You are on page 1of 9

toxicological sciences 132(1), 142–150 2013

doi:10.1093/toxsci/kfs321
Advance Access publication November 14, 2012

Expression of Human CAR Splicing Variants in BAC-Transgenic Mice


Yu-Kun Jennifer Zhang, Hong Lu, and Curtis D. Klaassen1
Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, Kansas 66160

1
To whom correspondence should be addressed at Department of Internal Medicine, University of Kansas Medical Center, 3901 Rainbow Blvd, Kansas City,
KS 66160. Fax: (913) 588-9197. E-mail: CKLAASSE@kumc.edu.

Received May 10, 2012; accepted November 7, 2012

heterodimerizes with the retinoid X receptor, binds to the pheno-


The nuclear receptor constitutive androstane receptor (CAR) is barbital (PB) responsive enhancer module elements, and induces
a key regulator for drug metabolism in liver. Human CAR (hCAR)
the transcription of numerous phase I and II enzymes and trans-
transcripts are subjected to alternative splicing. Some hCAR spli-
porters to facilitate the elimination of chemicals (Honkakoski
cing variants (SVs) have been shown to encode functional proteins
by reporter assays. However, in vivo research on the activity of these et  al., 1998). Examples of genes regulated by CAR are phase
hCAR SVs has been impeded by the absence of a valid model. This I enzymes such as cytochrome P450s (Cyps) (Huang et al., 2004;
study engineered an hCAR-BAC-transgenic (hCAR-TG) mouse Petrick and Klaassen, 2007), phase II enzymes such as UDP-
model by integrating the 8.5-kbp hCAR gene as well as 73-kbp glucuronosyltransferase 1a1 (Huang et al., 2004; Sugatani et al.,
upstream and 91-kbp downstream human genomic DNA into the 2001), glutathione S-transferase a1 (Huang et  al., 2004), and
genome of CAR-null mice. A  series of experiments demonstrate sulfotransferase 2a1 (Assem et al., 2004), as well as membrane
that (1) the expression of major hCAR mRNA SVs, SV0-4, in livers transporters such as multidrug resistance-associated protein 4
of hCAR-TG mice is comparable to that in human livers; (2) the (Assem et al., 2004; Petrick and Klaassen, 2007). Because of its
hCAR SVs are predominantly expressed in liver, which resembles constitutive activity and broad influence on xenobiotic metabol-
the tissue distribution of CAR in humans, but diverges from that in
ism, the hepatic expression level of CAR could potentially affect
mice; and (3) major hCAR mRNA SVs increase markedly in post-
the capability of an individual in eliminating drugs from the body
natal livers of hCAR-TG mice, which mimics the ontogeny of CAR
mRNA in humans. Thus, the transgene likely contains all the func- (Lamba et al., 2003; Vyhlidal et al., 2006).
tional regulatory elements controlling proper spatial and temporal Alternative splicing is a major source of protein diversity
expression of the hCAR gene. Moreover, hCAR-TG mice respond in higher eukaryotes and plays critical roles in differentiation,
to the hCAR-specific agonist 6-(4-chlorophenyl)imidazo[2,1-b] development, and disease. In humans, 35 splicing variants
[1,3]thiazole-5-carbaldehyde O-(3,4-dichlorobenzyl)oxime instead (SVs) of CAR mRNA have been identified (Ensembl:
of the mouse CAR agonist 1,4-bis[2-(3,5-dichloropyridyloxy)]ben- ENSG00000143257). In addition to the first-cloned SV0
zene, as well as the common CAR activator, phenobarbital, sug- (CAR1), SV1 (CAR2), and SV2 (CAR3) are the most abundant
gesting that hCAR is fully functional in livers of transgenic mice. human CAR (hCAR) SVs in human livers (Lamba et al., 2004).
In summary, the hCAR-TG mice developed by this study represent Although most of hCAR SVs do not encode functional proteins,
a valid model for studying in vivo function and regulation of hCAR
several in vitro assays showed that some hCAR SVs encode
and its splicing variants.
proteins that can be activated by chemicals that do not activate
Key Words: CAR; splicing variant; in vivo; BAC; transgenic mice.
the constitutively active wild-type (WT) hCAR (SV0 or CAR1)
(DeKeyser et al., 2009; Dring et al., 2010). For example, SV1
responds to the common plasticizer di(2-ethylhexyl)phthalate
Nuclear receptor constitutive androstane receptor (CAR; (DeKeyser et al., 2009), whereas SV2 exhibits low basal activity,
NR1I3) plays an important role in regulating the metabolism of but it is strongly activated by the prototypical hCAR agonist
xenobiotics and nutrients in the liver. Compared with many other 6-(4-chlorophenyl)imidazo[2,1-b] [1,3]thiazole-5-carbaldehyde
nuclear receptors, CAR is unique in that it is constitutively active O-(3,4-dichlorobenzyl)oxime (CITCO) (Auerbach et al., 2005).
and can be further activated by many endogenous and exogen- However, the in vivo function and expression profiles of these
ous chemicals (Swales and Negishi, 2004), including steroid hCAR SVs remain unknown, mainly due to the lack of a valid
hormones, pharmaceuticals, as well as environmental, dietary, animal model.
and occupational chemicals (Hernandez et al., 2009). Upon acti- Human hepatoma cell lines, human primary hepatocytes,
vation, CAR translocates from the cytoplasm into the nucleus, and human liver tissues are model systems that are commonly

© The Author 2012. Published by Oxford University Press on behalf of the Society of Toxicology.
All rights reserved. For permissions, please email: journals.permissions@oup.com
In Vivo Study of Human Car Splicing Variants 143

used to study the regulation and function of human genes. DNA from mCAR−/− mice served as the negative control. The homozygous
Significant disadvantages of these systems include compro- hCAR-TG mice from founder no. 22, as well as their WT littermates, were
used for this study.
mised regulatory properties (cell lines), lack of in vivo envir-
onment (cell lines and primary hepatocytes), and limited Animals and treatments.  All procedures were conducted in accordance
with the NIH Guidelines for the Care and Use of Laboratory Animals and were
accessibility (primary hepatocytes and human liver tissues).
approved by the University of Kansas Medical Center Institutional Animal
Laboratory mice provide useful models of human development Care and Use Committee. Mice were kept in a temperature/humidity controlled
and drug metabolism, but hCAR is different from mouse CAR facility. Food and water were available ad libitum. Light cycles were on a
(mCAR) in regard to ligand specificity and pre-mRNA alter- 14/10-h light/dark cycle with the light phase starting at 0600 h. For the devel-
native splicing. mCAR has a specific ligand, 1,4-bis[2-(3,5- opmental study, livers were collected from the hCAR-TG mice that were 2 days
before birth (both male and female) and 5, 10, 15, 20, and 60 days (male only)
dichloropyridyloxy)]benzene (TCPOBOP) (Nims et al., 1999),
of age. Livers were also collected from 60-day-old female hCAR-TG mice for
and does not respond to the hCAR-specific ligand CITCO. In the gender divergence study. For the tissue distribution study, the following
comparison to the 35 hCAR SVs, mice only express 2 SVs of organs were harvested from 2-month-old male hCAR-TG mice: liver, kidney,
CAR mRNA (Choi et al., 1997). hCAR transgenic mice pro- stomach, duodenum, jejunum, ileum, colon, lung, heart, testis, white adipose
vide a promising tool to study in vivo function and regulation of tissue, brown adipose tissue, and muscle. For the inducer study, male and/or
female hCAR-TG and WT mice (2 months old; n = 3–5) were dosed daily by
hCAR. Whereas two lines of hCAR-transgenic mice have been
ip injection with corn oil (vehicle), CITCO (20 mg/kg/d), or PB (100 mg/kg/
engineered, they either express only WT (SV0) hCAR (Zhang day; male only) for 3 days, and livers were removed 24 h after the last dose.
et al., 2002) or lack the promoter and 5′-untranslated region of TCPOBOP (3 mg/kg) was administrated by a single ip injection, and livers
hCAR gene (Scheer et al., 2008). Therefore, the purpose of this were removed 72 h later.
study was to engineer a new line of hCAR-transgenic mice by qRT-PCR.  Total RNA was prepared from various tissues of WT and
integrating the entire hCAR gene encoding region as well as hCAR-TG mice using RNA-Bee reagent (Tel-Test, Inc., Friendswood, TX)
73-kbp upstream and 91-kbp downstream of human genomic according to the manufacturer’s protocol. RNA was dissolved in diethyl pyro-
DNA into the mouse genome. Characterization of these hCAR- carbonate–treated deionized water, and RNA concentrations were determined
spectrophotometrically at 260 nm. RNA samples from five male Caucasian
transgenic mice indicates that they are suitable for studying the human livers were available in Dr. Klaassen’s laboratory (Xenotech, Lenexa,
developmental regulation and in vivo transcriptional function KS). RNA was reverse transcribed into cDNA with multiscript reverse tran-
of this human nuclear receptor. scriptase using High Capacity cDNA Reverse Transcription kits from Applied
Biosystems (Foster City, CA). Primers for detecting individual SVs of hCAR
were described previously (Jinno et al., 2004). The following primers were
Materials and Methods used to detect respective gene expression: Cyp2b10 (GenBank accession
number AF128849), 5′-aaggagaagtccaaccagca-3′ and 5′-ctctgcaacatggggg-
Chemicals and reagents.  CITCO was obtained from Enzo Life Science tact-3′; mCAR (GenBank accession number NM_009803), 5′-ctcaaggaaa-
(Farmingdale, NY). TCPOBOP and PB were obtained from Sigma Aldrich (St gcagggtcag-3′ and 5′-agttcctcggcccatattct-3′; Cyp3a11 (GenBank accession
Louis, MO). All other reagents were obtained from Sigma Aldrich. number NM_007818), 5′-acaaacaagcagggatggac-3′ and 5′-ggtagaggag-
caccaagctg-3′; and glyceraldehyde-3-phosphate dehydrogenase (Gapdh)
Generation of hCAR-BAC-transgenic mice.  The hCAR-BAC-transgenic (GenBank accession number M32599), 5′-aactttggcattgtggaagg-3′ and
(hCAR-TG) mouse founders were generated by the Transgenic and Gene- 5′-ggatgcagggatgatgttct-3′. The Power SYBR Green Master Mix (Applied
Targeting Institutional Facility at the University of Kansas Medical Center. Biosystems) was used for real-time PCR analysis. Differences in gene
Briefly, bacterial artificial chromosome (BAC) RP11-297K8 DNA, constructed expression between groups were calculated using cycle threshold (Ct) values,
by the Sanger Center Chromosome 1 Mapping Group, was linearized with which were normalized to Gapdh mRNA.
endonuclease NotI and microinjected at the concentration of 0.1 ng/µl into pro-
Immunoblot analysis.  Nuclear and cytoplasmic fractions were prepared
nuclei of fertilized eggs of FVB × C57BL/6 mice. Two founder lines (no. 22
from three individual mouse livers in each treatment group using NE-PER
and no.  25)  were generated, as detected by PCR analysis of genomic DNA
nuclear protein extraction kit (ThermoFisher Scientific Inc., Rockford, IL).
from tail biopsies.
Thirty micrograms of nuclear protein and 100 μg of the cytosol protein were
The transgenic founders were bred with CAR-null mice (provided by Dr.
separated by SDS-polyacrylamide gel electrophoresis and electrophoretically
Ivan Rusyn, University of North Carolina, Chapel Hill, NC) on the C57BL/6
transferred to a polyvinylidene fluoride membrane. Nuclear proteins were
background to generate hCAR+/−/mCAR+/− F1 hybrids. The F1 offspring
probed with an anti-CAR1/2 (M-150) polyclonal antibody (Santa Cruz
were further crossed with CAR-null mice to generate the hCAR+/−/mCAR−/−
Biotechnology, Inc., Santa Cruz, CA) and anti-TATA-binding protein (TBP)
F2 generation that expressed only hCAR but not mCAR as determined by
monoclonal antibody (ThermoFisher Scientific Inc.). Cytoplasmic proteins
PCR analysis. The F2 generation of hCAR+/−/mCAR−/− mice were bred with
were probed with rabbit anti-rat CYP2B1/2 polyclonal antibody (Xenotech)
their littermates to generate homozygous hCAR+/+/mCAR−/−. The zygosity
and rabbit β-actin polyclonal antibody (Abcam, Cambridge, MA). Secondary
of hCAR genomic DNA in hCAR-TG mice was determined by SYBR Green
anti-rabbit or mouse antibodies were diluted 10,000-fold. Immunoreactions
real-time quantitative PCR (real-time qPCR) as described (Haurogne et al.,
were detected by Pierce ECL Western blotting substrate (ThermoFisher
2007). PCR primers (5′-cagcgcattaaagtggtagcc-3′ and 5′-cccttgcagccct-
Scientific Inc.). The density of blots was quantified by Quantity One 1-D
cacaagt-3′), targeting the 3′-end of the hCAR gene and resulting in a 201-bp
Analysis Software (Bio-Rad Laboratories, Hercules, CA). The individual blot
band, were used to determine the zygosity of the hCAR gene in hCAR-TG
densities of CAR and Cyp2b10 were normalized to the nuclear and cytosol
mice. The mouse gap junction channel protein alpha 5 (mGJA5; GenBank:
loading control, TBP, and β-actin, respectively.
NM_008121) was used as the endogenous reference. Real-time qPCR prim-
ers for mGJA5 were 5′-accatggaggtggccttca-3′ and 5′-catgcagggtatccag- Statistical analysis.  Statistical significance was assessed by two-way
gaaga-3′. Genomic DNA from the F1 generation of transgenic mice, which ANOVA when two variables were examined or Student’s t-test when one vari-
is known to be heterozygous, was used as the quantity control. Genomic able was tested. The criterion for statistical significance was p < 0.05.
144 Zhang, Lu, And Klaassen

Results Comparison of Hepatic Expression of Major SVs of hCAR


mRNA in hCAR-TG Mice and Humans
Generation and Characterization of hCAR-TG Mice
Major SVs of hCAR mRNA reported in a previous study
BAC clone RP11-297K8 containing a 173-kbp DNA frag- (Jinno et  al., 2004) were determined in livers of male adult
ment from human chromosome 1 (Fig.  1A) was used to hCAR-TG mice and compared with those in human livers. As
generate hCAR-TG mice. In addition to the hCAR gene, RP11- shown in Figure 2, the WT hCAR SV0 was the predominant iso-
297K8 encompasses other human gene loci such as ubiquitin form (~60%), followed by SV2 (~30%), and SV1 (~8%) in both
specific protease 21 (USP21), protoporphyrinogen oxidase human and hCAR-TG mouse livers. Marked interindividual dif-
(PPOX), and apolipoprotein A2 (APOA2). Further details about ferences in hCAR SV0 were observed in human liver samples.
RP11-297K8 can be found on NCBI website http://www.ncbi. The expressions of SV0, SV1, SV2, and SV3 were comparable
nlm.nih.gov/clone/270914/. The transgenic founders were gen- in human livers and hCAR-TG mouse livers, whereas SV4 was
erated and bred with CAR-null mice on the C57BL/6 back- expressed lower in hCAR-TG mice than in humans.
ground for two generations to obtain the hCAR+/+/mCAR−/− F2
generation that expresses only hCAR but not mCAR (Fig. 1B). Comparison of Prototypical CAR-Target Genes Induced by
Homozygous hCAR-TG mice were identified by real-time Species-Specific CAR Activators in the Liver
qPCR as described (Haurogne et al., 2007). The primer designed Both WT and hCAR-TG mice (male) were subjected to the
for the hCAR gene was able to detect one Ct difference for two- following four treatments: corn oil (vehicle control), TCPOBOP
fold DNA quantities between heterozygous and homozygous, (mCAR-specific ligand), CITCO (hCAR-specific ligand), and
whereas the reference gene mGJA5 showed similar Ct values PB (nonligand activator for both mCAR and hCAR). As shown
in all examined samples (Fig. 1C). The homozygous hCAR-TG in Figure 3A, hCAR and mCAR mRNAs were only detected in
mice from line no. 22 were used in this study and their WT lit- livers of hCAR-TG and WT mice, respectively. The prototyp-
termates were used as controls. ical CAR-target gene Cyp2b10 was induced by TCPOBOP and

FIG. 1.  Generation of hCAR-TG mice. (A) The BAC clone RP11-297K8, containing about 73-kbp human DNA upstream of CAR gene and 91-kbp down-
stream of CAR gene, was integrated into mouse genome to generate the hCAR-TG mice. Abbreviations of genes surrounding hCAR gene locus were shown. For
detailed information about RP11-297K8, please refer to NCBI http://www.ncbi.nlm.nih.gov/clone/270914/. (B) PCR analysis of transgenic mice that contain
hCAR but not mCAR using hCAR− and mCAR-specific primers. L, DNA ladder; P, positive plasmid control; WT, wild-type mice; −/−, CAR-null mice; 1 and 2
are two hCAR-TG/mCAR-null mice. (C) Real-time quantitative PCR analysis to identify homozygous hCAR-TG mice. The mouse gap junction channel protein
alpha 5 (mGJA5; GenBank: NM_008121) was used as the endogenous reference. A, B, C, and D represent four hCAR-TG mice that are either homozygous (+/+)
or heterozygous (+/−) of hCAR gene.
In Vivo Study of Human Car Splicing Variants 145

mRNA constantly increased along with liver development in


the hCAR-TG mice. Specifically, hCAR SV0 was 2.9-fold and
Cyp2b10 was 7.2-fold higher in livers of 5-day than those in
−2-day-old mice. At 60  days of age, hepatic hCAR SV0 and
Cyp2b10 mRNA were both ~1.9-fold higher than those at
20 days of age.
In addition to hCAR, other human genes on the BAC construct,
such as apolipoprotein A2 (APOA2) and protoporphyrinogen
oxidase (PPOX), were also expressed in livers of the hCAR-TG
mice (Fig.  4B, right panels). APOA2 encodes an abundant
apolipoprotein, which stabilize the high-density lipoprotein
particles. Human APOA2 (hAPOA2) mRNA gradually
increased in hCAR-TG mice during liver development and
maturation. In contrast, the mRNA of human PPOX (hPPOX),
the enzyme that catalyzes the seventh step in heme production,
FIG. 2.  Expression of major hCAR mRNA SVs in livers of humans and was expressed in fetal livers and markedly reduced in neonates
hCAR-TG mice. Major SVs of hCAR mRNA were analyzed in livers of five and progressively decreased to a minimal level in adults.
male Caucasians and five male hCAR-TG mice (8 weeks old). The human liver
RNA samples were purchased from Xenotech. Tissue Distribution of hCAR SVs in hCAR-TG Mice and
Species-Specific Activation of CAR in Jejunum
PB in WT mice, but not by CITCO. On the contrary, Cyp2b10 The expression of hCAR SVs in major organs in male
mRNA was increased by CITCO and PB in livers of hCAR- hCAR-TG mice was shown in Figure  5A. The SVs of hCAR
TG mice, but not by TCPOBOP. Cyp3a11 is a shared target mRNA were predominantly expressed in liver. The expression
gene by CAR and pregnane X receptor. Cyp3a11 mRNA was of hCAR mRNA SVs in kidney and colon were ~2% of that
predominantly induced by TCPOBOP and PB in WT mice, in liver, even lower in small intestine, and almost undetectable
and by CITCO and PB in livers of hCAR-TG mice. However, in adipose tissues and muscle. In organs, where hCAR SVs
TCPOBOP also slightly increased Cyp3a11 mRNA in hCAR- were detected, SV0 and SV2 were more abundant than other
TG mice, suggesting off-target induction of Cyp3a11 by this SVs. In order to determine the function of hCAR in organs that
mCAR-specific ligand in mouse liver. Interestingly, the induc- express low levels of hCAR in the transgenic mice, mRNAs of
tion of CAR target genes by PB appears to be more potent in hCAR, mCAR, and Cyp2b10 were determined in jejunums of
livers of hCAR-TG mice than that in WT mice. Moreover, WT and hCAR-TG mice that were treated with species-specific
TCPOBOP in WT mice and CITCO in hCAR-TG mice both ligands of CAR or the common CAR activator, PB. As shown
increased nuclear accumulation of mCAR and hCAR proteins, in Figure 5B, hCAR mRNA was expressed at a very low level
respectively, as well as markedly induced Cyp2b10 protein in jejunums of the hCAR-TG mice, and neither CITCO nor
(Fig.  3B). In summary, the above data demonstrate that the PB induced the CAR-target gene, Cyp2b10. In comparison,
hCAR gene is functional in livers of the transgenic mice. WT mice expressed much more mCAR in jejunum. The
mCAR-specific ligand, TCOPOBOP, efficiently induced
Ontogenetic Expression of Major hCAR SVs, Cyp2b10, and Cyp2b10 mRNA in jejunums of WT mice. Interestingly, PB
Other Human Genes in Livers of hCAR-TG Mice was not able to increase Cyp2b10 expression in jejunums of
The expression of hCAR SVs was determined in livers of WT mice. Moreover, the basal expression of the CAR-target
male hCAR-TG mice at ages of 2 days before birth (−2 days), gene Cyp2b10 was also higher in jejunums of WT mice than
5, 20, and 60 days. hCAR mRNA SV0, 1, 2, 3, and 4 were detect- those in hCAR-TG mice, which is consistent with the higher
able in fetal livers of hCAR-TG mice. The expression of these expression of CAR in WT mice than in hCAR-TG mice. These
hCAR SVs was two- to seven-fold higher in neonates (5-day-old data indicate that, in contrast to the liver, in jejunums of the
hCAR-TG mice) livers than in fetal livers (Fig. 4A). Whereas hCAR-TG mice, hCAR is expressed at very low levels and the
the expression of SV1, SV2, SV3, and SV4 remained stable CAR-target genes are not inducible by hCAR activators.
from 5 to 60 days of age, the WT SV0 further increased in adult
mice (60 days old) compared with adolescents (20 days old). Gender Divergence of hCAR mRNA Expression and
Cyp2b10 is the prototypical target gene of CAR in mouse Transcriptional Function
livers. The mRNA of Cyp2b10 was determined along with Male and female hCAR-TG mice expressed similar levels
hCAR SV0 in male hCAR-TG mice at ages of 2 days before of each of the five SVs of hCAR mRNA in their livers (Fig. 6,
birth, 5, 10, 15, 20, and 60 days. As shown in Figure 4B (left upper panel). Female hCAR-TG mice expressed about onefold
panels), two major increases of hCAR SV0 were observed at more Cyp2b10 mRNA than male hCAR-TG mice, although
5 days (newborn) and 60 days (adult). In comparison, Cyp2b10 the difference was not statistically significant. The induction
146 Zhang, Lu, And Klaassen

FIG. 3.  Comparison of prototypical CAR-target genes induced by CAR activators in the liver. Male adult wild-type (WT) and hCAR-TG (TG) mice were
treated with corn oil (CO), TCPOBOP (TCP; mCAR agonist), CITCO (CIT; hCAR agonist), and PB. (A) Messenger RNA of hCAR, mCAR, and two CAR-target
genes, Cyp2b10 and Cyp3a11, in the liver. (B) Western blotting showing increased accumulation of CAR protein in the nucleus and markedly increased expression
of Cyp2b10 protein in cytoplasm after treatment with respective CAR agonist in both genotypes. Representative blots were shown on the left, and blot density
analysis of 3 individual samples of each group was presented on the right. Values were presented as mean ± SEM; n = 3–5 for mRNA data and n = 3 for protein data.
Student’s t-test (two sided) was performed. *Statistical differences between inducer-treated and corn oil–treated control groups within the same genotype, p < 0.05.

of Cyp2b10 by TCPOBOP in livers of WT mice and its induc- assays (DeKeyser et al., 2009; Dring et al., 2010; Omiecinski
tion by CITCO in hCAR-TG mice were both more profound et al., 2011). To obtain insights into the in vivo functions and
in females than in males (Fig. 6, lower panel). The hCAR-TG expression profiles of hCAR SVs, as well as to overcome the
mice appear to retain the female-predominant pattern of CAR marked species differences between human and mouse, this
activation in liver. study engineered a new line of hCAR transgenic mice utilizing
the BAC-transgenic technology.
Discussion Alternative splicing of mRNA is a common phenomenon in
mammals. The proper transcription and alternative splicing of
CAR together with its closest mammalian relative, the genes require the presence of not only the coding region of the
pregnane X receptor, are nuclear receptors that regulate hepatic gene, but also the promoter, untranslated regions, and even distant
genes that are responsible for the biotransformation and regulatory elements (Luco et al., 2010, 2011). The first hCAR-
distribution of drugs. Recently, numerous SVs of hCAR mRNA transgenic mouse was engineered by integrating hCAR cDNA
have been identified (Lamba et  al., 2004) and some of them (SV0) into the mouse genome (Zhang et al., 2002). Later, another
have been shown to have transcriptional activity by in vitro line of hCAR-transgenic mice was developed by knockin of the
In Vivo Study of Human Car Splicing Variants 147

higher level in human livers than in hCAR-TG mice livers. This


might be due to the small sample size we used (n = 5), consider-
ing the significant interindividual differences in hCAR expres-
sion observed in both humans and hCAR-TG mice. The hCAR
knockin mice, in which the hCAR gene is regulated by the
mCAR promoter, were also indicated to express hCAR SVs 2,
3, 4, and 9, although their expression levels were not specified
(Scheer et al., 2008). Later work demonstrated that the hCAR-
knockin mice expressed hCAR SV0 (CAR1), SV1 (CAR2), and
SV2 (CAR3) at similar levels to those in human livers (Ross
et al., 2010). The data from the hCAR knockin mice suggest
that hCAR gene-coding region contains necessary elements for
the pre-mRNA editing. However, to what extent these elements
within the hCAR gene and those outside of the coding region
contribute to hCAR alternative splicing is unknown. Comparing
the expression profiles of a full spectrum of hCAR SVs in the
hCAR-TG mice developed by this study and the hCAR knockin
mice will shed light on this topic.
Species-specific activation of CAR by certain chemicals is
a well-known phenomenon. In agreement with previous stud-
ies (Scheer et al., 2008), hCAR-TG mice expressed increased
amounts of Cyp2b10 mRNA and protein in liver in response
to the hCAR-specific ligand, CITCO, and the common CAR
activator, PB, but not to the mCAR-specific ligand, TCPOBOP
(Fig.  3). In WT mice, the opposite is the case; CAR is acti-
vated by TCPOBOP and PB, but not by CITCO. Therefore, the
hCAR-TG mice appear to mimic the responses of humans to
various CAR activators.
Differences in the potency of induction by CAR activators
were observed between WT and hCAR-TG mice (Fig. 3A). The
FIG.  4.  Developmental expression of hCAR SVs, Cyp2b10, and other
induction of Cyp2b10 and 3a11 by CITCO (20 mg/kg/day for 3
human genes in livers of male hCAR-TG mice. (A) The expression pattern of continuous days) in livers of hCAR-TG mice was ~40% lower
major hCAR mRNA SVs during liver development. (B) Developmental expres- than that by TCPOBOP (3 mg/kg for a single dose) in WT mice.
sion profiles of hCAR SV0, the prototypical CAR target gene, Cyp2b10, as well In contrast, PB (100 mg/kg/day for 3 continuous days) induced
as human genes hAPOA2 and hPPOX in livers of hCAR-TG mice. Values are both Cyp2b10 and Cyp3a11 to higher levels in hCAR-TG mice
presented as mean ± SEM; n = 3–5.
than in WT mice. At dosages used in this study, TCPOBOP
is a more effective activator for mCAR in WT mice, whereas
coding region (exons 2–9) of hCAR gene into the mCAR gene PB, instead of CITCO, is the stronger activator for hCAR in
allele, where hCAR replaced the endogenous mCAR gene and hCAR-TG mice. Higher doses of CITCO could be tested in the
was driven by the endogenous mCAR promoter (Scheer et  al., hCAR-TG mice in future studies. However, 50 mg/kg CITCO
2008). Obviously, neither of these lines of hCAR-transgenic was shown to nonspecifically induce Cyp2b10 in WT mice in a
mice contains the necessary human regulatory elements for the previous report (Scheer et al., 2008).
hCAR gene. In comparison, the hCAR-TG (hCAR+/+/mCAR−/−) It has been reported that CAR mRNA was expressed at
mice described in this study contain the full-length hCAR gene, low levels in fetal livers but increased after birth in humans
including the noncoding exon 1, as well as flanking fragments (Huang et al., 2003; Vyhlidal et al., 2006). Consistent with the
(73-kbp upstream and 91-kbp downstream) from the human gen- ontogenetic expression pattern of CAR in human livers, major
ome (Fig. 1). Therefore, the hCAR-TG mice engineered by this SVs of hCAR mRNA were expressed at low levels in livers of
study are more likely to recapitulate the in vivo transcriptional hCAR-TG fetus and markedly increased in postnatal livers of
regulation of the CAR gene in humans. hCAR-TG mice (Fig. 4A). The WT hCAR mRNA SV0 but not
The hepatic expression profiles of major SVs of hCAR in the the other hCAR SVs further increased when the hCAR-TG mice
hCAR-TG mice, namely SV0, SV1, SV2, and SV3, were com- matured at 60 days of age. The hepatic expression of Cyp2b10
parable to those in humans, suggesting that the pre-mRNA pro- mRNA, the prototypical CAR target gene, constantly increased
cessing of hCAR in hCAR-TG mice resembles that in humans during the maturation of hCAR-TG mice, including the two
(Fig. 2). SV4, a less-investigated hCAR SV, is expressed at a time points when hCAR SV0 increased. These data suggest that
148 Zhang, Lu, And Klaassen

FIG. 5.  Tissue distribution of hCAR SVs in adult male hCAR-TG mice and species-specific activation of CAR in jejunum. (A) Tissue distribution of various
hCAR SVs. Total hCAR was the sum of all five SVs in each tissue. Values are presented as mean ± SEM; n = 4–5. (B) The expression of hCAR and mCAR as well
as CAR-target gene, Cyp2b10, in jejunums of wild-type (WT) mice treated with TCPOBOP (TCP) and PB, as well as hCAR-TG (TG) mice treated with CITCO
(CIT) and PB. Values were presented as mean ± SEM; n = 3–5. Student’s t-test (two sided) was performed. *Statistical differences between inducer-treated and
corn oil–treated control groups within the same genotype, p < 0.05.

the expression of CAR-target genes during liver maturation mice reflected the transition of fetal liver from a hematopoietic
could be affected not only by the amount of CAR, but also by organ to a metabolic one around birth.
factors that could potentially activate CAR, as well as by other In mice, CAR mRNA is detected in several tissues with the
transcription factors. highest expression in liver and small intestine (Petrick and
In addition to hCAR, other human genes on the BAC clone Klaassen, 2007). In comparison, CAR mRNA is expressed at
were also expressed in livers of the hCAR-TG mice (Fig. 4B). a much higher level in liver than other tissues in humans (Baes
The hPPOX gene, encoding an enzyme involved in heme et al., 1994). In the hCAR-TG mice, the major SVs of hCAR
production, was expressed higher in livers of hCAR-TG mice mRNA were highly expressed in liver, but only at low levels in
before birth. The mRNA of the lipid metabolism-related gene, small intestine and other tissues (Fig. 5). Thus, the tissue dis-
APOA2, in contrast, increased after birth. It is known that tribution of hCAR in the hCAR-TG mice resembles the human
the hematopoietic cell transcriptional signature is dominant, pattern instead of the mouse pattern. Moreover, the low expres-
whereas most metabolic pathways are underexpressed in fetal sion of hCAR in jejunum was not sufficient to respond to the
livers (Lee et al., 2012). The developmental expression profiles hCAR-specific agonist, CITCO, to induce Cyp2b10 expression
of hPPOX, hAPOA2, hCAR, and Cyp2b10 in the hCAR-TG in the hCAR-TG mice. In summary, the temporal and spatial
In Vivo Study of Human Car Splicing Variants 149

in females (Fig. 6). After CITCO treatment, Cyp2b10 was more


profoundly induced in livers of female than in male hCAR-TG
mice, which resembles the female-predominant induction of
Cyp2b10 by TCPOBOP in WT mice. Therefore, the activation
of CAR is female-predominant in the hCAR-TG mice.
In summary, this study established a novel hCAR-transgenic
mouse model that resembles the hepatic expression profiles of
major hCAR SVs in humans in aspects including expression
level, ontogeny, tissue distribution, and ligand specificity. This
model is extremely useful for in vivo research on the function
and regulation of this important human xenobiotic sensor, espe-
cially, for studies on in vivo roles of hCAR SVs in metabolizing
environmental toxicants and pharmaceutical drugs.

Funding

National Institute of Health [DK-081461, ES-019487, and


ES-009649 to C.D.K.; and 5P20RR024214-05 to Y.K.Z.].

Acknowledgments

The authors would like to thank the Transgenic and Gene-


targeting Institutional Facility and Dr. Kenneth Peterson in the
Biochemistry Department at KUMC for helping to develop the
transgenic mice, Drs. Dan Li, Andrew Lickteig, and Jie Liu for
FIG.  6.  Gender differences in hepatic expression of hCAR mRNA SVs their assistance in harvesting tissues, as well as colleagues in Dr.
and Cyp2b10 in hCAR-TG mice. Upper panel: major hCAR SVs and prototypi- Klaassen’s laboratory for critically reviewing the manuscript.
cal CAR target gene, Cyp2b10, in livers of naïve male and female hCAR-TG
mice (8 weeks old). Lower panel: the induction of Cyp2b10 mRNA in wild-
type (WT) and hCAR-TG (TG) mice by mCAR agonist, TCPOBOP (TCP),
References
and hCAR agonist CITCO (CIT), respectively, in both genders. Control groups
were treated with corn oil (CO). Values are presented as mean ± SEM; n = 4–5.
Assem, M., Schuetz, E. G., Leggas, M., Sun, D., Yasuda, K., Reid, G., Zelcer,
Two-way ANOVA was performed with treatment and gender as main factors.
N., Adachi, M., Strom, S., Evans, R. M., et al. (2004). Interactions between
Significant main effects and interactions were followed by Student-Newman-
hepatic Mrp4 and Sult2a as revealed by the constitutive androstane receptor
Keuls comparisons to assess the differences between groups. *Statistical differ-
and Mrp4 knockout mice. J. Biol. Chem. 279, 22250–22257.
ences between inducer-treated and corn oil–treated control groups; #Statistical
differences between male and female mice; p < 0.05. Auerbach, S. S., Stoner, M. A., Su, S., and Omiecinski, C. J. (2005). Retinoid X
receptor-alpha-dependent transactivation by a naturally occurring structural
variant of human constitutive androstane receptor (NR1I3). Mol. Pharmacol.
expression patterns of the hCAR gene indicate that the trans- 68, 1239–1253.
gene likely contains all the functional regulatory elements Baes, M., Gulick, T., Choi, H. S., Martinoli, M. G., Simha, D., and Moore, D.
controlling the humanized expression of the hCAR gene in the D. (1994). A new orphan member of the nuclear hormone receptor superfam-
hCAR-TG mice. ily that interacts with a subset of retinoic acid response elements. Mol. Cell.
Female-predominant expression and activity of CAR have Biol. 14, 1544–1552.
been documented in mice (Ledda-Columbano et al., 2003). In Choi, H. S., Chung, M., Tzameli, I., Simha, D., Lee, Y. K., Seol, W., and Moore,
D. D. (1997). Differential transactivation by two isoforms of the orphan
humans, hepatic mRNA expression of both CAR and CYP2B6, nuclear hormone receptor CAR. J. Biol. Chem. 272, 23565–23571.
the human homologue of the mouse Cyp2b10, tend to be higher DeKeyser, J. G., Stagliano, M. C., Auerbach, S. S., Prabhu, K. S., Jones, A. D.,
in females than in males in certain ethnicities but not others and Omiecinski, C. J. (2009). Di(2-ethylhexyl) phthalate is a highly potent
(Lamba et al., 2003). Marked interindividual variation in CAR agonist for the human constitutive androstane receptor splice variant CAR2.
mRNA expression was observed in humans. The lower induc- Mol. Pharmacol. 75, 1005–1013.
tion of Cyp2b10 by CAR activators in males was postulated to Dring, A. M., Anderson, L. E., Qamar, S., and Stoner, M. A. (2010). Rational
be due to the male predominant hormone, androstanes, which quantitative structure-activity relationship (RQSAR) screen for PXR and
CAR isoform-specific nuclear receptor ligands. Chem. Biol. Interact. 188,
function as endogenous antagonists of CAR (Forman et  al., 512–525.
1998). This study shows that the five SVs of hCAR mRNA Forman, B. M., Tzameli, I., Choi, H. S., Chen, J., Simha, D., Seol, W., Evans,
were expressed at similar levels in livers of male and female R. M., and Moore, D. D. (1998). Androstane metabolites bind to and deacti-
hCAR-TG mice, whereas Cyp2b10 mRNA tended to be higher vate the nuclear receptor CAR-beta. Nature 395, 612–615.
150 Zhang, Lu, And Klaassen

Haurogne, K., Bach, J. M., and Lieubeau, B. (2007). Easy and rapid method of Luco, R. F., Allo, M., Schor, I. E., Kornblihtt, A. R., and Misteli, T. (2011).
zygosity determination in transgenic mice by SYBR Green real-time quanti- Epigenetics in alternative pre-mRNA splicing. Cell 144, 16–26.
tative PCR with a simple data analysis. Transgenic Res. 16, 127–131. Luco, R. F., Pan, Q., Tominaga, K., Blencowe, B. J., Pereira-Smith, O. M., and
Hernandez, J. P., Mota, L. C., and Baldwin, W. S. (2009). Activation of CAR Misteli, T. (2010). Regulation of alternative splicing by histone modifica-
and PXR by dietary, environmental and occupational chemicals alters tions. Science 327, 996–1000.
drug metabolism, intermediary metabolism, and cell proliferation. Curr. Nims, R. W., Beebe, L. E., Utermahlen, W. E. Jr., Thomas, P. E., and Lubet, R.
Pharmacogenomics Person. Med. 7, 81–105. A. (1999). Induction of hepatic cytochromes P450 by dietary 1,4-bis[2-(3,5-
Honkakoski, P., Zelko, I., Sueyoshi, T., and Negishi, M. (1998). The nuclear dichloropyridyloxy)]benzene (TCPOBOP) in the B6C3F1 mouse: Dose-
orphan receptor CAR-retinoid X receptor heterodimer activates the pheno- dependence and pharmacodynamics. Int J Toxicol 18, 123–130.
barbital-responsive enhancer module of the CYP2B gene. Mol. Cell. Biol. Omiecinski, C. J., Coslo, D. M., Chen, T., Laurenzana, E. M., and Peffer, R. C.
18, 5652–5658. (2011). Multi-species analyses of direct activators of the constitutive andros-
Huang, W., Zhang, J., Chua, S. S., Qatanani, M., Han, Y., Granata, R., and tane receptor. Toxicol. Sci. 123, 550–562.
Moore, D. D. (2003). Induction of bilirubin clearance by the constitutive Petrick, J. S., and Klaassen, C. D. (2007). Importance of hepatic induction of
androstane receptor (CAR). Proc. Natl. Acad. Sci. U.S.A. 100, 4156–4161. constitutive androstane receptor and other transcription factors that regulate
Huang, W., Zhang, J., Wei, P., Schrader, W. T., and Moore, D. D. (2004). Meclizine xenobiotic metabolism and transport. Drug Metab. Dispos. 35, 1806–1815.
is an agonist ligand for mouse constitutive androstane receptor (CAR) and an Ross, J., Plummer, S. M., Rode, A., Scheer, N., Bower, C. C., Vogel, O.,
inverse agonist for human CAR. Mol. Endocrinol. 18, 2402–2408. Henderson, C. J., Wolf, C. R., and Elcombe, C. R. (2010). Human constitu-
Jinno, H., Tanaka-Kagawa, T., Hanioka, N., Ishida, S., Saeki, M., Soyama, A., tive androstane receptor (CAR) and pregnane X receptor (PXR) support the
Itoda, M., Nishimura, T., Saito, Y., Ozawa, S., et al. (2004). Identification of hypertrophic but not the hyperplastic response to the murine nongenotoxic
novel alternative splice variants of human constitutive androstane receptor hepatocarcinogens phenobarbital and chlordane in vivo. Toxicol. Sci. 116,
and characterization of their expression in the liver. Mol. Pharmacol. 65, 452–466.
496–502. Scheer, N., Ross, J., Rode, A., Zevnik, B., Niehaves, S., Faust, N., and Wolf,
Lamba, J. K., Lamba, V., Yasuda, K., Lin, Y. S., Assem, M., Thompson, E., C. R. (2008). A novel panel of mouse models to evaluate the role of human
Strom, S., and Schuetz, E. (2004). Expression of constitutive androstane pregnane X receptor and constitutive androstane receptor in drug response.
receptor splice variants in human tissues and their functional consequences. J. Clin. Invest. 118, 3228–3239.
J. Pharmacol. Exp. Ther. 311, 811–821. Sugatani, J., Kojima, H., Ueda, A., Kakizaki, S., Yoshinari, K., Gong, Q. H.,
Lamba, V., Lamba, J., Yasuda, K., Strom, S., Davila, J., Hancock, M. L., Owens, I. S., Negishi, M., and Sueyoshi, T. (2001). The phenobarbital response
Fackenthal, J. D., Rogan, P. K., Ring, B., Wrighton, S. A., et  al. (2003). enhancer module in the human bilirubin UDP-glucuronosyltransferase
Hepatic CYP2B6 expression: Gender and ethnic differences and relationship UGT1A1 gene and regulation by the nuclear receptor CAR. Hepatology 33,
to CYP2B6 genotype and CAR (constitutive androstane receptor) expres- 1232–1238.
sion. J. Pharmacol. Exp. Ther. 307, 906–922. Swales, K., and Negishi, M. (2004). CAR, driving into the future. Mol.
Ledda-Columbano, G. M., Pibiri, M., Concas, D., Molotzu, F., Simbula, Endocrinol. 18, 1589–1598.
G., Cossu, C., and Columbano, A. (2003). Sex difference in the prolif- Vyhlidal, C. A., Gaedigk, R., and Leeder, J. S. (2006). Nuclear receptor expres-
erative response of mouse hepatocytes to treatment with the CAR ligand, sion in fetal and pediatric liver: Correlation with CYP3A expression. Drug
TCPOBOP. Carcinogenesis 24, 1059–1065. Metab. Dispos. 34, 131–137.
Lee, J. S., Ward, W. O., Knapp, G., Ren, H., Vallanat, B., Abbott, B., Ho, K., Zhang, J., Huang, W., Chua, S. S., Wei, P., and Moore, D. D. (2002). Modulation
Karp, S. J., and Corton, J. C. (2012). Transcriptional ontogeny of the devel- of acetaminophen-induced hepatotoxicity by the xenobiotic receptor CAR.
oping liver. BMC Genomics 13, 33. Science 298, 422–424.

You might also like