted using the Benjamini–Hochberg method, with a significance threshold set at <0.05. To minimize redundant enriched pathways and GO terms, we employed a series of analyses. First, for enriched GO terms, we used the ‘Wang’ algorithm from the R package ‘simplifyEnrichment’ to calculate the similarity between GO terms. Only connections with a similarity score greater than 0.7 were retained to construct the GO term similarity network. Subsequently, community analysis was performed using the ‘igraph’ R package to partition the network into distinct modules. The GO term with the smallest enrichment adjusted P value was chosen as the representative within each module. The same approach was applied to the enriched KEGG and Reactome pathways, with one slight modification. In this case, the ‘jaccard’ algorithm was used to calculate the similarity between pathways, and a similarity cutoff of 0.5 was employed for the Jaccard index. After removing redundant enriched pathways, we combined all the remaining GO terms and pathways. Subsequently, we calculated the similarity between these merged entities using the Jaccard index. This similarity analysis aimed to capture the overlap and relationships between the different GO terms and pathways. Using the same approach as before, we performed community analysis to identify distinct biological functional modules based on the merged GO terms and pathways.

Identification of functional modules

First, we used the ‘Wang’ algorithm for the GO database and the ‘jaccard’ algorithm for the KEGG and Reactome databases to calculate the similarity between pathways. The enriched pathways served as nodes in a similarity network, with edges representing the similarity between two nodes. Next, we employed the R package ‘igraph’ to identify modules within the network based on edge betweenness. By gradually removing edges with the highest edge betweenness scores, we constructed a hierarchical map known as a dendrogram, representing a rooted tree of the graph. The leaf nodes correspond to individual pathways, and the root node represents the entire graph 87 . We then merged pathways within each module, selecting the pathway with the smallest adjusted P value to represent the module. After this step, we merged pathways from all three databases into modules. Subsequently, we repeated the process by calculating the similarity between modules from all three databases using the ‘jaccard’ algorithm. Once again, we employed the same approach described above to identify the functional modules.

Metabolomics pathway enrichment

To perform pathway enrichment analysis for metabolomics data, we used the human KEGG pathway database. This database was obtained from KEGG using the R package ‘massDatabase’ 88 . For pathway enrichment analysis, we employed the hypergeometric distribution test from the ‘TidyMass’ project 89 . This statistical test allowed us to assess the enrichment of metabolites within each pathway. To account for multiple tests, P values were adjusted using the Benjamini–Hochberg method. We considered pathways with Benjamini–Hochberg-adjusted P values lower than 0.05 as significantly enriched.

Modified DE-SWAN

The DE-SWAN algorithm 14 was used. To begin, a unique age is selected as the center of a 20-year window. Molecule levels in individuals younger than and older than that age are compared using the Wilcoxon test to assess differential expression. P values are calculated for each molecule, indicating the significance of the observed differences. To ensure sufficient sample sizes for statistical analysis in each time window, the initial window ranges from ages 25 to 50. The left half of this window covers ages 25–40, whereas the right half spans ages 41–50. The window then moves in one-year steps; this is why Fig. 4 displays an age range of 40–65 years. To account for multiple comparisons, these P values are adjusted using Benjamini–Hochberg correction. To evaluate the robustness and relevance of the DE-SWAN results, the algorithm is tested with various parcel widths, including 15 years, 20 years, 25 years and 30 years. Additionally, different q value thresholds, such as <0.0001, <0.001, <0.01 and <0.05, are applied. By comparing the results obtained with these different parameters to results obtained by chance, we can assess the significance of the findings. To generate random results for comparison, the phenotypes of the individuals are randomly permuted, and the modified DE-SWAN algorithm is applied to the permuted dataset. This allows us to determine whether the observed results obtained with DE-SWAN are statistically significant and not merely a result of chance.

Reporting summary

Further information on research design is available in the Nature Portfolio Reporting Summary linked to this article.

Data availability

The raw data used in this study can be accessed without any restrictions on the National Institutes of Health Human Microbiome 2 project site ( https://portal.hmpdacc.org ). Both the raw and processed data are also available on the Stanford iPOP site ( http://med.stanford.edu/ipop.html ). Researchers and interested individuals can visit these websites to access the data. For further details and inquiries about the study, we recommend contacting the corresponding author, who can provide additional information and address any specific questions related to the research.

Code availability

The statistical analysis and data processing in this study were performed using R v.4.2.1, along with various base packages and additional packages. Detailed information about the specific packages used can be found in the Supplementary Note , which accompanies the manuscript. Furthermore, all the custom scripts developed for this study have been made openly accessible and can be found on the GitHub repository at https://github.com/jaspershen-lab/ipop_aging . By visiting this repository, researchers and interested individuals can access and use the custom scripts for their own analyses or to replicate the study’s findings.

Hou, Y. et al. Ageing as a risk factor for neurodegenerative disease. Nat. Rev. Neurol. 15 , 565–581 (2019).

Article   PubMed   Google Scholar  

Chen, R. et al. Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell 148 , 1293–1307 (2012).

Article   CAS   PubMed   PubMed Central   Google Scholar  

Valdes, A. M., Glass, D. & Spector, T. D. Omics technologies and the study of human ageing. Nat. Rev. Genet. 14 , 601–607 (2013).

Rutledge, J., Oh, H. & Wyss-Coray, T. Measuring biological age using omics data. Nat. Rev. Genet. 23 , 715–727 (2022).

Ahadi, S. et al. Personal aging markers and ageotypes revealed by deep longitudinal profiling. Nat. Med. 26 , 83–90 (2020).

Ram, U. et al. Age-specific and sex-specific adult mortality risk in India in 2014: analysis of 0.27 million nationally surveyed deaths and demographic estimates from 597 districts. Lancet Glob. Health 3 , e767–e775 (2015).

Rodgers, J. L. et al. Cardiovascular risks associated with gender and aging. J. Cardiovasc. Dev. Dis. 6 , 19 (2019).

CAS   PubMed   PubMed Central   Google Scholar  

Poewe, W. et al. Parkinson disease. Nat. Rev. Dis. Primers 3 , 17013 (2017).

Hy, L. X. & Keller, D. M. Prevalence of AD among whites: a summary by levels of severity. Neurology 55 , 198–204 (2000).

Article   CAS   PubMed   Google Scholar  

Nussbaum, R. L. & Ellis, C. E. Alzheimer’s disease and Parkinson’s disease. N. Engl. J. Med. 348 , 1356–1364 (2003).

Xiong, Y. et al. Vimar/RAP1GDS1 promotes acceleration of brain aging after flies and mice reach middle age. Commun. Biol. 6 , 420 (2023).

Sherwood, C. C. et al. Aging of the cerebral cortex differs between humans and chimpanzees. Proc. Natl Acad. Sci. USA 108 , 13029–13034 (2011).

Márquez, E. J. et al. Sexual-dimorphism in human immune system aging. Nat. Commun. 11 , 751 (2020).

Article   PubMed   PubMed Central   Google Scholar  

Lehallier, B. et al. Undulating changes in human plasma proteome profiles across the lifespan. Nat. Med. 25 , 1843–1850 (2019).

Fehlmann, T. et al. Common diseases alter the physiological age-related blood microRNA profile. Nat. Commun. 11 , 5958 (2020).

Shavlakadze, T. et al. Age-related gene expression signature in rats demonstrate early, late, and linear transcriptional changes from multiple tissues. Cell Rep. 28 , 3263–3273 (2019).

Vershinina, O., Bacalini, M. G., Zaikin, A., Franceschi, C. & Ivanchenko, M. Disentangling age-dependent DNA methylation: deterministic, stochastic, and nonlinear. Sci Rep. 11 , 9201 (2021).

Li, J. et al. Determining a multimodal aging clock in a cohort of Chinese women. Med 4 , 825–848 (2023).

Shen, X. Multi-omics microsampling for the profiling of lifestyle-associated changes in health. Nat. Biomed. Eng . 8 , 11–29 (2024).

Takahashi, T. A. & Johnson, K. M. Menopause. Med. Clin. North Am. 99 , 521–534 (2015).

Umbayev, B. et al. Role of a small GTPase Cdc42 in aging and age-related diseases. Biogerontology 24 , 27–46 (2023).

Yi, S.-J. & Kim, K. New insights into the role of histone changes in aging. Int. J. Mol. Sci. 21 , 8241 (2020).

Liguori, I. et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 13 , 757–772 (2018).

Borbolis, F. & Syntichaki, P. Cytoplasmic mRNA turnover and ageing. Mech. Ageing Dev. 152 , 32–42 (2015).

Kaushik, S. et al. Autophagy and the hallmarks of aging. Ageing Res. Rev. 72 , 101468 (2021).

Czibik, G. et al. Dysregulated phenylalanine catabolism plays a key role in the trajectory of cardiac aging. Circulation 144 , 559–574 (2021).

Rousselle, A. et al. CXCL5 limits macrophage foam cell formation in atherosclerosis. J. Clin. Invest. 123 , 1343–1347 (2013).

Fazeli, P. K., Lee, H. & Steinhauser, M. L. Aging is a powerful risk factor for type 2 diabetes mellitus independent of body mass index. Gerontology 66 , 209–210 (2019).

Allayee, H., Roth, N. & Hodis, H. N. Polyunsaturated fatty acids and cardiovascular disease: implications for nutrigenetics. J. Nutrigenet. Nutrigenomics 2 , 140–148 (2009).

Sacks, F. M. et al. Dietary fats and cardiovascular disease: a presidential advisory from the American Heart Association. Circulation 136 , e1–e23 (2017).

Qi, W. et al. The ω-3 fatty acid α-linolenic acid extends Caenorhabditis elegans lifespan via NHR-49/PPARα and oxidation to oxylipins. Aging Cell 16 , 1125–1135 (2017).

Bird, A. W. et al. Acetylation of histone H4 by Esa1 is required for DNA double-strand break repair. Nature 419 , 411–415 (2002).

Sivanand, S. et al. Nuclear acetyl-CoA production by ACLY promotes homologous recombination. Mol. Cell 67 , 252–265 (2017).

Zhao, S. et al. Regulation of cellular metabolism by protein lysine acetylation. Science 327 , 1000–1004 (2010).

Vericel, E. et al. Platelets and aging I.—Aggregation, arachidonate metabolism and antioxidant status. Thromb. Res. 49 , 331–342 (1988).

Gu, S. X. & Dayal, S. Redox mechanisms of platelet activation in aging. Antioxidants (Basel) 11 , 995 (2022).

Oikonomopoulou, K., Ricklin, D., Ward, P. A. & Lambris, J. D. Interactions between coagulation and complement–their role in inflammation. Semin. Immunopathol. 34 , 151–165 (2012).

Wasiak, S. et al. Downregulation of the complement cascade in vitro, in mice and in patients with cardiovascular disease by the bet protein inhibitor apabetalone (RVX-208). J. Cardiovasc. Transl. 10 , 337–347 (2017).

Article   Google Scholar  

Slack, M. A. & Gordon, S. M. Protease activity in vascular disease. Arterioscl. Thromb. Vasc. Biol. 39 , e210–e218 (2019).

Mari, D. et al. Hemostasis and ageing. Immun. Ageing 5 , 12 (2008).

Lowe, G. & Rumley, A. The relevance of coagulation in cardiovascular disease: what do the biomarkers tell us? Thromb. Haemostasis 112 , 860–867 (2014).

Li, Y. et al. Branched chain amino acids exacerbate myocardial ischemia/reperfusion vulnerability via enhancing GCN2/ATF6/PPAR-α pathway-dependent fatty acid oxidation. Theranostics 10 , 5623–5640 (2020).

McGarrah, R. W. & White, P. J. Branched-chain amino acids in cardiovascular disease. Nat. Rev. Cardiol. 20 , 77–89 (2023).

Arsenian, M. Potential cardiovascular applications of glutamate, aspartate, and other amino acids. Clin. Cardiol. 21 , 620–624 (1998).

Grajeda-Iglesias, C. & Aviram, M. Specific amino acids affect cardiovascular diseases and atherogenesis via protection against macrophage foam cell formation: review article. Rambam Maimonides Med. J. 9 , e0022 (2018).

Chen, H. et al. Comprehensive metabolomics identified the prominent role of glycerophospholipid metabolism in coronary artery disease progression. Front. Mol. Biosci. 8 , 632950 (2021).

Giammanco, A. et al. Hyperalphalipoproteinemia and beyond: the role of HDL in cardiovascular diseases. Life (Basel) 11 , 581 (2021).

CAS   PubMed   Google Scholar  

Zhu, Q. et al. Comprehensive metabolic profiling of inflammation indicated key roles of glycerophospholipid and arginine metabolism in coronary artery disease. Front. Immunol. 13 , 829425 (2022).

Yue, B. Biology of the extracellular matrix: an overview. J. Glaucoma 23 , S20–S23 (2014).

Zambrzycka, A. Aging decreases phosphatidylinositol-4,5-bisphosphate level but has no effect on activities of phosphoinositide kinases. Pol. J. Pharmacol. 56 , 651–654 (2004).

Lee, D. H., Oh, J.-H. & Chung, J. H. Glycosaminoglycan and proteoglycan in skin aging. J. Dermatol. Sci. 83 , 174–181 (2016).

Khan, A. U., Qu, R., Fan, T., Ouyang, J. & Dai, J. A glance on the role of actin in osteogenic and adipogenic differentiation of mesenchymal stem cells. Stem Cell Res. Ther. 11 , 283 (2020).

Lago, J. C. & Puzzi, M. B. The effect of aging in primary human dermal fibroblasts. PLoS ONE 14 , e0219165 (2019).

Pollard, T. D. Actin and actin-binding proteins. Cold Spring Harb. Perspect. Biol. 8 , a018226 (2016).

Lai, W.-F. & Wong, W.-T. Roles of the actin cytoskeleton in aging and age-associated diseases. Ageing Res. Rev. 58 , 101021 (2020).

Garcia, G., Homentcovschi, S., Kelet, N. & Higuchi-Sanabria, R. Imaging of actin cytoskeletal integrity during aging in C. elegans . Methods Mol. Biol. 2364 , 101–137 (2022).

Kim, Y. J. et al. Links of cytoskeletal integrity with disease and aging. Cells 11 , 2896 (2022).

Oosterheert, W., Klink, B. U., Belyy, A., Pospich, S. & Raunser, S. Structural basis of actin filament assembly and aging. Nature 611 , 374–379 (2022).

Bruzzone, A. et al. Dosage-dependent regulation of cell proliferation and adhesion through dual β2-adrenergic receptor/cAMP signals. FASEB J. 28 , 1342–1354 (2014).

McEver, R. P. & Luscinskas, F. W. Cell adhesion. In Hematology 7th edn (eds Hoffman, R. et al.) 127–134 (Elsevier, 2018).

Persa, O. D., Koester, J. & Niessen, C. M. Regulation of cell polarity and tissue architecture in epidermal aging and cancer. J. Invest. Dermatol. 141 , 1017–1023 (2021).

Canfield, C.-A. & Bradshaw, P. C. Amino acids in the regulation of aging and aging-related diseases. Transl. Med. Aging 3 , 70–89 (2019).

Chiabrando, D., Vinchi, F., Fiorito, V., Mercurio, S. & Tolosano, E. Heme in pathophysiology: a matter of scavenging, metabolism and trafficking across cell membranes. Front. Pharmacol. 5 , 61 (2014).

Aggarwal, S. et al. Heme scavenging reduces pulmonary endoplasmic reticulum stress, fibrosis, and emphysema. JCI Insight 3 , e120694 (2018).

Hodge, R. G. & Ridley, A. J. Regulating Rho GTPases and their regulators. Nat. Rev. Mol. Cell Biol. 17 , 496–510 (2016).

Siparsky, P. N., Kirkendall, D. T. & Garrett, W. E. Muscle changes in aging. Sports Health 6 , 36–40 (2014).

Johnson, A. A. & Stolzing, A. The role of lipid metabolism in aging, lifespan regulation, and age-related disease. Aging Cell 18 , e13048 (2019).

Paganelli, R., Scala, E., Quinti, I. & Ansotegui, I. J. Humoral immunity in aging. Aging Clin. Exp. Res. 6 , 143–150 (1994).

Article   CAS   Google Scholar  

Goronzy, J. J. & Weyand, C. M. Understanding immunosenescence to improve responses to vaccines. Nat. Immunol. 14 , 428–436 (2013).

Cunha, L. L., Perazzio, S. F., Azzi, J., Cravedi, P. & Riella, L. V. Remodeling of the immune response with aging: immunosenescence and its potential impact on COVID-19 immune response. Front. Immunol. 11 , 1748 (2020).

Lee, D., Son, H. G., Jung, Y. & Lee, S.-J. V. The role of dietary carbohydrates in organismal aging. Cell. Mol. Life Sci. 74 , 1793–1803 (2017).

Franco-Juárez, B. et al. Effects of high dietary carbohydrate and lipid intake on the lifespan of C. elegans . Cells 10 , 2359 (2021).

Gold, L. et al. Aptamer-based multiplexed proteomic technology for biomarker discovery. PLoS ONE 5 , e15004 (2010).

Galliera, E., Tacchini, L. & Corsi Romanelli, M. M. Matrix metalloproteinases as biomarkers of disease: updates and new insights. Clin. Chem. Lab. Med. 53 , 349–355 (2015).

Golusda, L., Kühl, A. A., Siegmund, B. & Paclik, D. Extracellular matrix components as diagnostic tools in inflammatory bowel disease. Biology (Basel) 10 , 1024 (2021).

Zhou, W. et al. Longitudinal multi-omics of host–microbe dynamics in prediabetes. Nature 569 , 663–671 (2019).

Schüssler-Fiorenza Rose, et al. A longitudinal big data approach for precision health. Nat. Med. 25 , 792–804 (2019).

Hornburg, D. et al. Dynamic lipidome alterations associated with human health, disease and ageing. Nat. Metab. 5 , 1578–1594 (2023).

Zhou, X. et al. Longitudinal profiling of the microbiome at four body sites reveals core stability and individualized dynamics during health and disease. Cell Host Microbe 32 , 506–526 (2024).

Contreras, P. H., Serrano, F. G., Salgado, A. M. & Vigil, P. Insulin sensitivity and testicular function in a cohort of adult males suspected of being insulin-resistant. Front. Med. (Lausanne) 5 , 190 (2018).

Evans, D. J., Murray, R. & Kissebah, A. H. Relationship between skeletal muscle insulin resistance, insulin-mediated glucose disposal, and insulin binding. Effects of obesity and body fat topography. J. Clin. Invest. 74 , 1515–1525 (1984).

Röst, H. L., Schmitt, U., Aebersold, R. & Malmström, L. pyOpenMS: a Python-based interface to the OpenMS mass-spectrometry algorithm library. Proteomics 14 , 74–77 (2014).

Shen, X. et al. Normalization and integration of large-scale metabolomics data using support vector regression. Metabolomics 12 , 89 (2016).

Shen, X. et al. metID: an R package for automatable compound annotation for LC−MS-based data. Bioinformatics 38 , 568–569 (2022).

Marabita, F. et al. Multiomics and digital monitoring during lifestyle changes reveal independent dimensions of human biology and health. Cell Syst. 13 , 241–255 (2022).

Wu, T. et al. clusterProfiler 4.0: a universal enrichment tool for interpreting omics data. Innovation (Camb.) 2 , 100141 (2021).

Newman, M. E. J. & Girvan, M. Finding and evaluating community structure in networks. Phys. Rev. E 69 , 026113 (2004).

Shen, X., Wang, C. & Snyder, M. P. massDatabase: utilities for the operation of the public compound and pathway database. Bioinformatics 38 , 4650–4651 (2022).

Shen, X. et al. TidyMass an object-oriented reproducible analysis framework for LC–MS data. Nat. Commun. 13 , 4365 (2022).

Download references

Acknowledgements

We sincerely thank all the research participants for their dedicated involvement in this study. We also thank A. Chen and L. Stainton for their valuable administrative assistance. Additionally, we are deeply grateful to A.T. Brunger’s support for this work. This work was supported by National Institutes of Health (NIH) grants U54DK102556 (M.P.S.), R01 DK110186-03 (M.P.S.), R01HG008164 (M.P.S.), NIH S10OD020141 (M.P.S.), UL1 TR001085 (M.P.S.) and P30DK116074 (M.P.S.) and by the Stanford Data Science Initiative (M.P.S.). The funders had no role in study design, data collection and analysis, decision to publish or preparation of the manuscript.

Author information

These authors contributed equally: Xiaotao Shen, Chuchu Wang.

Authors and Affiliations

Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA

Xiaotao Shen, Xin Zhou, Wenyu Zhou, Daniel Hornburg, Si Wu & Michael P. Snyder

Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore

Xiaotao Shen

School of Chemistry, Chemical Engineering and Biotechnology, Singapore, Singapore

Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA

Chuchu Wang

Department of Molecular and Cellular Physiology, Stanford University, Stanford, CA, USA

Stanford Center for Genomics and Personalized Medicine, Stanford, CA, USA

Xin Zhou & Michael P. Snyder

You can also search for this author in PubMed   Google Scholar

Contributions

X.S. and M.P.S. conceptualized and designed the study. X.Z. and W.Z. prepared the microbiome data. D.H. and W.S. prepared the lipidomics data. X.S. and C.W. conducted the data analysis. X.S. and C.W. prepared the figures. X.S., C.W. and M.P.S. contributed to the writing and revision of the manuscript, with input from other authors. M.S. and X.S. supervised the overall study.

Corresponding author

Correspondence to Michael P. Snyder .

Ethics declarations

Competing interests.

M.P.S. is a co-founder of Personalis, SensOmics, Qbio, January AI, Filtricine, Protos and NiMo and is on the scientific advisory boards of Personalis, SensOmics, Qbio, January AI, Filtricine, Protos, NiMo and Genapsys. D.H. has a financial interest in PrognomIQ and Seer. All other authors have no competing interests.

Peer review

Peer review information.

Nature Aging thanks Daniel Belsky and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended data fig. 1 demographic data of all the participants in the study..

a , The ages positively correlate with BMI. The shaded area around the regression line represents the 95% confidence interval. b , Gender with age. c , Ethnicity with age. d , Insulin response with age. e , biological sample collection for all the participants. f , Overlap of the different kinds of omics data. g , The age range for each participant in this study.

Extended Data Fig. 2 Most of the molecules change nonlinearly during human aging.

a , Differential expressional microbes in different age ranges compared to baselines (25 – 40 years old, two-sided Wilcoxon test, p -value < 0.05). b , Most of the linear changing molecules and microbiota are also included in the molecules/microbes that significantly dysregulated at least one age range.

Extended Data Fig. 3 Omics data can represent aging.

PCA score plot of metabolomics data ( a ), cytokine ( b ), and oral microbiome ( c ).

Extended Data Fig. 4 Functional analysis of molecules in different clusters.

a , The Jaccard index between clusters from different datasets. b , The overlap between clusters using different types of omics data. c , Functional module detection and identification. d , Functional analysis of nonlinear changing molecules for all clusters.

Extended Data Fig. 5 Function annotation for significantly dysregulated molecules in crest 1 and 2.

a , Transcriptomics data. b , Proteomics data. c , Metabolomics data.

Extended Data Fig. 6 Pathways enrichment results for crest 1 and 2.

a , The final functional modules identified for Crest 1 and 2. b , The pathway enrichment analysis results for transcriptomics data. c , The pathway enrichment analysis results for proteomics data. d , The pathway enrichment results for metabolomics data.

Supplementary information

Supplementary figs. 1–6, reporting summary, supplementary data analysis results of the study., rights and permissions.

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ .

Reprints and permissions

About this article

Cite this article.

Shen, X., Wang, C., Zhou, X. et al. Nonlinear dynamics of multi-omics profiles during human aging. Nat Aging (2024). https://doi.org/10.1038/s43587-024-00692-2

Download citation

Received : 09 December 2023

Accepted : 22 July 2024

Published : 14 August 2024

DOI : https://doi.org/10.1038/s43587-024-00692-2

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Quick links

  • Explore articles by subject
  • Guide to authors
  • Editorial policies

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

atherosclerosis case study pdf

Pardon Our Interruption

As you were browsing something about your browser made us think you were a bot. There are a few reasons this might happen:

  • You've disabled JavaScript in your web browser.
  • You're a power user moving through this website with super-human speed.
  • You've disabled cookies in your web browser.
  • A third-party browser plugin, such as Ghostery or NoScript, is preventing JavaScript from running. Additional information is available in this support article .

To regain access, please make sure that cookies and JavaScript are enabled before reloading the page.

U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Arq Bras Cardiol
  • v.103(3); 2014 Sep

Logo of arqbrascard

Case 4/2014 - A 66-Year-Old Man with Acute Myocardial Infarction and Death in Asystole after Primary Coronary Angioplasty

A 66-year-old man sought medical care at the hospital due to severe chest pain lasting for 24 hours. The patient was aware of being hypertensive and was a smoker. Without any prior symptom, he started to have severe chest pain and sought emergency medical care after about 24 hours, due to pain persistence.

At physical examination (August 13, 2005, 10 PM) he had a heart rate of 90 bpm and blood pressure of 110/70 mmHg. Lung examination showed no alterations. Heart assessment showed a systolic murmur in the lower left sternal border and mitral area.

The initial electrocardiogram (August 13, 2005, 22 h) showed HR of 100 bpm, sinus rhythm, 1 st -degree atrioventricular block (PR 240 ms), low-voltage QRS complexes in the frontal plane, QRS complex electrical alternans and extensive ongoing anterior wall infarction (QS V1 to V6, ST elevation in the same leads and QS in the inferior wall, II, III and aVF) ( Figure 1 ).

An external file that holds a picture, illustration, etc.
Object name is abc-103-03-0e31-g01.jpg

ECG: low QRS voltage in the frontal plane, electrical alternans of QRS complexes, electrically inactive lower wall area and extensive ongoing myocardial infarction.

Acetylsalicylic acid by oral route and 5 mg of intravenous metoprolol were administered. The patient had bradycardia and cardiorespiratory arrest in pulseless electrical activity, reversed after five minutes. He developed hypotension and peripheral hypoperfusion and was transferred to InCor (The Heart Institute).

On admission he had received heparin and continuous intravenous norepinephrine. BP was 60/30 mmHg.

The ECG (August 13, 2005, 11:36 PM) disclosed heart rate of 116 bpm, junctional escape rhythm with sinus arrest and atrial extrasystoles); low-voltage QRS complex in the frontal plane, extensive ongoing anterior acute myocardial infarction, inactive area in the inferior wall; presence of ST elevation at V1 to V5 and ST depression in leads I, II and aVF; ST elevation in aVR ( Figure 2 ).

An external file that holds a picture, illustration, etc.
Object name is abc-103-03-0e31-g02.jpg

ECG: low QRS voltage in the frontal plane, electrically inactive lower wall area and anterior myocardial infarction with increased ST elevation, still with positive T waves, "hyperacute phase of myocardial infaction".

Coronary angiography was indicated, which disclosed anterior interventricular branch occlusion and images suggestive of intracoronary thrombus, lesion of 70% in the circumflex artery, 50% in the right coronary artery and 70% in the ostium of the right posterior descending branch. Angioplasty was performed with stent implant in the anterior interventricular artery, but distal flow was not restored. This was followed by cardiac arrest in asystole, which did not respond to treatment and the patient died.

Clinical Aspects

This clinical case reports on a 66-year-old hypertensive patient, long-term smoker, who sought medical care due to acute chest pain. The main diagnostic hypothesis for this clinical case is of acute coronary syndrome.

Chest pain is one of the most common reasons for seeking emergency care and remains a challenge for the clinician, due to the difficulty in differentiating between non-emergency diagnoses and those of high morbidity and mortality, such as acute coronary syndrome (ACS), aortic dissection and pulmonary thromboembolism.

In the assessment of acute chest pain, there are three basic parameters for its management: clinical examination (clinical history and physical examination), electrocardiogram (ECG) and myocardial necrosis markers. They should be analyzed together to provide a safer approach to the patient, especially when it is necessary ruling out ACS. Chest radiography, chest Computed Tomography Angiography (CTA), echocardiography and other tests may be useful in the differential diagnosis.

Approximately 15-25% of patients presenting in the ER with chest pain are diagnosed with acute coronary syndrome, and this represents its more frequent clinical manifestation 1 . Therefore, in the first step of the evaluation, which is the clinical examination, the greater determinant of an ischemic etiology is the characteristic presence of angina.

Angina is often described as a burning or compression sensation or difficulty breathing, located in the precordial region or any other region of the chest, radiating to the neck, shoulder and left arm. It usually increases in intensity within minutes and may be accompanied by symptoms such as nausea and sweating. It can be triggered by physical or emotional stress and relieved by rest or use of nitrates. It should also be remembered that ACS can occur without obvious precipitating factors and be asymptomatic or present as ischemic equivalent, especially in the elderly and diabetic patients with autonomic dysfunction (dyspnea, syncope and pre-syncope).

On the other hand, there are characteristics of pain that make the diagnosis of ACS unlikely, such as pleuritic pain (reproduced by respiratory movements) located with the fingertip, pain in meso/hypogastrium region and reproduction of pain with local palpation or movement. These features raise the suspicion of other differential diagnoses such as pericarditis, pleuritis, gastrointestinal or musculoskeletal diseases.

In the present case report, the patient presented with prolonged chest pain, which does not rule out acute coronary syndrome (ACS), but raises the possibility of some condition associated with this coronary picture, such as pericarditis or mechanical complications.

Among the most important risk factors for atherosclerotic disease risk are dyslipidemia, diabetes mellitus, hypertension, male gender, older age, obesity/metabolic syndrome, smoking, sedentary lifestyle, chronic kidney disease, depression and stress. This patient had some risk factors that contributed to the development of coronary artery disease: age, male gender, hypertension and smoking.

Patients with chest pain and ACS often have a nonspecific physical examination, with less than 20% of them showing significant alterations in the initial evaluation 2 . This becomes important by helping in the detection of differential diagnoses (e.g., pericardial friction rub in pericarditis) or by inferring the presence of risk factors for coronary artery disease (abdominal or carotid murmur, among others).

However, when findings resulting from an ACS are present, they indicate a worse prognosis due to mechanical complications or due to a large area of myocardium at risk and ventricular dysfunction (hypotension, tachycardia, pulmonary edema and mitral regurgitation murmur secondary to ischemia).

The electrocardiogram is important in the diagnostic, prognostic and therapeutic approach and must be obtained within 10 minutes after the presentation of patients with ongoing chest pain 2 . A normal electrocardiographic tracing does not exclude the possibility of ACS and a serial ECG is indicated, which increases its sensitivity and helps differentiating between acute and chronic alterations.

The patient reported in this clinical case had, at the admission ECG performed at another service, ST-segment elevation in the anterior wall, suggesting the hypothesis of acute coronary syndrome with ST-segment elevation. However, this ECG also showed low voltage and electrical alternans of the QRS complex, which suggests large pericardial effusion or even cardiac tamponade.

The main hypothesis for this pericardial effusion is a mechanical complication of myocardial infarction: left ventricular free wall rupture. It occurs within 24 hours after infarction or between the third and fifth day, has an incidence of 0.8 to 6.2% and is more common in an extensive myocardial infarction, in the elderly, women and hypertensive patients. Its clinical course is variable 3 and may be acute and severe, leading to sudden death or subacute, with nonspecific clinical manifestations.

Other mechanical complications that may be present in myocardial infarction are papillary muscle and interventricular septum rupture. These complications do not present with significant pericardial effusion and normal pulmonary auscultation in this patient also makes the diagnosis of papillary muscle rupture less likely. This clinical condition presents with pulmonary congestion due to volume overload secondary to acute mitral regurgitation.

Another diagnostic hypothesis for this patient presenting with chest pain and pericardial effusion would be aortic dissection. Pain, in these cases, is usually of sudden onset and strong intensity since the beginning (unlike angina pain, which often increasingly escalates). It is often described as excruciating and its location reflects the site and progression of the dissection. Autonomic signs (pallor, profuse sweating) are greatly associated.

In aortic dissection, physical examination may disclose hypertensive crisis, differences between limb pulses, signs of pleural and pericardial effusion, diastolic murmur of aortic regurgitation, different from the systolic murmur detected in this clinical case. The extension of the dissection to other vessels can lead to other symptoms corresponding to ischemia of the organs irrigated by them: cerebrovascular accident, acute myocardial infarction, mesenteric ischemia, etc.

Another diagnostic hypothesis for the clinical case is pulmonary embolism. The absence of pulmonary symptoms, mainly dyspnea, makes this hypothesis less likely. It is the most common symptom of this disease, occurring in over 78% of the patients 4 . Sudden chest pain of sudden onset and very often pleuritic, affects up to 44% of patients 4 . Cough and hemoptysis may also occur. Additionally, there was no mention is made on admission at the other service, of right ventricular dysfunction manifestations, such as jugular stasis and hypotension.

The patient, an hour and 36 minutes after his admission at the Heart Institute, was submitted to coronary angiography with left anterior descending artery angioplasty. However, he developed asystole and cardiac arrest.

The main diagnoses for the final clinical picture are cardiogenic shock and/or distributive shock due to cardiac tamponade, discussed below.

The hypothesis of cardiogenic shock should be considered, as the patient had an extensive acute myocardial infarction without culprit artery reperfusion even after percutaneous revascularization attempt. However, this diagnosis cannot fully explain the patient's clinical condition, such as the absence of pulmonary congestion, which usually follows an acute myocardial failure.

Considering the patient's history, late cardiac tamponade seems to have been the main precipitating factor of the final clinical picture in this case. The electrocardiographic findings commonly observed in cardiac tamponade are low voltage and electrical alternans of the QRS complex, observed in the case. Although physical examination made no reference to clinical findings suggestive of tamponade, such as jugular stasis or muffled heart sounds, we cannot exclude this diagnostic hypothesis.

An echocardiography could have been performed to confirm this diagnosis, which is the most widely used noninvasive method for diagnostic investigation of this pathology. The ventriculography in this context would not be informative, as it was a free wall rupture with cardiac tamponade and thus, it would not allow the visualization of contrast leakage into the pericardial cavity.

This is a patient with myocardial infarction that came at the emergency room more than 24 hours after the onset of the event and who probably had a mechanical complication of myocardial infarction: ventricular free wall rupture.

Most deaths from myocardial infarction occur in the first hours of disease onset, with 40-65% occurring within the first hour and approximately 80% in the first 24 hours 5 , 6 . The recently implemented therapies for MI treatment have been proven to modify patient evolution and prognosis. However, the effectiveness of most of these measures is time-dependent and delay in seeking medical care may have been the factor that likely contributed to the clinical outcome of the patient in this case report. (Dr. Wilma Noia Ribeiro, Dr. Alice Tatsuko Yamada)

Diagnostic hypotheses: Acute myocardial infarction with mixed shock (cardiogenic - distributive) by mechanical complication - free wall rupture with tamponade (Dr. Wilma Noia Ribeiro, Dr. Alice Tatsuko Yamada)

The heart weighed 414 g. The myocardium of the left ventricular anterosseptal wall and right ventricular anterior wall was softened, slightly yellowish in color, characterizing extensive transmural acute myocardial infarction. There was an obvious narrowing of the affected anterosseptal wall, with ventricular septum rupture in the anterior region of its mid portion, with a ventricular septal defect measuring 10 mm in its longest axis. The other left ventricular walls showed to be slightly hypertrophic and there was a small area of fibrosis in the postero-inferior region of the ventricular septum. There was also moderate right ventricular dilation ( Figure 3 ).

An external file that holds a picture, illustration, etc.
Object name is abc-103-03-0e31-g03.jpg

Cross-section of the ventricles showing left ventricular transmural infarction of the anterosseptal wall (asterisks) and of the right ventricular anterior wall (arrows). The explorer shows the VSD secondary to septal rupture. Observe the ventricular wall thinning and the small area of fibrosis in the ventricular septum (arrowhead).

Histological analysis confirmed the presence of myocardial infarction, with marked neutrophil infiltration, confirming histological dating of 24-48 hours of onset. Another small ongoing microinfarction was observed in the posterior region of the ventricular septum, in addition to the previously described small area of fibrosis (healed infarction), compatible with approximately 7-10 days of evolution.

There was superficial fibrin deposition in the epicardium, with the presence of reactive inflammatory infiltrate. Examination of the initial segment of the left anterior descending artery showed fatty atherosclerotic plaques with areas of marked thinning of the fibrous cap that covered the lipid core and 80% of obstruction.

There were also areas of plaque rupture and hemorrhage, with acute thrombosis in the first and second centimeters of that artery ( Figs. 4 and ​ and5 5 ).

An external file that holds a picture, illustration, etc.
Object name is abc-103-03-0e31-g04.jpg

Histological section of the first centimeter of the left anterior descending artery showing large lipid-core atherosclerotic plaque, with internal area of fibrin deposition and hemorrhage (asterisk). Observe the area with marked thinning of the fibrous cap of the lipid core plaque (arrow), site of potential rupture and thrombosis. Hematoxylin-eosin, 2.5×.

An external file that holds a picture, illustration, etc.
Object name is abc-103-03-0e31-g05.jpg

Histological section of the second centimeter of the anterior interventricular artery showing large lipid-core atherosclerotic plaque with ruptured area (arrows) and occlusive luminal thrombosis (asterisk). Hematoxylin-eosin, 2.5×.

The lungs weighed 1,208 g together and showed alveolar edema. The kidneys showed irregular surface and retention cysts, with hyaline arteriolosclerosis on histological examination. The aorta showed mild / moderate degree of atherosclerosis. (Dr. Luiz Alberto Benvenuti)

Anatomopathological diagnoses. Coronary atherosclerosis; acute myocardial infarction involving the left ventricular anterosseptal wall and the right ventricular anterior wall; rupture of the ventricular septum, with VSD; acute pulmonary edema (cause of death) (Dr. Luiz Alberto Benvenuti) .

This is the case report of a 66-year-old man with systemic hypertension and a chronic smoker that presented with acute severe chest pain. After medical assessment, he was diagnosed with acute myocardial infarction and the patient underwent coronary angiography, which disclosed proximal occlusion of the left anterior descending artery with images suggesting the presence of thrombi. He was submitted to balloon-angioplasty in the affected segment without restoration of distal coronary flow (unsuccessful procedure) and the patient developed irreversible cardiac arrest and died.

The autopsy confirmed acute myocardial infarction, which was very extensive, affecting the left ventricular anterosseptal wall and the right ventricular anterior wall. Histological dating was 24-48 hours of onset, consistent with the clinical history. It is noteworthy the fact that the detailed examination of the ventricular septum showed the presence of two previous microinfarctions, an old (healed) one and an ongoing one.

The presence of atherosclerosis of the coronary arteries was identified, with massive plaques in the proximal segment of the left anterior descending artery, which resulted in chronic obstruction of 80% of the lumen. The fatty plaques had extensive lipid cores and there were areas of marked thinning of the fibrous cap that covered the cores, as well as areas of rupture associated with acute thrombosis of the remaining lumen in the first two centimeters of the left anterior descending artery. It is known that acute coronary occlusions with luminal thrombosis are usually associated with large lipid-core plaques, which undergo rupture due to the instability of their thin fibrous cap 7 , as observed in this case.

Aside from the great extent of the infarcted area, the patient developed an important mechanical complication of acute myocardial infarction, the occurrence of ventricular septal rupture with the establishment of VSD 8 - which certainly aggravated his hemodynamic condition, progressing to cardiogenic shock -, acute pulmonary edema and death. It should be emphasized that the patient had two classic risk factors for atherosclerosis and myocardial infarction: systemic hypertension and chronic smoking 9 . (Dr. Luiz Alberto Benvenuti)

Section Editor: Alfredo José Mansur ( rb.psu.rocni@rusnamja )

Associated Editors: Desidério Favarato ( rb.psu.rocni@otaravaflcd )

Vera Demarchi Aiello ( rb.psu.rocni@arevpna )

COMMENTS

  1. 21-Year-old male with severe coronary atherosclerosis

    The patient received a coronary artery bypass surgery at just 21 years of age. He exhibits traditional risk factors of CHD; hypertension, dyslipidemia, cigarette smoking, and obesity. CHD may be more prevalent in the younger population than once was thought, and it is important that risk factor reduction starts in early in life.>.

  2. 21-Year-old male with severe coronary atherosclerosis

    The patient received a coronary artery bypass surgery at just 21 years of age. He exhibits traditional risk factors of CHD; hypertension, dyslipidemia, cigarette smoking, and obesity. CHD may be more prevalent in the younger population than once was thought, and it is important that risk factor reduction starts in early in life.>.

  3. Case report: Coronary atherosclerosis in a patient with long-standing

    Importantly, this case demonstrates that coronary atherosclerosis can occur even with long-standing LDL-C less than 40 mg/dl when other risk factors such as hypertension and diabetes are present and otherwise well controlled. Reports of atherosclerosis in individuals with lifelong very low LDL-C are extremely rare . The patient in this instance ...

  4. Case Report: Multiple atherosclerotic plaques at its extreme in

    This case is an important reminder to all clinicians that atherosclerosis often involves multiple arterial territories. The weaknesses of the case report include significant omitted details regarding the history, physical examination, and objective findings. The timeline of the patient's management is also unclear.

  5. A Prospective Natural-History Study of Coronary Atherosclerosis

    Between October 29, 2004, and June 8, 2006, a total of 697 patients with acute coronary syndromes were enrolled after they had undergone successful percutaneous coronary intervention ( Table 1 ...

  6. PDF Pathophysiology and treatment of atherosclerosis

    Atherosclerosis is a chronic condition in which arteries harden through build-up of plaques. Main classical risk factors for atherosclerosis include dyslipoproteinaemia, di-abetes, cigarette smoking, hypertension and genetic abnor-malities. In this review, we present an update on the patho-physiology of atherosclerosis and related current and ...

  7. (PDF) EC CARDIOLOGY Mini Case Study Atherosclerosis Presenting as

    Case Report A 65 year old asymptomatic female was suspected to have a distal ascending aortic aneurysm and a moderate Aortic Regurgitation with some aortic sclerosis on routine health checkup.

  8. Progression of Early Subclinical Atherosclerosis (PESA) Study:

    Introduction. Cardiovascular disease (CVD) is the leading cause of mortality and morbidity worldwide and represents the highest proportion of costs to health care systems (1,2).Global CVD prevalence is growing, and if this trend continues, it will threaten the ability of health care systems to cope with its consequences ().Most forms of CVD originate in atherosclerosis, which is thus the main ...

  9. Arteriosclerosis and Atherosclerosis

    Arteriosclerosis and Atherosclerosis 1215. 3. Sawabe M, Takahashi R, Matsushita S, Ozawa T, Arai T, Hamamatsu A, Nakahara K, Chida K, Yamanouchi H, Murayama S, Tanaka N. Aortic pulse wave velocity and the degree of atherosclerosis in the elderly: a pathological study based on 304 autopsy cases. Atherosclerosis. 2005; 179:345-351.

  10. (PDF) A Multiscale and Patient-specific Computational Framework of

    A Multiscale and Patient-specific Computational Framework of Atherosclerosis Formation and Progression: A Case Study in the Aorta and Peripheral Arteri es.pdf Content available from CC BY-NC-ND 4.0:

  11. IJMS

    Atherosclerosis is the main risk factor for cardiovascular disease (CVD), which is the leading cause of mortality worldwide. Atherosclerosis is initiated by endothelium activation and, followed by a cascade of events (accumulation of lipids, fibrous elements, and calcification), triggers the vessel narrowing and activation of inflammatory pathways. The resultant atheroma plaque, along with ...

  12. Frontiers

    Importantly, this case demonstrates that coronary atherosclerosis can occur even with long-standing LDL-C less than 40 mg/dl when other risk factors such as hypertension and diabetes are present and otherwise well controlled. Reports of atherosclerosis in individuals with lifelong very low LDL-C are extremely rare . The patient in this instance ...

  13. Coronary Artery Disease

    Discovery of these common variants comes at the heel of large genome-wide association studies now being conducted on an increasingly larger scale. 2,3 As the name of these ... Atherosclerosis. 2011;219:799-806. doi: 10.1016/j.atherosclerosis ... a prospective case-control study. Lancet Diabetes Endocrinol. 2015;3:507-513. doi: 10. ...

  14. Atherosclerosis: Process, Indicators, Risk Factors and New Hopes

    Atherosclerosis is the major cause of morbidities and mortalities worldwide. In this study we aimed to review the mechanism of atherosclerosis and its risk factors, focusing on new findings in atherosclerosis markers and its risk factors. Furthermore, the role of antioxidants and medicinal herbs in atherosclerosis and endothelial damage has ...

  15. A Prospective Natural-History Study of Coronary Atherosclerosis

    Between October 29, 2004, and June 8, 2006, a total of 697 patients with acute coronary syn-dromes were enrolled after they had undergone successful percutaneous coronary intervention (Table 1 ...

  16. Cholesterol and Atherosclerotic Cardiovascular Disease: A Lifelong

    In this issue of the Journal of the American Heart Association (JAHA), Duncan et al apply the technique of trajectory analysis to a more‐contemporary cohort of the Framingham Heart Study, followed for 35 years to determine the relationship of lifelong exposure to elevated LDL cholesterol and low high‐density lipoprotein cholesterol to ASCVD and total mortality. 6 For LDL cholesterol, they ...

  17. Clinical case: The development of atherosclerosis in a patient 28 years

    Aim: The purpose of the study was to determine the presence of cardiovascular disease in young patient with a rare monogenic form of diabetes mellitus (MODY12). Clinical case: The development of atherosclerosis in a patient 28 years old with 12 mody diabetes - Atherosclerosis

  18. A 52-Year-Old Man With Atherosclerosis

    His family history is notable for a myocardial infarction (MI) in his father at the age of 52 years. He is a lifelong non-smoker. He does not take medications. His blood pressure was 110/75. His exam was notable for being overweight with a BMI of 27, but was otherwise unremarkable. His total cholesterol is 206 mg/dL, HDL-C is 46 mg/dL ...

  19. Atherosclerosis

    Atherosclerosis is a chronic inflammatory disease of the arteries and is the underlying cause of about 50% of all deaths in westernized society. It is principally a lipid-driven process initiated by the accumulation of low-density lipoprotein and remnant lipoprotein particles and an active inflammatory process in focal areas of arteries particularly at regions of disturbed non-laminar flow at ...

  20. Study on Correlation of Homocysteine Level and Ischemic Stroke

    Epidemiologic studies have identified hyperhomocysteinemia as a possible risk factor for atherosclerosis. We determined the risk of carotid-artery atherosclerosis in relation to both plasma ...

  21. Nonlinear dynamics of multi-omics profiles during human aging

    Aging is a complex and multifactorial process of physiological changes strongly associated with various human diseases, including cardiovascular diseases (CVDs), diabetes, neurodegeneration and ...

  22. Updates on Approaches for Studying Atherosclerosis

    Many animal models have been used to study atherosclerosis, as summarized in the recent American Heart Association Scientific Statement, 3 which provide suggestions on selecting appropriate animal models for a specific atherosclerosis study. Given its value and feasibility as a proof-of-principle model, mouse models have continued to be the most common species to study atherosclerosis.

  23. Case 5/2015

    The microscopic study of the coronary arteries revealed recent partial thrombosis on the seventh cm of the right coronary artery, in addition to atherosclerosis with 80% obstruction. The anterior interventricular and circumflex branches of the left coronary artery had stents placed several years before and were submitted to special processing ...

  24. Nervous Tissue-Case Study (pdf)

    Nervous System case study Name:_____ Going Under the Knife: A Case on Membrane Structure and Function Twenty-year-old Kevin groaned and clutched his abdomen as he lay on the emergency room gurney. He had just been diagnosed with acute appendicitis and was waiting to be taken to the operating room (OR). Although he desperately wanted the pain to stop, Kevin was terrified of having general ...

  25. Case 4/2014

    This is the case report of a 66-year-old man with systemic hypertension and a chronic smoker that presented with acute severe chest pain. After medical assessment, he was diagnosed with acute myocardial infarction and the patient underwent coronary angiography, which disclosed proximal occlusion of the left anterior descending artery with ...