Development of Dyslipidemia in Atherosclerosis and Modern Approaches to its Treatment

Authors

  • Alexander Blagov Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
  • Igor Sobenin Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Scientific Center of Surgery, 119991 Moscow, Russia; Laboratoty of Medical Genetics, Chazov National Medical Research Center of Cardiology, 121552 Moscow, Russia
  • Vasily Sukhorukov Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Scientific Center of Surgery, 119991 Moscow, Russia; Laboratoty of Medical Genetics, Chazov National Medical Research Center of Cardiology, 121552 Moscow, Russia
  • Victor Glanz Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Scientific Center of Surgery, 119991 Moscow, Russia
  • Elizaveta Pleshko Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
  • Alexander Orekhov Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia; Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Petrovsky Scientific Center of Surgery, 119991 Moscow, Russia

DOI:

https://doi.org/10.59958/hsf.7341

Keywords:

atherosclerosis, dyslipidemia, lipoproteins

Abstract

Dyslipidemia is one of the key features in the pathogenesis of atherosclerosis, however, many points in the progression of this pathological phenomenon remain unclear. In this review, we will consider the significance of various lipoproteins in the progression of atherosclerosis, the general pattern of dyslipidemia development and its role in the pathogenesis of atherosclerosis, as well as new approaches to the treatment of dyslipidemia, which will potentially reduce the disadvantages of existing therapies and increase drug efficacy.

References

Mohd Nor NS, Al-Khateeb AM, Chua YA, Mohd Kasim NA, Mohd Nawawi H. Heterozygous familial hypercholesterolaemia in a pair of identical twins: a case report and updated review. BMC Pediatrics. 2019; 19: 106.

Dichgans M, Pulit SL, Rosand J. Stroke genetics: discovery, biology, and clinical applications. The Lancet. Neurology. 2019; 18: 587–599.

Rafieian-Kopaei M, Setorki M, Doudi M, Baradaran A, Nasri H. Atherosclerosis: process, indicators, risk factors and new hopes. International Journal of Preventive Medicine. 2014; 5: 927–946.

Ala-Korpela M. The culprit is the carrier, not the loads: cholesterol, triglycerides and apolipoprotein B in atherosclerosis and coronary heart disease. International Journal of Epidemiology. 2019; 48: 1389–1392.

Jebari-Benslaiman S, Galicia-García U, Larrea-Sebal A, Olaetxea JR, Alloza I, Vandenbroeck K, et al. Pathophysiology of Atherosclerosis. International Journal of Molecular Sciences. 2022; 23: 3346.

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.

Bergheanu SC, Bodde MC, Jukema JW. Pathophysiology and treatment of atherosclerosis: Current view and future perspective on lipoprotein modification treatment. Netherlands Heart Journal: Monthly Journal of the Netherlands Society of Cardiology and the Netherlands Heart Foundation. 2017; 25: 231–242.

Tomkin GH, Owens D. The chylomicron: relationship to atherosclerosis. International Journal of Vascular Medicine. 2012; 2012: 784536.

Helkin A, Stein JJ, Lin S, Siddiqui S, Maier KG, Gahtan V. Dyslipidemia Part 1–Review of Lipid Metabolism and Vascular Cell Physiology. Vascular and Endovascular Surgery. 2016; 50: 107–118.

Kei AA, Filippatos TD, Tsimihodimos V, Elisaf MS. A review of the role of apolipoprotein C-II in lipoprotein metabolism and cardiovascular disease. Metabolism: Clinical and Experimental. 2012; 61: 906–921.

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. European Heart Journal. 2017; 38: 2459–2472.

Bandeali S, Farmer J. High-density lipoprotein and atherosclerosis: the role of antioxidant activity. Current Atherosclerosis Reports. 2012; 14: 101–107.

Rye KA, Barter PJ. Cardioprotective functions of HDLs. Journal of Lipid Research. 2014; 55: 168–179.

Rosenson RS, Brewer HB, Jr, Ansell BJ, Barter P, Chapman MJ, Heinecke JW, et al. Dysfunctional HDL and atherosclerotic cardiovascular disease. Nature Reviews. Cardiology. 2016; 13: 48–60.

Díaz-Aragón A, Ruiz-Gastélum E, Álvarez-López H. Knowing the basic mechanisms of lipid metabolism. Cardiovascular and Metabolic Science. 2021; 32: s147–s152.

Gyamfi D, Awuah EO, Owusu S. Lipid metabolism: an overview. The Molecular Nutrition of Fats. 2019; 17–32.

Alves-Bezerra M, Cohen DE. Triglyceride Metabolism in the Liver. Comprehensive Physiology. 2017; 8: 1–8.

Grundy SM, Stone NJ, Bailey AL, Beam C, Birtcher KK, Blumenthal RS, et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/ APhA /ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: Executive Summary. Journal of the American College of Cardiology. 2018; 73: 3168–3209.

Jialal I, Devaraj S. AHA/ACC/Multisociety Cholesterol Guidelines: highlights. Therapeutic Advances in Cardiovascular Disease. 2019; 13: 1753944719881579.

Kobayashi J, Miyashita K, Nakajima K, Mabuchi H. Hepatic Lipase: a Comprehensive View of its Role on Plasma Lipid and Lipoprotein Metabolism. Journal of Atherosclerosis and Thrombosis. 2015; 22: 1001–1011.

Goldstein JL, Brown MS. A century of cholesterol and coronaries: from plaques to genes to statins. Cell. 2015; 161: 161–172.

Wu MY, Li CJ, Hou MF, Chu PY. New Insights into the Role of Inflammation in the Pathogenesis of Atherosclerosis. International Journal of Molecular Sciences. 2017; 18: 2034.

Branchetti E, Bavaria JE, Grau JB, Shaw RE, Poggio P, Lai EK, et al. Circulating soluble receptor for advanced glycation end product identifies patients with bicuspid aortic valve and associated aortopathies. Arteriosclerosis, Thrombosis, and Vascular Biology. 2014; 34: 2349–2357.

Gimbrone MA, Jr, García-Cardeña G. Endothelial Cell Dysfunction and the Pathobiology of Atherosclerosis. Circulation Research. 2016; 118: 620–636.

Poggio P, Branchetti E, Grau JB, Lai EK, Gorman RC, Gorman JH, 3rd, et al. Osteopontin-CD44v6 interaction mediates calcium deposition via phospho-Akt in valve interstitial cells from patients with noncalcified aortic valve sclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology. 2014; 34: 2086–2094.

Förstermann U, Xia N, Li H. Roles of Vascular Oxidative Stress and Nitric Oxide in the Pathogenesis of Atherosclerosis. Circulation Research. 2017; 120: 713–735.

Pernow J, Shemyakin A, Böhm F. New perspectives on endothelin-1 in atherosclerosis and diabetes mellitus. Life Sciences. 2012; 91: 507–516.

Zanobini M, Saccocci M, Tamborini G, Veglia F, Di Minno A, Poggio P, et al. Postoperative Echocardiographic Reduction of Right Ventricular Function: Is Pericardial Opening Modality the Main Culprit? BioMed Research International. 2017; 2017: 4808757.

Tabas I, García-Cardeña G, Owens GK. Recent insights into the cellular biology of atherosclerosis. The Journal of Cell Biology. 2015; 209: 13–22.

Mitra R, O'Neil GL, Harding IC, Cheng MJ, Mensah SA, Ebong EE. Glycocalyx in Atherosclerosis-Relevant Endothelium Function and as a Therapeutic Target. Current Atherosclerosis Reports. 2017; 19: 63.

Wengrofsky P, Lee J, Makaryus AN. Dyslipidemia and Its Role in the Pathogenesis of Atherosclerotic Cardiovascular Disease: Implications for Evaluation and Targets for Treatment of Dyslipidemia Based on Recent Guidelines. In McFarlane SI (ed.) Dyslipidemia. IntechOpen: London, United Kingdom. 2019.

Sainger R, Grau JB, Poggio P, Branchetti E, Bavaria JE, Gorman JH, 3rd, et al. Dephosphorylation of circulating human osteopontin correlates with severe valvular calcification in patients with calcific aortic valve disease. Biomarkers: Biochemical Indicators of Exposure, Response, and Susceptibility to Chemicals. 2012; 17: 111–118.

Ramji DP, Davies TS. Cytokines in atherosclerosis: Key players in all stages of disease and promising therapeutic targets. Cytokine & Growth Factor Reviews. 2015; 26: 673–685.

Poggio P, Songia P, Cavallotti L, Barbieri SS, Zanotti I, Arsenault BJ, et al. PCSK9 Involvement in Aortic Valve Calcification. Journal of the American College of Cardiology. 2018; 72: 3225–3227.

Gisterå A, Hansson GK. The immunology of atherosclerosis. Nature Reviews. Nephrology. 2017; 13: 368–380.

Bleda S, de Haro J, Varela C, Ferruelo A, Acin F. Elevated levels of triglycerides and vldl-cholesterol provoke activation of nlrp1 inflammasome in endothelial cells. International Journal of Cardiology. 2016; 220: 52–55.

Riches K, Porter KE. Lipoprotein(a): Cellular Effects and Molecular Mechanisms. Cholesterol. 2012; 2012: 923289.

Lara-Guzmán OJ, Gil-Izquierdo Á, Medina S, Osorio E, Álvarez-Quintero R, Zuluaga N, et al. Oxidized LDL triggers changes in oxidative stress and inflammatory biomarkers in human macrophages. Redox Biology. 2018; 15: 1–11.

Zernecke A. Dendritic cells in atherosclerosis: evidence in mice and humans. Arteriosclerosis, Thrombosis, and Vascular Biology. 2015; 35: 763–770.

Tabas I, Lichtman AH. Monocyte-Macrophages and T Cells in Atherosclerosis. Immunity. 2017; 47: 621–634.

Subramanian M, Tabas I. Dendritic cells in atherosclerosis. Seminars in Immunopathology. 2014; 36: 93–102.

Wang J, Uryga AK, Reinhold J, Figg N, Baker L, Finigan A, et al. Vascular Smooth Muscle Cell Senescence Promotes Atherosclerosis and Features of Plaque Vulnerability. Circulation. 2015; 132: 1909–1919.

Ponticos M, Smith BD. Extracellular matrix synthesis in vascular disease: hypertension, and atherosclerosis. Journal of Biomedical Research. 2014; 28: 25–39.

Niedzielski M, Broncel M, Gorzelak-Pabiś P, Woźniak E. New possible pharmacological targets for statins and ezetimibe. Biomedicine & Pharmacotherapy = Biomedecine & Pharmacotherapie. 2020; 129: 110388.

Pinal-Fernandez I, Casal-Dominguez M, Mammen AL. Statins: pros and cons. Medicina Clinica. 2018; 150: 398–402.

Dayar E, Pechanova O. Targeted Strategy in Lipid-Lowering Therapy. Biomedicines. 2022; 10: 1090.

Zozina VI, Covantev S, Goroshko OA, Krasnykh LM, Kukes VG. Coenzyme Q10 in Cardiovascular and Metabolic Diseases: Current State of the Problem. Current Cardiology Reviews. 2018; 14: 164–174.

Meena AK, Ratnam DV, Chandraiah G, Ankola DD, Rao PR, Kumar MNVR. Oral nanoparticulate atorvastatin calcium is more efficient and safe in comparison to Lipicure in treating hyperlipidemia. Lipids. 2008; 43: 231–241.

Shaker MA, Elbadawy HM, Al Thagfan SS, Shaker MA. Enhancement of atorvastatin oral bioavailability via encapsulation in polymeric nanoparticles. International Journal of Pharmaceutics. 2021; 592: 120077.

Chen L, Nakano K, Kimura S, Matoba T, Iwata E, Miyagawa M, et al. Nanoparticle-mediated delivery of pitavastatin into lungs ameliorates the development and induces regression of monocrotaline-induced pulmonary artery hypertension. Hypertension (Dallas, Tex.: 1979). 2011; 57: 343–350.

Abd-Rabo MM, Wahman LF, El Hosary R, Ahmed IS. High-fat diet induced alteration in lipid enzymes and inflammation in cardiac and brain tissues: Assessment of the effects of Atorvastatin-loaded nanoparticles. Journal of Biochemical and Molecular Toxicology. 2020; 34: e22465.

Liu P, Liu Y, Li P, Zhou Y, Song Y, Shi Y, et al. Rosuvastatin- and Heparin-Loaded Poly(l-lactide- co-caprolactone) Nanofiber Aneurysm Stent Promotes Endothelialization via Vascular Endothelial Growth Factor Type A Modulation. ACS Applied Materials & Interfaces. 2018; 10: 41012–41018.

Esposto BS, Jauregi P, Tapia-Blácido DR, Martelli-Tosi M. Liposomes vs. chitosomes: Encapsulating food bioactives. Trends in Food Science & Technology. 2021; 108: 40–48.

Sharma K, Kumar K, Mishra N. Nanoparticulate carrier system: a novel treatment approach for hyperlipidemia. Drug Delivery. 2016; 23: 694–709.

Kurakula M, El-Helw AM, Sobahi TR, Abdelaal MY. Chitosan based atorvastatin nanocrystals: effect of cationic charge on particle size, formulation stability, and in-vivo efficacy. International Journal of Nanomedicine. 2015; 10: 321–334.

Yu H, Jin F, Liu D, Shu G, Wang X, Qi J, et al. ROS-responsive nano-drug delivery system combining mitochondria-targeting ceria nanoparticles with atorvastatin for acute kidney injury. Theranostics. 2020; 10: 2342–2357.

Tiwari R, Pathak K. Nanostructured lipid carrier versus solid lipid nanoparticles of simvastatin: comparative analysis of characteristics, pharmacokinetics and tissue uptake. International Journal of Pharmaceutics. 2011; 415: 232–243.

Beniwal A, Choudhary HC. Rosuvastatin calcium-loaded Solid Lipid Nanoparticles (SLN) using design of experiment approach for oral delivery. International Journal of Chemical and Lifesciences. 2017; 6: 2029–2038.

Duivenvoorden R, Tang J, Cormode DP, Mieszawska AJ, Izquierdo-Garcia D, Ozcan C, et al. A statin-loaded reconstituted high-density lipoprotein nanoparticle inhibits atherosclerotic plaque inflammation. Nature Communications. 2014; 5: 3065.

Dawoud MH, Fayez AM, Mohamed RA, Sweed NM. Enhancement of the Solubility of Rosuvastatin Calcium by Nanovesicular Formulation: A Systematic Study Based on a Quality by Design Approach. Proceedings. 2020; 78: 34.

Horton JD, Cohen JC, Hobbs HH. PCSK9: a convertase that coordinates LDL catabolism. Journal of Lipid Research. 2009; 50: S172–S177.

Pokrywka GS. PCSK9 inhibitors: a non-statin cholesterol-lowering treatment option. Postgraduate Medicine. 2018; 130: 287–298.

Fitzgerald G, Kiernan T. PCSK9 inhibitors and LDL reduction: pharmacology, clinical implications, and future perspectives. Expert Review of Cardiovascular Therapy. 2018; 16: 567–578.

Warden BA, Duell PB. Inclisiran: A Novel Agent for Lowering Apolipoprotein B-containing Lipoproteins. Journal of Cardiovascular Pharmacology. 2021; 78: e157–e174.

Leiter LA, Teoh H, Kallend D, Wright RS, Landmesser U, Wijngaard PLJ, et al. Inclisiran Lowers LDL-C and PCSK9 Irrespective of Diabetes Status: The ORION-1 Randomized Clinical Trial. Diabetes Care. 2019; 42: 173–176.

Ray KK, Stoekenbroek RM, Kallend D, Nishikido T, Leiter LA, Landmesser U, et al. Effect of 1 or 2 Doses of Inclisiran on Low-Density Lipoprotein Cholesterol Levels: One-Year Follow-up of the ORION-1 Randomized Clinical Trial. JAMA Cardiology. 2019; 4: 1067–1075.

Chackerian B, Remaley A. Vaccine strategies for lowering LDL by immunization against proprotein convertase subtilisin/kexin type 9. Current Opinion in Lipidology. 2016; 27: 345–350.

Kawakami R, Nozato Y, Nakagami H, Ikeda Y, Shimamura M, Yoshida S, et al. Development of vaccine for dyslipidemia targeted to a proprotein convertase subtilisin/kexin type 9 (PCSK9) epitope in mice. PloS One. 2018; 13: e0191895.

Wu D, Zhou Y, Pan Y, Li C, Wang Y, Chen F, et al. Vaccine Against PCSK9 Improved Renal Fibrosis by Regulating Fatty Acid β-Oxidation. Journal of the American Heart Association. 2020; 9: e014358.

Wang X, Chen X, Zhang X, Su C, Yang M, He W, et al. A small-molecule inhibitor of PCSK9 transcription ameliorates atherosclerosis through the modulation of FoxO1/3 and HNF1α. EBioMedicine. 2020; 52: 102650.

Salaheldin TA, Godugu K, Bharali DJ, Fujioka K, Elshourbagy N, Mousa SA. Novel oral nano-hepatic targeted anti-PCSK9 in hypercholesterolemia. Nanomedicine: Nanotechnology, Biology, and Medicine. 2022; 40: 102480.

Kersten S. ANGPTL3 as therapeutic target. Current Opinion in Lipidology. 2021; 32: 335–341.

Raal FJ, Rosenson RS, Reeskamp LF, Hovingh GK, Kastelein JJP, Rubba P, et al. Evinacumab for Homozygous Familial Hypercholesterolemia. The New England Journal of Medicine. 2020; 383: 711–720.

Qiu M, Glass Z, Chen J, Haas M, Jin X, Zhao X, et al. Lipid nanoparticle-mediated codelivery of Cas9 mRNA and single-guide RNA achieves liver-specific in vivo genome editing of Angptl3. Proceedings of the National Academy of Sciences of the United States of America. 2021; 118: e2020401118.

Fowler A, Sampson M, Remaley AT, Chackerian B. A VLP-based vaccine targeting ANGPTL3 lowers plasma triglycerides in mice. Vaccine. 2021; 39: 5780–5786.

Kastelein JJP, Hsieh A, Dicklin MR, Ditmarsch M, Davidson MH. Obicetrapib: Reversing the Tide of CETP Inhibitor Disappointments. Current Atherosclerosis Reports. 2024; 26: 35–44.

Ballantyne CM, Ditmarsch M, Kastelein JJ, Nelson AJ, Kling D, Hsieh A, et al. Obicetrapib plus ezetimibe as an adjunct to high-intensity statin therapy: A randomized phase 2 trial. Journal of Clinical Lipidology. 2023; 17: 491–503.

Nicholls SJ, Ditmarsch M, Kastelein JJ, Rigby SP, Kling D, Curcio DL, et al. Lipid lowering effects of the CETP inhibitor obicetrapib in combination with high-intensity statins: a randomized phase 2 trial. Nature Medicine. 2022; 28: 1672–1678.

Tummala R, Gupta M, Devanabanda AR, Bandyopadhyay D, Aronow WS, Ray KK, et al. Bempedoic acid and its role in contemporary management of hyperlipidemia in atherosclerosis. Annals of Medicine. 2022; 54: 1287–1296.

Published

2024-06-23

How to Cite

Blagov, A., Sobenin, I. ., Sukhorukov, V. ., Glanz, V. ., Pleshko, E. ., & Orekhov, A. . (2024). Development of Dyslipidemia in Atherosclerosis and Modern Approaches to its Treatment. The Heart Surgery Forum, 27(6), E680-E688. https://doi.org/10.59958/hsf.7341

Issue

Section

Review