Structural Elucidation of Irish Organic Farmed Salmon (Salmo salar) Polar Lipids with Antithrombotic Activities

Alexandros Tsoupras, Ronan Lordan, Martina Demuru, Katie Shiels, Sushanta Kumar Saha, Constantina Nasopoulou, Ioannis Zabetakis, Alexandros Tsoupras, Ronan Lordan, Martina Demuru, Katie Shiels, Sushanta Kumar Saha, Constantina Nasopoulou, Ioannis Zabetakis

Abstract

While several marine polar lipids (PL) have exhibited cardioprotective properties through their effects on the platelet-activating factor (PAF) pathways, salmon PL have not been tested so far. In this study, the antithrombotic activities of salmon PL were assessed in human platelets and the structural characterisation of bioactive salmon PL was performed by GC-MS and LC-MS analyses. PL from fillets of Irish organic farmed salmon (Salmo salar) were extracted and separated into several lipid subclasses by thin-layer chromatography (TLC), while their fatty acid profile was fully characterised by GC-MS. Salmon total lipids (TL), total neutral lipids (TNL), total polar lipids (TPL), and each PL subclass obtained by TLC were further assessed for their in vitro effects towards PAF-induced and thrombin-induced platelet aggregation in human platelets. Salmon PL exhibited antithrombotic effects on human platelet aggregation, mostly through their strong inhibitory effects against the PAF pathway with IC50 values comparable to other marine PL, but with lower effects towards the thrombin pathway. PL fractions corresponding to phosphatidylcholine and phosphatidylethanolamine derivatives exhibited the most potent anti-PAF effects, while LC-MS analysis putatively elucidated their structure/function relationship. Several diacyl-PC/PE and alkyl-acyl-PC/PE species containing mostly docosahexaenoic acid at their sn-2 glycerol-backbone may be responsible for the bioactivity. The data presented suggests that salmon contains PL with strong antithrombotic bioactivities.

Keywords: GC-MS; LC-MS; antithrombotic; docosahexaenoic acid (DHA); phosphatidylcholine; phosphatidylethanolamine; platelet aggregation; platelet-activating factor (PAF); polar lipids; polyunsaturated fatty acids (PUFA); salmon; thrombin.

Conflict of interest statement

The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
TLC analysis of the TPL of Irish organic farmed salmon fillets and the biological activity of each PL subclass towards PAF-induced platelet aggregation in hPRP. (A): Typical profile of the PL separation of salmon fillet on preparative TLC (Silica G). The elution system used for the separation of the TPL was chloroform:methanol:water 65:35:6 (v/v/v). PL: Polar lipids; E: mixture of standard PL from egg; Lyso-PC: lyso-phosphatidylcholine; SM: sphingomyelin; PC: phosphatidylcholine; Lyso-PE: lyso-phosphatidylethanolamine; PE: phosphatidylethanolamine; CL: cardiolipin; TLC; thin-layer chromatography. (B): Biological activities of the PL subclasses of the TLC bands towards PAF-induced platelet aggregation in hPRP. The results reflect the inhibitory strength of each lipid sample and are expressed as mean values of IC50 (μg of lipids in the aggregometer cuvette that causes 50% inhibition of PAF-induced aggregation of hPRP); The low IC50 value indicates strong inhibition of PAF-induced aggregation of hPRP; Lipid fractions of TLC bands 1 and 4 (corresponding to Lyso-PC and Lyso-PE) did not exhibit these bioactivities. hPRP: human platelet-rich plasma; PAF: platelet-activating factor; PL: polar lipids; TPL: total polar lipids.
Figure 2
Figure 2
A representative chart of the 50% inhibitory effect of Irish organic farmed salmon fillet PL towards PAF-induced platelet aggregation in hPRP. Trace 1 (blue) in both A and B demonstrate the maximum-reversible aggregation of hPRP induced by 2.6 × 10−8 M of PAF; Trace 2 (black) demonstrates the effect of preincubation of hPRP with the appropriate concentration (IC50 value) of salmon TPL (A) and the corresponding TLC Band of PC (B), which causes 50% inhibition of PAF-induced hPRP aggregation. PAF: platelet-activating factor; hPRP: human platelet-rich plasma; TPL: total polar lipids; PC: phosphatidylcholine.
Figure 3
Figure 3
Representative HPLC chromatograms of salmon PL LC-MS analysis. (A) Depicts a representative chromatogram of the TLC fraction corresponding to salmon phosphatidylethanolamine (PE) derivatives, whereas (B) depicts a representative chromatogram of the TLC fraction corresponding to salmon phosphatidylcholine (PC) derivatives. Analysis was performed using a HPLC (Agilent 1260 series) equipped with Q-TOF mass spectrometer (Agilent 6520). The column used for the separations was an Agilent C18 Poroshell 120 column (2.7 µm, 3.0 × 150 mm). The composition of mobile phase (A) was 2 mM ammonium acetate in water and 2 mM ammonium acetate in 95% acetonitrile for mobile phase (B). Chromatographic separation was performed by gradient elution starting with 60% B for 1 min, then increasing to 90% B over 2.5 min. Subsequently, 90% B was held for 1.5 min and increased afterward to 100% over 5 min. Then, 100% B was held for 4 min, reducing afterward to 60% B over 0.5 min and hold for 1 min until the next run. The mobile phase flow rate was 0.3 mL/min until 5 min elapsed, increasing up to 0.6 mL/min until 10 min had elapsed and held at this flow until the end of the run. The injection volume was 10 μL.
Figure 4
Figure 4
Representative mass spectra of PC species present in the relative TLC fraction of bioactive salmon PC. The LC-MS analysis demonstrated that the TLC fraction of the bioactive salmon PC contains a mixture of several PC species. (AC) Mass spectra depicting the most abundant PC molecules eluted at peaks 1, 2, and 3 of the LC-MS analysis, respectively. The most probable and proposed identities of the PC species for each acquired m/z values were verified using the LIPID MAPS: Nature Lipidomics Gateway (www.lipidmaps.org), by using the lowest delta values during identification, in combination with their content of FA that was acquired by both GC-MS and LC-MS analyses. In all 3 peaks, several PC molecules are proposed baring either EPA or DHA in the sn-2 position of the glycerol backbone, whereas lower but considerable quantities of alkyl-acyl moieties also seem to be present in all spectra (AC) that are outlined in squares. Interestingly, at peak 3c (elution time 3.2 min), a 1-O-alkyl (18:0)-2-sn-akyl (22:6, DHA)-3-glycerophosphocholine seems to be present in the TLC fraction of the bioactive salmon PC. FA: fatty acids; EPA: eicosapentaenoic acid; DHA: docosahexaenoic acid; PC: phosphatidylcholine.
Figure 4
Figure 4
Representative mass spectra of PC species present in the relative TLC fraction of bioactive salmon PC. The LC-MS analysis demonstrated that the TLC fraction of the bioactive salmon PC contains a mixture of several PC species. (AC) Mass spectra depicting the most abundant PC molecules eluted at peaks 1, 2, and 3 of the LC-MS analysis, respectively. The most probable and proposed identities of the PC species for each acquired m/z values were verified using the LIPID MAPS: Nature Lipidomics Gateway (www.lipidmaps.org), by using the lowest delta values during identification, in combination with their content of FA that was acquired by both GC-MS and LC-MS analyses. In all 3 peaks, several PC molecules are proposed baring either EPA or DHA in the sn-2 position of the glycerol backbone, whereas lower but considerable quantities of alkyl-acyl moieties also seem to be present in all spectra (AC) that are outlined in squares. Interestingly, at peak 3c (elution time 3.2 min), a 1-O-alkyl (18:0)-2-sn-akyl (22:6, DHA)-3-glycerophosphocholine seems to be present in the TLC fraction of the bioactive salmon PC. FA: fatty acids; EPA: eicosapentaenoic acid; DHA: docosahexaenoic acid; PC: phosphatidylcholine.
Figure 5
Figure 5
Representative mass spectra of PE species present in the relative TLC fraction of bioactive salmon PE. The LC-MS analysis showed that the TLC fraction of bioactive salmon PE contains a mixture of several PE species. (AC) Mass spectra depicting the most abundant PE molecules eluted at peaks 1, 3a, and 3b of the LC-MS analysis, respectively. The most probable and proposed identities of PE species for each acquired m/z values were verified using the LIPID MAPS: Nature Lipidomics Gateway (www.lipidmaps.org), by using the lowest delta values during identification, in combination with their content in FA that was acquired by both GC-MS and LC-MS analyses. In all 3 peaks, several PE molecules are proposed with DHA in the sn-2 position of the glycerol backbone, whereas lower but considerable quantities of alkyl-acyl moieties seem to be present only at B and C spectra of the PE molecules eluted at peak 3, which are outlined in squares. FA: fatty acids; EPA: eicosapentaenoic acid; DHA: docosahexaenoic acid; PE: phosphatidylethanolamine.
Figure 5
Figure 5
Representative mass spectra of PE species present in the relative TLC fraction of bioactive salmon PE. The LC-MS analysis showed that the TLC fraction of bioactive salmon PE contains a mixture of several PE species. (AC) Mass spectra depicting the most abundant PE molecules eluted at peaks 1, 3a, and 3b of the LC-MS analysis, respectively. The most probable and proposed identities of PE species for each acquired m/z values were verified using the LIPID MAPS: Nature Lipidomics Gateway (www.lipidmaps.org), by using the lowest delta values during identification, in combination with their content in FA that was acquired by both GC-MS and LC-MS analyses. In all 3 peaks, several PE molecules are proposed with DHA in the sn-2 position of the glycerol backbone, whereas lower but considerable quantities of alkyl-acyl moieties seem to be present only at B and C spectra of the PE molecules eluted at peak 3, which are outlined in squares. FA: fatty acids; EPA: eicosapentaenoic acid; DHA: docosahexaenoic acid; PE: phosphatidylethanolamine.

References

    1. Mori T.A. Marine OMEGA-3 fatty acids in the prevention of cardiovascular disease. Fitoterapia. 2017;123:51–58. doi: 10.1016/j.fitote.2017.09.015.
    1. Simopoulos A.P. The importance of the omega-6/omega-3 fatty acid ratio in cardiovascular disease and other chronic diseases. Exp. Biol. Med. 2008;233:674–688. doi: 10.3181/0711-MR-311.
    1. Lordan R., Tsoupras A., Zabetakis I. Phospholipids of animal and marine origin: Structure, function, and anti-inflammatory properties. Molecules. 2017;22:1964. doi: 10.3390/molecules22111964.
    1. Nasopoulou C., Tsoupras A.B., Karantonis H.C., Demopoulos C.A., Zabetakis I. Fish polar lipids retard atherosclerosis in rabbits by down-regulating PAF biosynthesis and up-regulating PAF catabolism. Lipids Health Dis. 2011;10:213. doi: 10.1186/1476-511X-10-213.
    1. Tsoupras A.B., Iatrou C., Frangia C., Demopoulos C.A. The implication of platelet-activating factor in cancer growth and metastasis: Potent beneficial role of PAF-inhibitors and antioxidants. Infect. Disord. Drug Targets. 2009;9:390–399. doi: 10.2174/187152609788922555.
    1. Tsoupras A.B., Chini M., Tsogas N., Fragopoulou E., Nomikos T., Lioni A., Mangafas N., Demopoulos C.A., Antonopoulou S., Lazanas M.C. Anti-platelet-activating factor effects of highly active antiretroviral therapy (HAART): A new insight in the drug therapy of HIV infection? AIDS Res. Hum. Retrovir. 2008;24:1079–1086. doi: 10.1089/aid.2007.0263.
    1. Demopoulos C.A., Karantonis H.C., Antonopoulou S. Platelet-activating factor—A molecular link between atherosclerosis theories. Eur. J. Lipid Sci. Technol. 2003;105:705–716. doi: 10.1002/ejlt.200300845.
    1. Tsoupras A., Lordan R., Zabetakis I. Inflammation, not cholesterol, is a cause of chronic disease. Nutrients. 2018;10:604. doi: 10.3390/nu10050604.
    1. Lordan R., Zabetakis I. Invited review: The anti-inflammatory properties of dairy lipids. J. Dairy Sci. 2017;100:4197–4212. doi: 10.3168/jds.2016-12224.
    1. Lordan R., Tsoupras A., Mitra B., Zabetakis I. Dairy fats and cardiovascular disease: Do we really need to be concerned? Foods. 2018;7:29. doi: 10.3390/foods7030029.
    1. Megson I.L., Whitfield P.D., Zabetakis I. Lipids and cardiovascular disease: Where does dietary intervention sit alongside statin therapy? Food Funct. 2016;7:2603–2614. doi: 10.1039/C6FO00024J.
    1. Zabetakis I. Food security and cardioprotection: The polar lipid link. J. Food Sci. 2013;78:1101–1104. doi: 10.1111/1750-3841.12194.
    1. Nasopoulou C., Karantonis H.C., Perrea D.N., Theocharis S.E., Iliopoulos D.G., Demopoulos C.A., Zabetakis I. In vivo anti-atherogenic properties of cultured gilthead sea bream (Sparus aurata) polar lipid extracts in hypercholesterolaemic rabbits. Food Chem. 2010;120:831–836. doi: 10.1016/j.foodchem.2009.11.023.
    1. Sioriki E., Nasopoulou C., Demopoulos C.A., Zabetakis I. Comparison of sensory and cardioprotective properties of olive-pomace enriched and conventional gilthead sea bream (Sparus aurata): The effect of grilling. J. Aquat. Food Prod. Technol. 2015;24:782–795. doi: 10.1080/10498850.2013.813100.
    1. Nasopoulou C., Smith T., Detopoulou M., Tsikrika C., Papaharisis L., Barkas D., Zabetakis I. Structural elucidation of olive pomace fed sea bass (Dicentrarchus labrax) polar lipids with cardioprotective activities. Food Chem. 2014;145:1097–1105. doi: 10.1016/j.foodchem.2013.08.091.
    1. Nasopoulou C., Psani E., Sioriki E., Demopoulos C.A., Zabetakis I. Evaluation of sensory and in vitro cardio protective properties of sardine (Sardina pilchardus): The effect of grilling and brining. Food Nutr. Sci. 2013;4:940–949.
    1. Nasopoulou C., Nomikos T., Demopoulos C., Zabetakis I. Comparison of antiatherogenic properties of lipids obtained from wild and cultured sea bass (Dicentrarchus labrax) and gilthead sea bream (Sparus aurata) Food Chem. 2007;100:560–567. doi: 10.1016/j.foodchem.2005.09.074.
    1. Panayiotou A., Samartzis D., Nomikos T., Fragopoulou E., Karantonis H.C., Demopoulos C.A., Zabetakis I. Lipid fractions with aggregatory and antiaggregatory activity toward platelets in fresh and fried cod (Gadus morhua): Correlation with platelet-activating factor and atherogenesis. J. Agric. Food Chem. 2000;48:6372–6379. doi: 10.1021/jf000701f.
    1. Rementzis J., Antonopoulou S., Argyropoulos D., Demopoulos C.A. Platelet-Activating Factor and Related Lipid Mediators 2. Springer; New York, NY, USA: 1996. Biologically active lipids from S. scombrus; pp. 65–72.
    1. Fragopoulou E., Nomikos T., Tsantila N., Mitropoulou A., Zabetakis I., Demopoulos C.A. Biological activity of total lipids from red and white wine/must. J. Agric. Food Chem. 2001;49:5186–5193. doi: 10.1021/jf0106392.
    1. Fragopoulou E., Antonopoulou S., Tsoupras A., Tsantila N., Grypioti A., Gribilas G., Gritzapi H., Konsta E., Skandalou E., Papadopoulou A. Antiatherogenic properties of red/white wine, musts, grape-skins, and yeast; Proceedings of the 45th International Conference on the Bioscience of Lipids, University of Ioannina; Ioannina, Greece. 25–29 May 2004; p. 66.
    1. Megalemou K., Sioriki E., Lordan R., Dermiki M., Nasopoulou C., Zabetakis I. Evaluation of sensory and in vitro anti-thrombotic properties of traditional greek yogurts derived from different types of milk. Heliyon. 2017;3:e00227. doi: 10.1016/j.heliyon.2016.e00227.
    1. Berdeaux O., Juaneda P., Martine L., Cabaret S., Bretillon L., Acar N. Identification and quantification of phosphatidylcholines containing very-long-chain polyunsaturated fatty acid in bovine and human retina using liquid chromatography/tandem mass spectrometry. J. Chromatogr. A. 2010;1217:7738–7748. doi: 10.1016/j.chroma.2010.10.039.
    1. Zabetakis I. Marine Oils (From Sea to Pharmaceuticals) Nova Science; Hauppauge, NY, USA: 2015.
    1. Raatz S.K., Johnson L.K., Rosenberger T.A., Picklo M.J. Twice weekly intake of farmed atlantic salmon (Salmo salar) positively influences lipoprotein concentration and particle size in overweight men and women. Nutr. Res. 2016;36:899–906. doi: 10.1016/j.nutres.2016.06.011.
    1. Zhang J., Wang C., Li L., Man Q., Song P., Meng L., Du Z.Y., Frøyland L. Inclusion of atlantic salmon in the chinese diet reduces cardiovascular disease risk markers in dyslipidemic adult men. Nutr. Res. 2010;30:447–454. doi: 10.1016/j.nutres.2010.06.010.
    1. Hagen I.V., Helland A., Bratlie M., Brokstad K.A., Rosenlund G., Sveier H., Mellgren G., Gudbrandsen O.A. High intake of fatty fish, but not of lean fish, affects serum concentrations of TAG and HDL-cholesterol in healthy, normal-weight adults: A randomised trial. Br. J. Nutr. 2016;116:648–657. doi: 10.1017/S0007114516002555.
    1. Dalum A., Tangen R., Falk K., Hordvik I., Rosenlund G., Torstensen B., Koppang E.O. Coronary changes in the atlantic salmon Salmo salar L.: Characterization and impact of dietary fatty acid compositions. J. Fish Dis. 2016;39:41–54. doi: 10.1111/jfd.12321.
    1. Raatz S.K., Golovko M.Y., Brose S.A., Rosenberger T.A., Burr G.S., Wolters W.R., Picklo M.J. Baking reduces prostaglandin, resolvin, and hydroxy-fatty acid content of farm-raised atlantic salmon (Salmo salar) J. Agric. Food Chem. 2011;59:11278–11286. doi: 10.1021/jf202576k.
    1. McLennan P.L., Owen A.J., Slee E.L., Theiss M.L. Myocardial function, ischaemia and n-3 polyunsaturated fatty acids: A membrane basis. Eur. J. Cardiovasc. Med. 2007;8:S15–S18. doi: 10.2459/01.JCM.0000289272.87803.ce.
    1. Rizos E.C., Ntzani E.E., Bika E., Kostapanos M.S., Elisaf M.S. Association between omega-3 fatty acid supplementation and risk of major cardiovascular disease events: A systematic review and meta-analysis. JAMA. 2012;308:1024–1033. doi: 10.1001/2012.jama.11374.
    1. Enns J.E., Yeganeh A., Zarychanski R., Abou-Setta A.M., Friesen C., Zahradka P., Taylor C.G. The impact of omega-3 polyunsaturated fatty acid supplementation on the incidence of cardiovascular events and complications in peripheral arterial disease: A systematic review and meta-analysis. BMC Cardiovasc. Disord. 2014;14:70. doi: 10.1186/1471-2261-14-70.
    1. Kwak S., Myung S., Lee Y., Seo H. Efficacy of omega-3 fatty acid supplements (eicosapentaenoic acid and docosahexaenoic acid) in the secondary prevention of cardiovascular disease: A meta-analysis of randomized, double-blind, placebo-controlled trials. Arch. Intern. Med. 2012;172:686–694.
    1. Walz C.P., Barry A.R., Koshman S.L. Omega-3 polyunsaturated fatty acid supplementation in the prevention of cardiovascular disease. Can. Pharm. J. 2016;149:166–173. doi: 10.1177/1715163516640812.
    1. Chowdhury R., Stevens S., Gorman D., Pan A., Warnakula S., Chowdhury S., Ward H., Johnson L., Crowe F., Hu F.B. Association between fish consumption, long chain omega-3 fatty acids, and risk of cerebrovascular disease: Systematic review and meta-analysis. BMJ. 2012;345:e6698. doi: 10.1136/bmj.e6698.
    1. Yanagita T., Nagao K. Functional lipids and the prevention of the metabolic syndrome. Asia Pac. J. Clin. Nutr. 2008;17:189–191. doi: 10.4010/jln.24.61.
    1. Küllenberg D., Taylor L.A., Schneider M., Massing U. Health effects of dietary phospholipids. Lipids Health Dis. 2012;11:3. doi: 10.1186/1476-511X-11-3.
    1. Verouti S.N., Tsoupras A.B., Alevizopoulou F., Demopoulos C.A., Iatrou C. Paricalcitol effects on activities and metabolism of platelet activating factor and on inflammatory cytokines in hemodialysis patients. Int. J. Artif. Organs. 2013;36:87–96. doi: 10.5301/ijao.5000187.
    1. Tsoupras A.B., Chini M., Mangafas N., Tsogas N., Stamatakis G., Tsantila N., Fragopoulou E., Antonopoulou S., Gargalianos P., Demopoulos C.A. Platelet-activating factor and its basic metabolic enzymes in blood of naive HIV-infected patients. Angiology. 2012;63:343–352. doi: 10.1177/0003319711420608.
    1. Ruiz-Lopez N., Stubhaug I., Ipharraguerre I., Rimbach G., Menoyo D. Positional distribution of fatty acids in triacylglycerols and phospholipids from fillets of atlantic salmon (Salmo salar) fed vegetable and fish oil blends. Mar. Drugs. 2015;13:4255–4269. doi: 10.3390/md13074255.
    1. Beppu F., Yasuda K., Okada A., Hirosaki Y., Okazaki M., Gotoh N. Comparison of the distribution of unsaturated fatty acids at the sn-2 position of phospholipids and triacylglycerols in marine fishes and mammals. J. Oleo Sci. 2017;66:1217–1227. doi: 10.5650/jos.ess17132.
    1. Peng J., Larondelle Y., Pham D., Ackman R.G., Rollin X. Polyunsaturated fatty acid profiles of whole body phospholipids and triacylglycerols in anadromous and landlocked atlantic salmon (Salmo salar L.) fry. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 2003;134:335–348. doi: 10.1016/S1096-4959(02)00263-4.
    1. Simopoulos A. An increase in the omega-6/omega-3 fatty acid ratio increases the risk for obesity. Nutrients. 2016;8:128. doi: 10.3390/nu8030128.
    1. Yeung E.H., Robledo C., Boghossian N., Zhang C., Mendola P. Developmental origins of cardiovascular disease. Curr. Epidemiol. Rep. 2014;1:9–16. doi: 10.1007/s40471-014-0006-4.
    1. O’Flaherty J.T., Tessner T., Greene D., Redman J.R., Wykle R.L. Comparison of 1-O-alkyl-, 1-O-alk-1′-enyl-, and 1-O-acyl-2-acetyl-sn-glycero-3-phosphoethanolamines and -3-phosphocholines as agonists of the platelet-activating factor family. Biochim. Biophys. Acta. 1994;1210:209–216. doi: 10.1016/0005-2760(94)90123-6.
    1. Tsantila N., Karantonis H.C., Perrea D.N., Theocharis S.E., Iliopoulos D.G., Antonopoulou S., Demopoulos C.A. Antithrombotic and antiatherosclerotic properties of olive oil and olive pomace polar extracts in rabbits. Mediat. Inflamm. 2007;2007:36204. doi: 10.1155/2007/36204.
    1. Hayashi H., Tanaka Y., Hibino H., Umeda Y., Kawamitsu H., Fujimoto H., Amakawa T. Beneficial effect of salmon roe phosphatidylcholine in chronic liver disease. Curr. Med. Res. Opin. 1999;15:177–184. doi: 10.1185/03007999909114089.
    1. Fukunaga K., Hossain Z., Takahashi K. Marine phosphatidylcholine suppresses 1,2-dimethylhydrazine-induced colon carcinogenesis in rats by inducing apoptosis. Nutr. Res. 2008;28:635–640. doi: 10.1016/j.nutres.2008.05.005.
    1. Hossain Z., Konishi M., Hosokawa M., Takahashi K. Effect of polyunsaturated fatty acid-enriched phosphatidylcholine and phosphatidylserine on butyrate-induced growth inhibition, differentiation and apoptosis in caco-2 cells. Cell Biochem. Funct. 2006;24:159–165. doi: 10.1002/cbf.1202.
    1. Bligh E.G., Dyer W.J. A rapid method of total lipid extraction and purification. Can. J. Biochem. Phys. 1959;37:911–917. doi: 10.1139/y59-099.
    1. Galanos D.S., Kapoulas V.M. Isolation of polar lipids from triglyceride mixtures. J. Lipid Res. 1962;3:134–136.
    1. Tsantila N., Tsoupras A.B., Fragopoulou E., Antonopoulou S., Iatrou C., Demopoulos C.A. In vitro and in vivo effects of statins on platelet-activating factor and its metabolism. Angiology. 2011;62:209–218. doi: 10.1177/0003319710375089.
    1. Nasopoulou C., Stamatakis G., Demopoulos C.A., Zabetakis I. Effects of olive pomace and olive pomace oil on growth performance, fatty acid composition and cardio protective properties of gilthead sea bream (Sparus aurata) and sea bass (Dicentrarchus labrax) Food Chem. 2011;129:1108–1113. doi: 10.1016/j.foodchem.2011.05.086.
    1. Otero P., Saha S.K., Gushin J.M., Moane S., Barron J., Murray P. Identification of optimum fatty acid extraction methods for two different microalgae phaeodactylum tricornutum and haematococcus pluvialis for food and biodiesel applications. Anal. Bioanal. Chem. 2017;409:4659–4667. doi: 10.1007/s00216-017-0412-9.
    1. Qandil A. Prodrugs of nonsteroidal anti-inflammatory drugs (NSAIDS), more than meets the eye: A critical review. Int. J. Mol. Sci. 2012;13:17244–17274. doi: 10.3390/ijms131217244.

Source: PubMed

3
Subscribe