Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 May 12.
Published in final edited form as: J Toxicol Sci. 2008 Oct;33(4):405–413. doi: 10.2131/jts.33.405

Regulation of insulin-like growth factor binding protein-1 and lipoprotein lipase by the aryl hydrocarbon receptor

Keiichi Minami 1, Miki Nakajima 1, Yuto Fujiki 1, Miki Katoh 1, Frank J Gonzalez 2, Tsuyoshi Yokoi 1
PMCID: PMC7217312  NIHMSID: NIHMS1584375  PMID: 18827440

Abstract

The aryl hydrocarbon receptor (Ahr), a ligand-activated transcriptional factor, mediates the transcriptional activation of a battery of genes encoding drug metabolism enzymes. In the present study, we investigated the hepatic mRNA expression profile in Ahr-null (Ahr KO) mice compared to wild-type mice by microarray analysis to find new Ahr target genes. Pooled total RNA samples of liver extracted from 7- and 60-week-old Ahr KO or wild-type mice were studied by DNA microarray representing 19,867 genes. It was demonstrated that 23 genes were up-regulated and 20 genes were down-regulated over 2 fold in Ahr KO mice compared with wild-type mice commonly within the different age groups. We focused on insulin-like growth factor binding protein-1 (Igfbp-1) and lipoprotein lipase (Lpl) that were up-regulated in Ahr KO mice. The higher expression in Ahr KO mice compared to wild-type mice were confirmed by real-time RT-PCR analysis. In the wild-type mice but not in the Ahr KO mice, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) treatment increased the Igfbp-1 and Lpl mRNA levels. The expression profile of Igfbp-1 protein was consistent with that of Igfbp-1 mRNA. Since Lpl is the primary enzyme responsible for hydrolysis of lipids in lipoproteins, the serum triglyceride levels were determined. Indeed, the serum triglyceride levels in Ahr KO mice was lower than that in wild-type mice in accordance with the Lpl mRNA levels. Contrary to our expectation, TCDD treatment significantly increased the serum triglyceride levels in wild-type, but did not in Ahr KO mice. These results suggest that serum triglyceride levels are not correlated with hepatic Lpl expression levels. In the present study, we found that Ahr paradoxically regulates Igfbp-1 and Lpl expressions in the liver.

Keywords: Aryl hydrocarbon receptor, Knockout mice, Igfbp-1, Lpl

INTRODUCTION

Aryl hydrocarbon receptor (Ahr) is a ligand-activated transcription factor and a member of the basic helix-loop-helix/Per-Arnt-Sim (bHLH/PAS) family of chemosensors and developmental regulators. Various kinds of environmental stimuli including 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) are well known as ligands of Ahr (Schmidt and Bradfield, 1996; Sogawa and Fujii-Kuriyama, 1997). Upon binding to the ligand, Ahr translocates to the nuclei, coincident with formation of a heterodimeric complex with Ahr nuclear translocator (Arnt). The ligand-activated Ahr mediates the transcriptional activation of a battery of genes encoding enzymes such as cytochrome P450 (CYP) 1 family, NAD(P)H: quinone oxidoreductase and glutathione S-transferase Ya subunit that function in the metabolism of xenobiotics and endobiotics (Bock, 1994; Rowlands and Gustafsson, 1997).

Gene knockout technology is a useful tool to estimate the roles of certain genes in vivo. Ahr-null (Ahr KO) mice on a C57BL/6 strain background were established by three research groups (Fernandez-Salguero et al., 1995; Schmidt et al., 1996; Mimura et al., 1997). The Ahr KO mice established by Fernandez-Salguero et al. (1995) exhibited 40-50% neonatal lethality, although survivors reached maturity and were fertile. The Ahr KO mice established by the latter two groups exhibited no neonatal lethality, but the growth rate was decreased in the first few weeks. In the Ahr KO mice, the size of the liver has been reported to be decreased compared with that in wild-type mice (Fernandez-Salguero et al., 1995; Schmidt et al., 1996). Hepatic portal fibrosis and hepatic vascular hypertrophy were observed in the Ahr KO mice (Fernandez-Salguero et al., 1995; Fernandez-Salguero et al., 1997). In addition, the accumulation of retinoid in the liver owing to reduced retinoic acid metabolism has also been documented (Andreola et al., 1997). The abnormality of retinoid homeostasis was considered to be the reason for the liver fibrosis in the Ahr KO mice (Andreola et al., 2004). Zaher et al. (1998) found that transforming growth factor β is overexpressed in the liver of Ahr KO mice, and this could be a causal factor of liver fibrosis. Thus, these findings suggest that Ahr expression in the liver is important for normal liver development. In the present study, we sought to determine the hepatic mRNA expression profile in Ahr KO mice compared with that in wild-type mice by microarray analysis to identify new targets of Ahr.

MATERIALS AND METHODS

Chemicals

CodeLink Expression Assay Reagent kit, Manual Prep and streptavidin-Cy5 were purchased from GE Healthcare Bio-Sciences (Piscataway, NJ, USA). QIAquick PCR Purification Kit and RNeasy Mini Kit were from Qiagen (Hilden, Germany). NEN Blocking Reagent and Biotin 11-UTP were from Perkin-Elmer Life Sciences (Boston, MA, USA). ReverTra Ace (Moloney Murine Leukemia Vims Reverse Transcriptase RNase H Minus) was from Toyobo (Osaka, Japan). SYBR Premix Ex Taq (Perfect Real Time) was from Takara (Shiga, Japan). Goat anti-mouse insulin-like growth factor binding protein 1 (Igfbp-1) antibody was purchased from Santa Cruz Biotechnology (Santa Cruz, CA, USA). Triglyceride E Test Wako was from Wako Pure Chemical Industries (Osaka, Japan). TCDD was from Cambridge Isotope Laboratories (Cambridge, MA, USA). All primers were commercially synthesized at Hokkaido System Sciences (Sapporo, Japan). Other chemicals were of the highest grade commercially available.

Animals and treatment

Ahr KO mice generated by Fernandez-Salguero et al. (1995) were used. Animals were housed in the institutional animal facility in a controlled environment (temperature 25 ± 1°C, humidity 50 ± 10% and 12 hr light/12 hr dark cycle) with access to food and water ad libitum. Animal maintenance and treatment were conducted in accordance with the National Institutes of Health Guide for Animal Welfare of Japan, as approved by the Institutional Animal Care and Use Committee of Kanazawa University. Genotyping of animals was carried out by polymerase chain reactions (PCRs) described previously (Takemoto et al., 2004). For the DNA microarray experiment, 7- and 60-week-old Ahr KO and wild-type mice were used. For the TCDD treatments, TCDD in corn oil (40 μg/kg body weight per day) was intraperitoneally administered to 35-week-old Ahr KO mice and 14-week-old wild-type mice for four days. Corn oil (2 ml/kg body weight) was administered as a control.

Total RNA preparation

Mice were sacrificed and the livers were collected and immediately frozen in liquid nitrogen and stored at −80°C until use. Total RNA from liver was isolated using ISOGEN (Nippon Gene, Tokyo, Japan) according to the manufacturer’s protocol. Equal amounts of total RNA from 5 - 7 mice were pooled.

Microarray analysis

Microarray analysis was performed using a CodeLink Bioarray Perfect System according to the manufacturer’s protocol (GE Healthcare Bio-Sciences). A Codelink UniSet Mouse 20K I Bioarray (GE Healthcare Bio-Sciences) consisting of 19,867 genes including expression sequence tags (ESTs) was used. Processed slides were scanned with an Agilent G2565BA Microarray Scanner using Agilent Scan Control Software (Agilent Technologies, Palo Alto, CA, USA) with the laser set to red (633 nm) and the photomultiplier tube value to 70%. The scanned images for each slide were analyzed using CodeLink Expression Analysis Software (GE Healthcare Bio-Sciences). The microarray data quality control was as follows: present, no flags (neither marginal nor absent); marginal, low quality spots judged by analysis software; absent, low signal density spots. Microarray data management was performed with GeneSpring software (Agilent Technologies). Comparison of the present genes, expression filtering and experiment normalization were performed. The individual gene expression for each array was normalized to their respective median value. Expression filters included the requirement that the genes be present in over 200% of controls for up-regulated genes and below 50% of controls for down-regulated genes.

Real-time RT-PCR

Total RNA (4 μg) was reverse transcribed using ReverTra Ace according to the manufacturer’s instructions and the resulting cDNA was amplified by PCR. Real-time PCR was performed using the Smart Cycler (Cepheid, Sunnyvale, CA, USA). PCR reactions were carried out as follows: A 1 μl portion of the reverse transcribed mixture was added to a PCR mixture containing 0.4 μM of each primer and SYBR Premix Ex Taq solution in a final volume of 25 μl. The primers used for PCR are shown in Table 1. The PCR condition for Igfbp-1, Lpl, Cyp17a1, and GAPDH was as follows: after an initial denaturation at 95°C for 30 sec, the amplification was performed by denaturation at 94°C for 4 sec, annealing and extension at 64°C for 20 sec for 45 cycles. The amplified products were monitored directly by measuring the increase of the dye intensity of the SYBR Green I. To normalize RNA loading and PCR variations, the signals of targets were corrected with the signals of GAPDH mRNA as the internal standard.

Table 1.

Primers used in the present study.

Primer Sequence
Cyp17a1 S 5’ -GTA TTC AGC ACC TTT TCC CT-3’
Cyp17a1 AS 5’ -AAT ATG TCC ACC AGA TCG CT-3’
IGFBP-1 S 5’ -CAA ACT GCA ACA AGA ATG G-3’
IGFBP-1 AS 5’ -TGT ATC AAG CAG TAT GTG G-3’
LPL S 5’ -AGA AGC AGC AAG ATG TAC CT-3’
LPL AS 5’ - GAA ACT TTC TCC CTA GCA CA-3’
GAPDH S 5’ -AAA TGG GGT GAG GCC GGT-3’
GAPDH AS 5’ -ATT GCT GAC AAT CTT GAG TGA-3’

Western blot analysis of Igfbp-1

Ahr KO and wild-type mice were sacrificed 24 hr after the last treatment with TCDD. The livers were homogenized with buffer (0.1 M Tris-HCl (pH 7.4), 0.1 M KCl, 1 mM EDTA, 1 mM Na3VO4, 1 mM NaF) and liver homogenates (100 μg protein) subjected to SDS-polyacrylamide gel electrophoresis with 10% polyacrylamide gels followed by Western blotting using a PVDF membrane (Immobilon-P, Millipore, Billerica, MA, USA). The membrane was incubated with goat anti-Igfbp-1 antibody at a dilution of 1:200. Biotinylated anti-goat IgG and a VECTASTAIN ABC kit (Vector Laboratories, Burlingame, CA) were used for diaminobenzidine staining. The quantitative analysis of protein expression was performed using ImageQuant TL software (GE Healthcare Bio-Sciences).

Serum triglycerides concentration

Blood samples were collected from the postcaval vein 24 hr after the last treatment with TCDD. The serum triglyceride concentration was measured using Triglyceride E Test Wako.

Statistical analysis

Statistical significance was determined by analysis of variance (ANOVA) followed by Dunnett’s test for multiple comparisons.

RESULTS

Comparison of gene expression profiles in Ahr KO and wild-type mice

Among 19,867 genes, 11,509 (58%) genes were categorized into 15 groups (Table 2). Among these, 7,255 (37%) genes showed sufficient spot density. In 7-week-old Ahr KO mice, the expression levels of 133 genes were elevated, whereas those of 95 genes were suppressed compared with age-matched wild-type mice. In 60-week-old Ahr KO mice, the expression levels of 76 genes were elevated, whereas those of 136 genes were suppressed compared with age-matched wild-type mice. In both 7- and 60-week-old Ahr KO mice, 23 genes were commonly elevated and 20 genes were commonly suppressed. The 43 common genes are shown in Table 3. Cyp1a2 and Ugt1a6, which are known to be highly regulated by Ahr, were down-regulated in Ahr KO mice. In addition, we found that Slc22a7 (organic anion transporter 2, Oat2) and Slc2a2 (facilitated glucose transporter 2) were also down-regulated in Ahr KO mice. These results suggest that these genes might be targets of Ahr regulation.

Table 2.

Number of genes of which expression were significantly changed in Ahr KO mice.

Up-regulated (> 2 fold)
Down-regulated (< 2 fold)
Category Total 1) Present 2) 7 w 60 w 7 & 60 w 7 w 60 w 7 & 60 w
Apoptosis regulator 265 198 5 3 0 0 5 0
Cancer 168 109 2 2 2 1 2 0
Cell cycle 386 252 5 3 1 0 0 0
Chaperone 429 276 4 3 0 4 9 0
Enzyme 3,829 2,615 59 28 12 54 59 11
Immunity 143 93 1 1 0 2 2 1
Microtubular 63 38 0 0 0 0 0 0
Motor 40 24 2 1 1 0 0 0
Nucleic acid binding 1,812 1,183 18 6 0 9 17 0
RNA 14 12 0 0 0 0 0 0
Signal transducer 839 387 6 6 2 7 6 1
Storage 11 5 0 0 0 0 0 0
Stractural protein 1,341 700 17 11 3 7 11 2
Transport 1,465 955 11 9 2 10 20 5
Others 704 408 3 3 1 1 5 0
11,509 7,255 133 76 23 95 136 20

Up-regulated and down-regulated genes showed more than 200% expression and less than 50% expression, respectively, compared with those in wild type mice.

1)

Number of genes of which categories were defined.

2)

Number of genes showing enough spot density.

Table 3.

Up- or down-regulated genes in Ahr KO mice.

Genbank
ID
Common
name
Relative mRNA
expression
Gene name 7 w 60 w
Up regulation (23 genes)
Cancer
    Rab38, member of RAS oncogene family NM_028238 Rab38 2.1 4.3
    Vav2 oncogene NM_009500 Vav2 2.1 2.2
Cell cycle regulator
    Cyclin B2 NM_007630 Ccnb2 3.8 2.1
Enzyme
    Aldo-keto reductase family 1, member B7 NM_009731 Akr1b7 2.0 2.6
    Arylacetamide deacetylase (esterase) NM_023383 Aadac 2.5 2.0
    Asparagine synthetase U38940 Asns 6.5 5.0
    Cis-retinol/3alpha hydroxysterol short-chain dehydrogenase-like BC018263 CRAD-L 3.2 3.1
    Cytochrome c oxidase, subunit VIIa 1 NM_009944 Cox7a1 2.8 2.1
    Cytochrome P450, family 4, subfamily a, polypeptide 14 NM_007822 Cyp4a14 3.6 4.1
    Cytochrome P450, family 17, subfamily a, polypeptide 1 NM_007809 Cyp17a1 12.5 2.1
    Glutaredoxin 2 (thioltransferase) NM_023505 Glrx2 2.3 2.2
    Hydroxysteroid (17-beta) dehydrogenase 9 NM_013786 Hsd17b9 3.0 3.5
    Lipoprotein lipase NM_008509 Lpl 14.0 5.5
    Methylmalonyl-Coenzyme A mutase NM_008650 Mut 23.6 9.8
    RIKEN cDNA 2310016A09 gene BC024580 RIKEN 4.7 2.7
Motor
    Dynein, axonemal, intermediate chain 1 AK004387 Dnaic1 4.1 8.2
Signal transducer
    Insulin-like growth factor binding protein 1 NM_008341 Igfbp1 3.2 4.8
Structural protein
    CD59a antigen NM_007652 Cd59a 8.4 3.7
    Collectin sub-family member 11 AK003121 Colec11 2.2 2.3
    Olfactory receptor 65 (Olfr65) NM_013617 Olfr65 2.3 2.5
Transport
    Fatty acid binding protein 5, epidermal NM_010634 Fabp5 2.3 2.7
    Solute carrier organic anion transporter family, member 1a4 NM_030687 Slco1a4 2.9 2.3
Others
    Ubiquitin-associated protein 1 NM_023305 Ubap1 5.8 3.3
Down regulation (20 genes)
Enzyme
    Betaine-homocysteine methyltransferase NM_016668 Bhmt 0.48 0.49
    Cytochrome P450, family 1, subfamily a, polypeptide 2 NM_009993 Cyp1a2 0.30 0.36
    Dopachrome tautomerase NM_010024 Dct 0.26 0.32
    Glutathione peroxidase 6 NM_145451 Gpx6 0.45 0.37
    Interferon gamma-induced GTPase NM_018738 Igtp 0.28 0.27
    Isovaleryl coenzyme A dehydrogenase NM_019826 Ivd 0.16 0.18
    NADH dehydrogenase (ubiquinone) Fe-S protein 5 NM_134104 Ndufs5 0.02 0.01
    Sulfotransferase family 5A, member 1 NM_020564 Sult5a1 0.30 0.29
    UDP glucuronosyltransferase 1 family, polypeptide A6 U16818 Ugt1a6 0.38 0.48
    Expressed sequence AI586015 NM_019992 AI586015 0.26 0.42
    RIKEN cDNA E430034L04 gene NM_011816 RIKEN 0.43 0.42
Immunity
    Plasminogen NM_008877 Plg 0.49 0.40
Signal transducer
    Transforming growth factor beta 1 induced transcript 4 NM_009366 Tgfb1i4 0.23 0.46
Structural protein
    Growth arrest specific 5 NM_013525 Gas5 0.27 0.46
    Prion protein NM_011170 Prnp 0.48 0.50
Transport
    Amiloride-sensitive cation channel 5, intestinal NM_021370 Accn5 0.12 0.23
    Aquaporin 8 NM_007474 Aqp8 0.46 0.39
    Solute carrier family 2 (facilitated glucose transporter), member 2 NM_031197 Slc2a2 0.47 0.26
    Solute carrier family 22 (organic anion transporter), member 7 NM_144856 Slc22a7 0.35 0.47
    Sorting nexin 1 NM_019727 Snx1 0.42 0.45

Up-regulated and down-regulated genes showed more than 200% and less than 50% expressions, respectively, compared with those in wild-type mice at the same weeks old.

Categories of first occurrence in Table 2 are listed.

The expression levels of methylmalonyl-Coenzyme A mutase, lipoprotein lipase (Lpl), and Cyp17a1 were highly (over 10 fold in 7-week-old mice) up-regulated in Ahr KO mice. Among these, the spot density of Lpl was highest. We additionally found that Igfbp-1 was unexpectedly up-regulated in Ahr KO mice, because previous studies reported that Igfbp-1 mRNA was induced by TCDD via Ahr activation (Adachi et al., 2004, Marchand et al., 2005). In a subsequent study, we investigated in detail the expression of Igfbp-1 and Lpl in the liver.

Real-time RT-PCR analysis

To confirm the results of the DNA microarray analysis, real-time RT-PCR analysis was performed (Fig. 1). The hepatic Igfbp-1 mRNA levels in Ahr KO mice were 7 fold (7-week-old) and 24 fold (60-week-old) higher than those in age-matched wild-type mice. The hepatic Lpl mRNA levels in Ahr KO mice were 8 fold (7-week-old) and 3 fold (60-week-old) higher than those in age-matched wild-type mice. Thus, the differences in the expression levels detected by microarray analysis were reproducible.

Fig. 1.

Fig. 1.

Relative expression levels of (A) Igfbp-1 and (B) Lpl mRNA in the liver of Ahr KO and wild-type mice determined by real-time RT-PCR. Total RNA was extracted from 7- and 60-week-old Ahr KO or wild-type mice. Samples from 5 - 7 mice were pooled within each group. The expression levels of Igfbp-1 and Lpl mRNA were normalized with the expression level of GAPDH as a control. Data are expressed as the mean of duplicate experiments.

Effects of TCDD treatment on Igfbp-1 and Lpl mRNA expression in Ahr KO and wild-type mouse livers

We investigated the effect of TCDD treatment on Igfbp-1 and Lpl mRNA expressions. Real-time RT-PCR analyses revealed that Igfbp-1 mRNA was significantly (7 fold) increased by TCDD in wild-type mice, but not in Ahr KO mice, showing 8-fold higher Igfbp-1 mRNA levels than those in wild-type mice (Fig. 2A). Lpl mRNA was also significantly (4 fold) increased by TCDD in wild-type mice, but not in Ahr KO mice, showing 4-fold higher Lpl mRNA levels than those in wild-type mice (Fig. 2B).

Fig. 2.

Fig. 2.

Effects of TCDD treatment on the expression levels of (A) Igfbp-1 and (B) Lpl mRNA in the liver of Ahr KO and wild-type mice determined by real-time RT-PCR analysis. TCDD (40 μg/kg weight) or corn oil was intraperitoneally administered to Ahr KO (35-week-old) and wild-type (14-week-old) mice for 4 days. The expression levels of Igfbp-1 and Lpl mRNA were normalized with the expression level of GAPDH as a control. Data are expressed as mean ± S.E. from 5 or 6 mice. **P < 0.01 by ANOVA.

Igfbp-1 protein expression

Western blot analysis demonstrated that Igfbp-1 protein was significantly (5 fold) induced by TCDD in wild-type mice (Fig. 3), but not in Ahr KO mice, showing 6-fold higher Igfbp-1 protein levels.

Fig. 3.

Fig. 3.

Effects of TCDD treatment on the expression level of Igfbp-1 protein level in the liver of Ahr KO and wild-type mice. TCDD (40 μg/kg weight) or corn oil was intraperitoneally administered to Ahr KO (35-week-old) and wild-type (14-week-old) mice for 4 days. Data are expressed as mean ± S.E. from 5 or 6 mice. *P < 0.05 by ANOVA.

Triglycerides concentration measurement

Since an antibody against Lpl is not commercially available, we sought to determine the Lpl activity to evaluate changes in the hepatic Lpl expression level. Lpl is the primary enzyme responsible for the metabolism of triglycerides. We investigated whether the differences in the Lpl expression level in liver might be inversely correlated with the serum triglyceride levels. The serum triglyceride level was lower in Ahr KO mice than in wild-type mice, being inversely correlated with the Lpl expression level. However, the serum triglyceride level was significantly (1.3 fold) increased by TCDD treatment in wild-type mice, but not in Ahr KO mice (Fig. 4).

Fig. 4.

Fig. 4.

Effects of TCDD treatment on serum triglyceride level in Ahr KO and wild-type mice. TCDD (40 μg/kg weight) or corn oil was administered to Ahr KO (35-week-old) and wild-type (14-week-old) mice for 4 days. Data are expressed as mean ± S.E. from 5 or 6 mice. *P < 0.05, ***P < 0.001 by ANOVA.

DISCUSSION

DNA microarray technology has been extensively used as a powerful tool for predicting unknown signaling pathways. Using DNA microarray analysis, the changes in mRNA expression levels in smooth muscle cells in Ahr KO mice were investigated by Guo et al. (2004) who found that transforming growth factor-beta 3 (Tgfb3) expression was higher in Ahr KO mice than in wild-type mice, indicating that Ahr suppresses Tgfb3 gene expression. It is well known that Ahr regulates various drug-metabolizing enzymes in liver. In the present study, we sought to investigate the mRNA profiles in liver from Ahr KO mice using microarray to find new Ahr gene targets. The overall gene expression profiles vary during development and the aging process. Therefore, we compared the data in young (7-week-old) and older (60-week-old) mice to determine common changes in gene expression by Ahr KO. The decreases in Cyp1a2 and Ugt1a6 in Ahr KO mice were consistent with those previously reported (Fernandez-Salguero et al., 1995). In addition, the decrease of Slc22a7 and increase of Cyp17a1 in livers in Ahr KO mice were consistent with a recent report by Tijet et al. (2006). These results suggest that our study was sufficiently reliable. In the present study, we first found that the Igfbp-1 and Lpl expression levels were higher in Ahr KO mice.

Despite the fact that the expression levels of Igfbp-1 and Lpl in the liver were increased in the Ahr KO, they were induced by TCDD in an Ahr-dependent manner. The latter results suggested that Ahr positively regulates Igfbp-1 and Lpl in the presence of the ligands. This is supported by a previous report indicating that a xenobiotic responsive element (XRE) to which Ahr binds is located in the promoter region of the Igfbp-1 gene at −87 (Marchand et al., 2005). In addition, we found two XRE sequences in the promoter region of the Lpl gene at −332 and −443 by a computer-assisted homology search. Further study will be necessary to determine whether the binding of Ahr to XRE might be responsible for the induction of Lpl by TCDD. Based on the higher expression levels of Igfbp-1 and Lpl in Ahr KO mice compared to those in the wild-type mice, Ahr might suppress Igfbp-1 and Lpl expressions in the absence of exogenous ligands and TCDD may interfere with the suppression. Alternatively, Ahr might positively regulate some suppressor of Igfbp-1 and Lpl expression in the absence of exogenous ligands.

IGFBP-1, one of the six IGFBPs, capable of sequestering insulin growth factors (IGF)s from their receptor. It was reported that transgenic mice of human IGFBP-1 gene showed postnatal growth retardation and impaired fecundity (Schneider et al., 2000). The pathophysiological abnormalities in Ahr KO mice such as decreased body weight, impaired fecundity as well as decrease liver size (Fernandez-Salguero et al., 1995) might be, in part, associated with Igfbp-1 overexpression.

LPL is produced by adipose tissue and then is transported to the endothelial cell surface (Matsumura, 1995). It is also expressed in heart, lung, adipose tissue, kidney, intestine and liver in mice (Kirchgessner et al., 1987). Lpl hydrolyses triglycerides. In Ahr KO mice, Lpl expression in adipocytes might also be increased in addition to that in liver, because the serum triglyceride levels in Ahr KO mice were decreased. The serum triglyceride level was increased by TCDD treatment in wild-type mice, being consistent with a previous report showing that adipose Lpl activity was decreased by TCDD treatment (Matsumura, 1995). TCDD inhibits the differentiation of preadipocytes to adipocytes (Alexander et al., 1998). The differentiation increases peroxisome proliferator activated receptor (PPAR) γ expression, which is a major regulator of Lpl in adipocytes. Therefore, the increase of serum trigrycerides by TCDD in wild-type mice was, in part, due to the inhibition of adipogenesis.

In summary, we found that hepatic Igfbp-1 and Lpl were paradoxically up-regulated by the activation and knockout of Ahr. Ahr would be responsible for glucose homeostasis and lipid metabolism.

ACKNOWLEDGEMENT

We thank Mr. Brent Bell for reviewing the manuscript.

REFERENCES

  1. Adachi J, Mori Y, Matsui S and Matsuda T (2004): Comparison of gene expression patterns between 2,3,7,8-tetrachlorodibenzo-p-dioxin and a natural arylhydrocarbon receptor ligand, indirubin. Toxicol. Sci, 80, 161–169. [DOI] [PubMed] [Google Scholar]
  2. Alexander DL, Ganem LG, Fernandez-Salguero P, Gonzalez F and Jefcoate CR (1998): Aryl-hydrocarbon receptor is an inhibitory regulator of lipid synthesis and of commitment to adipogenesis. J. Cell Sci, 111, 3311–3322. [DOI] [PubMed] [Google Scholar]
  3. Andreola F, Calvisi DF, Elizondo G, Jakowlew SB, Mariano J, Gonzalez FJ and De Luca LM (2004): Reversal of liver fibrosis in aryl hydrocarbon receptor null mice by dietary vitamin A depletion. Hepatology, 39, 157–166. [DOI] [PubMed] [Google Scholar]
  4. Andreola F, Fernandez-Salguero PM, Chiantore MV, Petkovich MP, Gonzalez FJ and De Luca LM (1997): Aryl hydrocarbon receptor knockout mice (AHR−/−) exhibit liver retinoid accumulation and reduced retinoic acid metabolism. Cancer Res., 57, 2835–2838. [PubMed] [Google Scholar]
  5. Bock KW (1994): Aryl hydrocarbon or dioxin receptor: biologic and toxic responses. Rev. Physiol. Biochem. Pharmacol, 125, 1–42. [DOI] [PubMed] [Google Scholar]
  6. Fernandez-Salguero P, Pineau T, Hilbert DM, McPhail T, Lee SS, Kimura S, Nebert DW, Rudikoff S, Ward JM and Gonzalez FJ (1995): Immune system impairment and hepatic fibrosis in mice lacking the dioxin-binding Ah receptor. Science, 268, 722–726. [DOI] [PubMed] [Google Scholar]
  7. Fernandez-Salguero PM, Ward JM, Sundberg JP and Gonzalez FJ (1997): Lesions of aryl-hydrocarbon receptor-deficient mice. Vet. Pathol, 34, 605–614. [DOI] [PubMed] [Google Scholar]
  8. Guo J, Sartor M, Karyala S, Medvedovic M, Kann S, Puga A, Ryan P and Tomlinson CR (2004): Expression of genes in the TGF-β signaling pathway is significantly deregulated in smooth muscle cells from aorta of aryl hydrocarbon receptor knockout mice. Toxicol. Appl. Pharmacol, 194, 79–89. [DOI] [PubMed] [Google Scholar]
  9. Kirchgessner TG, Svenson KL, Lusis AJ and Schotz MC (1987): The sequence of cDNA encoding lipoprotein lipase. A member of a lipase gene family. J. Biol. Chem, 262, 8463–8466. [PubMed] [Google Scholar]
  10. Marchand A, Tomkiewicz C, Marchandeau J-P, Boitier E, Barouki R and Garlatti M (2005): 2,3,7,8-Tetrachlorodibenzo-p-dioxin induces insulin-like growth factor binding protein-1 gene expression and counteracts the negative effect of insulin. Mol. Pharmacol, 67, 444–452. [DOI] [PubMed] [Google Scholar]
  11. Matsumura F (1995): Mechanism of action of dioxin-type chemicals, pesticides, and other xenobiotics affecting nutritional indexes. Am. J. Clin. Nutr, 61, 695–701. [DOI] [PubMed] [Google Scholar]
  12. Mimura J, Yamashita K, Nakamura K, Morita M, Takagi TN, Nakao K, Ema M, Sogawa K, Yasuda M, Katsuki M and Fujii-Kuriyama Y (1997): Loss of teratogenic response to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) in mice lacking the Ah (dioxin) receptor. Genes Cells, 2, 645–654. [DOI] [PubMed] [Google Scholar]
  13. Rowlands JC and Gustafsson JÅ (1997): Aryl hydrocarbon receptor-mediated signal transduction. Crit. Rev. Toxicol, 27, 109–134. [DOI] [PubMed] [Google Scholar]
  14. Schmidt JV and Bradfield CA (1996): Ah receptor signaling pathways. Annu. Rev. Cell Dev. Biol, 12, 55–89. [DOI] [PubMed] [Google Scholar]
  15. Schmidt JV, Su GH, Reddy JK, Simon MC and Bradfield CA (1996): Characterization of a murine Ahr null allele: involvement of the Ah receptor in hepatic growth and development. PNAS, 93, 6731–6736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Schneider MR, Lahm H, Wu M, Hoeflich A and Wolf E (2000): Transgenic mouse models for studying the functions of insulin-like growth factor-binding proteins. FASEB J., 14, 629–640. [DOI] [PubMed] [Google Scholar]
  17. Sogawa K and Fujii-Kuriyama Y (1997): Ah receptor, a novel ligand-activated transcription factor. J. Biochem, 122, 1075–1079. [DOI] [PubMed] [Google Scholar]
  18. Takemoto K, Nakajima M, Fujiki Y, Katoh M, Gonzalez FJ and Yokoi T (2004): Role of the aryl hydrocarbon receptor and Cyp1b1 in the antiestrogenic activity of 2,3,7,8-tetrachlorodibenzo-p-dioxin. Arch. Toxicol, 78, 309–315. [DOI] [PubMed] [Google Scholar]
  19. Tijet N, Boutros PC., Moffat ID, Okey AB, Tuomisto J and Pohjanvirta R (2006): Aryl hydrocarbon receptor regulates distinct dioxin-dependent and dioxin-independent gene batteries. Mol. Pharmacol, 69, 140–153. [DOI] [PubMed] [Google Scholar]
  20. Zaher H, Fernandez-Salguero PM, Letterio J, Sheikh MS, Fornace AJ Jr, Roberts AB and Gonzalez FJ (1998): The involvement of aryl hydrocarbon receptor in the activation of transforming growth factor-β and apoptosis. Mol. Pharmacol, 54, 313–321. [DOI] [PubMed] [Google Scholar]

RESOURCES