Skip to main content

Advertisement

Log in

Research Progress on the Involvement of ANGPTL4 and Loss-of-Function Variants in Lipid Metabolism and Coronary Heart Disease: Is the “Prime Time” of ANGPTL4-Targeted Therapy for Coronary Heart Disease Approaching?

  • Review Article
  • Published:
Cardiovascular Drugs and Therapy Aims and scope Submit manuscript

Abstract

Background

Multiple genetic studies have confirmed the definitive link among the loss-of-function variants of angiogenin-like protein 4 (ANGPTL4), significantly decreased plasma triglyceride (TG) levels, and reduced risk of coronary heart disease (CHD). The potential therapeutic effect of ANGPTL4 on dyslipidemia and CHD has been widely studied.

Objective

This review provides a detailed introduction to the research progress on the involvement of ANGPTL4 in lipid metabolism and atherosclerosis and evaluates the efficacy and safety of ANGPTL4 as a therapeutic target for CHD.

Relevant Findings

By inhibiting lipoprotein lipase (LPL) activity, ANGPTL4 plays a vital role in the regulation of lipid metabolism and energy balance. However, the role of ANGPTL4 in regulating lipid metabolism is tissue-specific. ANGPTL4 acts as a locally released LPL inhibitor in the heart, skeletal muscle and small intestine, while ANGPTL4 derived from liver and adipose tissue mainly acts as an endocrine factor that regulates systemic lipid metabolism. As a multifunctional protein, ANGPTL4 also inhibits the formation of foam cells in macrophages, exerting an anti-atherogenic role. The function of ANGPTL4 in endothelial cells is still uncertain. The safety of ANGPTL4 monoclonal antibodies requires further evaluation due to their potential adverse effects.

Conclusion

The biological characteristics of ANGPTL4 are much more complex than those demonstrated by genetic studies. Future studies must elucidate how to effectively reduce the risk of CHD while avoiding potential atherogenic effects and other complications before the “prime time” of ANGPTL4-targeted therapy arrives.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Rader DJ. New therapeutic approaches to the treatment of Dyslipidemia. Cell Metab. 2016;23:405–12.

    CAS  PubMed  Google Scholar 

  2. Jellinger PS, Smith DA, Mehta AE, Ganda O, Handelsman Y, Rodbard HW, et al. American Association of Clinical Endocrinologists' guidelines for Management of Dyslipidemia and Prevention of atherosclerosis. Endocr Pract. 2012;18(Suppl 1):1–78.

    PubMed  Google Scholar 

  3. Catapano AL, Graham I, De Backer G, Wiklund O, Chapman MJ, Drexel H, et al. 2016 ESC/EAS guidelines for the Management of Dyslipidaemias: the task force for the Management of Dyslipidaemias of the European Society of Cardiology (ESC) and European atherosclerosis society (EAS) developed with the special contribution of the European Assocciation for Cardiovascular Prevention & Rehabilitation (EACPR). Atherosclerosis. 2016;253:281–344.

    CAS  PubMed  Google Scholar 

  4. Toth PP, Granowitz C, Hull M, Liassou D, Anderson A, Philip S. High triglycerides are associated with increased cardiovascular events, medical costs, and resource use: a real-world administrative claims analysis of statin-treated patients with high residual cardiovascular risk. J Am Heart Assoc. 2018;7:e008740.

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Toth PP. Triglyceride-rich lipoproteins as a causal factor for cardiovascular disease. Vasc Health Risk Manag. 2016;12:171–83.

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Mach F, Baigent C, Catapano AL, Koskinas KC, Casula M, Badimon L, et al. 2019 ESC/EAS guidelines for the management of dyslipidaemias: lipid modification to reduce cardiovascular risk. Eur Heart J. 2020;41:111–88.

    PubMed  Google Scholar 

  7. Rosenson RS, Davidson MH, Hirsh BJ, Kathiresan S, Gaudet D. Genetics and causality of triglyceride-rich lipoproteins in atherosclerotic cardiovascular disease. J Am Coll Cardiol. 2014;64:2525–40.

    CAS  PubMed  Google Scholar 

  8. Kersten S. Physiological regulation of lipoprotein lipase. Biochim Biophys Acta. 1841;2014:919–33.

    Google Scholar 

  9. Dijk W, Kersten S. Regulation of lipid metabolism by angiopoietin-like proteins. Curr Opin Lipidol. 2016;27:249–56.

    CAS  PubMed  Google Scholar 

  10. Graham MJ, Lee RG, Brandt TA, Tai LJ, Fu W, Peralta R, et al. Cardiovascular and metabolic effects of ANGPTL3 antisense oligonucleotides. N Engl J Med. 2017;377:222–32.

    CAS  PubMed  Google Scholar 

  11. Dewey FE, Gusarova V, Dunbar RL, O'Dushlaine C, Schurmann C, Gottesman O, et al. Genetic and pharmacologic inactivation of ANGPTL3 and cardiovascular disease. N Engl J Med. 2017;377:211–21.

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Gusarova V, Alexa CA, Wang Y, Rafique A, Kim JH, Buckler D, et al. ANGPTL3 blockade with a human monoclonal antibody reduces plasma lipids in dyslipidemic mice and monkeys. J Lipid Res. 2015;56:1308–17.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Romeo S, Pennacchio LA, Fu Y, Boerwinkle E, Tybjaerg-Hansen A, Hobbs HH, et al. Population-based resequencing of ANGPTL4 uncovers variations that reduce triglycerides and increase HDL. Nat Genet. 2007;39:513–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Folsom AR, Peacock JM, Demerath E, Boerwinkle E. Variation in ANGPTL4 and risk of coronary heart disease: the atherosclerosis risk in communities study. Metabolism. 2008;57:1591–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Dewey FE, Gusarova V, O'Dushlaine C, Gottesman O, Trejos J, Hunt C, et al. Inactivating variants in ANGPTL4 and risk of coronary artery disease. N Engl J Med. 2016;374:1123–33.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Myocardial Infarction G, Investigators CAEC, Stitziel NO, Stirrups KE, Masca NG, Erdmann J, et al. Coding variation in ANGPTL4, LPL, and SVEP1 and the risk of coronary disease. N Engl J Med. 2016;374:1134–44.

    Google Scholar 

  17. Morisada T, Kubota Y, Urano T, Suda T, Oike Y. Angiopoietins and angiopoietin-like proteins in angiogenesis. Endothelium. 2006;13:71–9.

    CAS  PubMed  Google Scholar 

  18. Wan D, Gong Y, Qin W, Zhang P, Li J, Wei L, et al. Large-scale cDNA transfection screening for genes related to cancer development and progression. Proc Natl Acad Sci U S A. 2004;101:15724–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Kim I, Kim HG, Kim H, Kim HH, Park SK, Uhm CS, et al. Hepatic expression, synthesis and secretion of a novel fibrinogen/angiopoietin-related protein that prevents endothelial-cell apoptosis. Biochem J. 2000;346(Pt 3):603–10.

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Aryal B, Price NL, Suarez Y, Fernandez-Hernando C. ANGPTL4 in metabolic and cardiovascular disease. Trends Mol Med. 2019;25:723–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Ge H, Yang G, Huang L, Motola DL, Pourbahrami T, Li C. Oligomerization and regulated proteolytic processing of angiopoietin-like protein 4. J Biol Chem. 2004;279:2038–45.

    CAS  PubMed  Google Scholar 

  22. Grootaert C, Van de Wiele T, Verstraete W, Bracke M, Vanhoecke B. Angiopoietin-like protein 4: health effects, modulating agents and structure-function relationships. Expert Rev Proteomics. 2012;9:181–99.

    CAS  PubMed  Google Scholar 

  23. Lei X, Shi F, Basu D, Huq A, Routhier S, Day R, et al. Proteolytic processing of angiopoietin-like protein 4 by proprotein convertases modulates its inhibitory effects on lipoprotein lipase activity. J Biol Chem. 2011;286:15747–56.

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Zhang R. The ANGPTL3-4-8 model, a molecular mechanism for triglyceride trafficking. Open Biol. 2016;6:150272.

    PubMed  PubMed Central  Google Scholar 

  25. Dutton S, Trayhurn P. Regulation of angiopoietin-like protein 4/fasting-induced adipose factor (Angptl4/FIAF) expression in mouse white adipose tissue and 3T3-L1 adipocytes. Br J Nutr. 2008;100:18–26.

    CAS  PubMed  Google Scholar 

  26. Kersten S, Mandard S, Tan NS, Escher P, Metzger D, Chambon P, et al. Characterization of the fasting-induced adipose factor FIAF, a novel peroxisome proliferator-activated receptor target gene. J Biol Chem. 2000;275:28488–93.

    CAS  PubMed  Google Scholar 

  27. Catoire M, Alex S, Paraskevopulos N, Mattijssen F, Evers-van Gogh I, Schaart G, et al. Fatty acid-inducible ANGPTL4 governs lipid metabolic response to exercise. Proc Natl Acad Sci U S A. 2014;111:E1043–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Josephs T, Waugh H, Kokay I, Grattan D, Thompson M. Fasting-induced adipose factor identified as a key adipokine that is up-regulated in white adipose tissue during pregnancy and lactation in the rat. J Endocrinol. 2007;194:305–12.

    CAS  PubMed  Google Scholar 

  29. Mandard S, Zandbergen F, Tan NS, Escher P, Patsouris D, Koenig W, et al. The direct peroxisome proliferator-activated receptor target fasting-induced adipose factor (FIAF/PGAR/ANGPTL4) is present in blood plasma as a truncated protein that is increased by fenofibrate treatment. J Biol Chem. 2004;279:34411–20.

    CAS  PubMed  Google Scholar 

  30. Belanger AJ, Lu H, Date T, Liu LX, Vincent KA, Akita GY, et al. Hypoxia up-regulates expression of peroxisome proliferator-activated receptor gamma angiopoietin-related gene (PGAR) in cardiomyocytes: role of hypoxia inducible factor 1alpha. J Mol Cell Cardiol. 2002;34:765–74.

    CAS  PubMed  Google Scholar 

  31. Koliwad SK, Kuo T, Shipp LE, Gray NE, Backhed F, So AY, et al. Angiopoietin-like 4 (ANGPTL4, fasting-induced adipose factor) is a direct glucocorticoid receptor target and participates in glucocorticoid-regulated triglyceride metabolism. J Biol Chem. 2009;284:25593–601.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Kaplan R, Zhang T, Hernandez M, Gan FX, Wright SD, Waters MG, et al. Regulation of the angiopoietin-like protein 3 gene by LXR. J Lipid Res. 2003;44:136–43.

    CAS  PubMed  Google Scholar 

  33. Helgadottir A, Gretarsdottir S, Thorleifsson G, Hjartarson E, Sigurdsson A, Magnusdottir A, et al. Variants with large effects on blood lipids and the role of cholesterol and triglycerides in coronary disease. Nat Genet. 2016;48:634–9.

    CAS  PubMed  Google Scholar 

  34. Abid K, Trimeche T, Mili D, Msolli MA, Trabelsi I, Nouira S, et al. ANGPTL4 variants E40K and T266M are associated with lower fasting triglyceride levels and predicts cardiovascular disease risk in type 2 diabetic Tunisian population. Lipids Health Dis. 2016;15:63.

    PubMed  PubMed Central  Google Scholar 

  35. Smart-Halajko MC, Robciuc MR, Cooper JA, Jauhiainen M, Kumari M, Kivimaki M, et al. The relationship between plasma angiopoietin-like protein 4 levels, angiopoietin-like protein 4 genotype, and coronary heart disease risk. Arterioscler Thromb Vasc Biol. 2010;30:2277–82.

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Staiger H, Machicao F, Werner R, Guirguis A, Weisser M, Stefan N, et al. Genetic variation within the ANGPTL4 gene is not associated with metabolic traits in white subjects at an increased risk for type 2 diabetes mellitus. Metabolism. 2008;57:637–43.

    CAS  PubMed  Google Scholar 

  37. Talmud PJ, Smart M, Presswood E, Cooper JA, Nicaud V, Drenos F, et al. ANGPTL4 E40K and T266M: effects on plasma triglyceride and HDL levels, postprandial responses, and CHD risk. Arterioscler Thromb Vasc Biol. 2008;28:2319–25.

    CAS  PubMed  Google Scholar 

  38. Lichtenstein L, Berbee JF, van Dijk SJ, van Dijk KW, Bensadoun A, Kema IP, et al. Angptl4 upregulates cholesterol synthesis in liver via inhibition of LPL- and HL-dependent hepatic cholesterol uptake. Arterioscler Thromb Vasc Biol. 2007;27:2420–7.

    CAS  PubMed  Google Scholar 

  39. Lafferty MJ, Bradford KC, Erie DA, Neher SB. Angiopoietin-like protein 4 inhibition of lipoprotein lipase: evidence for reversible complex formation. J Biol Chem. 2013;288:28524–34.

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Beigneux AP, Allan CM, Sandoval NP, Cho GW, Heizer PJ, Jung RS, et al. Lipoprotein lipase is active as a monomer. Proc Natl Acad Sci U S A. 2019;116:6319–28.

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Dijk W, Beigneux AP, Larsson M, Bensadoun A, Young SG, Kersten S. Angiopoietin-like 4 promotes intracellular degradation of lipoprotein lipase in adipocytes. J Lipid Res. 2016;57:1670–83.

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Dijk W, Ruppert PMM, Oost LJ, Kersten S. Angiopoietin-like 4 promotes the intracellular cleavage of lipoprotein lipase by PCSK3/furin in adipocytes. J Biol Chem. 2018;293:14134–45.

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Davies BS, Beigneux AP, Barnes RH 2nd, Tu Y, Gin P, Weinstein MM, et al. GPIHBP1 is responsible for the entry of lipoprotein lipase into capillaries. Cell Metab. 2010;12:42–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Sonnenburg WK, Yu D, Lee EC, Xiong W, Gololobov G, Key B, et al. GPIHBP1 stabilizes lipoprotein lipase and prevents its inhibition by angiopoietin-like 3 and angiopoietin-like 4. J Lipid Res. 2009;50:2421–9.

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Mysling S, Kristensen KK, Larsson M, Kovrov O, Bensadouen A, Jorgensen TJ, et al. The angiopoietin-like protein ANGPTL4 catalyzes unfolding of the hydrolase domain in lipoprotein lipase and the endothelial membrane protein GPIHBP1 counteracts this unfolding. Elife. 2016;5.

  46. Beigneux AP, Davies BS, Gin P, Weinstein MM, Farber E, Qiao X, et al. Glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 plays a critical role in the lipolytic processing of chylomicrons. Cell Metab. 2007;5:279–91.

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Beigneux AP, Davies BS, Bensadoun A, Fong LG, Young SG. GPIHBP1, a GPI-anchored protein required for the lipolytic processing of triglyceride-rich lipoproteins. J Lipid Res. 2009;50(Suppl):S57–62.

    PubMed  PubMed Central  Google Scholar 

  48. Kersten S. Angiopoietin-like 3 in lipoprotein metabolism. Nat Rev Endocrinol. 2017;13:731–9.

    CAS  PubMed  Google Scholar 

  49. Chi X, Britt EC, Shows HW, Hjelmaas AJ, Shetty SK, Cushing EM, et al. ANGPTL8 promotes the ability of ANGPTL3 to bind and inhibit lipoprotein lipase. Mol Metab. 2017;6:1137–49.

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Haller JF, Mintah IJ, Shihanian LM, Stevis P, Buckler D, Alexa-Braun CA, et al. ANGPTL8 requires ANGPTL3 to inhibit lipoprotein lipase and plasma triglyceride clearance. J Lipid Res. 2017;58:1166–73.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Ge H, Cha JY, Gopal H, Harp C, Yu X, Repa JJ, et al. Differential regulation and properties of angiopoietin-like proteins 3 and 4. J Lipid Res. 2005;46:1484–90.

    CAS  PubMed  Google Scholar 

  52. Guo L, Li SY, Ji FY, Zhao YF, Zhong Y, Lv XJ, et al. Role of Angptl4 in vascular permeability and inflammation. Inflamm Res. 2014;63:13–22.

    CAS  PubMed  Google Scholar 

  53. Mead JR, Irvine SA, Ramji DP. Lipoprotein lipase: structure, function, regulation, and role in disease. J Mol Med (Berl). 2002;80:753–69.

    CAS  Google Scholar 

  54. Yoshida K, Shimizugawa T, Ono M, Furukawa H. Angiopoietin-like protein 4 is a potent hyperlipidemia-inducing factor in mice and inhibitor of lipoprotein lipase. J Lipid Res. 2002;43:1770–2.

    CAS  PubMed  Google Scholar 

  55. Mandard S, Zandbergen F, van Straten E, Wahli W, Kuipers F, Muller M, et al. The fasting-induced adipose factor/angiopoietin-like protein 4 is physically associated with lipoproteins and governs plasma lipid levels and adiposity. J Biol Chem. 2006;281:934–44.

    CAS  PubMed  Google Scholar 

  56. Aryal B, Singh AK, Zhang X, Varela L, Rotllan N, Goedeke L, et al. Absence of ANGPTL4 in adipose tissue improves glucose tolerance and attenuates atherogenesis. JCI Insight. 2018;3.

  57. Singh AK, Aryal B, Chaube B, Rotllan N, Varela L, Horvath TL, et al. Brown adipose tissue derived ANGPTL4 controls glucose and lipid metabolism and regulates thermogenesis. Mol Metab. 2018;11:59–69.

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Dijk W, Heine M, Vergnes L, Boon MR, Schaart G, Hesselink MK, et al. ANGPTL4 mediates shuttling of lipid fuel to brown adipose tissue during sustained cold exposure. Elife. 2015;4.

  59. Semenkovich CF, Chen SH, Wims M, Luo CC, Li WH, Chan L. Lipoprotein lipase and hepatic lipase mRNA tissue specific expression, developmental regulation, and evolution. J Lipid Res. 1989;30:423–31.

    CAS  PubMed  Google Scholar 

  60. Koster A, Chao YB, Mosior M, Ford A, Gonzalez-DeWhitt PA, Hale JE, et al. Transgenic angiopoietin-like (angptl)4 overexpression and targeted disruption of angptl4 and angptl3: regulation of triglyceride metabolism. Endocrinology. 2005;146:4943–50.

    CAS  PubMed  Google Scholar 

  61. Georgiadi A, Lichtenstein L, Degenhardt T, Boekschoten MV, van Bilsen M, Desvergne B, et al. Induction of cardiac Angptl4 by dietary fatty acids is mediated by peroxisome proliferator-activated receptor beta/delta and protects against fatty acid-induced oxidative stress. Circ Res. 2010;106:1712–21.

    CAS  PubMed  Google Scholar 

  62. Yu X, Burgess SC, Ge H, Wong KK, Nassem RH, Garry DJ, et al. Inhibition of cardiac lipoprotein utilization by transgenic overexpression of Angptl4 in the heart. Proc Natl Acad Sci U S A. 2005;102:1767–72.

    CAS  PubMed  PubMed Central  Google Scholar 

  63. An D, Pulinilkunnil T, Qi D, Ghosh S, Abrahani A, Rodrigues B. The metabolic "switch" AMPK regulates cardiac heparin-releasable lipoprotein lipase. Am J Physiol Endocrinol Metab. 2005;288:E246–53.

    CAS  PubMed  Google Scholar 

  64. Robciuc MR, Skrobuk P, Anisimov A, Olkkonen VM, Alitalo K, Eckel RH, et al. Angiopoietin-like 4 mediates PPAR delta effect on lipoprotein lipase-dependent fatty acid uptake but not on beta-oxidation in myotubes. PLoS One. 2012;7:e46212.

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Staiger H, Haas C, Machann J, Werner R, Weisser M, Schick F, et al. Muscle-derived angiopoietin-like protein 4 is induced by fatty acids via peroxisome proliferator-activated receptor (PPAR)-delta and is of metabolic relevance in humans. Diabetes. 2009;58:579–89.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Davies PJ, Berry SA, Shipley GL, Eckel RH, Hennuyer N, Crombie DL, et al. Metabolic effects of rexinoids: tissue-specific regulation of lipoprotein lipase activity. Mol Pharmacol. 2001;59:170–6.

    CAS  PubMed  Google Scholar 

  67. Seip RL, Angelopoulos TJ, Semenkovich CF. Exercise induces human lipoprotein lipase gene expression in skeletal muscle but not adipose tissue. Am J Phys. 1995;268:E229–36.

    CAS  Google Scholar 

  68. Seip RL, Mair K, Cole TG, Semenkovich CF. Induction of human skeletal muscle lipoprotein lipase gene expression by short-term exercise is transient. Am J Phys. 1997;272:E255–61.

    CAS  Google Scholar 

  69. Catoire M, Mensink M, Boekschoten MV, Hangelbroek R, Muller M, Schrauwen P, et al. Pronounced effects of acute endurance exercise on gene expression in resting and exercising human skeletal muscle. PLoS One. 2012;7:e51066.

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Thomson DM, Brown JD, Fillmore N, Ellsworth SK, Jacobs DL, Winder WW, et al. AMP-activated protein kinase response to contractions and treatment with the AMPK activator AICAR in young adult and old skeletal muscle. J Physiol. 2009;587:2077–86.

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Mattijssen F, Alex S, Swarts HJ, Groen AK, van Schothorst EM, Kersten S. Angptl4 serves as an endogenous inhibitor of intestinal lipid digestion. Mol Metab. 2014;3:135–44.

    CAS  PubMed  Google Scholar 

  72. Alex S, Lange K, Amolo T, Grinstead JS, Haakonsson AK, Szalowska E, et al. Short-chain fatty acids stimulate angiopoietin-like 4 synthesis in human colon adenocarcinoma cells by activating peroxisome proliferator-activated receptor gamma. Mol Cell Biol. 2013;33:1303–16.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Backhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy A, et al. The gut microbiota as an environmental factor that regulates fat storage. Proc Natl Acad Sci U S A. 2004;101:15718–23.

    PubMed  PubMed Central  Google Scholar 

  74. Tremaroli V, Backhed F. Functional interactions between the gut microbiota and host metabolism. Nature. 2012;489:242–9.

    CAS  PubMed  Google Scholar 

  75. Korecka A, de Wouters T, Cultrone A, Lapaque N, Pettersson S, Dore J, et al. ANGPTL4 expression induced by butyrate and rosiglitazone in human intestinal epithelial cells utilizes independent pathways. Am J Physiol Gastrointest Liver Physiol. 2013;304:G1025–37.

    CAS  PubMed  Google Scholar 

  76. Backhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc Natl Acad Sci U S A. 2007;104:979–84.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Adachi H, Fujiwara Y, Kondo T, Nishikawa T, Ogawa R, Matsumura T, et al. Angptl 4 deficiency improves lipid metabolism, suppresses foam cell formation and protects against atherosclerosis. Biochem Biophys Res Commun. 2009;379:806–11.

    CAS  PubMed  Google Scholar 

  78. Do R, Willer CJ, Schmidt EM, Sengupta S, Gao C, Peloso GM, et al. Common variants associated with plasma triglycerides and risk for coronary artery disease. Nat Genet. 2013;45:1345–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Georgiadi A, Wang Y, Stienstra R, Tjeerdema N, Janssen A, Stalenhoef A, et al. Overexpression of angiopoietin-like protein 4 protects against atherosclerosis development. Arterioscler Thromb Vasc Biol. 2013;33:1529–37.

    CAS  PubMed  Google Scholar 

  80. Makoveichuk E, Sukonina V, Kroupa O, Thulin P, Ehrenborg E, Olivecrona T, et al. Inactivation of lipoprotein lipase occurs on the surface of THP-1 macrophages where oligomers of angiopoietin-like protein 4 are formed. Biochem Biophys Res Commun. 2012;425:138–43.

    CAS  PubMed  Google Scholar 

  81. Moore KJ, Tabas I. Macrophages in the pathogenesis of atherosclerosis. Cell. 2011;145:341–55.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Doodnauth SA, Grinstein S, Maxson ME. Constitutive and stimulated macropinocytosis in macrophages: roles in immunity and in the pathogenesis of atherosclerosis. Philos Trans R Soc Lond Ser B Biol Sci. 2019;374:20180147.

    CAS  Google Scholar 

  83. Lichtenstein L, Mattijssen F, de Wit NJ, Georgiadi A, Hooiveld GJ, van der Meer R, et al. Angptl4 protects against severe proinflammatory effects of saturated fat by inhibiting fatty acid uptake into mesenteric lymph node macrophages. Cell Metab. 2010;12:580–92.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Aryal B, Rotllan N, Araldi E, Ramirez CM, He S, Chousterman BG, et al. ANGPTL4 deficiency in haematopoietic cells promotes monocyte expansion and atherosclerosis progression. Nat Commun. 2016;7:12313.

    CAS  PubMed  PubMed Central  Google Scholar 

  85. Gimbrone MA Jr, Bevilacqua MP, Cybulsky MI. Endothelial-dependent mechanisms of leukocyte adhesion in inflammation and atherosclerosis. Ann N Y Acad Sci. 1990;598:77–85.

    PubMed  Google Scholar 

  86. Foteinos G, Xu Q. Immune-mediated mechanisms of endothelial damage in atherosclerosis. Autoimmunity. 2009;42:627–33.

    CAS  PubMed  Google Scholar 

  87. Li H, Ge C, Zhao F, Yan M, Hu C, Jia D, et al. Hypoxia-inducible factor 1 alpha-activated angiopoietin-like protein 4 contributes to tumor metastasis via vascular cell adhesion molecule-1/integrin beta1 signaling in human hepatocellular carcinoma. Hepatology. 2011;54:910–9.

    CAS  PubMed  Google Scholar 

  88. Galaup A, Cazes A, Le Jan S, Philippe J, Connault E, Le Coz E, et al. Angiopoietin-like 4 prevents metastasis through inhibition of vascular permeability and tumor cell motility and invasiveness. Proc Natl Acad Sci U S A. 2006;103:18721–6.

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Huang RL, Teo Z, Chong HC, Zhu P, Tan MJ, Tan CK, et al. ANGPTL4 modulates vascular junction integrity by integrin signaling and disruption of intercellular VE-cadherin and claudin-5 clusters. Blood. 2011;118:3990–4002.

    CAS  PubMed  Google Scholar 

  90. Wang J, Xian X, Huang W, Chen L, Wu L, Zhu Y, et al. Expression of LPL in endothelial-intact artery results in lipid deposition and vascular cell adhesion molecule-1 upregulation in both LPL and ApoE-deficient mice. Arterioscler Thromb Vasc Biol. 2007;27:197–203.

    PubMed  Google Scholar 

  91. Stitziel NO, Khera AV, Wang X, Bierhals AJ, Vourakis AC, Sperry AE, et al. ANGPTL3 deficiency and protection against coronary artery disease. J Am Coll Cardiol. 2017;69:2054–63.

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was supported by National Natural Scientific Funding of China (No. 81672264 and No. 81871858).

Author information

Authors and Affiliations

Authors

Contributions

Danyan Xu conceived of the scope of the review and helped revise the manuscript. Jingmin Yang was involved in the collection of the relevant references and drafted the manuscript. Xiao Li helped revise the manuscript and created the figure and table. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Danyan Xu.

Ethics declarations

Conflict of Interest

There are no conflicts of interest.

Additional information

Publisher’s Note

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

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yang, J., Li, X. & Xu, D. Research Progress on the Involvement of ANGPTL4 and Loss-of-Function Variants in Lipid Metabolism and Coronary Heart Disease: Is the “Prime Time” of ANGPTL4-Targeted Therapy for Coronary Heart Disease Approaching?. Cardiovasc Drugs Ther 35, 467–477 (2021). https://doi.org/10.1007/s10557-020-07001-0

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10557-020-07001-0

Keywords

Navigation