On the Role of Liver X Receptors in Lipid Accumulation in Adipocytes
Lene K. Juvet,
Sissel M. Andresen,
Gertrud U. Schuster,
Knut Tomas Dalen,
Kari Anne R. Tobin,
Kristin Hollung,
Fred Haugen,
Severina Jacinto,
Stine M. Ulven,
Krister Bamberg,
Jan-Åke Gustafsson and
Hilde I. Nebb
Institute for Nutrition Research (L.K.J., S.M.A., K.T.D., K.A.R.T., K.H., F.H., S.M.U., H.I.N.), University of Oslo, N-0316 Oslo, Norway; Molecular Biology (S.J., K.B.), Research Area Cardiovascular & Gastrointestinal, AstraZeneca Mölndal, S-431 83 Mölndal, Sweden; and Department of Bioscience and Medical Nutrition (G.U.S., J.-Å.G.), Novum, S-141 86 Huddinge, Sweden
Address all correspondence and requests for reprints to: Hilde Irene Nebb, Institute for Nutrition Research, University of Oslo, P.O. Box 1046 Blindern, N-0316 Oslo, Norway. E-mail: h.i.nebb{at}basalmed.uio.no.
 |
ABSTRACT
|
---|
The pivotal role of liver X receptors (LXRs) in the metabolic conversion of cholesterol to bile acids in mice is well established. More recently, the LXR
promoter has been shown to be under tight regulation by peroxisome proliferator-activated receptors (PPARs), implying a role for LXR
in mediating the interplay between cholesterol and fatty acid metabolism. We have studied the role of LXR in fat cells and demonstrate that LXR is regulated during adipogenesis and augments fat accumulation in mature adipocytes. LXR
expression in murine 3T3-L1 adipocytes as well as in human adipocytes was up-regulated in response to PPAR
agonists. Administration of a PPAR
agonist to obese Zucker rats also led to increased LXR
mRNA expression in adipose tissue in vivo. LXR agonist treatment of differentiating adipocytes led to increased lipid accumulation. An increase of the expression of the LXR target genes, sterol regulatory binding protein-1 and fatty acid synthase, was observed both in vivo and in vitro after treatment with LXR agonists for 24 h. Finally, we demonstrate that fat depots in LXR
/ß-deficient mice are smaller than in age-matched wild-type littermates. These findings imply a role for LXR in controlling lipid storage capacity in mature adipocytes and point to an intriguing physiological interplay between LXR and PPAR
in controlling pathways in lipid handling.
 |
INTRODUCTION
|
---|
ADIPOCYTES PLAY a central role in maintaining lipid homeostasis and energy balance in vertebrates by storing triglycerides or releasing free fatty acids in response to changes in energy demands (1). Further understanding of this cell type is becoming increasingly important in the light of the rising incidence of obesity and its associated disorders such as type II diabetes, dyslipidemia, and cardiovascular diseases.
Considerable progress has been made during the past years in understanding the molecular mechanisms of adipogenesis. Three classes of transcription factors that directly influence adipogenesis have so far been identified. These are the peroxisome proliferator activated receptor (PPAR)-
, CCAAT/enhancer binding proteins (C/EBPs), and sterol regulatory element binding protein 1c (SREBP-1c, also called ADD1, adipocyte determination and differentiation factor) (2). The nuclear receptor PPAR
and its heterodimeric partner, retinoid X receptor (RXR), have been shown to play an obligatory role in the process of adipocyte differentiation (3, 4, 5). The PPAR
/RXR
heterodimer regulates transcription of adipocyte-specific genes by direct interactions with PPAR
response elements (PPREs) consisting of a direct repeat (AGGTCA) separated by one nucleotide, referred to as a direct repeat-1 motif (6, 7, 8).
Expression and activation of PPAR
in fibroblasts induces the adipocyte gene expression program (9). The ligands for PPAR
are diverse and include naturally occurring ligands, such as the eicosanoid 15-deoxy-12,14-prostaglandin J2 and oxidized low density lipoprotein particles (10, 11, 12, 13) as well as the synthetic thiazolidinediones, a new class of insulin-sensitizing drugs used in the treatment of type II diabetes (14). The effects of these ligands are mediated by changes in the rate of transcription of PPAR
target genes (15).
Liver X receptor (LXR)
(16, 17) and LXRß (18, 19) are members of the nuclear receptor family (20, 21). LXR
expression in adult animals is restricted predominantly to tissues known to play important roles in lipid metabolism, such as liver, kidney, small intestine, spleen, skeletal muscle, adipose tissue, pituitary, and adrenal gland (16, 17, 22), whereas LXRß is ubiquitously expressed (18). Both LXRs heterodimerize with RXR
and bind to a direct repeat-4 LXR response element for transcriptional regulation of specific target genes (23). The tissue distribution of LXRs, the identification of oxysterols as their ligands, and the identification of an LXR response element upstream of the mouse cholesterol 7
-hydroxylase (CYP7A1) gene were the first lines of evidence for the role of LXRs in cholesterol homeostasis (17, 24, 25). More recently, additional target genes for LXRs that are involved in cholesterol metabolism have been identified, including the cholesterol ester transfer protein (26), ATP-binding cassette transporter-A1 (27), -G1 (28), and apolipoprotein E (29). Additional support for the role of LXRs in cholesterol homeostasis came from the analysis of LXR
-deficient mice (LXR
-/-), in which the CYP7A1 gene and several other important lipid-associated genes are dysregulated (30).
LXR
expression was recently shown to be regulated by PPAR
in liver (31) and by PPAR
in macrophages (32), but this is not observed in skeletal muscle (33). A role for LXR in controlling lipogenesis in liver has also been shown (34, 35). Recent experiments revealed that LXRs directly controls the expression of SREBP-1c (36, 37), which regulates lipogenic enzymes in liver (38) such as fatty acid synthase (FAS) (39). Interestingly, LXRs also regulate FAS expression through direct interaction with the FAS promoter (39, 40). Treatment of wild-type mice with LXR agonists led to a marked increase in hepatic triglyceride content (34, 41), which was not observed in LXR
/ß double knockout mice (34). In wild-type mice, but not in LXR
knockout mice, hepatic triglyceride accumulation was also observed upon feeding high cholesterol diet (30). These findings implicate a broad role for LXRs in both sterol and fatty acid metabolism. A role for LXR
in regulation of several metabolic functions in adipocytes was also recently described, including glucose transport, glycogen synthesis, cholesterol synthesis, and nonesterified fatty acid release (42).
Although many of the studies have focused on LXRs function in the liver and macrophages, there is good evidence to suggest that LXRs activity may be important in lipid metabolism in adipocytes as well (26, 30, 36). In this study, we examined the role of LXRs in adipose tissue. Here we demonstrate that LXR
expression is increased directly by PPAR
activation both in cell lines and animals. Moreover, we show that LXR agonists trigger increased lipid accumulation. Treatment of adipocytes with LXR agonists both in vitro and in vivo results in increased expression of genes in the lipogenic pathway. In LXR
/ß knockout mice a significant decrease in adipose tissue was observed compared with wild-type mice. These observations suggest an important interrelationship between fatty acid and cholesterol signaling pathways in adipocytes and point to a physiological function for LXR in triglyceride accumulation.
 |
RESULTS
|
---|
PPAR
Activation Increases LXR
Expression in 3T3-L1 Adipocytes
The 3T3-L1 cell line derived from disaggregated mouse embryos and selected based on the propensity of these cells to differentiate into adipocytes in culture (43) is a widely used model for studying the adipocyte. Over the last 30 yr, this cell line has proven to be a faithful model for studying adipocyte biology, particularly adipogenesis and energy metabolism.
To determine possible changes in LXRs expression during adipocyte differentiation, 3T3-L1 cells were treated for 113 d with agents that induce differentiation. Cells were collected and total RNA was isolated and used in Northern analysis with LXR
, LXRß, PPAR
, SREBP-1, C/EBPß, and adipocyte fatty acid binding protein (aFABP, also called aP2) cDNAs as probes. As shown in Fig. 1
, there were significant changes in the expression of all the known adipocyte markers consistent with previous findings (44, 45). Interestingly, the expression of LXRß was detected in both resting and differentiated cells, whereas LXR
was dramatically induced starting at d 4 after the initiation of the differentiation program reaching maximum levels at d 8 and declining slowly after that. This supports the role of LXR
as an adipocyte differentiation marker gene.
Regulation of LXR
by PPARs has recently been demonstrated in hepatocytes (31) and macrophages (32) but not in skeletal muscle (33). We set out to investigate whether the endogenous LXR
gene could be induced by PPAR
agonists in adipocytes, the main energy storage organ. We performed Northern blot analysis of total RNA obtained from fully differentiated 3T3-L1 adipocytes treated for 24 h with the PPAR
agonists darglitazone (1 µM) or rosaglitazone (1 µM) (Fig. 2
). Both compounds gave a robust increase (
4-fold) in the steady-state LXR
mRNA levels in treated cells relative to untreated cells. A smaller effect was observed when the cells were treated with PPAR
agonists such as bezafibrate (10 µM) or tetradecylthioacetic acid (TTA) (50 µM). Increased expression of LXR
protein as a result of PPAR
activation was confirmed by Western blotting (data not shown).
To determine whether LXR
is a PPAR
target gene also in adipocytes, a luciferase reporter gene construct containing the mouse LXR
5'-flanking region [pLXR
(-1.5/+1.8 kb)-LUC] (31) was transiently transfected into fully differentiated 3T3-L1 adipocytes on d 13. Both darglitazone and rosaglitazone induced reporter gene activity in 3T3-L1 cells (data not shown). In 3T3-L1 cells, the functional PPRE in the 5'-flank of the LXR
promoter, localized at position -722 bp upstream of transcription start, is identical to the one previously identified using macrophages as model system (32).
The PPAR
-Mediated Up-Regulation of LXR
Gene Expression Is a Primary and Direct Effect of PPAR
Agonist-Activated Transcription
The protein synthesis inhibitor cycloheximide was used to determine whether regulation of LXR
by PPAR
requires new protein synthesis. 3T3-L1 cells were differentiated and either left untreated or treated with darglitazone (1 µM) in the presence or absence of cycloheximide. Cells were collected; total RNA was isolated and used in Northern analysis with LXR
as probe. As shown in Fig. 3
, cycloheximide treatment inhibited the basal levels of LXR
, indicating that ongoing protein synthesis is required for LXR
basal transcription. Despite this, an increased LXR
expression was observed when cells were treated with darglitazone and cycloheximide compared with the control cells treated with cycloheximide (Fig. 3
). This outcome suggests that the effect of PPAR
agonist on LXR
expression is a primary and direct effect of PPAR
agonist-activated transcription. A direct effect of PPAR
agonist was also shown for the known PPAR
target gene, aFABP (data not shown), except that the basal aFABP mRNA level was still high after 24 h of cycloheximide treatment (46).
Regulation of LXR
Expression in Obese Zucker Rats Treated with Thiazolidinediones
To investigate whether PPAR
activation induces changes in LXR
expression also in vivo, we examined the effect of darglitazone in Zucker rats, a rodent model of insulin resistance. Obese Zucker rats were treated with darglitazone for 3 wk, which normalized serum levels of triglycerides and insulin to those of the healthy, lean littermates (Table 1
) (47). RNA was isolated from the epididymal fat pads and analyzed by Northern blotting. The mRNA level of both LXR
and the known PPAR
target gene aFABP (48) increased 2-fold in darglitazone-treated obese animals compared with untreated controls (Fig. 4
, A and B). The increased LXR
mRNA levels in darglitazone-treated obese Zucker rats were approximately equal to those found in untreated lean littermates. In contrast to adipose tissue, LXR
expression in liver of darglitazone-treated obese Zucker rats remained unchanged (data not shown). Thus, regulation of LXR
expression in vivo by darglitazone occurs in adipocytes but not to the same extent in liver.

View larger version (47K):
[in this window]
[in a new window]
|
Figure 4. Effect of Darglitazone on LXR Levels in Zucker Rats
A, Obese Zucker rats were treated with vehicle or darglitazone for 3 wk. Each lane contains 20 µg total RNA prepared from white adipose tissue of individual animals. The levels of LXR and aFABP were measured after Northern blot analysis. B, The intensities were measured by densitometric scanning and were normalized against 36B4 examined in the same samples as a control. The results represent the average ± SD from all experimental animals. An asterisk indicates values significantly different from their respective controls determined by one-way ANOVA tests.
|
|
LXR
in Human Adipose Tissue Responds to Darglitazone and 22(R)-Hydroxycholesterol
Next we investigated the effect of PPAR
and LXR
activation on LXR
expression in human adipose tissue (Fig. 5
). Human adipose tissue was obtained from breast reduction surgery. Pieces of adipose tissue (
500 mg) were prepared for growth in primary culture under sterile conditions (49) and incubated with darglitazone (1 µM) or 22(R)-hydroxycholesterol [22(R)-HC] (5 µM) (50) for 48 h. Darglitazone treatment induced LXR
mRNA accumulation 4-fold, a similar response to that observed in 3T3-L1 cells. 22(R)-HC increased LXR
expression 2.5-fold in human adipose tissue (Fig. 5
); a similar 2-fold induction was observed in fully differentiated 3T3-L1 cells stimulated with 22(R)-HC (5 µM) for 24 h (data not shown). These data show that LXR
expression in human adipocytes is regulated by both PPAR
and LXR agonists in vitro.
A Physiological Role of LXR
in Adipocytes
To investigate the role of LXR during adipocyte differentiation, 3T3-L1 cells were treated with different oxy-sterols, 22(R)-HC (5 µM), 22(S)-HC (5 µM), or the synthetic LXR agonist, T0901317 (1 µM) (34), during the differentiation process. To show incorporation of lipids, the cells were stained with Oil Red-O on d 13. Cells treated with either 22(R)-HC or T0901317 during differentiation formed larger lipid droplets than those observed in control cells. In contrast, cells treated with the LXR antagonist 22(S)-HC accumulated less lipids compared with nontreated control (Fig. 6A
).

View larger version (75K):
[in this window]
[in a new window]
|
Figure 6. Effect of LXR Ligands on Lipid Accumulation during Differentiation of 3T3-L1 Adipocytes in Culture
A, Effect of LXR agonists on differentiation of 3T3-L1 cells. 3T3-L1 cells were cultured for 13 d and stimulated with either the oxysterols 22(R)-HC (5 µM), 22(S)-HC (5 µM), or the synthetic LXR ligand T0901317 (1 µM) during differentiation. Differentiation was induced by standard DMEM containing 1 µg/ml insulin, 0.5 mM isobutylmethylxanthine, and 1 nM dexamethasone. Each well was stained with Oil Red-O in parallel at d 13 after differentiation to determine the degree of lipid accumulation. Control (T) represents the control to the T0901317 treatment; this experiment was done separately. B, Effect of darglitazone treatment in combination with 22(R)-HC or 22(S)-HC on differentiation of 3T3-L1 cells. 3T3-L1 cells were treated for 13 d with either darglitazone alone or in combination with 22(R)-HC (5 µM) or 22(S)-HC (5 µM) during adipocyte differentiation. Differentiation and staining were performed as described above. C, Northern blot analysis of total RNA (20 µg) from 3T3-L1 adipocytes treated with either darglitazone (1 µM), 22(R)-HC (5 µM), or 22(S)-HC (5 µM) as indicated in the figure. The cells were treated for 13 d during 3T3-L1 adipocyte differentiation. Samples from each treatment at d 13 were subjected to Northern blot analysis and LXR , aFABP, CD36, SREBP, and 18S mRNA expression was examined using cDNA probes for each gene. Relative levels of the mRNA transcripts for each stimulation compared with control are shown.
|
|
We next assessed whether PPAR
-induced adipocyte differentiation was affected by LXR activation. 3T3-L1 cells were treated with darglitazone either alone or in presence of 22(R)-HC or 22(S)-HC during differentiation into adipocytes. Darglitazone-treated 3T3-L1 cells displayed a high degree of lipid accumulation as expected (51). Addition of 22(R)-HC together with darglitazone did not change this effect. Interestingly, however, cells stimulated with darglitazone and 22(S)-HC had less lipid accumulation compared with cells treated with darglitazone alone on d 13 (Fig. 6B
). These treatments affected the cellular triglyceride levels, demonstrated by lipid contents measurements, whereas the cholesterol and cholesterol ester levels were not affected to the same extent (Table 2
). Our results indicate a physiological function for LXRs in enhancing the amount of lipid loaded into mature adipocytes.
View this table:
[in this window]
[in a new window]
|
Table 2. Triglyceride, Cholesterol, and Cholesterol Ester Levels in 3T3-L1 Cells Treated with Darglitazone, 22(R)-HC, 22(S)-HC, or with Darglitazone and 22(S)-HC in Combination during the Differentiation
|
|
We therefore examined the possible effect of LXR ligands on the expression of adipocyte specific markers. Total RNA was isolated from 3T3-L1 adipocytes that were treated with 22(R)-HC, 22(S)-HC, or darglitazone during the whole differentiation process. Treatment with 22(R)-HC gave a small induction of gene expression of LXR
as well as genes involved in fatty acid transport such as aFABP and fatty acid translocase (CD36), although to a lower extent than with darglitazone treatment (Fig. 6C
). However, the total amount of SREBP-1a and 1c gene transcripts was not regulated when 3T3-L1 cells were treated for 13 d with LXR ligand.
T0901317 was able to induce FAS expression in fully differentiated 3T3-L1 cells after 24 h of treatment. A 4-fold increase of FAS expression was observed (Fig. 7A
). As for 22-(R)-HC treatment of human adipocytes, T0901317 treatment of 3T3-L1 cells induced expression of LXR
by 1.8-fold. Increased FAS, SREBP-1, and LXR
expression was also confirmed in mice fed with 50 mg/kg body weight (mpk) T0901317 for 24 h (Fig. 7B
).
To further confirm the role of LXRs in adipose tissue and fat accumulation, we analyzed 18-month-old mice with targeted disruption of both the LXR
and -ß genes [LXR
/ß double knockout mice (DOKO)]. Analysis of their body weight and adipose tissue indicated a significant decrease in white adipose tissue (WAT) and brown adipose tissue (BAT) compared with wild-type mice (Table 3
). The role of LXR in adipose tissue and fat accumulation was observed also in 3- to 4-month-old LXR DOKO mice showing reduced peritoneal fat pads compared with wild-type mice, whereas this was not observed in younger DOKO mice (data not shown). It was easier to obtain a clear significant decrease in WAT and BAT of older DOKO mice and therefore 18-month-old DOKO mice were used in our study. These data hence confirm our in vitro data and claim a role for LXRs in fat accumulation.
 |
DISCUSSION
|
---|
The pivotal role of LXRs in the metabolic conversion of cholesterol to bile acids in mice is well established (reviewed in Refs. 52, 53, 54). The observation that LXR
is fatty acid responsive (31) and expressed in tissues such as liver, kidney, small intestine, spleen, skeletal muscle, and adipose tissue (16, 17, 22, 33, 52) suggests that it also plays a role in fatty acid homeostasis. The analysis of gene expression in LXR
knockout mice confirmed that LXR
regulates a number of candidate target genes involved in both cholesterol and fatty acid metabolism (30, 55), which implicates LXR in a broader role in lipid metabolism. Furthermore, LXRs modifies the expression of lipogenic enzyme genes by regulating SREBP-1c expression. These studies provide a link between fatty acid and cholesterol metabolism (36, 37) and stimulated us to study the potential cross-regulation between sterol and lipid metabolism in adipocytes, which play a central role in maintaining lipid homeostasis and energy balance.
The work presented here describes a physiological role for LXRs in adipocytes. Activation of PPAR
induces LXR
gene expression through a PPRE located in the proximal promoter (-722 bp from transcription start) of the LXR
gene. This regulation of LXR
by PPAR
activation was observed in a murine adipocyte cell line, in human primary adipocytes and obese Zucker rats. We also found that LXR agonists are able to induce lipid accumulation in 3T3-L1 adipocytes in culture. The role of LXRs in storage of fat in adipocytes was further confirmed in LXR
/ß double knockout mice, in which the adipose tissue mass was decreased compared with wild-type mice.
Our results clearly show that LXR
is directly regulated by PPAR
in adipocytes. This cross-talk between sterol and lipid metabolism has also been shown in macrophages (32). A similar interrelationship has also been demonstrated between PPAR
and LXR
in liver, further supporting the importance of this cross-regulation in different metabolic tissues (31). Surprisingly, this association seems to be absent in skeletal muscle (33). Furthermore, we show an up-regulation of LXR
expression by PPAR
agonists in obese Zucker rats, in human adipocytes, and in mature 3T3-L1 adipocytes to further confirm the cross-talk between PPAR
and LXR
. In addition, we have also been able to demonstrate up-regulation of LXR
expression by an endogenous LXR agonist, 22-(R)-HC, in mature 3T3-L1 adipocytes, in human adipocytes in culture as well as by T0901317 in mice. LXR
autoregulation has also recently been observed in human macrophages; however, no autoregulation was reported in adipocytes (56, 57). Future studies are obviously needed to clarify this issue in adipose tissue.
Interestingly, increased lipid accumulation was observed when 3T3-L1 cells were treated with LXR agonists, 22(R)-HC and T0901317. Moreover, the LXR antagonist, 22(S)-HC, was unable to induce lipid accumulation during 3T3-L1 adipocyte differentiation. As expected, darglitazone stimulated 3T3-L1 cells accumulated large amounts of lipid. However, this accumulation was attenuated by the addition of 22(S)-HC. Interestingly, triglyceride levels in 22(R)-HC-treated cells increased to a similar extent as in cells treated with darglitazone, whereas neither cholesterol nor cholesterol ester intracellular levels changed. Our results therefore indicate that activation of LXRs increases triglyceride accumulation in adipocytes.
Because recent studies have established a role for LXRs in regulating triglyceride synthesis in liver (34, 36), we studied whether LXR ligands could modify the expression of genes in the lipogenic pathway in adipocytes. Our in vitro and in vivo studies demonstrate an increase in FAS and LXR
expression, after treatment with T0901317 for 24 h. Furthermore, in vivo studies also show an up-regulation of SREBP-1. The observed transcript is probably the SREBP-1c isoform because this is the most common isoform in tissues (45). The lack of any observed regulation of SREBP-1 in 3T3-L1 cells might be due to abundant expression of SREBP-1a compared with SREBP-1c. As only the SREBP-1c isoform is a direct target gene of LXRs, this might explain why SREBP-1 is not regulated in our cell culture systems. In contrast to our results, Ross et al. (42) did not report any effect of T0901317 on the accumulation of triacylglycerol under their culture conditions (42). Furthermore, they did not observe any effect on lipogenesis either in fully differentiated 3T3-L1 cells that expressed ectopic LXR
treated with 1 µM T0901317 or in female C57BL/6 mice injected with 50 mg of T0901317/kg·d daily for 1, 3, or 7 d. These authors rather demonstrated new metabolic roles for LXR
in adipose tissue with effects on metabolic pathways such as increased basal glucose uptake, glycogen synthesis, cholesterol synthesis, and release of nonesterified fatty acids. Our lipogenic effect of LXRs in adipose tissue is opposite to the lipolytic role of LXR
identified by Ross et al. (42). A role for LXRs in lipogenesis is clearly documented in liver (34, 35, 36). This lipogenic role of LXRs in other metabolic tissues has not been studied to the same extent. However, a recent study in skeletal muscle indicates a role for LXR
mainly in cholesterol metabolism, whereas it seems less important in fatty acid metabolism (33). This demonstrates that LXRs play different physiological roles in metabolic pathways in different tissues. Further investigations are needed to clarify the precise metabolic role of LXRs in the body.
As LXR
expression is induced late during the differentiation process, we do not believe that LXR plays an important role in the differentiation of adipocytes. Our results rather suggest that LXR plays a role in the late differentiated stage of adipocytes by inducing expression of genes important for fatty acid transport, fatty acid synthesis, and triglyceride accumulation. Supporting this hypothesis, the LXR
/ß double knockout mice contain fat, although less than wild-type mice, demonstrating that LXRs are not necessary for adipocyte differentiation. However, the reduced lipid accumulation clearly shows that LXRs play an important role in mediating fat metabolism and perhaps remodeling of mature adipocytes. To obtain a significant reduction in the fat depot, both LXR
and LXRß need to be abolished because a small but nonsignificant reduction of fat depot was obtained in LXR
knockout mice (data not shown). In addition, it is still unclear whether LXR
and LXRß are redundant in regulation of fatty acid metabolism. In the CYP7A1 pathway in mouse liver, LXRß is not able to compensate for LXR
(30). Nevertheless, LXR
is expressed during the differentiation of adipocytes and therefore seems the most plausible candidate for causing the effects observed in this study. However, because the LXR agonists are ligands for both isoforms, we cannot rule out an important role for LXRß in adipose tissue. Clearly, more studies are needed to unravel the precise role of LXR
and LXRß in adipocytes.
 |
MATERIALS AND METHODS
|
---|
Materials
DMEM, penicillin, streptomycin, and L-glutamine were obtained from Sigma (St. Louis, MO). Agarose was purchased from Bio-Rad Laboratories, Inc. (Richmond, CA). Multiple DNA labeling systems and radiolabeled [
-32P]deoxy-CTP were purchased from Amersham Pharmacia Biotech (Buckinghamshire, UK). Bio-Trans nylon filter was from ICN Biochemicals, Inc. (Irvine, CA). cDNA probes for human ribosomal protein L27 (ATCC-107385) and for the 18S ribosomal protein (ATCC-107382) were purchased from ATCC (Manassas, VA). pGL3-basic vector, dual luciferase assay, and TNT T7 Coupled Reticulo Lysate System were obtained from Promega Corp. (Madison, WI). TTA (C14-S-C2) was synthesized as previously described (58). Darglitazone, rosaglitazone, and T0901317 were obtained from Medicinal Chemistry, AstraZeneca Research and Development Mölndal (Mölndal, Sweden). Other chemicals were obtained from Sigma.
Cells
The 3T3-L1 cell line (ATCC) was maintained in DMEM supplemented with 10% fetal calf serum, 2 mM L-glutamine, and penicillin/streptomycin at 37 C. 3T3-L1 preadipocytes were seeded at passage 16, grown to confluence, and then exposed to adipogenic reagents for 3 d, followed by culturing for an additional 3 d in a medium containing insulin only as described elsewhere (59). The cells were then grown for an additional 7 d to ensure that all cells had become mature adipocytes (d 13). Insulin at a concentration of 1 µg/ml, isobutylmethylxanthine at 0.5 mM, and dexamethasone at 1 µM were used as adipogenic reagents unless otherwise stated in the figure legend. Darglitazone, rosaglitazone, and T0901317 were used at a concentration of 1 µM, 22(R)-hydroxycholesterol and 22(S)-hydroxycholesterol were used at a concentration of 5 µM, whereas Bezafibrate and TTA were used at concentrations of 10 µM and 50 µM, respectively.
Preparation and Analysis of RNA
Total RNA from differentiated 3T3-L1 adipocytes or adipose tissues were extracted with Trizol (Life Technologies, Inc., Gaithersburg, MD) as recommended by the manufacturer. Northern blot analysis of RNA was performed as described earlier (60). Twenty micrograms of total RNA were used and blots were probed with rat LXR
, LXRß, PPAR
, SREBP-1, FAS, C/EBPß, aFABP, and CD36 cDNAs. L27 mRNA (ATCC-107385), 36B4 mRNA, or 18S ribosomal protein mRNA (ATCC-107382) were used as controls for equal RNA loading.
Immunoblotting
Cells from culture or adipose tissue were homogenized in PBS containing 1% Nonidet P-40, 0.5% sodium deoxycholate, 0.1% sodium dodecyl sulfate, and protease inhibitors (Complete Protease Inhibitor Cocktail Tablets, Roche Molecular Biochemicals, Mannheim, Germany), and a soluble protein fraction was obtained. Protein concentration was determined using the BCA protein reagent (Pierce Chemical Co.). Aliquots of each sample (200 µg) were separated on a 10% SDS-PAGE, and transferred to a nitrocellulose membrane (Hybond-C-Extra, Amersham Pharmacia Biotech). LXR
proteins were detected immunochemically using a commercially available antibody (no. sc-1206, Santa Cruz Biotechnology, Inc., Santa Cruz, CA), and the enhanced chemiluminescence kit (Amersham Pharmacia Biotech).
Transfection and Luciferase Assay
3T3-L1 preadipocytes were grown to confluence in six-well dishes and differentiated as described above. 3T3-L1 differentiated adipocytes were transfected using 6 µl lipofectamine Plus reagent, 4 µl lipofectamine, and 1 µg of a plasmid construct containing the LXR
promoter fused to the luciferase reporter gene (31) and 100 ng pTK Renilla luciferase (as a control of transfection efficiency). In addition, 300 ng of RXR
or PPAR
2 were cotransfected. The transfections were carried out as recommended by the manufacturer (Life Technologies, Inc.), and the transfection efficiency was confirmed by transfection with pEGFP (CLONTECH Laboratories, Inc., Palo Alto, CA). Three hours after transfection, cells were cultured in medium containing serum and incubated for 24 h in the same medium containing appropriate agents, as indicated. Site-directed mutagenesis was performed using the QuikChange site-directed mutagenesis kit from Stratagene (La Jolla, CA), according to the manufacturers procedure. The luciferase activities were measured as recommended by the manufacturer (dual luciferase assay, Promega Corp.). All transfections were performed in triplicate.
Animal Experiments
Male obese Zucker rats (fa/fa) and lean Zucker littermates (Fa/?) were from Genetic Models, Inc. (Indianapolis, IN). The animals were fed either a control diet or diet containing darglitazone (6 mg/kg·d) beginning at 6 wk of age and continuing for an additional 3 wk. The animals were maintained at 22 C with a 12-h light, 12-h dark fixed cycle. Blood was collected from the tail vein; and plasma glucose, plasma insulin, and triglyceride concentrations were monitored throughout the dosing period.
Male C57Bl/6J mice 10 wk of age (30 g) were housed in temperature-controlled rooms (22 C) with 12-h/12-h fixed light-dark cycles with free access to water and standard pellet diet. Mice were gavage-fed twice (24 h and 8 h before the mice were killed) using mpk of T0901317 in a vehicle containing 0.9% carboxymethyl-cellulose (Sigma). Control mice were given vehicle only. Mice were killed by cervical dislocation and epididymal fat was immediately frozen in liquid nitrogen.
LXR
/ß double knockout mice were generated by gene targeting in our laboratory as described previously (55, 61). All mice used in our study, LXR
/ß double knockout mice and wild-type (controls), had mixed genetic background based on 129/Sv and C57Bl/6 strains, finally back-crossed in C57BL/6 mice for three generations, unless otherwise stated. Animals were housed with a regular 12-h light/12-h dark cycle and fed a low-fat standard rodent chow diet (R36 Lactamin AB, Vadstena, Sweden) ad libitum. For the determination of the adipose tissue content in LXR-deficient and wild-type mice, 18-month-old mice were anesthetized with light methoxyfluorane and killed by cervical dislocation. White adipose tissue (epididymal and peritoneal) and brown adipose tissues were removed for further analyses. Data are presented as mean ± SEM. The significance of differences between groups was tested by Students t test (Statistica software, Stat Soft, Tulsa OK).
Experiments were approved by the local ethical committee for animal experiments and the Guidelines for the Care and Use of Laboratory animals were followed.
Human Fat Explant Experiments
Source of Tissue.
Subcutaneous adipose tissue was obtained from nondiabetic patients undergoing mammoplastic surgery at the Volvat Medical Center. Written informed consent was obtained from the subjects. The local Ethical committee in Oslo, Norway, approved the study. Pieces of adipose tissue (5600 mg) were prepared under sterile conditions and used for incubations in plastic tubes essentially as described (49). Briefly, the tissue was preincubated for 3 d in a control medium (Parker Medium 199, Statens Bakteriologiska Laboratorium, Stockholm, Sweden) supplemented with 12.5 mmol/liter NaHCO3, 10 mmol/liter HEPES, 1% human serum albumin, 7175 pmol/liter insulin, and penicillin/streptomycin (pH adjusted to 7.4). During the next 48 h, 1 µM darglitazone and 5 µM 22-(R)-hydroxycholesterol was added as indicated in the figure legends. The adipose tissue was collected, lysed and mRNA was extracted with magnetic oligo deoxythymidine particles (Genovision, Oslo, Norway). Northern blot analysis of RNA was performed as described (60). Three micrograms of mRNA were analyzed for rat LXR
mRNA for the L27 mRNA (ATCC-107385).
Lipid Determination
Triglyceride, cholesterol, and cholesterol esters were extracted from total cell lysate as described previously (62). Dried lipids were resuspended in hexane. Identifications were by comigration with standards on thin layer chromatography plates using hexane/diethyl ether/acetic acid 80:20:1. Determination of cholesterol and triglyceride was performed with Cholesterol RTU (BioMérieux) and Triglyceride Enzymatique PAP150 (BioMérieux).
Oil Red-O Staining
Light microscopy and Oil Red-O staining were used to monitor the characteristic cell rounding and lipid accumulation during adipocyte differentiation, essentially as described previously (63).
 |
ACKNOWLEDGMENTS
|
---|
We are very grateful to Volvat Medical Center for providing us with sc adipose tissue from their patients undergoing mammoplastic surgery. We are grateful to Borghild M. Arntsen and Marie Green for excellent technical assistance, to Kirsten Robertson for assisting with the dissection of LXR-deficient mice, and Peter Åkerblad for critically reading the manuscript.
 |
FOOTNOTES
|
---|
The work was supported by Johan Throne Holst Foundation, AstraZeneca AB, Norwegian Research Council, The Novo Nordisk Foundation, The Diabetes Foundation, Norwegian Cancer Society, Thore Nilsson Foundation, and Swedish Medical Research Council (No. 13x-2819).
Abbreviations: aFABP, Adipocyte fatty acid binding protein; C/EBPs, CCAAT/enhancer binding proteins; CYP7A1, cholesterol 7
-hydroxylase; FAS, fatty acid synthase; LXR, liver X receptor; mpk, mg/kg body weight; PPAR, peroxisome proliferator-activated receptor; PPRE, PPAR
response element; RXR, retinoid X receptor; SREBP, sterol regulatory binding protein; TTA, tetradecylthioacetic acid.
Received for publication August 28, 2001.
Accepted for publication November 15, 2002.
 |
REFERENCES
|
---|
- Fruhbeck G, Gomez-Ambrosi J, Muruzabal FJ, Burrell MA 2001 The adipocyte: a model for integration of endocrine and metabolic signaling in energy metabolism regulation. Am J Physiol Endocrinol Metab 280:E827E847
- Rosen ED, Walkey CJ, Puigserver P, Spiegelman BM 2000 Transcriptional regulation of adipogenesis. Genes Dev 14:12931307[Free Full Text]
- Barak Y, Nelson MC, Ong ES, Jones YZ, Ruiz-Lozano P, Chien KR, Koder A, Evans RM 1999 PPAR
is required for placental, cardiac, and adipose tissue development. Mol Cell 4:585595[Medline]
- Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, Spiegelman BM, Mortensen RM 1999 PPAR
is required for the differentiation of adipose tissue in vivo and in vitro. Mol Cell 4:611617[Medline]
- Kubota N, Terauchi Y, Miki H, Tamemoto H, Yamauchi T, Komeda K, Satoh S, Nakano R, Ishii C, Sugiyama T, Eto K, Tsubamoto Y, Okuno A, Murakami K, Sekihara H, Hasegawa G, Naito M, Toyoshima Y, Tanaka S, Shiota K, Kitamura T, Fujita T, Ezaki O, Aizawa S, Kadowaki T 1999 PPAR
mediates high-fat diet-induced adipocyte hypertrophy and insulin resistance. Mol Cell 4:597609[Medline]
- Tontonoz P, Hu E, Graves RA, Budavari AI, Spiegelman BM 1994 mPPAR
2: tissue-specific regulator of an adipocyte enhancer. Genes Dev 8:12241234[Abstract]
- Tontonoz P, Graves RA, Budavari AI, Erdjument-Bromage H, Lui M, Hu E, Tempst P, Spiegelman BM 1994 Adipocyte-specific transcription factor ARF6 is a heterodimeric complex of two nuclear hormone receptors, PPAR
and RXR
. Nucleic Acids Res 22:56285634[Abstract]
- Kliewer SA, Umesono K, Noonan DJ, Heyman RA, Evans RM 1992 Convergence of 9-cis retinoic acid and peroxisome proliferator signalling pathways through heterodimer formation of their receptors. Nature 358:771774[CrossRef][Medline]
- Tontonoz P, Hu E, Spiegelman BM 1994 Stimulation of adipogenesis in fibroblasts by PPAR
2, a lipid-activated transcription factor [published erratum appears in Cell 1995 Mar 24;80(6):following 957]. Cell 79:11471156[Medline]
- Forman BM, Tontonoz P, Chen J, Brun RP, Spiegelman BM, Evans RM 1995 15-Deoxy-
12, 14-prostaglandin J2 is a ligand for the adipocyte determination factor PPAR
. Cell 83:803812[Medline]
- Kliewer SA, Lenhard JM, Willson TM, Patel I, Morris DC, Lehmann JM 1995 A prostaglandin J2 metabolite binds peroxisome proliferator-activated receptor
and promotes adipocyte differentiation. Cell 83:813819[Medline]
- Tontonoz P, Nagy L, Alvarez JG, Thomazy VA, Evans RM 1998 PPAR
promotes monocyte/macrophage differentiation and uptake of oxidized LDL. Cell 93:241252[Medline]
- Nagy L, Tontonoz P, Alvarez JG, Chen H, Evans RM 1998 Oxidized LDL regulates macrophage gene expression through ligand activation of PPAR
. Cell 93:229240[Medline]
- Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson TM, Kliewer SA 1995 An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor
(PPAR
). J Biol Chem 270:1295312956[Abstract/Free Full Text]
- Willson TM, Brown PJ, Sternbach DD, Henke BR 2000 The PPARs: from orphan receptors to drug discovery. J Med Chem 43:527550[CrossRef][Medline]
- Apfel R, Benbrook D, Lernhardt E, Ortiz MA, Salbert G, Pfahl M 1994 A novel orphan receptor specific for a subset of thyroid hormone-responsive elements and its interaction with the retinoid/thyroid hormone receptor subfamily. Mol Cell Biol 14:70257035[Abstract]
- Willy PJ, Umesono K, Ong ES, Evans RM, Heyman RA, Mangelsdorf DJ 1995 LXR, a nuclear receptor that defines a distinct retinoid response pathway. Genes Dev 9:10331045[Abstract]
- Song C, Kokontis JM, Hiipakka RA, Liao S 1994 Ubiquitous receptor: a receptor that modulates gene activation by retinoic acid and thyroid hormone receptors. Proc Natl Acad Sci USA 91:1080910813[Abstract/Free Full Text]
- Teboul M, Enmark E, Li Q, Wikstrom AC, Pelto-Huikko M, Gustafsson J-Å 1995 OR-1, a member of the nuclear receptor superfamily that interacts with the 9-cis-retinoic acid receptor. Proc Natl Acad Sci USA 92:20962100[Abstract]
- Mangelsdorf DJ, Evans RM 1995 The RXR heterodimers and orphan receptors. Cell 83:841850[Medline]
- Mangelsdorf DJ, Thummel C, Beato M, Herrlich P, Schutz G, Umesono K, Blumberg B, Kastner P, Mark M, Chambon P 1995 The nuclear receptor superfamily: the second decade. Cell 83:835839[Medline]
- Auboeuf D, Rieusset J, Fajas L, Vallier P, Frering V, Riou JP, Staels B, Auwerx J, Laville M, Vidal H 1997 Tissue distribution and quantification of the expression of mRNAs of peroxisome proliferator-activated receptors and liver X receptor-
in humans: no alteration in adipose tissue of obese and NIDDM patients. Diabetes 46:13191327[Abstract]
- Willy PJ, Mangelsdorf DJ 1997 Unique requirements for retinoid-dependent transcriptional activation by the orphan receptor LXR. Genes Dev 11:289298[Abstract]
- 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]
- 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]
- Luo Y, Tall AR 2000 Sterol upregulation of human CETP expression in vitro and in transgenic mice by an LXR element. J Clin Invest 105:513520[Abstract/Free Full Text]
- Repa JJ, Turley SD, Lobaccaro JA, Medina J, Li L, Lustig K, Shan B, Heyman RA, Dietschy JM, Mangelsdorf DJ 2000 Regulation of absorption and ABC1-mediated efflux of cholesterol by RXR heterodimers. Science 289:15241529[Abstract/Free Full Text]
- Venkateswaran A, Repa JJ, Lobaccaro JM, Bronson A, Mangelsdorf DJ, Edwards PA 2000 Human white/murine ABC8 mRNA levels are highly induced in lipid-loaded macrophages. A transcriptional role for specific oxysterols. J Biol Chem 275:1470014707[Abstract/Free Full Text]
- Laffitte BA, Repa JJ, Joseph SB, Wilpitz DC, Kast HR, Mangelsdorf DJ, Tontonoz P 2001 LXRs control lipid-inducible expression of the apolipoprotein E gene in macrophages and adipocytes. Proc Natl Acad Sci USA 98:507512[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]
- Tobin KA, Steineger HH, Alberti S, Spydevold O, Auwerx J, Gustafsson JA, Nebb HI 2000 Cross-talk between fatty acid and cholesterol metabolism mediated by liver X receptor-
. Mol Endocrinol 14:741752[Abstract/Free Full Text]
- Chawla A, Boisvert WA, Lee CH, Laffitte BA, Barak Y, Joseph SB, Liao D, Nagy L, Edwards PA, Curtiss LK, Evans RM, Tontonoz P 2001 A PPAR
-LXR-ABCA1 pathway in macrophages is involved in cholesterol efflux and atherogenesis. Mol Cell 7:161171[Medline]
- Muscat GE, Wagner BL, Hou J, Tangirala RK, Bischoff ED, Rohde P, Petrowski M, Li J, Shao G, Macondray G, Schulman IG 2002 Regulation of cholesterol homeostasis and lipid metabolism in skeletal muscle by liver X receptors. J Biol Chem 277:4072240728[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]
- Tobin KA, Ulven SM, Schuster GU, Steineger HH, Andresen SM, Gustafsson JA, Nebb HI 2002 Liver X receptors as insulin-mediating factors in fatty acid and cholesterol biosynthesis. J Biol Chem 277:1069110697[Abstract/Free Full Text]
- Repa JJ, Liang G, Ou J, Bashmakov Y, Lobaccaro JM, Shimomura I, Shan B, Brown MS, Goldstein JL, Mangelsdorf DJ 2000 Regulation of mouse sterol regulatory element-binding protein-1c gene (SREBP-1c) by oxysterol receptors, LXR
and LXRß. Genes Dev 14:28192830[Abstract/Free Full Text]
- Yoshikawa T, Shimano H, Amemiya-Kudo M, Yahagi N, Hasty AH, Matsuzaka T, Okazaki H, Tamura Y, Iizuka Y, Ohashi K, Osuga J, Harada K, Gotoda T, Kimura S, Ishibashi S, Yamada N 2001 Identification of liver X receptor-retinoid X receptor as an activator of the sterol regulatory element-binding protein 1c gene promoter. Mol Cell Biol 21:29913000[Abstract/Free Full Text]
- Kim JB, Spiegelman BM 1996 ADD1/SREBP1 promotes adipocyte differentiation and gene expression linked to fatty acid metabolism. Genes Dev 10:10961107[Abstract]
- Joseph SB, Laffitte BA, Patel PH, Watson MA, Matsukuma KE, Walczak R, Collins JL, Osborne TF, Tontonoz P 2002 Direct and indirect mechanisms for regulation of fatty acid synthase gene expression by LXRs. J Biol Chem 277:1101911025[Abstract/Free Full Text]
- Shimano H, Yahagi N, Amemiya-Kudo M, Hasty AH, Osuga J, Tamura Y, Shionoiri F, Iizuka Y, Ohashi K, Harada K, Gotoda T, Ishibashi S, Yamada N 1999 Sterol regulatory element-binding protein-1 as a key transcription factor for nutritional induction of lipogenic enzyme genes. J Biol Chem 274:3583235839[Abstract/Free Full Text]
- Grefhorst A, Elzinga BM, Voshol PJ, Plosch T, Kok T, Bloks VW, Van Der Sluijs FH, Havekes LM, Romijn JA, Verkade HJ, Kuipers F 2002 Stimulation of lipogenesis by pharmacological activation of the liver X receptor leads to production of large, triglyceride-rich very low density lipoprotein particles. J Biol Chem 277:3418234134[Abstract/Free Full Text]
- Ross SE, Erickson RL, Gerin I, DeRose PM, Bajnok L, Longo KA, Misek DE, Kuick R, Hanash SM, Atkins KB, Andresen SM, Nebb HI, Madsen L, Kristiansen K, MacDougald OA 2002 Microarray analyses during adipogenesis: understanding the effects of Wnt signaling on adipogenesis and the roles of liver X receptor
in adipocyte metabolism. Mol Cell Biol 22:59895999[Abstract/Free Full Text]
- Green H, Meuth M 1974 An established pre-adipose cell line and its differentiation in culture. Cell 3:127133[Medline]
- Wu Z, Xie Y, Morrison RF, Bucher NL, Farmer SR 1998 PPAR
induces the insulin-dependent glucose transporter GLUT4 in the absence of C/EBP
during the conversion of 3T3 fibroblasts into adipocytes. J Clin Invest 101:2232[Abstract/Free Full Text]
- Shimomura I, Shimano H, Horton JD, Goldstein JL, Brown MS 1997 Differential expression of exons 1a and 1c in mRNAs for sterol regulatory element binding protein-1 in human and mouse organs and cultured cells. J Clin Invest 99:838845[Abstract/Free Full Text]
- Melki SA, Abumrad NA 1993 Expression of the adipocyte fatty acid-binding protein in streptozotocin-diabetes: effects of insulin deficiency and supplementation. J Lipid Res 34:15271534[Abstract]
- Oakes ND, Thalen PG, Jacinto SM, Ljung B 2001 Thiazolidinediones increase plasma-adipose tissue FFA exchange capacity and enhance insulin-mediated control of systemic FFA availability. Diabetes 50:11581165[Abstract/Free Full Text]
- Harris PK, Kletzien RF 1994 Localization of a pioglitazone response element in the adipocyte fatty acid-binding protein gene. Mol Pharmacol 45:439445[Abstract]
- Ottosson M, Vikman-Adolfsson K, Enerback S, Olivecrona G, Bjorntorp P 1994 The effects of cortisol on the regulation of lipoprotein lipase activity in human adipose tissue. J Clin Endocrinol Metab 79:820825[Abstract]
- Janowski BA, Grogan MJ, Jones SA, Wisely GB, Kliewer SA, Corey EJ, Mangelsdorf DJ 1999 Structural requirements of ligands for the oxysterol liver X receptors LXR
and LXRß. Proc Natl Acad Sci USA 96:266271[Abstract/Free Full Text]
- Rosen ED, Spiegelman BM 2000 Molecular regulation of adipogenesis. Annu Rev Cell Dev Biol 16:145171[CrossRef][Medline]
- Repa JJ, Mangelsdorf DJ 2000 The role of orphan nuclear receptors in the regulation of cholesterol homeostasis. Annu Rev Cell Dev Biol 16:459481[CrossRef][Medline]
- Peet DJ, Janowski BA, Mangelsdorf DJ 1998 The LXRs: a new class of oxysterol receptors. Curr Opin Genet Dev 8:571575[CrossRef][Medline]
- Wolf G 1999 The role of oxysterols in cholesterol homeostasis. Nutr Rev 57:196198[Medline]
- Alberti S, Schuster G, Parini P, Feltkamp D, Diczfalusy U, Rudling M, Angelin B, Bjorkhem I, Pettersson S, Gustafsson JA 2001 Hepatic cholesterol metabolism and resistance to dietary cholesterol in LXRß-deficient mice. J Clin Invest 107:565573[Abstract/Free Full Text]
- Whitney KD, Watson MA, Goodwin B, Galardi CM, Maglich JM, Wilson JG, Willson TM, Collins JL, Kliewer SA 2001 LXR regulation of the LXR
gene in human macrophages. J Biol Chem 276:4350943515[Abstract/Free Full Text]
- Laffitte BA, Joseph SB, Walczak R, Pei L, Wilpitz DC, Collins JL, Tontonoz P 2001 Autoregulation of the human liver X receptor
promoter. Mol Cell Biol 21:75587568[Abstract/Free Full Text]
- Spydevold O, Bremer J 1989 Induction of peroxisomal ß-oxidation in 7800 C1 Morris hepatoma cells in steady state by fatty acids and fatty acid analogues. Biochim Biophys Acta 1003:7279[Medline]
- Lin FT, Lane MD 1992 Antisense CCAAT/enhancer-binding protein RNA suppresses coordinate gene expression and triglyceride accumulation during differentiation of 3T3L1 preadipocytes. Genes Dev 6:533544[Abstract]
- Sorensen HN, Gautvik KM, Bremer J, Spydevold O 1992 Induction of the three peroxisomal ß-oxidation enzymes is synergistically regulated by dexamethasone and fatty acids, and counteracted by insulin in Morris 7800C1 hepatoma cells in culture. Eur J Biochem 208:705711[Abstract]
- Schuster GU, Parini P, Wang L, Alberti S, Steffensen KR, Hansson GK, Angelin B, Gustafsson JA 2002 Accumulation of foam cells in liver X receptor-deficient mice. Circulation 106:11471153[Abstract/Free Full Text]
- Folch J, Lees M, Sloane-Stanley GH 1956 A simple method for the isolation and purification of total lipids from animal tissues. J Biol Chem 226:497509[Free Full Text]
- Wu Z, Bucher NL, Farmer SR 1996 Induction of peroxisome proliferator-activated receptor
during the conversion of 3T3 fibroblasts into adipocytes is mediated by C/EBPß, C/EBP
, and glucocorticoids. Mol Cell Biol 16:41284136[Abstract]