重生之星火燎原:ScienceDirect 7

来源:百度文库 编辑:九乡新闻网 时间:2024/05/03 04:58:59


Hub
Scopus
SciTopics
Applications
Register
Login
Go to SciVal Suite
Username:
Password:
Remember me
|Not Registered?
Forgotten your username or password?
Go to Athens / Institution login

Home
Browse
Search
My settings
My alerts
Help
Articles     All fields     AuthorAdvanced search
Images     Journal/Book title     Volume   Issue   PageSearch tips

',1)">
Supplementary content for this Article


Download this File View within Article 1
Related Articles
Quantitation of lysophosphatidylcholine molecular speci...Analytical Biochemistry
Characterisation of the glycerophospholipid fraction in...Food Chemistry
Relative contribution of individual oxidized components...Nutrition, Metabolism and Cardiovascular Diseases
Lipoprotein-Associated Phospholipase A2Clinics in Laboratory Medicine
Lysophospholipase D and its role in LPA productionCellular Signalling
  View more related articles

My Applications

Table Download






View Record in Scopus

PDF (501 K)
Export citation
E-mail article
Highlight keywords on
Thumbnails - selected |Full-Size images
Thumbnails - selected |Full-Size images
Article
Article - selected
Figures/Tables (6)
Figures/Tables (6)- selected
References (144)
References (144)- selected


Atherosclerosis
Volume 208, Issue 1, January 2010, Pages 10-18
doi:10.1016/j.atherosclerosis.2009.05.029 | How to Cite or Link Using DOI
Permissions & Reprints
Review
Metabolism and atherogenic disease association of lysophosphatidylcholine
Gerd Schmitz,a, and Katharina Ruebsaamena
a Institute for Clinical Chemistry and Laboratory Medicine, University of Regensburg, D-93042 Regensburg, Germany
Received 6 October 2008;
revised 27 April 2009;
accepted 25 May 2009.
Available online 9 June 2009.
Abstract
Lysophosphatidylcholine (LPC) is a major plasma lipid that has been recognized as an important cell signalling molecule produced under physiological conditions by the action of phospholipase A2 on phosphatidylcholine. LPC transports glycerophospholipid components such as fatty acids, phosphatidylglycerol and choline between tissues. LPC is a ligand for specific G protein-coupled signalling receptors and activates several second messengers. LPC is also a major phospholipid component of oxidized low-density lipoproteins (Ox-LDL) and is implicated as a critical factor in the atherogenic activity of Ox-LDL. Hence, LPC plays an important role in atherosclerosis and acute and chronic inflammation.
In this review we focus in some detail on LPC function, biochemical pathways, sources and signal-transduction system. Moreover, we outline the detection of LPC by mass spectrometry which is currently the best method for accurate and simultaneous analysis of each individual LPC species and reveal the pathophysiological implication of LPC which makes it an interesting target for biomarker and drug development regarding atherosclerosis and cardiovascular disorders.
Keywords: Lysophospholipids; Lipidomics; Lipid species; Signalling; G protein-coupled receptor; Phospholipase
Abbreviations: AA, arachidonic acid; AC, adenylate cyclase; cAMP, cyclic adenosine monophosphate; ESI, electrospray ionization; GPR, G protein-coupled receptor; LC, liquid chromatography; LCAT, lecithin-cholesterol acyltransferase; Lp(a), lipoprotein (a); LPA, lysophosphatidic acid; LPC, lysophosphatidylcholine; LPE, lysophosphatidylethanolamine; LPG, lysophosphatidylglycerol; LPI, lysophosphatidylinositol; LPS, lysophosphatidylserine; Lp-PLA2, lipoprotein-associated phospholipase A2; MAPK, mitogen-activated protein kinase; MS, mass spectrometry; Ox-LDL, oxidized low-density lipoprotein; PA, phosphatidic acid; PAF, platelet activating factor; PC, phosphatidylcholine; PLA1/2, phospholipase A1/2; PLD, phospholipase D; S1P, spingosine-1-phosphate; SPC, sphingosylphosphorylcholine
Article Outline
1.Lysophospholipids
2.Sources of LPC
2.1.Biochemical pathways 2.2.Occurrence of LPC
3.LPC binding to G protein-coupled receptors
4.LPC signalling and cell specific effects
5.Atherogenic disease association of LPC and pharmacological aspects
5.1.Secretory phospholipases A2 (sPLA2) 5.2.Lipoprotein-associated phospholipase A2 (Lp-PLA2) 5.3.Phospholipase A2 inhibitors
6.Determination of relevant LPC species
7.Future pathophysiological and molecular perspectives towards LPC based biomarker and drug target development
Acknowledgements
Appendix A.Supplementary data
References
1. Lysophospholipids
This phospholipid group includes glycerol- and sphingoid-based lipids which contain just one carbon chain: one fatty acid (in lysoglycerophospholipids) or the long chain of sphingosine alcohol (in lysosphingolipids). The different lysophospholipids are displayed inTable 1. Among them lysophosphatidic acid (LPA) and spingosine-1-phosphate (S1P) are probably the best characterized lysophospholipids and multiple G protein-coupled receptors (GPRs) have been identified so far with varying ligand specificities[1]. The characteristics of the currently identified lysophospholipid GPR are listed inSuppl. Table 1. However, additional as yet unidentified receptors may exist for a number of lysophospholipids which will increase this group of GPR (e.g. for LPG and LPE). Lysophospholipids have been found in a wide range of tissues and cell types, indicating their importance in many physiological and pathophysiological processes, including vascular development, reproduction, myelination, neuronal diseases and cancer[2]. Their role as intracellular signalling molecules beside the function of membrane phospholipid metabolites is increasingly appreciated. They regulate a wide variety of cellular activities including proliferation, wound healing, smooth muscle contraction and tumor cell invasiveness[3]. Lysophospholipids activate several second messengers including extracellular-signal-regulated kinases, mitogen-activated protein kinase (MAPK), phosphoinositide-3-kinase, Ca2+, adenylate cyclase (AC) and Rho[2] (Suppl. Table 1). These signalling pathways have been already covered extensively in reviews[4],[5],[6] and[7].
 
Table 1. Chemical structure of the different lysophospholipids. LIPID MAPS ID Short name Full name Structure
LMGP 01050018 LPC Lysophosphatidylcholine
LMGP 10050006 LPA Lysophosphatidic acid
LMGP 02050002 LPE lysophosphatidylethanolamine
LMGP 04050008 LPG Lysophosphatidylglycerol
LMGP 06050002 LPI Lysophosphatidylinositol
LMGP 03050002 LPS Lysophosphatidylserine
LMGP 01060010 Lyso-PAF Lyso-platelet activating factor
LMSP 01060001 SPC Sphingosylphosphorylcholine
LMSP 01050001 S1P Spingosine-1-phosphate
Full-size table
View Within Article
2. Sources of LPC
2.1. Biochemical pathways
The biochemical pathway of LPC is shown inFig. 1 and the relevant LPC enzymes are displayed inTable 2. LPC is derived from polar surface phosphatidylcholine (PC) of lipoproteins or from cell membrane-derived PC as a result of phospholipase A2 (PLA2)[8] and[9]. There is also a phospholipase A1 (PLA1), which is able to cleave the sn-1 ester bond[10]. Appreciable amounts of LPC are either formed in plasma by endothelial lipase[10] or by lecithin-cholesterol acyltransferase (LCAT) secreted from the liver to catalyse the transfer of fatty acids, preferentially linoleate which is in sn-2 position of phosphatidylcholine, to free cholesterol in plasma for the formation of cholesteryl esters[11]. Lysophospholipase substrates the reaction of LPC and water to carboxylate and glycerophosphocholine which is further converted to sn-glycerol-3-phosphate and choline by glycerophosphocholine phosphodiesterase. Glycerol-3-phosphate acyltransferase, located in both endoplasmic reticulum and mitochondria, catalyses the formation of the lysoproduct LPA by the acylation of glycerol 3-phosphate. LPA may also be synthesized by PLA2 catalysed deacylation of phosphatidic acid (PA). PA is generated by the hydrolysis of PC by PC-specific phospholipase D (PLD) or the acylation of LPA by LPA acyl transferases. PA is an important metabolite involved in phospholipid biosynthesis and membrane remodeling. PA appears to function in a manner similar to many other lipid second messengers: by promoting the binding of selected targets to specific regions of the cell membrane. Dephosphorylation of PA by phosphatidic acid phosphatase yields diacylglycerol which can be acylated at the sn-3 position to produce triacylglycerol by diacylglycerol acyltransferase. PA and diacylglycerol are both used as precursors for the major membrane glycerolipids. In addition, diacylglycerol is converted to PC by diacylglycerol-choline phosphotransferase.
Full-size image (75K)
Fig. 1.
Biosynthesis pathway of lysophosphatidylcholine (LPC).
View Within Article
Table 2. LPC relevant phospholipases and lysophospholipases. Enzyme Group Source Localization References
PLA1 Liver/bile/kidney/muscle Cytosolic[102] and[103]
PLA2 I B Pancreas/lung/kidney/spleen/liver/ovary/brain Secretory[68] and[104]
II A Synovial fluid/intestinal mucosa/lacrimal gland Secretory
II D Pancreas/spleen/mast cells Secretory
II E Brain/heart/uterus Secretory
II F Testis/placenta Secretory
III Kidney/heart/liver/skeletal muscle/placenta Secretory
IV A–D Ubiquitous Cytosolic
V Heart/lung/macrophages/neutrophils Secretory
VI A1/A2/B–F Ubiquitous Cytosolic
VII A/B Plasma/liver/kidney/macrophages Secretory
VIII A/B Brain Cytosolic
X Spleen/thymus/lung/pancreas/colon/leukocytes Secretory
XII A Heart/skeletal muscle/kidney/pancreas Secretory
XII B Liver/kidney/heart/skeletal muscle Secretory
XV Ubiquitous Lysosomal
PLC Ubiquitous Cytosolic[27] and[52]
PLD 1/2 Ubiquitous Cytosolic[105]
EL Brain/plasma Secretory[106]
HL Liver/plasma Secretory[107] and[108]
LCAT Plasma Secretory[106]
LYPL A1/A2 Ubiquitous Cytosolic[109]
D Liver/plasma/kidney/glandular gastric mucosa/myocytes/intestinal epithelial cells/testes Secretory[110] and[111]
LPCAT Lung/liver/adipose/pancreas/red blood cells/monocytes Cytosolic[112],[113],[114] and[115]
Full-size table
Abbreviations: phospholipase A1–2/C/D (PLA1/2/C/D), endothelial lipase (EL), hepatic lipase (HL), lysophospholipase (LYPL), lecithin-cholesterol acyltransferase (LCAT), and LPC-acyltransferase (LPCAT).
View Within Article
Direct hepatic secretion is a quantitatively important source of plasma unsaturated LPC[12]. LPC can also be formed when LDL is oxidatively altered in vitro by a number of different mechanisms. These include LDL exposure to different agents such as dissolved oxygen[13], transition metal ions[14], free radical initiators[15], UV radiation[16], specific iron or copper-containing proteins[17], lipoxygenase[18] and myeloperoxidase[19] as well as prolonged storage[20]. Also endothelial cells[14], smooth muscle cells[21] and[22], monocytes/macrophages and neutrophils[18] are capable of oxidizing LDL in vitro. Several cancer cell lines also release significant amounts of LPC into the culture medium which turned out to be a substrate for autotaxin, a tumor cell motility-stimulating factor[23]. The fact that autotaxin and lysophospholipase D are identical suggests that autocrine or paracrine production of LPA contributes to tumor cell motility, survival and proliferation[23].
2.2. Occurrence of LPC
The LPC molecule is wedge-shaped, consisting of one long hydrophobic fatty acyl chain and one large hydrophilic polar choline headgroup, attached to the glycerol backbone (Table 1)[24]. The amphipathic nature of LPC gives it surfactant- and detergent-like properties and LPC may lyse cells at high concentration[25]. Unlike other lipid signalling molecules, such as S1P, SPC and LPA, the physiological concentrations of LPC in body fluids are relatively high[26],[27],[28] and[29]. Independent analytical methods revealed normal blood levels of LPC between 140 and 150 μM[30] and[31], which increase to millimolar levels in hyperlipidemic subjects[32]. LPC, with one mole of fatty acid per mole of lipid in position sn-1, is also found in small amounts in most tissues. The functionally available concentrations of LPC in vivo and the activation of LPC receptors may be controlled by a balance between different physical forms of LPC, free, micellar or bound to LDL and hydrophobic serum proteins such as albumin, anti-phospholipid-immune complexes, or incorporated into plasma membranes[26] and[33]. Unlike LPA and S1P, which are active in the albumin-bound form for many of their biological effects, LPC signalling is strongly influenced by its physical form (free versus bound)[7]. In plasma most of the circulating LPC is likely bound to albumin, representing 5–20% of total phospholipids[26], but some is associated with lipoprotein particles, where it comprises 1–5% of the total PC content of LDL particles[34]. Another minor source of circulating LPC is represented by exosomes (2 nmol in 300 ml blood)[35] as well as microparticles, shedded from cells that are in contact to plasma, which contain about 2% LPC of all phospholipids[36] and[37]. Oxidation and fragmentation of the polyunsaturated sn-2 fatty acyl residues of PC, followed by the hydrolysis of the shortened fatty acyl residues by LDL-associated platelet activating factor (PAF) acetylhydrolase[38], also increase the content of LPC during oxidative modification of LDL that accompanies their conversion to atherogenic particles[39].
3. LPC binding to G protein-coupled receptors
LPC activates GRPs that are widely expressed and regulate distinct cellular functions. LPC activate two receptors of the OGR1 cluster GPR4 and G2A[5] as well as the orphan GPR119[40]. These can be regulated by both, specific lipid agonists and protons. However, the precise molecular mechanisms, by which dual regulation of these receptors takes place, remain to be determined. G2A receptors appear to play a functional role in migration and apoptosis of immune cells (lymphoid tissues, lymphocytes and macrophages)[41] and[42]. Mice in which G2A was genetically deleted suffered from a late-onset autoimmune syndrome that was attributed to insufficient T-cell apoptosis[43]. The GPR4 receptor is widely distributed, e.g. endothelial cells, overexpressed in cancer cells and contribute to osteoclastogenesis and angiogenesis, respectively. GRP4 is also involved in migration and impairment of endothelial barrier function[44] and[45]. GPR119 is predominantly expressed in the pancreas and involved in insulin secretion from β-cells[40]. LPC activates another receptor for phospholipids, the PAF receptor[46] and[47] which is involved in calcium signalling[48] and[49]. However, Marathe et al.[34] could not confirm that LPC is a PAF receptor agonist, therefore LPC may not be a direct ligand of orphan GPR119.Suppl. Table 1 provides a synopsis of the different GPR for LPC.
4. LPC signalling and cell specific effects
Interestingly, there are many indications of specific cell signalling mediated by LPC in accordance with the hypothesis of a specific receptor for LPC. However, because of its amphipathic character it is possible that LPC also induces non-specific cell responses. The biological effects and the signalling properties of LPC have been most extensively studied in vitro in cells related to atherosclerosis (for details see review articles in Refs.[24],[39],[50] and[51]). LPC has been demonstrated to be both atherogenic and antiatherogenic[50]. These effects are outlined inTable 3. One major signalling effect of LPC is the increase of [Ca2+][27] and[28] which is believed to be mediated mainly through PAF receptors[49]. LPC also inhibits or activates MAPK[52] such as p42/44 and p38[53], and activates protein kinase C[51] and PC-specific phospholipase C[27]. In addition LPC affects AC signalling to produce cyclic adenosine monophosphate (cAMP) as an intracellular signalling molecule in the monocytic cell line THP-1[54]. LPC activates a wide range of cell types and events in the vascular system, and therefore understanding its actions is relevant for multiple immunologic and inflammatory events. The cell specific effects are summarized inTable 4.
Table 3. Pro- and antiatherogenic effects of LPC. Proatherogenic Antiatherogenic
Arterial wall and smooth muscle cells Attenuation of cellular uptake of oxidized LDL[132]
Upregulation of adhesive molecules (VCAM-1/ICAM-1) and MCP-1[116],[117] and[118] Increase of extracellular superoxide dismutase expression in monocytes/macrophages[133]
Increase of cellular permeability and apoptosis[119],[120],[121] and[122] Stimulation of HDL-mediated cholesterol efflux from cells[134] and[135]
Release of arachidonic acid[123]
Inhibition of endothelial relaxation[124]
Cell proliferation, migration and overexpression of growth factor gene[125] and[126]
Macrophages
Production of IL-1β[127]
Cell activation[128]
Upregulation of scavenger receptor expression[129]
Increased expression of urokinase-type plasminogen activator and its receptor[130]
Tissue
Arachidonic acid formation leading to eicosanoid generation and signalling[131]
Full-size table
View Within Article
Table 4. Cell specific effects of LPC. Cell type Effect of LPC References
Immune cells Chemotaxis[131] and[136]
Monocytes Cell adhesion[137]
Macrophages Migration[130]
T-lymphocytes Growth[138]
Neutrophils Proinflammatory activity[131]
Lymphoid cells Cytokine synthesis[47] and[139]
Migration[140]
Smooth muscle cells Chemoattraction[126]
Proliferation[39]
Endothelial cells Growth factor expression[116] and[141]
Apoptotic cells Chemotactic attraction of phagocytes[142] and[143]
Pancreatic β-cells Induction of insulin secretion[40] and[144]
Full-size table
View Within Article
5. Atherogenic disease association of LPC and pharmacological aspects
In vivo, LPC is claimed to be an important component of Ox-LDL[55] and[56] which is increased 5-fold in Ox-LDL compared to normal LDL and 7-fold in oxidized lipoprotein (a) (Lp(a)) compared to normal Lp(a). Ox-LDL and oxidized Lp(a) induce apoptosis via oxidative stress which promotes atherogenesis[57]. LPC can be detected in atherosclerotic lesions[58] and[59] and exerts both pro- and anti-atherogenic roles (Table 3), depending on the arterial cell type and oxidation/inflammation status. Due to the elevation of LPC concentrations in atherogenic diseases, various attempts were undertaken to decrease LPC levels which are linked to PLA2 activity.
5.1. Secretory phospholipases A2 (sPLA2)
The sPLA2 family displayed inTable 2 is calcium-dependent and responsible for phospholipid hydrolysis at the sn-2 position to produce two potentially bioactive lipids that include non-esterified fatty acids (principally AA) and lysophospholipids such as LPC[60]. Among the sPLA2 enzymes groups IIa, V and X are highly expressed in human atherosclerotic lesions where the various groups have overlapping and dissimilar potential roles in atherosclerosis and lipoprotein metabolism[61]. The development of genetically manipulated mouse models lacking or overexpressing each of the three enzymes improved the understanding of the unique and shared functions of sPLA2-IIa, V and X in atherosclerosis. Two reviews about this topic including a detailed overview of group specific sPLA2 tissue and cell distribution were recently published[62] and[63] beside well-documented summaries of various clinical studies dealing with the questions of the potential role of sPLA2 in cardiovascular disorders[62],[63] and[64]. Concerning the role of lipoproteins as physiologic substrates of these three enzymes, sPLA2-V and X are considerably more active than sPLA2-IIA, an acute phase protein[65], to hydrolyze PC in HDL with no oxidation or modification of apolipoprotein A–I[66]. Thus, the reverse cholesterol efflux from lipid-loaded macrophages promoted by HDL is impaired. However, the potential in vivo consequences of HDL hydrolysis by sPLA2-V and X deserve further investigation. Furthermore, the interaction of apolipoprotein B containing lipoproteins with arterial proteoglycans in the intima is believed to be an important component of their atherogenicity[67]. In the extracellular intima sPLA2-V and X in contrast to sPLA2-IIa can be better suited for acting on apoliporotein B lipoproteins entrapped in this space[62] and LDL modified by sPLA2-V and X induce both foam cell formation[68]. sPLA2-IIa and V activity lead to fusion and enhanced binding of modified lipoproteins to extracellular proteoglycans that increase extracellular deposition of cholesterol-rich modified lipoproteins[69]. All these characteristics lead to the fact that sPLA2 enzymes have become a promising therapeutic target.
5.2. Lipoprotein-associated phospholipase A2 (Lp-PLA2)
Lp-PLA2, also known as platelet activating factor acetylhydrolase or type VIIA PLA2, is a calcium-independent phospholipase predominantly synthesized by macrophages. The biology of Lp-PLA2 is discussed in detail in a review article by Stafforini et al.[70]. In plasma Lp-PLA2 is bound to LDL and HDL lipoproteins with a greater affinity for the polar surface of LDL particles, particularly electronegative small LDL particles that have been minimally oxidatively modified[61]. Through hydrolysis of Ox-LDL particles, Lp-PLA2 generates two proinflammatory mediators, LPC and oxidized fatty acids, which activate redox-sensitive inflammatory pathways[71]. The products of Lp-PLA2 activity have been identified in human atherosclerotic vessel wall[72]. In preclinical animal studies, inhibition of the enzyme attenuates the inflammatory process and slows atherosclerotic disease progression, suggesting that it is not only a risk marker but also a candidate risk factor because it is intimately involved in the causal pathway of the intimal inflammatory cascade[73]. An overview of more than 25 published prospective epidemiologic studies showing an association of elevated Lp-PLA2 and higher cardiovascular risk is presented by recent reviews[64],[73] and[74]. Lp-PLA2 appears to be highly specific for vascular inflammation such that it appears to be unaffected by common infections or arthritis, and it has low biologic variability, similar to lipids[74]. The low biologic fluctuation makes it possible to use a single measurement for clinical decisions and also permits clinicians to follow serial assessment over time[74]. The level of Lp-PLA2 is independent of traditional risk factors, BMI and insulin resistance because Lp-PLA2 is produced by macrophages and foam cells in atherosclerotic plaques unlike systemic inflammatory markers, which may be produced by the liver in response to cytokines produced in mesenteric adipose tissue[74]. Lp-PLA2 is also incremental to high sensitivity C-reactive protein in risk assessment[73]. However, Lp-PLA2, unlike C-reactive protein and other biomarkers, is not recommended for general population screening. But, once a patient is determined to be at intermediate or high cardiovascular risk, elevated Lp-PLA2 might be used to move the patient to the next higher risk category, and low Lp-PLA2 might increase confidence that the patient is optimally treated or at lower risk[73]. Thus, the basic principle of preventive therapies is fulfilled to match the intensity of treatment to individual patient risk.
5.3. Phospholipase A2 inhibitors
To decrease LPC levels in the future a single drug molecule would be useful with a dual function, PLA2 inhibition and antioxidant activity, because PLA2 activation also results in AA-derived free radical intermediates. That could lead to PLA2 activity inhibition; scavenge free radicals released during AA metabolism and reduce pro-inflammatory molecules. Many secondary metabolites from plants and marine sponges like flavonoids, terpenes and alkaloids exhibit both anti-inflammatory and antioxidant properties. But in term of PLA2 inhibition and antioxidant activity, the structural aspects of flavonoids are well studied rather than terpenes and alkaloids[75]. Further selective PLA2 inhibitors have to be analyzed in randomized clinical trials and currently two specific inhibitors for Lp-PLA2 and sPLA2 are under evaluation[61]. Recently a review about PLA2 inhibitors was published by Rosenson et al.[61] showing that sPLA2 inhibition reduces LDL cholesterol levels, and both Lp-PLA2 and sPLA2 inhibitors mitigate inflammatory processes. The questions remain how much these enzymes should be inhibited to reduce cardiovascular events and how specific the inhibition should be to achieve efficiency. In relation to atherosclerosis treatment the inhibitor varespladib methyl emerged as a potential drug, especially in combination with statin therapy (pravastatin)[76].
6. Determination of relevant LPC species
The determination of LPC is often related to the determination of PLA2 activities[77]. However, potential effects of the detergent moiety on the enzyme activity cannot be excluded and the phospholipid bilayer structure is neglected. Beside mass spectrometry (MS) there exist other methods based on chromatography[78] and[79], UV- and fluorescence spectroscopy[80], NMR spectroscopy[81] and radioactivity[82] for analysis of lysophospholipids. The most common methods published for LPC analysis involve separation by thin-layer chromatography[83] and[84] or HPLC[85] and[86]. Studies analyzing LPC fatty acid composition used either TLC separation followed by gas chromatographic analysis[26] and[28] or HPLC coupled to electrospray ionization (ESI) MS[87]. The application of these methodologies in the routine laboratory, however, is limited because they are complicated, time-consuming procedures that are partly insensitive, lack specificity and accuracy. Within the last decade the analytical possibilities for detection of LPC species considerably improved. The detection and identification of phospholipids including LPC was made possible by further development of MS and meanwhile it is also possible to analyze all major lysophospholipids simultaneously[88] and[89]. Applying MALDI-TOF it was shown that the PC/LPC ratio was easily determined[90]. However most studies used liquid chromatography (LC) tandem MS[91],[92],[93] and[94] preferring ESI as the ion source of choice to couple LC with MS[95],[96] and[97]. It is concluded that ESI-MS/MS is the best and perhaps the only way to accurately determine the levels of each individual LPC species simultaneously in minimal amounts of body fluids such as blood. The “soft” ionization during usage of ESI-MS/MS does not cause extensive fragmentation, is highly sensitive, accurate, and reproducible without elaborate chromatographic separation.Suppl. Table 2 shows a protocol of an ESI-MS/MS methodology, developed by Liebisch et al.[97], which allows high-throughput quantification of distinct lysophosphatidylcholine (LPC) species from plasma and cell samples with an analysis time of 2 min/sample, simple sample preparation, and automated data analysis.
7. Future pathophysiological and molecular perspectives towards LPC based biomarker and drug target development
Research of the past decades has demonstrated that many important physiological and pathophysiological processes are regulated by lysophospholipids. However, there are still numerous open questions which require a better answer for the role of LPC in defined organs and diseases. Identification of enzymes controlling abnormal LPC levels in atherogenic disease and their relation to individual LPC species will be an initial step towards the development of novel therapeutics for treatment. LPC receptors need to be further characterized concerning ligand specificity and response, and attempts for pharmacological inhibition should be carried out. The fact that minor alterations of acyl chain length abolish LPC binding to GPR suggests that other structural modifications may uncover ligand mimetics with higher receptor binding affinities sufficient to effectively compete LPC binding[98]. The details of LPC contribution under conditions of atherogenic diseases have to be clarified after finding a lot of intriguing in vitro results for the important biological functions of LPC in cellular signalling and the development of different pathologies. It is also important to take into consideration that different LPC species, concerning fatty acid chain length and degree of saturation, could have different biological functions[99]. There are only a few studies related to inflammation which address this issue[100] and[101]. Many scientific and technical challenges need to be resolved to determine whether lysophospholipids or the enzymes producing lysophospholipids alone or in combination with other markers have the potential to contribute to early diagnosis. An emerging field of LPC based drug target development in atherosclerosis will certainly be inhibitors of the phospholipase A2 superfamily which serve as potential targets of therapy to reduce cardiovascular events as they are involved in lipoprotein modification, retention and oxidation; and activation of vascular and systemic inflammatory responses[61]. Together with Lp-PLA2 as a biomarker the final aim of preventive therapies might be fulfilled to match the intensity of treatment to individual patient risk.
Acknowledgements
This work was supported by Deutsche Forschungsgemeinschaft (SFB-TR 13/A3) (proposal number 013032) and by SSA – Danubian Biobank (WP4: enabling technologies) (proposal number 018822). This work was also supported by the seventh framework program of the EU-funded “LipidomicNet” (proposal number 202272). For more information see alsowww.lipidomicnet.org.
References
[1] T. Hla, M.J. Lee, N. Ancellin, J.H. Paik and M.J. Kluk, Lysophospholipids—receptor revelations, Science 294 (2001), pp. 1875–1878.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (345)
[2] E. Birgbauer and J. Chun, New developments in the biological functions of lysophospholipids, Cell Mol Life Sci 63 (2006), pp. 2695–2701.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (45)
[3] N. Fukushima, I. Ishii, J.J. Contos, J.A. Weiner and J. Chun, Lysophospholipid receptors, Annu Rev Pharmacol Toxicol 41 (2001), pp. 507–534.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (214)
[4] S.E. Gardell, A.E. Dubin and J. Chun, Emerging medicinal roles for lysophospholipid signaling, Trends Mol Med 12 (2006), pp. 65–75.Article | PDF (211 K) |View Record in Scopus |Cited By in Scopus (123)
[5] H.D. Meyer zu and K.H. Jakobs, Lysophospholipid receptors: signalling, pharmacology and regulation by lysophospholipid metabolism, Biochim Biophys Acta 1768 (2007), pp. 923–940.
[6] T.I. Torkhovskaya, O.M. Ipatova, T.S. Zakharova, M.M. Kochetova and E.M. Khalilov, Lysophospholipid receptors in cell signaling, Biochemistry (Mosc.) 72 (2007), pp. 125–131.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (10)
[7] Y. Xu, Y.J. Xiao and K. Zhu et al., Unfolding the pathophysiological role of bioactive lysophospholipids, Curr Drug Targets Immune Endocr Metabol Disord 3 (2003), pp. 23–32.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (35)
[8] K.P. Boggs, C.O. Rock and S. Jackowski, Lysophosphatidylcholine and 1-O-octadecyl-2-O-methyl-rac-glycero-3-phosphocholine inhibit the CDP-choline pathway of phosphatidylcholine synthesis at the CTP: phosphocholine cytidylyltransferase step, J Biol Chem 270 (1995), pp. 7757–7764.View Record in Scopus |Cited By in Scopus (106)
[9] D. Vance and J. Vance, Phospholipid biosynthesis in eukaryotes. In: D. Vance and J. Vance, Editors, Biochemistry of lipids, lipoproteins, and membranes, Elsevier, Amsterdam (2008), pp. 213–245.
[10] M. Gauster, G. Rechberger and A. Sovic et al., Endothelial lipase releases saturated and unsaturated fatty acids of high density lipoprotein phosphatidylcholine, J Lipid Res 46 (2005), pp. 1517–1525.View Record in Scopus |Cited By in Scopus (20)
[11] V.L.M. Lima, L.C.B.B. Coehlo, J.F. Kennedy and P.J. Dolphin, Lecithin-cholesterol acyltransferase (LCAT) as a plasma glycoprotein: an overview, Carbohyd Polym 55 (2004), pp. 179–191.Article | PDF (409 K) |View Record in Scopus |Cited By in Scopus (5)
[12] G. Sekas, G.M. Patton, E.C. Lincoln and S.J. Robins, Origin of plasma lysophosphatidylcholine: evidence for direct hepatic secretion in the rat, J Lab Clin Med 105 (1985), pp. 190–194.View Record in Scopus |Cited By in Scopus (24)
[13] J.R. Hessler, D.W. Morel, L.J. Lewis and G.M. Chisolm, Lipoprotein oxidation and lipoprotein-induced cytotoxicity, Arteriosclerosis 3 (1983), pp. 215–222.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (229)
[14] U.P. Steinbrecher, S. Parthasarathy, D.S. Leake, J.L. Witztum and D. Steinberg, Modification of low density lipoprotein by endothelial cells involves lipid peroxidation and degradation of low density lipoprotein phospholipids, Proc Natl Acad Sci USA 81 (1984), pp. 3883–3887.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (743)
[15] S. Ali, M.G. Davis, M.W. Becker and G.W. Dorn, Thromboxane A2 stimulates vascular smooth muscle hypertrophy by up-regulating the synthesis and release of endogenous basic fibroblast growth factor, J Biol Chem 268 (1993), pp. 17397–17403.View Record in Scopus |Cited By in Scopus (54)
[16] A. Negre-Salvayre, M. Lopez and T. Levade et al., Ultraviolet-treated lipoproteins as a model system for the study of the biological effects of lipid peroxides on cultured cells. II. Uptake and cytotoxicity of ultraviolet-treated LDL on lymphoid cell lines, Biochim Biophys Acta1045 (1990), pp. 224–232.Abstract | PDF (1109 K) |View Record in Scopus |Cited By in Scopus (42)
[17] E. Ehrenwald, G.M. Chisolm and P.L. Fox, Intact human ceruloplasmin oxidatively modifies low density lipoprotein, J Clin Invest 93 (1994), pp. 1493–1501.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (184)
[18] M.K. Cathcart, A.K. McNally and G.M. Chisolm, Lipoxygenase-mediated transformation of human low density lipoprotein to an oxidized and cytotoxic complex, J Lipid Res 32 (1991), pp. 63–70.View Record in Scopus |Cited By in Scopus (52)
[19] M.L. Savenkova, D.M. Mueller and J.W. Heinecke, Tyrosyl radical generated by myeloperoxidase is a physiological catalyst for the initiation of lipid peroxidation in low density lipoprotein, J Biol Chem 269 (1994), pp. 20394–20400.View Record in Scopus |Cited By in Scopus (252)
[20] J.A. Berliner, M.C. Territo and A. Sevanian et al., Minimally modified low density lipoprotein stimulates monocyte endothelial interactions, J Clin Invest 85 (1990), pp. 1260–1266.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (487)
[21] J.W. Heinecke, H. Rosen and A. Chait, Iron and copper promote modification of low density lipoprotein by human arterial smooth muscle cells in culture, J Clin Invest 74 (1984), pp. 1890–1894.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (250)
[22] D.W. Morel, P.E. DiCorleto and G.M. Chisolm, Endothelial and smooth muscle cells alter low density lipoprotein in vitro by free radical oxidation, Arteriosclerosis 4 (1984), pp. 357–364.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (317)
[23] M. Umezu-Goto, Y. Kishi and A. Taira et al., Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lysophosphatidic acid production, J Cell Biol 158 (2002), pp. 227–233.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (329)
[24] T. Matsumoto, T. Kobayashi and K. Kamata, Role of lysophosphatidylcholine (LPC) in atherosclerosis, Curr Med Chem 14 (2007), pp. 3209–3220.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (41)
[25] E. Liu, J.I. Goldhaber and J.N. Weiss, Effects of lysophosphatidylcholine on electrophysiological properties and excitation–contraction coupling in isolated guinea pig ventricular myocytes, J Clin Invest 88 (1991), pp. 1819–1832.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (35)
[26] M. Croset, N. Brossard, A. Polette and M. Lagarde, Characterization of plasma unsaturated lysophosphatidylcholines in human and rat, Biochem J 345 (Pt 1) (2000), pp. 61–67.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (88)
[27] F. Okajima, K. Sato and H. Tomura et al., Stimulatory and inhibitory actions of lysophosphatidylcholine, depending on its fatty acid residue, on the phospholipase C/Ca2+ system in HL-60 leukaemia cells, Biochem J 336 (Pt 2) (1998), pp. 491–500.View Record in Scopus |Cited By in Scopus (20)
[28] M. Okita, D.C. Gaudette, G.B. Mills and B.J. Holub, Elevated levels and altered fatty acid composition of plasma lysophosphatidylcholine (lysoPC) in ovarian cancer patients, Int J Cancer 71 (1997), pp. 31–34.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (81)
[29] T. Sasagawa, K. Suzuki, T. Shiota, T. Kondo and M. Okita, The significance of plasma lysophospholipids in patients with renal failure on hemodialysis, J Nutr Sci Vitaminol (Tokyo) 44 (1998), pp. 809–818.View Record in Scopus |Cited By in Scopus (40)
[30] R.A. Rabini, R. Galassi and P. Fumelli et al., Reduced Na(+)-K(+)-ATPase activity and plasma lysophosphatidylcholine concentrations in diabetic patients, Diabetes 43 (1994), pp. 915–919.View Record in Scopus |Cited By in Scopus (43)
[31] P.V. Subbaiah, C.H. Chen, J.D. Bagdade and J.J. Albers, Substrate specificity of plasma lysolecithin acyltransferase and the molecular species of lecithin formed by the reaction, J Biol Chem 260 (1985), pp. 5308–5314.View Record in Scopus |Cited By in Scopus (27)
[32] L. Chen, B. Liang and D.E. Froese et al., Oxidative modification of low density lipoprotein in normal and hyperlipidemic patients: effect of lysophosphatidylcholine composition on vascular relaxation, J Lipid Res 38 (1997), pp. 546–553.View Record in Scopus |Cited By in Scopus (50)
[33] Y. Xu, Sphingosylphosphorylcholine and lysophosphatidylcholine: G protein-coupled receptors and receptor-mediated signal transduction, Biochim Biophys Acta 1582 (2002), pp. 81–88.Article | PDF (112 K) |Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (122)
[34] G.K. Marathe, A.R. Silva and H.C. de Castro Faria Neto et al., Lysophosphatidylcholine and lyso-PAF display PAF-like activity derived from contaminating phospholipids, J Lipid Res 42 (2001), pp. 1430–1437.View Record in Scopus |Cited By in Scopus (25)
[35] C. Subra, K. Laulagnier, B. Perret and M. Record, Exosome lipidomics unravels lipid sorting at the level of multivesicular bodies, Biochimie 89 (2007), pp. 205–212.Article | PDF (463 K) |View Record in Scopus |Cited By in Scopus (19)
[36] E.M. Martin-Valmaseda, J. Sanchez-Yague, A. Hernandez-Hernandez and M. Llanillo, Vesiculation and changes in fluidity and lipid composition of platelet membranes after storage of sheep platelets in plasma or Seto solution, Thromb Haemost 80 (1998), pp. 668–676.View Record in Scopus |Cited By in Scopus (12)
[37] A.M. Weerheim, A.M. Kolb, A. Sturk and R. Nieuwland, Phospholipid composition of cell-derived microparticles determined by one-dimensional high-performance thin-layer chromatography, Anal Biochem 302 (2002), pp. 191–198.Abstract | PDF (140 K) |View Record in Scopus |Cited By in Scopus (44)
[38] K.E. Stremler, D.M. Stafforini, S.M. Prescott and T.M. McIntyre, Human plasma platelet-activating factor acetylhydrolase. Oxidatively fragmented phospholipids as substrates, J Biol Chem 266 (1991), pp. 11095–11103.View Record in Scopus |Cited By in Scopus (192)
[39] G.M. Chisolm III and Y. Chai, Regulation of cell growth by oxidized LDL, Free Radic Biol Med 28 (2000), pp. 1697–1707.Article | PDF (92 K) |View Record in Scopus |Cited By in Scopus (51)
[40] T. Soga, T. Ohishi and T. Matsui et al., Lysophosphatidylcholine enhances glucose-dependent insulin secretion via an orphan G-protein-coupled receptor, Biochem Biophys Res Commun 326 (2005), pp. 744–751.Article | PDF (456 K) |View Record in Scopus |Cited By in Scopus (76)
[41] C.G. Radu, L.V. Yang, M. Riedinger, M. Au and O.N. Witte, T cell chemotaxis to lysophosphatidylcholine through the G2A receptor, Proc Natl Acad Sci USA 101 (2004), pp. 245–250.View Record in Scopus |Cited By in Scopus (79)
[42] L.V. Yang, C.G. Radu, L. Wang, M. Riedinger and O.N. Witte, Gi-independent macrophage chemotaxis to lysophosphatidylcholine via the immunoregulatory GPCR G2A, Blood 105 (2005), pp. 1127–1134.View Record in Scopus |Cited By in Scopus (59)
[43] L.Q. Le, J.H. Kabarowski and Z. Weng et al., Mice lacking the orphan G protein-coupled receptor G2A develop a late-onset autoimmune syndrome, Immunity 14 (2001), pp. 561–571.Article | PDF (774 K) |View Record in Scopus |Cited By in Scopus (103)
[44] J. Qiao, F. Huang, R.P. Naikawadi, K.S. Kim, T. Said and H. Lum, Lysophosphatidylcholine impairs endothelial barrier function through the G protein-coupled receptor GPR4, Am J Physiol Lung Cell Mol Physiol 291 (2006), pp. L91–101.
[45] W.C. Sin, Y. Zhang and W. Zhong et al., G protein-coupled receptors GPR4 and TDAG8 are oncogenic and overexpressed in human cancers, Oncogene 23 (2004), pp. 6299–6303.Full Text via CrossRef
[46] Y. Aoki, M. Nakamura, H. Kodama, T. Matsumoto, T. Shimizu and M. Noma, A radioreceptor binding assay for platelet-activating factor (PAF) using membranes from CHO cells expressing human PAF receptor, J Immunol Methods 186 (1995), pp. 225–231.Article | PDF (653 K) |View Record in Scopus |Cited By in Scopus (16)
[47] Y.H. Huang, L. Schafer-Elinder, R. Wu, H.E. Claesson and J. Frostegard, Lysophosphatidylcholine (LPC) induces proinflammatory cytokines by a platelet-activating factor (PAF) receptor-dependent mechanism, Clin Exp Immunol 116 (1999), pp. 326–331.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (65)
[48] S. Ishii and T. Shimizu, Platelet-activating factor (PAF) receptor and genetically engineered PAF receptor mutant mice, Prog Lipid Res 39 (2000), pp. 41–82.Article | PDF (1089 K) |View Record in Scopus |Cited By in Scopus (170)
[49] T. Ogita, Y. Tanaka and T. Nakaoka et al., Lysophosphatidylcholine transduces Ca2+ signaling via the platelet-activating factor receptor in macrophages, Am J Physiol 272 (1997), pp. H17–H24.View Record in Scopus |Cited By in Scopus (34)
[50] S. Hara, T. Shike, N. Takasu and T. Mizui, Lysophosphatidylcholine promotes cholesterol efflux from mouse macrophage foam cells, Arterioscler Thromb Vasc Biol 17 (1997), pp. 1258–1266.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (23)
[51] N.V. Prokazova, N.D. Zvezdina and A.A. Korotaeva, Effect of lysophosphatidylcholine on transmembrane signal transduction, Biochemistry (Mosc.) 63 (1998), pp. 31–37.View Record in Scopus |Cited By in Scopus (54)
[52] B.V. Bassa, D.D. Roh, N.D. Vaziri, M.A. Kirschenbaum and V.S. Kamanna, Lysophosphatidylcholine activates mesangial cell PKC and MAP kinase by PLCgamma-1 and tyrosine kinase-Ras pathways, Am J Physiol 277 (1999), pp. F328–F337.View Record in Scopus |Cited By in Scopus (35)
[53] Q. Jing, S.M. Xin, W.B. Zhang, P. Wang, Y.W. Qin and G. Pei, Lysophosphatidylcholine activates p38 and p42/44 mitogen-activated protein kinases in monocytic THP-1 cells, but only p38 activation is involved in its stimulated chemotaxis, Circ Res 87 (2000), pp. 52–59.View Record in Scopus |Cited By in Scopus (52)
[54] Y. Yuan, S.M. Schoenwaelder, H.H. Salem and S.P. Jackson, The bioactive phospholipid, lysophosphatidylcholine, induces cellular effects via G-protein-dependent activation of adenyl cyclase, J Biol Chem 271 (1996), pp. 27090–27098.View Record in Scopus |Cited By in Scopus (66)
[55] N. Aiyar, J. Disa and Z. Ao et al., Lysophosphatidylcholine induces inflammatory activation of human coronary artery smooth muscle cells, Mol Cell Biochem 295 (2007), pp. 113–120.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (19)
[56] P. Kougias, H. Chai, P.H. Lin, A.B. Lumsden, Q. Yao and C. Chen, Lysophosphatidylcholine and secretory phospholipase A2 in vascular disease: mediators of endothelial dysfunction and atherosclerosis, Med Sci Monit 12 (2006), pp. RA5–16.
[57] K. Heermeier, R. Schneider, A. Heinloth, C. Wanner, S. Dimmeler and J. Galle, Oxidative stress mediates apoptosis induced by oxidized low-density lipoprotein and oxidized lipoprotein(a), Kidney Int 56 (1999), pp. 1310–1312.Full Text via CrossRef
[58] O.W. Portman and M. Alexander, Lysophosphatidylcholine concentrations and metabolism in aortic intima plus inner media: effect of nutritionally induced atherosclerosis, J Lipid Res 10 (1969), pp. 158–165.View Record in Scopus |Cited By in Scopus (84)
[59] A.K. Thukkani, J. McHowat, F.F. Hsu, M.L. Brennan, S.L. Hazen and D.A. Ford, Identification of alpha-chloro fatty aldehydes and unsaturated lysophosphatidylcholine molecular species in human atherosclerotic lesions, Circulation 108 (2003), pp. 3128–3133.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (70)
[60] J.E. Burke and E.A. Dennis, Phospholipase A2 biochemistry, Cardiovasc Drugs Ther 23 (2009), pp. 49–59.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (60)
[61] R.S. Rosenson, Future role for selective phospholipase A2 inhibitors in the prevention of atherosclerotic cardiovascular disease, Cardiovasc Drugs Ther 23 (2009), pp. 93–101.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (31)
[62] A.C. Jonsson-Rylander, S. Lundin, B. Rosengren, C. Pettersson and E. Hurt-Camejo, Role of secretory phospholipases in atherogenesis, Curr Atheroscler Rep 10 (2008), pp. 252–259.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (13)
[63] B. Rosengren, A.C. Jonsson-Rylander, H. Peilot, G. Camejo and E. Hurt-Camejo, Distinctiveness of secretory phospholipase A2 group IIA and V suggesting unique roles in atherosclerosis, Biochim Biophys Acta 1761 (2006), pp. 1301–1308.Article | PDF (344 K) |View Record in Scopus |Cited By in Scopus (24)
[64] W. Koenig and N. Khuseyinova, Lipoprotein-associated and secretory phospholipase A2 in cardiovascular disease: the epidemiological evidence, Cardiovasc Drugs Ther 23 (2009), pp. 85–92.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (19)
[65] M. Hartford, O. Wiklund and H.L. Mattsson et al., CRP, interleukin-6, secretory phospholipase A2 group IIA, and intercellular adhesion molecule-1 during the early phase of acute coronary syndromes and long-term follow-up, Int J Cardiol 108 (2006), pp. 55–62.Article | PDF (146 K) |View Record in Scopus |Cited By in Scopus (27)
[66] W. Pruzanski, G. Lambeau, M. Lazdunski, W. Cho, J. Kopilov and A. Kuksis, Hydrolysis of minor glycerophospholipids of plasma lipoproteins by human group IIA, V and X secretory phospholipases A2, Biochim Biophys Acta 1771 (2007), pp. 5–19.Article | PDF (1823 K) |View Record in Scopus |Cited By in Scopus (5)
[67] G. Camejo, E. Hurt-Camejo, O. Wiklund and G. Bondjers, Association of apo B lipoproteins with arterial proteoglycans: pathological significance and molecular basis, Atherosclerosis 139 (1998), pp. 205–222.Article | PDF (858 K) |View Record in Scopus |Cited By in Scopus (175)
[68] B.B. Boyanovsky and N.R. Webb, Biology of secretory phospholipase A2, Cardiovasc Drugs Ther 23 (2009), pp. 61–72.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (29)
[69] J.K. Hakala, K. Oorni, M.O. Pentikainen, E. Hurt-Camejo and P.T. Kovanen, Lipolysis of LDL by human secretory phospholipase A(2) induces particle fusion and enhances the retention of LDL to human aortic proteoglycans, Arterioscler Thromb Vasc Biol 21 (2001), pp. 1053–1058.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (71)
[70] D.M. Stafforini, Biology of platelet-activating factor acetylhydrolase (PAF-AH, lipoprotein associated phospholipase A2), Cardiovasc Drugs Ther 23 (2009), pp. 73–83.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (40)
[71] A. Zalewski and C. Macphee, Role of lipoprotein-associated phospholipase A2 in atherosclerosis: biology, epidemiology, and possible therapeutic target, Arterioscler Thromb Vasc Biol 25 (2005), pp. 923–931.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (153)
[72] G.K. Marathe, K.A. Harrison, R.C. Murphy, S.M. Prescott, G.A. Zimmerman and T.M. McIntyre, Bioactive phospholipid oxidation products, Free Radic Biol Med 28 (2000), pp. 1762–1770.Article | PDF (159 K) |View Record in Scopus |Cited By in Scopus (59)
[73] J.L. Anderson, Lipoprotein-associated phospholipase A2: an independent predictor of coronary artery disease events in primary and secondary prevention, Am J Cardiol 101 (2008), pp. 23F–33F.
[74] M.A. Corson, P.H. Jones and M.H. Davidson, Review of the evidence for the clinical utility of lipoprotein-associated phospholipase A2 as a cardiovascular risk marker, Am J Cardiol 101 (2008), pp. 41F–50F.
[75] B.L. Nanda, A. Nataraju, R. Rajesh, K.S. Rangappa, M.A. Shekar and B.S. Vishwanath, PLA2 mediated arachidonate free radicals: PLA2 inhibition and neutralization of free radicals by anti-oxidants—a new role as anti-inflammatory molecule, Curr Top Med Chem 7 (2007), pp. 765–777.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (18)
[76] Z. Shaposhnik, X. Wang, J. Trias, H. Fraser and A.J. Lusis, The synergistic inhibition of atherogenesis in apoE-/- mice between pravastatin and the sPLA2 inhibitor varespladib (A-002), J Lipid Res 50 (2009), pp. 623–629.View Record in Scopus |Cited By in Scopus (20)
[77] B.H. van den and A.J. Aarsman, A review on methods of phospholipase A determination, Agents Actions 9 (1979), pp. 382–389.
[78] S. Choi, Y.S. Lee, D.S. Na and Y.S. Yoo, Determination of enzymatic activity and properties of secretory phospholipase A2 by capillary electrophoresis, J Chromatogr A 853 (1999), pp. 285–293.Article | PDF (262 K) |View Record in Scopus |Cited By in Scopus (10)
[79] C.S. Lai, J.Z. Zhang and J. Joseph, Spin-label assay for phospholipase A2, Anal Biochem 172 (1988), pp. 397–402.Abstract |Article | PDF (483 K) |View Record in Scopus |Cited By in Scopus (7)
[80] A. Rawyler and P.A. Siegenthaler, A single and continuous spectrophotometric assay for various lipolytic enzymes, using natural, non-labelled lipid substrates, Biochim Biophys Acta 1004 (1989), pp. 337–344.Abstract | PDF (1284 K) |View Record in Scopus |Cited By in Scopus (11)
[81] B. Fuchs, J. Schiller, U. Wagner, H. Hantzschel and K. Arnold, The phosphatidylcholine/lysophosphatidylcholine ratio in human plasma is an indicator of the severity of rheumatoid arthritis: investigations by 31P NMR and MALDI-TOF MS, Clin Biochem 38 (2005), pp. 925–933.Article | PDF (396 K) |View Record in Scopus |Cited By in Scopus (43)
[82] S.D. Catz and N.B. Sterin-Speziale, Bradykinin stimulates phosphoinositide turnover and phospholipase C but not phospholipase D and NADPH oxidase in human neutrophils, J Leukoc Biol 59 (1996), pp. 591–597.View Record in Scopus |Cited By in Scopus (15)
[83] W.Q. Wang and A. Gustafson, One-dimensional thin-layer chromatographic separation of phospholipids and lysophospholipids from tissue lipid extracts, J Chromatogr 581 (1992), pp. 139–142.Abstract | PDF (356 K) |View Record in Scopus |Cited By in Scopus (19)
[84] K. Yokoyama, F. Shimizu and M. Setaka, Simultaneous separation of lysophospholipids from the total lipid fraction of crude biological samples using two-dimensional thin-layer chromatography, J Lipid Res 41 (2000), pp. 142–147.View Record in Scopus |Cited By in Scopus (21)
[85] Z. Guan, J. Grunler, S. Piao and P.J. Sindelar, Separation and quantitation of phospholipids and their ether analogues by high-performance liquid chromatography, Anal Biochem 297 (2001), pp. 137–143.Abstract | PDF (77 K) |View Record in Scopus |Cited By in Scopus (25)
[86] Z. Suchocka, D. Gronostajska, P. Suchocki and J. Pachecka, New HPLC method for separation of blood plasma phospholipids, J Pharm Biomed Anal 32 (2003), pp. 859–865.Article | PDF (206 K) |View Record in Scopus |Cited By in Scopus (14)
[87] W. Pruzanski, E. Stefanski, F.C. de Beer, M.C. de Beer, A. Ravandi and A. Kuksis, Comparative analysis of lipid composition of normal and acute-phase high density lipoproteins, J Lipid Res 41 (2000), pp. 1035–1047.View Record in Scopus |Cited By in Scopus (61)
[88] Y. Xiao, Y. Chen, A.W. Kennedy, J. Belinson and Y. Xu, Evaluation of plasma lysophospholipids for diagnostic significance using electrospray ionization mass spectrometry (ESI-MS) analyses, Ann N Y Acad Sci 905 (2000), pp. 242–259.View Record in Scopus |Cited By in Scopus (66)
[89] M. Murph, T. Tanaka and J. Pang et al., Liquid chromatography mass spectrometry for quantifying plasma lysophospholipids: potential biomarkers for cancer diagnosis, Methods Enzymol 433 (2007), pp. 1–25.Abstract |Article | PDF (273 K) |View Record in Scopus |Cited By in Scopus (17)
[90] M. Petkovic, J. Muller, M. Muller, J. Schiller, K. Arnold and J. Arnhold, Application of matrix-assisted laser desorption/ionization time-of-flight mass spectrometry for monitoring the digestion of phosphatidylcholine by pancreatic phospholipase A(2), Anal Biochem 308 (2002), pp. 61–70.Article | PDF (357 K) |View Record in Scopus |Cited By in Scopus (25)
[91] S. Cai, T. Huo, N. Li, Z. Xiong and F. Li, Lysophosphatidylcholine-biomarker of Metformin action: studied using UPLC/MS/MS, Biomed Chromatogr (2009).
[92] L.Q. Pang, Q.L. Liang, Y.M. Wang, L. Ping and G.A. Luo, Simultaneous determination and quantification of seven major phospholipid classes in human blood using normal-phase liquid chromatography coupled with electrospray mass spectrometry and the application in diabetes nephropathy, J Chromatogr B Analyt Technol Biomed Life Sci 869 (2008), pp. 118–125.Article | PDF (714 K) |View Record in Scopus |Cited By in Scopus (18)
[93] J.C. Smith, W. Hou, S.N. Whitehead, M. Ethier, S.A. Bennett and D. Figeys, Identification of lysophosphatidylcholine (LPC) and platelet activating factor (PAF) from PC12 cells and mouse cortex using liquid chromatography/multi-stage mass spectrometry (LC/MS3), Rapid Commun Mass Spectrom 22 (2008), pp. 3579–3587.Full Text via CrossRef
[94] A. Takatera, A. Takeuchi, K. Saiki, T. Morisawa, N. Yokoyama and M. Matsuo, Quantification of lysophosphatidylcholines and phosphatidylcholines using liquid chromatography-tandem mass spectrometry in neonatal serum, J Chromatogr B Analyt Technol Biomed Life Sci 838 (2006), pp. 31–36.Article | PDF (176 K) |View Record in Scopus |Cited By in Scopus (20)
[95] B. Davis, G. Koster and L.J. Douet et al., Electrospray ionization mass spectrometry identifies substrates and products of lipoprotein-associated phospholipase A2 in oxidized human low density lipoprotein, J Biol Chem 283 (2008), pp. 6428–6437.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (26)
[96] F.F. Hsu and J. Turk, Electrospray ionization/tandem quadrupole mass spectrometric studies on phosphatidylcholines: the fragmentation processes, J Am Soc Mass Spectrom 14 (2003), pp. 352–363.Article | PDF (408 K) |Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (108)
[97] G. Liebisch, W. Drobnik, B. Lieser and G. Schmitz, High-throughput quantification of lysophosphatidylcholine by electrospray ionization tandem mass spectrometry, Clin Chem 48 (2002), pp. 2217–2224.View Record in Scopus |Cited By in Scopus (46)
[98] J.H. Kabarowski, Y. Xu and O.N. Witte, Lysophosphatidylcholine as a ligand for immunoregulation, Biochem Pharmacol 64 (2002), pp. 161–167.Article | PDF (281 K) |View Record in Scopus |Cited By in Scopus (47)
[99] K. Leidl, G. Liebisch, D. Richter and G. Schmitz, Mass spectrometric analysis of lipid species of human circulating blood cells, Biochim Biophys Acta 1781 (2008), pp. 655–664.Article | PDF (1206 K) |View Record in Scopus |Cited By in Scopus (14)
[100] J.J. Yan, J.S. Jung and J.E. Lee et al., Therapeutic effects of lysophosphatidylcholine in experimental sepsis, Nat Med 10 (2004), pp. 161–167.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (108)
[101] W. Drobnik, G. Liebisch and F.X. Audebert et al., Plasma ceramide and lysophosphatidylcholine inversely correlate with mortality in sepsis patients, J Lipid Res 44 (2003), pp. 754–761.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (68)
[102] K. Hirano, T. Ito, H. Morihara, T. Tanaka and K. Satouchi, Cytosolic lysophosphatidylcholine/transacylase in the production of dipolyunsaturated phosphatidylcholine in bonito muscle, FEBS Lett 437 (1998), pp. 193–196.Article | PDF (92 K) |View Record in Scopus |Cited By in Scopus (4)
[103] R.M. Dawson, R.F. Irvine, N.L. Hemington and K. Hirasawa, The alkaline phospholipase A1 of rat liver cytosol, Biochem J 209 (1983), pp. 865–872.View Record in Scopus |Cited By in Scopus (9)
[104] F. Granata, B. Balestrieri, A. Petraroli, G. Giannattasio, G. Marone and M. Triggiani, Secretory phospholipases A2 as multivalent mediators of inflammatory and allergic disorders, Int Arch Allergy Immunol 131 (2003), pp. 153–163.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (29)
[105] G.M. Jenkins and M.A. Frohman, Phospholipase D: a lipid centric review, Cell Mol Life Sci 62 (2005), pp. 2305–2316.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (149)
[106] S. Chen and P.V. Subbaiah, Phospholipid and fatty acid specificity of endothelial lipase: potential role of the enzyme in the delivery of docosahexaenoic acid (DHA) to tissues, Biochim Biophys Acta 1771 (2007), pp. 1319–1328.Article | PDF (778 K) |View Record in Scopus |Cited By in Scopus (16)
[107] R.D. Shamburek, L.A. Zech, P.S. Cooper, J.M. Vandenbroek and C.C. Schwartz, Disappearance of two major phosphatidylcholines from plasma is predominantly via LCAT and hepatic lipase, Am J Physiol 271 (1996), pp. E1073–E1082.View Record in Scopus |Cited By in Scopus (5)
[108] B. Perret, L. Mabile, L. Martinez, F. Terce, R. Barbaras and X. Collet, Hepatic lipase: structure/function relationship, synthesis, and regulation, J Lipid Res 43 (2002), pp. 1163–1169.View Record in Scopus |Cited By in Scopus (82)
[109] A. Wang, R. Loo, Z. Chen and E.A. Dennis, Regiospecificity and catalytic triad of lysophospholipase I, J Biol Chem 272 (1997), pp. 22030–22036.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (43)
[110] Y. Nakagawa, M. Sugai and K. Karasawa et al., Possible influence of lysophospholipase on the production of 1-acyl-2-acetylglycerophosphocholine in macrophages, Biochim Biophys Acta 1126 (1992), pp. 277–285.Abstract | PDF (886 K) |View Record in Scopus |Cited By in Scopus (13)
[111] Y. Xie and K.E. Meier, Lysophospholipase D and its role in LPA production, Cell Signal 16 (2004), pp. 975–981.Article | PDF (137 K) |View Record in Scopus |Cited By in Scopus (13)
[112] S.K. Jackson and J. Parton, Lysophospholipid acyltransferases in monocyte inflammatory responses and sepsis, Immunobiology 209 (2004), pp. 31–38.Article | PDF (393 K) |View Record in Scopus |Cited By in Scopus (5)
[113] Y. Zhao, Y.Q. Chen and T.M. Bonacci et al., Identification and characterization of a major liver lysophosphatidylcholine acyltransferase, J Biol Chem 283 (2008), pp. 8258–8265.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (27)
[114] X. Chen, B.A. Hyatt, M.L. Mucenski, R.J. Mason and J.M. Shannon, Identification and characterization of a lysophosphatidylcholine acyltransferase in alveolar type II cells, Proc Natl Acad Sci USA 103 (2006), pp. 11724–11729.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (54)
[115] E. Soupene, H. Fyrst and F.A. Kuypers, Mammalian acyl-CoA: lysophosphatidylcholine acyltransferase enzymes, Proc Natl Acad Sci USA 105 (2008), pp. 88–93.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (23)
[116] N. Kume, M.I. Cybulsky and M.A. Gimbrone Jr., Lysophosphatidylcholine, a component of atherogenic lipoproteins, induces mononuclear leukocyte adhesion molecules in cultured human and rabbit arterial endothelial cells, J Clin Invest 90 (1992), pp. 1138–1144.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (533)
[117] J.X. Rong, J.W. Berman, M.B. Taubman and E.A. Fisher, Lysophosphatidylcholine stimulates monocyte chemoattractant protein-1 gene expression in rat aortic smooth muscle cells, Arterioscler Thromb Vasc Biol 22 (2002), pp. 1617–1623.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (36)
[118] N. Takahara, A. Kashiwagi, H. Maegawa and Y. Shigeta, Lysophosphatidylcholine stimulates the expression and production of MCP-1 by human vascular endothelial cells, Metabolism 45 (1996), pp. 559–564.Article | PDF (2629 K) |View Record in Scopus |Cited By in Scopus (60)
[119] S.M. Colles and G.M. Chisolm, Lysophosphatidylcholine-induced cellular injury in cultured fibroblasts involves oxidative events, J Lipid Res 41 (2000), pp. 1188–1198.View Record in Scopus |Cited By in Scopus (30)
[120] C.C. Hsieh, M.H. Yen, H.W. Liu and Y.T. Lau, Lysophosphatidylcholine induces apoptotic and non-apoptotic death in vascular smooth muscle cells: in comparison with oxidized LDL, Atherosclerosis 151 (2000), pp. 481–491.Article | PDF (309 K) |View Record in Scopus |Cited By in Scopus (48)
[121] M. Matsubara, K. Yao and K. Hasegawa, Benidipine, a dihydropyridine-calcium channel blocker, inhibits lysophosphatidylcholine-induced endothelial injury via stimulation of nitric oxide release, Pharmacol Res 53 (2006), pp. 35–43.Article | PDF (2203 K) |View Record in Scopus |Cited By in Scopus (14)
[122] S. Yan, H. Chai and H. Wang et al., Effects of lysophosphatidylcholine on monolayer cell permeability of human coronary artery endothelial cells, Surgery 138 (2005), pp. 464–473.Article | PDF (348 K) |Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (20)
[123] J.T. Wong, K. Tran, G.N. Pierce, A.C. Chan and P.C. Choy, Lysophosphatidylcholine stimulates the release of arachidonic acid in human endothelial cells, J Biol Chem 273 (1998), pp. 6830–6836.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (57)
[124] A. Ceylan, C. Karasu, F. Aktan and G. Ozansoy, Simvastatin treatment restores vasoconstriction and the inhibitory effect of LPC on endothelial relaxation via affecting oxidizing metabolism in diabetic rats, Diabetes Nutr Metab 17 (2004), pp. 203–210.View Record in Scopus |Cited By in Scopus (8)
[125] Y.C. Chai, P.H. Howe, P.E. DiCorleto and G.M. Chisolm, Oxidized low density lipoprotein and lysophosphatidylcholine stimulate cell cycle entry in vascular smooth muscle cells. Evidence for release of fibroblast growth factor-2, J Biol Chem 271 (1996), pp. 17791–17797.View Record in Scopus |Cited By in Scopus (109)
[126] M. Kohno, K. Yokokawa and K. Yasunari et al., Induction by lysophosphatidylcholine, a major phospholipid component of atherogenic lipoproteins, of human coronary artery smooth muscle cell migration, Circulation 98 (1998), pp. 353–359.View Record in Scopus |Cited By in Scopus (77)
[127] Y. Liu-Wu, E. Hurt-Camejo and O. Wiklund, Lysophosphatidylcholine induces the production of IL-1beta by human monocytes, Atherosclerosis 137 (1998), pp. 351–357.Article | PDF (149 K) |View Record in Scopus |Cited By in Scopus (55)
[128] S.S. Ousman and S. David, Lysophosphatidylcholine induces rapid recruitment and activation of macrophages in the adult mouse spinal cord, Glia 30 (2000), pp. 92–104.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (68)
[129] T. Kita, N. Kume and M. Yokode et al., Oxidized-LDL and atherosclerosis. Role of LOX-1, Ann N Y Acad Sci 902 (2000), pp. 95–100.
[130] H. Oka, K. Kugiyama and H. Doi et al., Lysophosphatidylcholine induces urokinase-type plasminogen activator and its receptor in human macrophages partly through redox-sensitive pathway, Arterioscler Thromb Vasc Biol 20 (2000), pp. 244–250.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (21)
[131] J. Oestvang and B. Johansen, PhospholipaseA2: a key regulator of inflammatory signalling and a connector to fibrosis development in atherosclerosis, Biochim Biophys Acta 1761 (2006), pp. 1309–1316.Article | PDF (518 K) |View Record in Scopus |Cited By in Scopus (19)
[132] M. Aviram and M. Rosenblat, Paraoxonases 1, 2, and 3, oxidative stress, and macrophage foam cell formation during atherosclerosis development, Free Radic Biol Med 37 (2004), pp. 1304–1316.Article | PDF (538 K) |View Record in Scopus |Cited By in Scopus (117)
[133] M. Yamamoto, H. Hara and T. Adachi, The expression of extracellular-superoxide dismutase is increased by lysophosphatidylcholine in human monocytic U937 cells, Atherosclerosis 163 (2002), pp. 223–228.Article | PDF (222 K) |View Record in Scopus |Cited By in Scopus (4)
[134] M. Rosenblat, J. Vaya, D. Shih and M. Aviram, Paraoxonase 1 (PON1) enhances HDL-mediated macrophage cholesterol efflux via the ABCA1 transporter in association with increased HDL binding to the cells: a possible role for lysophosphatidylcholine, Atherosclerosis 179 (2005), pp. 69–77.Article | PDF (360 K) |View Record in Scopus |Cited By in Scopus (60)
[135] M. Rosenblat, L. Gaidukov and O. Khersonsky et al., The catalytic histidine dyad of high density lipoprotein-associated serum paraoxonase-1 (PON1) is essential for PON1-mediated inhibition of low density lipoprotein oxidation and stimulation of macrophage cholesterol efflux, J Biol Chem 281 (2006), pp. 7657–7665.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (79)
[136] A.K. Ryborg, B. Deleuran, K. Thestrup-Pedersen and K. Kragballe, Lysophosphatidylcholine: a chemoattractant to human T lymphocytes, Arch Dermatol Res 286 (1994), pp. 462–465.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (17)
[137] C. Weber, W. Erl and P.C. Weber, Enhancement of monocyte adhesion to endothelial cells by oxidatively modified low-density lipoprotein is mediated by activation of CD11b, Biochem Biophys Res Commun 206 (1995), pp. 621–628.Abstract | PDF (407 K) |View Record in Scopus |Cited By in Scopus (48)
[138] T. Nakano, E.W. Raines, J.A. Abraham, M. Klagsbrun and R. Ross, Lysophosphatidylcholine upregulates the level of heparin-binding epidermal growth factor-like growth factor mRNA in human monocytes, Proc Natl Acad Sci USA 91 (1994), pp. 1069–1073.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (103)
[139] E. Nishi, N. Kume and H. Ochi et al., Lysophosphatidylcholine increases expression of heparin-binding epidermal growth factor-like growth factor in human T lymphocytes, Circ Res 80 (1997), pp. 638–644.View Record in Scopus |Cited By in Scopus (23)
[140] R.D. Hoffman, M. Kligerman, T.M. Sundt, N.D. Anderson and H.S. Shin, Stereospecific chemoattraction of lymphoblastic cells by gradients of lysophosphatidylcholine, Proc Natl Acad Sci USA 79 (1982), pp. 3285–3289.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (17)
[141] N. Kume and M.A. Gimbrone Jr., Lysophosphatidylcholine transcriptionally induces growth factor gene expression in cultured human endothelial cells, J Clin Invest 93 (1994), pp. 907–911.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (240)
[142] R.B. Mueller, A. Sheriff, U.S. Gaipl, S. Wesselborg and K. Lauber, Attraction of phagocytes by apoptotic cells is mediated by lysophosphatidylcholine, Autoimmunity 40 (2007), pp. 342–344.Full Text via CrossRef |View Record in Scopus |Cited By in Scopus (10)
[143] K. Lauber, E. Bohn and S.M. Krober et al., Apoptotic cells induce migration of phagocytes via caspase-3-mediated release of a lipid attraction signal, Cell 113 (2003), pp. 717–730.Article | PDF (675 K) |View Record in Scopus |Cited By in Scopus (289)
[144] W.Y. Fujimoto and J. Teague, Phasic effects of glucose, p-hydroxymercuribenzoate, and lysophosphatidylcholine on insulin secretion from HIT cells, Diabetes 38 (1989), pp. 625–628.View Record in Scopus |Cited By in Scopus (5)
Appendix A. Supplementary data
 

Download this File (382 K)
View Within Article
 
Corresponding author. Tel.: +49 941 944 6201; fax: +49 941 944 6202.
Copyright © 2009 Published by Elsevier Ltd.
Atherosclerosis
Volume 208, Issue 1, January 2010, Pages 10-18


',2)">
Home
Browse
Search
My settings
My alerts
Help

About ScienceDirectWhat is ScienceDirectContent detailsSet upHow to useSubscriptionsDevelopers
Contact and SupportContact and Support
About ElsevierAbout ElsevierAbout SciVerseAbout SciValTerms and ConditionsPrivacy policyInformation for advertisers
Copyright © 2011Elsevier B.V. All rights reserved. SciVerse® is a registered trademark of Elsevier Properties S.A., used under license. ScienceDirect® is a registered trademark of Elsevier B.V.