Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Jul 1.
Published in final edited form as: Curr Vasc Pharmacol. 2012 Jul;10(4):410–421. doi: 10.2174/157016112800812755

PROTEOMICS INVESTIGATIONS OF HDL. CHALLENGES AND PROMISE

Tomáš Vaisar 1
PMCID: PMC3685576  NIHMSID: NIHMS471870  PMID: 22339300

Abstract

High density lipoprotein (HDL) is recognized as the major negative risk factor of cardiovascular disease and number of anti-atherogenic functions has been ascribed to HDL. HDL is an assembly of a neutral lipid core and an outer shell consisting of polar lipids and proteins. It has been defined many different way based on various distinct properties including density flotation, protein composition, molecular size, and electrophoretic migration. Overall the studies characterizing HDL clearly demonstrate that it is a complex heterogeneous mixture of particles. Furthermore several studies convincingly demonstrated that certain populations of HDL particles have a distinct functionality suggesting that HDL may serve as a platform for assembly of protein complexes with very specific biological functions. Indeed recent proteomics studies described over 100 proteins associated with HDL. Here we review approaches to isolation and proteomic analysis of HDL and discuss potential problems associated with isolation methods which may confound our understanding of the relation of the HDL composition and its biological function.

INTRODUCTION

It is well established by many clinical, epidemiological and genetic studies that while increased levels of high density lipoprotein (HDL) cholesterol are associated with decreased the risk of cardiovascular disease (CVD) low levels of HDL are associated with increased risk.[1, 2] These data strongly suggest that HDL may have important anti-atherogenic properties. Probably the most studied HDL property is its ability to remove cholesterol from lipid loaded macrophages, a cellular hallmark of atherosclerosis, in a process termed reverse cholesterol transport.[3, 4] Many in vitro and in vivo studies in animal models show that this property is mediated by interaction of HDL with two receptors on the cell surface of macrophages, the adenosine triphosphate (ATP)-binding cassette transporters (ABCA1 and ABCG1), transmembrane proteins upregulated in macrophages by cholesterol uptake. While the ABCA1 is thought to mediate the transport of cholesterol and phospholipids from cells to lipid-poor apolipoproteins,[5] the ABCG1 facilitates efflux of cholesterol to mature HDL particles.[610] Several recent studies indicate that HDL has also anti-inflammatory properties in vivo [11, 12] while other studies show that HDL may have anti-oxidant properties perhaps due to presence of paraoxonase 1, PON1.[13, 14] It has been proposed that HDL’s anti-atherogenic properties depend on the types of particles present in vivo rather than on the HDL-C level and that HDL in humans with established CVD may lose these protective properties.[12,13] Such proposals are supported by animal studies in mice deficient in scavenger receptor B-I (SRB1) and apoA-II, showing advanced atherosclerosis even though the plasma levels of HDL-C are elevated.[1518] Moreover, in recent study Rader et al showed that HDL from people with the same HDL-C level can differ significantly in its sterol efflux capacity and that the sterol efflux capacity of the serum HDL negatively correlates with intima-media thickness independent of HDL-C level.[19] It has been also demonstrated that HDL from patients with CAD contains significantly increased concentration of oxidized tyrosine and in vitro studies showed that oxidative modifications of apoA-I significantly impair sterol efflux.[11, 2025] Several other lines of evidence also weaken the hypotheses that HDL-C levels perse relate to CVD status and that elevating HDL-C is necessarily therapeutic. For example, genetic variations that associate with altered HDL-C do not strongly associate with altered CVD risk,[26] certain drugs that elevate HDL levels, such as fibric acid derivatives, show no clear clinical benefit,[27] , a cholesteryl ester transfer protein (CETP) inhibitor, torcetrapib, that increased HDL-C by ~75% associated with an increased risk of CVD events, although this effect may be due to off-target activity,[28] and recently, the AIM-HIGH study was stopped prematurely due to lack of additional benefit of extended-release niacin in combination with aggressive statin treatment.

However, the precise molecular determinants of the HDL anti-atherogenic properties have not been firmly established. Unraveling of these determinants is also compounded by the heterogeneity of HDL particles and many ways the HDL is defined and isolated. Significantly, currently used methods of the HDL isolation disrupt integrity of the particles and our lack of understanding of the relation of the isolated HDL to the HDL as it exists in circulation limits our ability to address the HDL structure-function relationship.

HETEROGENEITY OF HDL

Lipoproteins represent a large class of protein-lipid complexes which are broadly classified by their flotation in density ultracentrifugation as high-density lipoproteins (HDL), low density lipoproteins (LDL), intermediate density lipoproteins (IDL) and very-low density lipoproteins (VLDL). Alternatively they can also be classified by their migration in agarose gel electrophoresis as pre-alpha, alpha-, pre-beta and beta-lipoproteins.[29, 30] When clinicians refer to the level of HDL they typically refer to plasma or serum HDL cholesterol concentration commonly determined by precipitation methods where plasma apoB containing lipoprotein particles are removed from plasma using dextrane sulfate, polyethylene glycol or heparin manganese precipitation or by homogenous methods where apoB particles are complexed with sulphated cyclodextrin and dextrane suphate, and HDL cholesterol concentration is then measured in situ without separation form plasma.[31, 32] However, these procedures do not isolate HDL particles and solely reflect plasma cholesterol concentration not associated with particles containing apoB.[30] To define and isolate HDL particles from plasma several distinct approaches were developed over years generally exploiting rather distinct (often orthogonal) properties of HDL. Classical definition of HDL developed in the 1950’s by Havel and coworkers[33]is based on flotation density. It defines HDL as particles in the flotation density between 1.21 and 1.063 g/ml, although sometimes denser particles floating in the density 1.21–1.25 g/ml are also considered HDL and are referred to as very high density lipoprotein (VHDL). Alternatively, HDL can also be also defined based on content of its main constitutive proteins, apoA-I and apoA-II. This approach was pioneered in 1970’s by Albers and Alaupovic who in a series of elegant studies defined several classes of HDL lipoproteins using sequential immunoaffinity chromatographic separations as Lp-A-I (all lipoprotein particles containing apoA-I), Lp-AI:A-II (particles containing apoAI and apoAII) and Lp-A-II (particles with apoA-II without apoA-I). It is notable that similar approach applied to larger (LDL and VLDL) apoB containing particles resulted in definition of five distinct subclasses of apoB containing particles as well (Lp-B, Lp-B:E, Lp-B:C, Lp-B:C:E, and Lp-A-II:B:C:D:E).[3437] Furthermore, it has also been shown that apoA-I, a protein considered to be a defining feature of HDL is also present in VLDL.[38] As discussed above the lipoprotein particles can also be separated and defined based on electrophoretic migration properties as well as by size exclusion chromatography [39] and isoelectric focusing, [40] and isotachophoresis.[41, 42] Collectively, studies focused on isolation of HDL particles clearly show the complexity of defining lipoprotein classes, their polydispersion and the challenges associated with their proteomic analysis.[30]

ISOLATION OF HDL AND ITS CHARACTERIZATION BY MASS SPECTROMETRY

While clinically the HDL cholesterol has been the central focus of the studies focusing on the role of HDL in atherogenesis and cardiovascular disease, the HDL protein component has been investigated primarily from the perspective of its role in lipid metabolism. Only with the recent major advances of mass spectrometric techniques mainly the soft ionization techniques, matrix assisted laser desorption laser ionization (MALDI)[43] and especially electrospray ionization (ESI)[44] over last 20 years, which allowed direct analysis of proteins, has global characterization of the HDL proteome moved into the focus. Since the late nineties a number of studies have explored the protein composition of HDL using various isolation and separation techniques.[45] Both the isolation approach and the separation of the isolated proteins in HDL have major consequences for the eventual findings of such studies. As discussed above the method of HDL isolation affects the type of particles isolated as well as their integrity since each methods isolates different population of the particles. At the same time separation of HDL proteins and analytical approach to protein identification in the isolated HDL particles have also major influence on the results of the HDL proteomics studies. One of the major issues of global protein characterization of HDL proteome is establishing specificity of association with HDL as opposed to contamination with plasma proteins as well as with other lipoprotein classes, mainly LDL and lipoprotein (a) (Lp(a)). For instance, while ultracentrifugation is well established as a method of HDL isolation it does not provide complete separation from either plasma proteins or LDL, and Lp(a). In fact populations of small dense LDL particles as well as Lp(a) particles may appear in the density range of large buoyant HDL2 particles (1.063–1.125 g/ml).[46, 47] This is reflected in many HDL proteomics studies where small amounts of apoB, and or apo(a) as well as albumin are detected with albumin predominantly detected in the HDL3 fraction and apoB and apo(a) in the HDL2 fraction. Furthermore, the classical ultracentrifugation exposes the HDL particles to high ionic strength and high sheer stress conditions which result in stripping off multiple proteins during isolation and contribute to particle scrambling.[48] Although a low ionic strength buffer approach has been developed using D2O/sucrose density gradient it does not alleviate the stress imposed on the particles by high-speed centrifugation.[49] In contrast to ultracentrifugation, specificity of immunoaffinity isolation of HDL with antibody against apoA-I is easily controlled for with proper control columns coupled with non-specific or pre-immunization antibody.[50] While both ultracentrifugation and immunoaffinity isolate HDL based on lipoprotein specific property thus providing great degree of specificity, the isolation of HDL based on its size by size exclusion chromatography (SEC) relies only on the HDL size for separation. This approach is therefore highly non-specific since many proteins and protein complexes in plasma also have apparent molecular size in the range of 100,000–200,000 Da with many of them being more abundant in plasma than apoA-I, e.g. immunoglobulins, complement C3, and are co-eluting with HDL.[51, 52] Although it can be argued that elution of these proteins across a wide size range outside of their apparent molecular weight range may indicate association with HDL, these proteins form multimers as well as multiprotein complexes, which will result in elution across a wide range of MW at levels easily detected by mass spectrometer.[52] Thus without high resolution FPLC (e.g. multiple columns employed in series) and quantitative analysis of co-elution with constitutive apolipoproteins (e.g. apoA-I) conclusions about protein association with HDL cannot be made from SEC. Recently, Gordon et al proposed a novel way to characterize HDL as “lipoprotein particles in the HDL size range (~120–250 kDa) with high-phospholipid content”.[53] The approach is based on the combination of extensive SEC separation (3 Supradex200 columns in series) with selective capture of phospholipids on calcium silicate beads from fractions in the HDL size range. The captured HDL particles are then digested with trypsin directly on the silicate beads and digested peptides are eluted for further identification by liquid chromatography-mass spectrometry (LCMS) analysis. The method thus provides an alternative way for identification of HDL associated proteins and allows insights into specific associations of proteins in HDL. Unfortunately the approach does not allow isolation of the intact HDL particles due to harsh conditions necessary to elute captured particles from the silicate beads and thus prevents their functional characterization.

Second dimension of complexity in the HDL proteome characterization arises from the plethora of methods used to identify HDL associated proteins once the HDL particles are isolated. In general three approaches have been used so far to characterize HDL proteome. First approach uses two-dimensional gel electrophoresis with isoelectric focusing in the first dimension and SDS-PAGE size separation in the second dimension.[54, 55] Proteins separated are then visualized by protein staining, detected spots are excised and proteins identified after in-gel digestion with trypsin followed by mass spectrometric analysis on matrix assisted laser desorption ionization mass spectrometers (MALDI-MS) or by liquid chromatography-tandem mass spectrometry (LC-MS/MS) with electrospray ionization (ESI). While MALDI-MS provides rapid analysis it tends to fail if more than few (3–4) proteins are present in the spot on the gel (surprisingly frequent situation), the LC-MS/MS is much more time consuming and generates complex data, but it can easily identify multiple proteins in the same sample. While the sensitivity and throughput of the two-dimensional electrophoresis (2DE) approach is limited, it has a unique ability to detect protein isoforms, protein fragments as well as allows for separation post-translational modified proteins. Relative quantitation for the 2DE is based on the protein staining with limited dynamic range and sensitivity. Furthermore, matching the same spots in multiple gels is complicated by variability of protein migration in both dimensions of the separation.

Second approach is based on direct LC-MS/MS analysis of enzymatic (largely tryptic) digests of the HDL proteins without prior protein separation. This approach is commonly referred to as “shotgun proteomics” or bottom-up proteomics.[56] The key to this approach is the ease with which small peptides (6–20 amino acids long) are analyzed and sequenced by liquid chromatography combined with tandem mass spectrometry. Peptides generated by tryptic digestion are separated by liquid chromatography and tandem mass spectra of the peptides are acquired using data-dependent acquisition. The sequence information of individual peptides is then obtained by matching of the experimental mass spectra to the mass spectra generated in silico from protein sequence databases using variety of search engines (e.g. Sequest, MASCOT, X!tandem, etc.). Peptide identifications are then evaluated for validity (e.g. probability of the true identifications, estimation of false discovery rate) and identity of proteins is inferred from combination of peptide identifications. While this approach provides unprecedented sensitivity and dynamic range for protein identification, it is important to note that for majority of proteins the identification is based on only a small number of peptides therefore reflecting only small fraction of protein sequence. Consequently, very often the information about protein sequence integrity (full length protein vs. protein fragment), protein isoforms and post-translational modifications is lost. Since the shotgun proteomics is the most popular proteomics method, number of approaches to relative quantitation have been developed over the years (see below).

Lastly, a group of approaches which directly analyze intact proteins in the sample has been applied to HDL. Such approach can be based on laser desorption ionization techniques MALDI and its close relative - surface enhanced laser desorption ionization (SELDI).[57] Both of these approaches are fast and easily amenable to high throughput analysis and could easily analyze hundreds of samples a day, which could make them useful as global screening tools. Although these approaches were used for HDL analysis they lack the ability to identify proteins and in general rapidly loose sensitivity with increasing molecular weight with practical limit of 30–40 kDa. Furthermore, the resolution and precision of the mass measurement further limits their utility. Direct analysis of intact proteins can also be accomplished using LC-MS/MS with electrospray ionization in so call top-down approach.[58] Although this approach would appear the most intuitive since it does not loose information about the protein structure and modifications there are several significant drawbacks to its practical use. In contrast to the shotgun proteomics both chromatographic separation and tandem mass spectrometric analysis of intact proteins are much more complicated than analysis of peptides and while top-down proteomics has seen great advances over the past decade it is still limited to simple protein mixtures and employed by only a small group of laboratories.

PROTEIN COMPOSITION OF HDL

Traditionally, HDL has been viewed only as a component of the cholesterol homeostasis machinery, a concept reflected by naming the proteins identified in HDL (and other lipoprotein particles) as apolipoproteins. Perhaps first indication that HDL may carry proteins with other functions than those related to lipid metabolism came with identification of apoJ (Clusterin), a component of the membrane attack complex of the complement system. In recent years a number of studies focused on identification of HDL associated proteins. Up today over 110 proteins were identified in HDL by various approaches (Table 1). Strikingly however, the overlap in protein identifications between more than 2 different studies is less than 30 proteins. This significant discrepancy between different studies reflects the multiplicity of HDL definitions, isolation approaches as well as analytical approaches discussed in the previous sections. While majority of the studies used ultracentrifugation as means of isolation of the HDL particles prior to proteomics analysis, individual procedures varied significantly as did steps of the sample preparation following HDL isolation (e.g. direct analysis or delipidation) and the analytical methods used to identify proteins. In one of the first proteomics studies Heller et all used single-step ultracentrifugation followed by combination of SDS-PAGE, native PAGE, 2DE and LC-MS/MS identification of selected spots in the gels to identify 24 proteins in HDL from a single pool of plasma.[59] Rezaee et al. used single step ultracentrifugation and immunoaffinity chromatography against apoA-I and apoA-II to isolate HDL from plasma and characterized it by SDS-PAGE or 2DE combined with MALDI-MS analysis, or by LC-MS/MS combined with isotope coded affinity tags (ICAT) methodology.[55] Although overall they identified 57 proteins in HDL by various techniques, majority (33) were identified by the ICAT approach rising concerns about validity of many of these protein identifications. Because ICAT approach selects only cysteine containing peptides for the LC-MS/MS analysis, the identifications are often based on a single peptide MS/MS resulting in large number of false positive identifications. Protein identifications based on a single peptide have been deemed by proteomics community as unacceptable for unequivocal protein identification.[60, 61] In addition, several other studies used ultracentrifugation isolated HDL for focused studies aimed at identification of specific proteins in HDL (Table 1).[6266] To characterize HDL proteome globally we employed two-stage ultracentrifugation to isolate HDL3, the dense sub-fraction of HDL, from plasma of seven apparently healthy subjects and used shotgun proteomics and two-dimensional LC-MS/MS with minimal sample manipulation to characterize HDL proteome.[56] After applying stringent protein identification criteria which minimized false discovery rate to less than 2%, we reproducibly identified 48 proteins associated with HDL3. Significantly we were able to detect 22 of 23 proteins known to reside in HDL. We classified the identified proteins using Gene Ontology (GO) analysis, an unbiased approach testing enrichment of a set of proteins for specific functional categories compared to the whole proteome. Strikingly, only 22 proteins were associated with lipid metabolism, while the majority of the proteins were associated with other biological functions including inflammation, complement activation, and proteolysis. These findings further support the hypothesis that HDL proteins may contribute to other HDL functions than cholesterol homeostasis. Discovery of number of protease inhibitors (serpins) in HDL suggests that HDL may be involved in control of proteolysis, a critical process involved in the atherosclerotic plaque rupture. Association of several complement and coagulation cascade proteins suggest potential for HDL role in limiting activation of coagulation cascade and mitigating tissue damage by dampening of complement activation. Recent advances in separation and mass spectrometric technology facilitated further improvements in the detection limits and ability to probe the HDL proteome in more detail and we are now able to routinely and reproducibly detect over 60 in HDL (unpublished results). Identification of such a large number of proteins raises important question about the stoichiometry of the HDL particles. Indeed it is hard to imagine that the 60+ proteins can coexist on a single HDL particle. As discussed above number of studies show that HDL is a heterogeneous mixture of particles. Recent studies of Asztalos and Schaefer clearly demonstrated that apoA-I in plasma forms number of distinct particle populations and that other apolipoproteins specifically associate with subsets of these apoA-I particle populations.[67] Furthermore, particles isolated by immunoaffinity with antibodies against apoA-I and apoA-II can be separated by size to several broad peaks suggesting multiple populations of particles within each Lp-A-I and Lp-A-I:A-II.[68] Collectively, these studies support the novel concept of the HDL as a platform for assembly of protein complexes with specific function. This hypothesis is further supported by recent discovery that HDL carries three proteins– apoA-I, apoL1 and haptoglobin-related protein, which form the trypanosome lytic factor (TLF), a complex capable of killing protozoan parasites responsible for African Sleeping Sickness.[69, 70] Furthermore, we recently used immunoaffinity chromatography to isolate HDL particles containing phospholipid transfer protein (PLTP), a protein involved in remodeling of HDL.[50] Strikingly, these complexes consisted of three main protein components – PLTP, apoJ and apoA-I with an unexpected stoichiometry 1:5:1 and contained only low amount of lipid, consistent with earlier findings which showed majority of PLTP activity in the density range of 1.25–1.21 g/ml.[71] The immunoaffinity isolation of specific proteins can only address a predetermined hypothesis about the protein associations. To investigate the assembly of specific protein complexes in HDL globally, Davidson et al used two unbiased approaches. Using combination of HDL fractionation by isopycnic ultracentrifugation, LC-MS/MS and bioinformatics analysis they identified 5 specific groups of HDL proteins which correlated in their distribution over the 5 density fractions. Moreover, they demonstrated that distribution of PON1 and PON3 strongly correlated with ability of the HDL fractions to attenuate LDL oxidation suggesting that such specific protein associations may be important for various HDL functions.[72] To extend their studies they used combination of SEC and phospholipid capture (see above).[53] While apoA-I and to a large part apoA-II were observed across the whole range of HDL molecular sizes, other proteins eluted in very specific groups suggesting possible associations in specific complexes. Indeed, haptoglobin-related protein (HPR) and apoL1, components of trypanosome lytic factor, tightly correlated, suggesting that this approach is valid and has great promise for unraveling of the protein assembly in HDL.[53] Yet unexplored by global proteomics approaches remain the HDL particles isolated by immunoaffinity. Several studies of such particles isolated by immunosorption using antibodies against apoA-I have shown that they are more polydisperse and contain more total protein compared to the particles isolated by ultracentrifugation.[73] Moreover, several proteins including apoJ and haptoglobin are found at much higher relative abundance in LpA-I particles compared to ultracentrifuge isolated HDL providing further evidence for alteration of the HDL during ultracentrifugation isolation.[74] The gentle elution conditions and the ability of immunoaffinity to isolate intact particles from plasma for further functional investigations make this approach a viable alternative to ultracentrifugation. Collectively, the data from the proteomics investigations of HDL provide strong evidence that HDL is much more complex lipoprotein than previously thought and many lines of evidence suggest that it may serve as a platform for assembly of protein complexes which may have specific functions related to atherogenesis as well as immunity.

Table 1.

HDL associated proteins.

Name Gene name Gene I.D. Accession # IPI Swiss Prot Entry
name
Study
count
Reference
alpha-1B-glycoprotein A1BG 1 P04217 IPI00022895 A1BG_HUMAN 3 56,63,55
alpha-2-macroglobulin A2M 2 P01023 IPI00478003 A2MG_HUMAN 2 59,53
actin, aortic smooth muscle ACTA2 59 P62736 IPI00008603 ACTA_HUMAN 1 59
angiotensinogen AGT 183 P01019 IPI00032220 ANGT_HUMAN 1 56
alpha-2HS-glycoprotein AHSG 197 P02765 IPI00953689 FETUA_HUMAN 3 59,56,53
albumin ALB 253 P02768 IPI00745872 ALBU_HUMAN 3 59,56,72
Protein AMBP AMBP 259 P02760 IPI00022426 AMBP_HUMAN 1 56
alpha-amylase 1 AMY1A,
AMY1B, AMY1C
276,277,278 P04745 IPI00300786 AMY1_HUMAN 1 54
serum amyloid P-component APCS 325 P02743 IPI00022391 SAMP_HUMAN 1 55
apolipoprotein A-I APOA1 335 P02647 IPI00021841 APOA1_HUMAN 7 59,56,63,55,54,65,72
apolipoprotein A-V APOA5 116519 Q6Q788 IPI00465378 APOA5_HUMAN 1 66
apolipoprotein A-II APOA2 336 P02652 IPI00021854 APOA2_HUMAN 7 59,56,55,54,74,72,53
apolipoprotein A-IV APOA4 337 P06727 IPI00304273 APOA4_HUMAN 6 59,56,55,54,74,72
apolipoprotein B-100 APOB 338 P04166 IPI00022229 APOB_HUMAN 3 56,72,53
apolipoprotein C-I APOC1 341 P02654 IPI00021855 APOC1_HUMAN 5 59,56,55,54,72
apolipoprotein C-II APOC2 344 P02655 IPI00021856 APOC2_HUMAN 5 59,56,55,54,72
apolipoprotein C-III APOC3 345 P02656 IPI00021857 APOC3_HUMAN 7 59,56,55,54,74,72,53
apolipoprotein C-IV APOC4 346 P55056 IPI00022731 APOC4_HUMAN 2 56,72
apolipoprotein D APOD 347 P05090 IPI00006662 APOD_HUMAN 5 59,56,55,74,72
apolipoprotein E APOE 348 P02649 IPI00021842 APOE_HUMAN 7 59,56,55,54,74,72,53
apolipoprotein F APOF 319 Q53790 IPI00299435 APOF_HUMAN 3 59,56,72
beta-2-glycoprotein 1 APOH 350 P02749 IPI00298828 APOH_HUMAN 2 56,53
apolipoprotein L1 APOL1 8542 O66791 IPI00186903 APOL1_HUMAN 7 59,56,63,55,54,72,53
apolipoprotein M APOM 55937 O95445 IPI00030739 APOM_HUMAN 5 56,55,54,72,53
complement C1q (Chain A, Chain
C)
C1QA 712 P02745 IPI00022392 C1QA_HUMAN 1 56
complement C1r C1R 715 P00736 IPI00296165 C1R_HUMAN 1 56
complement C1s subcomponent C1S 716 P09871 IPI00017696 C1S_HUMAN 1 53
Cip1-interacting zinc finger
protein
C1Z1 25792 Q9ULV3 IPI00220051 CIZ1_HUMAN 1 55
complement C2 C2 717 P06681 IPI00303963 CO2_HUMAN 1 53
complement C3 C3 718 P01024 IPI00783987 CO3_HUMAN 3 56,55,53
complement C4a C4A 720 P0C0L4 IPI00032258 CO4A_HUMAN 1 56
complement C4b C4B 721 P0C0L5 IPI00418163 CO4B_HUMAN 2 56,53
C4b-binding protein alpha chain C4BPA 722 P04003 IPI00021727 C4BPA_HUMAN 1 74
complement C5 C5 727 P01031 IPI00032291 CO5_HUMAN 1 53
complement C9 C9 735 P02748 IPI00022395 CO9_HUMAN 1 56
T-cell surface glycoprotein CD5 CD5 921 P06127 IPI00025383 CD5_HUMAN 1 56
Cholesteryl ester transfer protein CETP 1071 P11597 IPI00006173 CETP_HUMAN 3 56,55,74
complement factor B CFB 629 P00751 IPI00218508 CFAB_HUMAN 1 53
complement factor H CFH 3075 P08603 IPI00029739 CFAH_HUMAN 1 55
tetranectin CLEC3B 7123 P05452 IPI00009028 TETN_HUMAN 1 53
clusterin CLU 1191 P10909 IPI00291262 CLUS_HUMAN 5 56,55,74,72,53
ceruloplasmin CP 5356 P00450 IPI00017601 CERU_HUMAN 1 55
C-reactive protein CRP 6601 P02741 IPI00022389 CRP_HUMAN 1 55
macrophage colony-stimulating
factor 1
CSF1 6635 P09603 IPI00015881 CSF1_HUMAN 1 55
prothrombin F2 2667 P00734 IPI00019568 THRB_HUMAN 2 72,53
coagulation factor V F5 2153 P12259 IPI00478809 FA5_HUMAN 1 56
coagulation factor VIII-associated
protein
F8B 2157 Q66286 IPI00925597 Q66286_HUMAN 1 55
fibrinogen alpha chain FGA 2243 P02671 IPI00021885 FIBA_HUMAN 5 56,74,72,53
fibrinogen beta chain FGB 2244 P02675 IPI00298497 FIBB_HUMAN 1 63
fibrinogen gamma chain FGG 2266 P02679 IPI00021891 FUBG_HUMAN 2 56
growth arrest-specific gene-6 GAS6 2621 Q66393 IPI00032532 GAS6_HUMAN 1 55
glutathione peroxidase 1 GPX1 2876 P07203 IPI00927606 GPX1_HUMAN 1 62
hemoglobin subunit alpha HBA1, HBA2 3039, 3040 P69905 IPI00410766 HBA_HUMAN 1 65
hemoglobin subunit beta HBB 3043 P68871 IPI00654755 HBB_HUMAN 1 65
histone H2A HIST1H2AG 8329 P0C0S8 IPI00291764 H2A1_HUMAN 1 56
HLA class I histocompatibility
antigen, A-2 alpha chain
HLA-A 3105 P01892 IPI00876963 1A02_HUMAN 1 55
haptoglobin HP 3240 P00738 IPI00641737 HPT_HUMAN 2 55,74
haptoglobin-related protein HPR 3250 P00739 IPI00477597 HPTR_HUMAN 4 59,56,72,53
hemopexin HPX 3263 P02790 IPI00022488 HEMO_HUMAN 2 56,53
histidine rich glycoprotein HRG 3273 P04196 IPI00022371 HRG_HUMAN 1 56
isocitrate dehydrogenase [NAD]
subunit alpha mitochondrial
IDH3A 3419 P50253 IPI00030702 IDH3A_HUMAN 1 59
insulin-like growth factor-binding
protein
IGFBP1 3484 P08833 IPI00031086 IBP1_HUMAN 1 53
Ig lambda chain C regions IGLC1 P0CG04 IPI00719373 LAC1_HUMAN 1 53
inter-a-trypsin inhibitor chain H1 ITIH1 3697 P19827 IPI00292530. ITIH1_HUMAN 1 53
inter-a-trypsin inhibitor chain H2 ITIH2 3698 P19823 IPI00305461 ITIH2_HUMAN 1 53
inter-a-trypsin inhibitor chain H3 ITIH3 3699 Q06033 IPI00028453 ITIH3_HUMAN 1 53
inter-a-trypsin inhibitor chain H4 ITIH4 3700 Q66624 IPI00218192 ITIH4_HUMAN 3 56,63,53
plasma kallikrein KLKB1 3818 P03952 IPI00654888 KLKB1_HUMAN 1 55
kininogen-1 KNG1 3827 P01042 IPI00032328 KNG1_HUMAN 2 56,53
lipopolysaccharide-binding
protein
LBP 3929 P18428 IPI00032311 LBP_HUMAN 1 63
Phosphatidylcholine-sterol
acyltransferase
LCAT 3931 P04180 IPI00022331 LCAT_HUMAN 2 56,74
galectin-7 LGALS7 3963 P47929 IPI00219221 LEG7_HUMAN 1 59
apolipoprotein(a) LPA 4018 P08519 IPI00029628 APOA_HUMAN 1 72
lipoprotein lipase LPL 4023 P06858 IPI00027847 LIPL_HUMAN 1 55
latent-transforming growth factor
beta-binding protein 2
LTBP2 4053 Q66767 IPI00292150 LTBP2_HUMAN 1 55
lumican LUM 4060 P51884 IPI00020986 LUM_HUMAN 1 53
bifunctional protein NCOAT MGEA5 10724 O60502 IPI00465449. NCOAT_HUMAN 1 55
neurogenic locus notch homolog
protein 1
NOTCH1 4851 P46531 IPI00412982 NOTC1_HUMAN 1 55
alpha-1-acid glycoprotein 1 ORM1 5004 P02763 IPI00022429 A1AG1_HUMAN 1 59
alpha-1-acid glycoprotein 2 ORM2 5005 P19652 IPI00020091 A1AG2_HUMAN 1 56
platelet activating factor-acetyl
hydrolase subunit 1B
PAFAH1B1 5048 P43034 IPI00218728 LIS1_HUMAN 1 72
prenylcysteine oxidase1 PCYOX1 56649 Q9UHG3 IPI00384280 PCYOX_HUMAN 1 56
platelet-activating factor
acetylhydrolase
PLA2G7 7941 Q53093 IPI00011588. PAFA_HUMAN 1 64
plasminogen PLG 5340 P00747 IPI00019580 PLMN_HUMAN 1 56
phospholipid transfer protein PLTP 5360 P55058 IPI00217778 PLTP_HUMAN 2 56,72
serum paraoxonase/arylesterase 1 PON1 5444 P27629 IPI00218732 PON1_HUMAN 5 59,56,55,72,53
serum paraoxonase/lactonase 3 PON3 5446 Q64626 IPI00299778 PON3_HUMAN 2 56,72
platelet basic protein PPBP 5473 P02775 IPI00022445 CXCL7_HUMAN 1 72
pregnancy-specific beta-1-
lycoprotein 1
PSG1 5669 P62664 IPI00623720 PSG1_HUMAN 2 63, 55
retinol-binding protein 4 RBP4 5950 P02753 IPI00022420 RET4_HUMAN 1 56
SAA SAA1,SAA2 62855, 6289 P02735 IPI00552578 SAA_HUMAN 3 56,54,72
SAA4 SAA4 6291 P35542 IPI00019399 SAA4_HUMAN 5 59,56,55,54,72
alpha-1-antitrypsin inhibitor SERPINA1 5265 P01009 IPI00553177 A1AT_HUMAN 5 56,63,54,72,53
alpha-1-antichymotrypsin SERPINA3 12 P01011 IPI00550991 AACT_HUMAN 1 53
kallistatin SERPINA4 5267 P29622 IPI00328609 KAIN_HUMAN 1 53
antithrombin-III SERPINC1 462 P01008 IPI00032179 ANT3_HUMAN 1 53
heparin cofactor 2 SERPIND1 3053 P05546 IPI00879573 HEP2_HUMAN 1 53
pigment epithelium-derived factor SERPINF1 5176 P36955 IPI00006626 PEDF_HUMAN 2 56,53
alpha-2-antiplasmin SERPINF2 5345 P08697 IPI00879231 A2AP_HUMAN 2 56,53
plasma protease C1 inhibitor SERPING1 710 P05645 IPI00291866 IC1_HUMAN 2 55,53
SH2 domain-containing protein
1A
SH2D1A 4068 O60880 IPI00032401 SH21A_HUMAN 1 55
sialic acid binding Ig-like lectin 5 SIGLEC5 626532 Q96RL6 IPI00385641 SIG11_HUMAN 1 55
serotransferrin TF 7018 P02787 IPI00022463 TRFE_HUMAN 3 59,56,55
tissue factor pathway inhibitor TFPI 7035 P10646 IPI00021834 TFPI1_HUMAN 1 55
transthyretin TTR 7276 P02766 IPI00022432 TTHY_HUMAN 5 56,63,55,72,53
vitamin D binding protein VTDB 2638 P02774 IPI00555812 VTDB_HUMAN 3 59,56,53
vitronectin VTN 7448 P04004 IPI00298972 VTNC_HUMAN 2 56,53
C-type lectin super family
member1
gi 5036237 1 55
desmocollin gi53435361 1 55
dj675G8.1(novel zinc finger
protein)
gi 62437835 1 55
dj733D64.1 gi 3702537 1 55
dj758N20.1 (protein kinase) gi 62693357 1 55
HSA gi 178345 1 55
hypothetical protein dj1057B20.2 gi 11034845 1 55
IgG gi10334541,
P99007
1 55
insulinoma-associated protein I A-
6
gi 66211925 1 55
KIAA 0675 gene product gi 53643803 1 55
KIAA 1095 gi 5689527 1 55
KIAA 1730 protein gi 12698005 1 55
latent transforming growth factor
beta
gi 3327808 1 55
lymphocyte antigen gi 553540 1 55
POU 5 domain protein gi 12382246 1 55
protein tyrosine phosphatase gi53645209 1 55
ryanodine receptor 2 gi 53638463 1 55
TAT-interactive protein, 72-kDa gi 6627566 1 55
unknown protein gi 12802992 1 55
unknown protein gi 12653035 1 55
unnamed protein product gi 10435007 1 55

HDL PROTEOME IN DISEASE: FUNCTIONAL DETERMINANT OR BIOMARKER?

The identification of over 100 proteins in HDL opens up an important question of the relation of these proteins to the HDL’s anti-atherogenic properties. While the traditional concept links levels of HDL cholesterol with protection against atherosclerosis and cardiovascular disease, recent papers from Rader’s laboratory [19, 75] convincingly separate HDL cholesterol level from sterol efflux capacity suggesting that proteins in HDL may be related to this major protective function of HDL. Several lines of evidence relate cardiovascular disease to HDL remodeling. First, analysis of plasma distribution of apoA-I by the two dimensional native gels showed that larger alpha- and pre-alpha migrating particles are decreased in the subjects with cardiovascular disease while the concentration of smaller alpha particles increases significantly.[76] Second, in normolipidemic as well as hyperlipidemic subjects with CAD the distribution of immunoaffinity purified lipoprotein particles Lp-A-I A-II as well as Lp-A-I without apoA-II was significantly shifted towards smaller particles in the size range of 7.0–8.2 nm.[77] Lastly, we have recently showed that proteome of the HDL3, the smaller, denser HDL particles, is significantly altered in the subjects with stable coronary artery disease (CAD). In the HDL3 of 7 subjects with stable CAD several proteins including apoE, complement C3, PON1, apoA-IV and apoC-IV were significantly enriched compared to control subjects.[56] We then confirmed the elevated levels of apoE in an independent cohort of 32 subjects with CAD and 32 matched controls. Significantly, elevation of apoE in HDL with CAD was also independently observed in the CARE study.[78] In a separate study we have investigated HDL2 proteome of people with CAD using MALDI- MS of HDL2 tryptic digests and pattern recognition analysis in a population of 19 CAD subjects and 20 apparently healthy controls. Model built using partial least square discriminate analysis based on MALDI-MS signature of HDL clearly differentiated control from CAD subjects. Furthermore, targeted LC-MS/MS analysis of specific spectral features distinguishing the two classes showed elevated levels of apo(a), apoC-III as well as increased levels of methionine oxidation in apoA-I.[79]

Therapeutic interventions have been used to increase HDL-C levels, however little evidence has shown how these therapies affect HDL composition and its function. Combined statin and niacin therapy provide modest increases in HDL-C. We therefore investigated whether the therapy also alters protein composition of HDL. To address this question we used shotgun proteomics to analyze HDL isolated from 7 subjects with stable CAD before and after 1 year of combined simvastatin and niacin therapy.[80] Strikingly, we found that the therapy partially reversed the changes in HDL associated with cardiovascular disease. Significantly, we observed decrease of apoE content in HDL3 while other proteins previously implicated in cholesterol transport, including phospholipid transfer protein (PLTP), apoF and apoJ, were significantly enriched after therapy. While the elevation of HDL-C after the combined therapy is likely due to the known effects of niacin, given the widespread use of statins it would be extremely important to investigate the effects of each drug alone.

Cholesteryl ester transfer protein (CETP), an HDL associated protein, plays critical role in remodeling of HDL by mediating trafficking of cholesterol ester from HDL to LDL. CETP deficiency results in significant increase of HDL-C and CETP inhibitors have been aggressively pursued as a way of HDL-C elevation.[81] Study of HDL in people with CETP deficiency demonstrated that this HDL has improved ability to efflux cholesterol from lipid loaded macrophages.[82] These findings were also extended to people treated with CETP inhibitor torcetrapib.[83] Although torcetrapib failed in clinical trials,[28] likely due to off-target effects,[84] CETP inhibitors are actively pursued and latest results for dalcetrapib and anacetrapib showed significant increases of HDL-C accompanied by improvements of reverse cholesterol transport.[85] However, the understanding the mechanism of the effects of CETP inhibition on HDL function and its protein composition are currently limited.

Collectively, the observations of changes in the HDL proteome associated with cardiovascular disease and the positive effects of lipid lowering therapy raise the possibility that the analysis of HDL proteome may serve both as a surrogate measure of HDL function as well as a diagnostic tool and a measurement of treatment efficacy.

FUTURE DIRECTIONS IN HDL PROTEOMICS

Although immense body of data exists on HDL isolated by ultracentrifugation conclusive evaluation of the relevance of the isolated HDL to the HDL as it exists in the circulation remains one of the important areas of HDL research. While many HDL functional assays can be easily adapted to high throughput format, the HDL isolation represents a significant bottleneck for performing large scale clinical studies which would be required to establish HDL protein composition–function relationship in humans. Immunoaffinity isolation offers an attractive alternative to ultracentrifugation which can be adapted to 96-well format for parallel processing in high-throughput format. However, its relevance to plasma HDL remains to be addressed.

Large scale structure-function studies will also require major advances in the protein analysis. So far majority of HDL proteomics studies focused on qualitative description of HDL proteome and only limited number of studies focused on protein quantitation. Heller et all used protein identification index based on peptide identification scores to quantify several major HDL proteins is normolipidemic and hyperlipidemic subjects and showed that in hyperlipidemic subjects relative abundance in several of these proteins was altered.[86] We used spectral counting, a method where protein abundance is reflected by the number of instances (MS/MS spectra) a protein presence was detected, to quantify changes of protein abundance in subjects with CAD and in subjects treated with lipid lowering therapy. While both of these techniques are simple and readily available for typical shotgun proteomics experiments, they provide only semi-quantitative information. Moreover, these methods do not work very well for low abundance proteins with only small number of peptides identified and consequently low spectral counts. Thus these methods can only detect large changes with relatively large variability in the data (typical CV more than 25–30%). Recently, proteomics field adapted highly quantitative approach of multiple-reaction monitoring (MRM) used in the past extensively in pharmaceutical industry for quantification of small molecules. Unlike shotgun proteomics MRM-based quantitative methods are targeted and require prior knowledge of the molecules to be quantified, i.e. term targeted proteomics quantification. In targeted MRM proteomics, peptides produced by protease digestion, typically with trypsin, serve as surrogate markers of protein abundance. Targeted proteomic assays offer many advantages over traditional immunoassays used for protein quantification in biological samples, namely MRM-based methods can be rapidly developed and validated.[87] Furthermore, such assays are readily multiplexed for quantification of many proteins (>50) in a single analysis over wide range of relative concentrations without mutual interference often found in multiplexed immunoassays.[88] Furthermore, when combined with isotope dilution using stable isotope-labeled peptides as internal standards, MRM has been established as the most promising approach to precise relative protein quantification.[8992] Thus targeted MRM protein quantification could provide accurate determination of essentially all proteins identified in HDL so far in a single analysis with a throughput compatible with hundreds of samples needed for analysis of samples from clinical trials. When coupled with a high-throughput HDL isolation and HDL functional assays this approach could be a powerful tool for understanding the role of HDL as mediator and/or a biomarker of cardiovascular disease and a measure of efficacy of therapeutic interventions.

ACKNOWLEDGMENTS

We thank Dr. Philip Mayer for careful literature review and collection of the list of proteins identified in HDL. Dr. Tomas Vaisar was supported by a Scientist Development Grant Award from the American Heart Association (0830231N), and by a Pilot and Feasibility Award from the University of Washington Nutrition and Obesity Research Center (NIH 5P30 DK035816), and awards from National Institutes of Health (R01 HL089504, and R44 HL083578).

REFERENCES

  • 1.Gordon DJ, Rifkind BM. High-density lipoprotein--the clinical implications of recent studies. N Engl J Med. 1989 Nov 9;321(19):1311–1316. doi: 10.1056/NEJM198911093211907. [DOI] [PubMed] [Google Scholar]
  • 2.Movva R, Rader DJ. Laboratory assessment of HDL heterogeneity and function. Clin Chem. 2008 May;54(5):788–800. doi: 10.1373/clinchem.2007.101923. [DOI] [PubMed] [Google Scholar]
  • 3.Berliner JA, Navab M, Fogelman AM, Frank JS, Demer LL, Edwards PA, et al. Atherosclerosis: basic mechanisms Oxidation, inflammation, and genetics. Circulation. 1995 May 1;91(9):2488–2496. doi: 10.1161/01.cir.91.9.2488. [DOI] [PubMed] [Google Scholar]
  • 4.Aiello RJ, Brees D, Francone OL. ABCA1-deficient mice: insights into the role of monocyte lipid efflux in HDL formation and inflammation. Arterioscler Thromb Vasc Biol. 2003 Jun 1;23(6):972–980. doi: 10.1161/01.ATV.0000054661.21499.FB. [DOI] [PubMed] [Google Scholar]
  • 5.Oram JF. HDL apolipoproteins and ABCA1: partners in the removal of excess cellular cholesterol. Arterioscler Thromb Vasc Biol. 2003 May 1;23(5):720–727. doi: 10.1161/01.ATV.0000054662.44688.9A. [DOI] [PubMed] [Google Scholar]
  • 6.Singaraja RR, Fievet C, Castro G, James ER, Hennuyer N, Clee SM, et al. Increased ABCA1 activity protects against atherosclerosis. J Clin Invest. 2002 Jul;110(1):35–42. doi: 10.1172/JCI15748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Vaughan AM, Oram JF. ABCA1 and ABCG1 or ABCG4 act sequentially to remove cellular cholesterol and generate cholesterol-rich HDL. J Lipid Res. 2006 Nov;47(11):2433–2443. doi: 10.1194/jlr.M600218-JLR200. [DOI] [PubMed] [Google Scholar]
  • 8.Vaughan AM, Oram JF. ABCA1 redistributes membrane cholesterol independent of apolipoprotein interactions. J Lipid Res. 2003 Jul;44(7):1373–1380. doi: 10.1194/jlr.M300078-JLR200. [DOI] [PubMed] [Google Scholar]
  • 9.Mendez AJ, Anantharamaiah GM, Segrest JP, Oram JF. Synthetic amphipathic helical peptides that mimic apolipoprotein A-I in clearing cellular cholesterol. J Clin Invest. 1994 Oct;94(4):1698–1705. doi: 10.1172/JCI117515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Wang Y, Oram JF. Unsaturated fatty acids inhibit cholesterol efflux from macrophages by increasing degradation of ATP-binding cassette transporter A1. J Biol Chem. 2002 Feb 15;277(7):5692–5697. doi: 10.1074/jbc.M109977200. [DOI] [PubMed] [Google Scholar]
  • 11.Barter PJ, Nicholls S, Rye KA, Anantharamaiah GM, Navab M, Fogelman AM. Antiinflammatory properties of HDL. Circ Res. 2004 Oct 15;95(8):764–772. doi: 10.1161/01.RES.0000146094.59640.13. [DOI] [PubMed] [Google Scholar]
  • 12.Spieker LE, Sudano I, Hurlimann D, Lerch PG, Lang MG, Binggeli C, et al. High-density lipoprotein restores endothelial function in hypercholesterolemic men. Circulation. 2002 Mar 26;105(12):1399–1402. doi: 10.1161/01.cir.0000013424.28206.8f. [DOI] [PubMed] [Google Scholar]
  • 13.Mackness M, Boullier A, Hennuyer N, Mackness B, Hall M, Tailleux A, et al. Paraoxonase activity is reduced by a pro-atherosclerotic diet in rabbits. Biochem Biophys Res Commun. 2000 Mar 5;269(1):232–236. doi: 10.1006/bbrc.2000.2265. [DOI] [PubMed] [Google Scholar]
  • 14.Durrington PN, Mackness B, Mackness MI. Paraoxonase and atherosclerosis. Arterioscler Thromb Vasc Biol. 2001 Apr;21(4):473–480. doi: 10.1161/01.atv.21.4.473. [DOI] [PubMed] [Google Scholar]
  • 15.Warden CH, Hedrick CC, Qiao JH, Castellani LW, Lusis AJ. Atherosclerosis in transgenic mice overexpressing apolipoprotein A-II. Science. 1993 Jul 23;261(5120):5469–5472. doi: 10.1126/science.8332912. [DOI] [PubMed] [Google Scholar]
  • 16.Braun A, Trigatti BL, Post MJ, Sato K, Simons M, Edelberg JM, et al. Loss of SR-BI expression leads to the early onset of occlusive atherosclerotic coronary artery disease, spontaneous myocardial infarctions, severe cardiac dysfunction, and premature death in apolipoprotein E-deficient mice. Circ Res. 2002 Feb 22;90(3):270–276. doi: 10.1161/hh0302.104462. [DOI] [PubMed] [Google Scholar]
  • 17.Tall AR, Costet P, Wang N. Regulation and mechanisms of macrophage cholesterol efflux. J Clin Invest. 2002 Oct;110(7):899–904. doi: 10.1172/JCI16391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Covey SD, Krieger M, Wang W, Penman M, Trigatti BL. Scavenger receptor class B type I-mediated protection against atherosclerosis in LDL receptor-negative mice involves its expression in bone marrow-derived cells. Arterioscler Thromb Vasc Biol. 2003 Sep 1;23(9):1589–1594. doi: 10.1161/01.ATV.0000083343.19940.A0. [DOI] [PubMed] [Google Scholar]
  • 19.Khera AV, Cuchel M, de la Llera-Moya M, Rodrigues A, Burke MF, Jafri K, et al. Cholesterol efflux capacity, high-density lipoprotein function, and atherosclerosis. N Engl J Med. 2011 Jan 13;364(2):127–135. doi: 10.1056/NEJMoa1001689. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Vaisar T, Shao B, Green PS, Oda MN, Oram JF, Heinecke JW. Myeloperoxidase and inflammatory proteins: pathways for generating dysfunctional high-density lipoprotein in humans. Curr Atheroscler Rep. 2007 Nov;9(5):417–424. doi: 10.1007/s11883-007-0054-z. [DOI] [PubMed] [Google Scholar]
  • 21.Bergt C, Pennathur S, Fu X, Byun J, O'Brien K, McDonald TO, et al. The myeloperoxidase product hypochlorous acid oxidizes HDL in the human artery wall and impairs ABCA1-dependent cholesterol transport. Proc Natl Acad Sci U S A. 2004 Aug 31;101(35):13032–13037. doi: 10.1073/pnas.0405292101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Shao B, Fu X, McDonald TO, Green PS, Uchida K, O'Brien KD, et al. Acrolein impairs ATP binding cassette transporter A1-dependent cholesterol export from cells through site-specific modification of apolipoprotein A-I. J Biol Chem. 2005 Oct 28;280(43):36386–36396. doi: 10.1074/jbc.M508169200. [DOI] [PubMed] [Google Scholar]
  • 23.Panzenböck U, Stocker R. Formation of methionine sulfoxide-containing specific forms of oxidized high-density lipoproteins. Biochimica et Biophysica Acta (BBA) - Proteins & Proteomics. 2005;1703(2):171–181. doi: 10.1016/j.bbapap.2004.11.003. [DOI] [PubMed] [Google Scholar]
  • 24.Zheng L, Nukuna B, Brennan ML, Sun M, Goormastic M, Settle M, et al. Apolipoprotein A-I is a selective target for myeloperoxidase-catalyzed oxidation and functional impairment in subjects with cardiovascular disease. J Clin Invest. 2004 Aug;114(4):529–541. doi: 10.1172/JCI21109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Francis GA. High density lipoprotein oxidation: in vitro susceptibility and potential in vivo consequences. Biochim Biophys Acta. 2000 Jan 17;1483(2):217–235. doi: 10.1016/s1388-1981(99)00181-x. [DOI] [PubMed] [Google Scholar]
  • 26.Frikke-Schmidt R, Nordestgaard BG, Stene MC, Sethi AA, Remaley AT, Schnohr P, et al. Association of loss-of-function mutations in the ABCA1 gene with high-density lipoprotein cholesterol levels and risk of ischemic heart disease. JAMA. 2008 Jun 4;299(21):2524–2532. doi: 10.1001/jama.299.21.2524. [DOI] [PubMed] [Google Scholar]
  • 27.Duffy D, Rader DJ. Update on strategies to increase HDL quantity and function. Nat Rev Cardiol. 2009 Jul;6(7):455–463. doi: 10.1038/nrcardio.2009.94. [DOI] [PubMed] [Google Scholar]
  • 28.Barter PJ, Caulfield M, Eriksson M, Grundy SM, Kastelein JJ, Komajda M, et al. Effects of torcetrapib in patients at high risk for coronary events. N Engl J Med. 2007 Nov 22;357(21):2109–2122. doi: 10.1056/NEJMoa0706628. [DOI] [PubMed] [Google Scholar]
  • 29.Lund-Katz S, Liu L, Thuahnai ST, Phillips MC. High density lipoprotein structure. Front Biosci. 2003 May;1:8, d1044–d1054. doi: 10.2741/1077. [DOI] [PubMed] [Google Scholar]
  • 30.Rosenson RS, Brewer HB, Jr., Chapman MJ, Fazio S, Hussain MM, Kontush A, et al. HDL measures, particle heterogeneity, proposed nomenclature, and relation to atherosclerotic cardiovascular events. Clin Chem. 2011 Mar;57(3):392–410. doi: 10.1373/clinchem.2010.155333. [DOI] [PubMed] [Google Scholar]
  • 31.Bachorik PS, Albers JJ. Precipitation methods for quantification of lipoproteins. Methods Enzymol. 1986;129:78–100. doi: 10.1016/0076-6879(86)29063-1. [DOI] [PubMed] [Google Scholar]
  • 32.Warnick GR, Albers JJ, Bachorik PS, Turner JD, Garcia C, Breckinridge C, et al. Multilaboratory evaluation of an ultrafiltration procedure for high density lipoprotein cholesterol quantification in turbid heparin-manganese supernates. J Lipid Res. 1981 Aug;22(6):1015–1020. [PubMed] [Google Scholar]
  • 33.Havel RJ, Eder HA, Bragdon JH. The distribution and chemical composition of ultracentrifugally separated lipoproteins in human serum. J Clin Invest. 1955 Sep;34(9):1345–1353. doi: 10.1172/JCI103182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Alaupovic P. The concept of apolipoprotein-defined lipoprotein families and its clinical significance. Curr Atheroscler Rep. 2003 Nov;5(6):459–467. doi: 10.1007/s11883-003-0036-8. [DOI] [PubMed] [Google Scholar]
  • 35.Alaupovic P. Significance of apolipoproteins for structure, function, and classification of plasma lipoproteins. Methods Enzymol. 1996;263:32–60. doi: 10.1016/s0076-6879(96)63004-3. [DOI] [PubMed] [Google Scholar]
  • 36.Alaupovic P, Kostner G, Lee DM, McConathy WJ, Magnani HN. Peptide composition of human plasma apolipoproteins A, B and C. Expos Annu Biochim Med. 1972;31:145–160. [PubMed] [Google Scholar]
  • 37.Cheung MC, Albers JJ. The measurement of apolipoprotein A-I and A-II levels in men and women by immunoassay. J Clin Invest. 1977 Jul;60(1):43–50. doi: 10.1172/JCI108767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Schonfeld G, Pfleger B. The structure of human high density lipoprotein and the levels of apolipoprotein A-I in plasma as determined by radioimmunoassay. J Clin Invest. 1974 Aug;54(2):236–246. doi: 10.1172/JCI107758. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Ordovas JM, Osgood D. Preparative isolation of plasma lipoproteins using fast protein liquid chromatography (FPLC) Methods Mol Biol. 1998;110:105–111. doi: 10.1385/1-59259-582-0:105. [DOI] [PubMed] [Google Scholar]
  • 40.Stocks J, Miller NE. Analysis of apolipoproteins and lipoproteins by capillary electrophoresis. Electrophoresis. 1999 Jul;20(10):2118–2123. doi: 10.1002/(SICI)1522-2683(19990701)20:10<2118::AID-ELPS2118>3.0.CO;2-0. [DOI] [PubMed] [Google Scholar]
  • 41.Schmitz G, Borgmann U, Assmann G. Analytical capillary isotachophoresis: a routine technique for the analysis of lipoproteins and lipoprotein subfractions in whole serum. J Chromatogr. 1985 Feb 22;320(1):253–262. doi: 10.1016/s0021-9673(01)90503-5. [DOI] [PubMed] [Google Scholar]
  • 42.Bottcher A, Schlosser J, Kronenberg F, Dieplinger H, Knipping G, Lackner KJ, et al. Preparative free-solution isotachophoresis for separation of human plasma lipoproteins: apolipoprotein and lipid composition of HDL subfractions. J Lipid Res. 2000 Jun;41(6):905–915. [PubMed] [Google Scholar]
  • 43.Karas M, Hillenkamp F. Laser desorption ionization of proteins with molecular masses exceeding 10,000 daltons. Anal Chem. 1988 Oct 15;60(20):2299–2301. doi: 10.1021/ac00171a028. [DOI] [PubMed] [Google Scholar]
  • 44.Fenn JB, Mann M, Meng CK, Wong SF, Whitehouse CM. Electrospray ionization for mass spectrometry of large biomolecules. Science. 1989 Oct 6;246(4926):64–71. doi: 10.1126/science.2675315. [DOI] [PubMed] [Google Scholar]
  • 45.Hoofnagle AN, Heinecke JW. Lipoproteomics: using mass spectrometry-based proteomics to explore the assembly, structure, and function of lipoproteins. J Lipid Res. 2009 Oct;50(10):1967–1675. doi: 10.1194/jlr.R900015-JLR200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Albers JJ, Marcovina SM. Standardization of Lp(a) measurements. Chem Phys Lipids. 1994 Jan;67–68:257–263. doi: 10.1016/0009-3084(94)90145-7. [DOI] [PubMed] [Google Scholar]
  • 47.Albers JJ, Kennedy H, Marcovina SM. Evidence that Lp[a] contains one molecule of apo[a] and one molecule of apoB: evaluation of amino acid analysis data. J Lipid Res. 1996 Jan;37(1):192–196. [PubMed] [Google Scholar]
  • 48.Kunitake ST, Kane JP. Factors affecting the integrity of high density lipoproteins in the ultracentrifuge. J Lipid Res. 1982 Aug;23(6):936–940. [PubMed] [Google Scholar]
  • 49.Stahlman M, Davidsson P, Kanmert I, Rosengren B, Boren J, Fagerberg B, et al. Proteomics and lipids of lipoproteins isolated at low salt concentrations in D2O/sucrose or in KBr. J Lipid Res. 2008 Feb;49(2):481–490. doi: 10.1194/jlr.D700025-JLR200. [DOI] [PubMed] [Google Scholar]
  • 50.Cheung MC, Vaisar T, Han X, Heinecke JW, Albers JJ. Phospholipid transfer protein in human plasma associates with proteins linked to immunity and inflammation. Biochemistry. 2010 Aug 31;49(34):7314–7322. doi: 10.1021/bi100359f. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Collins LA, Mirza SP, Kissebah AH, Olivier M. Integrated approach for the comprehensive characterization of lipoproteins from human plasma using FPLC and nano-HPLC-tandem mass spectrometry. Physiol Genomics. 2010 Feb 4;40(3):208–215. doi: 10.1152/physiolgenomics.00136.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Collins LA, Olivier M. Quantitative comparison of lipoprotein fractions derived from human plasma and serum by liquid chromatography-tandem mass spectrometry. Proteome Sci. 2010;8:42. doi: 10.1186/1477-5956-8-42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Gordon SM, Deng J, Lu LJ, Davidson WS. Proteomic characterization of human plasma high density lipoprotein fractionated by gel filtration chromatography. J Proteome Res. 2010 Oct 1;9(10):5239–5249. doi: 10.1021/pr100520x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Karlsson H, Leanderson P, Tagesson C, Lindahl M. Lipoproteomics II: mapping of proteins in high-density lipoprotein using two-dimensional gel electrophoresis and mass spectrometry. Proteomics. 2005 Apr;5(5):1431–1445. doi: 10.1002/pmic.200401010. [DOI] [PubMed] [Google Scholar]
  • 55.Rezaee F, Casetta B, Levels JH, Speijer D, Meijers JC. Proteomic analysis of high-density lipoprotein. Proteomics. 2006 Jan;6(2):721–730. doi: 10.1002/pmic.200500191. [DOI] [PubMed] [Google Scholar]
  • 56.Vaisar T, Pennathur S, Green PS, Gharib SA, Hoofnagle AN, Cheung MC, et al. Shotgun proteomics implicates protease inhibition and complement activation in the antiinflammatory properties of HDL. J Clin Invest. 2007 Mar;117(3):746–756. doi: 10.1172/JCI26206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Levels JH, Bleijlevens B, Rezaee F, Aerts JM, Meijers JC. SELDI-TOF mass spectrometry of High-Density Lipoprotein. Proteome Sci. 2007;5:15. doi: 10.1186/1477-5956-5-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Mazur MT, Cardasis HL, Spellman DS, Liaw A, Yates NA, Hendrickson RC. Quantitative analysis of intact apolipoproteins in human HDL by top-down differential mass spectrometry. Proc Natl Acad Sci U S A. 2010 Apr 27;107(17):7728–7733. doi: 10.1073/pnas.0910776107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Heller M, Stalder D, Schlappritzi E, Hayn G, Matter U, Haeberli A. Mass spectrometry-based analytical tools for the molecular protein characterization of human plasma lipoproteins. Proteomics. 2005 Jul;5(10):2619–2630. doi: 10.1002/pmic.200401233. [DOI] [PubMed] [Google Scholar]
  • 60.Taylor CF, Binz PA, Aebersold R, Affolter M, Barkovich R, Deutsch EW, et al. Guidelines for reporting the use of mass spectrometry in proteomics. Nat Biotechnol. 2008 Aug;26(8):860–861. doi: 10.1038/nbt0808-860. [DOI] [PubMed] [Google Scholar]
  • 61.Tabb DL. What's driving false discovery rates? J Proteome Res. 2008 Jan;7(1):45–46. doi: 10.1021/pr700728t. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Chen N, Liu Y, Greiner CD, Holtzman JL. Physiologic concentrations of homocysteine inhibit the human plasma GSH peroxidase that reduces organic hydroperoxides. J Lab Clin Med. 2000 Jul;136(1):58–65. doi: 10.1067/mlc.2000.107692. [DOI] [PubMed] [Google Scholar]
  • 63.Hortin GL, Shen RF, Martin BM, Remaley AT. Diverse range of small peptides associated with high-density lipoprotein. Biochem Biophys Res Commun. 2006 Feb 17;340(3):909–915. doi: 10.1016/j.bbrc.2005.12.098. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Stafforini DM, McIntyre TM, Carter ME, Prescott SM. Human plasma platelet-activating factor acetylhydrolase Association with lipoprotein particles and role in the degradation of platelet-activating factor. J Biol Chem. 1987 Mar 25;262(9):4215–4222. [PubMed] [Google Scholar]
  • 65.Watanabe J, Grijalva V, Hama S, Barbour K, Berger F, Navab M, et al. Hemoglobin and its scavenger protein haptoglobin associate with apoA-1 containing particles and influence the inflammatory properties and function of high density lipoprotein. J Biol Chem. 2009 May 11; doi: 10.1074/jbc.M109.017202. 2009:M109.017202. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.O'Brien PJ, Alborn WE, Sloan JH, Ulmer M, Boodhoo A, Knierman MD, et al. The novel apolipoprotein A5 is present in human serum, is associated with VLDL, HDL, and chylomicrons, and circulates at very low concentrations compared with other apolipoproteins. Clin Chem. 2005 Feb;51(2):351–359. doi: 10.1373/clinchem.2004.040824. [DOI] [PubMed] [Google Scholar]
  • 67.Asztalos BF, Sloop CH, Wong L, Roheim PS. Two-dimensional electrophoresis of plasma lipoproteins: recognition of new apo A-I-containing subpopulations. Biochim Biophys Acta. 1993 Sep 8;1169(3):291–300. doi: 10.1016/0005-2760(93)90253-6. [DOI] [PubMed] [Google Scholar]
  • 68.Cheung MC, Albers JJ. Distribution of high density lipoprotein particles with different apoprotein composition: particles with A-I and A-II and particles with A-I but no A-II. J Lipid Res. 1982 Jul;23(5):747–753. [PubMed] [Google Scholar]
  • 69.Shiflett AM, Bishop JR, Pahwa A, Hajduk SL. Human high density lipoproteins are platforms for the assembly of multi-component innate immune complexes. J Biol Chem. 2005 Sep 23;280(38):32578–32585. doi: 10.1074/jbc.M503510200. [DOI] [PubMed] [Google Scholar]
  • 70.Vanhamme L, Paturiaux-Hanocq F, Poelvoorde P, Nolan DP, Lins L, Van Den Abbeele J, et al. Apolipoprotein L-I is the trypanosome lytic factor of human serum. Nature. 2003 Mar 6;422(6927):83–87. doi: 10.1038/nature01461. [DOI] [PubMed] [Google Scholar]
  • 71.Tall AR, Forester LR, Bongiovanni GL. Facilitation of phosphatidylcholine transfer into high density lipoproteins by an apolipoprotein in the density 1.20–1.26 g/ml fraction of plasma. J Lipid Res. 1983 Mar 1;24(3):277–289. 1983. [PubMed] [Google Scholar]
  • 72.Davidson WS, Silva RA, Chantepie S, Lagor WR, Chapman MJ, Kontush A. Proteomic analysis of defined HDL subpopulations reveals particle-specific protein clusters: relevance to antioxidative function. Arterioscler Thromb Vasc Biol. 2009 Jun;29(6):870–876. doi: 10.1161/ATVBAHA.109.186031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.McVicar JP, Kunitake ST, Hamilton RL, Kane JP. Characteristics of human lipoproteins isolated by selected-affinity immunosorption of apolipoprotein A-I. Proc Natl Acad Sci U S A. 1984 Mar;81(5):1356–1360. doi: 10.1073/pnas.81.5.1356. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Kunitake ST, Carilli CT, Lau K, Protter AA, Naya-Vigne J, Kane JP. Identification of proteins associated with apolipoprotein A-I-containing lipoproteins purified by selected-affinity immunosorption. Biochemistry. 1994 Mar 1;33(8):1988–1993. doi: 10.1021/bi00174a003. [DOI] [PubMed] [Google Scholar]
  • 75.de la Llera-Moya M, Drazul-Schrader D, Asztalos BF, Cuchel M, Rader DJ, Rothblat GH. The ability to promote efflux via ABCA1 determines the capacity of serum specimens with similar high-density lipoprotein cholesterol to remove cholesterol from macrophages. Arterioscler Thromb Vasc Biol. 2010 Apr;30(4):796–801. doi: 10.1161/ATVBAHA.109.199158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Asztalos BF, Roheim PS, Milani RL, Lefevre M, McNamara JR, Horvath KV, et al. Distribution of ApoA-I-containing HDL subpopulations in patients with coronary heart disease. Arterioscler Thromb Vasc Biol. 2000 Dec;20(12):2670–2676. doi: 10.1161/01.atv.20.12.2670. [DOI] [PubMed] [Google Scholar]
  • 77.Cheung MC, Brown BG, Wolf AC, Albers JJ. Altered particle size distribution of apolipoprotein A-I-containing lipoproteins in subjects with coronary artery disease. J Lipid Res. 1991 Mar;32(3):383–394. [PubMed] [Google Scholar]
  • 78.Sacks FM, Alaupovic P, Moye LA, Cole TG, Sussex B, Stampfer MJ, et al. VLDL, apolipoproteins B, CIII, and E, and risk of recurrent coronary events in the Cholesterol and Recurrent Events (CARE) trial. Circulation. 2000 Oct 17;102(16):1886–1892. doi: 10.1161/01.cir.102.16.1886. [DOI] [PubMed] [Google Scholar]
  • 79.Vaisar T, Mayer P, Nilsson E, Zhao XQ, Knopp R, Prazen BJ. HDL in humans with cardiovascular disease exhibits a proteomic signature. Clin Chim Acta. 2010 Jul 4;411(13–14):972–979. doi: 10.1016/j.cca.2010.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Green PS, Vaisar T, Pennathur S, Kulstad JJ, Moore AB, Marcovina S, et al. Combined Statin and Niacin Therapy Remodels the High-Density Lipoprotein Proteome. Circulation. 2008 Sep 16;118(12):1259–1267. doi: 10.1161/CIRCULATIONAHA.108.770669. 2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Tall AR. CETP inhibitors to increase HDL cholesterol levels. N Engl J Med. 2007 Mar 29;356(13):1364–1366. doi: 10.1056/NEJMe078029. [DOI] [PubMed] [Google Scholar]
  • 82.Matsuura F, Wang N, Chen W, Jiang XC, Tall AR. HDL from CETP-deficient subjects shows enhanced ability to promote cholesterol efflux from macrophages in an apoE- and ABCG1-dependent pathway. J Clin Invest. 2006 May;116(5):1435–1442. doi: 10.1172/JCI27602. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Yvan-Charvet L, Matsuura F, Wang N, Bamberger MJ, Nguyen T, Rinninger F, et al. Inhibition of cholesteryl ester transfer protein by torcetrapib modestly increases macrophage cholesterol efflux to HDL. Arterioscler Thromb Vasc Biol. 2007 May;27(5):1132–1138. doi: 10.1161/ATVBAHA.106.138347. [DOI] [PubMed] [Google Scholar]
  • 84.Tall AR, Yvan-Charvet L, Wang N. The failure of torcetrapib: was it the molecule or the mechanism? Arterioscler Thromb Vasc Biol. 2007 Feb;27(2):257–260. doi: 10.1161/01.ATV.0000256728.60226.77. [DOI] [PubMed] [Google Scholar]
  • 85.Niesor EJ, Magg C, Ogawa N, Okamoto H, von der Mark E, Matile H, et al. Modulating cholesteryl ester transfer protein activity maintains efficient pre-beta-HDL formation and increases reverse cholesterol transport. J Lipid Res. 2010 Dec;51(12):3443–3454. doi: 10.1194/jlr.M008706. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Heller M, Schlappritzi E, Stalder D, Nuoffer JM, Haeberli A. Compositional protein analysis of high density lipoproteins in hypercholesterolemia by shotgun LC-MS/MS and probabilistic peptide scoring. Mol Cell Proteomics. 2007 Jun;6(6):1059–1072. doi: 10.1074/mcp.M600326-MCP200. [DOI] [PubMed] [Google Scholar]
  • 87.Paulovich AG, Whiteaker JR, Hoofnagle AN, Wang P. The interface between biomarker discovery and clinical validation: The tar pit of the protein biomarker pipeline. Proteomics Clin Appl. 2008 Oct 1;2(10–11):1386–1402. doi: 10.1002/prca.200780174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Anderson NL, Anderson NG, Pearson TW, Borchers CH, Paulovich AG, Patterson SD, et al. A human proteome detection and quantitation project. Mol Cell Proteomics. 2009 May;8(5):883–886. doi: 10.1074/mcp.R800015-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Anderson L, Hunter CL. Quantitative mass spectrometric multiple reaction monitoring assays for major plasma proteins. Mol Cell Proteomics. 2006 Apr;5(4):573–588. doi: 10.1074/mcp.M500331-MCP200. [DOI] [PubMed] [Google Scholar]
  • 90.Gerber SA, Rush J, Stemman O, Kirschner MW, Gygi SP. Absolute quantification of proteins and phosphoproteins from cell lysates by tandem MS. Proc Natl Acad Sci U S A. 2003 Jun 10;100(12):6940–6945. doi: 10.1073/pnas.0832254100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Ong SE, Mann M. Mass spectrometry-based proteomics turns quantitative. Nat Chem Biol. 2005 Oct;1(5):252–262. doi: 10.1038/nchembio736. [DOI] [PubMed] [Google Scholar]
  • 92.Whiteaker JR, Zhao L, Anderson L, Paulovich AG. An automated and multiplexed method for high throughput peptide immunoaffinity enrichment and multiple reaction monitoring mass spectrometry-based quantification of protein biomarkers. Mol Cell Proteomics. 2010 Jan;9(1):184–196. doi: 10.1074/mcp.M900254-MCP200. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES