Differential Regulation of Rat and Human CYP7A1 by the Nuclear Oxysterol Receptor Liver X Receptor-
Bryan Goodwin,
Michael A. Watson,
Hwajin Kim,
Ji Miao,
Jongsook Kim Kemper and
Steven A. Kliewer
Nuclear Receptor Discovery Research (B.G., M.A.W.), GlaxoSmithKline Research and Development, Research Triangle Park, North Carolina 27709; Department of Molecular & Integrative Physiology (H.K., J.M., J.K.K.), University of Illinois at Urbana-Champaign, Urbana, Illinois 61801; and Department of Molecular Biology (S.A.K.), University of Texas Southwestern Medical Center, Dallas, Texas 75390-8594
Address all correspondence and requests for reprints to: Bryan Goodwin, Nuclear Receptor Discovery Research, GlaxoSmithKline Research and Development, Research Triangle Park, North Carolina 27709, E-mail: bryan.j.goodwin{at}gsk.com, and/or Steven A. Kliewer, Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390-8594. E-mail: steven.kliewer{at}utsouthwestern.edu.
 |
ABSTRACT
|
---|
In rodent liver, transcription of the gene encoding cholesterol 7
-hydroxylase (CYP7A1), which catalyzes the rate-limiting step in the classic bile acid synthetic pathway, is stimulated by the liver X receptor
(LXR
), a nuclear receptor for oxysterol metabolites of cholesterol. This feed-forward regulatory loop provides a mechanism for the elimination of excess cholesterol from the body. In this report, we demonstrate that in primary cultures of human hepatocytes, activation of LXR
has the opposite effect, repressing CYP7A1 expression. This repression is mediated, at least in part, through induction of the orphan nuclear receptor, short heterodimer partner (SHP), which is also induced by bile acids. We demonstrate that SHP is regulated directly by LXR
through a DNA response element that overlaps with the previously characterized bile acid response element. Our data reveal a fundamental difference in the regulation of CYP7A1 in rodent and human hepatocytes and provide evidence that different species employ distinct molecular strategies to regulate cholesterol homeostasis.
 |
INTRODUCTION
|
---|
HIGH LEVELS OF circulating cholesterol are a major risk factor for the development of atherosclerosis, one of the largest causes of mortality in the industrialized world. The level of plasma cholesterol is determined by cholesterol absorption, synthesis, storage, and excretion (1). Diets rich in cholesterol can result in hypercholesterolemia in humans as well as other species, including hamsters, rabbits, and African Green monkeys (2, 3, 4, 5). Interestingly, rats and mice fed a high cholesterol diet are resistant to hypercholesterolemia (3, 5). The reasons for these differences in cholesterol sensitivity among species are not fully understood.
The conversion of cholesterol to bile acids in the liver represents an important pathway for the elimination of cholesterol from the body. The first and rate-limiting step in the classic pathway for bile acid biosynthesis involves the 7
-hydroxylation of cholesterol, a reaction that is catalyzed by cytochrome P450 (CYP) 7A1. Because of the intrinsic toxicity of bile acids, their synthesis must be tightly regulated (5, 6, 7, 8). Bile acids feed back and suppress CYP7A1 transcription through a mechanism mediated in part by the farnesoid X receptor (FXR; NR1H4), a member of the nuclear receptor family of ligand-activated transcription factors (8). Suppression of CYP7A1 by FXR activation is not direct. Rather, the heterodimer formed between FXR and the 9-cis retinoic acid receptor-
(RXR
; NR2B1) induces the expression of small heterodimer partner (SHP; NR0B2), an atypical orphan nuclear receptor that lacks a DNA binding domain; SHP, in turn, represses the activity of liver receptor homolog 1 (NR5A2), which binds to the proximal CYP7A1 gene promoter (9, 10). In addition to this nuclear receptor-signaling cascade, there is also recent evidence for SHP-independent mechanisms for suppression of CYP7A1 expression (11, 12). Thus, feedback regulation of bile acid synthesis involves multiple signaling pathways.
In mice and rats, the expression of CYP7A1 is induced by high-cholesterol diets. Recent studies have demonstrated that this feed-forward regulation is mediated through the nuclear liver X receptor
(LXR
: NR1H3), which is activated by oxysterol metabolites of cholesterol (13, 14) and binds as an RXR heterodimer to a response element located in the proximal promoter of the CYP7A1 gene. LXR
-null mice fed a high-cholesterol diet accumulate copious amounts of cholesterol in their livers and eventually succumb to liver failure (15). In addition to CYP7A1, LXR
regulates a number of genes involved in cholesterol and/or lipid homeostasis including the ATP-binding cassette proteins (ABC) A1, G1, G4, G5, and G8, apolipoprotein E, cholesterol ester transport protein, lipoprotein lipase, fatty acid synthase, and the sterol-regulatory element binding protein 1 (SREBP-1) (16, 17, 18, 19). Thus, LXR
provides a mechanism for sensing cholesterol levels and regulating genes involved in cholesterol homeostasis accordingly.
The regulation of CYP7A1 by cholesterol varies across species. Thus, cholesterol induces CYP7A1 expression in mice, rats, dogs, and certain species of nonhuman primates (3, 20, 21, 22, 23), all species that adapt to high-cholesterol diets with little change in plasma cholesterol levels. In contrast, cholesterol has little effect or even suppresses CYP7A1 in other species including rabbit, hamster, and African Green monkey (2, 3, 24). All of these species develop hypercholesterolemia in response to dietary cholesterol. The effects of cholesterol on the regulation of CYP7A1 are not well characterized in man, but dietary cholesterol does not increase bile acid production at least in certain subjects (25, 26, 27). In agreement with these findings, the LXR response element (LXRE) is not conserved in the human (h) CYP7A1 promoter (28), and the hCYP7A1 promoter is not induced by cholesterol feeding in transgenic mice harboring the gene (29). Thus, there appear to be important interspecies differences in the regulation of CYP7A1. In this report, we have directly compared the effects of LXR agonists on CYP7A1 expression in human and rodent hepatocytes.
 |
RESULTS
|
---|
LXR Agonists Repress CYP7A1 in Human Hepatocytes
To better understand the regulation of hepatic cholesterol metabolism in different species, we compared the effects of the natural LXR agonist 22(R)-hydroxycholesterol (13), the synthetic LXR-selective agonist T0901317 (30), and the FXR-selective agonist GW4064 (9) on gene expression in primary cultures of rat or human hepatocytes treated for 48 h. 22(S)-hydroxycholesterol, which does not activate LXR, was included as a negative control. Northern analysis was performed using probes for either CYP7A1, SHP, or SREBP-1. As expected, treatment of rat hepatocytes with the selective LXR agonist T0901317 resulted in a robust induction of CYP7A1 (
9-fold) and SREBP-1 (
8-fold) mRNA levels (Fig. 1
, A and E, respectively). In contrast, the natural LXR agonist 22(R)-hydroxycholesterol had only very modest effects on CYP7A1 and SREBP-1 expression in the rat hepatocytes (Fig. 1
, A and E, respectively). Although the reason for this lack of activity is not clear, 22(R)-hydroxycholesterol may be either metabolized or inefficiently taken up by primary rat hepatocytes. Neither T0901317 nor 22(R)-hydroxycholesterol had any effect on SHP expression in rat hepatocytes (Fig. 1C
). As expected, treatment of rat hepatocytes with the FXR agonist GW4064 resulted in a marked induction of SHP (
70-fold) (Fig. 1C
) and complete suppression of CYP7A1 (Fig. 1A
). Thus, the regulation of SREBP-1, SHP, and CYP7A1 by LXR and FXR is recapitulated in primary cultures of rat hepatocytes.

View larger version (34K):
[in this window]
[in a new window]
|
Figure 1. Expression of CYP7A1, SHP, and SREBP-1 mRNAs in Primary Cultures of Rat and Human Hepatocytes Treated with LXR and FXR Activators
Primary cultures of rat (A, C, E, and G) and human (B, D, F, and H) hepatocytes were treated with 1) vehicle alone (0.1% dimethylsulfoxide), 2) 22(S)-hydroxycholesterol [22(S)-OH, 10 µM], 3) 22(R)-hydroxycholesterol [22(R)-OH, 10 µM], 4) T0901317 (1 µM), and 5) GW4064 (1 µM) for 48 h before harvest and isolation of total RNA. Northern analysis was performed using probes for CYP7A1 (A and B), SHP (C and D), and SREBP-1 (E and F) as described in Materials and Methods. Signal intensity was determined using a PhosphorImager (Molecular Dynamics, Inc.) and the relative mRNA abundance is shown graphically below each blot. To ensure equal loading of RNA samples, the blots were probed with radiolabeled cDNA corresponding to 18S rRNA (G and H).
|
|
The same experiments were performed in parallel in primary cultures of human hepatocytes. As previously reported, treatment with the FXR agonist GW4064 strongly induced SHP mRNA levels (7.5-fold) and effectively suppressed CYP7A1 expression in human hepatocytes (Fig. 1
, B and D). Thus, FXR agonists have identical effects on SHP and CYP7A1 expression in human and rodent hepatocytes. Surprisingly, treatment of human hepatocytes with either the natural LXR agonist 22(R)-hydroxycholesterol or the synthetic agonist T0901317 also resulted in the induction of SHP expression that was paralleled by a repression of CYP7A1 expression (Fig. 1
, B and D). In time course experiments, the effects of T0901317 on SHP and CYP7A1 expression were observed as early as 6 h after treatment (data not shown). In control experiments, 22(S)-hydroxycholesterol had virtually no effect on either CYP7A1 or SHP expression in the human liver cells (Fig. 1
, B and D, respectively). As expected, expression of the LXR target gene SREBP-1 was highly inducible by both 22(R)-hydroxycholesterol (
6-fold) and T0901317 (
8-fold) (Fig. 1F
). Expression of the bile salt export pump (ABCB11), an FXR target gene (31, 32), was induced by the FXR agonist GW4064 but not the LXR ligand T0901317 (data not shown). Similar effects of LXR agonists on gene expression were seen in three independent experiments performed using hepatocytes derived from different donors (data not shown). Thus, LXR agonists have completely different effects on SHP and CYP7A1 expression in human vs. rodent hepatocytes.
LXR
Activates the hSHP Promoter
We next examined whether LXR
directly regulates expression of the hSHP gene. Treatment of primary human hepatocytes with cycloheximide failed to block T0901317-dependent induction of hSHP expression (data not shown), indicating that LXR
directly regulates this gene. To further examine this issue, a reporter construct containing 571 bp of the hSHP promoter fused to the luciferase gene was cotransfected into human embryonal kidney cells (HEK293) either in the absence or presence of expression plasmids for hLXR
and/or hRXR
. T0901317 and/or the synthetic RXR agonist LG100268 had little or no effect on reporter activity in the absence of exogenously expressed LXR
and RXR
(Fig. 2A
). Reporter levels increased in response to expression of either receptor in the presence of its cognate ligand (Fig. 2A
). Introduction of both LXR
and RXR
in the absence of their ligands resulted in an approximately 5-fold increase in reporter activity (Fig. 2A
), which was stimulated an additional 2- to 4-fold by treatment with the LXR agonist T0901317 or the RXR agonist LG100268 (Fig. 2A
). Cotreatment with T0901317 and LG100268 resulted in additive effects on reporter activity (Fig. 2A
). Thus, the LXR
/RXR
heterodimer can directly activate the hSHP promoter.

View larger version (23K):
[in this window]
[in a new window]
|
Figure 2. Activation of the hSHP Promoter by LXR
A, The hSHP promoter (bases -571 to +10) was linked to a luciferase reporter gene (inset, panel A) and cotransfected into HEK293 cells in the presence of expression vectors for hLXR , hRXR , or both. After transfection, cells were treated with the LXR agonist T0901317 (1 µM), the RXR agonist LG100268 (1 µM), or both. Control transfections received vehicle (0.1% dimethylsulfoxide) alone. B, Identification of an LXRE in the hSHP promoter. Deletion mutants of the hSHP promoter were prepared as described in Materials and Methods and cotransfected into HEK293 cells with LXR and RXR expression vectors as indicated. Transfected cells were treated with either 1 µM LG100268 and T0901317 or vehicle alone. The pGL3-hSHP-413 LXRE plasmid harbors a mutated LXRE, whereas the pGL3-(hSHP-LXRE)-tk-Luc construct contains a single copy of the putative LXRE linked to a minimal tk promoter (bases -105 to +51) and a luciferase reporter gene. Cells were treated for 24 h before harvest and determination of luciferase and SPAP activity. Luciferase activities are normalized to SPAP and all data represent the mean ± SD of three individual transfections. Values are expressed as fold activation compared with the vehicle-treated, no-receptor control for each construct.
|
|
To delineate the LXR
/RXR
responsive region, we tested a series of deletion mutants in the hSHP promoter. Deletion to -288 did not reduce the responsiveness of the hSHP promoter to the LXR
/RXR
complex (Fig. 2B
, pGL-hSHP-288). However, further deletion to -266 essentially abolished LXR
/RXR
activity on the promoter (Fig. 2B
, pGL-hSHP-266). Within the region extending from -266 to -288, we identified an imperfect DR-4 motif with similarity to previously characterized LXR
/RXR
binding sites (Fig. 3A
). Interestingly, this motif shares a half-site with the previously characterized FXR/RXR binding site (inset, Fig. 2A
), which confers regulation by bile acids on the hSHP promoter. Site-directed mutagenesis of the DR-4 motif markedly decreased the response of the hSHP promoter to LXR
/RXR
(Fig. 2B
, pGL-hSHP-413
LXRE). Moreover, a single copy of this response element was sufficient to confer responsiveness to LXR
/RXR
on the heterologous thymidine kinase (tk) promoter [Fig. 2B
, pGL3-(hSHP-LXRE)-tk-Luc]. These data demonstrate that the DR-4 motif functions as an LXRE in cell-based reporter assays.
We performed EMSAs to test whether the LXR
/RXR
heterodimer binds directly to the DR-4 element in the hSHP promoter. As expected, LXR
bound efficiently as a heterodimer with RXR
to the LXRE from the rat CYP7A1 promoter (Fig. 3
, A and B). The LXR
/RXR
heterodimer also bound to the hSHP DR-4, albeit with lower affinity compared with the CYP7A1 LXRE (Fig. 3
, B and C). These data indicate that LXR
directly regulates expression of the hSHP by binding to a response element located in the promoter of the gene.
SHP Represses the hCYP7A1 Promoter
We next examined whether SHP was capable of repressing transcription of the hCYP7A1 gene in a manner analogous to that described for rat CYP7A1 (9, 10). Transfection of HepG2 cells with a SHP expression plasmid resulted in the repression of the endogenous CYP7A1 gene (Fig. 4
). These data suggest that induction of SHP contributes to the repression of CYP7A1 observed in primary cultures of human hepatocytes treated with LXR
agonists (Fig. 1
).

View larger version (28K):
[in this window]
[in a new window]
|
Figure 4. SHP Suppresses the hCYP7A1 Promoter
HepG2 cells were transfected with either control plasmid or SHP expression plasmid [Tf(SHP)], and SHP, CYP7A1, and ß-actin expression was measured by semiquantitative RT-PCR.
|
|
 |
DISCUSSION
|
---|
In rodents, the feed-forward regulation of CYP7A1 by oxysterols acting through LXR
results in increased catabolism of cholesterol to bile acids (16, 17, 19, 33). The importance of this pathway is highlighted in mice lacking LXR
: these animals accumulate copious amounts of cholesterol in their livers in response to a high-cholesterol diet (15). However, little is known about the role of LXR
in the regulation of hCYP7A1 gene. In this study, we compared the regulation of the rat and human CYP7A1 genes in primary hepatocytes. It was anticipated that LXR agonists might not induce CYP7A1 expression in human hepatocytes, since the LXR
/RXR
binding site is not conserved in the human gene promoter (28). Surprisingly, we found that CYP7A1 was effectively suppressed by LXR agonists in human hepatocytes. This repression of CYP7A1 was accompanied by a corresponding increase in the expression of SHP, which is known to be induced by bile acids and to contribute to the feedback repression of CYP7A1 in rodents (9, 10, 11, 12). In contrast, LXR agonists did not induce SHP expression in rats in vivo (data not shown) or in primary rat hepatocytes. These data indicate that there are fundamental differences in the regulation of CYP7A1 and cholesterol homeostasis in rodents and humans.
We speculate that the inhibition of CYP7A1 by LXR agonists may be a component of a broader physiological strategy to suppress the absorption of dietary cholesterol in the intestine. The obligate role of bile acids in the absorption of cholesterol from the intestinal lumen has been appreciated for more than 30 yr (34, 35, 36, 37). Decreases in bile acid pool size by either diversion from the intestinal lumen or sequestration with resins, such as cholestyramine, result in an inhibition of cholesterol absorption (25, 35, 38). Indeed, cholestyramine is reported to have utility in the treatment of patients with hypercholesterolemia (39). Additionally, targeted disruption of the murine genes encoding CYP7A1 or CYP27A1, two key enzymes in bile acid production, results in a reduction in the bile acid pool and a concomitant decrease in the solubilization and absorption of intestinal cholesterol (40, 41, 42). Thus, the repression of CYP7A1 and bile acid synthesis by dietary cholesterol in human hepatocytes may contribute to an overall reduction in cholesterol load by blocking its absorption in the intestine.
Consistent with this hypothesis, previous studies demonstrated that a high-cholesterol diet reduced CYP7A1 mRNA levels and enzymatic activity in the livers of African Green monkeys, a species that was studied because of its similarities to humans in lipoprotein and cholesterol metabolism (2). Rudel et al. (2) showed that this decrease in bile acid production was accompanied by a reduction in intestinal cholesterol absorption. Similar reductions in CYP7A1 mRNA and enzymatic activity after cholesterol feeding have been reported in other species, including hamsters and rabbits (3, 24). To date, little is known about the regulation of hCYP7A1 by cholesterol, and the overall response to dietary cholesterol consumption appears to be highly variable (26, 27, 43, 44, 45, 46, 47). However, there is evidence that bile acid synthesis does not increase and, in some individuals, the fraction of cholesterol absorbed decreases with increasing dietary cholesterol (26, 27, 44, 45).
Whereas rodents appear to cope with excess cholesterol by stimulating its conversion to bile acids for excretion from the body, African Green monkeys and perhaps other species, including humans, may have evolved an alternate strategy, in which the absorption of cholesterol in the intestine is reduced through decreases in bile acid production. These species differences may reflect the amount of cholesterol typically ingested in the diet, with primates having evolved on a lower cholesterol diet. Ultimately, the differences in the regulation of CYP7A1 may contribute to the propensity of humans and other species to develop hypercholesterolemia and other adverse events such as the formation of gall stones as a consequence of a high-cholesterol diet.
 |
MATERIALS AND METHODS
|
---|
Chemicals
22(R)-hydroxycholesterol, 22(S)-hydroxycholesterol, dexamethasone, and charcoal-stripped, delipidated calf serum were obtained from Sigma (St. Louis, MO). The human embryonal kidney cell line HEK293 was obtained from the American Type Culture Collection (ATCC no. CRL-1573, Manassas, VA). Unless otherwise stated, tissue culture reagents were purchased from Invitrogen (Carlsbad, CA). The synthetic LXR, RXR, and FXR agonists, T0901317 (30), LG100268 (48), and GW4064 (9), respectively, were synthesized in house.
Primary Culture of Rat and Human Hepatocytes and Northern Blot Analysis
Primary cultures of rat hepatocytes were prepared as described elsewhere (49). After isolation, hepatocytes were washed in DMEM supplemented with 5% fetal bovine serum, 1 µM dexamethasone, 4 µg/ml insulin, 50 U/ml penicillin, 50 µg/ml streptomycin, and 1.5 x 106 cells seeded into six-well plates coated with Matrigel (Becton Dickinson and Co., Franklin Lakes, NJ). Cells were allowed to attach for 4 h and thereafter maintained in serum-free Williams E medium containing 100 nM dexamethasone, 50 U/ml penicillin, 50 µg/ml streptomycin, and insulin-transferrin-selenium (Invitrogen). Primary human hepatocytes were obtained from BioWhittaker, Inc. (Walkersville, MD) and were similarly maintained in serum-free Williams E medium containing the supplements outlined above. Subsequently, cells were maintained and treated exactly as previously described (9). Total RNA was isolated and 10 µg resolved on a 1% agarose/2.2 M formaldehyde denaturing gel and transferred to a nylon membrane (Hybond N+, Pharmacia Biotech, Piscataway, NJ). Blots were sequentially hybridized with 32P-labeled cDNAs corresponding to hCYP7A1 (bases 991564, GenBank accession no. M93133), hSHP (GenBank accession no. L76571), and hSREBP-1 (bases 403-1204, GenBank accession no. NM 004176), or rat CYP7A1 (bases 235460, GenBank accession no. J05460), mouse SHP (bases 30783, GenBank accession no. L76567), mouse SREBP-1 (bases 969-1645, GenBank accession no. AF374266). To ensure equal loading of RNA samples, the blots were probed with radiolabeled cDNA corresponding to the rat 18S rRNA (bases 293970, GenBank accession no. X01117). The relative signal intensity for Northern blots was determined using a PhosphorImager (Molecular Dynamics, Inc., Sunnyvale, CA).
Plasmid Constructs
The mammalian expression vectors pcDNA3-hLXR
and pcDNA3-hRXR
were generated by subcloning the hLXR
and hRXR
open-reading frames from pSG5-hLXR
and pSG5-hRXR
(14), respectively, into pcDNA3.1(+) (Invitrogen). Preparation of the hSHP-luciferase reporter gene construct pGL3-hSHP-571 was previously described (9). 5'-Deletion mutants of this construct, namely pGL3-hSHP-413, pGL3-hSHP-288, pGL3-hSHP-266, and pGL3-hSHP-105, which contain bases -413 to +10, -288 to +10, -266 to +10, and -105 to +10, respectively, of the hSHP promoter inserted into the BglII site of the promoterless luciferase reporter vector pGL3-Basic were generated by PCR. Site-directed mutagenesis of the putative LXRE in the hSHP promoter was performed using the Transformer mutagenesis system (CLONTECH Laboratories, Inc., Palo Alto, CA) with the
LXRE (bases -294 to -258, 5'-CCTGAGTTAATGttCTTGTTTAggCACTTGAGTCATC). Mutated constructs were verified to be free of nonspecific base changes by sequencing. pß-actin-SPAP, an expression vector containing the human secreted placental alkaline phosphatase (SPAP) cDNA under the control of ß-actin promoter, was used as an internal control in all transfections.
Transient Transfection Assays
HEK293 cells were maintained in MEM containing 10% fetal bovine serum (Irvine Scientific, Santa Ana, CA), 50 U/ml penicillin, and 50 µg/ml streptomycin. Transient transfections were performed in 96-well plates using the FuGene6 transfection reagent (Roche Molecular Biochemicals, Indianapolis, IN) according to the manufacturers instructions. Typically, transfection mixes contained 10 ng SHP reporter gene, 2 ng pß-actin-SPAP as an internal control, 5 ng pcDNA3-hLXR
, and 3 ng pcDNA3-hRXR
. The final amount of plasmid DNA was adjusted to 80 ng with pBluescript II SK(+) (Stratagene, La Jolla, CA).
EMSAs
EMSAs were performed as described elsewhere (9). Competitor oligonucleotides were added at 25-, 100-, or 500-fold molar excess. The binding reactions were resolved on a preelectrophoresed 0.25x Tris-borate-EDTA, 4% polyacrylamide gel at room temperature. Human LXR
and RXR
proteins were synthesized from pSG5-hLXR
and pSG5-RXR
, respectively, using the TNT T7-coupled reticulocyte system (Promega Corp., Madison, WI). The following double-stranded oligonucleotides were used as probes and competitors in EMSA: rat CYP7A1 DR4, 5'-AGCTCTTTGGTCACTCAAGTTCAAGT-3'; hSHP DR4, 5'-AGCTTAATGACCTTGTTTATCCACTT-3'.
RT-PCR Analysis
HepG2 cells were grown in phenol red-free DMEM supplemented with 10% (vol/vol) charcoal dextran-stripped fetal bovine serum, 100 U/ml of penicillin, and 0.01% streptomycin. For transfection assays, confluent cells grown in six-well plates were transfected with 1 µg of either cytomegalovirus flag-hSHP or cytomegalovirus-flag plasmid (Stratagene) using Lipofectamine 2000 (Invitrogen) as directed by the manufacturer. Total RNA was isolated at 2436 h after transfection and subjected to RT-PCR analysis. Total RNA was isolated using Trizol (Life Technologies, Gaithersburg, MD), and cDNA was synthesized using oligo deoxythymidine and reverse transcriptase. hCYP7A1 and hSHP were measured by semiquantitative RT-PCR. ß-Actin served as an internal control for the RT-PCR analysis. The amplification of these mRNAs was linear up to 2530 cycles of PCR (data not shown). ß-Actin, CYP7A1, and SHP were amplified by 22, 28, and 25 cycles of PCR, respectively. The following primers were used to amplify CYP7A1, SHP, and ß-actin: CYP7A1 forward primer, 5'-CTAAGGAGGATTTCACTTGC-3'; CYP7A1 reverse primer, 5'-ACTGGTCCAAAGGTGGACAT-3'; SHP forward primer, 5'-CAAGAAGATTCTGCTGGAGG-3'; SHP reverse primer, 5'-GGATGTCAACATCTCCAATG-3'; ß-actin forward primer, 5'-GTCATCACCATTGGCAATGAG-3'; ß-actin reverse primer, 5'-CGTCATACTCCTGCTTGCTG-3'. The sizes of the CYP7A1, SHP, and ß-actin in PCR products were 350, 390, and 350 bp, respectively.
 |
ACKNOWLEDGMENTS
|
---|
We thank Dr. Larry Rudel for discussions, Drs. David Mangelsdorf and David Russell for their comments on the manuscript, Jason Holt and Cristin Galardi for assistance with transient transfections, and Dr. Heidi Kast for the rat 18S rRNA probe.
 |
FOOTNOTES
|
---|
Abbreviations: CYP, Cytochrome P450; CYP7A1, gene encoding cholesterol 7
-hydroxylase; FXR, farnesoid X receptor; h, human; HEK293, human embryonal kidney cell line; LXR, liver X receptor; LXRE, LXR response element; RXR
, 9-cis-retinoic acid receptor-
; SHP, short heterodimer partner; SPAP, secreted placental alkaline phosphatase; SREBP-1, sterol-regulatory element-binding protein 1; tk, thymidine kinase.
Received for publication July 15, 2002.
Accepted for publication December 6, 2002.
 |
REFERENCES
|
---|
- Goldstein JL, Brown MS 1990 Regulation of the mevalonate pathway. Nature 343:425430[CrossRef][Medline]
- Rudel L, Deckelman C, Wilson M, Scobey M, Anderson R 1994 Dietary cholesterol and downregulation of cholesterol 7
-hydroxylase and cholesterol absorption in African green monkeys. J Clin Invest 93:24632472[Medline]
- Horton JD, Cuthbert JA, Spady DK 1995 Regulation of hepatic 7
-hydroxylase expression and response to dietary cholesterol in the rat and hamster. J Biol Chem 270:53815387[Abstract/Free Full Text]
- Nguyen LB, Xu G, Shefer S, Tint GS, Batta A, Salen G 1999 Comparative regulation of hepatic sterol 27- hydroxylase and cholesterol 7
-hydroxylase activities in the rat, guinea pig, and rabbit: effects of cholesterol and bile acids. Metabolism 48:15421548[Medline]
- Davis RA, Miyake JH, Hui TY, Spann NJ 2002 Regulation of cholesterol-7
-hydroxylase. Barely missing a shp. J Lipid Res 43:533543[Abstract/Free Full Text]
- Russell DW, Setchell KD 1992 Bile acid biosynthesis. Biochemistry 31:47374749[Medline]
- Chiang JYL 1998 Regulation of bile acid synthesis. Front Biosci 3:D176D193
- Russell DW 1999 Nuclear orphan receptors control cholesterol catabolism. Cell 97:539542[Medline]
- Goodwin B, Jones SA, Price RR, Watson MA, McKee DD, Moore LB, Galardi C, Wilson JG, Lewis MC, Roth ME, Maloney PR, Willson TM, Kliewer SA 2000 A regulatory cascade of the nuclear receptors FXR, SHP-1, and LRH-1 represses bile acid biosynthesis. Mol Cell 6:517526[Medline]
- Lu TT, Makishima M, Repa JJ, Schoonjans K, Kerr TA, Auwerx J, Mangelsdorf DJ 2000 Molecular basis for feedback regulation of bile acid synthesis by nuclear receptors. Mol Cell 6:507515[Medline]
- Wang L, Lee YK, Bundman D, Han Y, Thevananther S, Kim CS, Chua SS, Wei P, Heyman RA, Karin M, Moore DD 2002 Redundant pathways for negative feedback regulation of bile acid production. Dev Cell 2:721731[Medline]
- Kerr TA, Saeki S, Schneider M, Schaefer K, Berdy S, Redder T, Shan B, Russell DW, Schwarz M 2002 Loss of nuclear receptor SHP impairs but does not eliminate negative feedback regulation of bile acid synthesis. Dev Cell 2:713720[Medline]
- Janowski BA, Willy PJ, Devi TR, Falck JR, Mangelsdorf DJ 1996 An oxysterol signalling pathway mediated by the nuclear receptor LXR
. Nature 383:728731[CrossRef][Medline]
- Lehmann JM, Kliewer SA, Moore LB, Smith-Oliver TA, Oliver BB, Su JL, Sundseth SS, Winegar DA, Blanchard DE, Spencer TA, Willson TM 1997 Activation of the nuclear receptor LXR by oxysterols defines a new hormone response pathway. J Biol Chem 272:31373140[Abstract/Free Full Text]
- Peet DJ, Turley SD, Ma W, Janowski BA, Lobaccaro JM, Hammer RE, Mangelsdorf DJ 1998 Cholesterol and bile acid metabolism are impaired in mice lacking the nuclear oxysterol receptor LXR
. Cell 93:693704[Medline]
- Laffitte BA, Tontonoz P 2002 Orphan nuclear receptors find a home in the arterial wall. Curr Atheroscler Rep 4:213221[Medline]
- Edwards PA, Kast HR, Anisfeld AM 2002 BAREing it all: the adoption of LXR and FXR and their roles in lipid homeostasis. J Lipid Res 43:212[Abstract/Free Full Text]
- Repa JJ, Berge KE, Pomajzl C, Richardson JA, Hobbs HH, Mangelsdorf DJ 2002 Regulation of ATP-binding cassette sterol transporters, ABCG5 and ABCG8, by the oxysterol receptors, LXR
and ß. J Biol Chem 277:1879318800[Abstract/Free Full Text]
- Tall AR, Costet P, Luo Y 2000 Orphans meet cholesterol. Nat Med 6:11041105[CrossRef][Medline]
- Lofland Jr HB, Clarkson TB, St Clair RW, Lehner ND 1972 Studies on the regulation of plasma cholesterol levels in squirrel monkeys of two genotypes. J Lipid Res 13:3947[Abstract/Free Full Text]
- Pertsemlidis D, Kirchman EH, Ahrens Jr EH 1973 Regulation of cholesterol metabolism in the dog. I. Effects of complete bile diversion and of cholesterol feeding on absorption, synthesis, accumulation, and excretion rates measured during life. J Clin Invest 52:23532367[Medline]
- Jelinek DF, Andersson S, Slaughter CA, Russell DW 1990 Cloning and regulation of cholesterol 7
-hydroxylase, the rate-limiting enzyme in bile acid biosynthesis. J Biol Chem 265:81908197[Abstract/Free Full Text]
- Pandak WM, Li YC, Chiang JY, Studer EJ, Gurley EC, Heuman DM, Vlahcevic ZR, Hylemon PB 1991 Regulation of cholesterol 7
-hydroxylase mRNA and transcriptional activity by taurocholate and cholesterol in the chronic biliary diverted rat. J Biol Chem 266:34163421[Abstract/Free Full Text]
- Xu G, Salen G, Shefer S, Ness GC, Nguyen LB, Parker TS, Chen TS, Zhao Z, Donnelly TM, Tint GS 1995 Unexpected inhibition of cholesterol 7
-hydroxylase by cholesterol in New Zealand white and Watanabe heritable hyperlipidemic rabbits. J Clin Invest 95:14971504[Medline]
- Grundy SM, Ahrens Jr EH, Davignon J 1969 The interaction of cholesterol absorption and cholesterol synthesis in man. J Lipid Res 10:304315[Abstract/Free Full Text]
- Nestel PJ, Poyser A 1976 Changes in cholesterol synthesis and excretion when cholesterol intake is increased. Metabolism 25:15911599[Medline]
- Kern Jr F 1994 Effects of dietary cholesterol on cholesterol and bile acid homeostasis in patients with cholesterol gallstones. J Clin Invest 93:11861194[Medline]
- Chiang JY, Kimmel R, Stroup D 2001 Regulation of cholesterol 7
-hydroxylase gene (CYP7A1) transcription by the liver orphan receptor (LXR
). Gene 262:257265[CrossRef][Medline]
- Agellon LB, Drover VA, Cheema SK, Gbaguidi GF, Walsh A 2002 Dietary cholesterol fails to stimulate the human cholesterol 7
-hydroxylase gene (CYP7A1) in transgenic mice. J Biol Chem 277:2013120134[Abstract/Free Full Text]
- Schultz JR, Tu H, Luk A, Repa JJ, Medina JC, Li L, Schwendner S, Wang S, Thoolen M, Mangelsdorf DJ, Lustig KD, Shan B 2000 Role of LXRs in control of lipogenesis. Genes Dev 14:28312838[Abstract/Free Full Text]
- Plass JR, Mol O, Heegsma J, Geuken M, Faber KN, Jansen PL, Muller M 2002 Farnesoid X receptor and bile salts are involved in transcriptional regulation of the gene encoding the human bile salt export pump. Hepatology 35:589596[CrossRef][Medline]
- Ananthanarayanan M, Balasubramanian N, Makishima M, Mangelsdorf DJ, Suchy FJ 2001 Human bile salt export pump promoter is transactivated by the farnesoid X receptor/bile acid receptor. J Biol Chem 276:2885728865[Abstract/Free Full Text]
- Repa JJ, Mangelsdorf DJ 1999 Nuclear receptor regulation of cholesterol and bile acid metabolism. Curr Opin Biotechnol 10:557563[CrossRef][Medline]
- Dietschy JM, Wilson JD 1970 Regulation of cholesterol metabolism. 2. N Engl J Med 282:11791183[Medline]
- Siperstein M, Chaikoff I, Reinhardt W 1952 Carbon14-cholesterol. V. Obligatory function of bile in intestinal absorption of cholesterol. J Biol Chem 198:111114[Free Full Text]
- Wilson MD, Rudel LL 1994 Review of cholesterol absorption with emphasis on dietary and biliary cholesterol. J Lipid Res 35:943955[Medline]
- Swell L, Trout E, Hopper Jr J, Field Jr H, Treadwell C 1958 Specific function of bile salts in cholesterol absorption. Proc Soc Exp Biol Med 98:174176
- Miettinen TA 1971 Relationship between faecal bile acids, absorption of fat and vitamin B 12, and serum lipids in patients with ileal resections. Eur J Clin Invest 1:452460[Medline]
- Ast M, Frishman WH 1990 Bile acid sequestrants. J Clin Pharmacol 30:99106[Abstract/Free Full Text]
- Schwarz M, Russell DW, Dietschy JM, Turley SD 1998 Marked reduction in bile acid synthesis in cholesterol 7
-hydroxylase-deficient mice does not lead to diminished tissue cholesterol turnover or to hypercholesterolemia. J Lipid Res 39:18331843[Abstract/Free Full Text]
- Schwarz M, Russell DW, Dietschy JM, Turley SD 2001 Alternate pathways of bile acid synthesis in the cholesterol 7
-hydroxylase knockout mouse are not upregulated by either cholesterol or cholestyramine feeding. J Lipid Res 42:15941603[Abstract/Free Full Text]
- Repa JJ, Lund EG, Horton JD, Leitersdorf E, Russell DW, Dietschy JM, Turley SD 2000 Disruption of the sterol 27-hydroxylase gene in mice results in hepatomegaly and hypertriglyceridemia. Reversal by cholic acid feeding. J Biol Chem 275:3968539692[Abstract/Free Full Text]
- Katan MB, Beynen AC 1987 Characteristics of human hypo- and hyperresponders to dietary cholesterol. Am J Epidemiol 125:387399[Abstract]
- McNamara DJ, Kolb R, Parker TS, Batwin H, Samuel P, Brown CD, Ahrens Jr EH 1987 Heterogeneity of cholesterol homeostasis in man. Response to changes in dietary fat quality and cholesterol quantity. J Clin Invest 79:17291739[Medline]
- Quintao E, Grundy SM, Ahrens Jr EH 1971 Effects of dietary cholesterol on the regulation of total body cholesterol in man. J Lipid Res 12:233247[Abstract/Free Full Text]
- Lin DS, Connor WE 1980 The long term effects of dietary cholesterol upon the plasma lipids, lipoproteins, cholesterol absorption, and the sterol balance in man: the demonstration of feedback inhibition of cholesterol biosynthesis and increased bile acid excretion. J Lipid Res 21:10421052[Abstract]
- Kern Jr F 1992 Altered cholesterol metabolism in patients with cholesterol gallstones: responses to reduced dietary cholesterol. Trans Am Clin Climatol Assoc 104:235240[Medline]
- Boehm MF, Zhang L, Zhi L, McClurg MR, Berger E, Wagoner M, Mais DE, Suto CM, Davies JA, Heyman RA, Nadzan AM 1995 Design and synthesis of potent retinoid X receptor selective ligands that induce apoptosis in leukemia cells. J Med Chem 38:31463155[Medline]
- LeCluyse EL 2001 Human hepatocyte culture systems for the in vitro evaluation of cytochrome P450 expression and regulation. Eur J Pharm Sci 13:343368[CrossRef][Medline]