Skip to content
Licensed Unlicensed Requires Authentication Published by De Gruyter September 21, 2011

A long and winding road: defining the biological role and clinical importance of paraoxonases

  • Richard W. James

Abstract

Paraoxonase-1 (PON1) is an enzyme belonging to a three-member gene family, each of which is highly conserved in mammalian evolution. Whilst there is consensus that the paraoxonase family members have a general protective influence, their precise biological role has remained elusive. A toxicological role, protecting from environmental poisoning by organophosphate derivatives, drove much of the earlier work on the enzymes. More recently, clinical interest has focused on a protective role in vascular disease via a hypothesised impact on lipoprotein lipid oxidation. Recent confirmation that the primary activity of the paraoxonases is that of a lactonase considerably expands the potential sources of biological substrates for the enzyme. Studies on such substrates may shed further light on different mechanisms by which paraoxonases beneficially influence atherosclerosis, as well as defining possible roles in limiting bacterial infection and in innate immunity.

Clin Chem Lab Med 2006;44:1052–9.


Corresponding author: Prof. R.W. James, Clinical Diabetes Unit, Service of Endocrinology, Diabetes and Nutrition, University Hospital, 24, rue Micheli-du-Crest, 1211 Geneva 14, Switzerland Phone: +41-22-3729304, Fax: +41-22-3729309,

References

1. Primo-Parmo SL, Sorenson RC, Teiber J, La Du BN. The human serum paraoxonase/arylesterase gene (PON1) is one member of a multigene family. Genomics 1996; 33:498–507.10.1006/geno.1996.0225Search in Google Scholar

2. Kobayashi M, Shinohara M, Sakoh C, Kataolsa M, Shimizu S. Lactone-ring-cleaving enzyme: genetic analysis, novel RNA editing, and evolutionary implications. Proc Natl Acad Sci USA 1998; 95:12787–92.10.1073/pnas.95.22.12787Search in Google Scholar

3. Ng CJ, Wadleigh DJ, Gangopadhyay A, Hama S, Grijalva VR, Navab M, et al. Paraoxonase-2 is a ubiquitously expressed protein with antioxidant properties and is capable of preventing cell-mediated oxidative modification of low density lipoprotein. J Biol Chem 2001; 276:44444–9.10.1074/jbc.M105660200Search in Google Scholar

4. Reddy ST, Wadleigh DJ, Grijalva V, Ng C, Hama S, Gangopadhyay A, et al. Human paraoxonase-3 is an HDL-associated enzyme with biological activity similar to paraoxonase-1 protein but is not regulated by oxidized lipids. Arterioscler Thromb Vasc Biol 2001; 21:542–7.10.1161/01.ATV.21.4.542Search in Google Scholar

5. Blatter M-C, James RW, Messmer S, Barja F, Pometta D. Identification of a distinct human high-density lipoprotein subspecies defined by a lipoprotein-associated protein, K-45. Identity of K-45 with paraoxonase. Eur J Biochem 1993; 211:871–9.Search in Google Scholar

6. Deakin S, Leviev I, Gomaraschi M, Calabresi L, Franceschini G, James RW. Enzymatically active paraoxonase-1 is located at the external membrane of producing cells and released by a high affinity, saturable, desorption mechanism. J Biol Chem 2002; 277:4301–8.10.1074/jbc.M107440200Search in Google Scholar

7. Mazur A. An enzyme in animal tissues capable of hydrolysing the phosphorus-fluorine bond of alkyl fluorophosphates. J Biol Chem 1946; 164:271–89.10.1016/S0021-9258(18)43068-2Search in Google Scholar

8. Aldridge WN, Reiner E. Serum esterase 1. Two types of esterase (A and B) hydrolysing p-nitrophenyl acetate, propionate and butyrate, and a method for their determination. Biochem J 1953; 53:227–9.Search in Google Scholar

9. Costa LG, Cole TB, Jarvik GP, Furlong CE. Functional genomic of the paraoxonase (PON1) polymorphisms: effects on pesticide sensitivity, cardiovascular disease, and drug metabolism. Annu Rev Med 2003; 54:371–92.10.1146/annurev.med.54.101601.152421Search in Google Scholar

10. La Du BN. Human serum paraoxonase/arylesterase. In: Kalow W, editor. Pharmacogenetics of drug metabolism. New York: Pergamon Press, 1992:51–91.Search in Google Scholar

11. Mackness MI, Thompson HM, Hardy AR, Walker CH. Distinction between ‘A’-esterases and arylesterases. Biochem J 1987; 245:293–6.10.1042/bj2450293Search in Google Scholar

12. Adkins S, Gan KN, Mody M, La Du BN. Molecular basis for the polymorphic forms of human serum paraoxonase/arylesterase: glutamine or arginine at position 191, for the respective A or B allozymes. Am J Hum Genet 1993; 52:598–608.Search in Google Scholar

13. Furlong CE, Costa LG, Hassett C, Richter RJ, Sundstrom JA, Adler DA, et al. Human and rabbit paraoxonases: purification, cloning, sequencing, mapping and role of polymorphism in organophosphate detoxification. Chem-Biol Interact 1993; 87:35–48.10.1016/0009-2797(93)90023-RSearch in Google Scholar

14. Davies HG, Richter RJ, Keifer M, Broomfield CA, Sowalla J, Furlong CE. The effect of the human serum paraoxonase polymorphism is reversed with diazoxon, soman and sarin. Nat Genet 1996; 14:334–6.10.1038/ng1196-334Search in Google Scholar PubMed

15. Augustinsson KB. The evolution of esterases. In: Van Thoai N, Roche J, editors. Homologous enzymes and biochemical evolution. New York: Gordon and Breach, 1968:299–311.Search in Google Scholar

16. Ray DE. Chronic effects of low level exposure to anticholinesterases – a mechanistic view. Toxicol Lett 1998; 102–103:527–33.10.1016/S0378-4274(98)00260-4Search in Google Scholar

17. Lotti M. The pathogenesis of organophosphate polyneuropathy. Crit Rev Toxicol 1991; 21:465–87.10.3109/10408449209089884Search in Google Scholar

18. Brealey CJ, Walker CH, Baldwin BC. A-esterase activities in relation to the differential toxicity of pirimiphos-methyl to birds and mammals. Pest Sci 1980; 11:546–54.10.1002/ps.2780110512Search in Google Scholar

19. Main AR. The role of A-esterase in the acute toxicity of paraoxon, TEPP, and parathion. Can J Biochem Physiol 1956; 34:197–216.10.1139/o56-023Search in Google Scholar

20. Li WF, Costa LG, Furlong CE. Serum paraoxonase status: a major factor in determining resistance to organophosphates. J Toxicol Environ Health 1993; 40:337–46.10.1080/15287399309531798Search in Google Scholar

21. Mackness MI, Arrol S, Durrington PN. Paraoxonase prevents accumulation of lipoperoxides in low-density lipoprotein. FEBS Lett 1991; 286:152–4.10.1016/0014-5793(91)80962-3Search in Google Scholar

22. Chisolm GM, Steinberg D. The oxidative modification hypothesis of atherogenesis: an overview. Free Radic Biol Med 2000; 28:1815–26.10.1016/S0891-5849(00)00344-0Search in Google Scholar

23. Aviram M, Rosenblat M, Bisgaier CL, Newton RS, Primo-Parma SL, La Du BN. Paraoxonase inhibits high-density lipoprotein oxidation and preserves its functions. A possible peroxidative role for paraoxonase. J Clin Invest 1998; 101:1581–90.10.1172/JCI1649Search in Google Scholar PubMed PubMed Central

24. Watson AD, Berliner JA, Hama SY, La Du BN, Faull KF, Fogelman AM, et al. Protective effect of high density lipoprotein associated paraoxonase. Inhibition of the biological activity of minimally oxidised low density lipoprotein. J Clin Invest 1995; 96:2882–91.10.1172/JCI118359Search in Google Scholar PubMed PubMed Central

25. van Himbergen TM, van Tits LJ, Hectors MP, de Graaf J, Roest M, Stalenhoef AF. Paraoxonase-1 and linoleic acid oxidation in familial hypercholesterolemia. Biochem Biophys Res Commun 2005; 333:787–93.10.1016/j.bbrc.2005.05.176Search in Google Scholar PubMed

26. Shah PK, Kaul S, Nilsson J, Cercek B. Exploiting the vascular protective effects of high-density lipoprotein and its apolipoproteins: an idea whose time for testing is coming, Part I. Circulation 2001; 104:2376–83.10.1161/hc4401.098467Search in Google Scholar

27. Assmann G, Nofer JR. Atheroprotective effects of high-density lipoproteins. Annu Rev Med 2003; 54:321–41.10.1146/annurev.med.54.101601.152409Search in Google Scholar

28. Ahmed Z, Ravandi A, Maguire GF, Emili A, Draganov D, La Du BN, et al. Apolipoprotein A-I promotes the formation of phosphatidylcholine core aldehydes that are hydrolyzed by paraoxonase (PON-1) during high density lipoprotein oxidation with a peroxynitrite donor. J Biol Chem 2001; 276:24473–81.10.1074/jbc.M010459200Search in Google Scholar

29. Rice-Evans C, Leake D, Bruckdorfer KR, Diplock AT. Practical approaches to low density lipoprotein oxidation: whys, wherefores and pitfalls. Free Radic Res 1996; 25:285–311.10.3109/10715769609149053Search in Google Scholar

30. Puhl H, Waeg G, Esterbauer H. Methods to determine oxidation of low-density lipoproteins. Methods Enzymol 1994; 233:425–41.10.1016/S0076-6879(94)33049-2Search in Google Scholar

31. Mackness MI, Arrol S, Abbot C, Durrington PN. Protection of low-density lipoprotein against oxidative modification by high-density lipoprotein associated paraoxonase. Atherosclerosis 1993; 104:129–35.10.1016/0021-9150(93)90183-USearch in Google Scholar

32. Hedrick CC, Thorpe SR, Fu MX, Harper CM, Yoo J, Kim SM, et al. Glycation impairs high-density lipoprotein function. Diabetologia 2000; 43:312–20.10.1007/s001250050049Search in Google Scholar PubMed

33. Kontush A, Chantepie S, Chapman MJ. Small, dense HDL particles exert potent protection of atherogenic LDL against oxidative stress. Arterioscler Thromb Vasc Biol 2003; 23:1881–8.10.1161/01.ATV.0000091338.93223.E8Search in Google Scholar PubMed

34. Shih DM, Gu L, Hama S, Xia Y-R, Navab M, Fogelman AM, et al. Genetic-dietary regulation of serum paraoxonase expression and its role in atherogenesis in a mouse model. J Clin Invest 1996; 97:1630–9.10.1172/JCI118589Search in Google Scholar PubMed PubMed Central

35. Mackness B, Quarck R, Verreth W, Mackness M, Holvoet P. Human paraoxonase-1 overexpression inhibits athero-sclerosis in a mouse model of metabolic syndrome. Arterioscler Thromb Vasc Biol 2006; 26:1545–50.10.1161/01.ATV.0000222924.62641.aaSearch in Google Scholar PubMed

36. Shih DM, Gu L, Xia Y-R, Navab M, Li W-F, Hama S, et al. Mice lacking serum paraoxonase are susceptible to organophosphate toxicity and atherosclerosis. Nature 1998; 394:284–7.10.1038/28406Search in Google Scholar

37. Van Lenten BJ, Hama SY, de Beer FC, Stafforini DM, McIntyre TM, Prescott SM, et al. Anti-inflammatory HDL becomes pro-inflammatory during the acute phase response. J Clin Invest 1995; 96:2758–67.10.1172/JCI118345Search in Google Scholar

38. Mackness B, Hine D, Liu Y, Mastorikou M, Mackness M. Paraoxonase-1 inhibits oxidised LDL-induced MCP-1 production by endothelial cells. Biochem Biophys Res Commun 2004; 318:680–3.10.1016/j.bbrc.2004.04.056Search in Google Scholar

39. Cao H, Girard-Globa A, Berthezene F, Moulin P. Paraoxonase protection of LDL against peroxidation is independent of its esterase activity towards paraoxon and is unaffected by the Q-R genetic polymorphism. J Lipid Res 1999; 40:133–9.10.1016/S0022-2275(20)33348-4Search in Google Scholar

40. Sarandol E, Safak O, Dirican M, Uncu G. Oxidizability of apolipoprotein B-containing lipoproteins and serum paraoxonase/arylesterase activities in preeclampsia. Clin Biochem 2004; 37:990–6.10.1016/j.clinbiochem.2004.06.013Search in Google Scholar PubMed

41. Rosenblat M, Gaidukov L, Khersonsky O, Vaya J, Oren R, Tawfik DS, 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 2006; 281:7657–65.10.1074/jbc.M512595200Search in Google Scholar PubMed

42. Shih DM, Xia YR, Wang XP, Miller E, Castellani LW, Subbanagounder G, et al. Combined serum paraoxonase knockout/apolipoprotein E knockout mice exhibit increased lipoprotein oxidation and atherosclerosis. J Biol Chem 2000; 275:17527–35.10.1074/jbc.M910376199Search in Google Scholar PubMed

43. Tward A, Xia YR, Wang XP, Shi YS, Park C, Castellani LW, et al. Decreased atherosclerotic lesion formation in human serum paraoxonase transgenic mice. Circulation 2002; 106:484–90.10.1161/01.CIR.0000023623.87083.4FSearch in Google Scholar

44. Bradshaw G, Gutierrez A, Miyake JH, Davis KR, Li AC, Glass CK, et al. Facilitated replacement of Kupffer cells expressing a paraoxonase-1 transgene is essential for ameliorating atherosclerosis in mice. Proc Natl Acad Sci USA 2005; 102:11029–34.10.1073/pnas.0502677102Search in Google Scholar PubMed PubMed Central

45. Mackness B, Durrington P, McElduff P, Yarnell J, Azam N, Watt M, et al. Low paraoxonase activity predicts coronary events in the Caerphilly Prospective Study. Circulation 2003; 107:2775–9.10.1161/01.CIR.0000070954.00271.13Search in Google Scholar PubMed

46. Jarvik GP, Rozek LS, Brophy VH, Hatsukami TS, Richter RJ, Schellenberg GD, et al. Paraoxonase (PON1) phenotype is a better predictor of vascular disease than is PON1(192) or PON1(55) genotype. Arterioscler Thromb Vasc Biol 2000; 20:2441–7.10.1161/01.ATV.20.11.2441Search in Google Scholar

47. Leviev I, Righetti A, James RW. Paraoxonase promoter polymorphism T(–107)C and relative paraoxonase deficiency as determinants of risk of coronary artery disease. J Mol Med 2001; 79:457–63.10.1007/s001090100240Search in Google Scholar

48. Mackness B, Davies GK, Turkie W, Lee E, Roberts DH, Hill E, et al. Paraoxonase status in coronary heart disease: are activity and concentration more important than genotype? Arterioscler Thromb Vasc Biol 2001; 21:1451–7.10.1161/hq0901.094247Search in Google Scholar

49. Abbott CA, Mackness MI, Kumar S, Boulton AJ, Durrington PN. Serum paraoxonase activity, concentration, and phenotype distribution in diabetes mellitus and its relationship to serum lipids and lipoproteins. Arterioscler Thromb Vasc Biol 1995; 15:1812–18.10.1161/01.ATV.15.11.1812Search in Google Scholar

50. Sakai T, Matsuura B, Onji M. Serum paraoxonase activity and genotype distribution in Japanese patients with diabetes mellitus. Intern Med 1998; 37:581–4.10.2169/internalmedicine.37.581Search in Google Scholar

51. Boemi M, Leviev I, Sirolla C, Pieri C, Marra M, James RW. Serum paraoxonase is reduced in type 1 diabetic patients compared to non-diabetic, first degree relatives; influence on the ability of HDL to protect LDL from oxidation. Atherosclerosis 2001; 155:229–35.10.1016/S0021-9150(00)00556-6Search in Google Scholar

52. Boemi M, Sirolla C, Testa R, Cenerelli S, Fumelli P, James RW. Smoking is associated with reduced serum levels of the antioxidant enzyme, paraoxonase, in type 2 diabetic patients. Diabet Med 2004; 21:423–7.10.1111/j.1464-5491.2004.01163.xSearch in Google Scholar

53. Mackness MI, Harty D, Bhatnagar D, Winocour PH, Arrol S, Ishola M, et al. Serum paraoxonase activity in familial hypercholesterolaemia and insulin-dependent diabetes mellitus. Atherosclerosis 1991; 86:193–9.10.1016/0021-9150(91)90215-OSearch in Google Scholar

54. Tomas M, Senti M, Garcia-Faria F, Vila J, Torrents A, Covas M, et al. Effect of simvastatin therapy on paraoxonase activity and related lipoproteins in familial hypercholesterolemic patients. Arterioscler Thromb Vasc Biol 2000; 20:2113–9.10.1161/01.ATV.20.9.2113Search in Google Scholar

55. Senti M, Tomas M, Fito M, Weinbrenner T, Covas MI, Sala J, et al. Antioxidant paraoxonase 1 activity in the metabolic syndrome. J Clin Endocrinol Metab 2003; 88:5422–6.10.1210/jc.2003-030648Search in Google Scholar PubMed

56. Blatter-Garin M, Kalix B, Morabia A, James RW. Small, dense lipoprotein particles and reduced paraoxonase-1 in patients with the metabolic syndrome. J Clin Endocrinol Metab 2005; 90:2264–9.10.1210/jc.2004-1295Search in Google Scholar

57. James RW, Leviev I, Righetti A. Smoking is associated with reduced serum paraoxonase activity and concentration in coronary artery disease patients. Circulation 2000; 101:2252–7.10.1161/01.CIR.101.19.2252Search in Google Scholar

58. Senti M, Aubo C, Tomas M. Differential effects of smoking on myocardial infarction risk according to the Gln/Arg 192 variants of the human paraoxonase gene. Metabolism 2000; 49:557–9.10.1016/S0026-0495(00)80026-8Search in Google Scholar

59. Aviram M, Rosenblat M, Billecke S, Erogul J, Sorenson R, Bisgaier CL, et al. Human serum paraoxonase (PON1) is inactivated by oxidised low density lipoprotein and preserved by antioxidants. Free Radic Biol Med 1999; 26:892–904.10.1016/S0891-5849(98)00272-XSearch in Google Scholar

60. Jarvik GP, Tsai NT, McKinstry LA, Wani R, Brophy VH, Richter RJ, et al. Vitamin C and E intake is associated with increased paraoxonase activity. Arterioscler Thromb Vasc Biol 2002; 22:1329–33.10.1161/01.ATV.0000027101.40323.3ASearch in Google Scholar

61. Aviram M, Dornfeld L, Rosenblat M, Volkova N, Kaplan M, Coleman R, et al. Pomegranate juice consumption reduces oxidative stress, atherogenic modifications to LDL, and platelet aggregation: studies in humans and in atherosclerotic apolipoprotein E-deficient mice. Am J Clin Nutr 2000; 71:1062–76.10.1093/ajcn/71.5.1062Search in Google Scholar PubMed

62. Beer S, Moren X, Ruiz J, James RW. Postprandial modulation of serum paraoxonase activity and concentration in diabetic and non-diabetic subjects. Nutr Metab Cardiovasc Dis 2006. In press, doi 10.1016/j.numecd.2005.09.005.10.1016/j.numecd.2005.09.005Search in Google Scholar PubMed

63. Deakin S, Moren X, James RW. Very low density lipoproteins provide a vector for secretion of paraoxonase-1 from cells. Atherosclerosis 2005; 179:17–25.10.1016/j.atherosclerosis.2004.08.039Search in Google Scholar PubMed

64. Deakin S, Leviev I, Guernier S, James RW. Simvastatin modulates expression of the PON1 gene and increases serum paraoxonase: a role for sterol regulatory element-binding protein-2. Arterioscler Thromb Vasc Biol 2003; 23:2083–9.10.1161/01.ATV.0000096207.01487.36Search in Google Scholar PubMed

65. Ota K, Suehiro T, Arii K, Ikeda Y, Kumon Y, Osaki F, et al. Effect of pitavastatin on transactivation of human serum paraoxonase 1 gene. Metabolism 2005; 54:142–50.10.1016/j.metabol.2004.06.018Search in Google Scholar PubMed

66. Cole TB, Jampsa RL, Walter BJ, Arndt TL, Richter RJ, Shih DM, et al. Expression of human paraoxonase (PON1) during development. Pharmacogenetics 2003; 13:357–64.10.1097/00008571-200306000-00007Search in Google Scholar PubMed

67. Furlong CE, Holland N, Richter RJ, Bradman A, Ho A, Eskenazi B. PON1 status of farmworker mothers and children as a predictor of organophosphate sensitivity. Pharmacogenet Genomics 2006; 16:183–90.10.1097/01.fpc.0000189796.21770.d3Search in Google Scholar PubMed

68. Benmoyal-Segal L, Vander T, Shifman S, Bryk B, Ebstein RP, Marcus EL, et al. Acetylcholinesterase/paraoxonase interactions increase the risk of insecticide-induced Parkinson's disease. FASEB J 2005; 19:452–4.10.1096/fj.04-2106fjeSearch in Google Scholar PubMed

69. Sklan EH, Lowenthal A, Korner M, Ritov Y, Landers DM, Rankinen T, et al. Acetylcholinesterase/paraoxonase genotype and expression predict anxiety scores in Health, Risk Factors, Exercise Training, and Genetics study. Proc Natl Acad Sci USA 2004; 101:5512–7.10.1073/pnas.0307659101Search in Google Scholar PubMed PubMed Central

70. Casida JE, Quistad GB. Organophosphate toxicology: safety aspects of nonacetylcholinesterase secondary targets. Chem Res Toxicol 2004; 17:983–98.10.1021/tx0499259Search in Google Scholar PubMed

71. Biggadike K, Angell RM, Burgess CM, Farrell RM, Hancock AP, Harker AJ, et al. Selective plasma hydrolysis of glucocorticoid gamma-lactones and cyclic carbonates by the enzyme paraoxonase: an ideal plasma inactivation mechanism. J Med Chem 2000; 43:19–21.10.1021/jm990436tSearch in Google Scholar PubMed

72. Jakubowski H. Calcium-dependent human serum homocysteine thiolactone hydrolase. A protective mechanism against protein n-homocysteinylation. J Biol Chem 2000; 275:3957–62.10.1074/jbc.275.6.3957Search in Google Scholar PubMed

73. Khersonsky O, Tawfik DS. Structure-reactivity studies of serum paraoxonase PON1 suggest that its native activity is lactonase. Biochemistry 2005; 44:6371–82.10.1021/bi047440dSearch in Google Scholar PubMed

74. Draganov DI, Teiber JF, Speelman A, Osawa Y, Sunahara R, La Du BN. Human paraoxonases (PON1, PON2, and PON3) are lactonases with overlapping and distinct substrate specificities. J Lipid Res 2005; 46:1239–47.10.1194/jlr.M400511-JLR200Search in Google Scholar PubMed

75. Serhan CN. Lipoxins and aspirin-triggered 15-epi-lipoxins are the first lipid mediators of endogenous anti-inflammation and resolution. Prostaglandins Leukot Essent Fatty Acids 2005; 73:141–62.10.1016/j.plefa.2005.05.002Search in Google Scholar PubMed

76. Libby P. Changing concepts of atherogenesis. J Intern Med 2000; 247:349–58.10.1046/j.1365-2796.2000.00654.xSearch in Google Scholar

77. Navab M, Ananthramaiah GM, Reddy ST, Van Lenten BJ, Ansell BJ, Fonarow GC, et al. The oxidation hypothesis of atherogenesis: the role of oxidized phospholipids and HDL. J Lipid Res 2004; 45:993–1007.10.1194/jlr.R400001-JLR200Search in Google Scholar

78. Splaver A, Lamas GA, Hennekens CH. Homocysteine and cardiovascular disease: biological mechanisms, observational epidemiology, and the need for randomized trials. Am Heart J 2004; 148:34–40.10.1016/j.ahj.2004.02.004Search in Google Scholar

79. McCully KS. Chemical pathology of homocysteine. I. Atherogenesis. Ann Clin Lab Sci 1993; 23:477–93.Search in Google Scholar

80. Jakubowski H. Molecular basis of homocysteine toxicity in humans. Cell Mol Life Sci 2005; 61:470–87.10.1007/s00018-003-3204-7Search in Google Scholar

81. Fuqua C, Parsek MR, Greenberg EP. Regulation of gene expression by cell-to-cell communication: acyl-homoserine lactone quorum sensing. Annu Rev Genet 2001; 35:439–68.10.1146/annurev.genet.35.102401.090913Search in Google Scholar

82. Chun CK, Ozer EA, Welsh MJ, Zabner J, Greenberg EP. Inactivation of a Pseudomonas aeruginosa quorum-sensing signal by human airway epithelia. Proc Natl Acad Sci USA 2004; 101:3587–90.10.1073/pnas.0308750101Search in Google Scholar

83. Ozer EA, Pezzulo A, Shih DM, Chun C, Furlong C, Lusis AJ, et al. Human and murine paraoxonase 1 are host modulators of Pseudomonas aeruginosa quorum-sensing. FEMS Microbiol Lett 2005; 253:29–37.10.1016/j.femsle.2005.09.023Search in Google Scholar

84. Muhlestein JB, Anderson JL. Chronic infection and coronary artery disease. Cardiol Clin 2003; 21:333–62.10.1016/S0733-8651(03)00054-7Search in Google Scholar

85. Procopiou PA, Biggadike K, English AF, Farrell RM, Hagger GN, Hancock AP, et al. Novel glucocorticoid antedrugs possessing a 17beta-(gamma-lactone) ring. J Med Chem 2001; 44:602–12.10.1021/jm001035cSearch in Google Scholar PubMed

86. Tougou K, Nakamura A, Watanabe S, Okuyama Y, Morino A. Paraoxonase has a major role in the hydrolysis of prulifloxacin (NM441), a prodrug of a new antibacterial agent. Drug Metab Dispos 1998; 26:355–9.Search in Google Scholar

87. Billecke S, Draganov D, Counsell R, Stetson P, Watson C, Hsu C, et al. Human serum paraoxonase (PON1) isozymes Q and R hydrolyze lactones and cyclic carbonate esters. Drug Metab Dispos 2000; 28:1335–42.Search in Google Scholar

Received: 2006-5-29
Accepted: 2006-6-20
Published Online: 2011-9-21
Published in Print: 2006-9-1

©2006 by Walter de Gruyter Berlin New York

Downloaded on 30.5.2024 from https://www.degruyter.com/document/doi/10.1515/CCLM.2006.207/html
Scroll to top button