Next Article in Journal
Expression of Fibrosis-Related Genes in Liver and Kidney Fibrosis in Comparison to Inflammatory Bowel Diseases
Next Article in Special Issue
Pregnane X Receptor Mediates Atherosclerosis Induced by Dicyclohexyl Phthalate in LDL Receptor-Deficient Mice
Previous Article in Journal
A Leucine Zipper Dimerization Strategy to Generate Soluble T Cell Receptors Using the Escherichia coli Expression System
Previous Article in Special Issue
Phosphorylation-Induced Ubiquitination and Degradation of PXR through CDK2-TRIM21 Axis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Nuclear Receptor PXR in Drug-Induced Hypercholesterolemia

1
Research Unit of Biomedicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014 Oulu, Finland
2
Research Unit of Internal Medicine, Biocenter Oulu, Medical Research Center Oulu, University of Oulu and Oulu University Hospital, P.O. Box 5000, FI-90014 Oulu, Finland
*
Author to whom correspondence should be addressed.
Cells 2022, 11(3), 313; https://doi.org/10.3390/cells11030313
Submission received: 13 December 2021 / Revised: 13 January 2022 / Accepted: 14 January 2022 / Published: 18 January 2022

Abstract

:
Atherosclerosis is a major global health concern. The central modifiable risk factors and causative agents of the disease are high total and low-density lipoprotein (LDL) cholesterol. To reduce morbidity and mortality, a thorough understanding of the factors that influence an individual’s cholesterol status during the decades when the arteria-narrowing arteriosclerotic plaques are forming is critical. Several drugs are known to increase cholesterol levels; however, the mechanisms are poorly understood. Activation of pregnane X receptor (PXR), the major regulator of drug metabolism and molecular mediator of clinically significant drug–drug interactions, has been shown to induce hypercholesterolemia. As a major sensor of the chemical environment, PXR may in part mediate hypercholesterolemic effects of drug treatment. This review compiles the current knowledge of PXR in cholesterol homeostasis and discusses the role of PXR in drug-induced hypercholesterolemia.

1. Introduction

Hypercholesterolemia, e.g., elevated LDL cholesterol (LDL-C) and imbalance of LDL-C and high-density lipoprotein cholesterol (HDL-C), is a central causative risk factor of atherosclerosis [1,2]. In fact, the retention of LDL-C and other cholesterol-rich apolipoprotein (Apo) B-containing lipoproteins within the arterial wall represents the key initiating event in atherogenesis [3]. Complications of atherosclerosis, ischemic heart disease and stroke, are among the leading causes of death [4]. In addition to atherosclerosis, excess cholesterol may also be involved in the pathogenesis of other diseases, such as non-alcoholic fatty liver disease and diabetes [5].
Multiple genetic and modifiable factors induce hypercholesterolemia. The most common LDL-C-elevating lifestyle factor is high saturated fat intake, but certain other nutritional factors and obesity also promote hypercholesterolemia [2,3]. Hypercholesterolemia can also be caused by secondary causes, like hypothyroidism [2]. Furthermore, multiple drugs may increase cholesterol. Some drug classes are well-known for their adverse effect on cholesterol, such as antipsychotics and immunosuppressants, but individual drugs from many different classes of drugs have been reported to affect cholesterol levels [2,6]. The mechanisms by which drugs increase cholesterol concentration are still largely unknown.
Nuclear receptor pregnane X receptor (PXR; NR1I2) is a ligand-activated transcription factor that regulates many phase I and II drug-metabolizing enzymes and drug transporters; induction of cytochrome P450 (CYP) 3A4 enzyme is a prime example [7]. Indeed, PXR is an important mediator of the induction type drug–drug interactions [8]. Unlike most other nuclear receptors, PXR accepts a wide array of structurally diverse chemicals as ligands. Many modern synthetic chemicals among environmental contaminants, industrial chemicals, and drugs are well-characterized ligands for PXR. In addition to the classical role in drug metabolism, PXR has been shown to play a role in other biological processes, such as inflammation, cellular proliferation, and glucose and lipid metabolism [9,10]. Due to its pleiotropic functions, PXR is actively studied as a contributing factor in disease pathophysiology and as a novel therapeutic target. Interestingly, PXR activation seems to adversely affect multiple metabolic functions, including lipid metabolism, glucose tolerance, and blood pressure [11]. Thus, it has been hypothesized that PXR activation may partly explain the adverse metabolic effects of environmental chemical exposure and drugs [12,13].
Exposure to environmental chemicals usually consists of a complex mixture of various low-dose exposures with variable amounts and exposure times, making the association of exposure and potential metabolic consequences challenging. In contrast, exposure to drugs is usually well-controlled and both the exposure time and the dose are known. For this reason, the effects of individual drugs on metabolic health are much better known than those of environmental chemicals, and this information could serve as a tool to understand the molecular mechanisms involved and subsequently help to predict the effects of other types of chemicals. This review focuses on the role of PXR in drug-induced hypercholesterolemia. We discuss the evidence for the hypercholesterolemic effect of PXR activation in humans and the mechanistic aspects characterized in mouse models. Finally, we survey the current knowledge on the cholesterol-elevating effect of clinically used drugs and discuss a putative role of PXR in these effects.

2. Evidence for Induction of Hypercholesterolemia by PXR Activation in Humans

It has long been known that PXR-activating and CYP enzyme-inducing antiepileptics (carbamazepine, phenytoin, and phenobarbital) [8] elevate cholesterol levels in patients with epilepsy [14,15,16,17]. For example, in one of the earliest prospective studies, phenytoin treatment elevated serum total cholesterol (TC) by 14% at 1-month and 11% at 3-month timepoints in 20 patients with epilepsy [16]. In a subset of 14 patients, 2-year treatment elevated TC by 19%. Similarly, in a prospective study in 36 patients with a more comprehensive lipid panel, 12-month carbamazepine treatment increased mean serum TC by 13%, LDL-C 13%, and HDL-C 14% [14]. In a subset of 19 patients, 5-year prospective measurements also demonstrated elevated levels for TC (by 12%) and HDL-C (26%); the effect on LDL-C was not significant (6%). Most of the cross-sectional studies display cholesterol-elevating effects of enzyme-inducing antiepileptics, while valproic acid, an antiepileptic without enzyme-inducing properties, does not elevate cholesterol [15,17,18,19]. Interestingly, phenobarbital is known to induce human hepatic 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR) protein, the target of statins, in vivo [20]. However, as enzyme-inducing antiepileptics also activate constitutive androstane receptor (CAR) [8], which is possibly involved in cholesterol metabolism regulation [21], the elevation of cholesterol cannot be solely attributed to PXR. The role of CAR activation alone in cholesterol elevation is difficult to study as there are no selective CAR agonists suitable for human volunteer and patient studies.
Among the drugs that activate PXR, the tuberculosis antibiotic rifampicin is a well-established selective agonist of the receptor [22]. Earlier rifampicin studies with a small number of healthy volunteers (21 days with n = 8, 6 days with n = 10, and 14 days with n = 7, respectively) did not observe an increase in TC [23,24,25], but serum lathosterol-to-cholesterol ratios, a marker of cholesterol synthesis [26], were increased during rifampicin treatment [24]. Furthermore, a trial (n = 12) with cholesterol measurement daily for 30 days reported that a 14-day rifampicin dosing elevated TC by 10% (not statistically significant) with gradual decrease to baseline after discontinuing the dosing [27].
We recently published combined results from two placebo-controlled cross-over trials [28,29] investigating the effect of 600 mg rifampicin for a week on plasma metabolomics, including all lipoprotein fractions and their lipid contents [30]. The study was the largest to date evaluating the effects of PXR activation on plasma metabolomics, with 34 healthy volunteers participating. Rifampicin significantly increased serum TC (by 7%) and LDL-C (12%), with all sizes of LDL particles (large, medium, small) elevated to a similar degree. Esterified (7%) and free cholesterol (7%), as well as intermediate-density lipoprotein (10%), were also elevated by rifampicin dosing. Changes in serum 4β-hydroxycholesterol, a marker of CYP3A4 activity [31] known to be elevated by PXR activation [11], correlated with these changes. The elevations of HDL-C (4%) and total ApoB (5%) by rifampicin were non-significant after correction for multiple testing, while the largest very low-density lipoprotein (VLDL) fractions tended to decrease [30]. Serum concentration of ApoB48, the intestinal form of ApoB, was decreased 14% by rifampicin dosing. During glucose challenge (2 h oral glucose tolerance test), PXR activation by rifampicin resulted in elevated concentrations of very small VLDL particles, remnant cholesterol, and total ApoB [30]. As in a previous study [24], rifampicin elevated serum lathosterol-to-cholesterol ratio [30], and, on the other hand, decreased serum concentrations of cholesterol synthesis precursors citrate and acetate, both suggesting increased cholesterol synthesis.
Another interesting PXR agonist with a strong hypercholesterolemic effect and some human mechanistic evidence on the induction of cholesterol synthesis is mitotane, a steroidogenesis inhibitor used in Cushing’s syndrome and a cytostatic treatment for adrenocortical carcinoma. Although its PXR-activating properties and high risk of drug–drug interactions were revealed only recently [32,33], mitotane has long been recognized as having a remarkable cholesterol-elevating effect. An early study on patients with Cushing’s syndrome demonstrated a 59% increase in serum TC and an 81% increase in LDL-C, with an accompanying 61% increase in ApoB concentration [34]. No significant effects on HDL-C were detected in this small study (n = between 6 and 21). In a recent study in patients with adrenocortical carcinoma (n = 39), mitotane treatment led to 43% increase in TC and 69% increase in LDL-C after 6-month dosing [35]. Additionally, HDL-C was elevated by 14%. Already in 1961, mitotane was suspected to induce cholesterol synthesis as indicated by an increased rate of incorporation of l4C-acetate into plasma cholesterol of patients [36]. Mitotane also elevates plasma levels of mevalonate, a marker of cholesterol synthesis [34].
These findings indicate that (1) drugs with PXR agonism elevate TC and LDL-C and that (2) increased cholesterol synthesis may play a role in drug-induced and PXR-mediated hypercholesterolemia. Genetic studies provide additional evidence, as PXR gene polymorphisms associate with plasma LDL-C levels [37].

3. Mechanisms of PXR-Induced Hypercholesterolemia

PXR is primarily expressed in the liver and intestine, which are the central tissues for drug metabolism but also those for cholesterol homeostasis. Several intestinal and hepatic mechanisms have been identified that may confer the hypercholesterolemic effect of PXR activation. However, the mechanistic evidence has been mainly gathered utilizing murine models, whose translational value is often limited by substantial differences in murine and human lipoprotein homeostasis. Mice lack cholesterol ester transfer protein (CETP), which leads to high HDL-C and very low LDL-C levels. Nevertheless, the fundamental mechanisms controlling cholesterol synthesis and several other steps in cholesterol homeostasis in humans and mice are similar, and the current evidence indicates important similarities in the mechanisms controlling PXR-mediated elevation of cholesterol in humans and mice. It should be kept in mind that the PXR ligand preference is species-specific and therefore the compound-specific results cannot be directly transferred from mouse or rat experiments to humans.

3.1. PXR in Cholesterol Synthesis

In mammals, almost all cells can synthesize cholesterol, but plasma cholesterol is only affected by cholesterol synthesis in the liver due to its central position in lipoprotein metabolism [38,39]. In the liver, as well as in other organs, the regulation of cholesterol synthesis is predominantly determined by the activity of the transcription factor sterol-regulatory element-binding protein 2 (SREBP2) (Figure 1) [40].
Inactive SREBP2 resides in the endoplasmic reticulum (ER) in a complex with SREPB-cleavage activating protein (SCAP) and insulin-induced gene 1 (INSIG1), in which the former functions as a sterol sensor and the latter as a negative regulator of SREBP2 activation [40]. Sterol depletion in the ER incites conformational changes to SCAP, which are required for the translocation of the SREBP2–SCAP complex to the Golgi apparatus. There, SREBP2 is proteolytically cleaved to yield active SREBP2 monomers, which homodimerize, translocate to the nucleus and induce gene expression (Figure 1). In the nucleus, SREBP2 induces genes for cholesterol synthesis, including the rate-controlling enzyme HMGCR. Repletion of sterols in the ER represses SCAP, thus forming a feedback loop to control cholesterol synthesis. In addition to fluctuating ER sterol levels, SREBP2 activity is regulated by several modulators of cellular energy metabolism [41]. For instance, factors affecting INSIG1 expression may affect SREBP2 activation, and SREBP2 is regulated by several post-translational modifications conveyed by hormonal signals and signaling cascades, such as insulin and mammalian target of rapamycin (mTOR). Thus, the need to adjust cholesterol synthesis rates may rise from multiple sources, and cholesterol synthesis presents a highly adaptable biological phenomenon.
Our recently published results indicate that rifampicin treatment increases human plasma cholesterol by activating PXR and hepatic cholesterol synthesis [30]. The same cholesterol synthesis-inducing effect could be repeated also in high-fat diet-fed mice, which were treated with selective murine PXR agonist, pregnenolone-16α-carbonitrile (PCN). This enabled us to study the molecular mechanisms in more detail. In the liver, PXR activation led to nuclear accumulation of active SREBP2 protein, widespread induction of its target genes, and increases in cholesterol and markers of cholesterol synthesis [30].
INSIG1 inhibits SREBP2 activation by retaining the SREBP2–SCAP complex in the ER [42]. PXR has been shown to transcriptionally induce Insig1 mRNA [43]. With this in mind, the stimulation of SREBP2 activity by PXR was an unexpected finding [30]. We also detected increased Insig1 mRNA expression in response to PXR activation but were unable to detect increased INSIG1 protein expression [30]. In their study, Roth and colleagues [43] did not report the INSIG1 protein level; furthermore, the study did not evaluate the effect of INSIG1 on the activity of SREBP2 but SREBP1, an SREBP isoform mainly controlling triglyceride synthesis [40]. SREBP1 inhibition by PXR activation is in line with studies describing how PXR induces hepatic steatosis independent of SREBP1 [44,45]. Altogether, these findings suggest that PXR may affect INSIG1 translation or protein degradation and selectively affect SREBP1 and SREBP2 activities. In addition to INSIG1, PXR has been shown to suppress fibroblast growth factor 21 (FGF21), a negative regulator of SREBP2 expression [46,47], which may play an additional role in PXR-stimulated SREBP2 activity, although Srebp2 mRNA expression remained unaffected in our experiments.
Cholesterol is synthesized from acetyl-CoA, which undergoes multiple enzymatic reactions to form squalene, subsequently converted to (S)-2,3-epoxysqualene by squalene epoxidase (SQLE) and further to lanosterol, which is converted to cholesterol either in the Bloch or in the Kandutsch–Russell pathway [48,49]. Recently, an antiretroviral efavirenz was shown to induce cholesterol synthesis in mice by activating PXR and inducing hepatic Sqle, a novel PXR target gene [50]. Importantly, the effect of efavirenz on cholesterol was abolished in mice lacking hepatic PXR. In our study, PCN treatment induced hepatic Sqle among other genes of cholesterol synthesis [30]. Furthermore, all PCN-treated mice had lower liver squalene levels than controls, which possibly indicated faster squalene metabolism and increased SQLE activity. Interestingly, besides HMGCR, SQLE is another rate-limiting enzyme of cholesterol synthesis and cholesterol constitutes its inhibitory feedback signal, in addition to downregulation of proteolytic processing of SREBP2, also by directly inhibiting SQLE activity [51,52]. Besides the direct PXR mediated regulation [50], distortion of normal inhibitory regulation of SREBP2 by PXR activation may in part account for the induction of Sqle gene expression [30].
We showed that increased cholesterol synthesis due to PXR activation was caused by the induction of the Kandutsch–Russell pathway, as evidenced by increased plasma and hepatic markers of the pathway, lathosterol and zymostenol, and induced DHCR24, an enzyme that directs cholesterol synthesis to the Kandutsch–Russell pathway [30]. PXR did not affect desmosterol, a marker of the Bloch pathway and a negative regulator of SREBP2 [53], meaning that by inducing the Kandutsch–Russell pathway instead of the Bloch pathway PXR may evade normal SREBP2 downregulation by desmosterol (Figure 1). Overall, these results are in line with the previous reports regarding PXR and cholesterol synthesis and, most importantly, with the human findings, and suggest that PXR activation stimulates hepatic SREBP2 activity with harmful effects on circulating atherogenic lipids.

3.2. PCSK9 Induction by PXR Activation

Identification of proprotein convertase subtilisin kexin-type 9 (PCSK9) and development of PCSK9 inhibitors for drug therapy has remarkably improved the understanding of plasma cholesterol regulation [54,55]. PCSK9 is secreted from the liver to the circulation, where it functions to induce the degradation of hepatic LDL receptors. This results in decreased LDL clearance and increased circulating LDL. Inhibition of plasma PCSK9 by antibodies has proven to be an efficient therapeutic strategy to lower plasma LDL [56].
In the liver, the expression of Pcsk9 is regulated by SREBP2 [57]. As PXR activation was shown to stimulate SREBP2 activity, this raised the hypothesis that PXR could also induce Pcsk9 expression. Indeed, this was found to be the case in both mice and humans [30]. LDL receptor is another SREBP2 target gene and could counteract the negative effect of PCSK9. Expectedly, the LDL receptor mRNA was also induced by PXR activation in mouse liver; however, the response was very minor compared with the Pcsk9 response [30].
Statins stimulate SREBP2 activity and Pcsk9 expression as a consequence of cholesterol synthesis inhibition and lower sterol content in the hepatocyte ER. Interestingly, lipophilic statins (atorvastatin, simvastatin) have been shown to increase PCSK9 more efficiently than hydrophilic statins (rosuvastatin, pravastatin) [58]. Atorvastatin and simvastatin are also PXR ligands, and it could be speculated that, in addition to the decreased cellular sterol content, the PXR agonism plays an additional role in the PCSK9 increase by these statins.
The current list of drugs that induce PCSK9 include statins, fibrates, mTOR inhibitors, nilotinib, and rifampicin [30,59,60,61,62]. The mechanisms are probably diverse but given the recently discovered role of PXR in Pcsk9 regulation, further research on the effects of PXR agonists on Pcsk9 expression is warranted. Of the currently known PCSK9-inducing drugs, several (atorvastatin, simvastatin, rifampicin, and nilotinib) are PXR ligands, although they involve also other cellular mechanisms.

3.3. PXR in Intestinal Cholesterol Absorption

Intestinal absorption of biliary and dietary cholesterol is an important contributor to hypercholesterolemia, and ezetimibe or food additives (plant stanols or sterols) that limit intestinal cholesterol absorption decrease circulating cholesterol [63,64]. Interestingly, Niemann-Pick C1-like 1 (NPC1L1), an intestinal cholesterol transporter protein and the molecular target of ezetimibe, has been shown to be directly regulated by PXR [65]. Tributyl citrate, a common plasticizer, and quetiapine, an atypical antipsychotic with known adverse effects on lipid metabolism, have been shown to induce hypercholesterolemia in mice by activating intestinal PXR, which is associated with increased Npc1l1 expression [50,65]. It is noteworthy that in mice NPC1L1 is expressed only in the intestine, whereas in humans NPC1L1 is also strongly expressed in the liver, which may affect the translational value of these findings [66]. Rifaximin is a gut-specific human PXR activator, which does not increase serum cholesterol in PXR-humanized mice, although it induces intestinal triglyceride absorption [67,68]. However, rifaximin induced hepatic cytochrome P450 7A1 (CYP7A1) enzyme, the gatekeeper of bile acid synthesis, which suggests that increased bile acid synthesis may have protected the mice from hypercholesterolemia.

3.4. PXR in Bile Acid Synthesis and Cholesterol Metabolism

Synthesis of bile acids from cholesterol and biliary secretion of bile acids and cholesterol present a major route of excess cholesterol disposal. Increasing bile acid disposal by inhibiting bile acid absorption from the intestine to the circulation by bile acid sequestrants, such as cholestyramine and colesevelam, lowers plasma cholesterol, highlighting the importance of hepatic cholesterol disposal as a regulatory mechanism of circulating cholesterol.
PXR activation has been shown to have a complex role in endogenous bile acid homeostasis. Activation of PXR by some bile acids is critical to accelerate the detoxification of otherwise hepatotoxic bile acids [69,70,71]. Bile acids are detoxified mainly by known PXR target genes; CYP3A enzymes, bile acid conjugation enzymes SULT2A1 and UGTs, and bile acid transporter MRP2 [72,73].
Several studies have reported that PXR activation represses CYP7A1, the rate-limiting enzyme of bile acid synthesis, thus forming a negative feedback loop to regulate bile acid homeostasis [72,74,75,76]. Although PXR activation represses CYP7A1 in several animal and cell models, rifampicin dosing does not seem to affect CYP7A1 expression in humans [77] and even increases bile acid synthesis, as evidenced by increased serum levels of 7α-hydroxy-4-cholestene-3-one, a marker of bile acid synthesis [24,77].

4. PXR in HDL Homeostasis

HDL is a central factor in reverse cholesterol transport, a mechanism by which tissues and cells get rid of excess cholesterol; HDL functions to transport cholesterol from extrahepatic tissues to the liver and intestine [78]. Although PXR activation appears to have mainly harmful effect on cholesterol homeostasis, there is some evidence that PXR activation could increase HDL-C. For instance, some drugs with PXR-activating properties, such as carbamazepine and phenytoin, appear to increase HDL-C [14,79,80,81]. Furthermore, we have reported that rifampicin induces CYP3A4-mediated formation of 4β-hydroxycholesterol (4βHC) and, as a liver X receptor (LXR) agonist, 4βHC in turn stimulates cholesterol efflux transporters in macrophages, possibly promoting HDL-C-mediated reverse cholesterol transport [82]. Thus, elevation of circulating 4βHC could partly explain the HDL-C increasing effect of PXR-activating drugs (PXR–4βHC–LXR circuit). For more information on this topic, please see our recent review [11].

5. Drug-Induced Hypercholesterolemia

Increase in circulating cholesterol during drug treatment is not uncommon, as several antihypertensives, antihyperglycemics, antipsychotics, antiretrovirals, and immunosuppressants may have an unfavorable effect on cholesterol. Many of these drugs are used long-term, increasing the potential for cardiovascular risk. Drug-induced hypercholesterolemia may be directly caused by the drug or secondarily caused by other effects of the drug, such as weight gain. For example, some antipsychotics, antidepressants, anticonvulsants, and hormones may increase weight and negatively affect cholesterol status [83].
To obtain a systematic overview on drug-induced hypercholesterolemia, we identified clinically used drugs that increase TC or LDL-C in the commercially available Drug Laboratory Effects database (Multirec Ltd., Turku, Finland) (Table 1). To increase the clinical significance of the table, we removed drugs that have been withdrawn from the market or were reported to have both lowering and increasing effects on cholesterol. Furthermore, we added bexarotene, mitotane, and rifampicin to the table, as they have reported effects on cholesterol but were not included in the original database [22,30,35].
Table 1 consists of 106 drugs, of which 48 drugs increase both LDL-C and TC, 54 drugs increase LDL-C, and 100 drugs increase TC. Among the drugs that increase LDL-C, the largest drug classes are immunosuppressants with 15 drugs (28%), antineoplastics with 10 drugs (19%), antipsychotics with eight drugs (15%), and antiretrovirals with six drugs (11%). Together these four classes comprise 72% of the drugs listed.
A similar pattern is seen in the 100 drugs that increase TC (Table 1): 62% of the drugs that increase cholesterol are either antineoplastic (22 drugs; 22%), immunosuppressants (19 drugs; 19%), antiretrovirals (12 drugs; 12%), or antipsychotics (9 drugs; 9%). In addition, the list of drugs that increase TC include several commonly used glucocorticoids, serotonin reuptake inhibitors, and non-steroidal anti-inflammatory drugs. Naturally, the lists of drugs increasing TC and LDL-C are heavily overlapping. It should be noted that sometimes the original publications used as the source for the database did not report all the relevant cholesterol values. Furthermore, in many cases there exist conflicting data on the effect of drugs on plasma cholesterol levels. Finally, among the drugs that have been reported to increase TC and/or LDL-C, the magnitude of effect and the clinical significance are very variable.
Some drug classes are clearly overrepresented in the list of cholesterol-elevating drugs and their effects may seem like a class effect of certain drugs. However, there are still significant differences between the drugs within the therapeutic groups. For instance, a recent meta-analysis described marked differences between atypical antipsychotics in the metabolic side effects, including TC and LDL-C, with olanzapine and clozapine displaying the worst profiles [130]. Furthermore, changing from quetiapine, risperidone, or olanzapine to aripiprazole has been reported to be beneficial for cholesterol status and 10-year cardiovascular disease risk [131]. Notably, these antipsychotics have also significant differences in their target receptor profiles that could potentially play a role in their varying effects on metabolic parameters.
The mechanistic understanding of the molecular mediators of drug-induced dyslipidemia is elusive and the data to date are limited by the lack of sophisticated human studies. The hypercholesterolemic effects of several antipsychotics have been linked to increased activation of SREBP2 due to inhibition of INSIG-2 in murine models [132]. Furthermore, INSIG-2 gene polymorphisms in schizophrenia patients are associated with weight gain and prevalence of metabolic syndrome [133].
Inflammation and drugs that modulate inflammation, non-steroidal anti-inflammatory drugs (NSAIDS), glucocorticoids, and immunosuppressants, are known regulators of cholesterol levels. Acute infections tend to decrease LDL cholesterol, and the more severe the infection, the more pronounced the effects on cholesterol [134,135]. The same seems to apply to chronic inflammatory diseases, as a decrease in HDL and LDL cholesterol is a common finding in cases of rheumatoid arthritis and the reductions in HDL and LDL correlate with disease severity in ankylosing spondylitis [136,137]. As inflammation attenuates during anti-inflammatory drug treatment, cholesterol levels return to normal, which may explain why many anti-inflammatory drugs seem to increase cholesterol [138,139]. However, some drugs used to treat inflammatory diseases may have more specific effects on lipid metabolism.
Interestingly, immunosuppressants used in organ transplant patients, cyclosporin (calcineurin inhibitor) and especially the mTOR inhibitors sirolimus, tacrolimus, and everolimus, increase cholesterol [140,141]. Immunosuppressant-mediated dyslipidemia is common and occurs in 60% of organ transplant patients [142]. The mechanisms by which immunosuppressants induce dyslipidemia are not well understood, but they seem to involve LDL metabolism and PCSK9 [143,144]. mTOR is a central regulator of lipid metabolism and SREBP, which already hints at mechanisms of mTOR inhibitor-induced dyslipidemia [145].
Antiretroviral therapies have improved the life expectancy of human immunodeficiency virus (HIV)-infected patients. This success has brought up HIV-associated risks for other comorbidities, including increased risk for cardiovascular diseases [146,147]. One significant contributing factor is the adverse effect of antiretrovirals on lipid metabolism [146]. Protease inhibitors, non-nucleoside reverse transcriptase inhibitors, and integrase inhibitors may all increase TC or LDL-C [148]. Most of the approved protease inhibitors and about a quarter of the approved reverse transcriptase inhibitors are included in the list of drugs that increase cholesterol (Table 1). Mechanistically, protease inhibitors have been shown to stimulate SREBP activity in the liver [148].

6. Identification of PXR Ligands among the Drugs Inducing Hypercholesterolemia

To gain a systematic overview of the potential role of PXR in drug-induced hypercholesterolemia, we identified in the literature the PXR-activating drugs among the drugs that increase TC or LDL-C (Table 1).
Only studies that provided direct evidence of PXR binding were considered. For instance, induction of classical PXR target genes, CYP3A and CYP2C, was not considered a proof of PXR agonism, as the expression of these genes is also regulated by other nuclear receptors. PXR activation is often evaluated based on CYP induction and consequent drug–drug interactions. However, this may not always reveal actual PXR activation status, as some inducers are also CYP inhibitors [149]. Indeed, several drugs that are PXR agonists also inhibit CYP3A4 activity. In these cases, the other effects of PXR activation could still be significant. On the other hand, the results of reporter gene assays or other in vitro methods may overestimate the receptor activation, especially if performed only with high ligand concentrations.
Among the 106 drugs that increase either LDL-C or TC, 23 (21.7%) were identified as PXR activators, and three, brigatinib, apalutamide, and ruxolitinib, as likely activators. Thus, altogether, of the 106 drugs that increase cholesterol levels, 26 (24.5%) are most likely to have PXR-activating property. Among the drugs that increase LDL cholesterol but have not been identified as PXR agonists, clozapine, and nevirapine have been shown to have some inducing effect on CYP3A4, but only nevirapine is a clinically significant CYP3A4 inducer. Among the drugs that increase TC, 10 drugs have not been identified as PXR agonists but induce CYP3A4.
PXR and its sister receptor constitutive androstane receptor (CAR) both regulate drug metabolism but also glucose and lipid metabolism and also share some ligands. However, while PXR activation is detrimental to metabolic performance, CAR activation has been shown to be beneficial. Of importance, CAR activation has been shown to lower cholesterol by increasing fecal cholesterol disposal and by suppressing cholesterol synthesis [21,150].

7. Conclusions

A large number of drugs have been reported to display adverse effects on plasma cholesterol level. For many, the effect size is rather small, but there is still a significant number of drugs for which the effect is considered clinically significant. In long-term treatment, the unfavorable effect of drug therapy on cholesterol may predispose to atherosclerosis, especially if combined with other risk factors.
Recent studies have indicated that activation of nuclear receptor PXR increases plasma cholesterol and several potential mechanisms have been reported, including increased hepatic cholesterol synthesis, induction of PCSK9, increased intestinal absorption, and decrease of bile acid synthesis. In humans, there is currently evidence for the first two of these mechanisms. So far, rifampicin, efavirenz, and quetiapine have been reported to induce hypercholesterolemia through PXR-mediated mechanisms, either in humans or mice [30,50,96]. Among the known cholesterol-increasing drugs, about a quarter are certain or possible PXR ligands. Thus, PXR activation is likely to at least partly mediate their hypercholesterolemic effects but verifying the role of PXR requires further studies in each case in the future.

Author Contributions

Conceptualization, M.K., J.H. (Janne Hukkanen) and J.H. (Jukka Hakkola); writing—original draft preparation, M.K.; writing—review and editing, J.H. (Janne Hukkanen) and J.H. (Jukka Hakkola). All authors have read and agreed to the published version of the manuscript.

Funding

The original research of the authors has been financially supported by the grants from the Academy of Finland (grants 286743 and 323706), the Novo Nordisk Foundation (grants NNF14OC0010653 and NNF15OC0015846), the Finnish Medical Foundation, the Finnish Foundation for Cardiovascular Research, the Northern Finland Health Care Support Foundation, and the Diabetes Research Foundation. The project EDCMET has received funding from the European Union’s Horizon 2020 research and innovation programme under grant agreement no. 825762.

Data Availability Statement

The data presented in this study are available within this article.

Acknowledgments

The authors acknowledge Multirec Ltd., Turku, Finland for the permission to use the Drug Laboratory Effects database information in this review article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Libby, P.; Buring, J.E.; Badimon, L.; Hansson, G.K.; Deanfield, J.; Bittencourt, M.S.; Tokgözoğlu, L.; Lewis, E.F. Atherosclerosis. Nat. Rev. Dis. Prim. 2019, 5, 56. [Google Scholar] [CrossRef] [PubMed]
  2. Karr, S. Epidemiology and management of hyperlipidemia. Am. J. Manag. Care 2017, 23, S139–S148. [Google Scholar] [PubMed]
  3. Mach, F.; Baigent, C.; Catapano, A.L.; Koskinas, K.C.; Casula, M.; Badimon, L.; Chapman, M.J.; De Backer, G.G.; Delgado, V.; Ference, B.A.; et al. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk. Eur. Heart J. 2020, 41, 111–188. [Google Scholar] [CrossRef] [PubMed]
  4. Abbafati, C.; Abbas, K.M.; Abbasi-Kangevari, M.; Abd-Allah, F.; Abdelalim, A.; Abdollahi, M.; Abdollahpour, I.; Abegaz, K.H.; Abolhassani, H.; Aboyans, V.; et al. Global burden of 369 diseases and injuries in 204 countries and territories, 1990-2019: A systematic analysis for the Global Burden of Disease Study 2019. Lancet 2020, 396, 1204–1222. [Google Scholar] [CrossRef]
  5. Song, Y.; Liu, J.; Zhao, K.; Gao, L.; Zhao, J. Cholesterol-induced toxicity: An integrated view of the role of cholesterol in multiple diseases. Cell Metab. 2021, 33, 1911–1925. [Google Scholar] [CrossRef] [PubMed]
  6. Chaggar, P.S.; Shaw, S.M.; Williams, S.G. Effect of antipsychotic medications on glucose and lipid levels. J. Clin. Pharmacol. 2011, 51, 631–638. [Google Scholar] [CrossRef]
  7. Kliewer, S.A.; Moore, J.T.; Wade, L.; Staudinger, J.L.; Watson, M.A.; Jones, S.A.; McKee, D.D.; Oliver, B.B.; Willson, T.M.; Zetterström, R.H.; et al. An Orphan Nuclear Receptor Activated by Pregnanes Defines a Novel Steroid Signaling Pathway. Cell 1998, 92, 73–82. [Google Scholar] [CrossRef] [Green Version]
  8. Hakkola, J.; Hukkanen, J.; Turpeinen, M.; Pelkonen, O. Inhibition and induction of CYP enzymes in humans: An update. Arch. Toxicol. 2020, 94, 3671–3722. [Google Scholar] [CrossRef] [PubMed]
  9. Cai, X.; Young, G.M.; Xie, W. The xenobiotic receptors PXR and CAR in liver physiology, an update. Biochim. Biophys. Acta Mol. Basis Dis. 2021, 1867, 166101. [Google Scholar] [CrossRef]
  10. Ihunnah, C.A.; Jiang, M.; Xie, W. Nuclear receptor PXR, transcriptional circuits and metabolic relevance. Biochim. Biophys. Acta Mol. Basis Dis. 2011, 1812, 956–963. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Hukkanen, J.; Hakkola, J. PXR and 4β-Hydroxycholesterol Axis and the Components of Metabolic Syndrome. Cells 2020, 9, 2445. [Google Scholar] [CrossRef] [PubMed]
  12. Hukkanen, J.; Hakkola, J.; Rysä, J. Pregnane X receptor (PXR)—A contributor to the diabetes epidemic? Drug Metabol. Drug Interact. 2014, 29, 3–15. [Google Scholar] [CrossRef]
  13. Toporova, L.; Balaguer, P. Nuclear receptors are the major targets of endocrine disrupting chemicals. Mol. Cell. Endocrinol. 2020, 502, 110665. [Google Scholar] [CrossRef] [PubMed]
  14. Isojärvi, J.I.T.; Myllylä, V.V.; Pakarinen, A.J. Serum lipid levels during carbamazepine medication. A prospective study. Arch. Neurol. 1993, 50, 590–593. [Google Scholar] [CrossRef] [PubMed]
  15. Eirís, J.; Novo-Rodríguez, M.I.; Del Río, M.; Meseguer, P.; Del Río, M.C.; Castro-Gago, M. The effects on lipid and apolipoprotein serum levels of long-term carbamazepine, valproic acid and phenobarbital therapy in children with epilepsy. Epilepsy Res. 2000, 41, 1–7. [Google Scholar] [CrossRef]
  16. Luoma, P.V.; Reunanen, M.I.; Sotaniemi, E.A. Changes in serum triglyceride and cholesterol levels during long-term phenytoin treatment for epilepsy. Acta Med. Scand. 1979, 206, 229–231. [Google Scholar] [CrossRef]
  17. Katsiki, N.; Mikhailidis, D.P.; Nair, D.R. The effects of antiepileptic drugs on vascular risk factors: A narrative review. Seizure 2014, 23, 677–684. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Calandre, E.P.; Rodriguez-Lopez, C.; Blazquez, A.; Cano, D. Serum lipids, lipoproteins and apolipoproteins A and B in epileptic patients treated with valproic acid, carbamazepine or phenobarbital. Acta Neurol. Scand. 1991, 83, 250–253. [Google Scholar] [CrossRef] [PubMed]
  19. Nikolaos, T.; Stylianos, G.; Chryssoula, N.; Irini, P.; Christos, M.; Dimitrios, T.; Konstantinos, P.; Antonis, T. The effect of long-term antiepileptic treatment on serum cholesterol (TC, HDL, LDL) and triglyceride levels in adult epileptic patients on monotherapy. Med. Sci. Monit. 2004, 10, MT50–MT52. [Google Scholar] [PubMed]
  20. Coyne, M.J.; Bonorris, G.G.; Goldstein, L.I.; Schoenfield, L.J. Effect of chenodeoxycholic acid and phenobarbital on the rate-limiting enzymes of hepatic cholesterol and bile acid synthesis in patients with gallstones. J. Lab. Clin. Med. 1976, 87, 281–291. [Google Scholar] [CrossRef] [PubMed]
  21. Sberna, A.L.; Assem, M.; Gautier, T.; Grober, J.; Guiu, B.; Jeannin, A.; Pais De Barros, J.P.; Athias, A.; Lagrost, L.; Masson, D. Constitutive androstane receptor activation stimulates faecal bile acid excretion and reverse cholesterol transport in mice. J. Hepatol. 2011, 55, 154–161. [Google Scholar] [CrossRef] [PubMed]
  22. Chen, J.; Raymond, K. Roles of rifampicin in drug-drug interactions: Underlying molecular mechanisms involving the nuclear pregnane X receptor. Ann. Clin. Microbiol. Antimicrob. 2006, 5, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Feely, J.; Clee, M.; Pereira, L.; Guy, E. Enzyme induction with rifampicin; lipoproteins and drug binding to alpha 1-acid glycoprotein. Br. J. Clin. Pharmacol. 1983, 16, 195–197. [Google Scholar] [CrossRef] [Green Version]
  24. Lütjohann, D.; Hahn, C.; Prange, W.; Sudhop, T.; Axelson, M.; Sauerbruch, T.; von Bergmann, K.; Reichel, C. Influence of rifampin on serum markers of cholesterol and bile acid synthesis in men. Int. J. Clin. Pharmacol. Ther. 2004, 42, 307–313. [Google Scholar] [CrossRef] [PubMed]
  25. Ohnhaus, E.E.; Kirchhof, B.; Peheim, E. Effect of enzyme induction on plasma lipids using antipyrine, phenobarbital, and rifampicin. Clin. Pharmacol. Ther. 1979, 25, 591–597. [Google Scholar] [CrossRef] [PubMed]
  26. Bjorkhem, I.; Miettinen, T.; Reihner, E.; Ewerth, S.; Angelin, B.; Einarsson, K. Correlation between serum levels of some cholesterol precursors and activity of HMG-CoA reductase in human liver. J. Lipid Res. 1987, 28, 1137–1143. [Google Scholar] [CrossRef]
  27. Kasichayanula, S.; Boulton, D.W.; Luo, W.L.; Rodrigues, A.D.; Yang, Z.; Goodenough, A.; Lee, M.; Jemal, M.; LaCreta, F. Validation of 4β-hydroxycholesterol and evaluation of other endogenous biomarkers for the assessment of CYP3A activity in healthy subjects. Br. J. Clin. Pharmacol. 2014, 78, 1122–1134. [Google Scholar] [CrossRef] [Green Version]
  28. Rysä, J.; Buler, M.; Savolainen, M.J.; Ruskoaho, H.; Hakkola, J.; Hukkanen, J. Pregnane X Receptor Agonists Impair Postprandial Glucose Tolerance. Clin. Pharmacol. Ther. 2013, 93, 556–563. [Google Scholar] [CrossRef] [PubMed]
  29. Hassani-Nezhad-Gashti, F.; Salonurmi, T.; Hautajärvi, H.; Rysä, J.; Hakkola, J.; Hukkanen, J. Pregnane X receptor activator rifampin increases blood pressure and stimulates plasma renin activity. Clin. Pharmacol. Ther. 2020, 108, 856–865. [Google Scholar] [CrossRef]
  30. Karpale, M.; Käräjämäki, A.J.; Kummu, O.; Gylling, H.; Hyötyläinen, T.; Orešič, M.; Tolonen, A.; Hautajärvi, H.; Savolainen, M.J.; Ala-Korpela, M.; et al. Activation of nuclear receptor PXR induces atherogenic lipids and PCSK9 through SREBP2-mediated mechanism. Br. J. Pharmacol. 2021, 178, 2461–2481. [Google Scholar] [CrossRef] [PubMed]
  31. Diczfalusy, U.; Nylén, H.; Elander, P.; Bertilsson, L. 4β-hydroxycholesterol, an endogenous marker of CYP3A4/5 activity in humans. Br. J. Clin. Pharmacol. 2011, 71, 183–189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Theile, D.; Haefeli, W.E.; Weiss, J. Effects of adrenolytic mitotane on drug elimination pathways assessed in vitro. Endocrine 2015, 49, 842–853. [Google Scholar] [CrossRef] [PubMed]
  33. Kroiss, M.; Quinkler, M.; Lutz, W.K.; Allolio, B.; Fassnacht, M. Drug interactions with mitotane by induction of CYP3A4 metabolism in the clinical management of adrenocortical carcinoma. Clin. Endocrinol. 2011, 75, 585–591. [Google Scholar] [CrossRef]
  34. Maher, V.; Trainer, P.J.; Scoppola, A.; Anderson, J.V.; Thompson, G.R.; Besser, G.M. Possible Mechanism and Treatment of o,p’DDD-induced Hypercholesterolaemia. Q. J. Med. 1992, 84, 671–679. [Google Scholar] [PubMed]
  35. Vikner, M.E.; Krogh, J.; Daugaard, G.; Andreassen, M. Metabolic and hormonal side effects of mitotane treatment for adrenocortical carcinoma: A retrospective study in 50 Danish patients. Clin. Endocrinol. 2021, 94, 141–149. [Google Scholar] [CrossRef] [PubMed]
  36. Molnar, G.D.; Nunn, S.L.; Tauxe, W.N. The effect of o,p’-DDD therapy on plasma cholesterol in adrenal carcinoma. Proc. Staff Meet. Mayo Clin. 1961, 36, 618–620. [Google Scholar] [PubMed]
  37. Lu, Y.; Feskens, E.J.M.; Boer, J.M.A.; Müller, M. The potential influence of genetic variants in genes along bile acid and bile metabolic pathway on blood cholesterol levels in the population. Atherosclerosis 2010, 210, 14–27. [Google Scholar] [CrossRef] [PubMed]
  38. Dietschy, J.M.; Turley, S.D. Control of cholesterol turnover in the mouse. J. Biol. Chem. 2002, 277, 3801–3804. [Google Scholar] [CrossRef] [Green Version]
  39. Mitsche, M.A.; McDonald, J.G.; Hobbs, H.H.; Cohen, J.C. Flux analysis of cholesterol biosynthesis in vivo reveals multiple tissue and cell-type specific pathways. Elife 2015, 4, e07999. [Google Scholar] [CrossRef]
  40. Shimano, H.; Sato, R. SREBP-regulated lipid metabolism: Convergent physiology—Divergent pathophysiology. Nat. Rev. Endocrinol. 2017, 13, 710–730. [Google Scholar] [CrossRef] [PubMed]
  41. Luo, J.; Yang, H.; Song, B.L. Mechanisms and regulation of cholesterol homeostasis. Nat. Rev. Mol. Cell Biol. 2019, 21, 225–245. [Google Scholar] [CrossRef]
  42. Sever, N.; Yang, T.; Brown, M.S.; Goldstein, J.L.; DeBose-Boyd, R.A. Accelerated Degradation of HMG CoA Reductase Mediated by Binding of Insig-1 to Its Sterol-Sensing Domain. Mol. Cell 2003, 11, 25–33. [Google Scholar] [CrossRef]
  43. Roth, A.; Looser, R.; Kaufmann, M.; Blättler, S.M.; Rencurel, F.; Huang, W.; Moore, D.D.; Meyer, U.A. Regulatory Cross-Talk between Drug Metabolism and Lipid Homeostasis: Constitutive Androstane Receptor and Pregnane X Receptor Increase Insig-1 Expression. Mol. Pharmacol. 2008, 73, 1282–1289. [Google Scholar] [CrossRef] [Green Version]
  44. Zhou, J.; Zhai, Y.; Mu, Y.; Gong, H.; Uppal, H.; Toma, D.; Ren, S.; Evans, R.M.; Xie, W. A novel pregnane X receptor-mediated and sterol regulatory element-binding protein-independent lipogenic pathway. J. Biol. Chem. 2006, 281, 15013–15020. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Bitter, A.; Rümmele, P.; Klein, K.; Kandel, B.A.; Rieger, J.K.; Nüssler, A.K.; Zanger, U.M.; Trauner, M.; Schwab, M.; Burk, O. Pregnane X receptor activation and silencing promote steatosis of human hepatic cells by distinct lipogenic mechanisms. Arch. Toxicol. 2015, 89, 2089–2103. [Google Scholar] [CrossRef]
  46. Ann Barretto, S.; Lasserre, F.; Fougerat, A.; Smith, L.; Fougeray, T.; Lukowicz, C.; Polizzi, A.; Smati, S.; Régnier, M.; Naylies, C.; et al. Gene expression profiling reveals that PXR activation inhibits hepatic PPARα activity and decreases FGF21 secretion in male C57BL6/J mice. Int. J. Mol. Sci. 2019, 20, 3767. [Google Scholar] [CrossRef] [Green Version]
  47. Lin, Z.; Pan, X.; Wu, F.; Ye, D.; Zhang, Y.; Wang, Y.; Jin, L.; Lian, Q.; Huang, Y.; Ding, H.; et al. Fibroblast growth factor 21 prevents atherosclerosis by suppression of hepatic sterol regulatory element-binding protein-2 and induction of adiponectin in mice. Circulation 2015, 131, 1861–1871. [Google Scholar] [CrossRef] [Green Version]
  48. Bloch, K. The biological synthesis of cholesterol. Science 1965, 150, 19–28. [Google Scholar] [CrossRef] [PubMed]
  49. Kandutsch, A.A.; Russell, A.E. Preputial gland tumor sterols. 2. The identification of 4 alpha-methyl-Delta 8-cholesten-3 beta-ol. J. Biol. Chem. 1960, 235, 2253–2255. [Google Scholar] [CrossRef]
  50. Gwag, T.; Meng, Z.; Sui, Y.; Helsley, R.N.; Park, S.H.; Wang, S.; Greenberg, R.N.; Zhou, C. Non-nucleoside reverse transcriptase inhibitor efavirenz activates PXR to induce hypercholesterolemia and hepatic steatosis. J. Hepatol. 2019, 70, 930–940. [Google Scholar] [CrossRef]
  51. Gill, S.; Stevenson, J.; Kristiana, I.; Brown, A.J. Cholesterol-dependent degradation of squalene monooxygenase, a control point in cholesterol synthesis beyond HMG-CoA reductase. Cell Metab. 2011, 13, 260–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Chua, N.K.; Howe, V.; Jatana, N.; Thukral, L.; Brown, A.J. A conserved degron containing an amphipathic helix regulates the cholesterol-mediated turnover of human squalene monooxygenase, a rate-limiting enzyme in cholesterol synthesis. J. Biol. Chem. 2017, 292, 19959–19973. [Google Scholar] [CrossRef] [Green Version]
  53. Spann, N.J.; Garmire, L.X.; McDonald, J.G.; Myers, D.S.; Milne, S.B.; Shibata, N.; Reichart, D.; Fox, J.N.; Shaked, I.; Heudobler, D.; et al. Regulated accumulation of desmosterol integrates macrophage lipid metabolism and inflammatory responses. Cell 2012, 151, 138–152. [Google Scholar] [CrossRef] [Green Version]
  54. Warden, B.A.; Fazio, S.; Shapiro, M.D. The PCSK9 revolution: Current status, controversies, and future directions. Trends Cardiovasc. Med. 2020, 30, 179–185. [Google Scholar] [CrossRef]
  55. Abifadel, M.; Varret, M.; Rabès, J.-P.; Allard, D.; Ouguerram, K.; Devillers, M.; Cruaud, C.; Benjannet, S.; Wickham, L.; Erlich, D.; et al. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat. Genet. 2003, 34, 154–156. [Google Scholar] [CrossRef]
  56. Seidah, N.G.; Awan, Z.; Chrétien, M.; Mbikay, M. PCSK9: A key modulator of cardiovascular health. Circ. Res. 2014, 114, 1022–1036. [Google Scholar] [CrossRef]
  57. Dong, B.; Wu, M.; Li, H.; Kraemer, F.B.; Adeli, K.; Seidah, N.G.; Park, S.W.; Liu, J. Strong induction of PCSK9 gene expression through HNF1alpha and SREBP2: Mechanism for the resistance to LDL-cholesterol lowering effect of statins in dyslipidemic hamsters. J. Lipid Res. 2010, 51, 1486–1495. [Google Scholar] [CrossRef] [Green Version]
  58. Sahebkar, A.; Simental-Mendía, L.E.; Guerrero-Romero, F.; Golledge, J.; Watts, G.F. Effect of statin therapy on plasma proprotein convertase subtilisin kexin 9 (PCSK9) concentrations: A systematic review and meta-analysis of clinical trials. Diabetes Obes. Metab. 2015, 17, 1042–1055. [Google Scholar] [CrossRef]
  59. Abumiya, M.; Akamine, Y.; Sato, S.; Takahashi, S.; Yoshioka, T.; Kameoka, Y.; Takahashi, N.; Miura, M. Effects of proprotein convertase subtilisin/kexin type 9 and nilotinib plasma concentrations on nilotinib-induced hypercholesterolaemia in patients with chronic myeloid leukaemia. J. Clin. Pharm. Ther. 2021, 46, 382–387. [Google Scholar] [CrossRef]
  60. Glerup, S.; Schulz, R.; Laufs, U.; Schlüter, K.-D. Physiological and therapeutic regulation of PCSK9 activity in cardiovascular disease. Basic Res. Cardiol. 2017, 112, 32. [Google Scholar] [CrossRef] [Green Version]
  61. Sato, S.; Akamine, Y.; Kagaya, H.; Saito, M.; Inoue, T.; Numakura, K.; Habuchi, T.; Satoh, S.; Miura, M. Changes in PCSK9 and LDL cholesterol concentrations by everolimus treatment and their effects on polymorphisms in PCSK9 and mTORC1. Pharmacol. Rep. 2020, 72, 622–630. [Google Scholar] [CrossRef] [PubMed]
  62. Simha, V.; Qin, S.; Shah, P.; Smith, B.H.; Kremers, W.K.; Kushwaha, S.; Wang, L.; Pereira, N.L. Sirolimus Therapy Is Associated with Elevation in Circulating PCSK9 Levels in Cardiac Transplant Patients. J. Cardiovasc. Transl. Res. 2017, 10, 9–15. [Google Scholar] [CrossRef] [Green Version]
  63. Phan, B.A.P.; Dayspring, T.D.; Toth, P.P. Ezetimibe therapy: Mechanism of action and clinical update. Vasc. Health Risk Manag. 2012, 8, 415–427. [Google Scholar]
  64. Miettinen, T.A.; Puska, P.; Gylling, H.; Vanhanen, H.; Vartiainen, E. Reduction of Serum Cholesterol with Sitostanol-Ester Margarine in a Mildly Hypercholesterolemic Population. N. Engl. J. Med. 1995, 333, 1308–1312. [Google Scholar] [CrossRef] [Green Version]
  65. Sui, Y.; Helsley, R.N.; Park, S.-H.; Song, X.; Liu, Z.; Zhou, C. Intestinal pregnane X receptor links xenobiotic exposure and hypercholesterolemia. Mol. Endocrinol. 2015, 29, 765–776. [Google Scholar] [CrossRef] [Green Version]
  66. Jia, L.; Betters, J.L.; Yu, L. Niemann-Pick C1-Like 1 (NPC1L1) protein in intestinal and hepatic cholesterol transport. Annu. Rev. Physiol. 2011, 73, 239–259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Ma, X.; Shah, Y.M.; Guo, G.L.; Wang, T.; Krausz, K.W.; Idle, J.R.; Gonzalez, F.J. Rifaximin is a gut-specific human pregnane X receptor activator. J. Pharmacol. Exp. Ther. 2007, 322, 391–398. [Google Scholar] [CrossRef] [PubMed]
  68. Cheng, J.; Krausz, K.W.; Tanaka, N.; Gonzalez, F.J. Chronic exposure to rifaximin causes hepatic steatosis in pregnane X receptor-humanized mice. Toxicol. Sci. 2012, 129, 456–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Sonoda, J.; Chong, L.W.; Downes, M.; Barish, G.D.; Coulter, S.; Liddle, C.; Lee, C.H.; Evans, R.M. Pregnane X receptor prevents hepatorenal toxicity from cholesterol metabolites. Proc. Natl. Acad. Sci. USA 2005, 102, 2198–2203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Xie, W.; Radominska-Pandya, A.; Shi, Y.; Simon, C.M.; Nelson, M.C.; Ong, E.S.; Waxman, D.J.; Evans, R.M. An essential role for nuclear receptors SXR/PXR in detoxification of cholestatic bile acids. Proc. Natl. Acad. Sci. USA 2001, 98, 3375–3380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. He, J.; Nishida, S.; Xu, M.; Makishima, M.; Xie, W. PXR Prevents Cholesterol Gallstone Disease by Regulating Biosynthesis and Transport of Bile Salts. Gastroenterology 2011, 140, 2095–2106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Staudinger, J.L.; Goodwin, B.; Jones, S.A.; Hawkins-Brown, D.; MacKenzie, K.I.; LaTour, A.; Liu, Y.; Klaassen, C.D.; Brown, K.K.; Reinhard, J.; et al. The nuclear receptor PXR is a lithocholic acid sensor that protects against liver toxicity. Proc. Natl. Acad. Sci. USA 2001, 98, 3369–3374. [Google Scholar] [CrossRef] [Green Version]
  73. Kast, H.R.; Goodwin, B.; Tarr, P.T.; Jones, S.A.; Anisfeld, A.M.; Stoltz, C.M.; Tontonoz, P.; Kliewer, S.; Willson, T.M.; Edwards, P.A. Regulation of multidrug resistance-associated protein 2 (ABCC2) by the nuclear receptors pregnane X receptor, farnesoid X-activated receptor, and constitutive androstane receptor. J. Biol. Chem. 2002, 277, 2908–2915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Bhalla, S.; Ozalp, C.; Fang, S.; Xiang, L.; Kemper, J.K. Ligand-activated pregnane X receptor interferes with HNF-4 signaling by targeting a common coactivator PGC-1α. Functional implications in hepatic cholesterol and glucose metabolism. J. Biol. Chem. 2004, 279, 45139–45147. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Li, T.; Chiang, J.Y.L. Mechanism of rifampicin and pregnane X receptor inhibition of human cholesterol 7α-hydroxylase gene transcription. Am. J. Physiol. Gastrointest. Liver Physiol. 2005, 288, 74–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Hassani-Nezhad-Gashti, F.; Kummu, O.; Karpale, M.; Rysä, J.; Hakkola, J. Nutritional status modifies pregnane X receptor regulated transcriptome. Sci. Rep. 2019, 9, 16728. [Google Scholar] [CrossRef]
  77. Marschall, H.-U.; Wagner, M.; Zollner, G.; Fickert, P.; Diczfalusy, U.; Gumhold, J.; Silbert, D.; Fuchsbichler, A.; Benthin, L.; Grundström, R.; et al. Complementary stimulation of hepatobiliary transport and detoxification systems by rifampicin and ursodeoxycholic acid in humans. Gastroenterology 2005, 129, 476–485. [Google Scholar] [CrossRef]
  78. Temel, R.E.; Brown, J.M. A new model of reverse cholesterol transport: EnTICEing strategies to stimulate intestinal cholesterol excretion. Trends Pharmacol. Sci. 2015, 36, 440–451. [Google Scholar] [CrossRef] [Green Version]
  79. Calandre, E.P.; Porta, B.S.; de la Calzada, D.G. The Effect of Chronic Phenytoin Treatment on Serum Lipid Profile in Adult Epileptic Patients. Epilepsia 1992, 33, 154–157. [Google Scholar] [CrossRef]
  80. Jakubus, T.; Michalska-Jakubus, M.; Łukawski, K.; Janowska, A.; Czuczwar, S.J. Atherosclerotic risk among children taking antiepileptic drugs. Pharmacol. Rep. 2009, 61, 411–423. [Google Scholar] [CrossRef]
  81. O’Neill, B.; Callaghan, N.; Stapleton, M.; Molloy, W. Serum elevation of high density lipoprotein (HDL) cholesterol in epileptic patients taking carbamazepine or phenytoin. Acta Neurol. Scand. 1982, 65, 104–109. [Google Scholar] [CrossRef]
  82. Salonurmi, T.; Nabil, H.; Ronkainen, J.; Hyötyläinen, T.; Hautajärvi, H.; Savolainen, M.J.; Tolonen, A.; Orešič, M.; Känsäkoski, P.; Rysä, J.; et al. 4β-Hydroxycholesterol Signals From the Liver to Regulate Peripheral Cholesterol Transporters. Front. Pharmacol. 2020, 11, 361. [Google Scholar] [CrossRef] [PubMed]
  83. Domecq, J.P.; Prutsky, G.; Leppin, A.; Sonbol, M.B.; Altayar, O.; Undavalli, C.; Wang, Z.; Elraiyah, T.; Brito, J.P.; Mauck, K.F.; et al. Drugs commonly associated with weight change: A systematic review and meta-analysis. J. Clin. Endocrinol. Metab. 2015, 100, 363–370. [Google Scholar] [CrossRef] [PubMed]
  84. Bertilsson, G.; Heidrich, J.; Svensson, K.; Åsman, M.; Jendeberg, L.; Sydow-Bäckman, M.; Ohlsson, R.; Postlind, H.; Blomquist, P.; Berkenstam, A. Identification of a human nuclear receptor defines a new signaling pathway for CYP3A induction. Proc. Natl. Acad. Sci. USA 1998, 95, 12208–12213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Grewal, G.K.; Singh, K.D.; Kanojia, N.; Rawat, C.; Kukal, S.; Jajodia, A.; Singhal, A.; Misra, R.; Nagamani, S.; Muthusamy, K.; et al. Exploring the Carbamazepine Interaction with Human Pregnane X Receptor and Effect on ABCC2 Using in Vitro and in Silico Approach. Pharm. Res. 2017, 34, 1444–1458. [Google Scholar] [CrossRef]
  86. Moreland, T.; Park, B.; Rylance, G. Microsomal enzyme induction in children: The influence of carbamazepine treatment on antipyrine kinetics, 6 beta-hydroxycortisol excretion and plasma gamma-glutamyltranspeptidase activity. Br. J. Clin. Pharmacol. 1982, 14, 861–865. [Google Scholar] [CrossRef] [Green Version]
  87. Oscarson, M.; Zanger, U.M.; Rifki, O.F.; Klein, K.; Eichelbaum, M.; Meyer, U.A. Transcriptional profiling of genes induced in the livers of patients treated with carbamazepine. Clin. Pharmacol. Ther. 2006, 80, 440–456. [Google Scholar] [CrossRef] [PubMed]
  88. Crawford, P.; Chadwick, D.; Martin, C.; Tjia, J.; Back, D.; Orme, M. The interaction of phenytoin and carbamazepine with combined oral contraceptive steroids. Br. J. Clin. Pharmacol. 1990, 30, 892. [Google Scholar] [CrossRef] [Green Version]
  89. Lynch, C.; Sakamuru, S.; Huang, R.; Niebler, J.; Ferguson, S.S.; Xia, M. Characterization of human pregnane X receptor activators identified from a screening of the Tox21 compound library. Biochem. Pharmacol. 2021, 184. [Google Scholar] [CrossRef]
  90. Duran, I.; Carles, J.; Bulat, I.; Hellemans, P.; Mitselos, A.; Ward, P.; Jiao, J.; Armas, D.; Chien, C. Pharmacokinetic Drug-Drug Interaction of Apalutamide, Part 1: Clinical Studies in Healthy Men and Patients with Castration-Resistant Prostate Cancer. Clin. Pharmacokinet. 2020, 59, 1135–1148. [Google Scholar] [CrossRef]
  91. Chortis, V.; Taylor, A.E.; Schneider, P.; Tomlinson, J.W.; Hughes, B.A.; O’Neil, D.M.; Libé, R.; Allolio, B.; Bertagna, X.; Bertherat, J.; et al. Mitotane therapy in adrenocortical cancer induces CYP3A4 and inhibits 5α-reductase, explaining the need for personalized glucocorticoid and androgen replacement. J. Clin. Endocrinol. Metab. 2013, 98, 161–171. [Google Scholar] [CrossRef] [Green Version]
  92. Van Erp, N.P.; Guchelaar, H.J.; Ploeger, B.A.; Romijn, J.A.; Den Hartigh, J.; Gelderblom, H. Mitotane has a strong and a durable inducing effect on CYP3A4 activity. Eur. J. Endocrinol. 2011, 164, 621–626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Harmsen, S.; Meijerman, I.; Maas-Bakker, R.F.; Beijnen, J.H.; Schellens, J.H.M. PXR-mediated P-glycoprotein induction by small molecule tyrosine kinase inhibitors. Eur. J. Pharm. Sci. 2012, 48, 644–649. [Google Scholar] [CrossRef]
  94. Danek, P.J.; Basińska-Ziobroń, A.; Wójcikowski, J.; Daniel, W.A. Levomepromazine and clozapine induce the main human cytochrome P450 drug metabolizing enzyme CYP3A4. Pharmacol. Rep. 2021, 73, 303–308. [Google Scholar] [CrossRef]
  95. Meng, Z.; Gwag, T.; Sui, Y.; Park, S.-H.H.; Zhou, X.; Zhou, C. The atypical antipsychotic quetiapine induces hyperlipidemia by activating intestinal PXR signaling. JCI Insight 2019, 4, e125657. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Hariparsad, N.; Nallani, S.C.; Sane, R.S.; Buckley, D.J.; Buckley, A.R.; Desai, P.B. Induction of CYP3A4 by efavirenz in primary human hepatocytes: Comparison with rifampin and phenobarbital. J. Clin. Pharmacol. 2004, 44, 1273–1281. [Google Scholar] [CrossRef] [PubMed]
  97. Mouly, S.; Lown, K.S.; Kornhauser, D.; Joseph, J.L.; Fiske, W.D.; Benedek, I.H.; Watkins, P.B. Hepatic but not intestinal CYP3A4 displays dose-dependent induction by efavirenz in humans. Clin. Pharmacol. Ther. 2002, 72, 1–9. [Google Scholar] [CrossRef] [Green Version]
  98. Fellay, J.; Marzolini, C.; Decosterd, L.; Golay, K.P.; Baumann, P.; Buclin, T.; Telenti, A.; Eap, C.B. Variations of CYP3A activity induced by antiretroviral treatment in HIV-1 infected patients. Eur. J. Clin. Pharmacol. 2005, 60, 865–873. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Sharma, D.; Lau, A.J.; Sherman, M.A.; Chang, T.K.H. Agonism of human pregnane X receptor by rilpivirine and etravirine: Comparison with first generation non-nucleoside reverse transcriptase inhibitors. Biochem. Pharmacol. 2013, 85, 1700–1711. [Google Scholar] [CrossRef]
  100. Schöller-Gyüre, M.; Kakuda, T.N.; Raoof, A.; De Smedt, G.; Hoetelmans, R.M.W. Clinical pharmacokinetics and pharmacodynamics of etravirine. Clin. Pharmacokinet. 2009, 48, 561–574. [Google Scholar] [CrossRef]
  101. Kakuda, T.N.; Van Solingen-Ristea, R.M.; Onkelinx, J.; Stevens, T.; Aharchi, F.; De Smedt, G.; Peeters, M.; Leopold, L.; Hoetelmans, R.M.W. The effect of single- and multiple-dose etravirine on a drug cocktail of representative cytochrome P450 probes and digoxin in healthy subjects. J. Clin. Pharmacol. 2014, 54, 422–431. [Google Scholar] [CrossRef]
  102. Tibotec Intelence (etravirine) full prescribing information 2011. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2011/022187s008lbl.pdf (accessed on 13 December 2021).
  103. Solas, C.; Poizot-Martin, I.; Drogoul, M.P.; Ravaux, I.; Dhiver, C.; Lafeuillade, A.; Allegre, T.; Mokhtari, M.; Moreau, J.; Lepeu, G.; et al. Therapeutic drug monitoring of lopinavir/ritonavir given alone or with a non-nucleoside reverse transcriptase inhibitor. Br. J. Clin. Pharmacol. 2004, 57, 436–440. [Google Scholar] [CrossRef] [Green Version]
  104. Mildvan, D.; Yarrish, R.; Marshak, A.; Hutman, H.W.; McDonough, M.; Lamson, M.; Robinson, P. Pharmacokinetic interaction between nevirapine and ethinyl estradiol/norethindrone when administered concurrently to HIV-infected women. J. Acquir. Immune Defic. Syndr. 2002, 29, 471–477. [Google Scholar] [CrossRef] [PubMed]
  105. Dailly, E.; Tribut, O.; Tattevin, P.; Arvieux, C.; Perré, P.; Raffi, F.; Jolliet, P. Influence of tenofovir, nevirapine and efavirenz on ritonavir-boosted atazanavir pharmacokinetics in HIV-infected patients. Eur. J. Clin. Pharmacol. 2006, 62, 523–526. [Google Scholar] [CrossRef]
  106. Svärd, J.; Spiers, J.P.; Mulcahy, F.; Hennessy, M. Nuclear receptor-mediated induction of CYP450 by antiretrovirals: Functional consequences of NR1I2 (PXR) polymorphisms and differential prevalence in whites and sub-Saharan Africans. J. Acquir. Immune Defic. Syndr. 2010, 55, 536–549. [Google Scholar] [CrossRef] [PubMed]
  107. Dussault, I.; Lin, M.; Hollister, K.; Wang, E.H.; Synold, T.W.; Forman, B.M. Peptide Mimetic HIV Protease Inhibitors Are Ligands for the Orphan Receptor SXR. J. Biol. Chem. 2001, 276, 33309–33312. [Google Scholar] [CrossRef] [Green Version]
  108. Ouellet, D.; Hsu, A.; Qian, J.; Locke, C.S.; Eason, C.J.; Cavanaugh, J.H.; Leonard, J.M.; Granneman, G.R. Effect of ritonavir on the pharmacokinetics of ethinyl oestradiol in healthy female volunteers. Br. J. Clin. Pharmacol. 1998, 46, 111–116. [Google Scholar] [CrossRef] [Green Version]
  109. Hsu, A.; Granneman, G.R.; Witt, G.; Locke, C.; Denissen, J.; Molla, A.; Valdes, J.; Smith, J.; Erdman, K.; Lyons, N.; et al. Multiple-dose pharmacokinetics of ritonavir in human immunodeficiency virus-infected subjects. Antimicrob. Agents Chemother. 1997, 41, 898. [Google Scholar] [CrossRef] [Green Version]
  110. Li, L.; Welch, M.A.; Li, Z.; Mackowiak, B.; Heyward, S.; Swaan, P.W.; Wang, H. Mechanistic insights of phenobarbital-mediated activation of human but not mouse pregnane X receptor. Mol. Pharmacol. 2019, 96, 345–354. [Google Scholar] [CrossRef]
  111. Burstein, S.; Klaiber, E.L. Phenobarbital-induced increase in 6-beta-hydroxycortisol excretion: Clue to its significance in human urine. J. Clin. Endocrinol. Metab. 1965, 25, 293–296. [Google Scholar] [CrossRef] [PubMed]
  112. Wallace, K.; Cowie, D.E.; Konstantinou, D.K.; Hill, S.J.; Tjelle, T.E.; Axon, A.; Koruth, M.; White, S.A.; Carlsen, H.; Mann, D.A.; et al. The PXR is a drug target for chronic inflammatory liver disease. J. Steroid Biochem. Mol. Biol. 2010, 120, 137–148. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Shukla, S.J.; Sakamuru, S.; Huang, R.; Moeller, T.A.; Shinn, P.; Van Leer, D.; Auld, D.S.; Austin, C.P.; Xia, M. Identification of Clinically Used Drugs That Activate Pregnane X Receptors. Drug Metab. Dispos. 2011, 39, 151. [Google Scholar] [CrossRef] [Green Version]
  114. Pan, Y.; Li, L.; Kim, G.; Ekins, S.; Wang, H.; Swaan, P.W. Identification and Validation of Novel Human Pregnane X Receptor Activators among Prescribed Drugs via Ligand-Based Virtual Screening. Drug Metab. Dispos. 2011, 39, 337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Pascussi, J.M.; Drocourt, L.; Fabre, J.M.; Maurel, P.; Vilarem, M.J. Dexamethasone induces pregnane X receptor and retinoid X receptor-alpha expression in human hepatocytes: Synergistic increase of CYP3A4 induction by pregnane X receptor activators. Mol. Pharmacol. 2000, 58, 361–372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Pichard, L.; Fabre, I.; Daujat, M.; Domergue, J.; Joyeux, H.; Maurel, P. Effect of corticosteroids on the expression of cytochromes P450 and on cyclosporin A oxidase activity in primary cultures of human hepatocytes. Mol. Pharmacol. 1992, 41, 1047–1055. [Google Scholar]
  117. McCune, J.S.; Hawke, R.L.; LeCluyse, E.L.; Gillenwater, H.H.; Hamilton, G.; Ritchie, J.; Lindley, C. In vivo and in vitro induction of human cytochrome P4503A4 by dexamethasone. Clin. Pharmacol. Ther. 2000, 68, 356–366. [Google Scholar] [CrossRef]
  118. Roberts, P.J.; Rollins, K.D.; Kashuba, A.D.M.; Paine, M.F.; Nelsen, A.C.; Williams, E.E.; Moran, C.; Lamba, J.K.; Schuetz, E.G.; Hawke, R.L. The Influence of CYP3A5 Genotype on Dexamethasone Induction of CYP3A Activity in African Americans. Drug Metab. Dispos. 2008, 36, 1465. [Google Scholar] [CrossRef] [Green Version]
  119. Watkins, P.B.; Murray, S.A.; Winkelman, L.G.; Heuman, D.M.; Wrighton, S.A.; Guzelian, P.S. Erythromycin breath test as an assay of glucocorticoid-inducible liver cytochromes P-450. Studies in rats and patients. J. Clin. Investig. 1989, 83, 688–697. [Google Scholar] [CrossRef] [Green Version]
  120. Van Duijnhoven, E.M.; Boots, J.M.M.; Christiaans, M.H.L.; Stolk, L.M.L.; Undre, N.A.; Van Hooff, J.P. Increase in tacrolimus trough levels after steroid withdrawal. Transpl. Int. 2003, 16, 721–725. [Google Scholar] [CrossRef]
  121. Press, R.R.; Ploeger, B.A.; Den Hartigh, J.; Van Der Straaten, T.; Van Pelt, H.; Danhof, M.; De Fijter, H.; Guchelaar, H.J. Explaining variability in ciclosporin exposure in adult kidney transplant recipients. Eur. J. Clin. Pharmacol. 2010, 66, 579. [Google Scholar] [CrossRef] [Green Version]
  122. Anglicheau, D.; Flamant, M.; Schlageter, M.H.; Martinez, F.; Cassinat, B.; Beaune, P.; Legendre, C.; Thervet, E. Pharmacokinetic interaction between corticosteroids and tacrolimus after renal transplantation. Nephrol. Dial. Transplant 2003, 18, 2409–2414. [Google Scholar] [CrossRef] [Green Version]
  123. Chen, X.P.; Tan, Z.R.; Huang, S.L.; Huang, Z.; Ou-Yang, D.S.; Zhou, H.H. Isozyme-specific induction of low-dose aspirin on cytochrome P450 in healthy subjects. Clin. Pharmacol. Ther. 2003, 73, 264–271. [Google Scholar] [CrossRef]
  124. Novotna, A.; Dvorak, Z. Omeprazole and lansoprazole enantiomers induce CYP3A4 in human hepatocytes and cell lines via glucocorticoid receptor and pregnane X receptor axis. PLoS ONE 2014, 9, e105580. [Google Scholar] [CrossRef]
  125. Wang, K.; Chen, S.; Xie, W.; Wan, Y.J.Y. Retinoids induce cytochrome P450 3A4 through RXR/VDR-mediated pathway. Biochem. Pharmacol. 2008, 75, 2204–2213. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Padda, S.K.; Chhatwani, L.; Zhou, L.; Jacobs, C.D.; Lopez-Anaya, A.; Wakelee, H.A. Phase I and pharmacokinetic study of bexarotene in combination with gefitinib in the third-line treatment of non-small-cell lung cancer: Brief report. Anticancer. Drugs 2013, 24, 731–735. [Google Scholar] [CrossRef] [PubMed]
  127. Wakelee, H.A.; Takimoto, C.H.; Lopez-Anaya, A.; Chu, Q.; Middleton, G.; Dunlop, D.; Ramlau, R.; Leighl, N.; Rowinsky, E.K.; Hao, D.; et al. The effect of bexarotene on atorvastatin pharmacokinetics: Results from a phase I trial of bexarotene plus chemotherapy in patients with advanced non-small cell lung cancer. Cancer Chemother. Pharmacol. 2012, 69, 563–571. [Google Scholar] [CrossRef] [PubMed]
  128. Novotna, A.; Kamenickova, A.; Pecova, M.; Korhonova, M.; Bartonkova, I.; Dvorak, Z. Profiling of enantiopure drugs towards aryl hydrocarbon (AhR), glucocorticoid (GR) and pregnane X (PXR) receptors in human reporter cell lines. Chem. Biol. Interact. 2014, 208, 64–76. [Google Scholar] [CrossRef]
  129. Rowland, A.; van Dyk, M.; Warncken, D.; Mangoni, A.A.; Sorich, M.J.; Rowland, A. Evaluation of modafinil as a perpetrator of metabolic drug–drug interactions using a model informed cocktail reaction phenotyping trial protocol. Br. J. Clin. Pharmacol. 2018, 84, 501–509. [Google Scholar] [CrossRef] [Green Version]
  130. Pillinger, T.; McCutcheon, R.A.; Vano, L.; Mizuno, Y.; Arumuham, A.; Hindley, G.; Beck, K.; Natesan, S.; Efthimiou, O.; Cipriani, A.; et al. Comparative effects of 18 antipsychotics on metabolic function in patients with schizophrenia, predictors of metabolic dysregulation, and association with psychopathology: A systematic review and network meta-analysis. Lancet Psychiatry 2020, 7, 64–77. [Google Scholar] [CrossRef]
  131. Stroup, T.S.; Byerly, M.J.; Nasrallah, H.A.; Ray, N.; Khan, A.Y.; Lamberti, J.S.; Glick, I.D.; Steinbook, R.M.; McEvoy, J.P.; Hamer, R.M. Effects of switching from olanzapine, quetiapine, and risperidone to aripiprazole on 10-year coronary heart disease risk and metabolic syndrome status: Results from a randomized controlled trial. Schizophr. Res. 2013, 146, 190–195. [Google Scholar] [CrossRef] [Green Version]
  132. Cai, H.L.; Tan, Q.Y.; Jiang, P.; Dang, R.L.; Xue, Y.; Tang, M.M.; Xu, P.; Deng, Y.; Li, H.D.; Yao, J.K. A potential mechanism underlying atypical antipsychotics-induced lipid disturbances. Transl. Psychiatry 2015, 5, e661. [Google Scholar] [CrossRef] [PubMed]
  133. Le Hellard, S.; Theisen, F.M.; Haberhausen, M.; Raeder, M.B.; Fernø, J.; Gebhardt, S.; Hinney, A.; Remschmidt, H.; Krieg, J.C.; Mehler-Wex, C.; et al. Association between the insulin-induced gene 2 (INSIG2) and weight gain in a German sample of antipsychotic-treated schizophrenic patients: Perturbation of SREBP-controlled lipogenesis in drug-related metabolic adverse effects? Mol. Psychiatry 2009, 14, 308–317. [Google Scholar] [CrossRef] [PubMed]
  134. Sammalkorpi, K.; Valtonen, V.; Kerttula, Y.; Nikkilä, E.; Taskinen, M.R. Changes in serum lipoprotein pattern induced by acute infections. Metabolism 1988, 37, 859–865. [Google Scholar] [CrossRef]
  135. Deniz, O.; Gumus, S.; Yaman, H.; Ciftci, F.; Ors, F.; Cakir, E.; Tozkoparan, E.; Bilgic, H.; Ekiz, K. Serum total cholesterol, HDL-C and LDL-C concentrations significantly correlate with the radiological extent of disease and the degree of smear positivity in patients with pulmonary tuberculosis. Clin. Biochem. 2007, 40, 162–166. [Google Scholar] [CrossRef]
  136. Georgiadis, A.N.; Papavasiliou, E.C.; Lourida, E.S.; Alamanos, Y.; Kostara, C.; Tselepis, A.D.; Drosos, A.A. Atherogenic lipid profile is a feature characteristic of patients with early rheumatoid arthritis: Effect of early treatment—A prospective, controlled study. Arthritis Res. Ther. 2006, 8, R82. [Google Scholar] [CrossRef] [Green Version]
  137. Choi, H.K.; Seeger, J.D. Lipid profiles among US elderly with untreated rheumatoid arthritis—The Third National Health and Nutrition Examination Survey. J. Rheumatol. 2005, 32, 2311–2316. [Google Scholar]
  138. Robertson, J.; Peters, M.J.; McInnes, I.B.; Sattar, N. Changes in lipid levels with inflammation and therapy in RA: A maturing paradigm. Nat. Rev. Rheumatol. 2013, 9, 513–523. [Google Scholar] [CrossRef]
  139. Heslinga, S.C.; Peters, M.J.; Ter Wee, M.M.; Van Der Horst-Bruinsma, I.E.; Van Sijl, A.M.; Smulders, Y.M.; Nurmohamed, M.T. Reduction of inflammation drives lipid changes in ankylosing spondylitis. J. Rheumatol. 2015, 42, 1842–1845. [Google Scholar] [CrossRef]
  140. Holdaas, H.; Potena, L.; Saliba, F. MTOR inhibitors and dyslipidemia in transplant recipients: A cause for concern? Transplant. Rev. 2015, 29, 93–102. [Google Scholar] [CrossRef]
  141. Kockx, M.; Kritharides, L. Cyclosporin A-Induced Hyperlipidemia. In Lipoproteins—Role in Health and Diseases; InTech: Pittsburgh, PA, USA, 2012. [Google Scholar]
  142. Hüsing, A.; Kabar, I.; Schmidt, H.H. Lipids in liver transplant recipients. World J. Gastroenterol. 2016, 22, 3315–3324. [Google Scholar] [CrossRef]
  143. Agarwal, A.; Prasad, G.V.R. Post-transplant dyslipidemia: Mechanisms, diagnosis and management. World J. Transplant. 2016, 6, 125. [Google Scholar] [CrossRef] [PubMed]
  144. Kockx, M.; Glaros, E.; Leung, B.; Ng, T.W.; Berbée, J.F.P.; Deswaerte, V.; Nawara, D.; Quinn, C.; Rye, K.A.; Jessup, W.; et al. Low-Density Lipoprotein Receptor-Dependent and Low-Density Lipoprotein Receptor-Independent Mechanisms of Cyclosporin A-Induced Dyslipidemia. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 1338–1349. [Google Scholar] [CrossRef] [Green Version]
  145. Bakan, I.; Laplante, M. Connecting mTORC1 signaling to SREBP-1 activation. Curr. Opin. Lipidol. 2012, 23, 226–234. [Google Scholar] [CrossRef] [PubMed]
  146. Hemkens, L.G.; Bucher, H.C. HIV infection and cardiovascular disease. Eur. Heart J. 2014, 35, 1373–1381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  147. Triant, V.A.; Grinspoon, S.K. Epidemiology of ischemic heart disease in HIV. Curr. Opin. HIV Aids 2017, 12, 540–547. [Google Scholar] [CrossRef]
  148. Feeney, E.R.; Mallon, P.W.G. HIV and HAART-Associated Dyslipidemia. Open Cardiovasc. Med. J. 2011, 5, 49–63. [Google Scholar] [CrossRef]
  149. Wei, Y.; Tang, C.; Sant, V.; Li, S.; Poloyac, S.M.; Xie, W. A Molecular Aspect in the Regulation of Drug Metabolism: Does PXR-Induced Enzyme Expression Always Lead to Functional Changes in Drug Metabolism? Curr. Pharmacol. Rep. 2016, 2, 187–192. [Google Scholar] [CrossRef] [Green Version]
  150. Sberna, A.L.; Assem, M.; Xiao, R.; Ayers, S.; Gautier, T.; Guiu, B.; Deckert, V.; Chevriaux, A.; Grober, J.; Le Guern, N.; et al. Constitutive androstane receptor activation decreases plasma apolipoprotein B-containing lipoproteins and atherosclerosis in low-density lipoprotein receptor-deficient mice. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 2232–2239. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Activation of PXR stimulates cholesterol synthesis and induces Pcsk9 in the liver. The processes and molecules upregulated and downregulated by PXR activation have been indicated with blue and red coloring in the figure, respectively. PXR activation leads to nuclear accumulation of SREBP2, which consequently induces cholesterogenic genes, including the rate-limiting enzyme HMGCR, and thus induces cholesterol synthesis. Furthermore, PXR activation directly induces Sqle to increase the rate of squalene epoxidation. Induction of DHCR24 enzyme targets cholesterol synthesis flux to the Kandutsch–Russell pathway instead of the Bloch pathway. PXR activation appears to bypass the usual negative feedback mechanism controlling cholesterol synthesis, including inhibition of SREBP2–SCAP complex translocation from the ER to the Golgi apparatus by high cellular cholesterol levels and desmosterol, as well as the inhibitory effect of cholesterol on SQLE activity. Accumulation of plasma LDL-C level is potentiated by induction of the hepatic Pcsk9 gene and a consequent increase in circulating PCSK9 levels.
Figure 1. Activation of PXR stimulates cholesterol synthesis and induces Pcsk9 in the liver. The processes and molecules upregulated and downregulated by PXR activation have been indicated with blue and red coloring in the figure, respectively. PXR activation leads to nuclear accumulation of SREBP2, which consequently induces cholesterogenic genes, including the rate-limiting enzyme HMGCR, and thus induces cholesterol synthesis. Furthermore, PXR activation directly induces Sqle to increase the rate of squalene epoxidation. Induction of DHCR24 enzyme targets cholesterol synthesis flux to the Kandutsch–Russell pathway instead of the Bloch pathway. PXR activation appears to bypass the usual negative feedback mechanism controlling cholesterol synthesis, including inhibition of SREBP2–SCAP complex translocation from the ER to the Golgi apparatus by high cellular cholesterol levels and desmosterol, as well as the inhibitory effect of cholesterol on SQLE activity. Accumulation of plasma LDL-C level is potentiated by induction of the hepatic Pcsk9 gene and a consequent increase in circulating PCSK9 levels.
Cells 11 00313 g001
Table 1. Cholesterol-increasing drugs and their potential to activate PXR and induce CYP3A4.
Table 1. Cholesterol-increasing drugs and their potential to activate PXR and induce CYP3A4.
Drug ClassDrugMechanismIncreasesPXR
Agonist
CYP3A4
Inducer
Clinically
Relevant CYP3A4
Inducer
AndrogenMethyl
testosterone
Androgen receptor
activation
LDL
AntiarrhythmicAmiodaroneBlocking of voltage gated K+ and Ca2+ channelsLDL
AntibioticRifampicinBacterial RNA synthesis inhibitionCHOL, LDLYes [22,84]Yes [22]Yes [22]
AnticonvulsantCarbamazepineBlocking of central Na+ channelCHOL, LDLYes [85]Yes [86,87,88]Yes [86,87,88]
TiagabineGABA reuptake
inhibition
CHOL
AntidepressantParoxetineSelective serotonin reuptake inhibitionCHOL
SertralineSelective serotonin reuptake inhibitionCHOL
DuloxetineSerotonin and
noradrenaline reuptake inhibition
CHOL
VenlafaxinSerotonin and
noradrenaline reuptake inhibition
CHOL
AntigonadotropicDanazolAndrogen receptor
activation
CHOL, LDL
AntigoutFebuxostatXanthine-oxidase
inhibition
CHOL
AntihyperglycemicErtugliflozinSGLT-2 inhibitionCHOL, LDL
SotagliflozinSGLT1/2 inhibitionCHOL, LDL
AntihypertensiveLacidipineCa2+ channel blockerLDLYes [89]
FurosemideDiureticCHOL, LDL
IndapamideDiureticCHOL, LDL
PropranololBeta-blockerCHOL
AntimycoticFluconazoleErgosterol synthesis
inhibition
CHOL
VoriconazoleErgosterol synthesis
inhibition
CHOL
AntineoplasticApalutamideAntiandrogenCHOL, LDLPossible [90]Yes [90]Yes [90]
AnastrozoleAromatase
inhibition
CHOL
LetrozoleAromatase
inhibition
CHOL
MitotaneAdrenal cortex
inhibition
CHOL, LDLYes [32]Yes [32,91]Yes [92]
AsparaginaseDepletion of circulating asparagineCHOL
HistrelinGnRH agonistCHOL
DegarelixGnRH blockerCHOL
PegvisomantIGF1 inhibitionCHOL
RuxolitinibJAK inhibitionCHOLPossible ****
RucaparibPARP inhibitionCHOL
VerteporfinPhototherapy
sensitizer
CHOL
CladribinePurine analogueCHOL
TegafurPyrimidine
analogue
CHOL, LDL
PadeliporfinRadiation therapy sensitizerCHOL, LDL
BrigatinibTyrosine kinase
inhibition
CHOL, LDLPossible *Yes *
CabozantinibTyrosine kinase
inhibition
CHOL, LDL
DasatinibTyrosine kinase
inhibition
CHOL, LDLYes [89]
LenvatinibTyrosine kinase
inhibition
CHOL, LDL Yes
LorlatinibTyrosine kinase
inhibition
CHOL, LDLYes **Yes **,***
NilotinibTyrosine kinase
inhibition
CHOL, LDLYes [93]
PazopanibTyrosine kinase
inhibition
CHOL
Antipsychotic, atypicalAmisulprideInhibition of D2 and
5-HT2A receptors
CHOL
AripiprazoleInhibition of D2 and
5-HT2A receptors
LDL
CariprazineInhibition of D2 and
5-HT2A receptors
CHOL, LDL
ClozapineInhibition of D2 and
5-HT2A receptors
CHOL, LDL Yes [94]
OlanzapineInhibition of D2 and
5-HT2A receptors
CHOL, LDL
PaliperidoneInhibition of D2 and
5-HT2A receptors
CHOL, LDL
QuetiapineInhibition of D2 and
5-HT2A receptors
CHOL, LDLYes [95]Yes [95]
RisperidoneInhibition of D2 and
5-HT2A receptors
CHOL, LDL
Antipsychotic, typicalFluphenazineInhibition of D2
receptors
CHOL
ZuclopenthixolInhibition of D2
receptors
CHOL, LDL
AntiretroviralCobicistatCYP3A inhibitionCHOL
RaltegravirIntegrase inhibitionCHOL, LDL
EfavirenzNon-nucleoside
reverse transcriptase
inhibition
CHOL, LDLYes [50]Yes [50,96,97]Yes [97,98]
EtravirineNon-nucleoside
reverse transcriptase
inhibition
CHOL, LDLYes [99]Yes [100,101,102]Yes [100,101]
NevirapineNon-nucleoside
reverse transcriptase
inhibition
CHOL, LDL Yes [103,104,105]Yes [103,104,105]
RilpivirineNon-nucleoside
reverse transcriptase
inhibition
CHOL, LDLYes [99]
DarunavirProtease inhibitionCHOLYes [50]
FosamprenavirProtease inhibitionCHOLYes [106]Yes [106]Yes [106]
IndinavirProtease inhibitionCHOL
LopinavirProtease inhibitionCHOLYes [50]
RitonavirProtease inhibitionCHOLYes [107]Yes [108,109]Yes [108,109]
SaquinavirProtease inhibitionCHOL, LDLYes [107]
TipranavirProtease inhibitionCHOL Yes *****Yes *****
AntithyroidMethimazoleThyroperoxidase
inhibition
LDL
AntiviralBoceprevirProtease inhibitionCHOL
BarbituratePhenobarbitalGABA stimulationLDLYes [110]Yes [111]Yes [111]
Emergency contraceptionUlipristalProgesterone
receptor modulation
CHOL
ImmunosuppressantCyclosporinCalcineurin
inhibition
CHOL, LDLYes [112]
TacrolimusCalcineurin
inhibition
CHOLYes [113]
RituximabCD20 inhibitionCHOL, LDL
BeclomethasoneGlucocorticoid
receptor activation
CHOLYes [114]
DexamethasoneGlucocorticoid
receptor activation
CHOLYes [115]Yes [116,117,118,119]Yes [117,118,119]
PrednisoloneGlucocorticoid
receptor activation
CHOL Yes [120,121]Yes [120,121]
PrednisoneGlucocorticoid
receptor activation
CHOL Yes [116,122]Yes [122]
AnakinraIL-1 inhibitionCHOL
RilonaceptIL-1 inhibitionCHOL, LDL
BasiliximabIL-2 inhibitionCHOL, LDL
SarilumabIL-6 inhibitionCHOL, LDL
SiltuximabIL-6 inhibitionCHOL
TocilizumabIL-6 inhibitionCHOL, LDL
BaricitinibJAK inhibitionCHOL, LDL
TofacitinibJAK inhibitionCHOL, LDL
EverolimusmTOR inhibitionCHOL, LDL
SirolimusmTOR inhibitionCHOL
TemsirolimusmTOR inhibitionCHOL, LDL
LeflunomidePyrimidine
synthesis inhibition
CHOL, LDL
Mycophenolate mofetilPurine synthesis
inhibition
CHOL
AdalimumabTNF inhibitionCHOL, LDL
Certolizumab PegolTNF inhibitionCHOL, LDL
GolimumabTNF inhibitionCHOL, LDL
InfliximabTNF inhibitionCHOL, LDL
Non-steroidal anti-inflammatory drugAcetylsalisylic acidCOX inhibitionCHOL Yes [123]
DiclofenacCOX inhibitionCHOL
IbuprofenCOX inhibitionCHOL
OtherAtalurenRibosome function modulationCHOL
Human normal
immunoglobulin
Improved pathogen
removal
CHOL
LeuprorelinGnRH analogueCHOL, LDL
Proton pump inhibitorLansoprazoleStomach acid
reduction
CHOLYes [124]Yes [124]
PantoprazoleStomach acid
reduction
CHOL
AntioxidantIdebenoneMitochondrial
electron transport chain stimulation
CHOL
RetinoidAlitretinoinRetinoid receptor
activation
CHOL Yes [125]
BexaroteneRetinoid receptor
activation
CHOL, LDL Yes [126,127]Yes [126,127]
IsotretinoinRetinoid receptor
activation
CHOL Yes [125]
TretinoinRetinoid receptor
activation
CHOL Yes [125]
StimulantModafinilDopaminergic
modulation
CHOLYes [128]Yes [129]
VitaminCholecalciferolVitamin D receptor
activation
CHOL, LDL
* U.S. Food and Drug Administration Center for Drug Evaluation and Research (2016) Chemistry review on Brigatinib (application number 208772Orig1s000), available at https://www.accessdata.fda.gov/drugsatfda_docs/nda/2017/208772Orig1s000ChemR.pdf (accessed on 13 December 2021). ** Pfizer (2021) Highlights of LORBRENA (lorlatinib) prescribing information, available at https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/210868s004lbl.pdf (accessed on 13 December 2021). *** European Medicines Agency (2021) Summary of Lorviqua (lorlatinib) product characteristics, available at https://www.ema.europa.eu/en/documents/product-information/lorviqua-epar-product-information_en.pdf (accessed on 13 December 2021). **** U.S. Food and Drug Administration Center for Drug Evaluation and Research (2011) Genomics group review on ruxolitinib (application number 202192Orig1s000), available at https://www.accessdata.fda.gov/drugsatfda_docs/nda/2011/202192Orig1s000ClinPharmR.pdf (accessed on 13 December 2021). ***** U.S. Food and Drug Administration Center for Drug Evaluation and Research (2004) Pharmacology/toxicology review and evaluation (application number NDA 21-814), available at https://www.accessdata.fda.gov/drugsatfda_docs/nda/2005/21814_000_Aptivus_pharmr1.pdf (accessed on 13 December 2021).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Karpale, M.; Hukkanen, J.; Hakkola, J. Nuclear Receptor PXR in Drug-Induced Hypercholesterolemia. Cells 2022, 11, 313. https://doi.org/10.3390/cells11030313

AMA Style

Karpale M, Hukkanen J, Hakkola J. Nuclear Receptor PXR in Drug-Induced Hypercholesterolemia. Cells. 2022; 11(3):313. https://doi.org/10.3390/cells11030313

Chicago/Turabian Style

Karpale, Mikko, Janne Hukkanen, and Jukka Hakkola. 2022. "Nuclear Receptor PXR in Drug-Induced Hypercholesterolemia" Cells 11, no. 3: 313. https://doi.org/10.3390/cells11030313

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop