Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Cellular retinol-binding protein (CRBP) type III (CRBP-III) belongs to the family of intracellular lipid-binding proteins, which includes the adipocyte-binding protein aP2. In the cytosol, CRBP-III binds retinol, the precursor of retinyl ester and the active metabolite retinoic acid. The goal of the present work is to understand the regulation of CRBP-III expression and its role in lipid metabolism. Using EMSAs, luciferase reporter assays, and chromatin immunoprecipitation assays, we found that CRBP-III is a direct target of peroxisome proliferator-activated receptor-gamma (PPARgamma). Moreover, CRBP-III expression was induced in adipose tissue of mice after treatment with the PPARgamma agonist rosiglitazone. To examine a potential role of CRBP-III in regulating lipid metabolism in vivo, CRBP-III-deficient (C-III-KO) mice were maintained on a high-fat diet (HFD). Hepatic steatosis was decreased in HFD-fed C-III-KO compared with HFD-fed wild-type mice. These differences were partly explained by decreased serum free fatty acid levels and decreased free fatty acid efflux from adipose tissue of C-III-KO mice. In addition, the lack of CRBP-III was associated with reduced food intake, increased respiratory energy ratio, and altered body composition, with decreased adiposity and increased lean body mass. Furthermore, expression of genes involved in mitochondrial fatty acid oxidation in brown adipose tissue was increased in C-III-KO mice, and C-III-KO mice were more cold tolerant than wild-type mice fed an HFD. In summary, we demonstrate that CRBP-III is a PPARgamma target gene and plays a role in lipid and whole body energy metabolism.

Free full text 


Logo of ajpendoLink to Publisher's site
Am J Physiol Endocrinol Metab. 2008 Dec; 295(6): E1358–E1368.
Published online 2008 Oct 7. https://doi.org/10.1152/ajpendo.90464.2008
PMCID: PMC2603557
PMID: 18840764

Cellular retinol-binding protein type III is a PPARγ target gene and plays a role in lipid metabolism

Associated Data

Supplementary Materials

Abstract

Cellular retinol-binding protein (CRBP) type III (CRBP-III) belongs to the family of intracellular lipid-binding proteins, which includes the adipocyte-binding protein aP2. In the cytosol, CRBP-III binds retinol, the precursor of retinyl ester and the active metabolite retinoic acid. The goal of the present work is to understand the regulation of CRBP-III expression and its role in lipid metabolism. Using EMSAs, luciferase reporter assays, and chromatin immunoprecipitation assays, we found that CRBP-III is a direct target of peroxisome proliferator-activated receptor-γ (PPARγ). Moreover, CRBP-III expression was induced in adipose tissue of mice after treatment with the PPARγ agonist rosiglitazone. To examine a potential role of CRBP-III in regulating lipid metabolism in vivo, CRBP-III-deficient (C-III-KO) mice were maintained on a high-fat diet (HFD). Hepatic steatosis was decreased in HFD-fed C-III-KO compared with HFD-fed wild-type mice. These differences were partly explained by decreased serum free fatty acid levels and decreased free fatty acid efflux from adipose tissue of C-III-KO mice. In addition, the lack of CRBP-III was associated with reduced food intake, increased respiratory energy ratio, and altered body composition, with decreased adiposity and increased lean body mass. Furthermore, expression of genes involved in mitochondrial fatty acid oxidation in brown adipose tissue was increased in C-III-KO mice, and C-III-KO mice were more cold tolerant than wild-type mice fed an HFD. In summary, we demonstrate that CRBP-III is a PPARγ target gene and plays a role in lipid and whole body energy metabolism.

Keywords: adipose tissue, retinoid, liver steatosis, lipolysis, brown adipose tissue, fatty acid-binding protein, intracellular lipid-binding protein, retinol-binding protein-7

white adipose is the primary tissue for storage of triglycerides and acts as an endocrine organ to secrete adipokines, which impact insulin sensitivity in nonadipose tissues (38, 43). Adipose tissue also plays a role in retinoid metabolism. It has been estimated that ~10–20% of total retinoids in the body are stored as retinyl ester in adipocytes (52). This storage appears not to merely serve as a passive storage tissue for retinoids. Rather, during insufficient vitamin A intake and low hepatic retinoid levels, adipose tissue stores appear to be readily mobilized (25, 33). Retinoids also act to modulate adipose tissue structure and metabolic function. All-trans retinoic acid (RA) can inhibit adipogenesis by activation of the RA nuclear hormone receptor (RAR) (10, 28). In contrast, activation of the retinoid X receptor (RXR) leads to enhancement of adipogenesis and insulin sensitivity (29). Recently, retinaldehyde (RAL), produced in the adipocytes, was shown to inhibit adipogenesis and, subsequently, the development of diet-induced obesity (55). In addition, data indicate that plasma retinol-binding protein, when secreted from adipose tissue, may act as an adipokine, reducing insulin sensitivity in mice (54). Intracellularly, retinol is the precursor for retinyl ester formation and RA synthesis, with RAL as the intermediate metabolite (5, 30). Similar to fatty acid sequestration by fatty acid-binding proteins in the cytosol, retinol is bound by intracellular cellular retinol-binding proteins (CRBPs) (21, 31). CRBP-I is ubiquitously expressed, with the highest expression in liver, kidney, testes, and eye. CRBP-III and CRBP-I have overlapping expression in heart, muscle, adipose, and mammary tissue (34, 36, 53). CRBP-III binds retinol, and we previously proposed that its function is to facilitate the esterification of retinol to retinyl ester in the mammary gland during lactation (36). Our next goal is to further determine the function of CRBP-III. Our first objective was to analyze the expression of CRBP-III during adipogenesis and, subsequently, determine how its expression may be regulated. We present data that CRBP-III is a peroxisome proliferator-activated receptor-γ (PPARγ) target gene. Since PPARγ is essential for adipogenesis and required for maintenance of normal lipid homeostasis (11, 20, 50), the objective of the present study was to understand the role of CRBP-III in lipid metabolism. We pursued our objective by examining CRBP-III-knockout (C-III-KO) mice during the metabolic challenge of a high-fat diet (HFD).

MATERIALS AND METHODS

Cell culture.

Mouse 3T3-L1 preadipocytes were cultured and differentiated as previously described (14).

Isolation of RNA and protein.

Total RNA was isolated using RNA-Bee (Tel-Test). Protein extracts were prepared in a buffer composed of Tris-Cl (20 mM, pH 7.5), EDTA (5 mM), EGTA (2 mM), NP-40 (1%), NaF (100 mM), NaVO3 (2 mM), NaP2O7 (10 mM), and proteinase and phosphatase inhibitor cocktails. Western blot analyses were essentially performed as described previously (36, 53). Antibodies for detection of CRBP-III have been described elsewhere (36). Antibodies to detect AMP-activated protein kinase and acetyl-CoA carboxylase (ACC) and their phosphorylation status were obtained from Cell Signaling (Danvers, MA). Separation of brown adipose tissue (BAT) into stromal vascular fraction and adipocytes was accomplished by collagenase digestion before Western blot analysis (53).

Quantitative real-time PCR.

cDNA was synthesized from total RNA (3 μg) using SuperScript III reverse transcriptase (Invitrogen). The final reaction for the quantitative real-time PCR consisted of 0.5 μM gene-specific primer, 1× SYBR Green PCR Master Mix (Applied Biosystems), and 10 μl of diluted cDNA. The reaction was carried out on a Stratagene cycler. (See supplemental Table S1 in the online version of this article for primer sequences.)

Reporter constructs for CRBP-III promoter and luciferase assays.

A 3.1-kb fragment upstream of the site of transcription was amplified from mouse genomic DNA (C57/Bl6) using a high-fidelity PCR system (Expand, Roche Diagnostics) and cloned into the Zero Blunt TOPO PCR vector (Invitrogen). The fragment was completely sequenced, and the results were compared with the published sequence. The 3.1-kb promoter fragment was cloned into the pGL3 basic vector (Promega) and designated pGL3k, which was used as a template for generation of two shorter promoter constructs, 2.5 kb (pGL2.5k) and 2 kb (pGL2k). A PCR approach was used to introduce mutations of the putative PPARγ response elements (PPREs). Putative PPRE1 (mut1), PPRE3 (mut3), or PPRE1 and PPRE3 (mut1 and mut3) were mutated. The primers used to introduce the mutations were 5′-GATCCATAACTGTGTACACAGCCGAAATTTCTGGAG-3 for PPRE1mut and 5′- CTGACTGACTTACACAGCCACAGTTTCCCTCTTACGGG-3′ for PPRE3mut. All constructs were sequenced to ensure the correct mutations. 3T3-L1 cells were differentiated in six-well plates and used at day 7 of differentiation. Cells were transfected with 0.2 μg of firefly luciferase DNA constructs (pGL3k, pGL2.5k, or pGL2k). COS-7 cells were plated for 24 h in 12-well plates in complete medium and transfected with firefly luciferase DNA constructs and with pcDNA3.1 alone or equivalent concentrations of pcDNA3.1-PPARγ2 and pcDNA3.1-RXRα, which overexpress PPARγ2 and RXRα, respectively. The Renilla luciferase vector (0.05 μg) was included in all transfection procedures as a control. Transfections were carried out overnight; on the next morning, the medium was replaced with complete medium containing 1 μM rosiglitazone or vehicle (ethanol). After 24 h, the dual luciferase reporter assay (Promega) was performed using a luminometer (TD-20/20, Turner Designs). Results were expressed as relative luciferase activity, with normalization of firefly luciferase activity to Renilla luciferase activity.

EMSA.

Nuclear proteins were isolated from COS-7 cells transiently expressing PPARγ2 and RXRα using a nuclear protein extraction kit (Pierce). The probes were prepared by annealing oligonucleotide and end-labeling the double-stranded DNA with [γ-32P]ATP using T4 kinase (Promega). The PPRE of the adipocyte-binding protein aP2 promoter was used as a positive control (50). Oligonucleotides were as follows: aP2-PPRE (5′-GATCTGTGACCTTTGACCTAGTAAG-3′, 5′-CTTACTAGGTCAAAGGTCACAGATC-3′), CRBP-III PPRE1 (5′-GAAATTTGGGCAGAGTTCACAGTTA-3′, 5′-TAACTGTGAACTCTGCCCAAATTTC-3′), CRBP-III PPRE2 (5′-ATGGATCCCTCAAAGGTCAAGGAAT-3′, 5′-ATTCCTTGACCTTTGAGGGATCCAT-3′), and CRBP-III PPRE3 (5′-TCTCCCTTGGAAAAGGTCACATTCA-3′, 5′-TGAATGTGACCTTTTCCAAGGGAGA-3′). The DNA-protein binding assays were performed as recommended by the manufacturer (Promega). A supershift assay was performed using PPARγ monoclonal (E8, Santa Cruz Biotechnology) or RXRα polyclonal (D-20, Santa Cruz Biotechnology) antibody. The binding reaction was incubated with the indicated antibody for 10 min at room temperature before addition of the labeled nucleotides. Electrophoresis of the DNA-protein complexes was performed on 6% DNA retardation gels (Invitrogen), which were placed on Whatman paper and dried using a vacuum gel dryer. The signals were detected using a Storm PhosphorImager (Amersham Biosciences).

Chromatin immunoprecipitation assay.

Differentiated 3T3-L1 adipocytes were used for the chromatin immunoprecipitation assays. After cross-linking of DNA and proteins in situ, the soluble chromatin was prepared according to the manufacturer's instructions (Upstate), and the DNA was sheared. The protein-chromatin complex was immunoprecipitated with PPARγ antibodies, and PCRs that included regions of PPRE1 and PPRE3 were performed.

Animal studies.

Three-month-old male wild-type (C57BL/6) mice (Jackson Laboratory, Bar Harbor, ME) were fed a chow diet or a diet supplemented with rosiglitazone (10 mg/kg body wt) for 21 days. Generation of mice lacking CRBP-III (C-III-KO) has previously been reported (36). The mice were backcrossed to the C57BL/6 genetic background for five generations before the experiments. C-III-KO and CRBP-III wild-type mice were fed an HFD providing 60% of calories from fat (Research Diets). The mice were fed the HFD for 20 wk and then killed, the liver was perfused with PBS, and tissues were excised, flash frozen in liquid nitrogen, and stored at −70°C until analysis. Experiments involving mice were performed with the approval of the Institutional Animal Care and Use Committee at Columbia University.

Histology.

Liver, white adipose tissue, and BAT were placed in 10% buffered formalin overnight and then embedded in paraffin. Tissue sections (5 μm) were stained with hematoxylin-eosin. Liver sections were also fresh frozen in OCT compound, sectioned (7 μm), and stained with oil red O for detection of neutral lipids.

Retinoid measures.

Retinol and retinyl ester levels in liver and adipose tissue were determined as previously described (36).

Lipolysis assay of isolated white adipose tissue.

Lipolysis assays were performed essentially as previously described (35, 42). White adipose tissue from HFD-fed wild-type and C-III-KO mice was excised, washed in PBS, and cut into small (~50 mg) pieces. The explants were incubated in Krebs-Ringer buffer for 3 h at 37°C in the absence or presence of isoproterenol (10 μM). At the end of the incubation, the plates were placed on ice, and the medium was removed for glycerol and free fatty acid analyses using commercial kits (Sigma Aldrich and Wako). Tissue pieces were washed and saved for protein analysis. For each animal, the assay was performed in quadruplicate.

Hepatic triglyceride levels.

Triglycerides were extracted from liver using a Folch extraction. Briefly, livers were homogenized in PBS and incubated in chloroform-methanol (2:1) in a rotator at room temperature for 1 h, and the chloroform phase was dried under a mild stream of nitrogen. The triglycerides were reconstituted in cold isopropanol, and levels were determined using a commercial kit (Thermo Scientific).

Serum parameters.

Blood for determination of lipid parameters, glucose, insulin, and adipokine levels was obtained from mice fasted during the day for 6 h (9 AM–3 PM) after 18 wk on the HFD. Serum was obtained after the blood samples had been allowed to clot. Commercially available kits were used to measure serum levels of nonessential fatty acids (Wako), free glycerol (Sigma Aldrich), β-hydroxybutyrate (Stanbio), total insulin (Mercodia), glucose (Invitrogen), and total cholesterol and triglyceride (Thermo Scientific) levels. Serum leptin and resistin levels were determined using a Multiplex Assay (Millipore).

Cold tolerance testing.

Cold tolerance was assessed in wild-type and C-III-KO mice fed the HFD for 20 wk by exposure to 4°C for 2 h. Rectal temperature was measured before the cold exposure and then at 1-h intervals during the cold challenge using a thermocouple probe (model IT-23) attached to a thermocouple amplifier (Physitemp BAT-10 Type I, Pharma).

Body composition analysis.

Body composition was measured by MRI using an EchoMRI (Echo Medical Systems).

Metabolic studies.

Animals were individually housed in metabolic chambers maintained at 20–22°C on a 12:12-h light-dark cycle, with lights on at 7 AM. Metabolic measurements (oxygen consumption, food intake, and locomotor activity) were obtained continuously using a Comprehensive Lab Animal Monitoring System (Columbus Instruments) open-circuit indirect calorimetry system. Metabolic data were collected over 5 consecutive days following 3 days of adaptation to the metabolic cages.

Statistical analysis.

Values are means ± SE. Statistical differences were determined using Student's t-test. Statistical significance was considered at P < 0.05.

RESULTS

CRBP-III expression is regulated by PPARγ in vitro and in vivo.

We previously showed that, in white adipose tissue, CRBP-III is expressed in the stromal vascular fraction and in adipocytes (53). To further understand the role of CRBP-III in adipocytes, we differentiated 3T3-L1 preadipocytes and analyzed gene expression at different stages of adipocyte differentiation. CRBP-III mRNA was detected 4 days and protein 7 days after the start of differentiation, suggesting posttranscriptional modification (Fig. 1, A and B). On the basis of the time of induction of CRBP-III expression, we hypothesized that CRBP-III could be regulated by PPARγ. After treatment of differentiated adipocytes with the PPARγ agonist rosiglitazone, CRBP-III expression levels were significantly increased compared with those measured in untreated cells (Fig. 1, A and B). Next, to test whether PPARγ also regulates CRBP-III expression in vivo, we fed wild-type mice a chow diet alone or a chow diet enriched with the PPARγ agonist rosiglitazone. CRBP-III expression levels were increased in white adipose tissue of mice fed the rosiglitazone-enriched diet compared with mice fed the unsupplemented chow diet (Fig. 1C). As a comparison, expression of aP2, a well-known target of PPARγ (49), also increased in mice fed the rosiglitazone-enriched diet (Fig. 1C).

An external file that holds a picture, illustration, etc.
Object name is zh10120855090001.jpg

Cellular retinol-binding protein type III (CRBP-III) is expressed in mature adipocytes, and its expression is regulated by peroxisome proliferator-activated receptor (PPAR)-γ. A and B: Northern and Western blot analysis, respectively, of CRBP-III expression during differentiation of 3T3-L1 preadipocytes into adipocytes and after no treatment (−) and rosiglitazone (Rosi) treatment (+). Days, days after start of differentiation; aP2, adipose fatty acid-binding protein (FABP4). C: Northern blot analysis of CRBP-III expression in white adipose tissue of mice fed rosiglitazone-enriched (+) and unsupplemented (−) diet. D: region located in CRBP-III gene untranslated region between 2.5 and 2 kb upstream of transcriptional start site is important for rosiglitazone response. Relative luciferase activity was assessed in differentiated 3T3-L1 adipocytes after treatment with vehicle (dark bar) or rosiglitazone (light bar). E: EMSA reveals 2 potential PPARγ response element (PPRE) regions (PPRE1 and PPRE3) in CRBP-III promoter that bind PPARγ/retinoid X receptor (RXR)-α heterodimer. PPRE located in aP2 promoter (aP2-PPRE) was used as a positive control. comp, Competitor; nonspec, nonspecific. F: PPARγ binds to PPRE1 and PPRE3 in vivo, as analyzed by chromatin immunoprecipitation assay. 1, Water control; 2, input; 3, anti-PPARγ antibody; 4, nonspecific anti-IgG; M, DNA size marker. G: single mutations in PPRE1 (PPRE1-mut) or PPRE3 (PPRE3-mut) lead to reduction in response to rosiglitazone (light bars), but response is abolished only in double mutant (PPRE1&3-mut). Luciferase assays show results of treatment of transfected COS-7 cells without (dark bars) and with (light bars) rosiglitazone. WT, wild-type. Values are means ± SE relative to untreated transfected cells (n = 3 per transfection, 2 independent experiments). *P < 0.05 vs. untreated.

CRBP-III promoter contains two functional PPREs.

PPARγ elicits its transcriptional effect through binding as a heterodimer with RXRα to specific PPREs in the noncoding sequences of genes. We cloned different-sized promoter fragments upstream of the transcriptional start site to understand whether a PPRE is present in the CRBP-III promoter. All promoter constructs (3, 2.5, and 2 kb) showed basal activity in fully differentiated 3T3-L1 adipocytes (Fig. 1D). However, treatment with rosiglitazone increased relative luciferase activity up to threefold over basal activity for a promoter fragment including the region 2.5 and 3 kb upstream of the transcriptional start site (Fig. 1D). In contrast, for the shorter (2-kb) promoter construct, no change relative to basal activity was observed after rosiglitazone treatment. Similar data were obtained when the promoter constructs were cotransfected with PPARγ and RXRα into COS-7 cells, where treatment with rosiglitazone only increased luciferase activity for the 3- and 2.5-kb constructs (see supplemental Fig. S1A). Taken together, these data indicate that a putative PPRE is present in a 500-bp region between 2.0 and 2.5 kb upstream of the transcriptional start site. Analyzing the DNA sequence of this 500-bp region, we identified three putative PPRE sites that contained the PPRE consensus sequence (22).

On the basis of EMSAs, PPARγ and RXRα bind to two PPRE sites (PPRE1 and PPRE3), forming a single DNA-protein complex (Fig. 1E). In contrast, there was no specific band for PPRE2. Binding of the labeled PPRE to the nuclear protein appears to be specific, inasmuch as it can be competed off with excess of specific competitor, but not with nonspecific DNA (Fig. 1E). When antibodies against PPARγ or RXRα were used, the DNA-protein complex supershifted to a higher molecular weight, further indicating that PPARγ and RXRα are binding to the specific DNA region in PPRE1 and PPRE3 (see supplemental Fig. S1B). Furthermore, on the basis of chromatin immunoprecipitation assay, PPARγ binds to the PPRE region including PPRE1 and PPRE3 in vivo (Fig. 1F). Because PPRE1 and PPRE3 are within close proximity (132 bp apart), we were unable to discern whether PPARγ binds to PPRE1 or PPRE3 or both. Therefore, we generated promoter constructs that contained mutations in one PPRE alone or in both. Mutations in either PPRE alone lead to a decreased response to rosiglitazone treatment. Importantly, the response to rosiglitazone was abolished only when both PPREs were mutated (Fig. 1G). Taken together, these data demonstrate that PPARγ agonists regulate CRBP-III expression in vivo and in vitro through two PPREs present in the promoter of CRBP-III.

Hepatic steatosis is reduced in HFD-fed C-III-KO mice.

PPARγ is essential for normal adipogenesis and critically required for maintaining normal glucose and lipid homeostasis (11, 20, 50). Since CRBP-III is regulated by PPARγ in adipose tissue, we sought to understand its role in regulating lipid homeostasis by studying mice with a gene-targeted deletion of CRBP-III (C-III-KO) (36).

Since excess dietary fat leads to an expansion of adipose tissue and, often, to perturbations in energy metabolism, we examined the effects of diet-induced obesity in C-III-KO compared with wild-type mice. Weight gain in HFD-fed animals was not different between the two groups (Fig. 2A). In addition, serum glucose and insulin levels did not differ between HFD-fed wild-type and C-III-KO mice (Table 1). Next, we examined liver, white adipose tissue, and BAT for differences between wild-type and C-III-KO mice. As anticipated, livers from HFD-fed wild-type mice contained abundant, large lipid droplets (Fig. 2B). In contrast, livers from C-III-KO mice contained significantly fewer lipid droplets (Fig. 2B). In agreement with these observations, hepatic triglyceride concentrations were significantly lower in C-III-KO than wild-type mice (Fig. 2C). Hepatic retinyl ester levels were not different between the two groups (Fig. 2D).

An external file that holds a picture, illustration, etc.
Object name is zh10120855090002.jpg

A: effect of high-fat diet (HFD) on body weight in WT and CRBP-III-deficient (C-III-KO) mice. B: lipid accumulation in livers from C-III-KO and WT mice, as judged by hematoxylin-eosin (top) and oil red O (bottom) staining. Images are representative of observations of 7 mice per group. C: triglyceride (TAG) content in livers is significantly lower in C-III-KO than WT mice fed HFD for 20 wk. Values are means ± SE (n = 9 for both genotypes). *P < 0.05 vs. WT. D: retinol and retinyl ester levels in the liver were not significantly different between WT and C-III-KO mice.

Table 1.

Fasted serum parameters in wild-type and C-III-KO mice fed HFD for 18 wk

WT (n = 9)C-III-KO (n = 10)
Glucose, mM12.3±1.513.5±2.7
Insulin, μg/l7.8±1.37.2±3.3
Triglycerides, mmol/l0.26±0.090.28±0.06
Resistin, pg/ml2,652±1793,163±336
Leptin, pg/ml10,588±2,1608,963±1,181

Values are means ± SE. Wild-type (WT) and cellular retinol-binding protein (CREB) type III-deficient (C-III-KO) mice were fasted for 6 h during the daytime before blood was drawn. HFD, high-fat diet. There was no significant difference between WT and C-III-KO for any parameter.

Since CRBP-III is not expressed in the liver but is expressed at high levels in adipose tissue (8, 36, 53), decreased hepatic triglyceride may be secondary to alterations in adipose tissue of C-III-KO mice. One possibility is that altered adipokine levels in C-III-KO mice may be responsible for the decreased fat accumulation in the liver. However, we detected no differences in levels for serum leptin and resistin (Table 1) and expression of adiponectin (see Table 3) between C-III-KO and wild-type mice. Similarly, genes involved in hepatic glucose and fatty acid metabolism were not altered in the liver of C-III-KO mice (Table 2). We detected no differences in the liver for expression of genes for glucose metabolism (GLUT2, phosphoenolpyruvate carboxykinase, and glucose-6-phosphatase), fatty acid synthesis (ACC, fatty acid synthase, stearoyl CoA-desaturase, and sterol regulatory element-binding protein-1c), or fatty acid oxidation (PPARα and acyl-coenzyme A dehydrogenase, long-chain). Similarly, in the liver, phosphorylation of AMP-activated kinase and its downstream target ACC were not different between HFD-fed C-III-KO and wild-type mice (Table 2). These data imply that there was no apparent change in fatty acid synthesis or oxidation in the liver of HFD-fed C-III-KO mice that would account for the decreased hepatic lipid accumulation.

Table 2.

Expression levels of genes involved in hepatic glucose and lipid metabolism in wild-type and C-III-KO mice fed HFD for 20 wk

GeneWTC-III-KO
GLUT20.59±0.130.59±0.08
PEPCK1.42±0.481.63±0.49
G6P1.93±0.381.68±0.69
ACC1.50±0.351.09±0.47
FAS0.67±0.220.52±0.15
SCD-12.46±1.151.50±0.50
SREBP-1c1.54±0.252.16±0.37
PPARα0.93±0.221.05±0.22
ACAD-M2.49±0.462.01±0.57
ACAD-L0.76±0.390.51±0.25
ACAD-VL0.69±0.220.70±0.37

Values (relative expression levels) are means ± SE (n = 4 for each genotype). PEPCK, phosphoenolpyruvate carboxykinase; G6P, glucose-6-phosphatase; ACC, acetyl-CoA carboxylase; FAS, fatty acid synthase; SCD-1, stearoyl-CoA desaturase-1; SREBP-1c, sterol regulatory element-binding protein-1c; PPARα, peroxisome proliferator-activated receptor-α; ACAD, acyl-coenzyme A dehydrogenase [medium (M), long (L), and very long (VL) chain]. There was no significant difference between WT and C-III-KO for any gene expression level.

Table 3.

Expression levels of genes in white adipose tissue in wild-type and C-III-KO mice fed HFD for 20 wk

GeneWTC-III-KO
Adiponectin0.99±0.390.74±0.32
ATGL0.38±0.040.39±0.09
HSL0.61±0.090.71±0.1
MGL0.83±0.481.02±0.31
aP21.24±0.200.75±0.12
PGC-1α0.18±0.040.23±0.05
ACAD-M0.60±0.060.55±0.17
COX II0.30±0.020.31±0.02
ADH10.68±0.380.69±0.38
RALDH19.44±3.427.03±2.16
RBP41.62±0.372.46±0.92
CRBP-I1.39±0.460.80±0.20
RetSDR12.67±1.272.95±0.49
Tgm23.13±1.972.31±1.22

Values (relative expression levels) are means ± SE (n = 4 for each genotype). ATGL, adipose triglyceride lipase; HSL, hormone-sensitive lipase; MGL, monoglyceride lipase; aP2, adipose fatty acid-binding protein (FABP4); PGC-1α, PPARγ coactivator-1α; COX II, cytochrome c oxidase type II; ADH1, alcohol dehydrogenase type 1; RALDH1, retinaldehyde dehydrogenase type 1; RBP4, retinol-binding protein; RetSDR1, short-chain dehydrogenase/reductase type 1; Tgm2, tissue transglutaminase type 2. There was no significant difference between WT and C-III-KO for any gene expression level.

Decreased serum free fatty acid levels and in vitro adipose lipolysis in HFD-fed C-III-KO mice.

In diet-induced obesity, the majority of free fatty acids used for hepatic triglycerol synthesis originate from adipose tissue (6). Lipolysis of adipose triglyceride leads to release of nonesterified free fatty acids and glycerol into the circulation, a process that is further increased in states of insulin resistance (24, 38). Therefore, we examined serum levels of free fatty acids and glycerol (Fig. 3, A and B). Serum free fatty acid and free glycerol levels were significantly lower in C-III-KO than wild-type mice. This is indicative of decreased free fatty acid efflux from the adipose tissue of C-III-KO mice. To further understand these changes, we performed in vitro lipolysis assays. We isolated white adipose tissue from HFD-fed wild-type and C-III-KO mice and measured glycerol and free fatty acid release under basal and isoproterenol-stimulated conditions in vitro. Under basal conditions, glycerol and free fatty acid release were significantly lower from adipose tissue of C-III-KO than wild-type mice (Fig. 3C). This finding is in agreement with the decreased levels observed in vivo. On stimulation of lipolysis by isoproterenol, significantly less glycerol was released from adipose tissue of C-III-KO than wild-type mice, whereas the decrease in free fatty acid release did not reach statistical significance (P = 0.08; Fig. 3C). Gene expression analysis of white adipose tissue from wild-type and C-III-KO mice revealed no differences in expression of the lipolytic enzymes adipose triglyceride lipase, hormone-sensitive lipase, or monoglyceride lipase (Table 3). In addition, expression of genes involved in fatty acid oxidation in white adipose tissue is not different between C-III-KO and wild-type mice (Table 3). In the liver, free fatty acids are reesterified to triglyceride but are also oxidized to ketone bodies (13, 15). Ketone body formation is another surrogate marker for increased fatty acid release from adipose tissue and for uptake and oxidation by the liver in states of insulin resistance, such as diet-induced obesity (15). The levels of the ketone body β-hydroxybutyrate were significantly lower in serum of C-III-KO mice (Fig. 3D), consistent with the idea that less free fatty acid is reaching the liver. Taken together, these data indicate that a decreased flux of free fatty acids from the adipose tissue to the liver may contribute to the decreased hepatic triglyceride content in C-III-KO mice.

An external file that holds a picture, illustration, etc.
Object name is zh10120855090003.jpg

Serum levels of nonessential free fatty acids (A) and free glycerol (B) are significantly lower in C-III-KO than WT mice. Values are means ± SE (n = 10 for each genotype). *P < 0.05 vs. WT. C: in vitro lipolysis assay of adipose tissue isolated from HFD-fed WT and C-III-KO mice. Glycerol and free fatty acid (FFA) release was significantly lower in the basal state in C-III-KO than WT mice. In the stimulated state, free glycerol release is significantly lower in C-III-KO mice, and the difference in free fatty acid release between WT and C-III-KO mice did not reach significance (P = 0.08). Values are means ± SE (n = 5 mice for each genotype performed in quadruplicate). *P < 0.05. D: serum levels of β-hydroxybutyrate are significantly lower in C-III-KO than WT mice. Values are means ± SE (n = 10 for each genotype). *P < 0.05 vs. WT.

White adipose tissue morphology of HFD-fed C-III-KO and wild-type mice showed the prevalence of large unilocular droplets, with no difference in adipocyte size at the end of the HFD (data not shown). Similar to the liver, no differences between wild-type and C-III-KO mice were detected for retinol or retinyl ester in the white adipose tissue (Table 4). In addition, expression in adipose tissue of genes related to retinoid metabolism, such as alcohol dehydrogenase type 1, retinal dehydrogenase type 1, CRBP-I, and retinol-binding protein, was not altered in C-III-KO compared with wild-type mice (Table 3). Although we did not directly measure RA levels, target genes activated by RA (short-chain dehydrogenase/reductase and tissue transglutaminase type 2) were not different between wild-type and C-III-KO mice in white adipose tissue (Table 3).

Table 4.

Retinol and retinyl ester levels in white adipose tissue from wild-type and C-III-KO mice fed HFD for 20 wk

WTC-III-KO
Retinol, μg/g0.31±0.060.23±0.02
Retinyl ester, μg/g1.89±0.211.76±0.90

Values are means ± SE (n = 8 for each genotype). There was no significant difference between WT and C-III-KO for retinol or retinyl ester.

Body composition, respiratory exchange ratio (RER), and food intake are altered in C-III-KO mice. To gain further mechanistic insight into the role of CRBP-III deficiency in lipid metabolism, we assessed body composition, RER, and food intake in wild-type and C-III-KO mice. After 20 wk of HFD feeding, body weights were not different between wild-type and C-III-KO mice (Figs. 2A and and4A).4A). However, MRI analysis of body composition revealed a small, but significant, reduction in fat mass accompanied by an increase in lean mass in C-III-KO compared with wild-type mice (Fig. 4A). To determine the basis for the decreased adiposity in C-III-KO mice, we assessed food intake during the HFD feeding. As shown in Fig. 4B, food intake was lower in C-III-KO than wild-type mice. Thus food intake may contribute to the reduction in adiposity. Because CRBP-III is not expressed in the brain (see supplemental Fig. S2), the reduced food intake in C-III-KO mice is likely due to central nervous system processing of the peripheral metabolic consequences of CRBP-III deficiency. Furthermore, there was a nonsignificant trend toward reduced oxygen consumption (Fig. 4C) and energy expenditure: 8.1 ± 0.1 and 7.6 ± 0.2 (daytime), 8.9 ± 0.1 and 8.5 ± 0.2 (nighttime), and 8.5 ± 0.1 and 8.1 ± 0.2 kcal·kg−1·h−1 (total) for wild-type and C-III-KO mice, respectively.

An external file that holds a picture, illustration, etc.
Object name is zh10120855090004.jpg

Metabolic analysis of WT and C-III-KO mice fed HFD for 20 wk. A: analysis of body composition of WT and C-III-KO mice. Total body weight was not different between WT and C-III-KO mice, but fat mass was decreased and lean mass was increased in C-III-KO compared with WT mice. Values are means ± SE (n = 6 for each genotype). B: C-III-KO mice consume less food than WT mice. Values are means ± SE (n = 6 for each genotype). C: oxygen consumption (Vo2) of HFD-fed WT and C-III-KO mice. Values are means ± SE (n = 4 for each genotype). D: respiratory exchange ratio (RER) was significantly higher for C-III-KO than WT mice during light and dark phases. Values are means ± SE (n = 4 for each genotype). E: activity levels were not different between WT and C-III-KO mice. Ambulatory and rearing activity were measured and combined to express total activity. Values are means ± SE (n = 4 for each genotype). *P < 0.05 vs. WT.

Inasmuch as CRBP-III is expressed in the muscle (53), we also performed gene expression studies in muscle. Soleus muscle expression of genes involved in fatty acid oxidation did not differ between wild-type and C-III-KO mice (see supplemental Table S2). This finding is further substantiated by results from indirect calorimetry. RER was slightly, but significantly, increased in C-III-KO compared with wild-type mice, indicating an increase in glucose, rather than fatty acid, oxidation (Fig. 4D). In addition, there was no difference in locomotor activity between wild-type and C-III-KO mice (Fig. 4E).

Maintenance of BAT function in HFD-fed C-III-KO mice.

CRBP-III is also highly expressed in BAT, in addition to white adipose tissue (see supplemental Fig. S3). Therefore, we examined this tissue following HFD consumption. Consistent with previous reports, morphology of BAT from HFD-fed wild-type mice was altered, with large unilocular lipid droplets and only a few multilocular lipid droplets characteristic of brown adipocytes (Fig. 5A). In contrast, BAT from C-III-KO mice is characterized by multilocular lipid droplets and contained only a few interspersed large lipid droplets (Fig. 5A). We next determined whether these changes were also reflected in altered gene expression between wild-type and C-III-KO mice. We found that genes involved in mitochondrial oxidative metabolism were expressed in higher levels in BAT from C-III-KO than wild-type mice. Expression of genes involved in oxidative phosphorylation (cytochrome c and cytochrome c oxidase II and VIII) and the citric acid cycle (aconitase) was significantly higher in BAT from C-III-KO than wild-type mice (Fig. 5B). In addition, PPARα levels and the expression of its downstream target genes for fatty acid oxidation (medium- and long-chain acyl-CoA dehydrogenase) were significantly higher in BAT from C-III-KO than wild-type mice (Fig. 5C). There was a trend for an increase in uncoupling protein-1 (UCP-1) expression levels in BAT of C-III-KO mice, but the difference did not reach statistical significance: 0.86 ± 0.07 and 1.40 ± 0.18 (SE) in wild-type and C-III-KO, respectively (P = 0.06, n = 4 each). To understand the functional implications of these differences for thermogenesis, we subjected mice to a 2-h cold challenge. C-III-KO mice were significantly more tolerant to the cold than were wild-type mice (Fig. 5D). Taken together, these results suggest that BAT from C-III-KO mice is phenotypically and functionally conserved during HFD feeding, in contrast to BAT from wild-type mice. This conclusion is further underscored by the fact that thermogenesis may be more preserved in C-III-KO than wild-type mice during a cold challenge.

An external file that holds a picture, illustration, etc.
Object name is zh10120855090005.jpg

Brown adipose tissue (BAT) remains more intact in HFD-fed C-III-KO than WT mice. A: hematoxylin-eosin staining of BAT in HFD-fed WT and C-III-KO mice. Images are representative of results from 7 mice per group. Expression of key genes involved in oxidative phosphorylation and citric acid cycle (B) and in fatty acid oxidation (C) was significantly higher in C-III-KO than WT mice. Expression is shown relative to expression of cyclophilin. Values are means ± SE (n = 4 for each genotype). *P < 0.05 vs. WT. COX, cytochrome c oxidase; ACAD-L/M, acyl-CoA dehydrogenase long-chain/medium-chain. D: changes in body temperature during exposure of HFD-fed C-III-KO and WT mice to 4°C for 2 h. C-III-KO mice were more cold tolerant than WT mice. *P < 0.05 vs. WT.

DISCUSSION

In the present study, we demonstrate that CRBP-III is a PPARγ target gene and plays a role in lipid homeostasis under conditions of diet-induced obesity.

CRBP-III is a small cytosolic protein belonging to the intracellular lipid-binding protein family, which, in addition to the CRBPs (I, II, III), includes cellular RA binding proteins (CRABP I and CRABP II) and fatty acid-binding proteins, such as aP2 (4, 31). CRBPs bind retinol, which can be metabolized intracellularly to RAL and RA, the latter being the major active retinoid (5, 31, 34). In addition, retinol can be esterified to retinyl ester for storage (5, 34). It is thought that the primary role of CRBP-I and CRBP-III is facilitatation of the esterification of retinol to retinyl ester catalyzed by lecithin:retinol acyltransferase (16, 36). Absence of CRBP-I leads to a significant reduction of hepatic retinyl ester storage (16). Similarly, the lack of CRBP-III leads to decreased retinol esterification during lactation (36). In the present study, we expanded the possible roles of CRBP-III. We show that the expression of CRBP-III can be directly modulated by PPARγ agonists. In this context, we elucidated that the promoter of CRBP-III harbors functional PPREs for binding of PPARγ in vitro and in vivo. PPARγ, through its transcriptional regulation of a plethora of genes, is essential for normal adipose tissue development and systemic energy homeostasis (2, 19, 23, 37, 50).

Therefore, to understand the potential role of CRBP-III, we examined gene-targeted mice lacking CRBP-III during diet-induced obesity. Overall, the lack of CRBP-III has distinct effects on 1) whole body energy balance, 2) the oxidative machinery in BAT, and 3) serum free fatty acid levels and white adipose tissue lipid metabolism.

CRBP-III and whole body energy metabolism.

In the present study, the lack of CRBP-III appears to affect whole body energy balance. Specifically, lack of CRBP-III resulted in a reduction in food intake and a nonsignificant trend toward reduced energy expenditure and oxygen consumption. Body weight was similar between C-III-KO and wild-type mice fed the HFD, suggesting that decreased food intake in C-III-KO mice may be balanced by decreased energy expenditure. However, fat mass was significantly reduced in C-III-KO compared with wild-type mice. This decrease in fat mass was offset by an increase in lean body mass, resulting in no net change in total mass relative to that of wild-type mice. The increased lean body mass would be predicted to increase the metabolic load in C-III-KO mice, which would, in turn, mitigate against a more significant reduction in oxygen consumption and energy expenditure.

The small, but significant, increase in RER in C-III-KO mice indicates increased carbohydrate, rather than fat, oxidation. This interpretation is consistent with our observation that expression of genes important in fatty acid oxidation was unchanged in skeletal muscle of C-III-KO compared with wild-type mice. Overall, the lack of CRBP-III induces a distinctive profile of behavioral and metabolic consequences, consisting of a change in nutrient partitioning, reduced feeding, decreased fat mass, and increased lean mass, resulting in an overall maintenance of body weight.

CRBP-III and BAT.

BAT is characterized by multilocular lipid droplets and a high density of mitochondria in mice fed a chow diet. The central function of BAT is oxidation of fatty acids and dissipation of energy in the form of heat as part of adaptive nonshivering thermogenesis to maintain body temperature in small mammals (7). On the basis of our data, C-III-KO mice likely maintained a better oxidative capacity, as reflected in higher expression of key genes involved in fatty acid oxidation and oxidative phosphorylation in BAT from C-III-KO than wild-type mice fed the HFD. In addition, C-III-KO mice were able to maintain body temperature during a cold challenge, indicating preserved thermogenesis in BAT of C-III-KO compared with wild-type mice. On the basis of this observation, lower body weights of C-III-KO than wild-type mice fed the HFD may be have been expected. In addition to the protection against cold, BAT has been suggested to play an important role in energy metabolism, but a direct role in body weight regulation remains controversial (26). Ablation of BAT or inactivation of all three β-adrenergic receptors rendered mice thermogenically inactive and susceptible to diet-induced obesity (3, 18, 27). In contrast, other mouse models, including inactivation of UCP-1 or failure to induce UCP-1, render chow- or HFD-fed mice cold sensitive but not obese (12, 4648). Several studies indicate that interscapular BAT may not play a direct role in regulating body weight but, rather, that diet-induced thermogenesis may be mediated by brown adipocytes or increased expression of BAT-specific genes in tissues such as skeletal muscle or white adipose tissue (1, 9, 26, 48). In the present mouse model, we may have a more preserved BAT, but it is not contributing to the phenotype, and/or its contribution may be blunted.

CRBP-III and lipolysis.

Significantly lower serum levels of free fatty acids and glycerol in C-III-KO than wild-type mice may have contributed to lower hepatic triglyceride accumulation in C-III-KO than wild-type mice. In addition to fatty acid reesterification to triglyceride in the liver, free fatty acids are used for ketogenesis, leading to increased ketone body levels in the circulation, which are significantly increased in states of insulin resistance and diabetes (15). Serum levels of ketone bodies were significantly lower in C-III-KO than wild-type mice, consistent with the notion that fatty acid flux to the liver is reduced.

Hepatic steatosis during obesity and insulin resistance are consequences of increased rates of lipogenesis in the liver and increased uptake of nonesterified fatty acids released by adipose tissue (6, 17, 24, 44). Excess triglyceride stored as cytosolic lipid droplets in hepatocytes contributes to the development of fatty liver and insulin resistance (6, 40, 41, 45). Since we found no evidence that changes in hepatic lipid synthesis or oxidation contributed to differences in hepatic steatosis, significantly lower serum free fatty acid levels in C-III-KO mice may offer an explanation for the lower hepatic triglyceride accumulation. Serum free fatty acid levels, especially under postabsorptive conditions, are mostly a function of fatty acid release from adipose tissue (13). During insulin resistance, white adipose tissue lipolysis is increased, leading to a rise in the release of free fatty acids and glycerol into the circulation. Consistent with decreased serum fatty acids in C-III-KO mice, in vitro lipolysis assays of adipose tissue from C-III-KO mice indicated diminished efflux of glycerol and free fatty acids from adipose tissue compared with wild-type mice in the basal state.

CRBP-III and PPARγ.

Given our findings that CRBP-III is a direct target of PPARγ activation, it is surprising that CRBP-III deficiency resulted in attenuation of hepatic steatosis and reduction of free fatty release from the adipose tissue. PPARγ agonists, such as thiazolidinediones, have been shown to decrease fatty acid levels in the circulation (32, 39, 51). In addition, adipose-specific deficiency of PPARγ abolished this effect of PPARγ activation on serum free fatty acids (19). The mechanism that leads to decreased free fatty acid efflux from adipose tissue of C-III-KO mice remains to be determined. It is unlikely that CRBP-III regulates PPARγ action, inasmuch as expression of PPARγ target genes was not altered in the absence of CRBP-III.

CRBP-III has been known to play a role in the mammary gland by facilitating retinol esterification during lactation (36). On the basis of this observation, we initially hypothesized that CRBP-III may have a similar role maintaining retinyl ester levels in adipose tissue. Therefore, CRBP-III would have an important role in keeping retinoid in the storage form, i.e., retinyl ester, and would effectively lower the amount of RA available for nuclear receptor activation. However, this is unlikely, given the similar levels of retinol and retinyl ester in adipose tissue from C-III-KO and wild-type mice, indicating that the actions of CRBP-III may be independent of its binding to retinol and may also be independent of retinoid metabolism.

Taken together, CRBP-III deficiency confers a protection for multiple pathological aspects in diet-induced obesity, resulting in reduced fatty liver and serum free fatty acid levels and decreased food intake and adiposity.

GRANTS

This work was supported by National Institute of Diabetes and Digestive and Kidney Diseases Grants RO1 DK-067512 (S. Vogel), DK-063608-06 (S. Vogel), and DK-066618 (G. J. Schwartz) and New York Obesity Research Center Grant 5P30 DK-026687.

Supplementary Material

[Supplemental Figures and Tables]

Notes

The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked “advertisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

REFERENCES

1. Almind K, Manieri M, Sivitz WI, Cinti S, Kahn CR. Ectopic brown adipose tissue in muscle provides a mechanism for differences in risk of metabolic syndrome in mice. Proc Natl Acad Sci USA 104: 2366–2371, 2007. [Europe PMC free article] [Abstract] [Google Scholar]
2. Anghel SI, Bedu E, Vivier CD, Descombes P, Desvergne B, Wahli W. Adipose tissue integrity as a prerequisite for systemic energy balance: a critical role for peroxisome proliferator-activated receptor-γ. J Biol Chem 282: 29946–29957, 2007. [Abstract] [Google Scholar]
3. Bachman ES, Dhillon H, Zhang CY, Cinti S, Bianco AC, Kobilka BK, Lowell BB. β-AR signaling required for diet-induced thermogenesis and obesity resistance. Science 297: 843–845, 2002. [Abstract] [Google Scholar]
4. Banaszak L, Winter N, Xu Z, Bernlohr DL, Cowan S, Jones TA. Lipid-binding proteins: a family of fatty acid and retinoid transport proteins. Adv Protein Chem 45: 89–151, 1994. [Abstract] [Google Scholar]
5. Blaner WS, Olsen JA. Retinol and retinoic acid metabolism. In: The Retinoids: Biology, Chemistry and Medicine, edited by Sporn MB, Roberts AB, Goodman DS. New York: Raven, 1994, p. 229–255.
6. Browning JD, Horton JD. Molecular mediators of hepatic steatosis and liver injury. J Clin Invest 114: 147–152, 2004. [Europe PMC free article] [Abstract] [Google Scholar]
7. Cannon B, Nedergaard J. Brown adipose tissue: function and physiological significance. Physiol Rev 84: 277–359, 2004. [Abstract] [Google Scholar]
8. Caprioli A, Zhu H, Sato T. CRBP-III:lacZ expression pattern reveals a novel heterogeneity of vascular endothelial cells. Genesis 40: 139–145, 2004. [Abstract] [Google Scholar]
9. Cederberg A, Gronning LM, Ahren B, Tasken K, Carlsson P, Enerback S. FOXC2 is a winged helix gene that counteracts obesity, hypertriglyceridemia, and diet-induced insulin resistance. Cell 106: 563–573, 2001. [Abstract] [Google Scholar]
10. Chawla A, Schwarz EJ, Dimaculangan DD, Lazar MA. Peroxisome proliferator-activated receptor (PPAR)-γ: adipose-predominant expression and induction early in adipocyte differentiation. Endocrinology 135: 798–800, 1994. [Abstract] [Google Scholar]
11. Duan SZ, Ivashchenko CY, Whitesall SE, D'Alecy LG, Duquaine DC, Brosius FC 3rd, Gonzalez FJ, Vinson C, Pierre MA, Milstone DS, Mortensen RM. Hypotension, lipodystrophy, and insulin resistance in generalized PPARγ-deficient mice rescued from embryonic lethality. J Clin Invest 117: 812–822, 2007. [Europe PMC free article] [Abstract] [Google Scholar]
12. Enerback S, Jacobsson A, Simpson EM, Guerra C, Yamashita H, Harper ME, Kozak LP. Mice lacking mitochondrial uncoupling protein are cold-sensitive but not obese. Nature 387: 90–94, 1997. [Abstract] [Google Scholar]
13. Frayn KN, Arner P, Yki-Jarvinen H. Fatty acid metabolism in adipose tissue, muscle and liver in health and disease. Essays Biochem 42: 89–103, 2006. [Abstract] [Google Scholar]
14. Frost SC, Lane MD. Evidence for the involvement of vicinal sulfhydryl groups in insulin-activated hexose transport by 3T3-L1 adipocytes. J Biol Chem 260: 2646–2652, 1985. [Abstract] [Google Scholar]
15. Fukao T, Lopaschuk GD, Mitchell GA. Pathways and control of ketone body metabolism: on the fringe of lipid biochemistry. Prostaglandins Leukotr Essent Fatty Acids 70: 243–251, 2004. [Abstract] [Google Scholar]
16. Ghyselnick NB, Bavik C, Sapin V, Mark M, Bonnies D, Hindelang C, Dierich A, Nilsson CB, Hakansson H, Sauvant P, Azais-Braesco V, Frasson M, Picaud S, Chambon P. Cellular retinol-binding protein I is essential for vitamin A homeostasis. EMBO J 18: 4903–4914, 1999. [Europe PMC free article] [Abstract] [Google Scholar]
17. Gibbons GF, Islam K, Pease RJ. Mobilisation of triacylglycerol stores. Biochim Biophys Acta 1483: 37–57, 2000. [Abstract] [Google Scholar]
18. Hamann A, Flier JS, Lowell BB. Decreased brown fat markedly enhances susceptibility to diet-induced obesity, diabetes, and hyperlipidemia. Endocrinology 137: 21–29, 1996. [Abstract] [Google Scholar]
19. He W, Barak Y, Hevener A, Olson P, Liao D, Le J, Nelson M, Ong E, Olefsky JM, Evans RM. Adipose-specific peroxisome proliferator-activated receptor-γ knockout causes insulin resistance in fat and liver but not in muscle. Proc Natl Acad Sci USA 100: 15712–15717, 2003. [Europe PMC free article] [Abstract] [Google Scholar]
20. Heikkinen S, Auwerx J, Argmann CA. PPARγ in human and mouse physiology. Biochim Biophys Acta 1771: 999–1013, 2007. [Europe PMC free article] [Abstract] [Google Scholar]
21. Hertzel AV, Bernlohr DA. The mammalian fatty acid-binding protein multigene family: molecular and genetic insights into function. Trends Endocrinol Metab 11: 175–180, 2000. [Abstract] [Google Scholar]
22. Jpenberg AI, Jeannin E, Wahli W, Desvergne B. Polarity and specific sequence requirements of peroxisome proliferator-activated receptor (PPAR)/retinoid X receptor heterodimer binding to DNA. A functional analysis of the malic enzyme gene PPAR response element. J Biol Chem 272: 20108–20117, 1997. [Abstract] [Google Scholar]
23. Jones JR, Barrick C, Kim KA, Lindner J, Blondeau B, Fujimoto Y, Shiota M, Kesterson RA, Kahn BB, Magnuson MA. Deletion of PPARγ in adipose tissues of mice protects against high fat diet-induced obesity and insulin resistance. Proc Natl Acad Sci USA 102: 6207–6212, 2005. [Europe PMC free article] [Abstract] [Google Scholar]
24. Lewis GF, Carpentier A, Adeli K, Giacca A. Disordered fat storage and mobilization in the pathogenesis of insulin resistance and type 2 diabetes. Endocr Rev 23: 201–229, 2002. [Abstract] [Google Scholar]
25. Liu L, Gudas LJ. Disruption of the lecithin:retinol acyltransferase gene makes mice more susceptible to vitamin A deficiency. J Biol Chem 280: 40226–40234, 2005. [Abstract] [Google Scholar]
26. Lowell BB, Bachman ES. β-Adrenergic receptors, diet-induced thermogenesis, and obesity. J Biol Chem 278: 29385–29388, 2003. [Abstract] [Google Scholar]
27. Lowell BB, S-Susulic V, Hamann A, Lawitts JA, Himms-Hagen J, Boyer BB, Kozak LP, Flier JS. Development of obesity in transgenic mice after genetic ablation of brown adipose tissue. Nature 366: 740–742, 1993. [Abstract] [Google Scholar]
28. Moon HS, Guo DD, Song HH, Kim IY, Jiang HL, Kim YK, Chung CS, Choi YJ, Lee HG, Cho CS. Regulation of adipocyte differentiation by PEGylated all-trans retinoic acid: reduced cytotoxicity and attenuated lipid accumulation. J Nutr Biochem 18: 322–331, 2007. [Abstract] [Google Scholar]
29. Mukherjee R, Davies PJ, Crombie DL, Bischoff ED, Cesario RM, Jow L, Hamann LG, Boehm MF, Mondon CE, Nadzan AM, Paterniti JR Jr, Heyman RA. Sensitization of diabetic and obese mice to insulin by retinoid X receptor agonists. Nature 386: 407–410, 1997. [Abstract] [Google Scholar]
30. Napoli JL Retinoic acid: its biosynthesis and metabolism. Prog Nucleic Acid Res Mol Biol 63: 139–188, 1999. [Abstract] [Google Scholar]
31. Noy N Retinoid-binding proteins: mediators of retinoid action. Biochem J 348: 481–495, 2000. [Europe PMC free article] [Abstract] [Google Scholar]
32. Oakes ND, Thalen PG, Jacinto SM, Ljung B. Thiazolidinediones increase plasma-adipose tissue FFA exchange capacity and enhance insulin-mediated control of systemic FFA availability. Diabetes 50: 1158–1165, 2001. [Abstract] [Google Scholar]
33. O'Byrne SM, Wongsiriroj N, Libien J, Vogel S, Goldberg IJ, Baehr W, Palczewski K, Blaner WS. Retinoid absorption and storage is impaired in mice lacking lecithin:retinol acyltransferase (LRAT). J Biol Chem 280: 35647–35657, 2005. [Europe PMC free article] [Abstract] [Google Scholar]
34. Ong DE, Newcomer ME, Chytil F. Cellular retinoid-binding proteins. In: The Retinoids: Biology, Chemistry, and Medicine, edited by Sporn MB, Roberts AB, Goodman DS. New York: Raven, 1994, p. 283–317.
35. Paglialunga S, Schrauwen P, Roy C, Moonen-Kornips E, Lu H, Hesselink MK, Deshaies Y, Richard D, Cianflone K. Reduced adipose tissue triglyceride synthesis and increased muscle fatty acid oxidation in C5L2 knockout mice. J Endocrinol 194: 293–304, 2007. [Abstract] [Google Scholar]
36. Piantedosi R, Ghyselinck N, Blaner WS, Vogel S. Cellular retinol-binding protein type III is needed for retinoid incorporation into milk. J Biol Chem 280: 24286–24292, 2005. [Abstract] [Google Scholar]
37. Rosen ED, Sarraf P, Troy AE, Bradwin G, Moore K, Milstone DS, Spiegelman BM, Mortensen RM. PPAR-γ is required for the differentiation of adipose tissue in vivo and in vitro. Mol Cell 4: 611–617, 1999. [Abstract] [Google Scholar]
38. Rosen ED, Spiegelman BM. Adipocytes as regulators of energy balance and glucose homeostasis. Nature 444: 847–853, 2006. [Europe PMC free article] [Abstract] [Google Scholar]
39. Saltiel AR, Olefsky JM. Thiazolidinediones in the treatment of insulin resistance and type II diabetes. Diabetes 45: 1661–1669, 1996. [Abstract] [Google Scholar]
40. Samuel VT, Liu ZX, Qu X, Elder BD, Bilz S, Befroy D, Romanelli AJ, Shulman GI. Mechanism of hepatic insulin resistance in non-alcoholic fatty liver disease. J Biol Chem 279: 32345–32353, 2004. [Abstract] [Google Scholar]
41. Savage DB, Choi CS, Samuel VT, Liu ZX, Zhang D, Wang A, Zhang XM, Cline GW, Yu XX, Geisler JG, Bhanot S, Monia BP, Shulman GI. Reversal of diet-induced hepatic steatosis and hepatic insulin resistance by antisense oligonucleotide inhibitors of acetyl-CoA carboxylases 1 and 2. J Clin Invest 116: 817–824, 2006. [Europe PMC free article] [Abstract] [Google Scholar]
42. Schweiger M, Schreiber R, Haemmerle G, Lass A, Fledelius C, Jacobsen P, Tornqvist H, Zechner R, Zimmermann R. Adipose triglyceride lipase and hormone-sensitive lipase are the major enzymes in adipose tissue triacylglycerol catabolism. J Biol Chem 281: 40236–40241, 2006. [Abstract] [Google Scholar]
43. Sethi JK, Vidal-Puig AJ. Adipose tissue function and plasticity orchestrate nutritional adaptation. J Lipid Res 48: 1253–1262, 2007. [Abstract] [Google Scholar]
44. Shimomura I, Bashmakov Y, Horton JD. Increased levels of nuclear SREBP-1c associated with fatty livers in two mouse models of diabetes mellitus. J Biol Chem 274: 30028–30032, 1999. [Abstract] [Google Scholar]
45. Shulman GI Cellular mechanisms of insulin resistance. J Clin Invest 106: 171–176, 2000. [Europe PMC free article] [Abstract] [Google Scholar]
46. Stefl B, Janovska A, Hodny Z, Rossmeisl M, Horakova M, Syrovy I, Bemova J, Bendlova B, Kopecky J. Brown fat is essential for cold-induced thermogenesis but not for obesity resistance in aP2-Ucp mice. Am J Physiol Endocrinol Metab 274: E527–E533, 1998. [Abstract] [Google Scholar]
47. Thomas SA, Palmiter RD. Thermoregulatory and metabolic phenotypes of mice lacking noradrenaline and adrenaline. Nature 387: 94–97, 1997. [Abstract] [Google Scholar]
48. Toh SY, Gong J, Du G, Li JZ, Yang S, Ye J, Yao H, Zhang Y, Xue B, Li Q, Yang H, Wen Z, Li P. Up-regulation of mitochondrial activity and acquirement of brown adipose tissue-like property in the white adipose tissue of fsp27 deficient mice. PLoS ONE 3: e2890, 2008. [Europe PMC free article] [Abstract] [Google Scholar]
49. Tontonoz P, Hu E, Graves RA, Budavari AI, Spiegelman BM. mPPAR-γ2: tissue-specific regulator of an adipocyte enhancer. Genes Dev 8: 1224–1234, 1994. [Abstract] [Google Scholar]
50. Tontonoz P, Hu E, Spiegelman BM. Stimulation of adipogenesis in fibroblasts by PPAR-γ2, a lipid-activated transcription factor. Cell 79: 1147–1156, 1994. [Abstract] [Google Scholar]
51. Tordjman J, Chauvet G, Quette J, Beale EG, Forest C, Antoine B. Thiazolidinediones block fatty acid release by inducing glyceroneogenesis in fat cells. J Biol Chem 278: 18785–18790, 2003. [Abstract] [Google Scholar]
52. Tsutsumi C, Okuno M, Tannous L, Piantedosi R, Allan M, Goodman DS, Blaner WS. Retinoids and retinoid-binding protein expression in rat adipocytes. J Biol Chem 267: 1805–1810, 1992. [Abstract] [Google Scholar]
53. Vogel S, Mendelsohn CL, Mertz J, Piantedosi R, Waldburger C, Gottesman ME, Blaner WS. Characterization of a new member of the fatty acid-binding protein family that binds all-trans-retinol. J Biol Chem 276: 1353–1360, 2001. [Abstract] [Google Scholar]
54. Yang Q, Graham TE, Mody N, Preitner F, Peroni OD, Zabolotny JM, Kotani K, Quadro L, Kahn BB. Serum retinol binding protein 4 contributes to insulin resistance in obesity and type 2 diabetes. Nature 436: 356–362, 2005. [Abstract] [Google Scholar]
55. Ziouzenkova O, Orasanu G, Sharlach M, Akiyama TE, Berger JP, Viereck J, Hamilton JA, Tang G, Dolnikowski GG, Vogel S, Duester G, Plutzky J. Retinaldehyde represses adipogenesis and diet-induced obesity. Nat Med 13: 695–702, 2007. [Europe PMC free article] [Abstract] [Google Scholar]

Articles from American Journal of Physiology - Endocrinology and Metabolism are provided here courtesy of American Physiological Society

Citations & impact 


Impact metrics

Jump to Citations
Jump to Data

Citations of article over time

Smart citations by scite.ai
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by EuropePMC if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
Explore citation contexts and check if this article has been supported or disputed.
https://scite.ai/reports/10.1152/ajpendo.90464.2008

Supporting
Mentioning
Contrasting
0
52
1

Article citations


Go to all (34) article citations

Data 


Data behind the article

This data has been text mined from the article, or deposited into data resources.

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.


Funding 


Funders who supported this work.

NIDDK NIH HHS (4)