Membrane plasmalogen composition and cellular cholesterol regulation: a structure activity study

Rishikesh Mankidy, Pearson Wk Ahiahonu, Hong Ma, Dushmanthi Jayasinghe, Shawn A Ritchie, Mohamed A Khan, Khine K Su-Myat, Paul L Wood, Dayan B Goodenowe, Rishikesh Mankidy, Pearson Wk Ahiahonu, Hong Ma, Dushmanthi Jayasinghe, Shawn A Ritchie, Mohamed A Khan, Khine K Su-Myat, Paul L Wood, Dayan B Goodenowe

Abstract

Background: Disrupted cholesterol regulation leading to increased circulating and membrane cholesterol levels is implicated in many age-related chronic diseases such as cardiovascular disease (CVD), Alzheimer's disease (AD), and cancer. In vitro and ex vivo cellular plasmalogen deficiency models have been shown to exhibit impaired intra- and extra-cellular processing of cholesterol. Furthermore, depleted brain plasmalogens have been implicated in AD and serum plasmalogen deficiencies have been linked to AD, CVD, and cancer.

Results: Using plasmalogen deficient (NRel-4) and plasmalogen sufficient (HEK293) cells we investigated the effect of species-dependent plasmalogen restoration/augmentation on membrane cholesterol processing. The results of these studies indicate that the esterification of cholesterol is dependent upon the amount of polyunsaturated fatty acid (PUFA)-containing ethanolamine plasmalogen (PlsEtn) present in the membrane. We further elucidate that the concentration-dependent increase in esterified cholesterol observed with PUFA-PlsEtn was due to a concentration-dependent increase in sterol-O-acyltransferase-1 (SOAT1) levels, an observation not reproduced by 3-hydroxy-3-methyl-glutaryl-CoA (HMG-CoA) reductase inhibition.

Conclusion: The present study describes a novel mechanism of cholesterol regulation that is consistent with clinical and epidemiological studies of cholesterol, aging and disease. Specifically, the present study describes how selective membrane PUFA-PlsEtn enhancement can be achieved using 1-alkyl-2-PUFA glycerols and through this action reduce levels of total and free cholesterol in cells.

Figures

Figure 1
Figure 1
Scheme showing the syntheses of phosphoethanolamine plasmalogen precursors C1-3, C6-10, and diacylglycerols C4 and C5, test compounds for the study. Reagents: (a) i R1Br, NaH/DMF or R1SO4CH3, NaH/THF, reflux; (ii) 10% HCl, reflux; (b) TBDMS-Cl, imidazole/DMF; (c) R2COCl, DMAP, Pyridine, Toluene; (d) TBAF/THF, Imidazole, - 20°C
Figure 2
Figure 2
Relative ethanolamine plasmalogens in DHAPAT-deficient cells (A). Plasmalogen content of CHO (C_V) and NRel-4 (N_V) cell lines. Values are an average of three independent experiments; error bars represent standard deviation. All transitions measured in NRel-4 cells were significantly different from control cells (p < 0.05). Cholesterol profile of DHAPAT-deficient cells (B). Values are an average of independent experiments; error bars represent standard deviation. Total, free and esterified cholesterol of N_V is significantly different from C_V ( p < 0.05).
Figure 3
Figure 3
Plasmalogen biosynthetic pathway showing therapeutic intervention.
Figure 4
Figure 4
Side chain-specific restoration of PlsEtn in NRel-4 cells. Effect of C1, C2, C3, C6-10 treatment of NRel-4 (N_V) cells on sn-1 specific PlsEtn pools.
Figure 5
Figure 5
Sn-2 rearrangement following C1, C2, C3, C6-10 treatment of NRel-4 (N_V) cells. Relative distribution of sn-2 fatty acids within each plasmalogen pool following treatments as above is displayed. All PlsEtn measurements are reported relative to the control CHO cells (C_V). Results are an average of three independent experiments. Error bars represent standard deviation.
Figure 6
Figure 6
Comparison of sn-2 fatty acid substitution (A) and sn-1bond type (B) on total DHA PlsEtn levels in NRel-4 cells. NRel-4 cells were treated with ethanol solvent (N_V), or with test compounds at 20 μM concentration. Compounds C1, C6-10 contain palmityl ether at sn-1 and different fatty acid moieties at sn-2 position; DHA (C1), oleic acid (C7), linoleic acid (C8), linolenic acid (C9), and arachidonic acid (C10), -OH refers to a free hydroxyl group at sn-2 position. Compounds C4 and C5 have acyl linkages at sn-1 and sn-2 positions. Total DHA containing ethanolamine plasmalogens were quantified, and expressed relative to the amount observed in wild-type CHO cells (C_V). Values were an average of three independent experiments. Error bars represent standard deviation.
Figure 7
Figure 7
Concentration response curve of PlsEtn precursor C1 in CHO and NRel-4 cells. Treatments were carried out at concentrations of 1 μM, 5 μM, and 20 μM of C1. (A): Relative restoration/augmentation of DHA PlsEtn; (B): Relative restoration/augmentation of total PlsEtn. Values were normalized to control CHO cells (C_V). Results are an average of three independent experiments. Error bars indicate standard deviation.
Figure 8
Figure 8
Cholesterol profile of CHO/NRel-4 cells. Cholesterol profile of NRel-4 cells following 48 hour treatments with PlsEtn precursors (C1, C6-10) and PtdEtn precursors (C4, C5) compared to control CHO cells. A: total cholesterol; B: free cholesterol; C: esterified cholesterol. Cholesterol is reported as μg per million cells. Results are an average of three independent experiments. Asterisk represents values that are significantly different from those observed in DHAPAT-deficient NRel-4 cells (p < 0.05).
Figure 9
Figure 9
Cholesterol profile of HEK293 cells treated for 48 hr with plasmalogen precursors. Cholesterol (total, free and esterified) content is reported as μg/million cells, and is an average of two independent experiments. Error bars indicate standard deviation. Asterisk indicates significantly lower total or free cholesterol, or significantly higher esterified cholesterol compared with control.
Figure 10
Figure 10
Immunoblots showing SOAT1 protein levels in wild-type HEK293 cells treated with a concentration range of C1 and pravastatin. β-actin was used as a loading control.

References

    1. Fielding CJ, Fielding PE. Membrane cholesterol and the regulation of signal transduction. Biochemical Society transactions. 2004;32(Pt 1):65–69. doi: 10.1042/BST0320065.
    1. Zwijsen RM, Oudenhoven IM, de Haan LH. Effects of cholesterol and oxysterols on gap junctional communication between human smooth muscle cells. European journal of pharmacology. 1992;228(2-3):115–120.
    1. Executive Summary of The Third Report of The National Cholesterol Education Program (NCEP) Expert Panel on Detection, Evaluation, And Treatment of High Blood Cholesterol In Adults (Adult Treatment Panel III) Jama. 2001;285(19):2486–2497. doi: 10.1001/jama.285.19.2486.
    1. Fowler SB, Kelly M, Ruh D, Johnson-Wells D. Management of lipid disorders for stroke prevention. J Neurosci Nurs. 2006;38(4 Suppl):282–287. doi: 10.1097/01376517-200609000-00003. 295.
    1. Corder EH, Saunders AM, Strittmatter WJ, Schmechel DE, Gaskell PC, Small GW, Roses AD, Haines JL, Pericak-Vance MA. Gene dose of apolipoprotein E type 4 allele and the risk of Alzheimer's disease in late onset families. Science (New York, NY) 1993;261(5123):921–923.
    1. Hager MH, Solomon KR, Freeman MR. The role of cholesterol in prostate cancer. Current opinion in clinical nutrition and metabolic care. 2006;9(4):379–385. doi: 10.1097/01.mco.0000232896.66791.62.
    1. Dietschy JM, Turley SD. Cholesterol metabolism in the brain. Current opinion in lipidology. 2001;12(2):105–112. doi: 10.1097/00041433-200104000-00003.
    1. Muse ED, Jurevics H, Toews AD, Matsushima GK, Morell P. Parameters related to lipid metabolism as markers of myelination in mouse brain. J Neurochem. 2001;76(1):77–86. doi: 10.1046/j.1471-4159.2001.00015.x.
    1. Martins IJ, Berger T, Sharman MJ, Verdile G, Fuller SJ, Martins RN. Cholesterol metabolism and transport in the pathogenesis of Alzheimer's disease. J Neurochem. 2009;111(6):1275–1308. doi: 10.1111/j.1471-4159.2009.06408.x.
    1. Breckenridge WC, Morgan IG, Zanetta JP, Vincendon G. Adult rat brain synaptic vesicles. II. Lipid composition. Biochimica et biophysica acta. 1973;320(3):681–686.
    1. Lohner K, Balgavy P, Hermetter A, Paltauf F, Laggner P. Stabilization of non-bilayer structures by the etherlipid ethanolamine plasmalogen. Biochimica et biophysica acta. 1991;1061(2):132–140. doi: 10.1016/0005-2736(91)90277-F.
    1. Glaser PE, Gross RW. Rapid plasmenylethanolamine-selective fusion of membrane bilayers catalyzed by an isoform of glyceraldehyde-3-phosphate dehydrogenase: discrimination between glycolytic and fusogenic roles of individual isoforms. Biochemistry. 1995;34(38):12193–12203. doi: 10.1021/bi00038a013.
    1. Bick RJ, Youker KA, Pownall HJ, Van Winkle WB, Entman ML. Unsaturated aminophospholipids are preferentially retained by the fast skeletal muscle CaATPase during detergent solubilization. Evidence for a specific association between aminophospholipids and the calcium pump protein. Archives of biochemistry and biophysics. 1991;286(2):346–352. doi: 10.1016/0003-9861(91)90050-S.
    1. Duhm J, Engelmann B, Schonthier UM, Streich S. Accelerated maximal velocity of the red blood cell Na+/K+ pump in hyperlipidemia is related to increase in 1-palmitoyl,2-arachidonoyl-plasmalogen phosphatidylethanolamine. Biochimica et biophysica acta. 1993;1149(1):185–188. doi: 10.1016/0005-2736(93)90040-7.
    1. Ford DA, Hale CC. Plasmalogen and anionic phospholipid dependence of the cardiac sarcolemmal sodium-calcium exchanger. FEBS letters. 1996;394(1):99–102. doi: 10.1016/0014-5793(96)00930-1.
    1. Nakayama R, Saito K. Presence of 1-O-alk-1'-enyl-2-O-acetyl glycerophosphocholine (vinyl form of PAF) in perfused rat and guinea pig hearts. Journal of biochemistry. 1989;105(4):494–496.
    1. Chao W, Olson MS. Platelet-activating factor: receptors and signal transduction. The Biochemical journal. 1993;292(Pt 3):617–629.
    1. Engelmann B, Brautigam C, Thiery J. Plasmalogen phospholipids as potential protectors against lipid peroxidation of low density lipoproteins. Biochemical and biophysical research communications. 1994;204(3):1235–1242. doi: 10.1006/bbrc.1994.2595.
    1. Brosche T, Platt D. The biological significance of plasmalogens in defense against oxidative damage. Experimental gerontology. 1998;33(5):363–369. doi: 10.1016/S0531-5565(98)00014-X.
    1. Kuczynski B, Reo NV. Evidence that plasmalogen is protective against oxidative stress in the rat brain. Neurochemical research. 2006;31(5):639–656. doi: 10.1007/s11064-006-9061-7.
    1. Zoeller RA, Grazia TJ, LaCamera P, Park J, Gaposchkin DP, Farber HW. Increasing plasmalogen levels protects human endothelial cells during hypoxia. American journal of physiology. 2002;283(2):H671–679.
    1. Farooqui AA, Rapoport SI, Horrocks LA. Membrane phospholipid alterations in Alzheimer's disease: deficiency of ethanolamine plasmalogens. Neurochemical research. 1997;22(4):523–527. doi: 10.1023/A:1027380331807.
    1. Guan Z, Wang Y, Cairns NJ, Lantos PL, Dallner G, Sindelar PJ. Decrease and structural modifications of phosphatidylethanolamine plasmalogen in the brain with Alzheimer disease. Journal of neuropathology and experimental neurology. 1999;58(7):740–747. doi: 10.1097/00005072-199907000-00008.
    1. Heymans HS, Schutgens RB, Tan R, van den Bosch H, Borst P. Severe plasmalogen deficiency in tissues of infants without peroxisomes (Zellweger syndrome) Nature. 1983;306(5938):69–70. doi: 10.1038/306069a0.
    1. Murphy EJ, Schapiro MB, Rapoport SI, Shetty HU. Phospholipid composition and levels are altered in Down syndrome brain. Brain research. 2000;867(1-2):9–18. doi: 10.1016/S0006-8993(00)02205-8.
    1. Schedin S, Sindelar PJ, Pentchev P, Brunk U, Dallner G. Peroxisomal impairment in Niemann-Pick type C disease. The Journal of biological chemistry. 1997;272(10):6245–6251. doi: 10.1074/jbc.272.10.6245.
    1. van den Bosch H, Schrakamp G, Hardeman D, Zomer AW, Wanders RJ, Schutgens RB. Ether lipid synthesis and its deficiency in peroxisomal disorders. Biochimie. 1993;75(3-4):183–189. doi: 10.1016/0300-9084(93)90076-5.
    1. Wells K, Farooqui AA, Liss L, Horrocks LA. Neural membrane phospholipids in Alzheimer disease. Neurochemical research. 1995;20(11):1329–1333. doi: 10.1007/BF00992508.
    1. Ginsberg L, Rafique S, Xuereb JH, Rapoport SI, Gershfeld NL. Disease and anatomic specificity of ethanolamine plasmalogen deficiency in Alzheimer's disease brain. Brain research. 1995;698(1-2):223–226. doi: 10.1016/0006-8993(95)00931-F.
    1. Han X, Holtzman DM, McKeel DW Jr. Plasmalogen deficiency in early Alzheimer's disease subjects and in animal models: molecular characterization using electrospray ionization mass spectrometry. J Neurochem. 2001;77(4):1168–1180. doi: 10.1046/j.1471-4159.2001.00332.x.
    1. Goodenowe DB, Cook LL, Liu J, Lu Y, Jayasinghe DA, Ahiahonu PW, Heath D, Yamazaki Y, Flax J, Krenitsky KF. Peripheral ethanolamine plasmalogen deficiency: a logical causative factor in Alzheimer's disease and dementia. Journal of lipid research. 2007;48(11):2485–2498. doi: 10.1194/jlr.P700023-JLR200.
    1. Stenvinkel P, Diczfalusy U, Lindholm B, Heimburger O. Phospholipid plasmalogen, a surrogate marker of oxidative stress, is associated with increased cardiovascular mortality in patients on renal replacement therapy. Nephrol Dial Transplant. 2004;19(4):972–976. doi: 10.1093/ndt/gfh035.
    1. Goodenowe DB. Methods for the diagnosis and risk assessment of plasmalogen deficiency mediated diseases of aging. 2008. In., G01N 33/483 (2006.01) edn. US PCT/CA2008/000659.
    1. Horrocks LA, VanRollins M, Yates AJ. The Molecular Basis of NeuroPathology. London; 1981.
    1. Maeba R, Maeda T, Kinoshita M, Takao K, Takenaka H, Kusano J, Yoshimura N, Takeoka Y, Yasuda D, Okazaki T. Plasmalogens in human serum positively correlate with high- density lipoprotein and decrease with aging. Journal of atherosclerosis and thrombosis. 2007;14(1):12–18.
    1. Rouser G, Yamamoto A. Curvilinear regression course of human brain lipid composition changes with age. Lipids. 1968;3(3):284–287. doi: 10.1007/BF02531202.
    1. Engelmann B, Streich S, Schonthier UM, Richter WO, Duhm J. Changes of membrane phospholipid composition of human erythrocytes in hyperlipidemias. I. Increased phosphatidylcholine and reduced sphingomyelin in patients with elevated levels of triacylglycerol-rich lipoproteins. Biochimica et biophysica acta. 1992;1165(1):32–37.
    1. Mandel H, Sharf R, Berant M, Wanders RJ, Vreken P, Aviram M. Plasmalogen phospholipids are involved in HDL-mediated cholesterol efflux: insights from investigations with plasmalogen-deficient cells. Biochemical and biophysical research communications. 1998;250(2):369–373. doi: 10.1006/bbrc.1998.9321.
    1. Munn NJ, Arnio E, Liu D, Zoeller RA, Liscum L. Deficiency in ethanolamine plasmalogen leads to altered cholesterol transport. Journal of lipid research. 2003;44(1):182–192. doi: 10.1194/jlr.M200363-JLR200.
    1. Bhatia SK, Hajdu J. Stereospecific Synthesis of Ether and Thioether Phospholipids. The Use of L-Glyceric Acid as a Chiral Phospholipid Precursor. J Org Chem. 1988;53:5034–5039. doi: 10.1021/jo00256a024.
    1. Sahai P, Vishwakarma RA. Phospholipase-A2-mediated stereoselective synthesis of (R)-1-O-alkylglycero-3-phosphate and alkyl-acyl analogues: application for synthesis of radiolabelled biosynthetic precursors of cell surface glycoconjugates of Leishmania donovani. J Chem Soc, Perkin Trans. 1997;1:1845–1849. doi: 10.1039/a700353f.
    1. Halldorsson A, Thordarson P, Kristinsson B, Magnusson C, Haraldsson GG. Lipase-catalyzed kinetic resolution of 1-O-alkylglycerols by sequential transesterification. Tetrahedron Asymmerty. 2004;15:2893–2899. doi: 10.1016/j.tetasy.2004.07.046.
    1. Bartolmas T, Heyn T, Mickeleit M, Fischer A, Reutter W, Danker K. Glucosamine-glycerophospholipids that activate cell-matrix adhesion and migration. Journal of medicinal chemistry. 2005;48(21):6750–6755. doi: 10.1021/jm050558n.
    1. Ramesha CS, Pickett WC, Murthy DV. Sensitive method for the analysis of phospholipid subclasses and molecular species as 1-anthroyl derivatives of their diglycerides. Journal of chromatography. 1989;491(1):37–48.
    1. Yashunsky DV, Borodkin VS, Ferguson MA, Nikolaev AV. The chemical synthesis of bioactive glycosylphosphatidylinositols from Trypanosoma cruzi containing an unsaturated fatty acid in the lipid. Angewandte Chemie (International ed) 2006;45(3):468–474. doi: 10.1002/anie.200502779.
    1. James PF, Lake AC, Hajra AK, Larkins LK, Robinson M, Buchanan FG, Zoeller RA. An animal cell mutant with a deficiency in acyl/alkyl-dihydroxyacetone-phosphate reductase activity. Effects on the biosynthesis of ether-linked and diacyl glycerolipids. The Journal of biological chemistry. 1997;272(38):23540–23546. doi: 10.1074/jbc.272.38.23540.
    1. Flint OP, Masters BA, Gregg RE, Durham SK. HMG CoA reductase inhibitor-induced myotoxicity: pravastatin and lovastatin inhibit the geranylgeranylation of low-molecular-weight proteins in neonatal rat muscle cell culture. Toxicology and applied pharmacology. 1997;145(1):99–110. doi: 10.1006/taap.1997.8174.
    1. Muzio G, Martinasso G, Trombetta A, Di Simone D, Canuto RA, Maggiora M. HMG-CoA reductase and PPARalpha are involved in clofibrate-induced apoptosis in human keratinocytes. Apoptosis. 2006;11(2):265–275. doi: 10.1007/s10495-006-3559-y.
    1. Yang YC, Ho TC, Chen SL, Lai HY, Wu JY, Tsao YP. Inhibition of cell motility by troglitazone in human ovarian carcinoma cell line. BMC cancer. 2007;7:216. doi: 10.1186/1471-2407-7-216.
    1. Liu D, Nagan N, Just WW, Rodemer C, Thai TP, Zoeller RA. Role of dihydroxyacetonephosphate acyltransferase in the biosynthesis of plasmalogens and nonether glycerolipids. Journal of lipid research. 2005;46(4):727–735. doi: 10.1194/jlr.M400364-JLR200.
    1. Nagan N, Hajra AK, Larkins LK, Lazarow P, Purdue PE, Rizzo WB, Zoeller RA. Isolation of a Chinese hamster fibroblast variant defective in dihydroxyacetonephosphate acyltransferase activity and plasmalogen biosynthesis: use of a novel two-step selection protocol. The Biochemical journal. 1998;332(Pt 1):273–279.
    1. Feulgen R, Voit K. Uber einen weiterbreiteten fasten Aldehyd. Arch Physiol. 1924;206:217–245.
    1. Klenk E, Debuch H. [Information on acetalphosphatides.] Hoppe-Seyler's Zeitschrift fur physiologische Chemie. 1954;296(3-4):179–188.
    1. Rapport MM, Franzl RE. The structure of plasmalogens. III. The nature and significance of the aldehydogenic linkage. J Neurochem. 1957;1(4):303–310. doi: 10.1111/j.1471-4159.1957.tb12086.x.
    1. Chakravarthy BR, Spence MW, Clarke JT, Cook HW. Rapid isolation of neuroblastoma plasma membranes on Percoll gradients. Characterization and lipid composition. Biochimica et biophysica acta. 1985;812(1):223–233. doi: 10.1016/0005-2736(85)90542-5.
    1. Deshmukh DS, Vorbrodt AW, Lee PK, Bear WD, Kuizon S. Studies on the submicrosomal fractions of bovine oligodendroglia: lipid composition and glycolipid biosynthesis. Neurochemical research. 1988;13(6):571–582. doi: 10.1007/BF00973300.
    1. Lange Y, Strebel F, Steck TL. Role of the plasma membrane in cholesterol esterification in rat hepatoma cells. The Journal of biological chemistry. 1993;268(19):13838–13843.
    1. Spohn M, Davison AN. Cholesterol metabolism in myelin and other subcellular fractions of rat brain. Journal of lipid research. 1972;13(5):563–570.
    1. Hill SA, McQueen MJ. Reverse cholesterol transport--a review of the process and its clinical implications. Clinical biochemistry. 1997;30(7):517–525. doi: 10.1016/S0009-9120(97)00098-2.
    1. Rothblat GH, Mahlberg FH, Johnson WJ, Phillips MC. Apolipoproteins, membrane cholesterol domains, and the regulation of cholesterol efflux. Journal of lipid research. 1992;33(8):1091–1097.
    1. Aviram M, Bierman EL, Oram JF. High density lipoprotein stimulates sterol translocation between intracellular and plasma membrane pools in human monocyte-derived macrophages. Journal of lipid research. 1989;30(1):65–76.
    1. Tabas I, Rosoff WJ, Boykow GC. Acyl coenzyme A:cholesterol acyl transferase in macrophages utilizes a cellular pool of cholesterol oxidase-accessible cholesterol as substrate. The Journal of biological chemistry. 1988;263(3):1266–1272.
    1. Holub BJ. Docosahexaenoic acid (DHA) and cardiovascular disease risk factors. Prostaglandins, leukotrienes, and essential fatty acids. 2009;81(2-3):199–204. doi: 10.1016/j.plefa.2009.05.016.
    1. Nissen SE, Tuzcu EM, Brewer HB, Sipahi I, Nicholls SJ, Ganz P, Schoenhagen P, Waters DD, Pepine CJ, Crowe TD. Effect of ACAT inhibition on the progression of coronary atherosclerosis. The New England journal of medicine. 2006;354(12):1253–1263. doi: 10.1056/NEJMoa054699.
    1. Tardif JC, Gregoire J, L'Allier PL, Anderson TJ, Bertrand O, Reeves F, Title LM, Alfonso F, Schampaert E, Hassan A. Effects of the acyl coenzyme A:cholesterol acyltransferase inhibitor avasimibe on human atherosclerotic lesions. Circulation. 2004;110(21):3372–3377. doi: 10.1161/01.CIR.0000147777.12010.EF.
    1. Meuwese MC, de Groot E, Duivenvoorden R, Trip MD, Ose L, Maritz FJ, Basart DC, Kastelein JJ, Habib R, Davidson MH. ACAT inhibition and progression of carotid atherosclerosis in patients with familial hypercholesterolemia: the CAPTIVATE randomized trial. Jama. 2009;301(11):1131–1139. doi: 10.1001/jama.301.11.1131.

Source: PubMed

3
Subskrybuj