DOI 10.1515/dmdi-2013-0009
Drug Metab Drug Interact 2013; 28(2): 79–93
Review
Ferdinand Molnár, Jenni Küblbeck, Johanna Jyrkkärinne, Viktória Prantner and
Paavo Honkakoski*
An update on the constitutive androstane
receptor (CAR)
Abstract: The constitutive androstane receptor (CAR;
NR1I3) has emerged as one of the main drug- and xenobiotic-sensitive transcriptional regulators. It has a major
effect on the expression of several oxidative and conjugative enzymes and transporters, and hence, CAR can contribute to drug/drug interactions. Novel functions for CAR
are also emerging: it is able to modulate the metabolic fate
of glucose, lipids, and bile acids, and it is also involved in
cell-cell communication, regulation of the cell cycle, and
chemical carcinogenesis. Here, we will review the recent
information available on CAR and its target gene expression, its interactions with partner proteins and mechanisms of action, interindividual and species variation,
and current advances in CAR ligand selectivity and methods used in interrogation of its ligands.
Keywords: constitutive androstane receptor (CAR); CYP
expression; in vitro assays; ligand-binding domain; ligand
specificity; nuclear receptor.
*Corresponding author: Paavo Honkakoski, School of Pharmacy,
Faculty of Health Sciences and Biocenter Kuopio, University of
Eastern Finland, PO Box 1627, FI-70211 Kuopio, Finland,
Phone: +358 40 355 2490, E-mail:
[email protected]
Ferdinand Molnár, Jenni Küblbeck, Johanna Jyrkkärinne and
Viktória Prantner: School of Pharmacy, Faculty of Health Sciences
and Biocenter Kuopio, University of Eastern Finland, Kuopio, Finland
Introduction
During the past 15 years, the constitutive androstane receptor (CAR; NR1I3) has been established as a key drug- and
xenobiotic-sensitive regulator of oxidative and conjugative
enzymes and transporters important for drug metabolism,
disposition, and drug interactions. Searching the PubMed
database in January 2013 with the phrase “constitutive
androstane receptor OR nr1i3” yields over 860 publications, and a wealth of information on CAR and its sister,
the pregnane X receptor (PXR; NR1I2), has been compiled
in excellent reviews [1–24] listed in Table 1. We advise the
readers to consult these reviews for details, and we will
highlight only most relevant and recent findings here.
Brief history
CAR, PXR, and the vitamin D receptor (VDR; NR1I1) form
the nuclear receptor (NR) subfamily 1, group I. In mid1990s, human and mouse CAR were identified as constitutively active NRs potentially modulating retinoic acid signaling, but the actual target genes of CAR were unknown at
that time [25, 26]. Studies on phenobarbital (PB)-inducible
expression of rodent cytochrome P450 (CYP) 2B genes [27,
28] led to the identification of PB-responsive DNA elements
mediating the response to several classes of xenobiotics
[29] and of CAR as the key factor interacting with these elements [30]. A string of studies in the early 2000s showed
the following: CYP2B genes in CAR null mice were unresponsive to PB-type inducers; the formation of reactive
metabolites from liver toxins was drastically modulated;
and liver hypertrophy and tumor promotion linked with
PB exposure were absent [31–34]. Efforts during the past
decade have shown that diverse chemical classes such as
pesticides, fire retardants, environmental contaminants,
drugs, and industrial chemicals can activate mammalian
CAR receptors, albeit with species-specific effects [2, 4, 35].
These findings reinforce the role of CAR as a crucial sensor
for xenobiotics, and some insights into the molecular basis
of xenobiotic recognition have been made [22]. CAR is
also important for the endobiotic metabolism of steroids,
bile acids, vitamin D, thyroid hormone, and bilirubin [21],
and evidence shows [36, 37] a disruption of the cellular
homeostasis by the inappropriate activation of CAR due to
xenobiotic exposure. Experiments in the past 5 years have
revealed that CAR is actively controlling hepatic glucose
and lipid metabolism, with CAR agonism producing beneficial effects in animal models of obesity and insulin
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
80
Molnár et al.: Constitutive androstane receptor
Table 1 Selected review articles on CAR.
Focus area of the review
References
General reviews on CAR and its
function
Honkakoski et al., 2003 [1]
Stanley et al., 2006 [2]
Timsit and Negishi, 2007 [3]
di Masi et al., 2009 [4]
Reschly and Krasowski, 2006 [5]
Graham and Lake, 2008 [6]
Lamba et al., 2005 [7]
Lamba, 2008 [8]
Tirona and Kim, 2005 [9]
Zhou et al., 2005 [10]
Tolson and Wang, 2010 [11]
Staudinger et al., 2010 [12]
Higgins and Hayes, 2011 [13]
Chai et al., 2013 [14]
Swales and Negishi, 2004 [15]
Pascussi et al., 2008 [16]
Li and Wang, 2010 [17]
Chai et al., 2013 [14]
Moreau et al., 2008 [18]
Wada et al., 2009 [19]
Gao and Xie, 2012 [20]
Chai et al., 2013 [14]
Wagner et al., 2010 [21]
Evolution and species
differences
Human pharmacogenetics
Target genes in phase I and II
drug metabolism and transport
Cross talk and mechanisms of
action
Role in energy (glucose and
lipid) metabolism
Role in metabolism of bilirubin
and bile acids
CAR ligands, activators, and
associated in silico and in vitro
methodology
Hepatocarcinogenesis in
human and animal models
Poso and Honkakoski, 2006 [22]
Raucy and Lasker, 2010 [23]
Köhle et al., 2008 [24]
resistance [20]. The role of CAR in chemical carcinogenesis and hepatic proliferation in rodents is currently
under intense research [24, 38, 39], but its significance for
humans is uncertain. The discovery and subsequent characterization of PXR (as cited in a review by Chai et al. [14])
during the same time revealed that both receptors have a
crucial role in regulation of drug metabolism and disposition. However, the elucidation of CAR- and PXR-mediated signaling is very complex due to overlapping CAR
and PXR ligand specificities and target gene profiles and
to the intricate cross talk with other transcription factors
(TFs) such as hepatocyte nuclear factor (HNF) 4α, cAMP
response element-binding protein, and the family of forkhead box (Fox) proteins [14, 40]. An additional complexity arises from the fact that CAR appears to be activated by
some CYP inducers such as PB indirectly via a cytoplasmic
dephosphorylation-dependent mechanism, culminating
in nuclear translocation of CAR [15]. Exciting results on
the physiological functions of CAR are expected because
knowledge of CAR properties and its connections with
other cellular processes is being accumulated.
Structural features of the NR CAR
Crystal structures of agonist-bound CAR
Three crystal structures of mouse or human CAR agonistbound ligand-binding domains (LBDs) (Table 2) conform
to the standard three-layer sandwich architecture seen in
other NRs [44]. The CAR LBDs contain 11 α-helices and 3
short β-strands, and helices 2 and 2′ assume the 310 conformation [41, 42] (Figure 1A). The unique structural features for CAR LBD include an additional helix called “X”
between helices 11 and 12 and an unusually short helix 12
(Figure 1A). The helix X is also present in VDR [45], retinoid-related orphan receptor (ROR) β [46], and RORα [47],
but the linker between helices X and 12 appears to be more
rigid in constitutively active RORs and CAR. The short
helix 12 is stabilized by interactions with a lysine residue
in helix 4 (K195 in human CAR) and intrahelical H-bonds
[42] (Figure 1B), contributing in part to the constitutive
activity. The two short 310 helices 2 and 2′ appear to form
a ligand entry point as postulated for the peroxisome
proliferator-activated receptor (PPAR) α [43, 48]. Similarly to other NRs, the CAR ligand-binding pocket (LBP)
is made up by about 30 residues in helices 2–7 and 10 and
in β-sheets 3 and 4 that form a mostly apolar lining of the
Table 2 The human and mouse CAR LBD crystal structures.
PDB ID
Protein
molecules
Co-crystallized
ligands
Co-regulator
peptide
1XVP
hCAR
hRXRα
hCAR
hRXRα
mCAR
hRXRα
mCAR
CITCO
Pentadecanoic acid
5β-Pregnanedione
C16–C18 fatty acids
TCPOBOP
9-cis-Retinoic acid
Androsten-3α-ol
1XV9
1XLS
1XNX
Resolution, Å
Completeness, %
References
SRC1
40.0–2.60
86.3
Xu et al., 2004 [41]
SRC1
40.0–2.70
86.4
Xu et al., 2004 [41]
TIF2
20.0–2.95
93.2
Suino et al., 2004 [42]
None
30.0–2.90
99.8
Shan et al., 2004 [43]
CITCO, 6-(4-chlorophenyl)imidazo[2,1-b][1,3]thiazole-5-carbaldehyde O-(3,4 dichlorobenzyl)oxime; TCPOBOP, 1,4-bis-[(3,5-dichloropyridyl)
oxy]benzene; SRC1, steroid receptor co-activator 1; TIF2, transcriptional intermediary factor 2.
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
Molnár et al.: Constitutive androstane receptor
81
Figure 1 The crystal structure of the human CAR complexed to CITCO.
(A) Overall view on the whole ligand-protein complex with highlighted helices discussed in the text. (B) Detailed view on the LBP with
some of the residues displayed that are discussed in the text. The interaction of the K195 with the terminal part of the H12 is schematically
depicted with green dashed line. The important features are illustrated in color. The co-activator peptide bearing the LXXLL motif derived
from steroid receptor co-activator 1 (SRC1/NCOA1) is in orange, helix 12 (H12) is in red, and helix X (HX) is in brown.
pocket, although two hydrophilic patches may allow the
formation of hydrogen bonds with the ligands. The LBP
volumes of CAR range from 525 to 675 Å3, placing them
in size between the classical steroid receptors and PXR.
Although the co-crystallized ligands are structurally different, they use the hydrophobic character of the cavities and
hydrogen bonds that are formed toward the polar residues
to orient the ligand. In mouse CAR, none of the ligands
makes a direct hydrogen bond contact with helix 12, but
1,4-bis-[2-(3,5-dichloropyridyloxy)]benzene
(TCPOBOP)
forms a number of hydrophobic interactions with helix
12 (L353) and the linker helix (L346, T350). Because these
interactions contribute toward the stabilization of helix
12, they may be responsible for the “superagonistic”
properties of this ligand. In human CAR, co-crystallized
ligands do not form direct contacts with helix 12. The
closest residue is L343, which is positioned at a distance
of 4.9 Å from the C21 of 5β-pregnane-3,20-dione and 3.9 Å
from the thiazole ring of 6-(4-chlorophenyl)imidazo[2,1-b]
[1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime
(CITCO). The barrier formed by residues of F161, N165,
F234, and Y326 excludes the possibility of a direct interaction between the ligand and helix 12 [41, 42, 49]
(Figure 1B). The structure of mouse CAR co-crystallized
with the inverse agonist androstenol indicates a structural
change in helices 10 and 11, which resembles the inactive
apo forms of NRs [43]. However, the lack of corepressor
peptide in this structure and the unavailability of the
ligand-free CAR crystals preclude further speculation on
the mechanisms of (inverse) agonism.
Molecular modeling studies
Before the crystal structures for CAR LBD became available in 2004, structural features were analyzed by creating
homology models [22]. The selection of the template had a
substantial effect on the modeled LBP volume and residue
orientation, as exemplified by the early excessively large
LBP volume estimates [50] and the relatively accurate prediction of the LBP performed later [49]. Prediction of the
protein flexibility is based on molecular dynamics (MD)
simulations [51]. Such studies have yielded information
on the basis of constitutive and agonist-induced activity of
CAR [51–53], and recently, on the probable mechanism of
inverse agonist-induced binding of NR corepressors [54].
Due to the limited number of agonist-bound CAR crystal
structures, information on binding of novel agonists must
be acquired by docking studies and supported by, e.g., sitedirected mutagenesis. The advances in speed and incorporation of protein flexibility in docking programs have
enabled more detailed analysis of ligand binding [54–57].
Due to the promiscuity for diverse ligands and the inherent flexibility of CAR, the building of pharmacophore/
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
82
Molnár et al.: Constitutive androstane receptor
quantitative structure-activity relationship models remains
problematic [22]. For a limited set of structurally similar
ligands, the pharmacophore alignment has been possible
[58–60], but for dissimilar ligands, an alignment based on
docking is almost a necessity [61–63].
Interspecies and interindividual
differences
Evolution and species differences
Invertebrates have a single protein orthologous to NR1I
genes that does not seem to respond to known xenobiotics [64]. In birds, the sole xenosensor appears to share
both CAR and PXR sequence similarity and ligand-binding properties [65], and similarly, fish and Caenorhabditis
elegans possess a single NR1I gene [66, 67]. The previous
notion that CAR evolved through gene duplication of a
single CAR/PXR ancestral gene has been challenged by a
new view that all NR1I genes result from whole genome
duplication [68]. This theory is supported by recent analysis showing that PXR/CAR duplication took place after the
split of tunicates and vertebrates but before that of fish
and land vertebrates [69]. In contrast to PXR, CAR genes
are not found in the fish lineages but are conserved in
all land vertebrates, including amphibians. Functionally, mammalians use both PXR and CAR as xenosensors,
whereas in nonmammalian land vertebrates, CAR may be
the predominant xenosensing receptor [69].
The sequence comparisons among NR1I members indicate that both CAR and PXR genes have been under positive
selection [70], presumably due to exposure to different dietderived xenobiotics. This divergent evolution may explain
the wide species differences in CYP induction and/or CAR
activation profiles, even though the basic mechanism of
receptor activation is well conserved. The sequence similarity between the mouse and human CAR LBDs is only 72%,
in contrast to more than 90% similarity in steroid hormone
receptors [5, 71]. Changes in the LBD residues contribute
to the different sizes, contours, and contact points with
the ligands between the mouse and human CAR LBPs. For
examples, residues F171, N175, F244, and Y336 forming the
“barrier” in mouse CAR do not appear to restrict the ligand
projecting toward helix 12 as much as the corresponding residues in human CAR do, enabling a direct contact
between the mouse-specific agonist TCPOBOP with helix
12 [41–43]. Second, mutagenesis studies have identified
key residues that dictate the species-specific response to
17α-ethinylestradiol, an inverse agonist for human CAR and
a partial agonist for mouse CAR (F243) and for TCPOBOP
(M340). Third, species differences exist in residues at positions critical for human CAR function [49]. However, the
role of these amino acid differences and extent for speciesspecific ligand-dependent activation remains enigmatic
because CAR has not been cloned and/or systematically
characterized from many other species relevant for drug
development such as the rat or the dog [72, 73].
Genetic variation in the human CAR
Exons 2 and 3 and part of exon 4 encode the DNA-binding
domain (DBD) and the hinge regions, whereas the LBD is
encoded by the rest of exon 4 and exons 5–9. Alternative
splicing has been shown to produce at least 26 splicing
variants, many of which contain a premature stop codon or
code for a variant protein [74, 75] and thus heavily influence
expression of functional CAR [76]. The most important isoforms are termed CAR1 (wild type), CAR2 (insertion of SPTV,
near LBP), and CAR3 (insertion of APYLT in the LBD/heterodimerization region) [7, 77, 78]. Although CAR1 has a high
basal activity, splice variants CAR2 and CAR3 display low
constitutive activity. Due to the changes in the LBD structures, it is not surprising that some differences in ligand
activation have been reported between the wild-type and
CAR2/CAR3 isoforms [60, 79]. Of note, similar splice variants are not present in experimental animals. At least 30
single-nucleotide polymorphisms (SNPs) have been identified [8, 80], albeit at a low frequency (<2%) in major populations. All five known nonsynonymous SNPs are located in
the LBD, and two of them disrupt CAR function: H246A was
inactive, whereas L380P had a reduced basal but normal
CITCO-elicited CYP3A4 reporter activity [81]. There is some
recent evidence of CAR polymorphisms being associated
with exposure to efavirenz, a selective substrate for human
CYP2B6 [82, 83]. However, the effects of more frequent
polymorphism in the CAR targets such as CYP2B6 [84] may
mask the relevance of CAR polymorphisms.
Regulation of CAR levels and
activity
CAR expression
The CAR gene is expressed in tissues with high capacity
for drug metabolism such as liver and intestine derived
from the endoderm. The key regulator in such cells is
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
Molnár et al.: Constitutive androstane receptor
the HNF4α, which recognizes a conserved element in the
proximal CAR gene promoter [85, 86]. Different isoforms
of HNF4α appear to either activate (isoform 1) or suppress (isoform 7) the expression of CAR in a co-activatordependent manner [86]. The integration of CAR to many
physiological processes controlled by other NRs gains
support from the findings that CAR expression and/or CYP
inducibility is increased by the glucocorticoid receptor [87]
and the retinoic acid receptor [88]. CAR expression is also
activated by PXR agonists (e.g., PCN, dexamethasone [87,
89]), potentially by peroxisome proliferators (e.g., fibrates
[88]) and is dependent on thyroid hormones [90]. The discovery of serum response elements in the CAR promoter
[91] provides a link to stress-activated protein kinase
pathways via the binding of the ETS domain-containing
protein Elk-1. This finding may explain why many growth
factors and the presence of serum inhibit PB-inducible
CYP expression in several experimental settings [92] and
why the dephosphorylation of CAR is associated with
its nuclear translocation [93]. Finally, CAR is under the
control of the circadian clock-related PAR-domain basic
leucine zipper TFs such as albumin gene D-site-binding
protein, thyrotroph embryonic factor, and hepatic leukemia factor [94].
Cytoplasmic CAR interactions
Groundbreaking work from the Negishi Laboratory
showed that CAR is complexed with heat shock protein
90 and a retaining CCRP protein in the liver cytoplasm
in unexposed animals [95] and that PB exposure leads to
nuclear translocation of CAR and to target gene activation.
The translocation process is influenced by phosphorylation status, with phosphorylation by extracellular signalregulated kinase 1/2 and protein kinase C affecting the
DBD (T38 in CAR) and retaining inactive CAR in the cytoplasm [93, 96], whereas dephosphorylation by a protein
phosphatase 1β (PP1β) and protein phosphatase 2A
(PP2A) [97] enhances nuclear translocation of active CAR
[98]. Also, AMP-activated protein kinase (AMPK) has been
shown to be involved in the induction of CYPs by PB [99].
Although CAR itself is not phosphorylated by AMPK, this
kinase seems to affect p300 and PPARγ co-activator (PGC)
1α, suggesting a possible mechanism for the observed liver
kinase B1/AMPK cascade activation by indirect inducers,
such as PB. These interactions are important as they link
CAR activation to other signal pathways activated by, e.g.,
stress and cell proliferation pathways. Indeed, cell cycle
proteins have been identified as CAR targets [100, 101].
CAR is required for chemically induced liver growth [31]
83
and signaling via phosphorylation has long been known
to affect CYP inducibility [102, 103].
PPP1R16A, the membrane subunit of PP1β, facilitates
the ligand-independent translocation of CAR into the
nucleus, indicating a novel mechanism for the translocation of NRs in which ligands and other receptors are not
involved [98]. However, the translocation effect is more
enhanced in the presence of PB. Given the fact that exposure to PB decreases hepatic cell-cell communication by
affecting the activity and levels of connexins [104, 105], it
is likely that novel cytoplasmic interactions of CAR remain
to be identified.
Interactions of CAR with DNA and
nuclear partner proteins
Specificity of DNA binding
Many of the CAR target genes have been listed in earlier
reviews (Table 1). They include the established genes of
enzymes of phase I and II biotransformation, uptake, and
efflux transporters (Table 3), but new targets continue
to emerge in genes responsible for endobiotic metabolism and cell cycle control [9–13]. The initially identified
binding site for CAR/RXR heterodimer was a direct repeat
5 (DR5; two AGGTCA-related hexamers separated by five
nucleotides) in retinoic acid-sensitive gene promoters
[25]. Later studies indicated that most efficacious PBresponsive enhancers consist of clusters of DR4 elements
in vicinity of other TF-binding sites in, e.g., CYP2B and
UGT1A1 genes [30, 106]. In addition, CAR is also able to
transactivate and/or bind the PXR-responsive DR3 and
everted repeat (ER) 6 elements present in the proximal
and distal regions of the CYP3A genes [107, 108] as well as
the PPAR-responsive DR1 elements [109–111]. Experiments
with in vitro-translated proteins have indicated that the
CAR/RXR heterodimer prefers DR4 over DR5, whereas
ER6–ER9 elements are recognized and DR1/3 show little
binding [29, 112]. CAR has been shown to bind DNA as a
monomer in human UGT1A1 and MDR1 promoter elements
and to be activated by ligands, which may point to a physiological role also for CAR monomers [112, 113]. Intriguingly, two nucleotides at the 5′ flank of each hexamer
motif appear to influence the binding of CAR/RXR or CAR
monomer by up to 20-fold [112].
The lack of high-quality antibodies for CAR has
precluded the assessment of true in vivo binding sites
by chromatin immunoprecipitation, and selection of
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
84
Molnár et al.: Constitutive androstane receptor
Table 3 CAR target genes.
Target gene
Phase I
Aldehyde dehydrogenases
Cytochrome P450s
P450 (cytochrome) oxidoreductase
Phase II
Glutathione S-transferases
Sulfotransferases
UDP-glucuronosyltransferases
Phase III
ATP-binding cassette family
Solute carrier transporters
Gene symbola
Species
Aldh1a1, 1a7
CYP2A6, 2B6, 2B10, 2C9, 2C19, 3A4, 3A11
Cyp1a1, 2a4, 2b10, 3a11
Cyp2b1, 2b2, 2c6, 2c7, 3a1
Por
M. musculus
H. sapiens
M. musculus
R. norvegicus
M. musculus
Gsta1, a2, a3, m1, m2
Gsta1, a2, a3, m1
Sult1a1, 2a1, 2a9
UGT1A1
Ugt1a1
Ugt1b2
M. musculus
R. norvegicus
M. musculus
H. sapiens
M. musculus
R. norvegicus
ABCB1, C2, C3
Abcb1a, c1, c2, c3, c4
Abcc2
Slco2a1
H. sapiens
M. musculus
R. norvegicus
M. musculus
R. norvegicus
Data were compiled from di Masi et al. [4] and Tirona and Kim [9]. M. musculus, Mus musculus; H. sapiens, Homo sapiens; R. norvegicus,
Rattus norvegicus. aApproved by the HUGO Gene Nomenclature Committee (http://www.genenames.org/).
random DNA sites by either in vitro amplification or yeast
genetics to identify CAR-binding sites has not been performed. This suggests that we do not yet have a full view
of DNA-binding specificity by CAR, whereas gene expression studies (with selective NR ligands, delivery of siRNA
or knockout animals) cannot distinguish between direct
DNA binding-mediated gene activation from responses
that are either secondary or mediated by protein/protein
interactions.
Interactions with the NR co-regulators
The interaction partners of CAR are summarized in Table
4. Most NR co-activators (CoAs) contain one or more NR
interaction boxes, bearing a short peptide motif LXXLL,
where L is leucine and X is any amino acid [114]. The
anchoring salt bridges at the ends of this motif help
orient it properly in the surface groove on the NR LBD,
whereas the leucines provide numerous van der Waals
interactions with the hydrophobic residues located
in LBD helices 3, 4–5, and 12 [42]. The only structural
information for CAR/CoA interactions is derived from
the steroid receptor co-activator (SRC) family members
NCOA1 (SRC1) and NCOA2 [transcriptional intermediary
factor 2 (TIF2)] [41, 42 ]. Many CoAs share characteristic
enzymatic activities such as histone acetyltransferase
activity, which targets histones or other proteins at
NR-regulated gene promoters for acetylation, which can
enhance the transcriptional activity [115].
CAR has been shown to physically interact with all
three members of the SRC family co-activators SRC1 [41,
49], TIF2 [34], and NCOA3 (receptor-associated cofactor 3, RAC3) [116] in vitro. Studies in cellular models
indicate that all three co-activators are redundant with
regard to enhancing CAR-mediated induction of CYP
genes. However, only NCOA3 is able to enhance CAR
transactivation in hepatic cells [38, 117]. Although CAR
interacts with another NR co-activator NCOA6 [118], its
liver-specific deletion does not interfere with the regulation of CAR target genes [119]. However, similar tissue-specific disruption of mediator of RNA polymerase
II transcription subunit 1 (MED1) resulted in the near
abrogation of TCPOBOP-activated gene expression and
acetaminophen-induced hepatotoxicity [120]. It has also
been shown that MED1 but not NCOA6 is required for
nuclear translocation of CAR in mouse liver [121]. The
critical effect of MED1 on CAR-mediated signaling could
be anticipated from the fact that MED1 is a key component of the mediator complex, which essential for transcriptional activation via a variety of TFs [122 ].
The PPARα-interacting cofactor (PRIC) complex component, PRIC320, associates with CAR in ligand-independent and ligand-dependent manner in vitro [123], but the
physiological consequences of this interaction have not
been explored further. The discovery of interaction between
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
Molnár et al.: Constitutive androstane receptor
85
Table 4 List of CAR-interacting proteins.
CAR-interacting protein
Group/function
Full name
Gene symbola
Steroid receptor co-activator 1 (SRC1)
Transcriptional intermediary factor 2 (TIF2)
Receptor-associated co-activator 3 (RAC3)
Activating signal co-integrator 2 (ASC2)
PPAR-binding protein (PBP)
PPARα-interacting cofactor 320 (PRIC320)
PPARγ co-activator 1α (PGC-1α)
Forkhead box O1 (FoxO1)
Growth arrest and DNA damage-inducible 45β (Gadd45β)
Protein phosphatase 1 regulatory subunit 16A (PPP1R16A)
Splicing factor 3a, subunit 3, 60 kDa (SF3a)
Nuclear receptor corepressor (NCoR)
Silencing mediator for retinoid or thyroid hormone receptors (SMRT)
NCOA1
NCOA2
NCOA3
NCOA6
MED1
CHD9
PPARGC1A
FOXO1
GADD45B
PPP1R16A
SF3A3
NCOR1
NCOR2
p160 Co-activator
p160 Co-activator
p160 Co-activator
General NR co-activator
Mediator TRIP/TRAP co-activator
General transcription machinery-interacting protein
General NR co-activator
Metabolic transcriptional factor
Cell cycle-regulating factor
Regulator of signal transduction
Splicing/inhibitor of CAR signaling
General NR corepressor
General NR corepressor
For references, see the section Interactions of CAR with other nuclear proteins. aApproved by the HUGO Gene Nomenclature Committee
(http://www.genenames.org/).
CAR and PGC-1α again highlights the connections among
energy metabolism and detoxification [124]. Later in vivo
studies using knockout animals demonstrated that fasting
upregulates CAR expression and ligand-independent CAR
activity that involves the interaction with PGC-1α [85].
The interaction of CAR with prototypic NR corepressors NCoR (NCOR1) and silencing mediator of
retinoid or thyroid hormone receptors (SMRT; NCOR2)
in vitro explains the mechanism of inverse agonist suppression of CAR activity [57, 125, 126]. Ex vivo, an association of CAR with SMRT on CYP24A1 gene promoter
has been reported, thus mediating cross talk with VDR
signaling [127].
Interactions of CAR with other nuclear
proteins
Additional interaction partners of CAR are listed in
Table 4. In analogy to most NRs, CAR makes heterodimers
with retinoid X receptor (RXR) isotypes [30, 112]; thus, the
lack of RXRα reduces expression of CAR target genes. In
addition to this natural partner, CAR has been reported
to interact with by small heterodimer partner (SHP,
NR0B1) and NR0B2, resulting in the suppression of CAR
activity and target gene expression [128, 129]. SMRT and
NCoR can inhibit CAR-mediated signaling independent of
SHP, demonstrating that they may bind to distinct sites
[128]. The recently identified SHP-interacting leucine
zipper protein (SMILE) [130] is able to interact with CAR,
competing with co-activators TIF2 and PGC-1α in vitro
and in vivo [131].
In vitro and cell-based assays have shown that
CAR interacts directly with FoxO1 and represses FoxO1mediated transcription of the insulin-responsive phosphoenolpyruvate carboxykinase 1 (PEPCK1) and glucose
6-phosphatase (G6Pase) promoters [32, 40]. These findings provide a mechanistic basis to following observations: long-term treatment with PB is known to decrease
plasma glucose levels, improve insulin sensitivity in
diabetic patients [132 ], and repress rodent PEPCK1
and G6Pase [32, 133]. In lipogenesis, CAR counters the
effect of PXR by suppressing lipogenic genes such as
sterol regulatory element-binding protein 1C and fatty
acid synthase [134]. Therefore, CAR is able to modulate
glucose and lipid metabolism, and its activators may be
potential candidate drugs for hepatobiliary and metabolic diseases.
Gadd45β is a growth arrest- and DNA damageinducible protein that interacts with CAR in a liganddependent way and enhances liver growth in mice. The
administration of TCPOBOP in mice results in druginduced hyperplasia, which is associated with dramatic
and rapid hepatocyte growth [135]. Although the proliferation seems to be intact in Gadd45b null mice, the
hepatic growth is delayed and the early transcriptional
stimulation of CAR target genes is weaker [39]. Another
CAR partner, a component of the splicing factor 3a, has
been identified via yeast two-hybrid screening and confirmed in other interaction assays [136].
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
86
Molnár et al.: Constitutive androstane receptor
CAR ligands and associated
methods
Variability of CAR ligands
Only few selective CAR agonists and inverse agonists are
currently known because many reported ligands have
turned out to modulate other NRs or TFs, hampering
their use as tools to interrogate CAR biology. Examples of
this low selectivity include many drugs, pesticides, and
polychlorinated biphenyls (CAR and PXR), phthalates
(CAR and PPAR), estrogens (CAR and estrogen receptor, albeit at different affinities), and oltipraz (CAR and
nuclear factor erythroid 2-related factor).
Meanwhile, the list of CAR-activating chemicals is
rapidly expanding (Table 5), including steroids [144],
natural compounds [145], pesticides [139], industrial
chemicals [146], drugs [62, 63], and various synthetic compounds including thiazolidin-4-ones, sulfoamides [59],
and flexible diaryl compounds [54, 56]. The activity of CAR
is also thought to be modulated by the so-called indirect
activators (acetaminophen, bilirubin, 6,7-dimethylsculetin, PB, and phenytoin) that stimulate the nuclear translocation of CAR and the expression of its target genes but
without binding directly to the LBD [15, 147]. However, at
least for phenytoin and PB, this view has been challenged
because assays with natural CAR or its variants have
shown increased reporter activity by these compounds
[56, 79, 148–150].
The inverse agonists bind the CAR LBD and cause
a reduction in CAR transcriptional activity due to the
recruitment of corepressors. These include different steroids, the isoquinoline carboxamide PK11195 [143] and the
novel compound 1-[(2-methylbenzofuran-3-yl)methyl]3-(thiophen-2-ylmethyl) urea (S07662) [54, 57]. In some
cases, reports on ligand binding and ligand-elicited CAR
activation are controversial such as for clotrimazole [58,
138] and meclizine [57, 140]. This might be due to different cell lines with variable co-regulator contents used in
the studies. The activation of CAR can be also decreased
or increased by retinoid-like substances, but the mechanisms remain unknown [151, 152].
Assays to discover novel ligands
One significant reason behind this expansion of CAR
ligands has been the development of assays for the
measurement of ligand-dependent CAR activation and/
or interaction. Most commonly, various reporter gene
assays measure the activation of human CAR and thus
indirectly assess CAR/ligand interaction [56, 57, 59, 61,
141, 142, 145, 153]. Naturally occurring splice variants
(CAR3) or mutated CAR LBDs are suggested to improve
the assay sensitivity due to the lower basal activity of the
modified receptor [73, 79, 146, 154]. Another approach to
lower the basal activity has been the addition of a CAR
inverse agonist [55, 60, 108]. Recently, a careful selection
of the cell line used for transfection has made it possible
to use the wild-type human CAR without any modification to the LBD structure or the addition of any inverse
agonists [56, 57]. The mammalian two-hybrid assay
measures the ligand-dependent interaction of CAR with
a selected co-regulator peptide. This assay appears to
be more sensitive in identifying weak or partial agonists
that may elicit both co-activator and corepressor recruitment and very useful in dissecting the co-regulator
profile of human CAR [57] and to gain support for human
CAR/ligand interactions [35, 63].
Similar CAR/co-regulator assays, which resulted in
the identification of the potent agonist CITCO, can be
designed for in vitro screening [138]. An LBD assembly assay, originally described by Pissios et al. [155] for
mouse CAR, is also useful in identifying novel human
CAR ligands [62, 156, 157]. More recently, surface plasmon
resonance has been utilized in the identification of novel
ligands and species-specificity studies on human CAR
[62, 137]. Here, a solution with CAR LBD protein and
increasing concentrations of agonist is flushed over the
surface bound by a co-activator peptide, and the resulting optical change of the surface is then monitored.
Because the detection measures any binding reactions
taking place on the surface, it must be carefully controlled for and verified for dependency on the CAR LBD
using, e.g., a mutated CAR.
Because the CAR resides in the hepatocyte cytoplasm in the absence of its activators, the reporter gene
measurements have sometimes been complemented
with nuclear translocation assays. This requires the
transfection of primary hepatocytes with, e.g., constructs encoding yellow fluorescent protein-tagged CAR.
The translocation of CAR into the nucleus in response
to compound exposure can be monitored by confocal
microscopy [23, 158].
The direct assessment of CAR/ligand interactions
in biochemical assays in vitro has lagged behind the
reporter assays. There is limited evidence that the presence of an agonist increases DNA binding by human
CAR/RXR heterodimers [108]. Both agonists and inverse
agonists provide increased protection for human CAR
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
Molnár et al.: Constitutive androstane receptor
87
Table 5 Latest additions to human CAR ligands and/or activators.
Compounds
Steroids
Androstan-3α-ol and androsten-3α-ol
3,17β-Estradiol and 17α-ethinylestradiol
5β-Pregnanedione
Pesticides
Pyrethroids (e.g., permethrin, cypermethrin)
Carbamates (e.g., benfuracarb)
Organochlorines (e.g., methoxychlor, PCB153, o,p′-DDT)
Drugs
Clotrimazole
Meclizine
Artemisinin and some derivatives
Carbamazepine
Nevirapine
Phenytoin
Natural polyphenols
Food-derived flavonoids (e.g., chrysin)
Alcohol-derived flavonoids (e.g., ellagic acid)
Plasticizers
Triaryl phosphates
Di(2-ethylhexyl)phthalate
Synthetic compounds
CITCO
Flexible diaryl compounds (FL81)
Thiazolidin-4-ones
Sulfonamides
A series of chemotypes
PK11195
S07662
Effect on human CAR
References
IA (h>m)
IA (h), A (m)
A (h), IA (m)
Dau et al., 2013 [137]
Dau et al., 2013 [137]
Maglich et al., 2003 [138]
A
A
A
Küblbeck et al., 2011 [56]
Abass et al., 2012 [139]
Küblbeck et al., 2011 [56]
IA or A
IA or inactive
A
A
A
Activator or A
Jyrkkärinne et al., 2008 [61]
Lynch et al. 2012 [63]
Huang et al., 2004 [140]
Burk et al., 2012 [62]
Faucette et al., 2007 [79]
Faucette et al., 2007 [79]
Küblbeck et al., 2011 [56]
A
A
Yao et al., 2011 [141]
Yao et al., 2011 [141]
A
A for hCAR2
Jyrkkärinne et al., 2008 [61]
DeKeyser et al., 2009 [142]
A
A
A
A
A
IA
Maglich et al., 2003 [138]
Küblbeck et al., 2011 [56]
Küblbeck et al., 2008 [59]
Küblbeck et al., 2008 [59]
Li et al., 2008 [143]
Küblbeck et al., 2011 [57]
Lynch et al., 2012 [63]
Küblbeck et al., 2011 [57]
IA
A, agonist; IA, inverse agonist; activator, indirect activation, no evidence of direct binding; h, human CAR; m, mouse CAR.
LBD against proteolytic digestion [56, 57]. Displacement
of labeled clotrimazole from the CAR LBD by test compounds [159] has the disadvantage that it cannot distinguish between agonists and inverse agonists. Due to the
high basal activity and complex activation mechanisms
of CAR as well as rather tedious protocols and/or technical issues, these assays are only low-throughput and/or
prone to false positives [17].
Future directions
To elucidate the diverse biological functions of human
CAR in more detail, we must first develop more potent
and selective CAR agonists and inverse agonists. Nevertheless, the combination of molecular modeling
and biological assays [57, 59, 63] has proven a very
fruitful approach in raising the range and diversity of
CAR ligands. It is expected that advances in structural
biology, such as the determination of ligand-free and
corepressor-bound CAR LBD structures, and in comparative molecular modeling will resolve the frequent
problem of PXR activation by many of the currently available CAR ligands. Second, the identification of liganddependent CAR/co-regulator and cytoplasmic interactions constitutes an important avenue in deciphering
the mechanisms of CAR activation and in helping the
identification of novel, primary CAR target genes. This
in turn should highlight the role of CAR in processes
of liver growth, cell-cell communication, intermediate
metabolism, and in discovering new absorption, distribution, metabolism, and excretion (ADME)-related
CAR targets in addition to CYP2B6. Finally, the development of comprehensive assays for reliable screening of
CAR activation will help in the prediction of its in vivo
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
88
Molnár et al.: Constitutive androstane receptor
relevance, in the study of its ramifications in ADME and
drug safety research.
Acknowledgments: We acknowledge the financial support
of the Academy of Finland, National Agency for Technology and Innovation, FinPharma Doctoral Program, Ministry of Agriculture and Forestry, and the Finnish Cultural
Foundation in our earlier and current research. We apologize to colleagues for omission of citations of their contributions to this research field due to space limitations.
Conflict of interest statement
Authors’ conflict of interest disclosure: The authors stated
that there are no conflicts of interest regarding the publication of this article.
Research funding: None declared.
Employment or leadership: None declared.
Honorarium: None declared.
Received February 1, 2013; accepted April 17, 2013; previously published online May 13, 2013
References
1. Honkakoski P, Sueyoshi T, Negishi M. Drug-activated nuclear
receptors CAR and PXR. Ann Med 2003;35:172–82.
2. Stanley LA, Horsburgh BC, Ross J, Scheer N, Wolf CR. PXR
and CAR: nuclear receptors which play a pivotal role in drug
disposition and chemical toxicity. Drug Metab Rev 2006;38:
515–97.
3. Timsit YE, Negishi M. CAR and PXR: the xenobiotic-sensing
receptors. Steroids 2007;72:231–46.
4. di Masi A, De Marinis E, Ascenzi P, Marino M. Nuclear receptors
CAR and PXR: molecular, functional, and biomedical aspects.
Mol Asp Med 2009;30:297–343.
5. Reschly EJ, Krasowski MD. Evolution and function of the NR1I
nuclear hormone receptor subfamily (VDR, PXR, and CAR)
with respect to metabolism of xenobiotics and endogenous
compounds. Curr Drug Metab 2006;7:349–65.
6. Graham MJ, Lake BG. Induction of drug metabolism:
species differences and toxicological relevance. Toxicology
2008;254:184–91.
7. Lamba J, Lamba V, Schuetz E. Genetic variants of PXR (NR1I2)
and CAR (NR1I3) and their implications in drug metabolism and
pharmacogenetics. Curr Drug Metab 2005;6:369–83.
8. Lamba JK. Pharmacogenetics of the constitutive androstane
receptor. Pharmacogenomics 2008;9:71–83.
9. Tirona RG, Kim RB. Nuclear receptors and drug disposition gene
regulation. J Pharmacol Sci 2005;94:1169–86.
10. Zhou J, Zhang J, Xie W. Xenobiotic nuclear receptor-mediated
regulation of UDP-glucuronosyl-transferases. Curr Drug Metab
2005;6:289–98.
11. Tolson AH, Wang H. Regulation of drug-metabolizing enzymes
by xenobiotic receptors: PXR and CAR. Adv Drug Deliv Rev
2010;62:1238–49.
12. Staudinger JL, Xu C, Cui YJ, Klaassen CD. Nuclear receptormediated regulation of carboxylesterase expression and
activity. Expert Opin Drug Metab 2010;6:261–71.
13. Higgins LG, Hayes JD. Mechanisms of induction of cytosolic
and microsomal glutathione transferase (GST) genes by
xenobiotics and pro-inflammatory agents. Drug Metab Rev
2011;43:92–137.
14. Chai X, Zeng S, Xie W. Nuclear receptors PXR and CAR:
implications for drug metabolism regulation, pharmacogenomics and beyond. Expert Opin Drug Metab 2013;9:253–66.
15. Swales K, Negishi M. CAR, driving into the future. Mol
Endocrinol 2004;18:1589–98.
16. Pascussi J-M, Gerbal-Chaloin S, Duret C, Daujat-Chavanieu M,
Vilarem M-J, Maurel P. The tangle of nuclear receptors that
controls xenobiotic metabolism and transport: crosstalk and
consequences. Annu Rev Pharmacol Toxicol 2008;48:1–32.
17. Li H, Wang H. Activation of xenobiotic receptors: driving into the
nucleus. Expert Opin Drug Metab 2010;6:409–26.
18. Moreau A, Vilarem MJ, Maurel P, Pascussi JM. Xenoreceptors
CAR and PXR activation and consequences on lipid metabolism,
glucose homeostasis, and inflammatory response. Mol Pharmaceutics 2008;5:35–41.
19. Wada T, Gao J, Xie W. PXR and CAR in energy metabolism. Trends
Endocr Met 2009;20:273–9.
20. Gao J, Xie W. Targeting xenobiotic receptors PXR and CAR for
metabolic diseases. Trends Pharmacol Sci 2012;33:552–8.
21. Wagner M, Zollner G, Trauner M. Nuclear receptor regulation of
the adaptive response of bile acid transporters in cholestasis.
Semin Liver Dis 2010;30:160–77.
22. Poso A, Honkakoski P. Ligand recognition by drug-activated
nuclear receptors PXR and CAR: structural, site-directed
mutagenesis and molecular modeling studies. Mini Rev Med
Chem 2006;6:937–47.
23. Raucy JL, Lasker JM. Current in vitro high throughput screening
approaches to assess nuclear receptor activation. Curr Drug
Metab 2010;11:806–14.
24. Köhle C, Schwarz M, Bock KW. Promotion of hepatocarcinogenesis in humans and animal models. Arch Toxicol
2008;82:623–31.
25. Baes M, Gulick T, Choi H, Stinoli MG, Simha D, Moore DD. A new
orphan member of the nuclear hormone receptor superfamily
that interacts with a subset of retinoic acid response elements.
Mol Cell Biol 1994;14:1544–52.
26. Choi HS, Chung M, Tzameli I, Simha D, Lee YK, Seol W, et al.
Differential transactivation by two isoforms of the orphan
nuclear hormone receptor CAR. J Biol Chem 1997;272:
23565–71.
27. Trottier E, Belzil A, Stoltz C, Anderson A. Localization of a
phenobarbital-responsive element (PBRE) in the 5′-flanking
region of the rat CYP2B2 gene. Gene 1995;158:263–8.
28. Honkakoski P, Negishi M. Characterization of a phenobarbitalresponsive enhancer module in mouse P450 Cyp2b10 gene.
J Biol Chem 1997;272:14943–9.
29. Honkakoski P, Moore R, Washburn KA, Negishi M. Activation
by diverse xenochemicals of the 51-base pair phenobarbital-
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
Molnár et al.: Constitutive androstane receptor
responsive enhancer module in the CYP2B10 gene. Mol
Pharmacol 1998;53:597–601.
30. Honkakoski P, Zelko I, Sueyoshi T, Negishi M. The nuclear
orphan receptor car-retinoid X receptor heterodimer activates
the phenobarbital-responsive enhancer module of the CYP2B
gene. Mol Cell Biol 1998;18:5652–8.
31. Wei P, Zhang J, Egan-Hafley M, Liang S, Moore DD. The nuclear
receptor CAR mediates specific xenobiotic induction of drug
metabolism. Nature 2000;407:920–3.
32. Ueda A, Hamadeh HK, Webb HK, Yamamoto Y, Sueyoshi T,
Afshari CA, et al. Diverse roles of the nuclear orphan receptor
CAR in regulating hepatic genes in response to phenobarbital.
Mol Pharmacol 2002;61:1–6.
33. Yamamoto Y, Moore R, Goldsworthy TL, Negishi Monpot RR.
The orphan nuclear receptor constitutive active/androstane
receptor is essential for liver tumor promotion by phenobarbital
in mice. Cancer Res 2004;64:7197–200.
34. Zhang Q, Bae Y, Kemper JK, Kemper B. Analysis of multiple
nuclear receptor binding sites for CAR/RXR in the phenobarbital
responsive unit of CYP2B2. Arch Biochem Biophys
2006;451:119–27.
35. Lau AJ, Yang G, Rajaraman G, Baucom CC, Chang TK. Speciesdependent and receptor-selective action of bilobalide on the
function of constitutive androstane receptor and pregnane X
receptor. Drug Metab Dispos 2012;40:178–86.
36. Huang W, Zhang J, Washington M, Liu J, Parant JM, Lozano G,
et al. Xenobiotic stress induces hepatomegaly and liver tumors
via the nuclear receptor constitutive androstane receptor. Mol
Endocrinol 2005;19:1646–53.
37. Kiyosawa N, Kwekel JC, Burgoon LD, Dere E, Williams KJ,
Tashiro C, et al. Species-Specific regulation of PXR/CAR/
ER-target genes in the mouse and rat liver elicited by o,p′-DDT.
BMC Genomics 2008;9:487.
38. Chen T, Chen Q, Xu Y, Zhou Q, Zhu J, Zhang H, et al. SRC-3 is
required for car-regulated hepatocyte proliferation and drug
metabolism. J Hepatol 2012;56:210–7.
39. Tian J, Huang H, Hoffman B, Liebermann DA, Ledda-Columbano
GM, Columbano A, et al. Gadd45Β is an inducible coactivator
of transcription that facilitates rapid liver growth in mice. J Clin
Invest 2011;121:4491–502.
40. Kodama S, Koike C, Negishi M, Yamamoto Y. Nuclear receptors
CAR and PXR cross talk with FOXO1 to regulate genes that
encode drug-metabolizing and gluconeogenic enzymes. Mol
Cell Biol 2004;24:7931–40.
41. Xu RX, Lambert MH, Wisely BB, Warren EN, Weinert EE, Waitt GM,
et al. A structural basis for constitutive activity in the human
CAR/RXRα heterodimer. Mol Cell 2004;16:919–28.
42. Suino K, Peng L, Reynolds R, Li Y, Cha JY, Repa JJ, et al. The
nuclear xenobiotic receptor CAR: structural determinants
of constitutive activation and heterodimerization. Mol Cell
2004;16:893–905.
43. Shan L, Vincent J, Brunzelle JS, Dussault I, Lin M, Ianculescu I,
et al. Structure of the murine constitutive androstane receptor
complexed to androstenol: a molecular basis for inverse
agonism. Mol Cell 2004;16:907–17.
44. Moras D, Gronemeyer H. The nuclear receptor ligandbinding domain: structure and function. Curr Opin Cell Biol
1998;10:384–91.
45. Rochel N, Wurtz JM, Mitschler A, Klaholz B, Moras D. The crystal
structure of the nuclear receptor for vitamin D bound to its
natural ligand. Mol Cell 2000;5:173–9.
89
46. Stehlin C, Wurtz JM, Steinmetz A, Greiner E, Schüle R, Moras D,
et al. X-ray structure of the orphan nuclear receptor RORβ
ligand-binding domain in the active conformation. EMBO J
2001;20:5822–31.
47. Kallen JA, Schlaeppi JM, Bitsch F, Geisse S, Geiser M, Delhon I,
et al. X-ray structure of the hRORα LBD at 1.63 A: structural and
functional data that cholesterol or a cholesterol derivative is the
natural ligand of RORα. Structure (Camb) 2002;10:1697–707.
48. Gampe RT, Montana VG, Lambert MH, Wisely GB, Milburn MV,
Xu HE. Structural basis for autorepression of retinoid X
receptor by tetramer formation and the AF-2 helix. Genes Dev
2000;14:2229–41.
49. Jyrkkärinne J, Windshügel B, Mäkinen J, Ylisirniö M, Peräkylä M,
Poso A, et al. Amino acids important for ligand specificity
of the human constitutive androstane receptor. J Biol Chem
2005;280:5960–71.
50. Dussault I, Lin M, Hollister K, Fan M, Termini J, Sherman MA,
et al. A structural model of the constitutive androstane receptor
defines novel interactions that mediate ligand-independent
activity. Mol Cell Biol 2002;22:5270–80.
51. Windshügel B, Jyrkkärinne J, Poso A, Honkakoski P, Sippl W.
Molecular dynamics simulations of the human CAR ligandbinding domain: deciphering the molecular basis for
constitutive activity. J Mol Model 2005;11:69–79.
52. Windshügel B, Jyrkkärinne J, Vanamo J, Poso A, Honkakoski P,
Sippl W. Comparison of homology models and x-ray structures
of the nuclear receptor CAR: assessing the structural basis of
constitutive activity. J Mol Graph 2007;25:644–57.
53. Windshügel B, Poso A. Constitutive activity and ligand-dependent
activation of the nuclear receptor car-insights from molecular
dynamics simulations. J Mol Recogn 2011;24:875–82.
54. Jyrkkärinne J, Küblbeck J, Pulkkinen J, Honkakoski P, Laatikainen
R, Poso A, et al. Molecular dynamics simulations for human CAR
inverse agonists. J Chem Inf Model 2012;52:457–64.
55. Repo S, Jyrkkärinne J, Pulkkinen JT, Laatikainen R, Honkakoski P,
Johnson MS. Ligand specificity of constitutive androstane
receptor as probed by induced-fit docking and mutagenesis.
J Med Chem 2008;51:7119–31.
56. Küblbeck J, Laitinen T, Jyrkkärinne J, Rousu T, Tolonen A,
Abel T, et al. Use of comprehensive screening methods to
detect selective human CAR activators. Biochem Pharmacol
2011;82:1994–2007.
57. Küblbeck J, Jyrkkärinne J, Molnár F, Kuningas T, Patel J,
Windshügel B, et al. New in vitro tools to study human
constitutive androstane receptor (CAR) biology: discovery
and comparison of human CAR inverse agonists. Mol Pharm
2011;8:2424–33.
58. Jyrkkärinne J, Mäkinen J, Gynther J, Savolainen H, Poso A,
Honkakoski P. Molecular determinants of steroid inhibition
for the mouse constitutive androstane receptor. J Med Chem
2003;46:4687–95.
59. Küblbeck J, Jyrkkärinne J, Poso A, Turpeinen M, Sippl W,
Honkakoski P, et al. Discovery of substituted sulfonamides and
thiazolidin-4-one derivatives as agonists of human constitutive
androstane receptor. Biochem Pharmacol 2008;76:1288–97.
60. Dring AM, Anderson LE, Qamar S, Stoner MA. Rational
quantitative structure-activity relationship (RQSAR) screen for
PXR and CAR isoform-specific nuclear receptor ligands. Chem
Biol Interact 2010;188:512–25.
61. Jyrkkärinne J, Windshügel B, Rönkkö T, Tervo AJ, Küblbeck J,
Lahtela-Kakkonen M, et al. Insights into ligand-elicited
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
90
Molnár et al.: Constitutive androstane receptor
activation of human constitutive androstane receptor based on
novel agonists and three-dimensional quantitative structureactivity relationship. J Med Chem 2008;51:7181–92.
62. Burk O, Piedade R, Ghebreghiorghis L, Fait JT, Nussler AK,
Gil JP, et al. Differential effects of clinically used derivatives
and metabolites of artemisinin in the activation of constitutive
androstane receptor isoforms. Br J Pharmacol 2012;
167:666–81.
63. Lynch C, Pan Y, Li L, Ferguson SS, Xia M, Swaan PW, et al. Identification of novel activators of constitutive androstane receptor
from FDA-approved drugs by integrated computational and
biological approaches. Pharm Res 2013;30:489–501.
64. Ekins S, Reschly EJ, Hagey LR, Krasowski MD. Evolution of
pharmacologic specificity in the pregnane X receptor. BMC Evol
Biol 2008;8:103.
65. Handschin C, Podvinec M, Meyer UA. CXR, a chicken xenobioticsensing orphan nuclear receptor, is related to both mammalian
pregnane X receptor (PXR) and constitutive androstane receptor
(CAR). Proc Natl Acad Sci USA 2000;97:10769–74.
66. Lindblom TH, Pierce GJ, Sluder AE. A C. elegans orphan nuclear
receptor contributes to xenobiotic resistance. Curr Biol
2001;11:864–8.
67. Maglich JM, Caravella JA, Lambert MH, Willson TM, Moore JT,
Ramamurthy L. The first completed genome sequence from
a teleost fish (Fugu rubripes) adds significant diversity
to the nuclear receptor superfamily. Nucleic Acids Res
2003;31:4051–8.
68. Makino T, McLysaght A. Ohnologs in the human genome are
dosage balanced and frequently associated with disease. Proc
Natl Acad Sci USA 2010;107:9270–4.
69. Mathäs M, Burk O, Qiu H, Nusshag C, Gödtel-Armbrust U,
Baranyai D, et al. Evolutionary history and functional characterization of the amphibian xenosensor CAR. Mol Endocrinol
2012;26:14–26.
70. Krasowski MD, Yasuda K, Hagey LR, Schuetz EG. Evolution of the
pregnane X receptor: adaptation to cross-species differences in
biliary bile salts. Mol Endocrinol 2005;19:1720–39.
71. Krasowski MD, Yasuda K, Hagey LR, Schuetz EG. Evolutionary
selection across the nuclear hormone receptor superfamily
with a focus on the NR1I subfamily (vitamin D, pregnane X,
and constitutive androstane receptors). Nucl Receptor
2005;3:2.
72. Yoshinari K, Sueyoshi T, Moore R, Negishi M. Nuclear receptor
CAR as a regulatory factor for the sexually dimorphic induction
of CYB2B1 gene by phenobarbital in rat livers. Mol Pharmacol
2001;59:278–84.
73. Omiecinski CJ, Coslo DM, Chen T, Laurenzana EM, Peffer RC.
Multi-species analyses of direct activators of the constitutive
androstane receptor. Toxicol Sci 2011;123:550–62.
74. Arnold KA, Eichelbaum M, Burk O. Alternative splicing affects
the function and tissue-specific expression of the human
constitutive androstane receptor. Nucl Receptor 2004;2:1.
75. Lamba JK, Lamba V, Yasuda K, Lin YS, Assem M, Thompson E,
et al. Expression of constitutive androstane receptor splice
variants in human tissues and their functional consequences.
J Pharmacol Exp Ther 2004;311:811–21.
76. DeKeyser JG, Laurenzana EM, Peterson EC, Chen T, Omiecinski CJ.
Selective phthalate activation of naturally occurring human
constitutive androstane receptor splice variants and the
pregnane X receptor. Toxicol Sci 2011;120:381–91.
77. Auerbach SS, Ramsden R, Stoner MA, Verlinde C, Hassett C,
Omiecinski CJ. Alternatively spliced isoforms of the human
constitutive androstane receptor. Nucleic Acids Res
2003;31:3194–207.
78. Jinno H, Tanaka-Kagawa T, Hanioka N, Ishida S, Saeki M, Soyama
A, et al. Identification of novel alternative splice variants of
human constitutive androstane receptor and characterization of
their expression in the liver. Mol Pharmacol 2004;65:496–502.
79. Faucette SR, Zhang T-C, Moore R, Sueyoshi T, Omiecinski CJ,
LeCluyse EL, et al. Relative activation of human pregnane
X receptor versus constitutive androstane receptor defines
distinct classes of CYP2B6 and CYP3A4 inducers. J Pharmacol
Exp Ther 2007;320:72–80.
80. Thompson EE, Kuttab-Boulos H, Krasowski MD, Di Rienzo A.
Functional constraints on the constitutive androstane receptor
inferred from human sequence variation and cross-species
comparisons. Hum Genomics 2005;2:168–78.
81. Ikeda S, Kurose K, Jinno H, Sai K, Ozawa S, Hasegawa R,
et al. Functional analysis of four naturally occurring variants
of human constitutive androstane receptor. Mol Gen Metab
2005;86:314–9.
82. Swart M, Whitehorn H, Ren Y, Smith P, Ramesar RS, Dandara C.
PXR and CAR single nucleotide polymorphisms influence plasma
efavirenz levels in South African HIV/AIDS patients. BMC Med
Genet 2012;13:112–23.
83. Cortes CP, Siccardi M, Chaikan A, Owen A, Zhang G, Porte CJ.
Correlates of efavirenz exposure in Chilean patients affected
with human immunodeficiency virus reveals a novel association
with a polymorphism in the constitutive androstane receptor.
Ther Drug Monitor 2013;35:78–83.
84. Turpeinen M, Zanger UM. Cytochrome P450 2B6: function,
genetics, and clinical relevance. Drug Metabol Drug Interact
2012;27:185–97.
85. Ding X, Lichti K, Kim I, Gonzalez FJ, Staudinger JL. Regulation
of constitutive androstane receptor and its target genes
by fasting, camp, hepatocyte nuclear factor alpha, and the
coactivator peroxisome proliferator-activated receptor gamma
coactivator-1α. J Biol Chem 2006;281:26540–51.
86. Pascussi JM, Robert A, Moreau A, Ramos J, Bioulac-Sage P,
Navarro F, et al. Differential regulation of constitutive
androstane receptor expression by hepatocyte nuclear factor-4
alpha isoforms. Hepatology 2007;45:1146–53.
87. Pascussi JM, Gerbal-Chaloin S, Fabre JM, Maurel P, Vilarem MJ.
Dexamethasone enhances constitutive androstane
receptor expression in human hepatocytes: consequences
on cytochrome P450 gene regulation. Mol Pharmacol
2000;58:1441–50.
88. Saito K, Kobayashi K, Mizuno Y, Fukuchi Y, Furihata T, Chiba K.
Peroxisome proliferator-activated receptor alpha (PPARalpha)
agonists induce constitutive androstane receptor (CAR) and
cytochrome P450 2B in rat primary hepatocytes. Drug Metab
Pharmacokinet 2010;25:108–11.
89. Maglich JM, Stoltz CM, Goodwin B, Hawkins-Brown D, Moore JT,
Kliewer SA. Nuclear pregnane X receptor and constitutive
androstane receptor regulate overlapping but distinct sets of
genes involved in xenobiotic detoxification. Mol Pharmacol
2002;62:638–46.
90. Ooe H, Kon J, Oshima H, Mitaka T. Thyroid hormone is necessary
for expression of constitutive androstane receptor in rat
hepatocytes. Drug Metab Dispos 2009;37:1963–9.
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
Molnár et al.: Constitutive androstane receptor
91. Osabe M, Sugatani J, Takemura A, Kurosawa M, Yamazaki Y,
Ikari A, et al. Up-regulation of CAR expression through Elk-1 in
HepG2 and SW480 cells by serum starvation stress. FEBS Lett
2009;583:885–9.
92. Koike C, Moore R, Negishi M. Extracellular signal-regulated
kinase is an endogenous signal retaining the nuclear
constitutive active/androstane receptor (CAR) in the cytoplasm
of mouse primary hepatocytes. Mol Pharmacol 2007;71:
1217–21.
93. Osabe M, Negishi M. Active ERK1/2 protein interacts
with the phosphorylated nuclear constitutive active/
androstane receptor (CAR; NR1I3), repressing dephosphorylation and sequestering CAR in the cytoplasm. J Biol Chem
2011;286:35763–9.
94. Gachon F, Olela FF, Schaad O, Descombes P, Schibler U. The
circadian PAR-domain basic leucine zipper TFs DBP, TEF, and
HLF modulate basal and inducible xenobiotic detoxification.
Cell Metab 2006;4:25–36.
95. Kobayashi K, Sueyoshi T, Inoue K, Moore R, Negishi M.
Cytoplasmic accumulation of the nuclear receptor CAR by a
tetratricopeptide repeat protein in HepG2 cells. Mol Pharmacol
2003;64:1069–75.
96. Mutoh S, Osabe M, Inoue K, Moore R, Pedersen L, Perera L,
et al. Dephosphorylation of threonine 38 is required for
nuclear translocation and activation of human xenobiotic
receptor CAR (NR1I3). J Biol Chem 2009;284:34785–92.
97. Hosseinpour F, Moore R, Negishi M, Sueyoshi T. Serine 202
regulates the nuclear translocation of constitutive active/
androstane receptor. Mol Pharmacol 2006;69:1095–102.
98. Sueyoshi T, Moore R, Sugatani J, Matsumura Y, Negishi M.
PPP1R16A, the membrane subunit of protein phosphatase
1β, signals nuclear translocation of the nuclear receptor
constitutive active/androstane receptor. Mol Pharmacol
2008;73:1113–21.
99. Blättler SM, Rencurel F, Kaufmann MR, Meyer UA. In the
regulation of cytochrome P450 genes, phenobarbital targets
LKB1 for necessary activation of AMP-activated protein kinase.
Proc Natl Acad Sci USA 2007;104:1045–50.
100. Chakraborty S, Kanakasabai S, Bright JJ. Constitutive
androstane receptor agonist CITCO inhibits growth
and expansion of brain tumour stem cells. Br J Cancer
2011;104:448–59.
101. Kamino H, Negishi M. The nuclear receptor constitutive
active/androstane receptor arrests DNA-damaged human
hepatocellular carcinoma Huh7 cells at the G2/M phase. Mol
Carcinogen 2012;51:206–12.
102. Sidhu JS, Omiecinski CJ. cAMP-associated inhibition of
phenobarbital-inducible cytochrome P450 gene expression in
primary rat hepatocyte cultures. J Biol Chem 1995;270:12762–73.
103. Honkakoski P, Negishi M. Protein serine/threonine
phosphatase inhibitors suppress phenobarbital-induced
Cyp2b10 gene transcription in mouse primary hepatocytes.
Biochem J 1998;330:889–95.
104. Ito S, Tsuda M, Yoshitake A, Yanai T, Masegi T. Effect of
phenobarbital on hepatic gap junctional intercellular
communication in rats. Toxicol Pathol 1998;26:253–9.
105. Warner KA, Fernstrom MJ, Ruch RJ. Inhibition of mouse
hepatocyte gap junctional intercellular communication by
phenobarbital correlates with strain-specific hepatocarcinogenesis. Toxicol Sci 2003;71:190–7.
91
106. Sugatani J, Sueyoshi T, Negishi M, Miwa M. Regulation of the
human UGT1A1 gene by nuclear receptors constitutive active/
androstane receptor, pregnane X receptor, and glucocorticoid
receptor. Method Enzymol 2005;400:92–104.
107. Goodwin B, Hodgson E, D’Costa DJ, Robertson GR, Liddle
C. Transcriptional regulation of the human CYP3A4 gene
by the constitutive androstane receptor. Mol Pharmacol
2002;62:359–65.
108. Mäkinen J, Frank C, Jyrkkärinne J, Gynther J, Carlberg C,
Honkakoski P. Modulation of mouse and human phenobarbitalresponsive enhancer module by nuclear receptors. Mol
Pharmacol 2002;62:366–78.
109. Kassam A, Winrow CJ, Fernandez-Rachubinski F, Capone JP,
Rachubinski RA. The peroxisome proliferator response element
of the gene encoding the peroxisomal beta-oxidation enzyme
enoyl-CoA hydratase/3-hydroxyacyl-CoA dehydrogenase
is a target for constitutive androstane receptor beta/9-cisretinoic acid receptor-mediated transactivation. J Biol Chem
2000;275:4345–50.
110. Miao J, Fang S, Bae Y, Kemper JK. Functional inhibitory
cross-talk between constitutive androstane receptor and
hepatic nuclear factor-4 in hepatic lipid/glucose metabolism
is mediated by competition for binding to the DR1 motif and
to the common coactivators, GRIP-1 and PGC-1α. J Biol Chem
2006;281:14537–46.
111. Kachaylo EM, Yarushkin AA, Pustylnyak VO. Constitutive
androstane receptor activation by 2,4,6-triphenyl-1,3-dioxane
suppresses the expression of the gluconeogenic genes. Eur J
Pharmacol 2012;679:139–43.
112. Frank C, Gonzalez MM, Oinonen C, Dunlop TW, Carlberg C.
Characterization of DNA complexes formed by the nuclear
receptor constitutive androstane receptor. J Biol Chem
2003;278:43299–310.
113. Burk O, Katja AA, Geick A, Tegude H, Eichelbaum M. A role for
constitutive androstane receptor in the regulation of human
intestinal MDR1 expression. Biol Chem 2005;386:503–13.
114. Heery DM, Kalkhoven E, Hoare S, Parker MG. A signature motif
in transcriptional co-activators mediates binding to nuclear
receptors. Nature 1997;387:733–6.
115. McKenna NJ, Lanz RB, O’Malley BW. Nuclear receptor
coregulators: cellular and molecular biology. Endocr
Rev 1999;20:321–44.
116. Molnár F, Matilainen M, Carlberg C. Structural determinants
of the agonist-independent association of human peroxisome
proliferator-activated receptors with coactivators. J Biol Chem
2005;280:26543–56.
117. Xia J, Liao L, Sarkar J, Matsumoto K, Reddy JK, Xu J, et al.
Redundant enhancement of mouse constitutive androstane
receptor transactivation by p160 coactivator family members.
Arch Biochem Biophys 2007;468:49–57.
118. Choi E, Lee S, Yeom S-Y, Kim GH, Lee JW, Kim S-W. Characterization of activating signal cointegrator-2 as a novel
transcriptional coactivator of the xenobiotic nuclear
receptor constitutive androstane receptor. Mol Endocrinol
2005;19:1711–9.
119. Jia Y, Guo GL, Surapureddi S, Sarkar J, Qi C, Guo D, et al.
Transcription coactivator peroxisome proliferator-activated
receptor-binding protein/mediator 1 deficiency abrogates
acetaminophen hepatotoxicity. Proc Natl Acad Sci USA
2005;102:12531–6.
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
92
Molnár et al.: Constitutive androstane receptor
120. Sarkar J, Qi C, Guo D, Ahmed MR, Jia Y, Usuda N, et al.
Transcription coactivator PRIP, the peroxisome proliferatoractivated receptor (PPAR)-interacting protein, is redundant
for the function of nuclear receptors pparalpha and CAR,
the constitutive androstane receptor, in mouse liver. Gene
Expression 2007;13:255–69.
121. Guo D, Sarkar J, Ahmed MR, Viswakarma N, Jia Y, Yu S, et al.
Peroxisome proliferator-activated receptor (PPAR)-binding
protein (PBP) but not PPAR-interacting protein (PRIP) is
required for nuclear translocation of constitutive androstane
receptor in mouse liver. Biochem Biophys Res Commun
2006;347:485–95.
122. Malik S, Roeder RG. The metazoan mediator co-activator
complex as an integrative hub for transcriptional regulation.
Nat Rev Genet 2010;11:761–72.
123. Surapureddi S, Viswakarma N, Yu S, Guo D, Rao MS, Reddy JK.
PRIC320, a transcription coactivator, isolated from peroxisome
proliferator-binding protein complex. Biochem Biophys Res
Commun 2006;343:535–43.
124. Shiraki T, Sakai N, Kanaya E, Jingami H. Activation of orphan
nuclear constitutive androstane receptor requires subnuclear
targeting by peroxisome proliferator-activated receptor-γ
coactivator-1α. A possible link between xenobiotic response
and nutritional state. J Biol Chem 2003;278:11344–50.
125. Mäkinen J, Reinisalo M, Niemi K, Viitala P, Jyrkkärinne J, Chung
H, et al. Dual action of oestrogens on the mouse constitutive
androstane receptor. Biochem J 2003;376:465–72.
126. Lempiäinen H, Molnár F, Macias Gonzalez M, Peräkylä M,
Carlberg C. Antagonist- and inverse agonist-driven
interactions of the vitamin D receptor and the constitutive
androstane receptor with corepressor protein. Mol Endocrinol
2005;19:2258–72.
127. Konno Y, Kodama S, Moore R, Kamiya N, Negishi M. Nuclear
xenobiotic receptor pregnane X receptor locks corepressor
silencing mediator for retinoid and thyroid hormone receptors
(SMRT) onto the CYP24A1 promoter to attenuate vitamin D3
activation. Mol Pharmacol 2009;75:265–71.
128. Bae Y, Kemper JK, Kemper B. Repression of CAR-mediated
transactivation of CYP2B genes by the orphan nuclear receptor,
short heterodimer partner (SHP). DNA Cell Biol 2004;23:81–91.
129. Laurenzana EM, Chen T, Kannuswamy M, Sell BE, Strom SC,
Li Y, et al. The orphan nuclear receptor DAX-1 functions as a
potent corepressor of the constitutive androstane receptor
(NR1I3). Mol Pharmacol 2012;82:918–28.
130. Xie Y-B, Lee O-H, Nedumaran B, Seong H-A, Lee K-M, Ha H,
et al. SMILE, a new orphan nuclear receptor SHP-interacting
protein, regulates SHP-repressed estrogen receptor transactivation. Biochem J 2008;416:463–73.
131. Xie Y-B, Nedumaran B, Choi H-S. Molecular characterization
of SMILE as a novel corepressor of nuclear receptors. Nucleic
Acids Res 2009;37:4100–15.
132. Lahtela JT, Arranto AJ, Sotaniemi EA. Enzyme inducers improve
insulin sensitivity in non-insulin-dependent diabetic subjects.
Diabetes 1985;34:911–6.
133. Argaud D, Halimi S, Catelloni F, Leverve XM. Inhibition of
gluconeogenesis in isolated rat hepatocytes after chronic
treatment with phenobarbital. Biochem J 1991;280:663–9.
134. Gao J, He J, Zhai Y, Wada T, Xie W. The constitutive androstane
receptor is an anti-obesity nuclear receptor that improves
insulin sensitivity. J Biol Chem 2009;284:25984–92.
135. Ledda-Columbano GM, Pibiri M, Loi R, Perra A, Shinozuka H,
Columbano A. Early increase in cyclin D1 expression and
accelerated entry of mouse hepatocytes into S phase after
administration of the mitogen 1,4-bis[2-(3,5-dichloropyridyloxy)] benzene. Am J Pathol 2000;156:91–7.
136. Yun HJ, Kwon J, Seol W. Specific inhibition of transcriptional
activity of the constitutive androstane receptor (CAR) by the
splicing factor SF3a. Biol Chem 2008;389:1313–8.
137. Dau PT, Sakai H, Hirano M, Ishibashi H, Tanaka Y, Kameda K,
et al. Quantitative analysis of the interaction of constitutive
androstane receptor with chemicals and steroid receptor
coactivator 1 using surface plasmon resonance biosensor
systems: a case study of the baikal seal (Pusa sibirica) and the
mouse. Toxicol Sci 2013;131:116–27.
138. Maglich JM, Parks DJ, Moore LB, Collins JL, Goodwin B,
Billin AN, et al. Identification of a novel human constitutive
androstane receptor (CAR) agonist and its use in the identification of CAR target genes. J Biol Chem 2003;278:17277–83.
139. Abass K, Lämsä V, Reponen P, Küblbeck J, Honkakoski P,
Mattila S, et al. Characterization of human cytochrome P450
induction by pesticides. Toxicology 2012;294:17–26.
140. Huang W, Zhang J, Wei P, Schrader WT, Moore DD. Meclizine is
an agonist ligand for mouse constitutive androstane receptor
(CAR) and an inverse agonist for human CAR. Mol Endocrinol
2004;18:2402–8.
141. Yao R, Yasuoka A, Kamei A, Kitagawa Y, Rogi T, Taieishi N, et al.
Polyphenols in alcoholic beverages activating constitutive
androstane receptor CAR. Biosci Biotechnol Biochem
2011;75:1635–7.
142. DeKeyser JG, Stagliano MC, Auerbach SS, Prabhu KS, Jones
AD, Omiecinski CJ. Di(2-ethylhexyl)phthalate is a highly potent
agonist for the human constitutive androstane receptor splice
variant CAR2. Mol Pharmacol 2009;75:1005–13.
143. Li L, Chen T, Stanton JD, Sueyoshi T, Negishi M, Wang H. The
peripheral benzodiazepine receptor ligand 1-(2-chlorophenylmethylpropyl)-3-isoquinoline-carboxamide is a novel
antagonist of human constitutive androstane receptor. Mol
Pharmacol 2008;74:443–53.
144. Kawamoto T, Kakizaki S, Yoshinari K, Negishi M. Estrogen
activation of the nuclear orphan receptor CAR (constitutive
active receptor) in induction of the mouse Cyp2b10 gene. Mol
Endocrinol 2000;14:1897–905.
145. Yao R, Yasuoka A, Kamei A, Kitagawa Y, Tateishi N, Tsuruoka N,
et al. Dietary flavonoids activate the constitutive androstane
receptor (CAR). J Agric Food Chem 2010;58:2168–73.
146. Imai J, Yamazoe Y, Yoshinari K. Novel cell-based reporter assay
system using epitope-tagged protein for the identification
of agonistic ligands of constitutive androstane receptor
(CAR). Drug Metab Pharmacokinet 2012. DOI: http://dx.doi.
org/10.2133/dmpk.DMPK-12-RG-112.
147. Huang W, Zhang J, Chua SS, Qatanani M, Han Y, Granata R,
et al. Induction of bilirubin clearance by the constitutive
androstane receptor (CAR). Proc Natl Acad Sci USA
2003;100:4156–61.
148. Chen T, Tompkins LM, Li L, Li H, Kim G, Zheng Y, et al. A single
amino acid controls the functional switch of human constitutive
androstane receptor (CAR) 1 to the xenobiotic-sensitive splicing
variant CAR3. J Pharmacol Exp Ther 2010;332:106–15.
149. Lau AJ, Yang G, Chang TK. Isoform-selective activation of
human constitutive androstane receptor by Ginkgo biloba
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM
Molnár et al.: Constitutive androstane receptor
150.
151.
152.
153.
154.
extract: functional analysis of the SV23, SV24, and SV25 splice
variants. J Pharmacol Exp Ther 2011;339:704–15.
Choi E-J, Jang Y-J, Cha E-Y, Shin J-G, Lee SS. Identification
and characterization of novel alternative splice variants
of human constitutive androstane receptor in liver
samples of Koreans and Caucasians. Drug Metab
Dispos 2013;41:888–96.
Tzameli I, Chua SS, Cheskis B, Moore DD. Complex effects of
rexinoids on ligand dependent activation or inhibition of the
xenobiotic receptor, CAR. Nucl Receptor 2003;1:2.
Chen S, Wang K, Wan Y-J. Retinoids activate RXR/
CAR-mediated pathway and induce CYP3A. Biochem
Pharmacol 2010;79:270–6.
Howe K, Sanat F, Thumser AE, Coleman T, Plant N. The
statin class of HMG-CoA reductase inhibitors demonstrate
differential activation of the nuclear receptors PXR, CAR and
FXR, as well as their downstream target genes. Xenobiotica
2011;41:519–29.
Kanno Y, Inouye Y. A consecutive three alanine residue
insertion mutant of human CAR: a novel CAR ligand screening
system in HepG2 cells. J Toxicol Sci 2010;35:515–25.
93
155. Pissios P, Tzameli I, Kushner P, Moore DD. Dynamic
stabilization of nuclear receptor ligand binding domains by
hormone or corepressor binding. Mol Cell 2000;6:245–53.
156. Burk O, Arnold KA, Nussler AK, Schaeffeler E, Efimova E, Avery
BA, et al. Antimalarial artemisinin drugs induce cytochrome
P450 and MDR1 expression by activation of xenosensors
pregnane X receptor and constitutive androstane receptor. Mol
Pharmacol 2005;67:1954–65.
157. Kobayashi K, Saito K, Takagi S, Chiba K. Ligand-dependent
assembly of pregnane X receptor, constitutive androstane
receptor and liver X receptor is applicable to identify ligands.
Drug Metab Lett 2010;4:88–94.
158. Li H, Chen T, Cottrell J, Wang H. Nuclear translocation of
adenoviral-enhanced yellow fluorescent protein-taggedhuman constitutive androstane receptor (hCAR): a novel tool
for screening hCAR activators in human primary hepatocytes.
Drug Metab Dispos 2009;37:1098–106.
159. Moore LB, Parks DJ, Jones SA, Bledsoe RK, Consler TG, Stimmel
JB, et al. Orphan nuclear receptors constitutive androstane
receptor and pregnane X receptor share xenobiotic and steroid
ligands. J Biol Chem 2000;275:15122–7.
Brought to you by | University of Oulu
Authenticated |
[email protected]
Download Date | 6/7/13 10:06 AM