ISSN: 1885-5857 Impact factor 2023 7.2
Vol. 57. Num. 6.
Pages 557-569 (June 2004)

The Antioxidant Function of High Density Lipoproteins: A New Paradigm in Atherosclerosis

Función antioxidante de las lipoproteínas de alta densidad: un nuevo paradigma en la arteriosclerosis

Marta TomásaGlòria LatorreaMariano SentíbJaume Marrugatc

Options

The one disease associated with the greatest morbidity and mortality in industrialized countries is coronary heart disease (CHD). High density lipoprotein (HDL) is one of the most important independent protective factors for the arteriosclerosis which underlies CHD. Paraoxonase1 (PON1) is an enzyme that confers antioxidant properties to HDL. In vitro, PON1 hydrolyzes a large variety of endogenous or exogenous substrates, some of which are clearly involved in the progression of arteriosclerosis. A close relationship between PON1 deficiency and accelerated progression of arteriosclerosis has been found in animal models. Moreover, PON1 activity is reduced in high oxidative stress diseases such as CHD, dyslipoproteinemias, inflammatory processes, diabetes and certain neuropathies. Reduced PON1 enzyme activity is associated with several arteriosclerosis-related diseases. The most thoroughly studied genetic variant of PON1 is PON1-192, in which the R allele is associated with elevated paraoxonase activity. This allele, present in 24.8% of the Italian population, is found in up to 78.9% of the population of Ecuadorian Cayapa Indians. A metaanalysis of studies on the relationship between CHD and the R allele showed the latter to be an independent risk factor for this disease, with an odds ratio of 1.18 (95% confidence interval, 1.10-1.27). The PON1 enzyme is a potentially useful new qualitative indicator in addition to the well known reverse cholesterol transport capacity associated with high plasma levels of HDL.

Keywords

Paraoxonase
Arteriosclerosis
Antioxidants
Oxidative stress

High-density lipoprotein (HDL) is today regarded as one of the most important protective factors against arteriosclerosis. Traditionally, HDL's protective function has been attributed to its active participation in the reverse transport of cholesterol. Since 1975, numerous cohort studies and clinical trials have confirmed the association between a low HDL-cholesterol concentration and an increased risk of coronary heart disease.1­5 In experimental animals, an inverse correlation between HDL concentration and the development of arteriosclerosis has been demonstrated. However, it has been observed that arteriosclerotic lesions tend to regress in vivo as the concentration of HDL or its apolipoproteins increases.6­8 On the other hand, certain genetically inherited diseases that are characterized by an abnormally low HDL level, such as Tangier disease, fish-eye disease and diseases linked to apo A1 gene mutations, are frequently associated with arteriosclerosis and premature ischemic heart disease.9­12 Recently, it has been shown that increasing the HDL-cholesterol concentration is artificially inhibited by cholesterol ester transfer protein. However, the effect an increased concentration has on the risk of coronary disease remains unknown.13

The "French paradox" in cardiovascular disease refers to the coexistence of a diet rich in saturated fats with low mortality from coronary heart disease. In Spain, despite the fact that coronary heart disease affects more than 68 000 people annually and costs the lives of more than 40% of those individuals, it has also been confirmed that a low incidence of acute myocardial infarction (AMI) coexists with a high prevalence of classic cardiovascular risk factors.14,15 For this reason, it makes sense to study the variables that could explain this remarkable situation, including, importantly, HDL and it properties. The significant antioxidant and anti-inflammatory effects of HDL, which have been discovered in the last decade, could substantially modify our understanding of the mechanisms that prevent arteriosclerosis.

In the Spanish population, HDL concentrations are similar to or slightly higher than those normally found in individuals from countries with a higher incidence of coronary heart disease, such as the United States, Australia, and the countries of northern Europe.16 Consequently, the absolute HDL concentration does not appear to be the determining factor in explaining the observed geographical differences in disease incidence.16 It could be that specific qualitative characteristics of the HDL particle increase HDL's antioxidant capacity and, thereby, enable it to provide greater protection against cardiac ischemia.16 Paraoxonase-1 (PON1), the enzyme responsible for HDL's antioxidant function, is closely bound to the HDL particle. PON1's enzymatic activity is highly regulated by environmental factors such as diet and physical activity, by certain drugs, and by genetic factors, especially certain genetic polymorphisms in the paraoxonase-1 gene, PON1.17­19 The antioxidant capacity of PON1 and the variability observed in its activity, which can be evaluated using the paraoxon substrate in a variety of contexts, ensure that the results of investigations into PON1 will be of great interest in the field of arteriosclerotic disease.

PROPERTIES AND CHARACTERISTICS OF PARAOXONASE-1

PON1 is synthesized in the mammalian liver and circulates in blood bound to HDL apolipoprotein (apo) A-1 and apo J. PON1 expression is inhibited by proatherogenic stimuli.20­23 There are 2 other proteins in the same family that probably also have an antioxidant function: PON2 and PON3. PON2 is ubiquitously expressed within cells, whereas PON3 exhibits a basal constitutive antioxidant activity and is essentially bound to HDL.22,24­26 In this review, we will focus on PON1, whose relationship with numerous pathological conditions and with environmental and genetic factors has been the most extensively studied of the 3 proteins.

PON1 has a number of enzymatic activities (Table). We will concentrate on its antioxidant activity and its paraoxonase activity, that is, the activity that is quantified using the pesticide paraoxon as a substrate.

Paraoxonase Activity (Paraoxon Substrate)

PON1 was first identified in the field of toxicology because it was able to hydrolyze synthetic organophosphates such as pesticides (e.g., paraoxon, and oxon metabolites of chlorpyrifos and diazinon) and nerve gases (e.g., sarin and soman).27 Paraoxon is a toxic metabolite produced in the liver from parathion, which is a relatively innocuous compound.28 In mammals, when paraoxon enters the bloodstream, it is hydrolyzed and inactivated by HDL PON1. This does not occur in birds, fish and insects, which generally lack PON1 and are easily poisoned by these compounds.29

It is important to note that paraoxonase activity is reduced in patients with conditions associated with arteriosclerosis, such as AMI, diabetes mellitus and familial hypercholesterolemia (FH).30­33 It has been reported that PON1's level of interaction with the paraoxon substrate is highly correlated with the protein's concentration.34 However, despite strong binding of the enzyme to the HDL particle, there does not always appear to be a correlation between HDL concentration and paraoxonase activity.35

Other Enzymatic Activities of Paraoxonase-1: Arylesterase (Phenylacetate Substrate) and Lactonase (Lactone Substrate) Activities

PON1 is able to hydrolyze arylesters such as phenylacetate by means of its arylesterase activity, which also appears to be related to the concentration of the protein.34,36,37 It also has the capacity to hydrolyze more than 30 different lactone molecules, including endogenous molecules, such as homocysteine thiolactone and glucocorticoid gamma-lactones, and exogenous molecules, such as statins.38,39

Antioxidant Activity

PON1 endows HDL with its antioxidant properties and is probably responsible for the principal mechanism inhibiting the oxidation of both low-density lipoproteins (LDLs) and HDL itself, a process that is directly involved in the initial phases of arteriosclerosis, as illustrated in Figure 1.40­43 In vitro, PON1 neutralizes hydrogen peroxide and peroxidized lipids that are either free or present in atherosclerotic lesions or in minimally oxidized LDL.42,44­46 However, although there is still no general agreement on the subject, PON1 may be able to activate platelet activating factor acetylhydrolase, an enzyme that hydrolyzes this well-known proinflammatory factor. This process could give PON1 its anti-inflammatory properties.47,48

Fig. 1. The antioxidant role of paraoxonase-1 in arteriosclerotic plaque formation. ABCA1 indicates ATP-binding cassette A1; apo A-I, apolipoprotein A-I; CETP, cholesterol ester transfer protein; HDL, high-density lipoprotein; LDL, low-density lipoprotein; LDLox, oxidized LDL; HL, hepatic lipase; LPL, lipoprotein lipase; PON1, paraoxonase-1; Cm, chylomicron; ROS, reactive oxygen species; TG, triglycerides; VLDL, very-low-density lipoprotein.

Experimental evidence for the antioxidant function of PON1 comes from in vivo studies carried out in PON1/apo E double knock-out mice. These studies have demonstrated that PON1 is effective in inhibiting the oxidation of both LDL and intermediate-density lipoproteins, substances with proven atherogenic effects.49 PON1-knock-out mice fed an atherogenic diet rich in fat and cholesterol developed more arteriosclerosis than wild-type mice. In addition, their HDL was incapable of counteracting LDL oxidation, being both LDL and HDL itself more easily oxidized.18 Animals lacking the PON1 gene are in a state of increased oxidative stress.50,51 In contrast, mice that overexpress PON1 demonstrate less lipid hydroperoxide production.52 Moreover, when human PON1 is injected into apo E-knock-out mice that exhibit accelerated arteriosclerosis development, the lipid peroxide content of peritoneal macrophages decreases significantly.53

Factors Regulating the Enzymatic Activity of Paraoxonase-1

Lipid peroxides inhibit the paraoxonase, arylesterase and antioxidant activities of PON1, probably via interactions with a sulfur group on the enzyme.54,55 One important consequence of this phenomenon is that, if HDL is oxidized, there will be an accompanying reduction in paraoxonase activity and, therefore, also a reduction in the enzyme's protective activity against LDL oxidation.56

In addition to being influenced by lipid peroxides, the variations observed in PON1's enzymatic activity also depend on specific amino acids in the protein (such as a cysteine at position 283), the plasma concentration of the calcium ion Ca2+, and the presence of genetic variants in gene-coding regions, such as PON1-192 and PON1-55, and in the gene promoter, such as PON1-(107).38,54 Figure 2 illustrates the frequency of occurrence of the R allele of the PON1-192 polymorphism in different parts of the world.

Fig. 2. The frequency of occurrence of the R allele of the PON1-192 polymorphism in different parts of the world. Adapted from Scacchi et al159 with permission (© 2003 Wayne State University Press, Detroit, Michigan, USA). Data on the Spanish population are taken from Senti et al.134

PARAOXONASE-1 AND DISEASES RELATED TO ARTERIOSCLEROSIS

Diseases associated with arteriosclerosis and with various environmental factors are, to a large extent, related to by PON1 activity. However, PON1 is known to be an inducible enzyme as various stimuli can modify its expression.22 To date, the published results of studies into the relationship between specific genetic polymorphisms in the PON1 gene and coronary heartdisease have been clearly inconsistent. This is the case with the PON1-192 polymorphism. We carried out a meta-analysis on this polymorphisms as part of this review. See Figure 3. The principal finding was that the presence of the R allele of the PON1-192 polymorphism was weakly but significantly associated with the presence of coronary heart disease when the results of all the studies that dealt with the subject were combined. This finding was not affected by whether (Figure 3a) or not (Figure 3b) the studies looked at certain subgroups of patients, such as those with diabetes or FH, or those who belonged to a specific age group. It would seem logical, therefore, that PON1 genotype, as well as PON1 activity and concentration, must be taken into account in attempting to predict the risk of coronary heart disease.19,57,58 However, to date, few studies have looked at PON1 polymorphisms in combination with paraoxonase or arylesterase activities.59­62 Nevertheless, there are some indications that the interindividual variability in PON1 activity found in healthy subjects is principally due to factors that are independent of genotype.63

Fig. 3a. Meta-analysis of the relationship between the frequency of occurrence of the R allele of the PON1-192 polymorphism and the presence of coronary heart disease in 33 studies. *Diabetics; aage <62 years; bage <45 years; dprospective placebo-controlled study; eprospective study of fluvastatin treatment; ¼familial hypercholesterolemia; OR indicates odds ratio; CI, confidence interval

Fig. 3b. Meta-analysis of the relationship between the frequency of occurrence of the R allele of the PON1-192 polymorphism and the presence of coronary haert disease in 25 studies that did not include diabetics, that did not have an age limit, that did not involve prospective assessment of interventions, and that did not compare control subjects with patients with familial hypercholesterolemia. OR indicates odds ratio; CI, confidence interval

Coronary Heart Disease, Inflammation and Paraoxonase-1

It is well established that arteriosclerosis and its clinical manifestations are often associated with a reduced concentration of HDL, the type of lipoprotein on which PON1 is located. In fact, we know that a low HDL concentration is usually accompanied by a reduction in PON1 activity or concentration, such as occurs in Tangier disease and fish-eye disease.64,65

Cellular factors associated with inflammation have been found in arteriosclerotic lesions. Some studies have shown that the incidence of arteriosclerosis is related to the presence of certain infectious agents, such as Chlamydia pneumoniae and cytomegalovirus, and to the presence of conditions such as chronic bronchitis.66­69 Moreover, certain molecules implicated in acute-phase inflammatory responses, such as interleukin-1 (IL-1), IL-6, endotoxins, and oxidized phospholipids, reduce both PON1 expression and its paraoxonase activity.70,71 Similarly, it has been reported that proinflammatory cytokines, such as IL-1β, tumor necrosis factor-alpha (TNF-α), and IL-6, have a coordinated effect on the regulation of PON1 transcription in hepatocytes.72 The formation of immune complexes directed against oxidized apo A-I that has been observed in mice fed on a short-term atherogenic diet could be the mechanism by which oxidized HDL is eliminated. This process could lead to a reduction in paraoxonase activity without affecting messenger RNA (mRNA) levels.73

Reduced paraoxonase activity and the concomitant loss of HDL's antioxidant properties have been observed during the acute-phase response in patients who have undergone cardiac surgery.74 In addition, reduced paraoxonase activity has also been observed in subjects with antiphospholipid autoantibodies, in individuals infected by the influenza virus, and in patients with rheumatoid arthritis.75­77 In these individuals, mortality from coronary heart disease appears to be increased.

Paraoxonase activity is diminished after an AMI and remains reduced from 2 hours to approximately 40 days after the event.30,32,78 Moreover, activity may also be diminished for a short time before the onset of symptoms. Serum arylesterase activity, which declines during heart failure, increases again after appropriate treatment is given.79 In addition, diseases that are closely linked to arteriosclerosis, such as FH, the metabolic syndrome and AMI, are also associated with reduced paraoxonase activity, as was observed in some of the studies illustrated in Figure 4.31,33,80,81 Nevertheless, in the absence of an analytical test based on lipid peroxide hydrolysis and of accurate knowledge of PON1's physiological substrate, it would make sense to determine paraoxonase and diazoxonase activity or the PON1 concentration in all epidemiological studies looking at the relationship between PON1 and coronary disease.34,59

Fig. 4. Paraoxonase activity (mean ± standard deviation) in untreated patients with familial hypercholesterolemia, in patients with familial hypercholesterolemia receiving simvastatin, in stable patients with a history of acute myocardial infarction (AMI) or unstable angina, in patients who had had a first AMI 6 months previously and who had a low high-density lipoprotein cholesterol (HDL-C) level, and in patients with the metabolic syndrome, each compared with healthy control subjects. P indicates significance of the statistical difference between patients and control subjects; NS, not significant; *median and interquartile range.

Individuals with a paraoxonase activity level in the highest quintile have less risk of coronary heart disease than those whose activity level lies in the lowest quintile. In addition, the difference was found to be particularly striking in patients taking part in studies on the secondary prevention of coronary heart disease.82 The paraoxonase concentration and paraoxonase activity are reduced by as much as 50% in patients with coronary heart disease, irrespective of whether they have the PON1-192 or PON1-55 genotype.59,83 Moreover, some of these patients have normal HDL levels. It has been suggested that their type of HDL is insufficiently protective against monocyte migration to atherosclerotic plaque.84 In addition, it has been observed that the PON1 concentration tends to be lower in populations in which there is a high prevalence of coronary heart disease.62 In this context, it is worthwhile noting that the PON1 concentration predicts the occurrence of coronary heart disease in a similar way to other parameters, such as the ratio of the apo J concentration to paraoxonase activity or the ratio of the total cholesterol concentration to the HDL cholesterol concentration.59,84

Certain pecularieties have has also been reported in patients who present with chronic complaints that are frequently associated with coronary heart disease exhibit certain peculiarities. For example, renal transplant patients, with or without coronary heart disease, have reduced levels of paraoxonase and arylesterase activity.61 In contrast, diabetic patients with coronary heart disease present with higher PON1 activity levels than those without coronary heart disease.37

Some authors regard paraoxonase activity as being a better predictor of coronary heart disease risk than the PON1-(-107), PON1-55, or PON1-192 genotypes alone. Nevertheless, it is important to remember that enzyme activity levels overlap considerably between patients with different genotypes of the same polymorphism.57,59,85,86

Familial Hypercholesterolemia and Paraoxonase-1

The prevalence of the heterozygotic form of FH in the general population is 0.2%. The disease is characterized by a reduced number of LDL receptors, which is due to mutations in the receptor gene. The majority of patients with FH present with type-IIa hyperlipidemia with a smaller proportion presenting with phenotype-IIb hyperlipidemia, where the different forms of hyperlipidemia are defined according to the World Health Organization's adaptation of Fredrickson's hyperlipidemia classification.87 Affected patients often have early arteriosclerosis.88,89 It has been observed that paraoxonase activity in patients with FH is 50-80% of that found in unaffected individuals despite there is no difference in HDL concentration.31,90 A similar situation occurs when the hypercholesterolemic phenotype is reproduced in animals fed on a saturated fat-rich diet. These animal models demonstrate significant reductions in paraoxonase activity in the absence of any appreciable change in HDL level.91

Statin treatment has been observed to increase paraoxonase activity significantly in patients with FH.31,92,93 Factors associated with transcription may by involved in this phenomenon since it is known that simvastatin can increase the transcriptional activity of the PON1 gene promoter by 2.5 times through a mechanism that depends on mevalonate, a metabolic product of 3-hydroxymethylglutaryl coenzyme A (HMG-CoA) reductase.94

Statins are hypolipidemic compounds that inhibit HMG-CoA reductase, a key enzyme in the intracellular biosynthesis of cholesterol. In general, when used for either primary or secondary disease prevention, statins bring about a reduction in total and LDL cholesterol levels, and alter the HDL concentration to a variable extent.95­98 In addition to modifying the lipid profile, simvastatin, like other statins, can have pleiotropic effects that are not directly related to lipids. These can either prevent arteriosclerosis or bring about its regression by means of a variety of mechanisms.99 In particular, both in vitro and ex vivo experiments indicate that simvastatin itself can act as an antioxidant. Adding simvastatin to a medium containing LDL and HDL reduces the rate of formation of conjugated dienes, products of fatty acid oxidation. Similarly, ex vivo experiments in patients treated with simvastatin show that there is less oxidation of their LDL and HDL.95 Moreover, PON1 can hydrolyze statins, which contain lactone rings.38 Some derivatives of atorvastatin have been observed to inhibit LDL and HDL oxidation in vitro and to maintain paraoxonase activity.100 This latter effect has not been observed with fluvastatin, whose administration is associated with a reduction in PON1's paraoxonase, arylesterase and lactonase activities in rats. However, fluvastatin does reduce oxidative stress in these animals.101

The fibrates form another group of widely used hypolipidemic medications. Their principal pharmacological actions are to reduce the triglyceride concentration and to increase the HDL cholesterol concentration. Although some serious side effects have been reported, combined statin and fibrate administration may be indicated in patients with hypercholesterolemia and hypertriglyceridemia.89 Numerous clinical trials have confirmed that fibrates can slow arteriosclerosis progression and can reduce cardiovascular morbidity and mortality. These benefits are due to the compounds' hypolipidemic activity and possibly also to their pleiotropic effects, which are anti-inflammatory and antioxidant in nature and can improve endothelial function.102

There is no general agreement about how fibrates influence paraoxonase activity. Although, initially, gemfibrozil was reported to increase paraoxonase activity, this observation has not been duplicated in later studies on gemfibrozil, bezafibrate, ciprofibrate or fenofibrate.103­106

Nevertheless, some reports indicate that gemfibrozil helps preserve paraoxonase activity and inhibits LDL and HDL oxidation in vitro.100,107

Diabetes Mellitus and Paraoxonase-1

Type-I diabetes mellitus is associated with an elevated level of oxidative stress, increased susceptibility to coronary heart disease, and reductions in PON1 concentration and paraoxonase activity.108 It has been reported that 67% of type-I diabetics have a low level of paraoxonase activity, irrespective of HDL cholesterol concentration, compared with 50% of the healthy population.90

Type-II diabetes mellitus is characterized by a raised blood glucose level, hypertriglyceridemia, increased oxidative metabolism, reduced HDL cholesterol concentration, high prevalence of obesity, and accelerated arteriosclerosis.109­111 Type-II diabetes is also associated with the occurrence of cardiovascular events, which are probably linked to low HDL cholesterol concentrations rather than to elevated LDL cholesterol concentrations.112 Moreover, type-II diabetics present with a lower paraoxonase activity level and a lower ratio of paraoxonase activity to HDL cholesterol level than those found in health control subjects.113­115 One striking observation in these patients is that there is no correlation between paraoxonase activity and HDL cholesterol concentration.115 In addition, diabetic patients who present with complications such as coronary heart disease, retinopathy or neuropathy37,114,116 have lower paraoxonase activity levels than diabetics without these complications.90,113,117 In our laboratory, we have recently observed that paraoxonase activity decreases as the number of metabolic alterations linked to the metabolic syndrome rises and that this decrease is accompanied by increasingly high concentrations of lipid peroxides.81 Both individuals with impaired glucose tolerance and recently diagnosed diabetics have normal enzymatic activity levels even though they exhibit an abnormally high level of LDL oxidation.118 Apparently, paraoxonase activity declines as diabetes progresses and reaches a particularly low level in advanced stages of the disease. In patients with obesity associated with an elevated leptin concentration, reduced PON1 paraoxonase, arylesterase and lactonase activities and increased oxidative stress have been observed. These phenomena could explain, at least in part, the relationship between obesity and arteriosclerosis in hyperleptinemics.119 It has also been observed that paraoxonase activity and HDL concentrations are lower in diabetic patients undergoing hemodialysis than in non-diabetic patients undergoing hemodialysis.120

In vitro, an elevated glucose concentration reduces HDL's antioxidant capacity, partly because both paraoxonase activity and the ratio of paraoxonase activity to HDL cholesterol level decrease while the concentration of oxidation markers simultaneously increases.117 In rats with streptozocin-induced diabetes, serum paraoxonase activity declines progressively with time.121

Neurological Disease and Paraoxonase-1

The majority of organophosphate compounds are neurotoxic. Chronic exposure can induce neuropathies and has neuropsychiatric effects.122 It has been suggested that PON1's ability to detoxify both organophosphates and lipid peroxides could make the enzyme a key element in determining susceptibility to neurological disease, such as Alzheimer disease and Parkinson disease.123

One notable observation is that farmers who disinfect sheep with organophosphates that have diazoxon as a metabolite can become ill if PON1's ability to hydrolyze diazoxon is reduced.124 Low PON1 activity levels have been observed in Persian Gulf War veterans, some of whom presented with neurotoxic effects.125,126

Kidney Failure and Paraoxonase-1

Chronic kidney failure is generally associated with dyslipidemia, an elevated risk of coronary heart disease, and an elevated level of oxidative stress, which is probably due to the inadequate antioxidant enzymatic activities of superoxide dismutase, catalase and PON1.127­129 Although paraoxonase activity, aryles terase activity and the HDL level are all reduced in patients who are undergoing hemodialysis or who have uremia, this not the case for the ratio of paraoxonase activity to HDL cholesterol level or for the ratio of paraoxonase activity to apo A-I level in diabetics with nephropathy.113,129­132 Kidney transplantation appears to re-establish normal paraoxonase activity and may also increase arylesterase activity.61,133

CONCLUSIONS

The ability of HDL to protect against arteriosclerosis and coronary heart disease has traditionally been attributed to its well-known participation in the reverse transport of cholesterol. Within the last few years, another mechanism has been discovered: HDL's function as a LDL antioxidant. This additional property of HDL could be vital since its effect is to prevent or delay the formation of atheromatous plaque. Consequently, our understanding of the mechanisms by which HDL exerts a protective effect has been substantially refined. It is now believed that it is not only the quantity of HDL that is important but also the qualitative nature of its antioxidant capacity. The presence in HDL particles of the antioxidant enzyme PON1, which determines HDL's ability to restrain the process of LDL oxidation in subendothelial tissue, strengthens this belief. If, as it appears, the antioxidant capacity of HDL depends almost exclusively on PON1, it is logical to search for factors that can bring about an increase in PON1 activity. This discovery clearly has important clinical implications and may contribute to explaining part of the southern European paradox, which is illustrated, for example, by the fact that both the mortality and the incidence of myocardial infarction in Spain are lower than would be expected from the observed prevalence of known risk factors.

Full English text available at: www.revespcardiol.org

ABBREVIATIONS
AMI: acute myocardial infarction.
apo: apolipoprotein.
FH: familial hypercholesterolemia.
HDL: high-density lipoprotein.
HMG-CoA: 3-hydroxy-3-methylglutaryl coenzyme A.
IL: interleukin.
LDL: low-density lipoprotein.
mRNA: messenger ribonucleic acid.
PON: paraoxonase.
TNF: tumor necrosis factor.

This review was partly funded by grants FIS 99/0013/01, FEDER 1FD97/0626, and FIS G03/45 (Red Temática de Grupos HERACLES) awarded by the Instituto de Salud Carlos III, Spain, and by grant FIS C03/01 (Red Temática de Centros RECAVA) awarded by the Fondo de Investigación Sanitaria, Spain.


Correspondence: Dr. J. Marrugat.
Unitat de Lípids i Epidemiologia Cardiovascular.
Institut Municipal d'Investigació Mèdica (IMIM).
Dr. Aiguader, 80. 08003 Barcelona. España.
E-mail: jmarrugat@imim.es

Bibliography
[1]
Miller GJ, Miller NE..
Plasma-high-density-lipoprotein concentration and development of ischaemic heart-disease..
Lancet, (1975), 1 pp. 16-9
[2]
Gordon T, Castelli WP, Hjortland MC, Kannel WB, Dawber TR..
High density lipoprotein as a protective factor against coronary heart disease. The Framingham Study..
Am J Med, (1977), 62 pp. 707-14
[3]
Rhoads GG, Gulbrandsen CL, Kagan A..
Serum lipoproteins and coronary heart disease in a population study of Hawaii Japanese men..
N Engl J Med, (1976), 294 pp. 293-8
[4]
The Lipid Research Clinics Coronary Primary Prevention Trial results..
I. Reduction in incidence of coronary heart disease..
JAMA, (1984), 251 pp. 351-64
[5]
Gordon DJ, Probstfield JL, Garrison RJ, Neaton JD, Castelli WP, Knoke JD, et al..
High-density lipoprotein cholesterol and cardiovascular disease. Four prospective American studies..
Circulation, (1989), 79 pp. 8-15
[6]
Miyazaki A, Sakuma S, Morikawa W, Takiue T, Miake F, Terano T, et al..
Intravenous injection of rabbit apolipoprotein A-I inhibits the progression of atherosclerosis in cholesterol-fed rabbits..
Arterioscler Thromb Vasc Biol, (1995), 15 pp. 1882-8
[7]
Badimon JJ, Badimon L, Fuster V..
Regression of atherosclerotic lesions by high density lipoprotein plasma fraction in the cholesterol-fed rabbit..
J Clin Invest, (1990), 85 pp. 1234-41
[8]
Nissen SE, Tsunoda T, Tuzcu EM, Schoenhagen P, Cooper CJ, Yasin M, et al..
Effect of recombinant apo A-I Milano on coronary atherosclerosis in patients with acute coronary syndromes: a randomized controlled trial..
JAMA, (2003), 290 pp. 2292-300
[9]
Srivastava N..
ATP binding cassette transporter A1­key roles in cellular lipid transport and atherosclerosis..
Mol Cell Biochem, (2002), 237 pp. 155-64
[10]
Kuivenhoven JA, Pritchard H, Hill J, Frohlich J, Assmann G, Kastelein J..
The molecular pathology of lecithin:cholesterol acyltransferase (LCAT) deficiency syndromes..
J Lipid Res, (1997), 38 pp. 191-205
[11]
Ordovas JM, Cassidy DK, Civeira F, Bisgaier CL, Schaefer EJ..
Familial apolipoprotein A-I, C-III, and A-IV deficiency and premature atherosclerosis due to deletion of a gene complex on chromosome 11..
J Biol Chem, (1989), 264 pp. 16339-42
[12]
Matsunaga T, Hiasa Y, Yanagi H, Maeda T, Hattori N, Yamakawa K, et al..
Apolipoprotein A-I deficiency due to a codon 84 nonsense mutation of the apolipoprotein A-I gene..
Proc Natl Acad Sci U S A, (1991), 88 pp. 2793-7
[13]
Brousseau ME, Schaefer EJ, Wolfe ML, Bloedon LT, Digenio AG, Clark RW, et al..
Effects of an inhibitor of cholesteryl ester transfer protein on HDL cholesterol..
N Engl J Med, (2004), 350 pp. 1505-15
[14]
Marrugat J, Elosua R, Martí H..
Epidemiología de la cardiopatía isquémica en España: estimación del número de casos y de las tendencias entre 1997 y 2005..
Rev Esp Cardiol, (2002), 55 pp. 337-46
[15]
Masià R, Pena A, Marrugat J, Sala J, Vila J, Pavesi M, et al..
High prevalence of cardiovascular risk factors in Gerona, Spain, a province with low myocardial infarction incidence..
J Epidemiol Community Health, (1998), 52 pp. 707-15
[16]
Sentí M, Masià R, Pena A, Elosua R, Aubó C, Bosch M, et al..
Determinantes antropométricos y dietéticos de la concentración sérica del colesterol de las lipoproteínas de alta densidad en un estudio de base poblacional. El estudio REGICOR..
Rev Esp Cardiol, (1998), 51 pp. 979-87
[17]
Durrington PN, Mackness B, Mackness MI..
Paraoxonase and atherosclerosis..
Arterioscler Thromb Vasc Biol, (2001), 21 pp. 473-80
[18]
Shih DM, Gu L, Xia YR, Navab M, Li W, Hama S, et al..
Mice lacking serum paraoxonase are susceptible to organophosphate toxicity and atherosclerosis..
Nature, (1998), 394 pp. 284-7
[19]
Mackness B, Mackness MI, Arrol S, Turkie W, Durrington PN..
Effects of the human serum paraoxonase 55 and 192 genetic polymorphisms on the protection by high density lipoprotein against low density lipoprotein oxidative modification..
FEBS Lett, (1998), 423 pp. 57-60
[20]
Mackness MI..
Possible medical significance of human serum paraoxonase. En: Reiner E, Aldridge WN, Hoskin FC, editors. Enzymes hydrolysing organophosphorus compounds. UK: Ellis-Horwood.
Possible medical significance of human serum paraoxonase. En: Reiner E, Aldridge WN, Hoskin FC, editors. Enzymes hydrolysing organophosphorus compounds. UK: Ellis-Horwood, (1989), pp. 202-13
[21]
Blatter MC, 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 pp. 871-9
[22]
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 pp. 542-7
[23]
Kelso GJ, Stuart WD, Richter RJ, Furlong CE, Jordan-Starck TC, Harmony JA..
Apolipoprotein J is associated with paraoxonase in human plasma..
Biochemistry, (1994), 33 pp. 832-9
[24]
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 pp. 498-507
[25]
Mochizuki H, Scherer SW, Xi T, Nickle DC, Majer M, Huizenga JJ, et al..
Human PON2 gene at 7q213: cloning, multiple mRNA forms, and missense polymorphisms in the coding sequence..
Gene, (1998), 213 pp. 149-57
[26]
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 pp. 44444-9
[27]
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 pp. 334-6
[28]
Human serum paraoxonase/arylesterase. In: Kalow W, editor. Pharmacogenetics of drug metabolism. New York: Pergamon Press, 1992.
[29]
Brealey CJ, Walker CH, Bladwin BC..
A-esterase activities in relation to the differential toxicity of pirimiphos-methyl to birds and mammals..
Pestic Sci, (1980), 11 pp. 546-54
[30]
Ayub A, Mackness MI, Arrol S, Mackness B, Patel J, Durrington PN..
Serum paraoxonase after myocardial infarction..
Arterioscler Thromb Vasc Biol, (1999), 19 pp. 330-5
[31]
Tomás M, Senti M, García-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 pp. 2113-9
[32]
McElveen J, Mackness NI, Colley CM, Peard T, Warner S, Walker CH..
Distribution of paraoxon hydrolytic activity in the serum of patients after myocardial infarction..
Clin Chem, (1986), 32 pp. 671-3
[33]
Mackness B, Mackness MI, Arrol S, Turkie W, Julier K, Abuasha B, et al..
Serum paraoxonase (PON1) 55 AND 192 polymorphism and paraoxonase activity and concentration in non-insulin dependent diabetes mellitus..
Atherosclerosis, (1998), 139 pp. 341-9
[34]
Blatter Garin MC, Abbott C, Messmer S, Mackness M, Durrington P, Pometta D, et al..
Quantification of human serum paraoxonase by enzyme-linked immunoassay: population differences in protein concentrations..
Biochem J, (1994), 304 pp. 549-54
[35]
Rantala M, Silaste ML, Tuominen A, Kaikkonen J, Salonen JT, Alfthan G, et al..
Dietary modifications and gene polymorphisms alter serum paraoxonase activity in healthy women..
J Nutr, (2002), 132 pp. 3012-7
[36]
Eckerson HW, Wyte CM, La Du BN..
The human serum paraoxonase/arylesterase polymorphism..
Am J Hum Genet, (1983), 35 pp. 1126-38
[37]
Garin MC, James RW, Dussoix P, Blanche H, Passa P, Froguel P, et al..
Paraoxonase polymorphism Met-Leu54 is associated with modified serum concentrations of the enzyme. A possible link between the paraoxonase gene and increased risk of cardiovascular disease in diabetes..
J Clin Invest, (1997), 99 pp. 62-6
[38]
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 pp. 1335-42
[39]
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 pp. 19-21
[40]
Mackness MI, Arrol S, Durrington PN..
Paraoxonase prevents accumulation of lipoperoxides in low- density lipoprotein [published erratum appears in FEBS Lett 1991 Nov 4;292(1-2):307]..
FEBS Lett, (1991), 286 pp. 152-4
[41]
Mackness MI, Arrol S, Abbott C, Durrington PN..
Protection of low-density lipoprotein against oxidative modification by high- density lipoprotein associated paraoxonase..
Atherosclerosis, (1993), 104 pp. 129-35
[42]
Aviram M, Rosenblat M, Bisgaier CL, Newton RS, Primo-Parmo 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 pp. 1581-90
[43]
Berliner JA, Navab M, Fogelman AM, Frank JS, Demer LL, Edwards PA, et al..
Atherosclerosis: basic mechanisms. Oxidation, inflammation, and genetics..
Circulation, (1995), 91 pp. 2488-96
[44]
Aviram M, Hardak E, Vaya J, Mahmood S, Milo S, Hoffman A, et al..
Human Serum Paraoxonases (PON1) Q and R Selectively Decrease Lipid Peroxides in Human Coronary and Carotid Atherosclerotic Lesions: PON1 Esterase and Peroxidase-Like Activities..
Circulation, (2000), 101 pp. 2510-7
[45]
Watson AD, Subbanagounder G, Welsbie DS, Faull KF, Navab M, Jung ME et al..
Structural identification of a novel pro-inflammatory epoxyisoprostane phospholipid in mildly oxidized low density lipoprotein..
J Biol Chem, (1999), 274 pp. 24787-98
[46]
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 oxidized low density lipoprotein..
J Clin Invest, (1995), 96 pp. 2882-91
[47]
Rodrigo L, Mackness B, Durrington PN, Hernandez A, Mackness MI..
Hydrolysis of platelet-activating factor by human serum paraoxonase..
Biochem J, (2001), 354 pp. 1-7
[48]
Marathe GK, Zimmerman GA, McIntyre TM..
Platelet-activating factor acetylhydrolase, and not paraoxonase-1, is the oxidized phospholipid hydrolase of high density lipoprotein particles..
J Biol Chem, (2003), 278 pp. 3937-47
[49]
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 pp. 17527-35
[50]
Rozenberg O, Rosenblat M, Coleman R, Shih DM, Aviram M..
Paraoxonase (PON1) deficiency is associated with increased macrophage oxidative stress: studies in PON1-knockout mice..
Free Radic Biol Med, (2003), 34 pp. 774-84
[51]
Rozenberg O, Shih DM, Aviram M..
Human serum paraoxonase 1 decreases macrophage cholesterol biosynthesis: possible role for its phospholipase-A2-like activity and lysophosphatidylcholine formation..
Arterioscler Thromb Vasc Biol, (2003), 23 pp. 461-7
[52]
Oda MN, Bielicki JK, Ho TT, Berger T, Rubin EM, Forte TM..
Paraoxonase 1 overexpression in mice and its effect on high-density lipoproteins..
Biochem Biophys Res Commun, (2002), 290 pp. 921-7
[53]
Fuhrman B, Volkova N, Aviram M..
Oxidative stress increases the expression of the CD36 scavenger receptor and the cellular uptake of oxidized low-density lipoprotein in macrophages from atherosclerotic mice: protective role of antioxidants and of paraoxonase..
Atherosclerosis, (2002), 161 pp. 307-16
[54]
Aviram M, Billecke S, Sorenson R, Bisgaier C, Newton R, Rosenblat M, et al..
Paraoxonase active site required for protection against LDL oxidation involves its free sulfhydryl group and is different from that required for its arylesterase/paraoxonase activities: selective action of human paraoxonase allozymes Q and R..
Arterioscler Thromb Vasc Biol, (1998), 18 pp. 1617-24
[55]
Aviram M, Rosenblat M, Billecke S, Erogul J, Sorenson R, Bisgaier CL, et al..
Human serum paraoxonase (PON 1) is inactivated by oxidized low density lipoprotein and preserved by antioxidants..
Free Radic Biol Med, (1999), 26 pp. 892-904
[56]
Jaouad L, Milochevitch C, Khalil A..
PON1 paraoxonase activity is reduced during HDL oxidation and is an indicator of HDL antioxidant capacity..
Free Radic Res, (2003), 37 pp. 77-83
[57]
Brophy VH, Jarvik GP, Richter RJ, Rozek LS, Schellenberg GD, Furlong CE..
Analysis of paraoxonase (PON1) L55M status requires both genotype and phenotype..
Pharmacogenetics, (2000), 10 pp. 453-60
[58]
Furlong CE, Cole TB, Jarvik GP, Costa LG..
Pharmacogenomic considerations of the paraoxonase polymorphisms..
Pharmacogenomics, (2002), 3 pp. 341-8
[59]
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 pp. 1451-7
[60]
Pati N, Pati U..
Paraoxonase gene polymorphism and coronary artery disease in Indian subjects..
Int J Cardiol, (1998), 66 pp. 165-8
[61]
Hasselwander O, Savage DA, McMaster D, Loughrey CM, McNamee PT, Middleton D, et al..
Paraoxonase polymorphisms are not associated with cardiovascular risk in renal transplant recipient..
Kidney International, (1999), 56 pp. 289-98
[62]
Mackness B, Mackness MI, Durrington PN, Arrol S, Evans AE, McMaster D, et al..
Paraoxonase activity in two healthy populations with differing rates of coronary heart disease..
Eur J Clin Invest, (2000), 30 pp. 4-10
[63]
Mackness MI, Mackness B, Durrington PN, Connelly PW, Hegele RA..
Paraoxonase: biochemistry, genetics and relationship to plasma lipoproteins..
Curr Opin Lipidol, (1996), 7 pp. 69-76
[64]
Mackness MI, Walker CH, Carlson LA..
Low A-esterase activity in serum of patients with fish-eye disease..
Clin Chem, (1987), 33 pp. 587-8
[65]
Mackness MI, Peuchant E, Dumon MF, Walker CH, Clerc M..
Absence of A-esterase activity in the serum of a patient with Tangier disease..
Clin Biochem, (1989), 22 pp. 475-8
[66]
Ross R..
The pathogenesis of atherosclerosis: a perspective for the 1990s..
Nature, (1993), 362 pp. 801-9
[67]
Libby P, Egan D, Skarlatos S..
Roles of infectious agents in atherosclerosis and restenosis: an assessment of the evidence and need for future research..
Circulation, (1997), 96 pp. 4095-103
[68]
Britt WJ..
Vaccines against human cytomegalovirus: time to test..
Trends Microbiol, (1996), 4 pp. 34-8
[69]
Jousilahti P, Vartiainen E, Tuomilehto J, Puska P..
Symptoms of chronic bronchitis and the risk of coronary disease..
[70]
van Lenten BJ, Wagner AC, Navab M, Fogelman AM..
Oxidized phospholipids induce changes in hepatic paraoxonase and ApoJ but not monocyte chemoattractant protein-1 via interleukin-6..
J Biol Chem, (2001), 276 pp. 1923-9
[71]
Feingold KR, Memon RA, Moser AH, Grunfeld C..
Paraoxonase activity in the serum and hepatic mRNA levels decrease during the acute phase response..
Atherosclerosis, (1998), 139 pp. 307-15
[72]
Kumon Y, Suehiro T, Ikeda Y, Hashimoto K..
Human paraoxonase-1 gene expression by HepG2 cells is downregulated by interleukin-1beta and tumor necrosis factor-alpha, but is upregulated by interleukin-6..
Life Sci, (2003), 73 pp. 2807-15
[73]
Hedrick CC, Hassan K, Hough GP, Yoo JH, Simzar S, Quinto CR, et al..
Short-term feeding of atherogenic diet to mice results in reduction of HDL and paraoxonase that may be mediated by an immune mechanism..
Arterioscler Thromb Vasc Biol, (2000), 20 pp. 1946-52
[74]
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. Loss of protective effect of HDL against LDL oxidation in aortic wall cell cocultures..
J Clin Invest, (1995), 96 pp. 2758-67
[75]
Lambert M, Boullier A, Hachulla E, Fruchart JC, Teissier E, Hatron PY, et al..
Paraoxonase activity is dramatically decreased in patients positive for anticardiolipin antibodies..
Lupus, (2000), 9 pp. 299-300
[76]
Tillett HE, Smith JW, Gooch CD..
Excess deaths attributable to influenza in England and Wales: age at death and certified cause..
Int J Epidemiol, (1983), 12 pp. 344-52
[77]
Tanimoto N, Kumon Y, Suehiro T, Ohkubo S, Ikeda Y, Nishiya K, et al..
Serum paraoxonase activity decreases in rheumatoid arthritis..
Life Sci, (2003), 72 pp. 2877-85
[78]
Mackness B, Durrington PN, Abuashia B, Boulton AJ, Mackness MI..
Low paraoxonase activity in type II diabetes mellitus complicated by retinopathy..
Clin Sci (Colch), (2000), 98 pp. 355-63
[79]
Xie XM, Zhao SP..
Congestive heart failure and paraoxonase..
Hunan Yi Ke Da Xue Xue Bao, (2002), 27 pp. 157-8
[80]
Weinbrenner T, Cladellas M, Isabel CM, Fitó M, Tomás M, Sentí M, et al..
High oxidative stress in patients with stable coronary heart disease..
Atherosclerosis, (2003), 168 pp. 99-106
[81]
Sentí M, Tomás M, Fitó M, Weinbrenner T, Covas MI, Sala J, et al..
Antioxidant paraoxonase 1 activity in the metabolic syndrome..
J Clin Endocrinol Metab, (2003), 88 pp. 5422-6
[82]
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..
[83]
Karakaya A, Ibis S, Kural T, Kose SK, Karakaya AE..
Serum paraoxonase activity and phenotype distribution in Turkish subjects with coronary heart disease and its relationship to serum lipids and lipoproteins..
Chem Biol Interact, (1999), 118 pp. 193-200
[84]
Navab M, Hama-Levy S, van Lenten BJ, Fonarow GC, Cardinez CJ, Castellani LW, et al..
Mildly oxidized LDL induces an increased apolipoprotein J/paraoxonase ratio..
J Clin Invest, (1997), 99 pp. 2005-19
[85]
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 pp. 2441-7
[86]
Jarvik GP, Hatsukami TS, Carlson C, Richter RJ, Jampsa R, Brophy VH, et al..
Paraoxonase activity, but not haplotype utilizing the linkage disequilibrium structure, predicts vascular disease..
Arterioscler Thromb Vasc Biol, (2003), 23 pp. 1465-71
[87]
Rubiés-Prat J..
Temas actuales. Hiperlipidemias y arteriosclerosis. Barcelona: Espaxs.
Temas actuales. Hiperlipidemias y arteriosclerosis. Barcelona: Espaxs, (1992), pp. 148-51
[88]
Gotto AM Jr..
Alteraciones de los lípidos y lipoproteínas. Primer de Cardiología preventiva. Barcelona: Medical Trends, SL.
Alteraciones de los lípidos y lipoproteínas. Primer de Cardiología preventiva. Barcelona: Medical Trends, SL, (1996), pp. 96-9
[89]
Banegas Banegas JR, Casasnovas Lenguas JA, Gil López E, Maiques Galan A, Mata López P, Pérez Jiménez F, et al..
. Un instrumento para la prevención cardiovascular. Madrid: Ministerio de Sanidad y Consumo.
. Un instrumento para la prevención cardiovascular. Madrid: Ministerio de Sanidad y Consumo, (2000), pp. 20-7
[90]
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 pp. 193-9
[91]
Boullier A, Hennuyer N, Tailleux A, Furman C, Duverger N, Caillaud JM, et al..
Increased levels of high-density lipoprotein cholesterol are ineffective in inhibiting the development of immune responses to oxidized low-density lipoprotein and atherosclerosis in transgenic rabbits expressing human apolipoprotein (apo) A-I with severe hypercholesterolaemia..
Clin Sci (Lond), (2001), 100 pp. 343-55
[92]
Leviev I, James R..
Simvastatin increases plasma levels of the antioxidants enzyme paraoxonase by PON1 gene activation..
Atherosclerosis, (2000), 151 pp. 41
[93]
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 pp. 1329-33
[94]
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 pp. 2083-9
[95]
Girona J, la Ville AE, Sola R, Plana N, Masana L..
Simvastatin decreases aldehyde production derived from lipoprotein oxidation..
Am J Cardiol, (1999), 83 pp. 846-51
[96]
MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20, 536 high-risk individuals: a randomised placebo-controlled trial..
[97]
Pedersen TR, Olsson AG, Faergeman O, Kjekshus J, Wedel H, Berg K, et al..
Lipoprotein changes and reduction in the incidence of major coronary heart disease events in the Scandinavian Simvastatin Survival Study (4S)..
Circulation, (1998), 97 pp. 1453-60
[98]
Lagrost L, Athias A, Lemort N, Richard JL, Desrumaux C, Chatenet-Duchene L, et al..
Plasma lipoprotein distribution and lipid transfer activities in patients with type IIb hyperlipidemia treated with simvastatin..
Atherosclerosis, (1999), 143 pp. 415-25
[99]
Kwak B, Mulhaupt F, Veillard N, Pelli G, Mach F..
The HMG-CoA reductase inhibitor simvastatin inhibits IFN-gamma induced MHC class II expression in human vascular endothelial cells..
Swiss Med Wkly, (2001), 131 pp. 41-6
[100]
Aviram M, Rosenblat M, Bisgaier CL, Newton RS..
Atorvastatin and gemfibrozil metabolites but not the parent drugs, are potent antioxidants against lipoprotein oxidation..
Atherosclerosis, (1998), 138 pp. 271-80
[101]
Beltowski J, Wojcicka G, Jamroz A..
Differential effect of 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitors on plasma paraoxonase 1 activity in the rat..
Pol J Pharmacol, (2002), 54 pp. 661-71
[102]
Elisaf M..
Effects of fibrates on serum metabolic parameters..
Curr Med Res Opin, (2002), 18 pp. 269-76
[103]
Paragh G, Balogh Z, Seres I, Harangi M, Boda J, Kovacs P..
Effect of gemfibrozil on HDL-Associated serum paraoxonase activity and lipoprotein profile in patients with hyperlipidaemia..
Clinical Drug Investigation, (2000), 19 pp. 277-82
[104]
Balogh Z, Seres I, Harangi M, Kovacs P, Kakuk G, Paragh G..
Gemfibrozil increases paraoxonase activity in type 2 diabetic patients..
A new hypothesis of the beneficial action of fibrates? Diabetes Metab, (2001), 27 pp. 604-10
[105]
Durrington PN, Mackness MI, Bhatnagar D, Julier K, Prais H, Arrol S, et al..
Effects of two different fibric acid derivatives on lipoproteins, cholesteryl ester transfer, fibrinogen, plasminogen activator inhibitor and paroxonase avtivity in type IIb hyperlipoproteinaemia..
Atherosclerosis, (1998), 138 pp. 217-25
[106]
Turay J, Grniakova V, Valka J..
Changes in paraoxonase and apolipoprotein A-I, B, C-III and E in subjects with combined familiar hyperlipoproteinemia treated with ciprofibrate..
Drugs Exp Clin Res, (2000), 26 pp. 83-8
[107]
Tsimihodimos V, Kakafika A, Tambaki AP, Bairaktari E, Chapman MJ, Elisaf M, et al..
Fenofibrate induces HDL-associated PAF-AH but attenuates enzyme activity associated with apoB-containing lipoproteins..
J Lipid Res, (2003), 44 pp. 927-34
[108]
Mackness B, Durrington PN, Boulton AJ, Hine D, Mackness MI..
Serum paraoxonase activity in patients with type 1 diabetes compared to healthy controls..
Eur J Clin Invest, (2002), 32 pp. 259-64
[109]
Malin R, Rantalaiho V, Huang XH, Wirta O, Pasternack A, Leinonen JS, et al..
Association between M/L55-polymorphism of paraoxonase enzyme and oxidative DNA damage in patients with type 2 diabetes mellitus and in control subjects..
Hum Genet, (1999), 105 pp. 179-80
[110]
Syvanne M, Ahola M, Lahdenpera S, Kahri J, Kuusi T, Virtanen KS, et al..
High density lipoprotein subfractions in non-insulin-dependent diabetes mellitus and coronary artery disease..
J Lipid Res, (1995), 36 pp. 573-82
[111]
Tkac I, Kimball BP, Lewis G, Uffelman K, Steiner G..
The severity of coronary atherosclerosis in type 2 diabetes mellitus is related to the number of circulating triglyceride-rich lipoprotein particles..
Arterioscler Thromb Vasc Biol, (1997), 17 pp. 3633-8
[112]
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 pp. 133-9
[113]
Ikeda Y, Suehiro T, Inoue M, Nakauchi Y, Morita T, Arii K, et al..
Serum paraoxonase activity and its relationship to diabetic complications in patients with non-insulin-dependent diabetes mellitus..
Metabolism, (1998), 47 pp. 598-602
[114]
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 pp. 1812-8
[115]
Sakai T, Matsuura B, Onji M..
Serum paraoxonase activity and genotype distribution in Japanese patients with diabetes mellitus..
Intern Med, (1998), 37 pp. 581-4
[116]
Pfohl M, Koch M, Enderle MD, Kühn R, Füllhase J, Karsch KR, et al..
Paraoxonase 192 Gln/Arg gene polymorphism, coronary artery disease, and myocardial infarction in type 2 diabetes..
Diabetes, (1999), 48 pp. 623-7
[117]
Hedrick CC, Thorpe SR, Fu MX, Harper CM, Yoo J, Kim SM, et al..
Glycation impairs high-density lipoprotein function..
Diabetologia, (2000), 43 pp. 312-20
[118]
Kopprasch S, Pietzsch J, Kuhlisch E, Graessler J..
Lack of association between serum paraoxonase 1 activities and increased oxidized low-density lipoprotein levels in impaired glucose tolerance and newly diagnosed diabetes mellitus..
J Clin Endocrinol Metab, (2003), 88 pp. 1711-6
[119]
Beltowski J, Wojcicka G, Jamroz A..
Leptin decreases plasma paraoxonase 1 (PON1) activity and induces oxidative stress: the possible novel mechanism for proatherogenic effect of chronic hyperleptinemia..
Atherosclerosis, (2003), 170 pp. 21-9
[120]
Zhang B, Eto S, Fan P, Bian C, Shimoji E, Saito T, et al..
Paraoxonase (Pon1) Q192R polymorphism and serum Pon1 activity in diabetic patients on maintenance hemodialysis..
Clin Nephrol, (2003), 60 pp. 257-65
[121]
Patel BN, Mackness MI, Harty DW, Arrol S, Boot-Handford RP, Durrington PN..
Serum esterase activities and hyperlipidaemia in the streptozotocin- diabetic rat..
Biochem Biophys Acta, (1990), 1035 pp. 113-6
[122]
Haley RW, Marshall WW, McDonald GG, Daugherty MA, Petty F, Fleckenstein JL..
Brain abnormalities in Gulf War syndrome: evaluation with 1H MR spectroscopy..
[123]
Paragh G, Balla P, Katona E, Seres I, Egerhazi A, Degrell I..
Serum paraoxonase activity changes in patients with Alzheimer's disease and vascular dementia..
Eur Arch Psychiatry Clin Neurosci, (2002), 252 pp. 63-7
[124]
Cherry N, Mackness M, Durrington P, Povey A, Dippnall M, Smith T, et al..
Paraoxonase (PON1) polymorphisms in farmers attributing ill health to sheep dip..
Lancet, (2002), 359 pp. 763-4
[125]
Mackness B, Durrington PN, Mackness MI..
Low paraoxonase in Persian Gulf War veterans self-reporting Gulf War syndrome..
Biochem Biophys Res Commun, (2000), 276 pp. 729-33
[126]
Hotopf M, Mackness MI, Nikolaou V, Collier DA, Curtis C, David A, et al..
Paraoxonase in Persian Gulf War veterans..
J Occup Environ Med, (2003), 45 pp. 668-75
[127]
Trznadel K, Pawlicki L, Kedziora J, Luciak M, Blaszczyk J, Buczynski A..
Superoxide anion generation, erythrocytes superoxide dismutase activity, and lipid peroxidation during hemoperfusion and hemodialysis in chronic uremic patients..
Free Radic Biol Med, (1989), 6 pp. 393-7
[128]
Ak G, Ozgonul M, Sozmen EY, Aslan SL, Sozmen B..
Renal cortical thickness and PON1 activity both decrease in chronic renal failure..
J Nephrol, (2002), 15 pp. 144-9
[129]
Itahara T, Suehiro T, Ikeda Y, Inoue M, Nakamura T, Kumon Y, et al..
Serum paraoxonase and arylesterase activities in hemodialysis patients..
J Atheroscler Thromb, (2000), 7 pp. 152-8
[130]
Biasioli S, Schiavon R, Petrosino L, de Fanti E, Cavalcanti G, Battaglia P, et al..
Paraoxonase activity and paraoxonase 1 gene polymorphism in patients with uremia..
ASAIO J, (2003), 49 pp. 295-9
[131]
Schiavon R, Battaglia P, de Fanti E, Fasolin A, Biasioli S, Targa L, et al..
HDL3-related decreased serum paraoxonase (PON) activity in uremic patients: comparison with the PON1 allele polymorphism..
Clin Chim Acta, (2002), 324 pp. 39
[132]
Juretic D, Tadijanovic M, Rekic B, Simeon-Rudolf V, Reiner E, Baricic M..
Serum paraoxonase activities in hemodialyzed uremic patients: cohort study..
Croat Med J, (2001), 42 pp. 146-50
[133]
Dantoine TF, Debord J, Charmes JP, Merle L, Marquet P, Lachatre G et al..
Decrease of serum paraoxonase activity in chronic renal failure..
J Am Soc Nephrol, (1998), 9 pp. 2082-8
[134]
Sentí M, Tomás M, Elosua R, Sala J, Masià R, Marrugat J..
The paraoxonase-1 codon 192 polymorphism is associated with fasting total cholesterol and LDL-cholesterol concentrations only in postmenopausal women. The REGICOR study..
Clin Chem Lab Med, (2002), 40 pp. 677-83
[135]
Ruiz J, Blanche H, James RW, Garin MC, Vaisse C, Charpentier G, et al..
Gln-Arg192 polymorphism of paraoxonase and coronary heart disease in type 2 diabetes..
Lancet, (1995), 346 pp. 869-72
[136]
Serrato M, Marian AJ..
A variant of human paraoxonase/arylesterase (HUMPONA) gene is a risk factor for coronary artery disease..
J Clin Invest, (1995), 96 pp. 3005-8
[137]
Antikainen M, Murtomaki S, Syvanne M, Pahlman R, Tahvanainen E, Jauhiainen M, et al..
The Gln-Arg191 polymorphism of the human paraoxonase gene (HUMPONA) is not associated with the risk of coronary artery disease in Finns..
J Clin Invest, (1996), 98 pp. 883-5
[138]
Herrmann SM, Blanc H, Poirier O, Arveiler D, Luc G, Evans A, et al..
The Gln/Arg polymorphism of human paraoxonase (PON 192) is not related to myocardial infarction in the ECTIM Study..
Atherosclerosis, (1996), 126 pp. 299-303
[139]
Suehiro T, Nakauchi Y, Yamamoto M, Arii K, Itoh H, Hamashige N, et al..
Paraoxonase gene polymorphism in Japanese subjects with coronary heart disease..
Int J Cardiol, (1996), 57 pp. 69-73
[140]
Odawara M, Tachi Y, Yamashita K..
Paraxonase polymorphism (Gln 192-Arg) is associated with coronary heart disease in Japanese noninsulin-dependent diabetes mellitus..
J Clin Endocrinol Metab, (1997), 82 pp. 2257-60
[141]
Rice GI, Ossei-Gerning N, Stickland MH, Grant PJ..
The paraoxonase GIn-Arg 192 polymorphism in subjects with ischaemic heart disease..
Coron Artery Dis, (1997), 8 pp. 677-82
[142]
Sanghera DK, Saha N, Aston CE, Kamboh MI..
Genetic polymorphism of paraoxonase and the risk of coronary heart disease..
Arterioscler Thromb Vasc Biol, (1997), 17 pp. 1067-73
[143]
Zama T, Murata M, Matsubara Y, Kawano K, Aoki N, Yoshino H, et al..
A 192Arg variant of the human paraoxonase (HUMPONA) gene polymorphism is associated with an increased risk for coronary artery disease in the Japanese..
Arterioscler Thromb Vasc Biol, (1997), 17 pp. 3565-9
[144]
Ko YL, Ko YS, Wang SM, Hsu LA, Chang CJ, Chu PH, et al..
The Gln-Arg 191 polymorphism of the human paraoxonase gene is not associated with the risk of coronary artery disease among Chinese in Taiwan..
Atherosclerosis, (1998), 141 pp. 259-64
[145]
Cascorbi I, Laule M, Mrozikiewicz PM, Mrozikiewicz A, Andel C, Baumann G, et al..
Mutations in the human paraoxonase 1 gene: frequencies, allelic linkages, and association with coronary artery disease..
Pharmacogenetics, (1999), 9 pp. 755-61
[146]
Aubó C, Sentí M, Marrugat J, Tomás M, Vila J, Sala J, et al..
Risk of myocardial infarction associated with Gln/Arg 192 polymorphism in the human paraoxonase gene and diabetes mellitus. The REGICOR Investigators..
Eur Heart J, (2000), 21 pp. 33-8
[147]
Aynacioglu AS, Kepekci Y..
The human paraoxonase Gln-Argl92 (Q/R) polymorphism in turkish patients with coronary artery disease..
Int J Cardiol, (2000), 74 pp. 33-7
[148]
Gardemann A, Philipp M, Hess K, Katz N, Tillmanns H, Haberbosch W..
The paraoxonase leu-Met54 and gln-Arg191 gene polymorphisms are not associated with the risk of coronary heart disease..
Atherosclerosis, (2000), 152 pp. 421-31
[149]
Imai Y, Morita H, Kurihara H, Sugiyama T, Kato N, Ebihara A, et al..
Evidence for association between paraoxonase gene polymorphisms and atherosclerotic diseases..
Atherosclerosis, (2000), 149 pp. 435-42
[150]
Sen-Banerjee S, Siles X, Campos H..
Tobacco smoking modifies association between gln-Arg192 polymorphism of human paraoxonase gene and risk of myocardial infarction..
Arterioscler Thromb Vasc Biol, (2000), 20 pp. 2120-6
[151]
Leus FR, Zwart M, Kastelein JJ, Voorbij HA..
PON(2) gene variants are associated with clinical manifestations of cardiovascular disease in familial hypercholesterolemia patients..
Atherosclerosis, (2001), 154 pp. 641-9
[152]
Senti M, Tomas M, Vila J, Marrugat J, Elosua R, Sala J, et al..
Relationship of age-related myocardial infarction risk and Gln/Arg 192 variants of the human paraoxonase1 gene: the REGICOR study..
Atherosclerosis, (2001), 156 pp. 443-9
[153]
Turban S, Fuentes F, Ferlic L, Brugada R, Gotto AM, Ballantyne CM, et al..
A prospective study of paraoxonase gene Q/R192 polymorphism and severity, progression and regression of coronary atherosclerosis, plasmlipid levels, clinical events and response to fluvastatin..
Atherosclerosis, (2001), 154 pp. 633-40
[154]
Ferre N, Tous M, Paul A, Zamora A, Vendrell JJ, Bardaji A, et al..
Paraoxonase Gln-Arg(192) and Leu-Met(55) gene polymorphisms and enzyme activity in a population with a low rate of coronary heart disease..
Clin Biochem, (2002), 35 pp. 197-203
[155]
Voetsch B, Benke KS, Damasceno BP, Siqueira LH, Loscalzo J..
Paraoxonase 192 Gln->Arg polymorphism: an independent risk factor for nonfatal arterial ischemic stroke among young adults..
Stroke, (2002), 33 pp. 1459-64
[156]
Zuliani G, Cherubini A, Volpato S, Palmieri E, Mecocci P, De Rango P, et al..
Genetic factors associated with the absence of atherosclerosis in octogenarians..
J Gerontol A Biol Sci Med Sci, (2002), 57 pp. M611-5
[157]
Fortunato G, Rubba P, Panico S, Trono D, Tinto N, Mazzaccara C, et al..
A paraoxonase gene polymorphism, PON 1 (55), as an independent risk factor for increased carotid intima-media thickness in middle-aged women..
Atherosclerosis, (2003), 167 pp. 141-8
[158]
Ombres D, Pannitteri G, Montali A, Candeloro A, Seccareccia F, Campagna F, et al..
The gln-Arg192 polymorphism of human paraoxonase gene is not associated with coronary artery disease in italian patients..
Arterioscler Thromb Vasc Biol, (1998), 18 pp. 1611-6
[159]
Scacchi R, Gambina G, Martini MC, Broggio E, Vilardo T, Corbo RM..
Different pattern of association of paraoxonase Gln192­>Arg polymorphism with sporadic late-onset Alzheimer's disease and coronary artery disease..
Neurosci Lett, (2003), 339 pp. 17-20
[160]
Sentí M, Tomás M, Marrugat J, Elosua R..
Paraoxonase1-192 polymorphism modulates the nonfatal myocardial infarction risk associated with decreased HDLs..
Arterioscler Thromb Vasc Biol, (2001), 21 pp. 415-20
Are you a healthcare professional authorized to prescribe or dispense medications?