839
Views
19
CrossRef citations to date
0
Altmetric
Review Article

Cytochrome P450 — physiological key factor against cholesterol accumulation and the atherosclerotic vascular process

Pages 359-370 | Published online: 08 Jul 2009

Abstract

In the early 1960s liver cytochrome P450 (P450) was known as an enzyme in drug metabolism. By the late 1970s, P450 induction was associated with elevation of plasma high‐density lipoprotein cholesterol and apolipoprotein AI indicating a reduced risk of atherosclerotic disease. Later on, 57 human P450 genes have been identified. One P450 enzyme participates in cholesterol synthesis, and several others catabolize it to oxysterols and other metabolites. Oxysterols are physiological ligands specific for liver X receptors (LXRs) in the activation of ATP‐binding cassette (ABC) transporter and other cholesterol‐lowering genes. Elevation of cholesterol leads to an endogenous induction of P450 and consequently to enhanced generation of oxysterols and activation of genes coding proteins which efflux cholesterol out of cells, transport it to the liver, catabolize and excrete cholesterol into bile, and prevent absorption of cholesterol in the intestine in the processes that maintain cellular cholesterol homeostasis and protect arteries from atherosclerosis. Peroxisome proliferator‐activated receptors (PPARs) co‐operate with LXRs and ABC transporters in cholesterol regulation. Secretion of oxysterol is a direct pathway for cellular cholesterol elimination. Several compounds induce P450 and other genes regulating cholesterol balance and prevent or regress atherosclerosis, whereas inhibition of P450 blocks oxidative reactions, promotes cholesterol accumulation, and enhances the atherosclerotic vascular process.

Abbreviations
ABC=

ATP‐binding cassette

apo=

apolipoprotein

ATP=

adenosine 5′‐triphosphate

cAMP=

cyclic adenosine monophosphate

CAR=

constitutive androstane receptor

CYP=

cytochrome

FXR=

farnesoid X receptor

HDL=

high‐density lipoprotein

HMG‐CoA=

hydroxymethyl‐glutaryl coenzyme A

LDL=

low‐density lipoprotein

LXR=

liver X receptor

OSBP=

oxysterol‐binding protein

ORP=

OSBP‐related protein

P450=

cytochrome P450

PPAR=

peroxisome proliferator‐activated receptor

RXR=

retinoid X receptor

SR‐B1=

scavenger receptor B1

SREBP=

sterol regulatory element binding protein

TZD=

thiazolidinedione

VDR=

vitamin D3 receptor

Introduction

Despite enormous research efforts and advances in treatment, atherosclerosis remains the leading cause of death in the industrialized world. Lipoproteins are intimately associated with the development of atherosclerosis. Persons with high plasma high‐density lipoprotein (HDL) cholesterol or apolipoprotein AI (apo AI), have a reduced risk of coronary heart disease (CHD) and prolonged life expectancy, whereas those with high low‐density lipoprotein (LDL) cholesterol or apo B have an increased risk of CHD and death.

Cytochrome P450 (P450, CYP), a cellular chromophore, was first named in 1961 because in spectral analysis the pigment had a peak at 450 nm Citation1,2. At that time it was thought to be one enzyme, by the mid 1960s it was connected with drug and steroid metabolism, and by the late 1970s with apo AI and HDL Citation3,4. Our original observations showed that apo AI and HDL cholesterol levels in plasma vary proportionately to microsomal P450 concentrations in the liver Citation3,Citation5. They also revealed that the increase of apo AI and HDL cholesterol in persons treated with drugs inducing protein synthesis parallels the extent of P450 induction, and put forward the hypothesis that P450 and the induction phenomenon have beneficial exploitable effects against atherosclerosis Citation3,Citation5. The decrease in plasma LDL cholesterol in proportion to the induction supported this possibility Citation6,7.

Improved analytical methods in 1980s and thereafter have revealed 57 P450 genes which code enzymes catalysing numerous reactions in important life processes in man Citation2. One P450 enzyme participates in cholesterol synthesis, and several others catabolize cholesterol to oxysterols and other metabolites. Studies in the mid 1990s identify oxysterols as signalling molecules which, through the activation of nuclear receptors, induce the transcription of genes that regulate cholesterol balance in the body Citation8,9 and as metabolites for cholesterol elimination via direct secretion Citation10–12. Several compounds induce P450 and other genes coding proteins in lipid metabolism including apo AI, the major protein constituent of HDL important in cholesterol transport. This review focuses on P450 enzymes and gene activation together with nuclear receptors and transport proteins in maintaining cholesterol homeostasis and protecting arteries from atherosclerosis.

Key messages

  • Cytochrome P450 enzymes are essential in the synthesis, metabolism, and elimination of cholesterol and affect the development of atherosclerosis in many ways.

  • Elevated cholesterol upregulates P450 that turns on mechanisms eliminating surplus cholesterol in the process of maintaining cholesterol homeostasis and atheroprotection.

  • Several compounds induce P450 and other genes that regulate cholesterol balance and prevent or regress atherosclerosis, whereas inhibition of P450 suppresses oxidative reactions and promotes atherogenesis.

P450, lipids, and apolipoproteins—earlier studies

Apo AI and HDL pick up cholesterol that cells transfer to their outer membranes and shuttle it to the liver for biliary excretion and disposal Citation13–15. The original studies in the 1970s connecting P450, apo AI, and HDL led to further investigations clarifying the effects of P450 and gene activation on lipid and protein metabolism (reviewed in Citation16). The studies linked plasma apo AI and HDL cholesterol with liver protein and phospholipid concentrations and P450 induction Citation17, and subsequently the P450 inducers were found to enhance Citation18–23 and P450 inhibitors Citation22,23 to prevent apo AI synthesis. Studies in the 1990s on transgenic animals further emphasized the significance of apo AI in atheroprotection by showing that overexpression of human apo AI gene raises HDL cholesterol, enhances reverse cholesterol transport and inhibits atherogenesis Citation24, and protects from diet‐induced atherosclerosis Citation25. In humans, an infusion of recombinant proapolipoprotein AI was found to promote faecal cholesterol excretion, implying a stimulation of reverse cholesterol transport Citation26.

P450 enzymes, oxysterols, and bile acids

Cholesterol is essential for normal cell function, but when accumulated in excess, it has deleterious effects. This is particularly true for the cells of the arterial wall during the process of atherosclerosis. Highly sophisticated regulatory systems take care to eliminate excess cholesterol out of the cells. P450 enzymes have a physiological key role in this process in catabolizing cholesterol to oxysterols and other metabolites and turning on the cholesterol‐lowering mechanisms Citation8–12,Citation27 (Tables and ).

Table I. Cytochrome P450 enzymes in the synthesis and metabolism of major oxysterols, bile acids and cholesterol.

Table II. Effects of P450‐inducing and ‐inhibiting compounds on plasma HDL cholesterol (HDL‐C) and apo AI levels, apo AI synthesis (‐S), ABCA1 and/or ABCG1 activity (‐A), development of atherosclerosisa and morbidity/mortality b.

Oxysterols are biologically active signalling molecules which as specific ligand activators for liver X receptors (LXRs) induce transcription of genes regulating cholesterol homeostasis in the body, i.e. its efflux, transport, excretion, and absorption Citation8,9,Citation28. The conversion of cholesterol to oxysterol is necessary for transcriptional activity; neither free cholesterol nor cholesterol esters have been identified as ligands for LXRs Citation29. Most oxysterols are produced through the actions of P450 enzymes that are particularly prevalent in the liver Citation30. Major oxysterols present in circulation, such as 7α‐hydroxycholesterol, 27‐hydroxycholesterol, 24S‐hydroxycholesterol, and 4β‐hydroxycholesterol, are generated by CYP7A1, CYP27A1, CYP46A1, and CYP3A4 enzymes, respectively Citation31. P450 enzymes metabolize oxysterols further to bile acids via two principal pathways: the classic or neutral pathway, and alternative or acidic pathway Citation30.

CYP7A1, cholesterol 7α‐hydroxylase, is the first and rate‐limiting hepatic enzyme in the neutral, quantitatively most significant pathway for bile acid synthesis and plays a major role in maintaining cholesterol balance. 7α‐Hydroxycholesterol is an intermediate in the synthesis of primary bile acids, cholic acid, and chenodeoxycholic. CYP8B1, sterol 12α‐hydroxylase, is essential for cholate synthesis Citation2.

CYP27A1, mitochondrial sterol 27‐hydroxylase, is found in almost all tissues and cell types including macrophages. 27‐Hydroxycholesterol is a ligand for liver X receptor (LXR) in the activation of genes coding ATP‐binding cassette (ABC) transporters which efflux cholesterol from macrophages and other cells Citation32, and it contributes to cholesterol elimination via direct secretion Citation12. The enzyme could also independently of ABC transporters enhance the efflux of non‐oxidized cholesterol to extracellular acceptors Citation33. 27‐Hydroxycholesterol is further hydroxylated by microsomal oxysterol 7α‐hydroxylase, CYP7B1, and CYP7A1 Citation34. This pathway produces mainly chenodeoxycholic acid.

CYP46A1, a P450 enzyme in neuronal cells of central nervous system, plays an active role in cholesterol metabolism in the brain Citation35,36. The enzyme generates 24S‐hydroxycholesterol which can readily pass the blood brain barrier, and the efflux into the circulation represents a pathway of cholesterol turnover in the brain and a part of reverse cholesterol transport Citation37. Hepatic CYP39A, another oxysterol 7α‐hydroxylase, converts it to bile acid Citation38. 24S‐hydroxycholesterol also is a potent activator of LXR Citation9, and could hence through transcriptional mechanisms regulate cholesterol balance in brain cells Citation39,40. It has also been found to increase ABCA1 expression and apo AI‐dependent cholesterol efflux from cultured brain endothelial cells Citation41. Elevated plasma 24S‐hydroxycholesterol levels have been measured in early stages of Alzheimer's disease, possibly reflecting on‐going demyelinization Citation42, and, in contrast, reduced levels in the advanced Alzheimer's disease, probably reflecting the loss of neuronal cells containing CYP46A1 Citation35.

CYP3A4, the predominant hepatic P450 enzyme, which is responsible for biotransformation of approximately half of commercially available drugs, also influences cholesterol and bile acid metabolism. It catabolizes cholesterol to 4β‐hydroxycholesterol, a major oxysterol found in circulation, particularly in persons treated with P450 inducers such as phenobarbital, phenytoin, or carbamazepine Citation43.

CYP51A1 gene, which is ubiquitously expressed, codes lanosterol‐14α‐demethylase, the only P450 enzyme in the biosynthesis of cholesterol. It demethylates lanosterol to cholesterol in a reaction that produces oxysterols inhibiting hydroxymethyl‐glutaryl coenzyme A (HMG‐CoA) reductase and sterol synthesis Citation44. One major intermediate in cholesterol synthesis, squalene, is in a shunt pathway metabolized to 24S,25‐epoxycholesterol, an efficient activator of LXR that may act as a signalling molecule in the feedback regulation of cholesterol synthesis Citation9.

Oxysterol binding protein (OSBP) and related proteins form a cytosolic family of OSBP‐related protein (ORPs) including 12 members Citation45,46. Several ORPs bind intracellular oxysterols which are mainly generated by P450 enzymes. Recent studies with yeast have identified ORPs as non‐vesicular transport proteins moving oxysterols among intracellular compartments Citation47, but it is still unclear whether this could apply to mammals. It is believed that ORPs act as sterol sensors co‐ordinating cellular lipid metabolism and membrane trafficking Citation46.

Oxysterols, LXRs, PPARs, and ABC transporters

LXRs, LXRα and LXRβ, are nuclear receptors which are activated by physiological concentrations of several oxysterols Citation28,Citation48. Oxysterols are the natural ligands for LXRs to induce multiple genes in the regulation of cholesterol balance Citation8,9,Citation32. LXR upregulates ABC transporters such as ABCA1, ABCG1, ABCG4, ABCG5 and ABCG8 acting in intracellular cholesterol trafficking.

ABCA1 has been identified as the key protein responsible for cellular lipid efflux Citation49–52. It transfers cholesterol and phospholipid across cellular membranes, where they are removed by apo AI Citation13. Mice deficient in LXRs accumulate foam cells in multiple peripheral tissues Citation53,54, and upregulation of ABCA1 results in protection against atherosclerosis Citation55. The significance of ABCA1 is demonstrated in Tangier disease which is characterized by a defective ABCA1 gene and lack of cholesterol and phospholipid efflux, accumulation of cholesterol esters in tissues enriched with macrophages, and premature onset of atherosclerosis Citation49–52. Liver ABCA1 has a major role in maintaining circulating HDL cholesterol levels Citation56,57, and intestinal ABCA1 contributes to it Citation58. Intestinal ABCA1 also regulates the absorption of dietary cholesterol by directing it from the enterocytes to intestinal lumen Citation59.

ABCG1, which is expressed in numerous tissues, and ABCG4 expressed in the brain, are transporters which have been found to efflux cholesterol to HDL2 and HDL3 Citation14,15. ABCG5 and ABCG8 are transporters required for biliary and intestinal excretion of cholesterol Citation60, and in the case of impaired biliary excretion enterocytes can compensate Citation61. Mutations in the genes encoding ABCG5 and ABCG8 cause sitosterolaemia, a rare recessive disorder of sterol metabolism, that is characterized by hypercholesterolaemia accompanied by xanthomas and premature coronary atherosclerosis Citation62.

The PPARs, PPARα, PPARγ, and PPARδ, are ligand‐activated transcription factors that affect lipid metabolism and the development of atherosclerosis in many ways. They mediate induction of apo AI synthesis and co‐operate with LXR and ABC transporters to efflux cholesterol from macrophages to extracellular acceptors Citation48,Citation63,64.

All members of the apolipoprotein E, CI, CII, CIV gene cluster respond to LXR activation and have been shown to serve in the ABCA1‐mediated cholesterol efflux Citation28,Citation65. Apo AIV, synthesized in the intestine and to a lesser extent in the liver, is also a direct target gene of LXRs and may contribute to the antiatherogenic effects of LXR activation Citation66,67. In addition, LXR increases the expression of lipoprotein demodelling enzymes such as cholesterol ester transfer protein, phospholipid transfer protein and lipoprotein lipase Citation28, and SREBPs (sterol regulatory element binding proteins) which mediate the synthesis of cholesterol, fatty acids, and triglycerides Citation68,69, and liver scavenger receptor B1 (SR‐B1), that mediates the selective uptake of HDL associated cholesterol esters to liver Citation70. SREBP‐1c enhances preferentially transcription of genes in fatty acid synthesis and may cause hypertriglyceridaemia, a significant adverse effect of LXR agonists. SR‐B1 is also expressed by macrophages and may, in addition to ABC transporters, contribute to cholesterol efflux Citation71.

P450 enzymes, bile acids, and nuclear receptors

The liver has an essential function to eliminate cholesterol from the body, owing to its ability to synthesize bile acids and transport cholesterol into bile. Bile acids generated by P450 enzymes activate nuclear receptors in the regulation of bile acid balance, such as farnesoid X receptor (FXR), pregnane X receptor (PXR), vitamin D3 receptor (VDR) and constitutive androstane receptor (CAR). This chapter is a short summary of P450 enzymes and bile acid regulation; for further details, see recent reviews Citation72,73.

FXR has a key role in the regulation of bile acid levels Citation72. It is expressed particularly in the liver, and also intestine, kidneys, and adrenals, and forms a heterodimer with retinoid X receptor (RXR). Accumulation of bile acids stimulates the FXR‐mediated suppression of CYP7A1and CYP8B1, key enzymes in bile acid synthesis. FXR has been found to inhibit the expression of CYP7A1 also by inducing intestinal fibroblast growth factor 19 (FGF‐19) Citation72,73. FXR also induces expression of CYP3A4 that reduces the toxicity of bile acids by oxidizing them Citation73, bile acid transporters ABCB11 and ABCC2, and intestinal bile acid binding protein, IBABP, a mediator in the enterohepatic circulation of bile acids Citation72,73.

PXR and VDR, which form heterodimers with RXR, are sensors of secondary bile acids and affect their metabolism in the liver and intestine Citation72,73. They mediate the expression of CYP3A4 gene in the metabolism of toxic bile acids into less toxic derivatives Citation72,Citation74. PXR can also downregulate CYP7A1 in response to increased levels of intracellular bile acids Citation72. CAR which is known for its role in xenobiotic metabolism also ameliorates the effects of hyperbilirubinaemia and toxic bile acids Citation75.

Induction of genes in cholesterol metabolism by drugs and other compounds

Several drugs and other compounds affecting lipid and protein metabolism are P450‐inducers. They include drugs indicated for dyslipidaemias, e.g. fibrates such as gemfibrozil, fenofibrate, and bezafibrate Citation16,Citation76,77, statins such as simvastatin, atorvastatin, fluvastatin, lovastatin Citation78,79, and pitavastatin Citation80, and cholestyramine Citation16, antidiabetic thiazolidinediones (TZDs), pioglitazone, and rosiglitazone Citation81, anticonvulsants such as phenobarbital, phenytoin, and carbamazepine Citation16, steroid hormones dexamethasone and prednisone Citation16, alcohol Citation23,Citation82,83, adenosine 2A receptor agonists Citation84, retinoids such as 9‐cis‐retinoid acid, and all‐trans retinoic acid Citation85.

Fibrates are PPARα activators which enhance apo AI synthesis, raise plasma HDL cholesterol and apo AI, and reduce LDL cholesterol and triglyceride levels Citation20–22,Citation86. They induce LXRα expression, upregulate LXRα‐dependent ABCA1 gene, promote cholesterol efflux to apo AI Citation63,64,Citation87, and inhibit cholesterol absorption in the intestine Citation88. Ketoconazole, a specific inhibitor of P450, prevents fibrate‐caused P450 induction and suppresses oxysterol generation and the activation of PPARα, LXRα, ABCA1 Citation89, and apo A1 Citation22 genes.

Statins reduce plasma LDL cholesterol and raise HDL cholesterol and apo AI. Simvastatin and atorvastatin have been found to upregulate ABCA1 and ABG1 genes in cholesterol‐loaded macrophages and to promote cholesterol efflux to apo AI and HDL3 Citation90, and pitavastatin to upregulate ABCA1 Citation91 and apo AI Citation92 genes in HepG2 cells. Several statins are ligands for PXR and CAR receptors which, via induction of P450 enzymes Citation78,79,Citation93, enhance oxysterol generation. The promoting effect of statins on cholesterol elimination has been linked with oxysterol generation, inhibition of HMG‐CoA reductase and induction of PPARα Citation92, PPARγ Citation94, LXRα Citation91, apo AI Citation80,Citation92, ABCA1, and ABCG1 genes.

Cholestyramine is a CYP7A1 inducer which in addition to reducing LDL cholesterol stimulates apo AI synthesis and raises plasma HDL cholesterol Citation95 and apo AI Citation96.

TZDs are P450 inducers and PPARγ ligands Citation97,98 that increase apo AI synthesis and plasma HDL cholesterol Citation99, and through the activation of LXR, ABCA1, ABCG1, apo E, and SR‐B1 genes promote cholesterol efflux from macrophages Citation48,Citation100,101. A PPARγ agonist may also via upregulation of CYP27A1 contribute to cholesterol removal from macrophages Citation102,103.

Anticonvulsants such as phenobarbital, phenytoin, and carbamazepine induce expression of P450 enzymes including CYP3A4, which is a target gene for nuclear receptors such as PXR Citation104, CAR Citation105, and VDR Citation106. Persons treated with these drugs show elevation of plasma HDL cholesterol and apo AI in proportion to P450 induction Citation3,Citation17, and increased generation of oxysterols, particularly 4β‐hydroxycholesterol Citation43. This oxysterol activates LXR Citation8 and could hence affect cholesterol balance through transcriptional regulation. The inverse relation of LDL cholesterol to the extent of P450 induction Citation6 could reflect increased oxysterol generation and consequent downregulation of HMG‐CoA reductase, and upregulation of the LDL receptor gene.

Alcohol is a P450 inducer Citation23,Citation82, which enhances apo AI synthesis Citation19,Citation23, raises HDL cholesterol and apo AI in proportion to P450 induction Citation83, and promotes cyclic adenosine monophosphate (cAMP)‐stimulated and particularly ABCA1‐dependent cholesterol efflux from macrophages Citation107.

AdenosineA2 receptor agonists have been found to upregulate 27‐hydroxylase and ABCA1 expression and thus have potential to enhance cholesterol elimination Citation84.

Vitamin A derivatives, retinoids exert their biological effects as ligands for RXR. They heterodimerize with many other nuclear receptors and upregulate genes involved in reverse cholesterol transport, including CYP27A1, LXRα, PPARγ, ABCA1, ABCG1, and apolipoproteins CI, CII, CIV, and E Citation102,Citation108. Significant adverse effects, including hypertriglyceridaemia, have limited the clinical usage of retinoids.

P450 enzymes, atherosclerosis, morbidity, and mortality

Both endogenous and exogenous P450 induction protects arteries from atherosclerosis and P450 inhibition has an opposite effect. Studies in the 1960s and 1970s demonstrated that carbon monoxide (CO) inactivates P450 by binding to its ferrous haem, blocks oxidative reactions Citation1, promotes cholesterol accumulation, and enhances the atherosclerotic process Citation109. More recent studies show that an exposure to CO Citation110 or environmental tobacco smoke Citation111 reduces HDL cholesterol and apo AI, and that cardiovascular events and deaths increase with the degree of CO exposure Citation112. Ketoconazole, another inhibitor of P450, suppresses oxysterol generation and the induction of LXRα, PPARα, ABCA1 Citation89, and apo AI Citation22 genes. Interferon‐γ reduces the activity of CYP27A1 Citation113 and other P450 enzymes Citation114, and ABCA1 Citation115; it also impedes reverse cholesterol transport and promotes atherogenesis Citation113,Citation115.

A genetic defect in P450 may also enhance the atherosclerotic process. Mutation in CYP27A1 gene causes a sterol storage disease, cerebrotendinous xanthomatosis (CTX), which is characterized by xanthomas in tendons and brain and premature atherosclerosis Citation116, and a defect in CYP7A1 gene could lead to hepatic cholesterol accumulation and hypercholesterolaemia, and enhance atherogenesis Citation117. A change in P450 activity and oxysterol generation also affects the LXR‐mediated induction of several proteins which participate in cholesterol transport, such as ABC transporters, apolipoproteins and lipoprotein‐modifying enzymes, and SREBPs.

Several P450‐inducing drugs and other compounds retard the progression or even regress coronary atherosclerosis (). Positive angiographic results have been obtained with gemfibrozil Citation118, bezafibrate Citation119, fenofibrate Citation120, and with statins such as simvastatin Citation121 and lovastatin Citation122, and cholestyramine Citation123. Recently, also pioglitazone was found to reduce carotid intima‐media thickness as assessed by ultrasonography in type 2 diabetic subjects Citation124. Phenobarbital inhibits cholesterol accumulation in the arterial wall and the formation of atherosclerotic plaque Citation125,126, and moderate alcohol consumption associates with less carotid Citation127 and coronary atherosclerosis Citation128.

Several large‐scale trials have evaluated the effects of fibrates, statins, and other gene‐activating compounds on cardiovascular morbidity and mortality. Recent follow‐up data from the Helsinki Heart Study Citation129 revealed that gemfibrozil reduces CHD mortality and also all‐cause mortality in the subgroup of obese persons with high triglycerides Citation130. Bezafibrate has been found to reduce the incidence of coronary events Citation119, and fenofibrate that of non‐fatal myocardial infarctions Citation131. Statins also reduce coronary events, strokes, and in addition all‐cause mortality Citation132–135. A recent meta‐analysis including 14 randomized trials of statins showed that the decrease in total mortality reflects a reduction in coronary mortality Citation135. Cholestyramine reduces the risk of definite CHD death and/or non‐fatal myocardial infarction Citation136, and pioglitazone that of the composite of all‐cause mortality, non‐fatal myocardial infarction, and stroke in type 2 diabetics who have a high risk of macrovascular events Citation137. Moderate alcohol consumption decreases CHD morbidity and mortality and also total mortality, and increases the lifespan Citation138.

Discussion and conclusions

Studies in the past three decades have identified P450 enzymes as active participants in the control of cellular homeostasis, instead of the original belief of being one enzyme in the drug detoxication system. They respond to both endogenous and exogenous signals and generate new signalling molecules acting in important metabolic processes. Our studies originating from the 1970s evaluated the effects of P450 and gene activation on the fate of lipids and proteins in the atherosclerotic vascular process. Beneficial changes in lipid and apolipoprotein levels with P450 induction were seen in different patient groups, including for example diabetic Citation139, non‐diabetic, and epileptic subjects Citation3,Citation16, and persons using alcohol regularly Citation83. Also healthy, sober subjects without any drug therapy show high plasma HDL cholesterol and HDL2 cholesterol together with high microsomal P450 activity in the liver Citation140, which was assessed by determining plasma elimination rate of orally taken antipyrine Citation7. Clinicopharmacological studies associate high P450 level and high hydroxylase activity in the liver Citation141, an exposure to two or three P450 inducers results in greater increase in P450 and HDL cholesterol Citation3,Citation16 than the use of one inducer only, and HDL cholesterol levels are related to plasma concentrations of P450‐inducing agents Citation142. Persons undergoing P450‐inducing drug therapy show reduced LDL cholesterol and elevated HDL cholesterol levels together with a rapid antipyrine elimination rate Citation7. These investigations presented a novel mechanism to prevent and treat atherosclerotic disease, i.e. induction of P450 and other genes coding proteins that regulate the cholesterol balance and protect arteries from atherosclerosis Citation3,Citation7,Citation16,17, which has then been associated with interesting findings in different population Citation82,Citation143,144, clinical Citation19,Citation82,Citation145,146 and experimental studies Citation18,Citation23,Citation147. The studies now reviewed identify P450s as enzymes which are upregulated by elevated cholesterol and gene‐activating compounds and which via cholesterol hydroxylation turn on mechanisms that eliminate excess cholesterol and counteract the atherosclerotic process, and as enzymes the inhibition of which suppresses oxidative reactions and enhances the development of atherosclerosis.

Important investigations in the 1990s discovering LXR and identifying oxysterols as ligands for it Citation8,9, and as metabolites for a direct removal of intracellular cholesterol Citation12, uncovered mechanisms for cholesterol elimination. Identification of the endogenous ligands and physiological functions for PPARs Citation48,Citation63, FXR, and several other nuclear receptors Citation148 and ABC transporters Citation28 further clarified processes controlling cholesterol homeostasis. The liver is the principal site for the synthesis and elimination of lipoproteins circulating in plasma. The effect of liver structure and function on hepatic P450 and other enzymes, lipids and proteins has been evaluated in investigations including patients with different drug regimens and liver diseases, such as fatty liver, cirrhosis Citation3,Citation5, and polycystic liver Citation149. Studies clarifying the effect of liver size on lipids and proteins associate P450 induction with a favourable change in plasma HDL cholesterol level, and the HDL/LDL profile proportionately to the increase in metabolically active liver mass as assessed by relative liver weight (liver weight/body weight) Citation150. The effect of liver size has been found important also in the evaluation of circulating 24S‐hydroxycholesterol levels that reflect the balance between cerebral production and hepatic metabolism of the oxysterol Citation151. A recent study showing that increased hepatic LXRα expression protects from atherosclerosis in susceptible mice which were fed a Western diet demonstrated that LXRα activation in the liver also has beneficial effects Citation152. The benefit of the LXRα activation was lost upon treatment with synthetic LXR agonist raising triglycerides due to activation of SREBP‐1c pathway. The study shows an important difference between endogenous oxysterols and pharmacological LXR agonists that promote lipogenesis and also emphasizes the need of new agonists without such effects.

The ability of different compounds to activate key proteins controlling cholesterol homeostasis and to attenuate the development of atherosclerosis has led to intensive search for new agents to prevent and treat atherosclerotic disease. Studies on LXR agonists in mouse models concluded that activation of LXR inhibits the development of atherosclerotic lesions, probably via direct actions of the LXR ligands on vascular gene expression Citation153–155. A recent study also showed that macrophage LXR expression is necessary for the antiatherogenic effects of LXR agonists and that an LXR agonist increases the expression of ABCA1 within pre‐existing atherosclerotic lesions Citation156. It also demonstrated that an LXR agonist not only inhibits the development but also induces regression of atherosclerotic lesions. Another recent study presented a direct proof that administration of a synthetic LXR agonist substantially increases reverse cholesterol transport of macrophage‐derived cholesterol in vivoCitation157. It also established that LXR agonist promotes cholesterol efflux from macrophages into liver and ultimately into bile and faeces.

Acknowledgements

The author is grateful for excellent collaboration especially to Eero Sotaniemi (†), Olavi Pelkonen, Markku Savolainen, and Vilho Myllylä from the Medical Faculty, University of Oulu, Finland; Christian Ehnholm from the National Public Health Institute, Helsinki; and Heikki Vapaatalo from the Department of Pharmacology, University of Helsinki. The studies were supported by the Academy of Finland and the Paavo Nurmi Foundation, Finland.

References

  • Omura T., Sato R. The carbon‐monoxide‐binding pigment in liver microsomes. II. Solubilization, purification and properties. J Biol Chem 1964; 239: 2379–85
  • Nebert D. W., Russell D. W. Clinical importance of the cytochromes P450. Lancet 2002; 360: 1155–62
  • Luoma P. V., Sotaniemi E. A., Pelkonen R. O., Myllylä V. V. Plasma high density lipoprotein cholesterol and hepatic cytochrome P‐450 concentrations in epileptics undergoing anticonvulsant treatment. Scand J Clin Lab Invest 1980; 40: 163–7
  • Luoma P. V., Sotaniemi E. A., Pelkonen R. O., Ehnholm C. Plasma high density lipoprotein and liver microsomal enzyme activity in man. The Medical Research Council 1977–1979, A. S Pilli‐Sihvola, T. H Laaksovirta. The Academy of Finland, Helsinki 1980; 71
  • Luoma P. V., Sotaniemi E. A., Pelkonen R. O., Arranto A., Ehnholm C. Plasma high density lipoproteins and hepatic microsomal induction—relation to histological changes in the liver. Eur J Clin Pharmacol 1982; 23: 275–82
  • Luoma P. V., Sotaniemi E. A., Pelkonen R. O. Inverse relation of serum LDL cholesterol and the LDL/HDL cholesterol ratio to liver microsomal induction in man. Res Commun Chem Pathol Pharmacol 1983; 42: 173–6
  • Luoma P. V., Sotaniemi E. A., Arranto A. J. Serum LDL cholesterol and LDL/HDL cholesterol ratio and liver microsomal induction evaluated by antipyrine kinetics. Scand J Clin Lab Invest 1983; 43: 671–5
  • Janowski B. A., Willy P. J., Devi T. R., Falck J. R., Mangelsdorf D. J. An oxysterol signalling pathway mediated by the nuclear receptor LXRα. Nature 1996; 383: 728–31
  • Lehmann J. M., Kliewer S. A., Moore L. B., Smith‐Oliver T. A., Oliver B. B., Su J. L., et al. Activation of the nuclear receptor LXR by oxysterols defines a new hormone response pathway. J Biol Chem 1997; 272: 3137–40
  • Björkhem I., Andersson O., Diczfalusy U., Sefastik B., Xiu R. ‐J., Duan C., et al. Atherosclerosis and sterol 27‐hydroxylase: Evidence for a role of this enzyme in elimination of cholesterol from human macrophages. Proc Natl Acad Sci U S A 1994; 91: 8592–6
  • Reiss A. B., Martin K. O., Javitt N. B., Martin D. W., Grossi E. A., Galloway A. C. Sterol 27‐hydroxylase: high levels of activity in vascular endothelium. J Lipid Res 1994; 35: 1026–30
  • Björkhem I., Diczfalusy U., Lütjohann D. Removal of cholesterol from extrahepatic sources by oxidative mechanisms. Curr Opin Lipidol 1999; 10: 161–5
  • Wang N., Silver D. L., Costet P., Tall A. R. Specific binding of apoA‐I, enhanced cholesterol efflux, and altered plasma membrane morphology in cells expressing ABCA1. J Biol Chem 2000; 275: 33053–8
  • Wang N., Lan D., Chen W., Matsuura F., Tall A. R. ATP‐binding cassette transporters G1 and G4 mediate cellular cholesterol efflux to high‐density lipoproteins. Proc Natl Acad Sci U S A 2004; 101: 9774–9
  • Klucken J., Büchler C., Orsó E., Kaminski W. E., Porsch‐Özcϋrϋmez M., Liebisch G., et al. ABCG1 (ABC8), the human homolog of the drosophila white gene, is regulator of macrophage cholesterol and phospholipid transport. Proc Natl Acad Sci U S A 2000; 97: 817–22
  • Luoma P. V. Gene activation, apolipoprotein A‐I/ high density lipoprotein, atherosclerosis prevention and longevity. Pharmacol Toxicol 1997; 81: 57–64
  • Luoma P. V., Savolainen M. J., Sotaniemi E. A., Pelkonen R. O., Arranto A. J., Ehnholm C. Plasma high density lipoprotein and liver lipids and proteins in man. Relation to hepatic histology and microsomal enzyme induction. Acta Med Scand 1983; 214: 103–9
  • Chao Y. U., Pickett C. B., Yamin T. T., Guo L. S., Alberts A., Kroon P. A. Phenobarbital induces rat liver apoliprotein A‐I mRNA. Mol Pharmacol 1985; 27: 394–8
  • Malmendier C. L., Delcroix C. Effect of alcohol intake on high and low density lipoprotein metabolism in healthy volunteers. Clinica Chimica Acta 1985; 152: 281–8
  • Malmendier C., Delcroix C. Effects of fenofibrate on high and low density lipoprotein metabolism in heterozygous familial hypercholesterolemia. Atherosclerosis 1985; 55: 161–9
  • Saku K., Gartside P. S., Hynd B. A., Kashyap M. L. Mechanism of action of gemfibrozil on lipoprotein metabolism. J Clin Invest 1985; 75: 1702–12
  • Tam S. P. Effects of gemfibrozil and ketoconazole on human apolipoprotein A‐I and E levels in two hepatoma cell lines HepG2 and HepG3. Atherosclerosis 1991; 91: 51–61
  • Tam S. P. Effect of ethanol on lipoprotein secretion in two human hepatoma cell lines HepG2 and HepG3. Alcohol Clin Exp Res 1992; 16: 1021–8
  • Rubin E. M., Krauss M. R., Spangler E. A., Verstuyft J. G., Clift M. S. Inhibition of early atherogenesis in transgenic mice by human apoprotein A‐I. Nature 1991; 353: 265–7
  • Duverger N., Kruth N. H., Emmanuel F., Caillaud J. M., Viglietta C., Castro G., et al. Inhibition of atherosclerosis development in cholesterol‐fed human apolipoprotein A‐I transgenic rabbits. Circulation 1996; 94: 713–7
  • Eriksson M., Carlson L. A., Miettinen T. A., Angelin B. Stimulation of fecal steroid excretion after infusion of recombinant proapolipoprotein A‐I. Potential reverse cholesterol transport in humans. Circulation 1999; 100: 594–8
  • Russell D. W. Oxysterol biosynthetic enzymes. Biochim Biophys Acta 2000; 1529: 126–35
  • Tontonoz P., Mangelsdorf D. J. Liver X receptors signalling pathways in cardiovascular disease. Mol Endocrinol 2003; 17: 985–93
  • Ory D. S. Nuclear receptor signalling in the control of cholesterol homeostasis: have the orphans found a home. Circ Res 2004; 95: 660–70
  • Russell D. W. The enzymes, regulation and genetics of bile acid synthesis. Annu Rev Biochem 2003; 72: 137–74
  • Björkhem I., Diczfalusy U. Oxysterols–friends, foes or just fellow passengers. Arterioscler Thromb Vasc Biol 2002; 22: 734–42
  • Fu X., Menke J. G., Chen Y., Zhou G., MacNaul K. L., Wright S. D., et al. 27–hydroxycholesterol is an endogenous ligand for liver X receptor in cholesterol loaded cells. J Biol Chem 2001; 276: 38378–87
  • Escher G., Krozowski Z., Croft K. D., Sviridov D. Expression of sterol 27‐hydroxylase (CYP27A1) enhances cholesterol efflux. J Biol Chem 2003; 278: 11015–9
  • Norlin M., Andersson U., Björkhem I., Wikwall K. Oxysterol 7α‐hydroxylase activity by cholesterol 7α‐hydroxylase (CYP7A). J Biol Chem 2000; 275: 34046–53
  • Björkhem I., Meaney S. Brain cholesterol: long secret life behind a barrier. Arterioscler Thromb Vasc Biol 2004; 24: 806–15
  • Dietschy J. M., Turley S. D. Cholesterol metabolism in the central nervous system during early development and in the mature animal. J Lipid Res 2004; 45: 1375–97
  • Lütjohann D., Breuer O., Ahlborg G., Nennesmo I., Sidén Å., Diczfalusy U., et al. Cholesterol homeostasis in human brain: Evidence for an age‐dependent flux of 24S‐hydroxycholesterol from the brain into circulation. Proc Natl Acad Sci U S A 1996; 93: 9799–804
  • Li‐Hawkins J., Lund E. G., Bronson A. D., Russell D. W. Expression cloning of an oxysterol 7α‐hydroxylase selective for 24‐hydroxycholesterol. J Biol Chem 2000; 275: 16543–48
  • Abildayeva K., Jansen P. J., Hirsch‐Reinshagen V., Bloks W. V., Bakker A. H. F., Ramaekers F. C. S., et al. 24(S)‐hydroxycholesterol participates in a liver X receptor‐controlled pathway in astrocytes that regulates apolipoprotein E‐mediated cholesterol efflux. J Biol Chem 2006; 281: 12799–808
  • Pfrieger F. W. Outsourcing in the brain: do neurons depend on cholesterol delivery by astrocytes. Bioessays 2002; 25: 72–8
  • Panzenboeck U., Balazs Z., Sovic A., Hrzenjak A., Levak‐Frank S., Wintersperger A., et al. ABCA1 and scavenger receptor class B, type I, are modulators of reverse cholesterol transport at an in vitro blood‐brain barrier constituted porcine brain capillary endothelial cells. J Biol Chem 2002; 277: 42781–9
  • Lütjohann D., Papassotiropoulos A., Björkhem I., Locatelli S., Bagli M., Oehring R. D., et al. Plasma 24S‐hydroxycholesterol (cerebrosterol) is increased in Alzheimer and vascular demented patients. J Lipid Res 2000; 41: 195–8
  • Bodin K., Bretillon L., Aden Y., Bertilsson L., Broome U., Einarsson C., et al. Antiepileptic drugs increase plasma levels of 4β‐hydroxycholesterol in humans. J Biol Chem 2001; 276: 38685–9
  • Gibbons G. F. The role of cytochrome P450 in the regulation of cholesterol biosynthesis. Lipids 2002; 37: 1163–70
  • Ridgway N. D., Dawson P. A., Ho Y. K., Brown M. S., Goldstein J. L. Translocation of oxysterol binding protein to Golgi apparatus triggered by ligand binding. J Cell Biol 1992; 116: 307–19
  • Olkkonen V. M., Johansson M., Suchanek M., Yan D., Hynynen R., Ehnholm C., et al. The OSBP‐related proteins (ORPS): global sterol sensors for co‐ordination of cellular lipid metabolism, membrane trafficking and signalling processes. Biochem Soc Transact 2006; 34: 389–91
  • Raychaudhuri S., Im Y. J., Hurley J. H., Prinz W. A. Nonvesicular sterol movement from plasma membrane to ER requires oxysterol‐binding protein related proteins and phosphoinositides. J Cell Biol 2006; 173: 107–19
  • Chawla A., Boisvert W., Lee C. ‐H., Laffitte B. A., Barak Y., Joseph S. B., et al. A PPARγ– LXR–ABCA1 pathway in macrophages is involved in cholesterol efflux and atherogenesis. Mol Cell 2001; 7: 161–71
  • Bodzioch M., Orso E., Klucken J., Langmann T., Bottcher A., Diederich W., et al. The gene encoding ATP‐binding cassette transporter 1 is mutated in Tangier disease. Nat Genet 1999; 22: 347–51
  • Brooks‐Wilson A., Marcil M., Clee S. M., Zhang L. H., Roomp K., van Dam M., et al. Mutations in ABC1 in Tangier disease and familial high‐density lipoprotein deficiency. Nat Genet 1999; 22: 336–45
  • Rust S., Rosier M., Funke H., Real J., Amoura Z., Piett J. C., et al. Tangier disease is caused by mutations in the gene encoding ATP‐binding cassette transporter 1. Nat Genet 1999; 22: 352–5
  • Lawn R. M., Wade D. P., Garvin M. R., Wang X., Schwartz K., Porter J. G., et al. The Tangier disease gene product ABC1 controls the cellular apolipoprotein‐mediated lipid removal pathway. J Clin Invest 1999; 104: R25–31
  • Tangirala R. K., Bischoff E. D., Joseph S. B., Wagner B. L., Walczak R., Laffitte B. A., et al. Identification of macrophage liver X receptors as inhibitors of atherosclerosis. Proc Natl Acad Sci U S A 2002; 99: 11896–901
  • Schuster G. U., Parini P., Wang L., Alberti S. A., Steffensen K. R., Hansson G. K., et al. Accumulation of foam cells in liver X receptor‐deficient mice. Circulation 2002; 106: 1147–53
  • Singaraja R. R., Fievet C., Castro G., James E. R., Hennuyer N., Clee S. M., et al. Increased ABCA1 activity protects against atherosclerosis. J Clin Invest 2002; 110: 35–42
  • Brewer H. B., Jr, Remaley A. T., Neufeld E. B., Basso F., Jouce C. Regulation of plasma high‐density lipoprotein levels by the ABCA1 transporter and the emerging role of high‐density lipoprotein in the treatment of cardiovascular disease. Arterioscler Thromb Vasc Biol 2004; 24: 1755–60
  • Timmins J. M., Lee J. ‐Y., Boydyguina E., Kluckman K. D., Brunham L. R., Mulya A., et al. Targeted inactivation of hepatic Abc 1 causes profound hypoalphalipoproteinemia and kidney hypercatabolism of apoA‐I. J Clin Invest 2005; 115: 1333–42
  • Brunham L. R., Kruit K. J., Iqbal J., Fievet C., Timmins J. M., Pape T., et al. Intestinal ABCA1 directly contributes to HDL biogenesis in vivo. J Clin Invest 2006; 116: 1052–62
  • Repa J. J., Turley S. D., Lobaccaro J. A., Medina J., Li L., Lustig K., et al. Regulation of absorption of ABCA1‐mediated efflux of cholesterol by RXR heterodimers. Science 2000; 289: 1524–9
  • Yu L., Li‐Hawkins J., Hammer R. E., Berge K. E., Horton J. D., Cohen J. C., et al. Overexpression of ABCG5 and ABCG8 promotes biliary cholesterol secretion and reduces fractional absorption of dietary cholesterol. J Clin Invest 2002; 110: 671–80
  • Kruit J. K., Plösch T., Havinga R., Boverhof R., Groot P. E. H., Groen A. K., et al. Increased fecal neutral steroid loss upon liver‐X‐receptor activation is independent of biliary sterol secretion in mice. Gastroenterology 2005; 128: 147–56
  • Berge K. E., Tian H., Graf G. A., Yu L., Grishin N. V., Schutz J., et al. Accumulation of dietary cholesterol in sitosterolemia caused by mutations in adjacent ABC transporters. Science 2000; 290: 1771–5
  • Chinetti G., Lestavel S., Bocher V., Remaley A. T., Neve B., Torra I. P., et al. PPARα and PPARγ activators induce cholesterol removal from human macrophage foam cells through stimulation of the ABCA1 pathway. Nat Med 2001; 7: 53–8
  • Li A. C., Glass C. K. PPAR‐ and LXR‐dependent pathways controlling lipid metabolism and the development of atherosclerosis. J Lipid Res 2004; 45: 2161–73
  • Mak P. A., Laffitte B. A., Resrumaux C., Joseph S. B., Curtiss L. K., Mangelsdorf D. J., et al. Regulated expression of the apolipoproteinE/C‐I/C‐IV/C‐CII gene cluster in murine and human macrophage. A critical role for nuclear liver X receptors α and β. J Biol Chem 2002; 277: 31900–8
  • Duverger N., Tremp G., Caillaud J. M., Emmanuel F., Castro G., Fruchart J. M., et al. Protection against atherogenesis in mice mediated by human apolipoprotein AIV. Science 1996; 273: 966–8
  • Liang Y., Jiang X. ‐C., Liu R., Liang G., Beyer T. P., Gao H., et al. Liver X receptors (LXRs) regulate apolipoprotein AIV—implications of the antiatherosclerotic effect of LXR agonists. Mol Endocrinol 2004; 18: 2000–10
  • Repa J. J., Liang G., Ou J., Bashmakov Y., Lobaccaro J. M. A., Shimomura I., et al. Regulation of mouse sterol regulatory element‐binding protein‐1c (SREBP‐1c) by oxysterol receptors LXRα and LXRβ. Genes Dev 2000; 14: 2819–30
  • Horton J. D., Goldstein J. L., Brown M. S. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J Clin Invest 2002; 109: 1125–31
  • Malerod L., Juvet L. K., Hanssen‐Bauer A., Eskild W., Berg T. Oxysterol‐activated LXRalpha/RXR induces hSR‐BI‐promoter activity in hepatoma cells and preadipocytes. Biochem Biophys Res Commun 2002; 299: 916–23
  • Ji Y., Jian B., Wang N., Sun Y., de la Llera Moya M., Phillips M. C., et al. Scavenger receptor BI promotes high density lipoprotein‐mediated cellular cholesterol efflux. J Biol Chem 1997; 272: 20982–5
  • Eloranta J. J., Kullak‐Ublick G. A. Coordinate transcriptional regulation of bile acid homeostasis and drug metabolism. Arch Biochem Biophys 2005; 433: 397–412
  • Claudel T., Staels B., Kuipers F. The farnesoid X receptor—a molecular link between bile acid and lipid and glucose metabolism. Arterioscler Thromb Vasc Biol 2005; 25: 2020–31
  • Bodin K., Lindbom U., Diczfalusy U. Novel pathways of bile acid metabolism involving CYP3A4. Biochim Biophys Acta 2005; 1687: 84–93
  • Goodwin B., Moore J. T. CAR: detailing new models. Trends Pharmacol Sci 2004; 25: 437–41
  • Prueksaritanont T., Richards K. M., Qui Y., Strong‐Basalyga K., Miller A., Li C., et al. Comparative effects of fibrates on drug metabolizing enzymes in human hepatocytes. Pharmaceut Res 2005; 22: 71–8
  • Amacher D., Beck D., Schomaker S. J., Kenny C. V. Hepatic microsomal enzyme induction, β‐oxidation, and cell proliferation following administration of clofibrate, gemfibrozil or bezafibrate in the CD rat. Toxicol Appl Pharmacol 1997; 142: 143–50
  • Kocarek T., Dahn M. S., Cai H., Strom S. C., Mercer‐Haines N. A. Regulation of CYP2B6 and CYP3A expression by hydroxymethylglutaryl coenzyme A inhibitors in primary cultured human hepatocytes. Drug Metab Dispos 2002; 30: 1400–5
  • Kobayashi K., Yamanaka Y., Iwazaki N., Nakajo I., Hosokawa M., Negishi M., et al. Identification of HMG‐CoA reductase inhibitors as activators for human, mouse and rat constitutive androstane receptor. Drug Metab Dispos 2005; 33: 924–9
  • Fan P., Zhang B., Kuroki S., Saku K. Pitavastatin, a potent hydroxymethylglutaryl coenzyme A reductase inhibitor, increases cholesterol 7α‐hydroxylase gene expression in HepG2 cells. Circ J 2004; 68: 1061–6
  • Sahi J., Black C. B., Hamilton G. A., Zheng X., Jolley S., Rose K. A., et al. Comparative effects of thiazolidinediones on in vitro P450 enzyme induction and inhibition. Drug Metab Dispos 2003; 31: 439–46
  • Lieber C. S. To drink (moderately) or not to drink?. N Engl J Med 1984; 310: 846–8
  • Luoma PV Sotaniemi E. A., Pelkonen R. O., Ehnholm C. High‐density lipoproteins and hepatic microsomal enzyme induction in alcohol consumers. Res Commun Chem Pathol Pharmacol 1982; 37: 91–6
  • Reiss A. B., Rahman M., Chan E. S., Montesinos C., Awadallah N. W., Cronstein B. N. Adenosine A2A receptor occupancy stimulates expression of proteins involved in reverse cholesterol transport and inhibits foam cell formation in macrophages. J Leucoc Biol 2004; 76: 727–34
  • Langmann T., Liebisch G., Moehle C., Schifferer R., Dayoab R., Heiduczek S., et al. Gene expression profiling identifies retinoids as potent inducers of macrophage lipid efflux. Biochim Biophys Acta 2005; 1740: 155–61
  • Staels B., Dallongville D., Auwerx J., Schhoonjans K., Leitersdorf E., Fruchart J. C. Mechanism of action of fibrates on lipid and lipoprotein metabolism. Circulation 1998; 98: 2088–93
  • Arakawa R., Tamehiro N., Nishimaki‐Mogami T., Ueda K., Yokoyama S. Fenofibric acid, an active form of fenofibrate increases apolipoprotein A‐I‐mediated high‐density lipoprotein biogenesis by enhancing transcription of ATP‐binding cassette transporter AI gene in a liver X receptor‐dependent manner. Arterioscler Thromb Vasc Biol 2005; 25: 1193–7
  • Knight B. L., Patel D. D., Humphreys S. M., Wiggins D., Gibbons G. F. Inhibition of cholesterol absorption associated with a PPARα‐dependent increase in ABC binding cassette transporter A1 in mice. J Lipid Res 2003; 44: 2049–58
  • Guan J. ‐Z., Tamasava N., Murakami H., Matsui J., Yamato K., Suda T. Clofibrate, a peroxisome‐proliferator, enhances reverse cholesterol transport through cytochrome P450 activation and oxysterol generation. Tohoku J Exp Med 2003; 201: 251–9
  • Argmann C. A., Edwards J. Y., Sawyez C. G., O'Neil C. H., Hegele R. A., Pickering J. G., et al. Regulation of macrophage cholesterol efflux through hydroxymethylglutaryl‐CoA reductase inhibition. J Biol Chem 2005; 280: 22212–21
  • Maejima T., Yamazaki H., Aoki T., Tamaki T., Sato F., Kitahara M., et al. Y. Effect of pitavastatin on apolipoprotein A‐I production in HepG2 cell. Biochem Biophys Res Commun 2004; 324: 835–9
  • Martin G., Duez H., Blanquart C., Berezowski V., Poulain P., Fruchart J. C., et al. Statin‐induced inhibition of the Rho‐signaling pathway activates PPARα and induces HDL apoA‐I. J Clin Invest 2001; 107: 1423–32
  • Lehmann J. M., McKee D. D., Watson M. A., Willson T. M., Moore J. T., Kliewer S. A. Human orphan nuclear receptor PXR is activated by compounds that regulate CYP3A4 gene expression and cause drug interactions. J Clin Invest 1998; 102: 1016–23
  • Fajas L., Schoonjans K., Gelman L., Kim J. B., Najib J., Martin G., et al. Regulation of peroxisome proliferator‐activated receptor gamma expression by adipocyte differentation and determination factor1/sterol regulatory element binding protein 1: implications for adipocyte differentiation and metabolism. Mol Cell Biol 1999; 19: 5495–503
  • Glueck C. J. Nonpharmacologic and pharmacologic alteration of high‐density lipoprotein cholesterol: therapeutic approaches to prevention of atherosclerosis. Am Heart J 1985; 110: 1107–15
  • Shepherd J. The effect of cholestyramine on high density lipoprotein metabolism. Atherosclerosis 1979; 33: 433–44
  • Forman B. M., Tontonoz P., Chen J., Brun R. P., Spiegelman B. M., Evans R. M. 15‐deoxy‐Δ 12,14‐prostaglandin J2 is a ligand for the adipocyte determination factor PPARγ. Cell 1995; 83: 803–12
  • Lehmann J. M., Moore L. B., Smith‐Oliver T. A., Wilkison W. O., Wilson T. M., Kliewer S. A. An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator‐activated receptor PPARγ. J Biol Chem 1995; 270: 12953–6
  • Myers C. D., Kashyap M. L. Pharmacological augmentation of high‐density lipoproteins: mechanisms of currently available and emerging therapies. Curr Opin Cardiol 2005; 20: 307–12
  • Akiyama T. E., Sakai S., Lambert G., Nicol C. J., Matsusue K., Pimprale S., et al. Conditional disruption of the peroxisome proliferator‐activated receptor γ gene in mice results in lowered expression of ABCA1, ABCG1 and apo E in macrophages and reduced cholesterol efflux. Mol Cell Biol 2002; 22: 2607–19
  • Chinetti G., Gbaguidi F. G., Griglio S., Mallat Z., Antonucci M., Poulain P., et al. CLA‐1/SR‐BI is expressed in atherosclerotic lesion macrophages and regulated by activators of peroxisome proliferator‐activated receptors. Circulation 2000; 101: 2411–7
  • Szanto A., Benko S., Szatmari I., Balint L. B., Furtos I., Rϋhl R., et al. Transcriptional regulation of human CYP27 integrates retinoid, peroxisome proliferator‐activated receptor, and liver X receptor signalling in macrophages. Mol Cell Biol 2004; 24: 8154–66
  • Quinn C. M., Jessup W., Wong J., Kritharides L., Brown A. J. Expression and regulation of sterol 27‐hydroxylase (CYP27A1) in human macrophages: a role for RXR and PPARγ ligands. Biochem J 2005; 385: 823–30
  • Luo G., Cunningham M., Kim S., Burn T., Lin J., Sinz M., et al. 3A4 induction by drugs: correlation between a pregnane x receptor receptor gene assay and CYP3A4 expression in human hepatocytes. Drug Metab Disp 2002; 30: 795–802
  • Moore L. B., Parks D. J., Jonest S. A., Bledsoe R. K., Consler T. G., Stimmel J. B., et al. Orphan nuclear receptors constitutive androstane receptor and Pregnane receptor share xenobiotic and steroid ligands. J Biol Chem 2000; 275: 15122–7
  • Makishima M., Lu T. T., Xie W., Whitfield G. K., Domoto H., Evans R. M., et al. Vitamin D receptor as an intestinal bile acid sensor. Science 2002; 296: 1313–6
  • Beulens J. W., Sierksma A., van Tol A., Fournier N., van Gent T., Paul J. ‐L., et al. Moderate alcohol consumption increases cholesterol efflux mediated by ABCA1. J Lipid Res 2004; 45: 1716–23
  • Costet P., Lalanne F., Gerbod‐Gionnone M., Molina J. R., Fu X., Lund E. G., et al. Retinoid acid receptor‐mediated induction of ABCA1 in macrophages. Mol Cell Biol 2003; 23: 7756–66
  • Astrup P., Kjeldsen K., Wanstrup J. Effects of carbon monoxide exposure on the arterial walls. Ann NY Acad Sci 1970; 174: 294–300
  • Haustein K. O., Krause J., Haustein H., Rasmussen T., Cort N. Comparison of the effects of combined nicotine replacement therapy vs. cigarette smoking in males. Nicotine Tob Res 2003; 5: 195–203
  • Moffatt R. J., Stamford B. A., Biggerstaff K. D. Influence of worksite tobacco smoke on serum lipoprotein profiles of female nonsmokers. Metabolism 1995; 44: 1536–9
  • Hedblad B., Ögren M., Engström G., Wollmer P., Janzon L. Heterogeneity of cardiovascular risk among smokers is related to degree of carbon monoxide exposure. Atherosclerosis 2005; 179: 177–83
  • Reiss A. B., Patel C. A., Rahman M. M., Chan E. S. L., Hasneen K., Montesinos M. M., et al. Interferon‐γ impedes reverse cholesterol transport and promotes foam cell transformation in TPH‐1 human monocytes/macrophages. Med Sci Monit 2004; 10: BR420–5
  • Williams J. F. Cytochrome isoforms. Regulation during infection, inflammation and by cytokines. J Fla Med Assoc 1991; 78: 517–9
  • Panousis V. G., Zuckerman S. H. Interferon‐γ induces downregulation of Tangier disease gene (ATP‐binding cassette transporter 1) in macrophage derived foam cells. Arterioscler Thromb Vasc Biol 2000; 20: 1565–71
  • von Bahr S., Movin T., Papadogiannakis N., Pikuleva I., Rönnow P., Diczfalusy U., et al. Mechanism of accumulation of cholesterol and cholestanol in tendons and the role of sterol 27‐hydroxylase (CYP27A1). Arterioscler Thromb Vasc Biol 2002; 22: 1129–35
  • Pullinger C. R., Eng C., Salen G., Shefer S., Barra A. K., Erickson S. K., et al. Human cholesterol 7α‐hydroxylase (CYP7A1) deficiency has a hypercholesterolemic phenotype. J Clin Invest 2002; 110: 109–17
  • Frick M. H., Syvänne M., Nieminen M. S., Kauma H., Majahalme S., Virtanen V., et al. Prevention of the angiographic progression of coronary vein‐graft atherosclerosis by gemfibrozil after coronary by‐pass surgery in men with low levels of HDL cholesterol. Circulation 1997; 96: 2137–43
  • Ericsson C. G., de Faire U., Grip L., Svane B., Hamsten A., Nilsson J. Angiographic assessment of effects of bezafibrate on progression of coronary artery disease in young male postinfarction patients. Lancet 1996; 347: 849–53
  • Effect of fenofibrate on progression of coronary artery disease in type 2 diabetes: the Diabetes Atherosclerosis Intervention Study, a randomised study. Lancet 2001; 357: 905–10
  • Effect of simvastatin on coronary atheroma. The Multicentre Anti‐Atheroma Study (MAAS). Lancet 1994; 344: 633–8
  • Blankenhorn D. H., Azen S. P., Kramsch D. M., Mack W. J., Cashin‐Hemphill L., Hodis H. N., , the MARS Research Group, et al. Coronary angiographic changes with lovastatin therapy; The Monitored Atherosclerosis Regression Study (MARS). Ann Intern Med 1993; 119: 969–76
  • Watts G. F., Lewis B., Brunt J. N. H., Lewis E. s., Coltart D. J., Smith L. D., et al. Effects on coronary artery disease of lipid lowering diet, or diet plus cholestyramine. In St Thomas Atherosclerosis Regression Study (STARS). Lancet 1992; 339: 563–9
  • Langenfeld M. R., Forst T., Hohberg C., Kann P., Lubben G., Konrad T., et al. Pioglitazone decreases carotid intima‐media thickness independently of glycemic control in patients with type 2 diabetes mellitus. Results from a controlled randomized study. Circulation 2005; 111: 2525–31
  • Myasnikov A. L. Influence of some factors on development of experimental cholesterol atherosclerosis. Circulation 1958; 17: 99–113
  • Salvador R. A., Atkins C., Haber S., Kozma C., Conney A. H. Effect of phenobarbital and chlorcyclizine on the development of atheromatosis in the cholesterol‐fed rabbit. Biochem Pharmacol 1970; 19: 1975–81
  • Kiehl S., Willeit J., Rungger G., Egger G., Oberhollenzer F., Bonora E., et al. Alcohol consumption and atherosclerosis: What is the relation. Prospective results from the Bruneck study. Stroke 1998; 29: 900–7
  • Femia R., Natali A., L'Abbate A., Ferrannini E. Coronary atherosclerosis and alcohol consumption. Angiographic and mortality data. Arterioscler Thromb Vasc Biol 2006; 26: 1607–12
  • Frick M. H., Elo O., Haapa K., Heinonen O. P., Heinsalmi P., Helo P., et al. Helsinki Heart Study: primary prevention trial with gemfibrozil in middle‐aged men with dyslipidemia. New Engl J Med 1987; 317: 1237–45
  • Tenkanen L., Mänttäri M., Kovanen P. T., Virkkunen H., Manninen V. Gemfibrozil in the treatment of dyslipidemia. An 18‐year follow‐up of the Helsinki Heart Study. Arch Intern Med 2006; 166: 743–8
  • Keech A., Simes R. J., Barter J., Best J., Scott R., Taskinen M. R., et al. Effects of long‐term fenofibrate therapy on cardiovascular events in 9759 people with type 2 diabetes mellitus (the FIELD Study): randomised controlled trial. Lancet 2005; 366: 1849–61
  • Randomised trial of cholesterol lowering in 4444 patients with coronary heart disease: the Scandinavian Simvastatin Survival Study (4S). Lancet 1994; 344: 1383–9
  • Heart Protection Study Collaborative Group. MRC/BHF Heart Protection Study of cholesterol lowering with simvastatin in 20,536 high‐risk individuals: a randomised placebo‐controlled trial. Lancet 2002; 360: 7–22
  • Cheung B. M. Y., Lauder I. J., Lau C. ‐P., Kumana C. Meta‐analysis of large randomized controlled trials to evaluate the impact of statins on cardiovascular outcomes. Br J Clin Pharmacol 2004; 57: 640–51
  • Baigent C., Keech A., Kearney P. M., Blackwell L., Buck G., Pollicino C., , Cholesterol Treatment Trialists' (CTT) Collaborators, et al. Efficacy and safety of cholesterol‐lowering treatment: prospective meta‐analysis of data from 90 056 participants in 14 randomised trials of statins. Lancet 2005; 366: 1267–78
  • The Lipid Research Clinics Coronary Primary Prevention Trial: results. I. Reduction in the incidence of coronary heart disease. JAMA 1984; 251: 351–64
  • Dormandy J. A., Charbonnel B., Eckland D. A. J., Erdmann E., Massi‐Benedetti M., Moules I. K., et al. Secondary prevention of macrovascular events in patients with type 2 diabetes in the Proactive Study (PROspective pioglitAzone Clinical Trial In macroVascular Events): a randomised controlled trial. Lancet 2005; 366: 1279–89
  • Poikolainen K. Alcohol and mortality: a review. J Clin Epidemiol 1995; 48: 455–65
  • Luoma P. V., Savolainen MJ., Sotaniemi E. A., Arranto A. J., Pelkonen R. O. Plasma HDL cholesterol and blood glucose in non‐insulin‐dependent diabetics related to liver lipids and microsomal enzyme activity. Acta Med Scand 1985; 217: 473–9
  • Luoma P. V., Rautio A., Stengård J., Sotaniemi E. A., Marniemi J. High‐density lipoprotein subfractions, apolipoproteins and antipyrine clearance in normal subjects. Eur J Clin Pharmacol 1990; 38: 625–7
  • Tokola O., Pelkonen O., Karki N. T., Luoma P. Hepatic drug‐oxidizing enzyme systems and urinary D‐glucaric acid excretion in patients with congestive heart failure. Br J Clin Pharmacol 1975; 2: 429–36
  • Luoma P. V., Myllylä V. V., Hokkanen E. Relationship between plasma high‐density lipoprotein cholesterol and anticonvulsant levels in epileptics. J Cardiovasc Pharmacol 1982; 4: 1024–7
  • LaPorte R., Valvo‐Gerard L., Kuller L., Wanju R., Bates M., Cresanta J., et al. The relationship between alcohol consumption, liver enzymes and high‐density lipoprotein cholesterol. Circulation 1981; 64(suppl III)67–72
  • Steyn K., Fourie J., Benade A. J., Roussow J. E., Langenhoven M. L., Joubert G., et al. Factors associated with high density lipoprotein cholesterol in a population with high high density lipoprotein cholesterol levels. Arteriosclerosis 1989; 9: 390–7
  • Dixon R. M., Borden E. C., Keim N. L., Anderson S., Spennetta T. L., Tormey D. C., et al. Decreases in serum high‐density‐lipoprotein cholesterol and total cholesterol resulting from naturally produced recombinant DNA‐derived leucocyte interferon. Metabolism 1984; 33: 400–4
  • Andrade R. J., Escolar J. L., Valdivielso P., Gonzelez‐Santos P. Apolipoprotein distribution in plasma HDL subfractions in alcohol consumers. Drug Alcohol Depend 1990; 26: 161–8
  • Robertson R. G., Krause B. R., Welty D. F., Wolfgang G. H. I., Grazian M. J., Pilcher G. D., et al. Hepatic microsomal induction profile of carbamic acid [[2,6‐bis(1‐methylethyl) phenoxy] sulfonyl]‐2,6‐bis(1‐methylethyl) phenyl ester, monosodium salt (PD138142‐15), a novel lipid regulating agent. Biochem Pharmacol 1995; 49: 799–808
  • Evans R. M. The nuclear receptor superfamily: a rosette stone for physiology. Mol Endocrinol 2005; 19: 1429–38
  • Luoma P. V., Sotaniemi E. A., Ehnholm C. Low high‐density lipoprotein and reduced antipyrine metabolism in members of a family with polycystic liver disease. Scand J Gastroenterol 1980; 15: 869–73
  • Luoma P. V., Sotaniemi E. A., Pelkonen R. O., Pirttiaho H. I. Serum low density and high density lipoprotein cholesterol, and liver size in subjects on drugs inducing hepatic microsomal enzymes. Eur J Clin Pharmacol 1985; 28: 615–8
  • Bretillon L., Lütjohann D., Ståhle L., Widhe T., Bindl L., Eggertsen G., et al. Plasma levels of 24S‐hydroxycholesterol reflect the balance between cerebral production and hepatic metabolism and are inversely related to body surface. J Lipid Res 2000; 41: 840–5
  • Lehrke M., Lebherz C., Millington S. C., Guan H. P., Millar J., Rader D. J., et al. Diet‐dependent cardiovascular lipid metabolism controlled by hepatic LXRα. Cell Metab 2005; 1: 297–308
  • Joseph S. B., Mckilligan E., Pei L., Watson M. A., Collins A. R., Laffitte B. A., et al. Synthetic LXR ligand inhibits the development of atherosclerosis in mice. Proc Natl Acad Sci U S A 2002; 99: 7604–9
  • Terasaka N., Hiroshima A., Koieyama T., Ubukata N., Morikawa Y., Nakai D., et al. T‐0901317, a synthetic liver X receptor ligand, inhibits development of atherosclerosis in LDL receptor‐deficient mice. FEBS Lett 2003; 536: 6–11
  • Kovanen P. T., Pentikäinen M. O. Pharmacological evidence for a role of liver X receptors in atheroprotection. FEBS Lett 2003; 536: 3–5
  • Levin N., Bischoff E. D., Daige C. L., Thomas D., Vu C. T., Heyman R. A., et al. Macrophage liver X receptor is required for anti‐atherogenic activity of LXR agonists. Arterioscler Thromb Vasc Biol 2005; 25: 135–42
  • Naik S. U., Wang X., Da Silva J. S., Jaye M., Macphee C. H., Reilly M. R., et al. Pharmacological activation of liver X receptors promotes reverse cholesterol transport in vivo. Circulation 2006; 113: 90–7

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.