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 


Vitamin K is a fat-soluble vitamin that serves as a coenzyme for vitamin K-dependent carboxylase. Besides its canonical action, vitamin K binds to the steroid and xenobiotic receptor (SXR)/pregnane X receptor (PXR) and modulates gene transcription. To determine if the osteoprotective action of vitamin K is the result of the PXR/SXR pathway, we screened by two-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis the PXR/SXR target genes in an osteoblastic cell line (MC3T3-E1) treated with a vitamin K2 (menaquinone 4 [MK4]). Osteoblastic differentiation of MC3T3-E1 cells was induced by MK4. Msx2, an osteoblastogenic transcription factor, was identified as an MK4-induced gene. Functional analysis of the Msx2 gene promoter mapped a vitamin K-responsive element (PXR-responsive element [PXRE]) that was directly bound by a PXR/retinoid X receptor alpha heterodimer. In a chromatin immunoprecipitation analysis, PXR was recruited together with a coactivator, p300, to the PXRE in the Msx2 promoter. MK4-bound PXR cooperated with estrogen-bound estrogen receptor alpha to control transcription at the Msx2 promoter. Knockdown of either PXR or Msx2 attenuated the effect of MK4 on osteoblastic differentiation. Thus, the present study suggests that Msx2 is a target gene for PXR activated by vitamin K and suggests that the osteoprotective action of MK4 in the human mediates, at least in part, a genomic pathway of vitamin K signaling.

Free full text 


Logo of molcellbLink to Publisher's site
Mol Cell Biol. 2007 Nov; 27(22): 7947–7954.
Published online 2007 Sep 17. https://doi.org/10.1128/MCB.00813-07
PMCID: PMC2169156
PMID: 17875939

This article has been retracted. Retraction in: Mol Cell Biol. 2014 March; 34(5): 918    See also: PMC Retraction Policy

This article has been retracted. Retraction in: Mol Cell Biol. 2013 June; 33(12): 2508    See also: PMC Retraction Policy

Vitamin K Induces Osteoblast Differentiation through Pregnane X Receptor-Mediated Transcriptional Control of the Msx2 Gene[down-pointing small open triangle]

Abstract

Vitamin K is a fat-soluble vitamin that serves as a coenzyme for vitamin K-dependent carboxylase. Besides its canonical action, vitamin K binds to the steroid and xenobiotic receptor (SXR)/pregnane X receptor (PXR) and modulates gene transcription. To determine if the osteoprotective action of vitamin K is the result of the PXR/SXR pathway, we screened by two-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis the PXR/SXR target genes in an osteoblastic cell line (MC3T3-E1) treated with a vitamin K2 (menaquinone 4 [MK4]). Osteoblastic differentiation of MC3T3-E1 cells was induced by MK4. Msx2, an osteoblastogenic transcription factor, was identified as an MK4-induced gene. Functional analysis of the Msx2 gene promoter mapped a vitamin K-responsive element (PXR-responsive element [PXRE]) that was directly bound by a PXR/retinoid X receptor α heterodimer. In a chromatin immunoprecipitation analysis, PXR was recruited together with a coactivator, p300, to the PXRE in the Msx2 promoter. MK4-bound PXR cooperated with estrogen-bound estrogen receptor α to control transcription at the Msx2 promoter. Knockdown of either PXR or Msx2 attenuated the effect of MK4 on osteoblastic differentiation. Thus, the present study suggests that Msx2 is a target gene for PXR activated by vitamin K and suggests that the osteoprotective action of MK4 in the human mediates, at least in part, a genomic pathway of vitamin K signaling.

The K vitamins are a group of fat-soluble vitamins that occur in two natural forms: phyloquinones (K1) and menaquinones (K2). Vitamin K (VK) has classically been associated with blood coagulation (31). In its canonical role, VK serves as a coenzyme for VK-dependent carboxylase. This enzyme converts glutamate residues into γ-carboxyglutamate (Gla) residues in VK-dependent proteins, such as prothrombin, and factors IX and X (6, 10, 29). Such VK-induced protein modification also occurs in osteocalcin (7, 21) and matrix Gla protein (MGP) (22). Thus, VK may exert beneficial effects on bone formation and remodeling. In fact, animal studies suggest that VK deficiency results in a reduction in bone mass together with hypocarboxylation of osteocalcin (25).

Clinically, the most common form of K2, menaquinone 4 (MK4), has been shown to prevent bone fractures (3). This osteoprotective effect is more pronounced in K2 than in K1, and hence MK4 has been used to treat osteoporotic patients in Japan (9, 11, 28). However, the bone phenotypic abnormalities of mice deficient in osteocalcin and MGP do not fully support the classical view that the osteoprotective action of VK is the result of the modification of skeletal proteins. These mice, which are genetically deficient for osteocalcin or MGP, exhibited bone mass increases instead of losses (4). This suggests that the osteoprotective action of VK is mediated by another pathway.

MK4 recently has been shown to act as a ligand for the steroid and xenobiotic receptor (SXR) in human osteoblastic cells (33). It transcriptionally regulates gene expression and represents a new pathway of VK action. The SXR and its mouse homolog, the pregnane X receptor (PXR), respond to xenobiotics and pregnenones. PXR and SXR are members of the nuclear receptor (NR) gene superfamily and bind to specific DNA elements (PXR-responsive elements [PXRE]) as heterodimers with one of the retinoid X receptor (RXR) subtypes (α, β, and γ) (1, 2, 15, 16, 18). Like the other NR members, ligand binding to PXR/SXR induces dissociation of corepressors and recruitment of coactivators for ligand-induced transactivation in the target gene promoters (17). Thus, these findings suggest that it is feasible that the osteoprotective VK action mediates its transcriptional control of the VK target genes via PXR/SXR. In fact, several PXR/SXR genes recently have been shown to transcriptionally respond to VK (8).

To test this idea, we screened VK target genes in an osteoblastic cell line (MC3T3-E1) treated with MK4 with two-dimensional sodium dodecyl sulfate-polyacrylamide gel electrophoresis (2D SDS-PAGE). A prime osteoblastogenic factor, Msx2, was identified, and a PXRE was located in its gene promoter. MK4 interacted with the PXRE via PXR/RXRα binding in vivo and in vitro. Osteoblast genesis in MC3T3-E1 cells was induced by MK4, but knockdown of Msx2 by RNA interference (RNAi) abrogated the MK4 effect. The present study suggests that Msx2 is a target gene for VK-activated PXR/SXR. It implies that the osteoprotective VK action takes place, at least in part, on a genomic level by stimulating osteoblast differentiation through Msx2 gene induction.

MATERIALS AND METHODS

Plasmids.

The full-length cDNA for the mouse PXR (15) was subcloned into a pcDNA3 expression vector (Invitrogen) tagged with the hemagglutinin epitope at the N terminus. The mouse Msx2 promoters, the sequences of which were derived from the Ensembl genome browser (http://www.ensembl.org/index.html), were subcloned into a pGL3-Basic vector (Promega).

Animals.

Estrogen receptor α knockout (ERαKO) mice were kindly provided by P. Chambon (19). Genotyping for ERαKO mice was routinely performed on DNA isolated from tail snips by a PCR procedure (19).

Cell culture, transient transfection, and luciferase assay.

MC3T3-E1 and ST2 cells were cultured with alpha minimum essential medium (α-MEM) (GIBCO) containing 10% fetal bovine serum (FBS) (GIBCO) at 37°C and 5% CO2 (31). HEK 293T cells were cultured with Dulbecco's modified Eagle's medium (DMEM) containing 10% FBS (GIBCO) at 37°C and 5% CO2. For transfection, cells were plated in the corresponding medium supplemented with 10% charcoal-stripped FBS in 12-well plates 1 day before transfection. Transfection was performed with Lipofectamine (Invitrogen) with Plus reagent (Invitrogen) or Polyfect (QIAGEN) as directed by the manufacturer's protocol. After 3 h, 17β-estradiol (E2) (10 nM) and MK4 (10 μM) were added to α-MEM containing 2% FBS, and the cells were incubated continuously at 37°C for 24 h. As a reference to normalize transfection efficiency, 2.5 ng/well of pRL-CMV plasmid (Promega) was cotransfected in all experiments. Luciferase activity was determined using the luciferase assay system (Promega) (5).

Osteoblast primary culture.

Calvaria of newborn mice were digested for 80 min at 37°C in phosphate-buffered saline (PBS) containing 0.1% collagenase A (Roche). Cells were plated with α-MEM containing 10% FBS. Media were changed every 3 days (32).

ALP staining.

For alkaline phosphatase (ALP) staining, primary osteoblasts, MC3T3-E1 and ST2 cells, were washed twice with PBS, fixed in 3.7% formaldehyde, and stained with a mixture of 0.1 mg/ml naphthol AS-MX phosphate (Sigma), 0.6 mg/ml fast-blue BB salt (Sigma), 2 mM MgCl2, 5 μl/ml N,N-dimethylforamide (Wako), and 100 mM Tris-HCl (pH 8.8) buffer at 37°C for 5 to 10 min. When the cells turned blue, the cells were washed twice with PBS (32).

ALP assay.

The ALP assay was performed using the LabAssay ALP kit (Wako) according to the manufacturer's instructions. In brief, primary osteoblasts, MC3T3-E1 and ST2 cells, were washed twice with ice-cold PBS and then were solubilized with lysis buffer (20 mM Tris-HCl [pH 7.9], 1% NP-40, 1 mM EDTA, 150 mM NaCl, 2.5 mM MgCl2, 5% glycerol) containing protease inhibitors, followed by determination of the ALP activity in lysates. The protein concentration was determined with a Bio-Rad protein assay kit.

qRT-PCR.

For quantitative real-time reverse transcription-PCR (qRT-PCR), 1 μg of total RNA from each sample was reverse transcribed into first-strand cDNA with random hexamers using Superscript III reverse transcriptase (Invitrogen). Primer sets for all genes were purchased from Takara Bio Inc. (Tokyo, Japan). Real-time RT-PCR was performed using SYBR premix EX Taq (Takara) with the thermal cycler Dice RealTime System TP800 (Takara) according to the manufacturer's instructions. Experimental samples were matched to a standard curve generated by amplifying serially diluted products using the same PCR protocol. To correct for variability in RNA recovery and the efficiency of reverse transcription, glyceraldehyde-3-phosphate dehydrogenase cDNA was amplified and quantified in each cDNA preparation. Normalization and calculation steps were performed as reported previously (34).

2D SDS-PAGE analysis.

For 2D SDS-PAGE analysis, MC3T3-E1 cells were washed with ice-cold PBS, collected by centrifugation at 2,000 × g, resuspended in 50 μl lysis buffer (20 mM Tris-HCl [pH 7.9], 1% NP-40, 1 mM EDTA, 150 mM NaCl, 2.5 mM MgCl2, 5% glycerol) containing protease inhibitors, incubated on ice for 30 min, and then centrifuged for 30 min at 12,000 × g. After centrifugation, the supernatants were cleaned with a 2D-Clean-Up kit (Amersham). These samples were separated with the 2D SDS-PAGE system (Bio-Rad). The 2D SDS-PAGE gels were visualized with the SilverQuest silver staining kit (Invitrogen).

ABCD assay.

For the avidin-biotin complex DNA (ABCD) assay, sense and antisense oligonucleotide DNAs that were biotinylated at the 3′ terminus were incubated at 100°C for DNA annealing and then were cooled slowly at room temperature. To prepare the beads-DNA complex, biotin-conjugated double-stranded oligonucleotide DNA was mixed with a 50% slurry of avidin beads (Tetralink tetrameric avidin resin; Promega). Cells were lysed with lysis buffer (10 mM Tris-Cl [pH 7.8], 1 mM EDTA, 150 mM NaCl, 0.1% NP-40) containing protease inhibitors for 1 h at 4°C. Whole-cell lysates were clarified by centrifugation, mixed with the 50% slurry of avidin beads, and pelleted at 3,000 × g. Supernatants were added to the beads-DNA complex and rotated for 1 h at 4°C to mix. The beads were collected by centrifugation at 3,000 × g and were washed successively in the lysis buffer. Proteins were resolved with SDS-PAGE, and Western blotting was performed with the corresponding antibody (12).

ChIP analysis.

Chromatin immunoprecipitation (ChIP) analysis was performed using the ChIP assay kit (Upstate Biotechnology Inc.) (13, 38) according to the manufacturer's instructions. MC3T3-E1 cells were cultured in the presence or absence of MK4 and E2. Soluble chromatin prepared from 1 × 106 cells was immunoprecipitated with antibodies against the indicated proteins.

Generation of adenovirus.

Recombinant adenoviruses carrying RNAi of Msx2 and PXR were constructed using the RNAi-Ready pSIREN-Shuttle vector (BD Biosciences Clontech) first and then were moved to the Adeno-X vector (Clontech) by being spliced into the I-CeuI and PI-SceI site. The parental virus genomes in HEK 293 cells (ATCC) were constructed according to the manufacturer's protocol (20). MC3T3-E1 cells then were infected by incubation with the recombinant adenovirus.

RESULTS

Vitamin K2 promotes osteoblast differentiation.

We explored the osteoprotective effects of K2 on a molecular level to better understand how K2 clinically prevents osteoporosis. We first tested the effects of MK4, a K2. The effect of MK4 was measured by ALP assay of primary cultured mice osteoblastic cells, derived from calvaria, to detect osteoblastic differentiation. As shown in the lower panels of Fig. Fig.11 A and B (lanes 1 to 4), ALP activity was induced in the primary cultured osteoblastic cells treated with MK4 for 24 h. Likewise, E2 was stimulatory, and the actions of MK4 and E2 were additive. To clarify the stages of differentiation at which MK4 and E2 act, we tested ST2 cells and MC3T3-E1 cells. ST2 cells are bipotential cells derived from mouse bone marrow mesenchymal stem cells that differentiate into either osteoblasts or adipocytes under the proper conditions (32). MC3T3-E1 cells are derived from mouse calvaria and are preosteoblastic cells that can be induced to differentiate into mature osteoblasts (30). As shown in the middle panels of Fig. 1A and B (lanes 5 to 8), ALP induction following treatment with MK4 and/or E2 for 24 h was observed in MC3T3-E1 cells but not in ST2 cells, as shown in the bottom panels of Fig. 1A and B (lanes 9 to 12). The additive action of MK4 and E2 in MC3T3-E1 cells was more pronounced following a longer culture period (Fig. 1C and D). Our results suggest that MK4 and E2 act from the stage at which preosteoblasts differentiate into mature osteoblasts but not at the earlier stage of the cell fate decision. To confirm this, we measured the expression of osteoblast genesis marker genes in the MC3T3-E1 cells treated with MK4 and E2 by qRT-PCR. Early osteoblast genesis marker genes (encoding type I collagen α and ALP) were induced by both MK4 and E2 treatments, while mRNA levels of late osteoblast genesis marker genes (encoding osteopontin and osteocalcin) were unaltered following 24 h of treatment (Fig. (Fig.1E1E).

An external file that holds a picture, illustration, etc.
Object name is zmb0220771260001.jpg

VK and estrogen cooperatively promote mature osteoblast formation. (A) The effect of MK4 and E2 on osteoblastic differentiation in the presence or absence of MK4 (1 μM) and E2 (1 nM). After 24 h, osteoblast differentiation was detected by ALP staining. Upper panel, primary osteoblasts from mouse calvaria; middle panel, MC3T3-E1 cells; bottom panel, ST2 cells. (B) The effect of MK4 and E2 on osteoblastic differentiation in the presence or absence of MK4 (10 μM) and E2 (10 nM). After 24 h, ALP activity was measured using the optical density at 405 nm. All values are means ± standard deviations for at least three independent experiments. (C) The effect of MK4 and E2 in the early stage of osteoblastic differentiation in MC3T3-E1 cells. After 24-, 48-, and 72-h treatments, ALP staining was performed. (D) The effect of MK4 and E2 in the early stage of osteoblastic differentiation in MC3T3-E1 cells. After 24-, 48-, and 72-h treatments, ALP activity was measured. (E) Relative expression levels of the marker genes of osteoblastic differentiation. The total RNA for the qRT-PCR was extracted using the ISOGEN kit (Nippon Gene) from MC3T3-E1 cells treated with MK4 and E2 or left untreated. OPN, osteopontin; OC, osteocalcin.

Msx2 is a target gene for MK4 and E2 in MC3T3-E1 cells.

To elucidate the molecular pathway through which MK4 and E2 induce osteoblast genesis, we screened for MK4 target proteins in untreated and treated MC3T3-E1 cells with 2D SDS-PAGE (Fig. 2A and B). E2 was used for cotreatment, since it potentiated MK4 action in osteoblastic differentiation. As shown in Fig. Fig.2B,2B, expression levels of several candidate proteins were up-regulated or down-regulated by the treatment, and we tried to identify them by matrix-assisted laser desorption ionization-time-of-flight mass spectroscopy analysis. One of these was Msx2, a protein that was shown to be a critical factor for inducing mesenchymal stem cells to differentiate into preosteoblasts (26). We reasoned that Msx2 mediates MK4 action as an MK4 target gene. Indeed, induction of Msx2 gene expression by MK4 alone was visible, and the additive induction with E2 was at both the transcriptional level (qRT-PCR) and the translational level (Western blotting) (Fig. 2C and D).

An external file that holds a picture, illustration, etc.
Object name is zmb0220771260002.jpg

Identification of Msx2 as a regulatory target of VK and estrogen in MC3T3-E1 cells. (A) Strategy for isolating MK4 targets. MC3T3-E1 cells were incubated for 24 h in the presence or absence of MK4 (1 μM) and E2 (1 nM). Total cell lysates then were separated by 2D SDS-PAGE, and the bands were examined with matrix-assisted laser desorption ionization-time-of-flight mass spectroscopy (labeled TOF-MS) for protein identification. (B) Comparison of protein expression by 2D SDS-PAGE in the presence or absence of MK4 and E2. M.W., molecular size. (C) The effect of MK4 and E2 on Msx2 gene expression. Msx2 mRNA expression was analyzed by qRT-PCR. (D) The effect of MK4 and E2 on Msx2 protein expression. Msx2 protein levels were determined by immunoblotting. β-Actin was used as a control.

Msx2 promoter confers responsiveness to MK4 and E2 through cognate NRs.

To test the idea that MK4 acts as a PXR ligand in the transcriptional control of the mouse Msx2 gene, we performed a promoter analysis of the Msx2 gene by using a luciferase reporter assay on MC3T3-E1 cells. The responsiveness to MK4 and E2 was tested with a luciferase gene reporter driven by the mouse Msx2 gene promoter region (−1 to −3000) (designated Msx2-3000). Either MK4 or E2 was sufficient to activate transcription alone, and synergy of MK4 with E2 was observed (Fig. (Fig.3A).3A). Knockdown of PXR and ERα by RNAi (Fig. 3B and C) abolished the MK4- and E2-induced transactivation of the Msx2 gene promoter (Fig. (Fig.3A,3A, lanes 5 to 16). Other known PXR ligands also were tested with this promoter. Though MK4 was effective in stimulating the transcription of the Msx2 gene promoter, none of the other tested ligands (pregnenolone 16α-carbonitrile, 17α-hydroxypregnenolone, mifepristone, and dexamethazone 21-acetate) stimulated transcription alone. Additionally, none demonstrated synergy with E2 (Fig. (Fig.3D).3D). We used a series of promoter deletion mutants to map an MK4-responsive element in the proximal promoter (−1 to −390) (data not shown) that contained a consensus PXR/RXR binding-related element (designated Msx2-PXRE) at around bp −230 (Fig. (Fig.3E).3E). Additionally, a consensus element (designated Msx2-ERE) related to the ER-responsive element (ERE) was found in the distal promoter region (−2672 to −2658) (see Fig. Fig.3E3E and and4A).4A). Short elements containing either Msx2-PXRE or Msx2-ERE conferred the expected response to the cognate NR ligand (data not shown). To verify the impact of the response elements, we introduced two point mutations into each of the elements (Fig. (Fig.3E).3E). As shown in Fig. Fig.3F,3F, Msx2-M1, which was mutated at Msx2-ERE, lost the E2 response; likewise, the MK4 response was abolished by the mutation (Msx2-M2) in Msx2-PXRE. As expected, no NR ligand response was seen in the mutant (Msx2-M3) that was mutated at both Msx2-PXRE and Msx2-ERE (Fig. (Fig.3F3F).

An external file that holds a picture, illustration, etc.
Object name is zmb0220771260003.jpg

Identification of PXRE-like and ERE-like elements in the Msx2 gene promoter region. (A) Functional analysis of the Msx2 gene promoter. Plasmid constructs pSIREN-Shuttle-PXR (100 ng) and pSIREN-Shuttle-ERα (100 ng), together with the Msx2-3000 reporter (400 ng), were transfected in MC3T3-E1 cells, and the transfected cells were cultured for 24 h. Promoter activity was measured by luciferase (luc.) activity. All values are means ± standard deviations for at least three independent experiments. (B and C) Gene-specific knockdown using PXR (B) and ERα (C) RNAi vectors. The RNAi vectors were transfected into MC3T3-E1 cells for 48 h, and the protein levels were determined by immunoblotting. β-Actin was used as a control. (D) Regulation of the Msx2-3000 reporter by known PXR ligands, such as pregnenolone 16α-carbonitrile (PCN), 17α-hydroxypregnenolone (POH), mifepristone (MIF), and dexamethasone 21-acetate (DAC), in MC3T3-E1 cells. The amounts of each transfected plasmid are listed above for panel A. (E) Mutation of the Msx2 gene promoters. These mutant promoters were generated with a mutagenesis kit (Stratagene). (F) Analysis of the mutated Msx2 gene promoters by MK4 and E2 using endogenous expression receptors in MC3T3-E1 cells. The amounts of each transfected plasmid are described above for panel A.

An external file that holds a picture, illustration, etc.
Object name is zmb0220771260004.jpg

Direct recruitment of PXR/RXRα to Msx2-PXRE in the Msx2 gene promoter. (A) The position of Msx2-PXRE and Msx2-ERE in the Msx2 gene promoter. (B) The recruitment of PXR/RXRα and ERα to Msx2-PXRE and Msx2-ERE in the Msx2 gene promoter in vitro. The ABCD assay was performed with two copies of Msx2-PXRE and Msx2-ERE that were conjugated to the beads and incubated with whole-cell extracts from HEK 293T cells that overexpressed PXR, RXRα, and ERα together with the respective ligands for the NRs. The proteins interacting with DNA-conjugated beads were detected by immunoblotting. α-ER, anti-ER antibody; α-PXR, anti-PXR antibody; α-p300, anti-p300 antibody, con., consensus. (C) The recruitment of PXR/RXRα and ERα to the Msx2 gene promoter in vivo. ChIP analysis was performed with soluble chromatin prepared from MC3T3-E1 cells treated with MK4 and E2 for 1 h and immunoprecipitated with the indicated antibodies. Extracted DNA samples were amplified using primer pairs to detect the regions containing either Msx2-PXRE or Msx2-ERE. IgG, immunoglobulin G.

Direct DNA binding of PXR on the Msx2 promoter.

To determine whether PXR bound to the proposed PXRE (Fig. (Fig.4A),4A), we examined the DNA binding ability of PXR and ERα on the Msx2 gene promoter with an ABCD assay. We tested whole-cell extracts of MC3T3-E1 cells transiently overexpressing PXR, RXRα, or ERα. PXR/RXRα bound to Msx2-PXRE, while ERα binding was not seen for Msx2-PXRE (Fig. (Fig.4B).4B). Conversely, ERα bound, as expected, to Msx2-ERE (Fig. 4A and B). Moreover, in this assay, p300 also associated with the receptors. To verify if the observed DNA binding of PXR and ERα in vitro reflected the physiological events in the Msx2 promoter, we performed a ChIP analysis on the endogenous promoter of the Msx2 gene. E2-induced recruitment of the ERα, but not PXR, was observed in the upstream region of Msx2-ERE. In contrast, the ligand-independent association of PXR took place in the region of the distal promoter containing Msx2-PXRE (Fig. (Fig.4C).4C). Consistent with the type of recruited receptors, the known NR coactivator p300 also was recruited to the promoter (Fig. (Fig.4C4C).

MK4-induced osteoblast genesis is mediated by PXR.

To confirm the physiological impact of PXR function on VK-induced osteoblast genesis, we knocked down either Msx2 or PXR in MC3T3-E1 cells by RNAi with adenovirus (Fig. 5A and B). Expression of endogenous Msx2 and PXR was significantly attenuated by RNAi (Fig. 5A and B). As expected, osteoblast genesis induced by MK4 was attenuated when cells were infected with Msx2 RNAi adenovirus (the middle panels of Fig. 5C and D [lanes 5 to 8]). Moreover, in primary osteoblasts derived from ERαKO mice, osteoblast genesis induced by E2 also was attenuated (Fig. (Fig.5E).5E). The induction of marker genes (encoding type I collagen α and ALP) was, as expected, also abrogated with Msx2 RNAi adenovirus (Fig. (Fig.5F,5F, lanes 5 to 8 and 17 to 20). Moreover, PXR RNAi also inhibited MK4-dependent osteoblast genesis, as shown in the bottom panels of Fig. 5C, D (lanes 9 to 12), and F (lanes 9 to 12 and 21 to 24).

An external file that holds a picture, illustration, etc.
Object name is zmb0220771260005.jpg

VK and estrogen cooperatively promote osteoblast differentiation through Msx2 gene induction. (A and B) Gene-specific knockdown using Msx2 (A) and PXR (B) RNAi adenovirus. MC3T3-E1 cells were cultured with the RNAi adenovirus for 48 h, and the protein levels were determined by immunoblotting with each antibody. β-Actin antibody (α-β-actin) was used as a control. α-Msx2, anti-Msx2 antibody; α-PXR, anti-PXR antibody. (C) Msx2 mediates the effect of MK4 and E2 in osteoblast differentiation. MC3T3-E1 cells infected with the RNAi adenovirus for 48 h were incubated with MK4 and E2 for 24 h. Osteoblast differentiation was detected by ALP staining. (D) Msx2 mediates the effect of MK4 and E2 in osteoblast differentiation. MC3T3-E1 cells infected with the RNAi adenovirus for 48 h were incubated with MK4 and E2 for 24 h, and then ALP activity was measured. (E) The effect of E2 and MK4 in primary ERαKO osteoblast differentiation. Primary osteoblasts derived from ERαKO mice were incubated with MK4 and E2 for 24 h. The osteoblast differentiation was measured by ALP assay. (F) The expression levels of osteoblast differentiation marker genes induced by MK4 and E2 with Msx2 or PXR RNAi. MC3T3-E1 cells were infected with the RNAi (control, Msx2, or PXR) adenovirus for 48 h, followed by either no treatment or treatment with MK4 and E2 for 24 h. The total RNA then was extracted and used for qRT-PCR. (G) A schematic view of the VK and E2 actions at the Msx2 gene promoter.

DISCUSSION

This study demonstrates that Msx2 contains a PXRE in its gene promoter and that it is a direct target gene for MK4-bound PXR. Reflecting the similar effects of E2 and MK4, an ERE also was mapped in the Msx2 gene promoter. Although each of the response elements alone was not remarkable, the combined effects of MK4 and E2 were evident in transactivation. In osteoblastic differentiation induced by MK4, E2, or both, the knockdown of endogenous Msx2 abrogated the effects of the NR ligands. Thus, Msx2 likely mediates, at least in part, the effects of both MK4 and E2 on osteoblastic differentiation. Msx2 is one of the prime osteoblastogenic factors in an intact animal (26). Our observation that osteoblast genesis is induced by MK4 in cultured preosteoblastic cells suggests that MK4 exerts its osteoprotective action by upregulating Msx2 gene expression and increasing osteoblast genesis (Fig. (Fig.5G).5G). Since extracellular matrix Tsukushi genes also are VK target genes (8), the osteoprotective effects of VK in the human are mediated both by the classical pathway of protein γ-carboxylation and through genomic action via PXR/SXR.

PXR/SXR is activated by a number of endogenous and exogenous ligands (23). PXR/SXR plays a prominent role in the detoxification of chemicals (36). PXR/SXR is considered a global sensor for low-molecular-mass fat-soluble drugs that also is responsible for the timely degradation of these compounds (14). Thus, in contrast to steroid hormone receptors, PXR/SXR responds to a variety of ligands with unrelated structures. The three-dimensional structure of the PXR/SXR ligand binding domain (LBD) is known (35). The LBD is flexible, with a wider cave than those of steroid receptors (37). This permits PXR/SXR to capture a variety of ligands, including MK4. Shifting of the C-terminal transactivation helix H12 in the ERα LBD alters the conformation of the receptor, depending on whether an agonist or antagonist is bound. This plasticity enables ERα to respond to a variety of different ligands (27). It is possible that MK4 binding induces an H12 shift that is distinct from those induced by the other ligands, since the transactivation of PXR in the Msx2 promoters was induced by MK4 but not by the other known PXR/SXR ligands in the osteoblastic cell lines. Thus, the MK4-induced angle in the H12 shifting of PXR might be preferential to recruit coactivator/coactivator complexes to the Msx2 gene promoter in preosteoblasts. In fact, the MK4-induced association of PXR with the well-characterized coactivator (p300) was detectable in both an ABCD assay and a ChIP analysis of the Msx2 gene promoter. Our observations are a starting point for investigating coregulators (17, 24) associating with MK4-bound PXR in osteoblastic cells. The identification of PXR coregulators will enhance the understanding of the osteoprotective effects of VK at a molecular level.

Acknowledgments

We thank P. Chambon for kindly providing ERαKO mice; S. Takezawa, F. Ohtake, M. S. Kim, S. Fujiyama, Y. Mezaki, R. Fujiki, M. Kouzu-Fujita, T. Matsumoto, and Y. Imai for technical assistance; T. Yoshizawa and H. Kawashima (Niigata University) for the kind gift of Max2 expression vector; Eisai Co., Ltd., for the gift of MK4; T. Matsumoto, D. Inoue, and R. Okazaki for helpful discussions; and H. Higuchi and K. Hiraga for manuscript preparation.

This work was supported in part by a Grant-in-Aid for Basic Research Activities for Innovative Bioscience (BRAIN).

Footnotes

[down-pointing small open triangle]Published ahead of print on 17 September 2007.

REFERENCES

1. Bertilsson, G., J. Heidrich, K. Svensson, M. Åsman, L. Jendeberg, M. Syndow-Bäckman, R. Ohlsson, H. Postlind, P. Blomquist, and A. Berkenstam. 1998. Identification of a human nuclear receptor defines a new signaling pathway for CYP3A4 induction. Proc. Natl. Acad. Sci. USA 95:12208-12213. [Europe PMC free article] [Abstract] [Google Scholar]
2. Blumberg, B., W. Sabbagh, Jr., H. Juguilon, J. Bolado, Jr., C. M. van Meter, E. S. Ong, and R. M. Evans. 1998. SXR, a novel steroid and xenobiotic-sensing nuclear receptor. Gene Dev. 12:3195-3205. [Europe PMC free article] [Abstract] [Google Scholar]
3. Booth, S. L., K. L. Tucker, H. Chen, M. T. Hannan, D. R. Gagnon, L. A. Cupples, P. W. F. Wilson, J. Ordovas, E. J. Schaefer, B. Dawson-Hughes, and D. P. Kiel. 2000. Dietary vitamin K intakes are associated with hip fracture but not with bone mineral density in elderly men and women. Am. J. Clin. Nutr. 71:1201-1208. [Abstract] [Google Scholar]
4. Ducy, P., C. Desbois, B. Boyce, G. Pinero, B. Story, C. Dunstan, E. Smith, J. Bonadio, S. Goldstein, C. Gundberg, A. Bradley, and G. Karsenty. 1996. Increased bone formation in osteocalcin-deficient mice. Nature 382:448-452. [Abstract] [Google Scholar]
5. Fujiki, R., M. S. Kim, Y. Sasaki, K. Yoshimura, H. Kitagawa, and S. Kato. 2005. Ligand-induced transrepression by VDR through association of WSTF with acetylated histones. EMBO J. 24:3881-3894. [Europe PMC free article] [Abstract] [Google Scholar] Retracted
6. Fujikawa, K., A. R. Thompson, M. E. Legaz, R. G. Meyer, and E. W. Davie. 1973. Isolation and characterization of bovine factor IX (Christmas factor). Biochemistry 12:4938-4945. [Abstract] [Google Scholar]
7. Hauschka, P. V., and M. L. Reid. 1978. Vitamin K dependence of a calcium-binding protein containing γ-carboxyglutamic acid in chicken bone. J. Biol. Chem. 253:9063-9068. [Abstract] [Google Scholar]
8. Ichikawa, T., K. Horie-Inoue, K. Ikeda, B. Blumberg, and S. Inoue. 2006. Steroid and xenobiotic receptor SXR mediates vitamin K2-activated transcription of extracellular matrix-related genes and collagen accumulation in osteoblastic cells. J. Biol. Chem. 281:16927-16934. [Abstract] [Google Scholar]
9. Iwamoto, I., S. Kosha, S. Noguchi, M. Murakami, T. Fujimoto, T. Douchi, and Y. Nagata. 1999. A longitudinal study of the effect of vitamin K2 on bone mineral density in postmenopausal women a comparative study with vitamin D3 and estrogen-progestin therapy. Maturitas 31:161-164. [Abstract] [Google Scholar]
10. Jackson, C. M., and D. J. Hanahan. 1968. Studies on bovine factor X. II. Characterization of purified factor X. Observations on some alterations in zone electrophoretic and chromatographic behavior occurring during purification. Biochemistry 7:4506-4517. [Abstract] [Google Scholar]
11. Kaneki, M., S. J. Hedges, T. Hosoi, S. Fujiwara, A. Lyons, S. J. Crean, N. Ishida, M. Nagasawa, M. Takechi, Y. Sano, Y. Mizuno, S. Hoshino, M. Miyao, S. Inoue, K. Horiki, M. Shiraki, Y. Ouchi, and H. Orimo. 2001. Japanese fermented soybean food as the major determinant of the large geographic difference in circulating levels of vitamin K2 possible implications for hip-fracture risk. Nutrition 17:315-321. [Abstract] [Google Scholar]
12. Kim, M. S., R. Fujiki, A. Murayama, H. Kitagawa, K. Yamaoka, Y. Yamamoto, M. Mihara, K. Takeyama, and S. Kato. 2007. 1α, 25 (OH) 2D3-induced transrepression by vitamin D receptor through E-box-type elements in the human parathyroid hormone gene promoter. Mol. Endocrinol. 21:334-342. [Abstract] [Google Scholar]
13. Kitagawa, H., R. Fujiki, K. Yoshimura, Y. Mezaki, Y. Uematsu, D. Matsui, S. Ogawa, K. Unno, M. Okubo, A. Tokita, T. Nakagawa, T. Ito, Y. Ishimi, H. Nagasawa, T. Matsumoto, J. Yanagisawa, and S. Kato. 2003. The chromatin-remodeling complex WINAC targets a nuclear receptor to promoters and is impaired in Williams syndrome. Cell 113:905-917. [Abstract] [Google Scholar]
14. Kliewer, S. A., B. Goodwin, and T. M. Willson. 2002. The nuclear pregnane X receptor: a key regulator of xenobiotic metabolism. Endocr. Rev. 23:687-702. [Abstract] [Google Scholar]
15. Kliewer, S. A., J. T. Moore, L. Wade, J. L. Staudinger, M. A. Watson, S. A. Jones, D. D. McKee, B. B. Oliver, T. M. Willson, R. H. Zetterström, T. Perlmann, and J. M. Lehmann. 1998. An orphan nuclear receptor activated by pregnanes defines a novel steroid signaling pathway. Cell 92:73-82. [Abstract] [Google Scholar]
16. Lehmann, J. M., D. D. McKee, M. A. Watson, T. M. Willson, J. T. Moore, and S. A. Kliewer. 1998. The human orphan nuclear receptor PXR is activated by compounds that regulated CYR3A4 gene expression and cause drug interactions. J. Clin. Investig. 102:1016-1023. [Europe PMC free article] [Abstract] [Google Scholar]
17. Li, B., M. Carey, and J. L. Workman. 2007. The role of chromatin during transcription. Cell 128:707-719. [Abstract] [Google Scholar]
18. Mani, S., H. Huang, S. Sundarababu, W. Liu, G. Kalpana, A. B. Smith, and S. B. Horwitz. 2005. Activation of the steroid and xenobiotic receptor (human pregnane X receptor) by nontaxane microtubule-stabilizing agents. Clin. Cancer Res. 11:6359-6369. [Abstract] [Google Scholar]
19. Nakamura, T., Y. Imai, T. Matsumoto, S. Sato, K. Takeuchi, K. Igarashi, Y. Harada, Y. Azuma, A. Krust, Y. Yamamoto, H. Nishina, S. Takeda, H. Takayanagi, D. Metzger, J. Kanno, K. Takaoka, T. J. Martin, P. Chambon, and S. Kato. 2007. Estrogen prevents bone loss via estrogen receptor α and induction of Fas ligand in osteoclast. Cell 130:811-823. [Abstract] [Google Scholar]
20. Ohtake, F., A. Baba, I. Takada, M. Okada, K. Iwasaki, H. Miki, S. Takahashi, A. Kouzmenko, K. Nohara, T. Chiba, Y. Fujii-Kuriyama, and S. Kato. 2007. Dioxin receptor is a ligand-dependent E3 ubiqoutin ligase. Nature 446:562-566. [Abstract] [Google Scholar]
21. Price, P. A., A. S. Otsuka, J. W. Poser, J. Kristaponis, and N. Raman. 1976. Characterization of a γ-carboxyglutamic acid-containing protein from bone. Proc. Natl. Acad. Sci. USA 73:1447-1451. [Europe PMC free article] [Abstract] [Google Scholar]
22. Price, P. A., and M. K. Williamson. 1985. Primary structure of bovine matrix Gla protein, a new vitamin K-dependent bone protein. J. Biol. Chem. 260:14971-14975. [Abstract] [Google Scholar]
23. Reschly, E. J., and M. D. Krasowski. 2006. Evolution and fuction of the NR11 nuclear hormone receptor subfamily (VDR, PXR, and CAR) with respect to metabolism of xenobiotics and endogenous compounds. Curr. Drug Metab. 7:349-365. [Europe PMC free article] [Abstract] [Google Scholar]
24. Rosenfeld, M. G., V. V. Lunyak, and C. K. Glass. 2006. Sensors and signals: a coactivator/corepressor/epigenetic code for integrating signal-dependent programs of transcriptional response. Gene Dev. 20:1405-1428. [Abstract] [Google Scholar]
25. Sato, T., Y. Ohtani, Y. Yamada, S. Saitoh, and H. Harada. 2002. Difference in the metabolism of vitamin K between liver and bone in vitamin K-deficient rats. Br. J. Nutr. 87:307-314. [Abstract] [Google Scholar]
26. Satokata, I., L. Ma, H. Ohshima, M. Bei, I. Woo, K. Nisizawa, T. Maeda, Y. Takano, M. Uchiyama, S. Heaney, H. Peters, Z. Tang, R. Maxson, and R. Maas. 2000. Msx2 deficiency in mice causes pleiotropic defects in bone growth and ectodermal organ formation. Nat. Genet. 24:391-395. [Abstract] [Google Scholar]
27. Shiau, A. K., D. Barstad, J. T. Radek, M. J. Meyers, K. W. Nettles, B. S. Katzenellenbogen, J. A. Katzenellenbogen, D. A. Agard, and G. L. Greene. 2002. Structural characterization of a subtype-selective ligand reveals a novel mode of estrogen receptor antagonism. Nat. Struct. Biol. 9:359-364. [Abstract] [Google Scholar]
28. Shiraki, M., Y. Shiraki, C. Aoki, and M. Miura. 2000. Vitamin K2 (menatetrenone) effectively prevents fracture and sustains lumbar bone mineral density in osteoporosis. J. Bone Miner. Res. 15:515-521. [Abstract] [Google Scholar]
29. Stenflo, J., P. Fernlund, W. Egan, and P. Roepstorff. 1974. Vitamin K dependent modifications of glutamic acid residues in prothrombin. Proc. Natl. Acad. Sci. USA 71:2730-2733. [Europe PMC free article] [Abstract] [Google Scholar]
30. Sudo, H., H. A. Kodama, Y. Amagi, S. Yamamoto, and S. Kasai. 1983. In vitro differentiation and calcification in a new clonal osteogenic cell line derived from newborn mouse calvaria. J. Cell Biol. 96:191-198. [Europe PMC free article] [Abstract] [Google Scholar]
31. Suttie, J. W. 1980. Mechanism of action of vitamin K: synthesis of γ-carboxyglutamic acid. CRC Crit. Rev. Biochem. 8:191-223. [Abstract] [Google Scholar]
32. Suzawa, M., I. Takada, J. Yanagisawa, F. Ohtake, S. Ogawa, T. Yamauchi, T. Kadowaki, Y. Takeuchi, H. Shibuya, Y. Gotoh, K. Matsumoto, and S. Kato. 2003. Cytokines suppress adipogenesis and PPAR-γ function through the TAK1/TAB1/NIK cascade. Nat. Cell Biol. 5:224-230. [Abstract] [Google Scholar]
33. Tabb, M. M., A. Sun, C. Zhou, F. Grün, J. Errandi, K. Romero, H. Pham, S. Inoue, S. Mallick, M. Lin, B. M. Forman, and B. Blumberg. 2003. Vitamin K2 regulation of bone homeostasis is mediated by the steroid and xenobiotic receptor SXR. J. Biol. Chem. 278:43919-43927. [Abstract] [Google Scholar]
34. Takezawa, S., A. Yokoyama, M. Okada, R. Fujiki, A. Iriyama, Y. Yanagi, H. Ito, I. Takada, M. Kishimoto, A. Miyajima, K. Takeyama, K. Umesono, H. Kitagawa, and S. Kato. 2007. A cell cycle-dependent co-repressor mediates photoreceptor cell-specific nuclear receptor function. EMBO J. 26:764-774. [Europe PMC free article] [Abstract] [Google Scholar] Retracted
35. Watkins, R. E., G. B. Wisely, L. B. Moore, J. L. Collins, M. H. Lambert, S. P. Williams, T. M. Willson, S. A. Kliewer, and M. R. Redinbo. 2001. The human nuclear xenobiotic receptor PXR: structural determinants of directed promiscuity. Science 292:2329-2333. [Abstract] [Google Scholar]
36. Willson, T. M., and S. A. Kliewer. 2002. PXR, CAR and drug metabolism. Nat. Rev. Drug Discov. 1:259-266. [Abstract] [Google Scholar]
37. Wurtz, J. M., W. Bourguet, J. P. Renaud, V. Vivat, P. Chambon, D. Moras, and H. Gronemeyer. 1996. A canonical structure for the ligand-binding domain of nuclear receptors. Nat. Struct. Biol. 3:87-94. [Abstract] [Google Scholar]
38. Yanagisawa, J., H. Kitagawa, M. Yanagida, O. Wada, S. Ogawa, M. Nakagomi, H. Oishi, Y. Yamamoto, H. Nagasawa, S. B. McMahon, M. D. Cole, L. Tora, N. Takahashi, and S. Kato. 2002. Nuclear receptor function requires a TFTC-type histone acetyl transferase complexes. Mol. Cell. 9:553-562. [Abstract] [Google Scholar]

Articles from Molecular and Cellular Biology are provided here courtesy of Taylor & Francis

Citations & impact 


Impact metrics

Jump to Citations
Jump to Data

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/2107002
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/2107002

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/10.1128/mcb.00813-07

Supporting
Mentioning
Contrasting
1
53
1

Article citations


Go to all (56) article citations

Data