Susceptibility of low-density lipoprotein particles to aggregate depends on particle lipidome, is modifiable, and associates with future cardiovascular deaths

Maija Ruuth, Su Duy Nguyen, Terhi Vihervaara, Mika Hilvo, Teemu D Laajala, Pradeep Kumar Kondadi, Anton Gisterå, Hanna Lähteenmäki, Tiia Kittilä, Jenni Huusko, Matti Uusitupa, Ursula Schwab, Markku J Savolainen, Juha Sinisalo, Marja-Liisa Lokki, Markku S Nieminen, Antti Jula, Markus Perola, Seppo Ylä-Herttula, Lawrence Rudel, Anssi Öörni, Marc Baumann, Amos Baruch, Reijo Laaksonen, Daniel F J Ketelhuth, Tero Aittokallio, Matti Jauhiainen, Reijo Käkelä, Jan Borén, Kevin Jon Williams, Petri T Kovanen, Katariina Öörni, Maija Ruuth, Su Duy Nguyen, Terhi Vihervaara, Mika Hilvo, Teemu D Laajala, Pradeep Kumar Kondadi, Anton Gisterå, Hanna Lähteenmäki, Tiia Kittilä, Jenni Huusko, Matti Uusitupa, Ursula Schwab, Markku J Savolainen, Juha Sinisalo, Marja-Liisa Lokki, Markku S Nieminen, Antti Jula, Markus Perola, Seppo Ylä-Herttula, Lawrence Rudel, Anssi Öörni, Marc Baumann, Amos Baruch, Reijo Laaksonen, Daniel F J Ketelhuth, Tero Aittokallio, Matti Jauhiainen, Reijo Käkelä, Jan Borén, Kevin Jon Williams, Petri T Kovanen, Katariina Öörni

Abstract

Aims: Low-density lipoprotein (LDL) particles cause atherosclerotic cardiovascular disease (ASCVD) through their retention, modification, and accumulation within the arterial intima. High plasma concentrations of LDL drive this disease, but LDL quality may also contribute. Here, we focused on the intrinsic propensity of LDL to aggregate upon modification. We examined whether inter-individual differences in this quality are linked with LDL lipid composition and coronary artery disease (CAD) death, and basic mechanisms for plaque growth and destabilization.

Methods and results: We developed a novel, reproducible method to assess the susceptibility of LDL particles to aggregate during lipolysis induced ex vivo by human recombinant secretory sphingomyelinase. Among patients with an established CAD, we found that the presence of aggregation-prone LDL was predictive of future cardiovascular deaths, independently of conventional risk factors. Aggregation-prone LDL contained more sphingolipids and less phosphatidylcholines than did aggregation-resistant LDL. Three interventions in animal models to rationally alter LDL composition lowered its susceptibility to aggregate and slowed atherosclerosis. Similar compositional changes induced in humans by PCSK9 inhibition or healthy diet also lowered LDL aggregation susceptibility. Aggregated LDL in vitro activated macrophages and T cells, two key cell types involved in plaque progression and rupture.

Conclusion: Our results identify the susceptibility of LDL to aggregate as a novel measurable and modifiable factor in the progression of human ASCVD.

Figures

Figure 1
Figure 1
Measurement of the susceptibility of low-density lipoprotein (LDL) from healthy human subjects and from coronary artery disease patients to aggregate ex vivo. (A) LDL is isolated from blood plasma by ultracentrifugation and aggregation was induced by incubation with human recombinant secretory sphingomyelinase (hrSMase) at pH 5.5. The size of LDL particles was measured before hrSMase treatment (time = 0 h), and formation of LDL aggregates was followed in real time by measuring their size with dynamic light scattering. (B) LDL particles were isolated from 100 plasma samples collected from the Finnish Health 2000 Health Examination Survey and the aggregation susceptibility of the particles was analysed. Based on LDL aggregate size at 2 h, the particles were divided into quartiles. (C) Size distributions of LDL aggregates at the 2 h time point. The box encompasses the middle 50% of the measured values; the horizontal line within each box shows the median of the measured values; the whiskers encompass the most extreme data point that is still no further from the margins of the box than 1.5 times the interquartile range. (D) Patients (n = 48) from the Corogene study, having >50% stenosis in their coronary arteries were divided into two groups: (i) CAD death group, in which patients died of coronary events during an average 2.5-year follow-up period and (ii) stable CAD group, having no cardiovascular events during the follow-up period. The patients were matched for the conventional cardiovascular risk factors. LDL was isolated and LDL aggregation was induced by treatment with hrSMase. The box plot diagram shows the distribution of aggregate sizes after incubation for 2 h in the two groups from Corogene study and in 100 subjects from the Health 2000 study (all quartiles from C combined). Statistical differences between the groups were determined using Kruskal–Wallis test followed by Dunn’s test. P < 0.001 by Kruskal–Wallis test; *P < 0.05, ***P <0.001 by Dunn’s test.
Figure 2
Figure 2
The susceptibility of low-density lipoprotein (LDL) to aggregate strongly correlates with the surface lipid composition of the particles. (A) LDL was isolated from plasma and LDL lipidome was analysed using mass spectrometry. Volcano plots showing Spearman correlation coefficients of LDL aggregate size at 2 h vs. LDL surface lipids in (B) Health 2000 samples and (C) in Corogene samples. Red circles indicate positive correlations, and blue circles indicate negative correlations. The identities of only those lipids with significance correlation values (P < 0.05) are indicated. Cer, ceramide; LPC, lysophosphatidylcholine; PC, phosphatidylcholine; SM, sphingomyelin.
Figure 3
Figure 3
The susceptibility of low-density lipoprotein (LDL) to aggregate strongly correlates with the core lipid composition of the particles. Volcano plots showing Spearman correlation coefficients of LDL aggregate size at 2 h vs. LDL core lipids (A) in Health 2000 samples and (B) in Corogene samples. Red circles indicate positive correlations, and blue circles indicate negative correlations. The identities of only those lipids with significance correlation values (P < 0.05) are indicated. CE, cholesteryl ester; TAG, triacylglycerol.
Figure 4
Figure 4
A dietary intervention and PCSK9 inhibition in human subjects improves their low-density lipoprotein (LDL) composition and renders their particles less susceptible to aggregate. Plasma samples were obtained from the SYSDIET-study, where participants were placed on either an isocaloric healthy Nordic diet (n = 33) or a control diet (n = 25) for 18 or 24 weeks and from the EQUATOR study, a randomized placebo-controlled phase II trial of a monoclonal antibody inhibiting the function of PCSK9, RG7652, (n = 25), or placebo (n = 15) for 29 days. LDL was isolated, and aggregation analysed from samples before and after the diet/treatment period. (A and B) LDL aggregate sizes at the 2-h time point are shown in the diet group and control group before and after the diet period. Each line represents one subject and blue lines show a decrease and red lines an increase in aggregate size. (D and E) LDL aggregate sizes at the 2-h time point are shown in the PSCK9 inhibitor group and placebo group before and after the treatment period. Each line represents one subject and blue lines show a decrease and red lines an increase in aggregate size. (C and F) Volcano plot showing the Spearman correlation coefficients of LDL aggregate size at 2 h vs. LDL surface lipids in the SYSDIET study and in the EQUATOR study. PC, phosphatidylcholine; PE, phosphatidylethanolamine; SM, sphingomyelin.
Figure 5
Figure 5
Pharmacological and genetic interventions that favourably alter low-density lipoprotein (LDL) lipid composition in vivo in hypercholesterolaemic mice render their LDL aggregation-resistant. (A) Human APOB transgenic/LDLr−/− mice were given a single intravenous injection of large ‘empty’ vesicles (LEVs) made from PC 16:0/18:1 or equivalent volume of PBS (control, n= 8 per group). After 1 h, plasma was collected, LDL isolated, and aggregation induced by treatment with hrSMase. LDL aggregation was followed by dynamic light scattering at the indicated time points. The insert shows LDL aggregation for up to 24 h. (B) LDLr−/−/Apob100/100 mice were simultaneously started on an atherogenic western-type diet and intraperitoneal injections three times per week of either myriocin, and inhibitor of SM biosynthesis, or PBS (n = 11 mice per group). Diet and injections continued for 10 weeks. LDL was isolated, then treated with hrSMase, and particle aggregation was followed by dynamic light scattering at the indicated time points. (C) Representative aortas from a control mouse and a myriocin-treated mouse stained with Sudan IV. The extent of atherosclerosis was determined by measuring the amounts of Sudan IV positive areas in the aortas. (D) LDL was isolated from the plasma of human APOB transgenic/LDLr−/−/Soat2−/− mice and human APOB transgenic/LDLr−/−/Soat2+/+ littermates. LDL was treated with hrSMase and LDL aggregation followed by dynamic light scattering at the indicated time points. The line and column graphs display averages ± standard deviations. *P <0.05, **P < 0.01, and ***P < 0.001 by Student’s t-test.
Figure 6
Figure 6
The effect of aggregated low-density lipoprotein (LDL) on macrophages and T-cells. (A) Oil Red O-stained human monocyte-derived macrophages incubated for 20 h with 100 µg/mL of LDL or aggregated LDL (24-h treatment with hrSMase). (B) The amounts of cholesteryl esters in monocyte-derived macrophages incubated in the presence of 100 µg/mL of the variously treated LDL preparations for 20 h. (C) The amount of MMPs secreted from the cells was determined by a Multiplex array. Only secretion of MMP-7 was induced by SMase-treated LDL. (D) Activation of human apoB-100-specific T-cell hybridoma 48-5 measured by IL-2 secretion after 24 h co-culture with antigen presenting cells and 10 µg/mL LDL with increasing amount of sphingomyelinase modification (left panel) or oxidation (right panel). The column graphs show averages ± standard deviations. Statistical differences between the groups were determined using Kruskal–Wallis test followed by post hoc pairwise comparison using Dunn’s test. *P < 0.05 and **P < 0.01.
Take home figure
Take home figure
Development of a measurement of LDL aggregation susceptibility revealed the importance of qualitative differences in LDL particles in ASCVD. The circulating LDL particles of patients succumbing in CAD have a high proportion of sphingomyelins (SM) and ceramides. When such aggregation-prone LDL particles enter the arterial intima, they easily aggregate upon modification. Intimal LDL aggregates promote atherogenesis, inflammation and plaque rupture by inducing foam cell formation, secretion of matrix metalloproteinase 7 (MMP7) and by activating T-cells, ultimately increasing the risk for fatal CAD. Therefore, decreasing the aggregation susceptibility of circulating LDL could reduce CAD risk. In vitro and in mouse models, aggregation-prone LDL particles can be rendered aggregation-resistant by reducing their sphingolipid content. In humans, a healthy diet or treatment with a PCSK9 inhibitor improves the quality of LDL particles resulting in aggregation-resistant phosphatidylcholine (PC)- and lysophosphatidylcholine (LPC)-rich LDL particles.
https://www.ncbi.nlm.nih.gov/pmc/articles/instance/6047440/bin/ehy319f7a.jpg

References

    1. Ference BA, Ginsberg HN, Graham I, Ray KK, Packard CJ, Bruckert E, Hegele RA, Krauss RM, Raal FJ, Schunkert H, Watts GF, Borén J, Fazio S, Horton JD, Masana L, Nicholls SJ, Nordestgaard BG, van de Sluis B, Taskinen M-R, Tokgözoğlu L, Landmesser U, Laufs U, Wiklund O, Stock JK, Chapman MJ, Catapano 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;38:2459–2472.
    1. Piepoli MF, Hoes AW, Agewall S, Albus C, Brotons C, Catapano AL, Cooney MT, Corra U, Cosyns B, Deaton C, Graham I, Hall MS, Hobbs FDR, Lochen ML, Lollgen H, Marques-Vidal P, Perk J, Prescott E, Redon J, Richter DJ, Sattar N, Smulders Y, Tiberi M, van der Worp HB, van Dis I, Verschuren WMM, Binno S. 2016 European Guidelines on cardiovascular disease prevention in clinical practice: The Sixth Joint Task Force of the European Society of Cardiology and Other Societies on Cardiovascular Disease Prevention in Clinical Practice (constituted by representatives of 10 societies and by invited experts)Developed with the special contribution of the European Association for Cardiovascular Prevention & Rehabilitation (EACPR). Eur Heart J 2016;37:2315–2381.
    1. Sabatine MS, Giugliano RP, Keech AC, Honarpour N, Wiviott SD, Murphy SA, Kuder JF, Wang H, Liu T, Wasserman SM, Sever PS, Pedersen TR.. Evolocumab and clinical outcomes in patients with cardiovascular disease. N Engl J Med 2017;376:1713–1722.
    1. Kaasenbrood L, Boekholdt SM, van der Graaf Y, Ray KK, Peters RJ, Kastelein JJ, Amarenco P, LaRosa JC, Cramer MJ, Westerink J, Kappelle LJ, de Borst GJ, Visseren FL.. Distribution of estimated 10-year risk of recurrent vascular events and residual risk in a secondary prevention population. Circulation 2016;134:1419–1429.
    1. Catapano AL, Graham I, De Backer G, Wiklund O, Chapman MJ, Drexel H, Hoes AW, Jennings CS, Landmesser U, Pedersen TR, Reiner Z, Riccardi G, Taskinen MR, Tokgozoglu L, Verschuren WMM, Vlachopoulos C, Wood DA, Zamorano JL, Cooney MT. 2016 ESC/EAS Guidelines for the Management of Dyslipidaemias. Eur Heart J 2016;37:2999–3058.
    1. Williams KJ, Tabas I.. Lipoprotein retention—and clues for atheroma regression. Arterioscler Thromb Vasc Biol 2005;25:1536–1540.
    1. Borén J, Williams KJ.. The central role of arterial retention of cholesterol-rich apolipoprotein-B-containing lipoproteins in the pathogenesis of atherosclerosis: a triumph of simplicity. Curr Opin Lipidol 2016;27:473–483.
    1. Öörni K, Pentikäinen MO, Ala-Korpela M, Kovanen PT. Aggregation, fusion, and vesicle formation of modified low density lipoprotein particles: molecular mechanisms and effects on matrix interactions. J Lipid Res 2000;41:1703–1714.
    1. Hoff HF, Morton RE.. Lipoproteins containing apo B extracted from human aortas. Structure and function. Ann N Y Acad Sci 1985;454:183–194.
    1. Aviram M, Maor I, Keidar S, Hayek T, Oiknine J, Barel Y, Adler Z, Kertzman V, Milo S.. Lesioned low density lipoprotein in atherosclerotic apolipoprotein E-deficient transgenic mice and in humans is oxidized and aggregated. Biochem Biophys Res Commun 1995;216:501–513.
    1. Schissel SL, Tweedie-Hardman J, Rapp JH, Graham G, Williams KJ, Tabas I.. Rabbit aorta and human atherosclerotic lesions hydrolyze the sphingomyelin of retained low-density lipoprotein. Proposed role for arterial-wall sphingomyelinase in subendothelial retention and aggregation of atherogenic lipoproteins. J Clin Invest 1996;98:1455–1464.
    1. Lehti S, Nguyen SD, Belevich I, Vihinen H, Heikkilä HM, Soliymani R, Käkelä R, Saksi J, Jauhiainen M, Grabowski GA, Kummu O, Hörkkö S, Baumann M, Lindsberg PJ, Jokitalo E, Kovanen PT, Öörni K.. Extracellular lipid accumulates in human carotid arteries as distinct three-dimensional structures with proinflammatory properties. Am J Pathol 2018;188:525–538.
    1. Plihtari R, Hurt-Camejo E, Oörni K, Kovanen PT.. Proteolysis sensitizes LDL particles to phospholipolysis by secretory phospholipase A2 group V and secretory sphingomyelinase. J Lipid Res 2010;51:1801–1809.
    1. Öörni K, Kovanen PT.. PLA2-V: a real player in atherogenesis. Arterioscler Thromb Vasc Biol 2007;27:445–447.
    1. Schissel SL, Jiang X-C, Tweedie-Hardman J, Jeong T-S, Camejo EH, Najib J, Rapp JH, Williams KJ, Tabas I.. Secretory sphingomyelinase, a product of the acid sphingomyelinase gene, can hydrolyze atherogenic lipoproteins at neutral pH. Implications for atherosclerotic lesion development. J Biol Chem 1998;273:2738–2746.
    1. Tabas I, Li Y, Brocia RW, Xu SW, Swenson TL, Williams KJ.. Lipoprotein lipase and sphingomyelinase synergistically enhance the association of atherogenic lipoproteins with smooth muscle cells and extracellular matrix. A possible mechanism for low density lipoprotein and lipoprotein(a) retention and macrophage foam cell formation. J Biol Chem 1993;268:20419–20432.
    1. Grosheva I, Haka AS, Qin C, Pierini LM, Maxfield FR.. Aggregated LDL in contact with macrophages induces local increases in free cholesterol levels that regulate local actin polymerization. Arterioscler Thromb Vasc Biol 2009;29:1615–1621.
    1. Ketelhuth DF, Hansson GK.. Adaptive response of T and B cells in atherosclerosis. Circ Res 2016;118:668–678.
    1. Libby P, Hansson GK.. Inflammation and immunity in diseases of the arterial tree: players and layers. Circ Res 2015;116:307–311.
    1. Aromaa A, Koskinen S.. Health and Functional Capacity in Finland: Baseline Results of the Health 2000 Health Examination Survey, Vol. B12/200. Publications of the National Public Health Institute; Helsinki, Finland: 2004.
    1. Vaara S, Nieminen MS, Lokki ML, Perola M, Pussinen PJ, Allonen J, Parkkonen O., Sinisalo J.. Cohort Profile: the Corogene study. Int J Epidemiol 2012;41:1265–1271.
    1. Magnusdottir OK, Landberg R, Gunnarsdottir I, Cloetens L, Åkesson B, Önning G, Jonsdottir SE, Rosqvist F, Schwab U, Herzig K-H, Savolainen MJ, Brader L, Hermansen K, Kolehmainen M, Poutanen K, Uusitupa M, Thorsdottir I, Risérus U.. Plasma alkylresorcinols reflect important whole-grain components of a healthy Nordic diet. J Nutr 2013;143:1383–1390.
    1. Uusitupa M, Hermansen K, Savolainen MJ, Schwab U, Kolehmainen M, Brader L, Mortensen LS, Cloetens L, Johansson-Persson A, Onning G, Landin-Olsson M, Herzig K-H, Hukkanen J, Rosqvist F, Iggman D, Paananen J, Pulkki KJ, Siloaho M, Dragsted L, Barri T, Overvad K, Bach Knudsen KE, Hedemann MS, Arner P, Dahlman I, Borge GIA, Baardseth P, Ulven SM, Gunnarsdottir I, Jónsdóttir S, Thorsdottir I, Orešič M, Poutanen KS, Risérus U, Akesson B.. Effects of an isocaloric healthy Nordic diet on insulin sensitivity, lipid profile and inflammation markers in metabolic syndrome—a randomized study (SYSDIET). J Intern Med 2013;274:52–66.
    1. Baruch A, Mosesova S, Davis JD, Budha N, Vilimovskij A, Kahn R, Peng K, Cowan KJ, Harris LP, Gelzleichter T, Lehrer J, Davis JC, Tingley WG.. Effects of RG7652, a monoclonal antibody against PCSK9, on LDL-C, LDL-C subfractions, and inflammatory biomarkers in patients at high risk of or with established coronary heart disease (from the Phase 2 EQUATOR Study). Am J Cardiol 2017;119:1576–1583.
    1. Havel RJ. Biology of cholesterol, lipoproteins and atherosclerosis. Clin Exp Hypertens A 1989;11:887–900.
    1. Melchior JT, Sawyer JK, Kelley KL, Shah R, Wilson MD, Hantgan RR, Rudel LL.. LDL particle core enrichment in cholesteryl oleate increases proteoglycan binding and promotes atherosclerosis. J Lipid Res 2013;54:2495–2503.
    1. Sneck M, Nguyen SD, Pihlajamaa T, Yohannes G, Riekkola ML, Milne R, Kovanen PT, Oörni K.. Conformational changes of apoB-100 in SMase-modified LDL mediate formation of large aggregates at acidic pH. J Lipid Res 2012;53:1832–1839.
    1. Laaksonen R, Ekroos K, Sysi-Aho M, Hilvo M, Vihervaara T, Kauhanen D, Suoniemi M, Hurme R, März W, Scharnagl H, Stojakovic T, Vlachopoulou E, Lokki M-L, Nieminen MS, Klingenberg R, Matter CM, Hornemann T, Jüni P, Rodondi N, Räber L, Windecker S, Gencer B, Pedersen ER, Tell GS, Nygård O, Mach F, Sinisalo J, Lüscher TF.. Plasma ceramides predict cardiovascular death in patients with stable coronary artery disease and acute coronary syndromes beyond LDL-cholesterol. Eur Heart J 2016;37:1967–1976.
    1. Hilvo M, Simolin H, Metso J, Ruuth M, Öörni K, Jauhiainen M, Laaksonen R, Baruch A.. PCSK9 inhibition alters the lipidome of plasma and lipoprotein fractions. Atherosclerosis 2018;269:159–165.
    1. Rodrigueza WV, Mazany KD, Essenburg AD, Pape ME, Rea TJ, Bisgaier CL, Williams KJ.. Large versus small unilamellar vesicles mediate reverse cholesterol transport in vivo into two distinct hepatic metabolic pools. Implications for the treatment of atherosclerosis. Arterioscler Thromb Vasc Biol 1997;17:2132–2139.
    1. Hojjati MR, Li Z, Zhou H, Tang S, Huan C, Ooi E, Lu S, Jiang X-C.. Effect of myriocin on plasma sphingolipid metabolism and atherosclerosis in apoE-deficient mice. J Biol Chem 2005;280:10284–10289.
    1. Willner EL, Tow B, Buhman KK, Wilson M, Sanan DA, Rudel LL, Farese RV.. Deficiency of acyl CoA: cholesterol acyltransferase 2 prevents atherosclerosis in apolipoprotein E-deficient mice. Proc Natl Acad Sci USA 2003;100:1262–1267.
    1. Hermansson A, Ketelhuth DFJ, Strodthoff D, Wurm M, Hansson EM, Nicoletti A, Paulsson-Berne G, Hansson GK.. Inhibition of T cell response to native low-density lipoprotein reduces atherosclerosis. J Exp Med 2010;207:1081–1093.
    1. Khokha R, Murthy A, Weiss A.. Metalloproteinases and their natural inhibitors in inflammation and immunity. Nat Rev Immunol 2013;13:649–665.
    1. Tabas I, Williams KJ, Boren J.. Subendothelial lipoprotein retention as the initiating process in atherosclerosis: update and therapeutic implications. Circulation 2007;116:1832–1844.
    1. Devlin CM, Leventhal AR, Kuriakose G, Schuchman EH, Williams KJ, Tabas I.. Acid sphingomyelinase promotes lipoprotein retention within early atheromata and accelerates lesion progression. Arterioscler Thromb Vasc Biol 2008;28:1723–1730.
    1. Abbas A, Aukrust P, Russell D, Krohg-Sørensen K, Almås T, Bundgaard D, Bjerkeli V, Sagen EL, Michelsen AE, Dahl TB, Holm S, Ueland T, Skjelland M, Halvorsen B.. Matrix metalloproteinase 7 is associated with symptomatic lesions and adverse events in patients with carotid atherosclerosis. PLoS One 2014;9:e84935..
    1. Ketelhuth DFJ, Gisterå A, Johansson DK, Hansson GK.. T cell-based therapies for atherosclerosis. Curr Pharm Des 2013;19:5850–5858.
    1. Frahnow T, Osterhoff MA, Hornemann S, Kruse M, Surma MA, Klose C, Simons K, Pfeiffer AFH.. Heritability and responses to high fat diet of plasma lipidomics in a twin study. Sci Rep 2017;7:3750..
    1. Deevska GM, Sunkara M, Morris AJ, Nikolova-Karakashian MN.. Characterization of secretory sphingomyelinase activity, lipoprotein sphingolipid content and LDL aggregation in ldlr-/- mice fed on a high-fat diet. Biosci Rep 2012;32:479–490.
    1. Jeong TS, Schissel SL, Tabas I, Pownall HJ, Tall AR, Jiang X.. Increased sphingomyelin content of plasma lipoproteins in apolipoprotein E knockout mice reflects combined production and catabolic defects and enhances reactivity with mammalian sphingomyelinase. J Clin Invest 1998;101:905–912.
    1. Friedman M, Byers SO, Rosenman RH.. Resolution of aortic atherosclerotic infiltration in the rabbit by phosphatide infusion. Proc Soc Exp Biol Med 1957;95:586–588.
    1. Jiang XC, Paultre F, Pearson TA, Reed RG, Francis CK, Lin M, Berglund L, Tall AR.. Plasma sphingomyelin level as a risk factor for coronary artery disease. Arterioscler Thromb Vasc Biol 2000;20:2614–2618.
    1. Fernandez C, Sandin M, Sampaio JL, Almgren P, Narkiewicz K, Hoffmann M, Hedner T, Wahlstrand B, Simons K, Shevchenko A, James P, Melander O.. Plasma lipid composition and risk of developing cardiovascular disease. PLoS One 2013;8:e71846..

Source: PubMed

3
Subscribe