Abstract

HomeArteriosclerosis, Thrombosis, and Vascular BiologyVol. 21, No. 12ApoA-II Versus ApoA-I Free AccessEditorialPDF/EPUBAboutView PDFView EPUBSections ToolsAdd to favoritesDownload citationsTrack citationsPermissions ShareShare onFacebookTwitterLinked InMendeleyReddit Jump toFree AccessEditorialPDF/EPUBApoA-II Versus ApoA-ITwo for One Is Not Always a Good Deal Lawrence W. Castellani and Aldons J. Lusis Lawrence W. CastellaniLawrence W. Castellani Departments of Medicine (L.W.C., A.J.L.) and Microbiology (A.J.L.), Immunology and Molecular Genetics, and Molecular Biology Institute (A.J.L.), University of California, Los Angeles. Search for more papers by this author and Aldons J. LusisAldons J. Lusis Departments of Medicine (L.W.C., A.J.L.) and Microbiology (A.J.L.), Immunology and Molecular Genetics, and Molecular Biology Institute (A.J.L.), University of California, Los Angeles. Search for more papers by this author Originally published15 Mar 2018https://doi.org/10.1161/atvb.21.12.1870Arteriosclerosis, Thrombosis, and Vascular Biology. 2001;21:1870–1872Apolipoprotein A-I (apoA-I), the major protein of HDL, is probably among the most intensively studied of all proteins. It functions in HDL assembly, in the removal of excess cholesterol from cells, and in the transport of cholesterol from peripheral tissues to the liver, a process known as “reverse cholesterol transport.” It is a cofactor for lecithin:cholesterol acyl transferase (LCAT), the enzyme responsible for cholesterol esterification in HDL, and it stabilizes certain antioxidant enzymes, such as serum paraoxonase, that are carried on HDL.1,2 Rare null mutations of apoA-I in human populations are associated with early coronary heart disease,3 and overexpression of apoA-I in transgenic mice protects against atherogenesis.4 An exciting recent development was the 4-angstrom-resolution x-ray structure for the helical part of apoA-I, encompassing residues 44 to 243 (reviewed by Segrest et al5).See page 1977In contrast, apolipoprotein A-II (apoA-II), the second most abundant protein of HDL, has no known function, and it has been the subject of few detailed studies. Its presence in HDL has been reported to decrease,6 or have no influence on, cholesterol efflux from cells,7,8 nor is it a cofactor for any known enzyme. Rare individuals lacking apoA-II appear to have normal plasma lipids.9 In fact, all of the known effects of apoA-II are deleterious. In studies with mice and humans, apoA-II levels have been associated with increased susceptibility to atherosclerosis,10–13 increased free fatty acid levels,14,15 increased body fat,15,16 and increased insulin resistance.15,17 Undoubtedly, apoA-II has some unknown beneficial functions, possibly related to fatty acid metabolism or host defense.In this issue of Arteriosclerosis, Thrombosis, and Vascular Biology, Holvoet et al18 report studies of a chimeric protein between apoA-I and apoA-II, in which the central domain of apoA-I (Arg123-Tyr166) is substituted with the Ser12-Ala75 segment of apoA-II. Mice expressing this chimeric protein were compared with transgenic mice expressing similar levels of apoA-I. Although cholesterol levels were similar between the apoA-I/apoA-II transgenic mice and the apoA-I transgenic mice, atherosclerotic lesions (studied in an apoE null background) were much larger in apoA-I/apoA-II transgenics (about 2.7-fold). The apoA-I/apoA-II transgenic mice also exhibited increased macrophage homing in the aortic root (2.7-fold) and increased macrophage to smooth muscle cell ratios in lesions (2.1-fold). Thus, although apoA-I and apoA-II are close cousins, both consisting largely of a common lipid-association structural motif, the substitution of the central region of apoA-I by a portion of apoA-II has major functional consequences. The results suggest that the region is critical for functions involved in oxidative stress and inflammation but not cholesterol transport. A summary of the effects of apoA-I, apoA-II, and the apoA-I/A-II chimera are given in the Table. Table 1. Comparison of Effects of ApoAI, ApoAII, and the ApoAI/ApoAII Chimera as Studied in Transgenic MiceApoA-IApoA-IIApoA-I/ApoA-II ChimeraFFA indicates free fatty acid.Plasma HDL cholesterolIncreased because it promotes formation of nascent HDL and activates LCATIncreased because of impaired catabolismNo effectPlasma triglyceridesApoA-I Milano variant increases triglycerides in humans and transgenic miceIncreased because increased production and decreased clearance of VLDLNo effectPlasma FFA levelsNo known effectsIncreased possibly because of altered FFA metabolism mediated through CD36 or as a consequence of peripheral insulin resistanceNo effectSR-BI receptorIncreases binding and cholesterol exchange compared with apoA-IIDecreases HDL binding and cholesterol exchange?CD36 receptorIncreased binding and cholesterol exchange compared with apoA-IIDecreases HDL binding and cholesterol exchange?ABCA-1 receptorSimilar binding as apoA-IISimilar binding as apoA-I?AtherosclerosisProtects: promotes reverse cholesterol transport; stabilizes HDL-associated anti-oxidant enzymes, PON1 and PAF-AHPromotes: inhibits several aspects of reverse cholesterol transport and lowers PON1 activity in transgenic micePromotes: increases oxidative stress through a reduction in the HDL anti-oxidant enzymes PON1 and PAF-AHInsulin resistanceNo known effectsClearly promotes insulin resistance in mice; probably promotes insulin resistance in humans?Lipid-binding affinityLower affinity than apoA-IIHigher affinity than apoA-INo effectReverse cholesterol transportPromotes all aspectsEither has no effect or impairs all aspectsNo effectParaoxonaseIncreases PON1 activityPON1 activity reduced by 50%, based on HDL protein, in apoA-II transgenic micePON1 activity reduced compared with apoA-I transgenic mice on an apoE-null backgroundPAF acetyl-hydrolaseIncreases activity in apoA-I transgenic mice; activity decreased in apoA-I variantsActivity not affected in apoA-II transgenic micePAF-AH activity reducedOxidized stressAnti-oxidant, partly through stabilizing PON1 and PAF-AHPro-oxidant: lowers PON1 activity and increases apoB containing lipoproteinsPro-oxidant: decreased PAF-AH activityHow does apoA-I transport lipids and contribute to the anti-inflammatory properties of HDL? ApoA-I can interact with phospholipids to form discoidal (hockey puck-shaped) complexes that are precursors of mature, spherical HDL. These apoA-I-only (LpA-I) particles subsequently become loaded with cholesteryl esters through the action of LCAT and acquire other lipoproteins, including apoA-II (LpA-I, A-II) (reviewed by Frank and Marcel19). Previous studies have revealed domains of apoA-I associated with various functions. The N-terminus (residues 1 to 43) forms a globular domain while the C-terminus (residues 44 to 241) consists largely of a series of amphipathic α-helices that that have a narrow hydrophobic face on one side and a hydrophilic face on the other. Mutagenesis and other approaches have defined a number of regions as important in lipid binding: residues 44 to 65, 100 to 121, 122 to 143, and 210 to 241. Residues 144 to 186, however, contribute relatively little to lipid binding. The LCAT activation domain encompasses helices 144 to 165 and 166 to 186. The C-terminal region of apoA-I seems to be important for the interaction of lipid-free apoA-I with macrophages and for specific lipid efflux.If, indeed, the amphipathic helices are a key to the antiatherogenic functions of apoA-I, why then does apoA-II not protect similarly, since it is also composed largely of amphipathic helices? The answer seems to be that the precise sequence of the helices, not simply their amphipathic nature, is crucial for the functions of apoA-I.20 It is probably significant that apoA-I helices contain both Lys and Arg as positively charged amino acids, whereas apoA-II contains only Lys. Some studies suggest that that Arg residues at the polar-nonpolar interface of apoA-I are important for LCAT activation.21 There are also other differences between the helices of apoA-I and apoA-II, such as the presence of bulky Ile and Phe residues in apoA-II. Such differences could conceivably influence interactions with lipids, cell membranes, or proteins. It is noteworthy that Anantharmaiah and colleagues20 have produced α-helical peptides that form peptido-lipid complexes similar to those formed by intact apoA-I. Like intact apoA-I, these peptides are capable of inhibiting atherogenesis and of clearing oxidized pro-inflammatory lipids. Thus, it seems that certain short amphipathic helical domains of apoA-I can mimic at least some of the properties of intact apoA-I.It is likely that the effects of the A-I/A-II chimera on atherosclerosis and inflammation are caused, in part, by interactions with two anti-inflammatory enzymes carried on HDL, platelet activating factor acetyl hydrolase (PAF-AH) and serum paraoxonase (PON1). In tissue cultures of vascular cells, HDL isolated from transgenic mice overexpressing apoA-II promoted the formation of lipid hydroperoxides and the activation of inflammatory genes in cells.8 Such apoA-II transgenic HDL had only about 50% of the PON1 activity compared with HDL from nontransgenic mice, and PON1 supplementation of HDL from the apoA-II transgenic mice significantly reduced the proinflammatory properties of the HDL.8 Overexpression of apoA-I, however, resulted in increased PON1 and PAF-AH activities.22 Although plasma paraoxonase activities were not determined in the present study, the activity of PAF-AH was significantly reduced. It would seem that the region of apoA-I substituted in the present study by Holvoet et al18, is important for stabilizing PAF-AH activity and possibly paroxonase. Other regions of apoA-I important for stabilizing paraoxonase have been identified by using mutagenesis studies.23The increased lesion formation observed in the present study, as well as in the apoA-II transgenic mice, may also be mediated in part through interaction with cell surface receptors, and the scavenger receptor CD36 is a good candidate. The CD36 receptor is expressed in a variety of cell types including endothelial cells, and it binds both native and oxidized lipoproteins. The latter have been demonstrated to induce a variety of pro-inflammatory genes through a second messenger system involving nuclear factor-κB.24 HDL from apoA-II transgenic mice exhibited reduced binding to the CD36 receptor.15 Recent studies with apoA-II transgenic mice provide strong evidence that HDL can induce insulin resistance in skeletal muscle,15 suggesting such receptor interactions. It is interesting to speculate that HDL with the chimeric apoA-I may exhibit altered interaction with CD36.Elevated expression of apoA-II dramatically increased plasma levels of VLDL, LDL, and remnant particles, most likely through both increased synthesis and decreased catabolism.8,10,15 Still another potential pathway by which the apoA-I/A-II chimera may influence atherosclerosis and vascular inflammation is via effects on triglyceride-rich lipoproteins. Overexpression of apoA-II also resulted in displacement of apoE and apoCs from HDL to VLDL, LDL, and remnant-sized particles, which may contribute to their altered catabolism.10 Although in the present study no differences in plasma lipid concentrations were observed, βVLDL from the apoA-I/A-II transgenic mice were significantly different from βVLDL from the apoA-I transgenic mice with respect to stimulation of macrophage homing.These results by Holvoet and colleagues18 remind us that we still have much to learn about the functions of apoA-I and apoA-II. Most studies have focused on the lipid transport aspects of these molecules, but it is clear that functions independent of lipid transport are likely to be very important with respect to atherogenesis. In this regard, it is interesting that a recent report indicates that ABCA-1-null mice have a near absence of HDL cholesterol, but, nevertheless, have normal biliary secretion rates of cholesterol, bile salts, and phospholipids.25 This observation requires further study, but it suggests that the anti-inflammatory properties of HDL may be of primary importance in atherosclerosis.FootnotesCorrespondence to Lawrence W. Castellani, PhD, Department of Medicine/Division of Cardiology, 47-123 CHS, University of California, Los Angeles, CA 90095. E-mail [email protected] References 1 Sviridov D, Hoang A, Sawyer WH, Fidge NH. Identification of a sequence of apolipoprotein A-I associated with the activation of lecithin:cholesterol acyltransferase. J Biol Chem. 2000; 275: 19707–19712.CrossrefMedlineGoogle Scholar2 Sorenson RC, Bisgaier CL, Aviram M, Hsu C, Billecke S, La Du BN. Human serum paraoxonase/arylesterase’s retained hydrophobic N-terminal leader sequence associates with HDLs by binding phospholipids: apolipoprotein A-I stabilizes activity. Arterioscler Thromb Vasc Biol. 1999; 19: 2214–2225.CrossrefMedlineGoogle Scholar3 Ng DS, Vezina C, Wolever TS, Kuksis A, Hegele RA, Connelly PW. Apolipoprotein A-I deficiency: biochemical and metabolic characteristics. Arterioscler Thromb Vasc Biol. 1995; 15: 2157–2164.CrossrefMedlineGoogle Scholar4 Rubin EM, Krauss RM, Spangler EA, Verstuyft JG, Clift SM. Inhibition of early atherogenesis in transgenic mice by human apolipoprotein AI. Nature. 1991; 353: 265–267.CrossrefMedlineGoogle Scholar5 Segrest JP, Li L, Anantharamaiah GM, Harvey SC, Liadaki KN, Zannis V. Structure and function of apolipoprotein A-I and high-density lipoprotein. Curr Opin Lipidol. 2000; 11: 105–115.CrossrefMedlineGoogle Scholar6 Barbaras R, Puchois P, Fruchart JC, Ailhaud G. Cholesterol efflux from cultured adipose cells is mediated by LpAI particles but not by LpAI:AII particles. Biochem Biophys Res Comm. 1987; 142: 63–69.CrossrefMedlineGoogle Scholar7 Castro G, Nihoul LP, Dengremont C, de Geiter C, Delfly B, Tailleux A, Fievet C, Duverger N, Denefle P, Fruchart JC, Rubin EM. Cholesterol efflux, lecithin-cholesterol acyltransferase activity, and pre-beta particle formation by serum from human apolipoprotein A-I and apolipoprotein A-I/apolipoprotein A-II transgenic mice consistent with the latter being less effective for reverse cholesterol transport. Biochemistry. 1997; 36: 2243–2249.CrossrefMedlineGoogle Scholar8 Castellani LW, Navab M, Lenten BJ, Hedrick CC, Hama SY, Goto AM, Fogelman AM, Lusis AJ. Overexpression of apolipoprotein AII in transgenic mice converts high density lipoproteins to proinflammatory particles. J Clin Invest. 1997; 100: 464–474.CrossrefMedlineGoogle Scholar9 Deeb SS, Takata K, Peng RL, Kajiyama G, Albers JJ. A splice-junction mutation responsible for familial apolipoprotein A-II deficiency: a splice-junction mutation responsible for familial apolipoprotein A-II deficiency. Am J Hum Genet. 1990; 46: 822–827.MedlineGoogle Scholar10 Warden CH, Hedrick CC, Qiao JH, Castellani LW, Lusis AJ. Atherosclerosis in transgenic mice overexpressing apolipoprotein A-II. Science. 1993; 261: 469–472.CrossrefMedlineGoogle Scholar11 Mehrabian M, Qiao J-H, Hyman R, Ruddle D, Laughton, Lusis AJ. Influence of the apoA-II gene locus on HDL levels and fatty streak development in mice. Arterioscler Thromb. 1993; 13: 1–10.CrossrefMedlineGoogle Scholar12 Escola-Gil JC, Marzal-Casacuberta A, Julve-Gil J, Ishida BY, Ordonez-Llanos J, Chan L, Gonzalez-Sastre F, Blanco-Vaca F. Human apolipoprotein A-II is a pro-atherogenic molecule when it is expressed in transgenic mice at a level similar to that in humans: evidence of a potentially relevant species-specific interaction with diet. J Lipid Res. 1998; 39: 457–462.CrossrefMedlineGoogle Scholar13 Schultz JR, Gong EL, McCall MR, Nichols AV, Clift SM, Rubin EM. Expression of human apolipoprotein A-II and its effect on high density lipoproteins in transgenic mice. J Biol Chem. 1992; 267: 21630–21636.CrossrefMedlineGoogle Scholar14 Warden CH, Daluiski A, Bu X, Purcell-Huynh DA, De Meester C, Shieh BH, Puppione DL, Gray RM, Reaven GM, Chen YD et al. Evidence for linkage of the apolipoprotein A-II locus to plasma apolipoprotein A-II and free fatty acid levels in mice and humans. Proc Nat Acad Sci U S A. 1993; 90: 10886–10890.CrossrefMedlineGoogle Scholar15 Castellani LW, Goto AM, Lusis AJ. Studies with apolipoprotein A-II transgenic mice indicate a role for HDLs in adiposity and insulin resistance. Diabetes. 2001; 50: 643–651.CrossrefMedlineGoogle Scholar16 Van’t Hooft FM, Ruotolo G, Boquist S, de Faire U, Eggertsen G, Hamsten A. Human evidence that the apolipoprotein a-II gene is implicated in visceral fat accumulation and metabolism of triglyceride-rich lipoproteins. Circulation. 2001; 104: 1223–1228.CrossrefMedlineGoogle Scholar17 Weng W, Breslow JL. Dramatically decreased high density lipoprotein cholesterol, increased remnant clearance, and insulin hypersensitivity in apolipoprotein A-II knockout mice suggest a complex role for apolipoprotein A-II in atherosclerosis susceptibility. Proc Nat Acad Sci USA. 1996; 93: 14788–14794.CrossrefMedlineGoogle Scholar18 Holvoet P, Peeters K, Lund-Katz S, Mertens A, Verhamme P, Quarck R, Stengel D, Lox M, Deridder E, Bernar H, Nickel M, Theilmeier G, Ninio E, Phillips MC. Arg123-Tyr166 domain of human apoA-I is critical for HDL-mediated inhibition of macrophage honing and early atherosclerosis in mice. Arterioscler Thromb Vasc Biol. 2001; 21: 1977–1983.CrossrefMedlineGoogle Scholar19 Frank PG, Marcel YL. Apolipoprotein A-I. structure-function relationships. J Lipid Res. 2000; 41: 853–872.CrossrefMedlineGoogle Scholar20 Garber D, Datta G, Chaddha M, Palgunachari MN, Hama SY, Navab M, Fogelman AM, Segrest JP, Anantharamaiah GM. A new synthetic class A amphipathic peptide analogue protects mice from diet-induced atherosclerosis. J Lipid Res. 2001; 42: 545–552.CrossrefMedlineGoogle Scholar21 Roosbeek S, Vanloo B, Duverger N, Caster H, Breyne J, De Beun I, Patel H, Vanderkerckhove J, Shoulders C, Rossenue M, Peelman F. Three arginine residues in apolipoprotein A-I are critical for activation of lecithin:cholesterol acyltransferase. J Lipid Res. 2001; 42: 31–40.CrossrefMedlineGoogle Scholar22 De Geest B, Stengel D, Landeloos M, Lox M, Le Gat L, Collen D, Holvoet P, and Ninio E. Effect of overexpression of human apoA-I in C57BL/6 and C57BL/6 apoE-deficient mice on 2 lipoprotein-associated enzymes, platelet activating factor acetylhydrolase and paraoxonase. Comparison of adenovirus-mediated human apoA-I gene transfer and human apoA-I transgenesis. Arterioscler Thromb Vasc Biol. 2000; 20 (10): E68–E75.CrossrefMedlineGoogle Scholar23 Oda MN, Bielicki JK, Berger T, Forte TM. Cysteine substitutions in apolipoprotein A-I primary structure modulate paraoxonase activity. Biochemistry. 2001; 40: 1710–1718.CrossrefMedlineGoogle Scholar24 Yakub MJ, Yamashita S, Matsumoto K, Nozaki S, Matsuzawa Y. Oxidized low density lipoprotein-induced activation of NF-kB and subsequent expression of a variety of proinflammatory and proatherogenic genes are defective in monocyte-derived macrophages from CD36-deficient mice. Circulation. 1999; 100 (suppl 1): I-684–685.MedlineGoogle Scholar25 Groen AK, Bloks VW, Bandsma RH, Ottenhoff R, Chimini G, Kuipers F. Hepatobiliary cholesterol transport is not impaired in Abca1-null mice lacking HDL. J Clin Invest. 2001; 108: 843–850.CrossrefMedlineGoogle Scholar Previous Back to top Next FiguresReferencesRelatedDetailsCited By Sun X, Chen R, Yan G, Chen Z, Yuan H, Huang W and Lu Y (2020) Gender-specific associations between apolipoprotein A1 and arterial stiffness in patients with nonalcoholic fatty liver disease, PeerJ, 10.7717/peerj.9757, 8, (e9757) Chen H, Shao Z, Gao Y, Yu X, Huang S and Zeng P (2020) Are blood lipids risk factors for fracture? Integrative evidence from instrumental variable causal inference and mediation analysis using genetic data, Bone, 10.1016/j.bone.2019.115174, 131, (115174), Online publication date: 1-Feb-2020. Rodriguez-Cuenca S, Carobbio S, Barceló-Coblijn G, Prieur X, Relat J, Amat R, Campbell M, Dias A, Bahri M, Gray S and Vidal-Puig A (2018) P465L-PPARγ mutation confers partial resistance to the hypolipidaemic action of fibrates, Diabetes, Obesity and Metabolism, 10.1111/dom.13370, 20:10, (2339-2350), Online publication date: 1-Oct-2018. Lek M, Cruz S, Ibe N, Beck W, Bielicki J, Weers P, Narayanaswami V and Feng Y (2017) Swapping the N- and C-terminal domains of human apolipoprotein E3 and AI reveals insights into their structure/activity relationship, PLOS ONE, 10.1371/journal.pone.0178346, 12:6, (e0178346) Moradi H, Said H and Vaziri N (2013) Post-transcriptional nature of uremia-induced downregulation of hepatic apolipoprotein A-I production, Translational Research, 10.1016/j.trsl.2012.11.001, 161:6, (477-485), Online publication date: 1-Jun-2013. Hajhosseiny R, Khavandi K and Goldsmith D (2012) Cardiovascular disease in chronic kidney disease: untying the Gordian knot, International Journal of Clinical Practice, 10.1111/j.1742-1241.2012.02954.x, 67:1, (14-31), Online publication date: 1-Jan-2013. Chan D, Ng T and Watts G (2011) Apolipoprotein A-II: Evaluating its significance in dyslipidaemia, insulin resistance, and atherosclerosis, Annals of Medicine, 10.3109/07853890.2011.573498, 44:4, (313-324), Online publication date: 1-Jun-2012. Chan D, Watts G, Ooi E, Chan D, Wong A and Barrett P (2011) Apolipoprotein A-II and adiponectin as determinants of very low-density lipoprotein apolipoprotein B-100 metabolism in nonobese men, Metabolism, 10.1016/j.metabol.2011.03.003, 60:10, (1482-1487), Online publication date: 1-Oct-2011. Walldius G and Jungner I (2007) Apolipoprotein A-I versus HDL cholesterol in the prediction of risk for myocardial infarction and stroke, Current Opinion in Cardiology, 10.1097/HCO.0b013e3281bd8849, 22:4, (359-367), Online publication date: 1-Jul-2007. Dahlbäck B and Nielsen L (2006) Apolipoprotein M – a novel player in high-density lipoprotein metabolism and atherosclerosis, Current Opinion in Lipidology, 10.1097/01.mol.0000226122.10005.88, 17:3, (291-295), Online publication date: 1-Jun-2006. Táborský L, Adam P, Sobek O, Dostál M, Dvořáková J and Dubská L (2003) Levels of apolipoprotein A-II in cerebrospinal fluid in patients with neuroborreliosis are associated with lipophagocytosis, Folia Microbiologica, 10.1007/BF02931523, 48:6, (849-855), Online publication date: 1-Dec-2003. Allayee H, Ghazalpour A and Lusis A (2003) Using Mice to Dissect Genetic Factors in Atherosclerosis, Arteriosclerosis, Thrombosis, and Vascular Biology, 23:9, (1501-1509), Online publication date: 1-Sep-2003.Parhami F, Basseri B, Hwang J, Tintut Y and Demer L (2002) High-Density Lipoprotein Regulates Calcification of Vascular Cells, Circulation Research, 91:7, (570-576), Online publication date: 4-Oct-2002.Brown B, Cheung M, Lee A, Zhao X and Chait A (2002) Antioxidant Vitamins and Lipid Therapy, Arteriosclerosis, Thrombosis, and Vascular Biology, 22:10, (1535-1546), Online publication date: 1-Oct-2002. Blanco Vaca F (2002) La saturación de la grasa en la dieta afecta la concentración de apolipoproteína AII y la composición de las HDL en las mujeres menopáusicas, Clínica e Investigación en Arteriosclerosis, 10.1016/S0214-9168(02)78849-9, 14:3, (167-168), Online publication date: 1-Jan-2002. December 2001Vol 21, Issue 12 Advertisement Article InformationMetrics https://doi.org/10.1161/atvb.21.12.1870PMID: 11742857 Originally publishedMarch 15, 2018 PDF download Advertisement

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call