当前位置: 首页 > 期刊 > 《动脉硬化血栓血管生物学》 > 2004年第9期 > 正文
编号:11330438
Homeobox Protein Hex Facilitates Serum Responsive Factor–Mediated Activation of the SM22 Gene Transcription in Embryonic Fibroblasts
http://www.100md.com 《动脉硬化血栓血管生物学》
     From the Department of Medicine and Biological Science (Y.O., K.K.-K., K.S., M.S., H.S., M.Y., Y.O., Y.A., T.I., E.O., M.K.), Graduate School of Medicine, University of Gunma, Gunma; and the Department of Cardiovascular Medicine (R.N.), Graduate School of Medicine, University of Tokyo, Tokyo, Japan.

    ABSTRACT

    Objective— Hex (hematopoietically expressed homeobox), a member of homeobox family of transcription factors, has been implicated in the vascular development because of its expression in hemangioblast, a hypothetical stem cell that gives rise to both angioblasts and hematopoietic lineages. In the present study, we examined the role of Hex in the differentiation of vascular smooth muscle cells.

    Methods and Results— We constructed adenovirus expressing Hex, to which we refer to as AxCA/Hex, and transduced murine embryonic fibroblasts, 10T1/2 cells. Northern blot analyses showed that Hex increased the mRNA levels of smooth muscle -actin and SM22 but not of calponin and smooth muscle myosin heavy chain. Transient transfection assays showed that Hex activates the transcription from the SM22 promoter in a CArG box-dependent manner. Electrophoretic mobility shift assays demonstrate that Hex is not able to bind to CArG box, but binding of serum responsive factor (SRF) to CArG box is enhanced in AxCA/Hex-transduced cells. Recombinant Hex protein produced by in vitro translation system augmented the binding activity of SRF to CArG box. Immunoprecipitation experiments revealed the physical association between Hex and SRF.

    Conclusions— Hex induces transcription of the SM22 gene by facilitating the interaction between SRF and its cognate binding site in pluripotent embryonic fibroblasts.

    This study demonstrates that Hex, a hematopoietically expressed homeobox protein, induces transcription of the SM22 gene by facilitating the interaction between SRF and its cognate binding site in embryonic fibroblasts. These findings will provide the clue for understanding the mechanisms by which bone marrow-derived SMC precursor cells undergo differentiation.

    Key Words: transcriptional factor ? smooth muscle cells ? atherosclerosis ? homeobox ? serum response factor

    Introduction

    Homeobox transcription factors control a range of cellular activity including induced expression of a set of lineage-specific genes.1 A family of vertebrate homeobox genes related to the Drosophila tinman (tin) gene plays a crucial role for cardiogenic differentiation. Nkx2.3, Nkx2.5, and Nkx2.7 are expressed in the developing heart tissues.2,3 In particular, Nkx2.5 is selectively expressed in cardiac cell progenitors before cardiogenic differentiation4,5 and activates cardiac muscle-specific genes, including cardiac -actin,6 myosin heavy chain,5 and atrial natriuretic peptide genes.7 Ablation of Nkx2.5 gene function in the mouse leads to atypical cardiac development.8,9 In contrast, little is currently known about the transcription factors involved in formation of the embryonic vasculature, although many angiogenic peptides and their signal transduction cascades have been identified that regulate the process of vasculogenesis and angiogenesis.

    The divergent homeobox gene Hex, also known as Prh, is expressed in early endothelial precursors and angioblasts, as well as in a range of multipotent hematopoietic progenitor cells.10,11 Hex is expressed in the nascent blood islands of the visceral yolk sac and later in embryonic angioblasts.11,12 Endothelial expression of Hex is transient and commences slightly after expression of the receptor tyrosine kinase gene, flk-1, which is known to be essential for vascular development.12 Once endothelial cell differentiation commences, Hex expression starts to decline.11 Such a downregulation of Hex expression appears to be required for terminal differentiation to occur. Overexpression of Xenopus laevis Hex (XHex) in the frog embryo causes disruption to developing vascular structure and an increase in the number of vascular endothelial cells.12 Because endothelial precursors are originated from blood islands, and because smooth muscle cells (SMC) arise from multiple types of progenitors including endothelial cells, we hypothesize that Hex, which is implicated in the endothelial and blood differentiation, is also involved in SMC differentiation.

    Recently, we have identified that Hex expression is induced in neointima after balloon injury.13 Because Hex expression is associated with the expression of the embryonic/nonmuscle isoform of myosin heavy chain (SMemb/NMHC-B) gene in neointima, we have examined whether Hex regulates the expression of this gene. We found that Hex activates the promoter activity of the SMemb/NMHC-B gene through cAMP-responsive element binding protein (CREB) binding site in vitro. However, it is unlikely that interaction between Hex and CREB plays a role in regulating the SMC differentiation marker genes, including SM -actin and SM22,14 which are known to be expressed in neointima, because no previous studies have identified functional CRE within SMC marker gene promoters. Thus, the role of Hex in regulating the expression of SMC-specific genes remains to be determined.

    In the present study, we demonstrated that Hex activates the endogenous smooth muscle -actin (SM -actin) and SM22 genes in non-SMC. In addition, we showed that such an induction is dependent on intact CArG (CC(A/T)6GG) box, which serves as the efficient binding site for serum responsive factor (SRF) and plays a crucial role in the expression of the SMC differentiation marker genes. By using electrophoretic mobility shift assays of in vitro-translated proteins of Hex and SRF, we then asked how this induction was mediated. Although Hex was not able to bind to CArG box, it enhanced the binding activity of SRF to CArG box. Immunoprecipitation assays showed that Hex interacts in vitro with SRF. On the basis of the results presented here, we propose that Hex can impart the SMC-specific function to the CArG box of SM22 by physically interacting with SRF.

    Methods

    Cell Cultures

    The 10T1/2 cells (murine embryonic fibroblast cells) have been previously described.15 Cells were grown at 37°C in a 5% CO2 atmosphere in Dulbecco modified Eagle’s medium (DMEM) supplement with 10% fetal bovine serum and 1% penicillin/streptomycin. Rat aorta-derived SMCs have been previously described.16

    Recombinant Adenovirus Expression Constructs

    The recombinant adenovirus vectors were generated as previously described.17 AxCA/LacZ and AxCA/Hex were prepared by inserting the ?-galactosidase or Hex cDNAs, respectively, into the Ad E1-deleted region under the control the CAG promoter.

    Northern Blot Analysis

    Total RNA from 10T1/2 cells transduced with either AxCA/LacZ or AxCA/Hex was isolated using the ISOGEN reagent (Nippongene) in accordance with the manufacturer’s instructions. Northern blot analysis was performed as previously described.18

    Western Blot Analysis

    Cytosolic extracts from 10T1/2 cells infected with either AxCA/LacZ or AxCA/Hex were electrophoresed as previously described18 and blotted with anti-Hex antibody. Anti-Hex antibody was synthesized as previously described.13

    Plasmid Constructs

    The mouse SM22 promoter/luciferase fusion plasmid (SM22/pGL2) was kindly provided by Dr K. Hasegawa (Kyowahakko Co Ltd, Tokyo, Japan). For generation of serial deletion construct, SM22/pGL2 was digested with SacI and HindIII (SM22-1309Luc), PstI and HindIII (SM22-445Luc), SmaI and HindIII (SM22-115Luc), gel-purified and subcloned into pGL3 (Promega). SM22-158Luc and SM22-171Luc were prepared by polymerase chain reaction (PCR) reaction with SM22/pGL2 as a template. The forward and reverse primers used for generation of SM22-171Luc were 5'-CCCGAGCTCGCTCCAACTTGGTG-TCTTTCC-3' and 5'-GGGAAGCTT-GAAGGAGAGTAGCTTCGGTGT-3' with SacI and HindIII sites (italic), respectively. The forward and reverse primers used for generation of SM22-158Luc were 5'-CCCGAGCTCTTTC-CCCAAATATGGAGCCTG -3' and 5'-GGGAAGCTTGAAGGAGAGTAGCTT-CGGTGT-3'. The PCR products were gel-purified, digested, and subcloned into pGL3.

    For generation of mutants, the following forward and reverse primers were used in a PCR reaction with SM22-158Luc as a template. The primers (mutations of wild-type sequence appear in bold) for SM22-158m1Luc, SM22-158m2Luc, and SM22-158m3Luc were as follows: SM22-158m1Luc, 5'-CCCGAGCTCGGGAACCAAATATGGAGCCTG-3', SM22-158m2Luc, 5'-CCCGAGCTCTTTCCAACCCTATGGAGCCTG-3', and for SM22-158m3Luc was 5'-CCCGAGCTCTTTCCCCAAAGCGTTAGCCTG -3'.

    Hex expression plasmid (Hex/cytomegalovirus ) has been previously described.13 The expression plasmid SRF/pME18S, the coding region of mouse SRF, (kindly provided by Dr T. Miwa) has been described.19

    Transfection and Luciferase Assays

    The 10T1/2 cells were transfected with a series of the SM22 reporter construct (1 μg), along with 1 μg of expression plasmid by a modified calcium phosphate precipitation method as previously described.18 Cells were harvested in cell lysis buffer at 48 hours after transfection. Luciferase activities were assayed according to the manufacturer’s specifications (Promega).

    Electrophoretic Mobility Shift Assays

    Nuclear extracts from 10T1/2 cells were prepared as previously described.20 The sequences for sense strand of double-stranded oligonucleotides used as probes or competitors in electrophoretic mobility shift assays (EMSA) were as follows (with a consensus motif in italics and mutations of wild-type sequence in bold): SM22 (-158/-133), 5'-GTCTTTCCCCAAATATGGAGCCTGTG-3'; mut-1, 5'- GTCGGGAACCAAAT-ATGGAGCCTGTG -3'; mut-2, 5'- GTCTTTCCAACCCTATGGAGCCTGTG-3'; mut-3, 5'- GTCTTTCCCCAAAGCGTTAGCCTGTG-3'’; mut-4, 5'- GTCTTTCCCCAAATATGGCTAAGGTG-3'; Homeobox protein, 5'-TGCCAC-TTAATCATTAAGGGAGCC-3'; CArG, 5'-TCGAGCCCCAAATATGGAGC-CCCAAATATGGAGC-3'; and Sp1, 5'-ATTCGATCGGGGCGGGGCGAGC-3'. Binding reactions were performed as previously described.21

    In Vitro Synthesis of Hex and SRF

    In vitro synthesis of Hex and SRF were performed using TNT T7 Quick Coupled Translation System (Promega) with Hex/pcDNA3 and SRF/pGEM2 as templates. For generation of Hex/pcDNA3, mouse Hex cDNA was inserted into pcDNA3.1 (Invitrogen). SRF/pGEM2, the coding region of human SRF, was kindly provided by Dr I. Manabe (University of Tokyo, Japan).

    Immunoprecipitation Assay

    The 10T1/2 cells were transfected with SRF/pME18S and Hex/pcDNA3.1 (HA epitope tagged). At 24 hours after transfection, the protein lysis solution was immunoprecipitated with normal rabbit IgG, anti-SRF, and anti-HA probe antibodies (Santa Cruz Biotechnology, Inc). Immunoprecipitated protein lysis solution was electrophoresed on 15% SDS-polyacrylamide gels and then transferred onto nitrocellulose membrane (Schleicher and Schuell, Inc).

    Results

    Hex Induces SMC-Specific Marker Gene Expression

    To determine the role of Hex in the process of SMC differentiation, we first constructed AxCA/Hex, which contains mouse Hex cDNA. Figure 1A and 1B show the mRNA and protein expression of Hex in the 10T1/2 cells transduced with AxCA/Hex. Figure 1C shows that Hex expression is localized in the nuclei of 10T1/2 cells.

    Figure 1. Construction of Adex-Hex. A, Northern blot analysis of 10T1/2 cells transduced with AxCA/Hex. 10T1/2 cells were incubated in 10-cm dishes and transduced with indicated amount of AxCA/Hex or AxCA/LacZ. Twenty-four hours after infection, total cellular RNA (15 μg) was prepared and analyzed by Northern blotting for Hex. B, Western blot analysis of 10T1/2 cells transduced with AxCA/Hex. Proteins from 10T1/2 cells transduced with AxCA/Hex (100 multiplicity of infection ) or AxCA/LacZ (100 moi) for 24 hours were electrophoresed on 15% SDS-polyacrylamide gels, transferred onto a nitrocellulose membrane, and immunostained with anti-Hex antibody. C, Immunostaining of 10T1/2 cells transduced with AxCA/Hex. Twenty-four hours after infection with AxCA/Hex (100 moi), cells were immunostained with anti-Hex antibody.

    We next transduced 10T1/2 cells with AxCA/Hex and performed Northern blot analyses. As shown in Figure 2, the mRNA levels of SMC-specific genes, SM -actin and SM22, were significantly increased in AxCA/Hex-transduced 10T1/2 cells as compared with AxCA/LacZ-transduced 10T1/2 cells. In contrast, Hex did not affect the mRNA levels of these SMC-specific genes in rat aortic SMCs in which the SM -actin and SM22 genes are highly expressed under basal condition.

    Figure 2. Hex induces SMC-specific marker gene expression. Northern blot analysis. 10T1/2 cells (left) and rat aorta-derived SMCs (right) were cultured in 10-cm dishes and were harvested 24 hours after infection with AxCA/Hex (100 moi) or AxCA/LacZ (100 moi). Total RNA (15 μg) from 10T1/2 cells and rat aortic SMCs were transferred to nylon membranes. SM -actin and SM22 cDNAs were used as probes.

    Mapping of the Hex Responsive Region Within the SM22 Promoter

    To investigate whether Hex regulates the SM22 expression at the transcriptional level, we performed transient transfection assays. We cotransfected 10T1/2 cells with SM22 (–1309/+41) Luc/reporter gene, designated as SM22-1309Luc and Hex expression vector (Hex/CMV). As shown in Figure 3A, Hex/CMV increased luciferase activity of SM22-1309Luc by 3-fold.

    Figure 3. Mapping of the Hex-responsive region within the SM22 promoter. A, A series of 5'-deletion reporter constructs containing the SM22 promoter region and the luciferase gene were transfected into 10T1/2 cells along with pRC/CMV (empty vector) or Hex/CMV (Hex-expressing vector). Transfection into 10T1/2 cells was performed by a modified calcium phosphate precipitation method. After 48 hours of post-transfection, cells were harvested and luciferase activities were measured. Luciferase activities are expressed relative to that of pGL3, which set at 1.0. Values are mean±SEM for 3 separate experiments. B, Schematic representation of the base substitutions into SM22-158Luc. Mutations of wild-type sequence appear in bold. C, Effect of the mutation of CArG box on the SM22 promoter activity. Wild-type (SM22-158Luc) or mutated constructs (SM22-158m1Luc, SM22-158mut2Luc, and SM22-158mut3Luc) were cotransfected with either pRC/CMV or Hex/CMV. Luciferase activities are expressed relative to that of pGL3, which is set at 1.0. Values are mean±SEM for 3 separate experiments.

    To delineate the sequence responsible for the activation of SM22 promoter by Hex, progressive deletion of 5'-flanking region was performed. The SM22-promoter constructs SM22-445Luc and SM22-171Luc, in which the 5' ends correspond to –445 and –171, respectively, were activated by Hex, whereas SM22-115Luc and promoterless construct pGL3 were unresponsive. These data indicate that Hex-responsive sequence is localized between –171 and –115. Sequence analysis of this region revealed a sequence identical to CArG box, 5'-CCAAATATGG-3', at –150. To test whether CArG box is necessary for the activation of the SM22 promoter by Hex, we made constructs in which mutation was introduced into CArG box in the context of SM22-158Luc (Figure 3B and 3C). The construct SM22-158m1Luc, which contains mutation outside CArG box, exhibited the same activation in promoter activity as the wild-type promoter construct SM22-158Luc in response to Hex/CMV, whereas the constructs SM22-158m2Luc and SM22-158m3Luc both contain mutation within the CArG motif and did not respond to Hex/CMV.

    Identification of Nuclear Factor Binding Sites in the SM22 Promoter

    To investigate the molecular mechanisms underlying the activation of the CArG-dependent transcription by Hex, we performed EMSA to determine whether Hex is capable of binding directly to CArG box. We used double-stranded oligonucleotide containing sequence from –158 to –133 of the SM22 gene as a probe (shown in Figure 4A) and incubated with nuclear extracts from 10T1/2 cells transduced with AxCA/Hex. As shown in Figure 4B, prominent shifted complex C1 was observed. Formation of C1 was sequence-specific because although addition of 100 molar excess of unlabeled probe or m1, m4, and CArG sequence almost completely abolished the complex formations, m2, m3, and Hex binding sequence had no effects on complex formation. Based on the sequence mutated in m2 and m3, these results indicate that sequence involved in the complex C1 formation is localized to CArG box between –150 and –141. We next performed supershift assay. Incubation of the nuclear extracts with anti-SRF antibodies completely supershifted complex C1. In contrast, anti-Hex and anti-Sp1 antibody had no effect on complex C1 formation (Figure 4C). These results suggest that C1 complex is constituted by SRF alone and Hex is not a measurable component of C1 complex.

    Figure 4. Identification of nuclear factor binding sites in the SM22 promoter. A, Nucleotide sequences of the oligonucleotides used in the competitive EMSA were shown. Probe sequence is derived from the SM22 promoter spanning between –158 and –133. Five successive mutations introduced into this sequence are indicated by bold. B, EMSA. Nuclear extracts (10 μg) prepared from AxCA/Hex transduced 10T1/2 cells were incubated with 32P-labeled –158/–133 probe, in the absence or presence of a 100-fold molar excess of indicated unlabeled competitors. Position of the sequence-specific DNA: protein complex C1 is indicated. Consensus binding sequences for Hex and CArG are shown in Methods. C, Supershift assays using nuclear extracts prepared from AxCA/Hex-transduced 10T1/2 cells were performed using indicated antibodies. Supershifted complex (SS) is indicated by arrow. D, Western blot analysis. 10T1/2 cells transduced with either AxCA/Hex (100 moi) or AxCA/LacZ (100 moi) for 24 hours. Protein was harvested from adeno-infected cells. Immunostaining was performed using anti-SRF antibody.

    Overexpression of Hex Enhances Binding of SRF to CArG Box but Does not Increase SRF Expression

    To explore the mechanisms whereby Hex enhanced the promoter activity in CArG box-dependent manner, we first performed Western blot analysis and examined whether Hex had an effect on SRF protein levels. As shown in Figure 4D, Western blot analysis of 10T1/2 cells, which were transduced with either AxCA/LacZ or AxCA/Hex showed no discernable increase in Hex protein levels in Hex-transduced cells compared with LacZ-transduced cells. We then tested whether Hex increases the DNA binding of SRF. As shown in Figure 5A, the intensity of complex C1, which is constituted by SRF, was clearly increased in the nuclear extracts prepared from the cells transduced with AxCA/Hex. Intensity of Sp1 binding was equivalent between samples used in the EMSA, indicating that observed change in intensity of C1 complex is not caused by the difference or variation in quality and quantity of the nuclear extracts used (Figure 5B).

    Figure 5. Hex increases the binding of SRF to CArG box. A, 10T1/2 cells were transduced with either AxCA/LacZ or AxCA/Hex. Nuclear extracts prepared from 10T1/2 cells were incubated with –158/–133 probe. Position of the sequence-specific DNA: protein complex (C1) is indicated. B, Same experiments were performed using the Sp1 consensus sequence as a probe. Consensus binding sequences for Sp1 are shown in Methods. C, EMSA were performed with in vitro-translated SRF (rSRF) and –158/–133 probe. Various amounts of in vitro-translated Hex (rHex) were added to binding reactions. Unprogrammed (unprog) reticulocyte lysates and rHex alone were incubated with –158/–133 probe as a control. D, Recombinant SRF and Hex were labeled with 35S-Methionine (Amersham) and electrophoresed on 12% SDS-polyacrylamide gels.

    Recombinant Hex Enhances Binding of SRF to CArG Box

    To further examine the interaction between Hex and SRF, we next performed EMSA using recombinant Hex (rHex) and SRF (rSRF) produced in a reticulocyte lysate system. As shown in Figure 5C, radioactive complex was barely detected when incubated with 1 μL of rSRF and 32P-labeled probe SM22 (–158/–133), which contains CArG box. Incubation of 2 μL of rSRF with the probe gave rise to a weakly shifted complex. Addition of rHex markedly enhanced SRF binding to CArG box in a dose-dependent manner. No retarded bands were observed when rHex or unprogrammed lysates were incubated with probe. Addition of unprogrammed lysate did not change SRF binding. SDS-PAGE of rSRF and rHex was performed for control (Figure 5D).

    Hex Directly Binds to SRF

    We asked whether the enhancement of SRF binding to CArG box in the presence of Hex was mediated through protein–protein association. We performed immunoprecipitation assay to investigate the interaction of SRF and Hex. The 10T1/2 cells were transfected with SRF/pME18S and Hex expression vector expressing the HA epitope-tagged Hex protein. Normal rabbit IgG, anti-HA probe antibody, and anti-SRF antibody were used for immunoprecipitation. The resulting immunoprecipitates were resolved by SDS-PAGE, transferred onto nitrocellulose membrane, and subjected to immunoblot analysis. As shown in Figure 6, the anti-SRF antibody coimmunoprecipitated HA epitope-tagged Hex and the anti-HA antibody coimmunoprecipitated SRF. Our results indicated that Hex and SRF form complexes in 10T1/2 cells.

    Figure 6. Hex directly binds to SRF. Immunoprecipitation assay of 10T1/2 cells. 10T1/2 cells were transfected with combination of SRF expression vector (15 μg) and Hex expression vector (15 μg). Immunoprecipitation was performed using normal rabbit IgG, anti-SRF,and anti-HA antibodies. Immunoprecipitated proteins were electrophoresed and immunostained with anti-SRF (upper) and anti-HA antibodies (lower).

    Discussion

    In the present study, we showed that Hex activates endogenous SM -actin and SM22 expression in 10T1/2 fibroblasts. In addition, we found that Hex induces SM22 promoter activity via proximal CArG element. With EMSA, using the nuclear extracts from AxCA/Hex-transduced cells and in vitro-translated Hex and SRF proteins, we demonstrated that Hex does not bind to CArG element of the SM22 promoter but rather increases the binding of SRF to this element. Furthermore, the immunoprecipitation assay showed that Hex physically interacts with SRF. Given that SRF plays a central role in the activation of the SMC marker genes, our data suggest that Hex may contribute to SMC differentiation by serving as positively acting cofactor for SRF.

    Accumulating evidence indicates that homeodomain factors, which contain distinct DNA-binding domains, achieve their transactivating action not only through DNA–protein interaction but also through protein–protein interaction. However, the fact that Hex is capable of increasing the binding activity of SRF to CArG box deserves particular attention because there are only a few precedent reports that describe similar observation. Grueneberg et al22 have shown that paired homeobox protein Phox enhances the DNA binding activity of SRF to the c-fos SRE. By EMSA, using recombinant SRF and GST-Phox proteins, they suggested that the ternary Phox-SRF-SRE complex was transient or unstable under EMSA conditions, because formation and dissociation of this complex were rapid in the presence of Phox. Likewise, Hautmann et al23 showed that Mhox, which is the murine homologue of Phox, increases SRF binding to the CArG box in SM -actin promoter. In our study, although we did not determine the formation of the ternary Hex-SRF-CArG complex in our EMSA condition, we did observe that in vitro-translated Hex increased the binding of SRF to CArG box. Thus, Hex-SRF-CArG complex may be either transient or unstable. Other factors or post-transcriptional modification of Hex and SRF do not appear to be required to stabilize this complex.

    The molecular mechanisms of how Hex increases the binding of SRF to CArG box of SM22 promoter remain unanswered. At least 3 possibilities are envisioned, which are not mutually exclusive. First, Hex could accelerate the exchange of a new SRF molecule onto the SRE. Second, Hex could act by eliciting a conformational change in SRF. Third, SRF and Hex could form stable cooperative complexes on DNA. We favor the third model because we detected the Hex/SRF complex by immunoprecipitation assays. Chen and Schwartz have described the possibility that coassociation of Nkx2.5 and SRF in the CArG box might be stabilized through a short peptide encompassing the amino terminus and helix 1, and perhaps helix 2 of Nkx2.5.24 Consistent with this view, overexpression of plasmid encoding mutated Hex, which lacks homeodomain, was not able to activate SM22 promoter (data not shown). Further studies to define the interactive regions shared by Hex and SRF are needed.

    We have previously shown that Hex expression is induced in neointima after balloon injury in rats.13 Phenotypic modulation of vascular SMC, which occurs in response to vascular injury, has long been known to be associated with the downregulation of virtually every SMC differentiation marker and upregulation of dedifferentiated SMC markers, including the SMemb/NMHC-B.14,25 Among SMC differentiation markers, SM -actin and SM22, but not calponin and smooth muscle myosin heavy chain 1 and 2 (SM1 and SM2), are expressed in neointima, although their expression levels are reduced in neointima as compared with those in media. Interestingly, our Northern blot analysis showed that Hex induces SM -actin and SM22 genes, but it has no effects on calponin and SM1/2 gene expression (data not shown). Thus, it is intriguing to speculate that Hex plays a role in inducing or maintaining the proliferating phenotype characterizing SMC in neointima. Our data suggest that Hex functions in a cell type-specific manner, because Hex overexpression does not induce SMC marker gene expression in bovine aortic endothelial cells. This result conforms to our hypothesis that Hex acts as a cofactor of SRF, which plays a crucial role in SMC gene expression. Furthermore, it is likely that Hex plays a dispensable role in maintaining the undifferentiated phenotype, because overexpression of Hex, which acts as an inhibitor of Hex, has no effects on SM -actin and SM22 gene expression in vascular SMC (data not shown). Then, what is the molecular mechanism by which Hex is capable of inducing limited set of SMC marker gene expression, even though all of the SMC marker genes contain functional CArG box within their promoters? Taking into consideration that the SM -actin and SM22 genes are induced at early stage of SMC differentiation, and that the calponin and SM1/2 genes are induced at later stage of differentiation,14 one of the most plausible explanations for differential expression of the SMC marker genes by Hex is that SRF levels may be only rate-limiting in activation of early SMC differentiation marker, and Hex is sufficient to drive transcription of these genes through increasing SRF binding activity. However, combinatorial interactions of SRF with many transcription factors such as GATA6, MEF2, and SRF cofactors may be required for the later stage of SMC differentiation markers to be expressed and, if so, Hex overexpression may not be sufficient for activating these genes. We are currently testing this hypothesis.

    In conclusion, the present study showed that the SMC marker genes were induced by Hex through the interaction between Hex and SRF on CArG box. Regulatory role of Hex in SRF-dependent genes through protein–protein interaction supports the concept that function of SRF is modulated by a variety of SRF cofactors, including myocardin26 and C-reactive protein.27 Further investigation of the molecular basis for enhancement of the binding of SRF to CArG box by Hex will provide the clue for the understanding of the context-dependent and signal-responsive regulation of phenotypic modulation of vascular SMC.

    Acknowledgments

    This work was supported in part by a grant-in-aid for scientific research from the Ministry of Education, Science, Sport, and Culture of Japan and by a grant from the Japan Cardiovascular Foundation (to R.N. and M.K.).

    References

    Wright CV, Cho KW, Oliver G, De Robertis EM. Vertebrate homeodomain proteins: families of region-specific transcription factors. Trends Biochem Sci. 1989; 14: 52–56.

    Harvey RP. NK-2 homeobox genes and heart development. Dev Biol. 1996; 178: 203–216.

    Newman CS, Krieg PA. tinman-related genes expressed during heart development in Xenopus. Dev Genet. 1998; 22: 230–238.

    Komuro I, Izumo S. Csx: a murine homeobox-containing gene specifically expressed in the developing heart. Proc Natl Acad Sci U S A. 1993; 90: 8145–8149.

    Lints TJ, Parsons LM, Hartley L, Lyons I, Harvey RP. Nkx-2.5: a novel murine homeobox gene expressed in early heart progenitor cells and their myogenic descendants. Development. 1993; 119: 419–431.

    Chen CY, Croissant J, Majesky M, Topouzis S, McQuinn T, Frankovsky MJ, Schwartz RJ. Activation of the cardiac -actin promoter depends upon serum response factor, Tinman homologue, Nkx-2.5, and intact serum response elements. Dev Genet. 1996; 19: 119–130.

    Durocher D, Chen CY, Ardati A, Schwartz RJ, Nemer M. The atrial natriuretic factor promoter is a downstream target for Nkx-2.5 in the myocardium. Mol Cell Biol. 1996; 16: 4648–4655.

    Lyons I, Parsons LM, Hartley L, Li R, Andrews JE, Robb L, Harvey RP. Myogenic and morphogenetic defects in the heart tubes of murine embryos lacking the homeo box gene Nkx2–5. Genes Dev. 1995; 9: 1654–1666.

    Biben C, Harvey RP. Homeodomain factor Nkx2–5 controls left/right asymmetric expression of bHLH gene eHand during murine heart development. Genes Dev. 1997; 11: 1357–1369.

    Manfioletti G, Gattei V, Buratti E, Rustighi A, De Iuliis A, Aldinucci D, Goodwin GH, Pinto A. Differential expression of a novel proline-rich homeobox gene (Prh) in human hematolymphopoietic cells. Blood. 1995; 85: 1237–1245.

    Thomas PQ, Brown A, Beddington RS. Hex: a homeobox gene revealing peri-implantation asymmetry in the mouse embryo and an early transient marker of endothelial cell precursors. Development. 1998; 125: 85–94.

    Newman CS, Chia F, Krieg PA. The XHex homeobox gene is expressed during development of the vascular endothelium: overexpression leads to an increase in vascular endothelial cell number. Mech Dev. 1997; 66: 83–93.

    Sekiguchi K, Kurabayashi M, Oyama Y, Aihara Y, Tanaka T, Sakamoto H, Hoshino Y, Kanda T, Yokoyama T, Shimomura Y, Iijima H, Ohyama Y, Nagai R. Homeobox protein Hex induces SMemb/nonmuscle myosin heavy chain-B gene expression through the cAMP-responsive element. Circ Res. 2001; 88: 52–58.

    Owens GK. Regulation of differentiation of vascular smooth muscle cells. Physiol Rev. 1995; 75: 487–517.

    Adam PJ, Regan CP, Hautmann MB, Owens GK. Positive- and negative-acting Kruppel-like transcription factors bind a transforming growth factor ? control element required for expression of the smooth muscle cell differentiation marker SM22 in vivo. J Biol Chem. 2000; 275: 37798–37806.

    Travo P, Barrett G, Burnstock G. Differences in proliferation of primary cultures of vascular smooth muscle cells taken from male and female rats. Blood Vessels. 1980; 17: 110–116.

    Miyake S, Makimura M, Kanegae Y, Harada S, Sato Y, Takamori K, Tokuda C, Saito I. Efficient generation of recombinant adenoviruses using adenovirus DNA-terminal protein complex and a cosmid bearing the full-length virus genome. Proc Natl Acad Sci U S A. 1996; 93: 1320–1324.

    Watanabe N, Kurabayashi M, Shimomura Y, Kawai-Kowase K, Hoshino Y, Manabe I, Watanabe M, Aikawa M, Kuro-o M, Suzuki T, Yazaki Y, Nagai R. BTEB2, a Kruppel-like transcription factor, regulates expression of the SMemb/Nonmuscle myosin heavy chain B (SMemb/NMHC-B) gene. Circ Res. 1999; 85: 182–191.

    Miwa T, Kedes L. Duplicated CArG box domains have positive and mutually dependent regulatory roles in expression of the human -cardiac actin gene. Mol Cell Biol. 1987; 7: 2803–2813.

    Dignam JD, Lebovitz RM, Roeder RG. Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids Res. 1983; 11: 1475–1489.

    Kawai-Kowase K, Kurabayashi M, Hoshino Y, Ohyama Y, Nagai R. Transcriptional activation of the zinc finger transcription factor BTEB2 gene by Egr-1 through mitogen-activated protein kinase pathways in vascular smooth muscle cells. Circ Res. 1999; 85: 787–795.

    Grueneberg DA, Natesan S, Alexandre C, Gilman MZ. Human and Drosophila homeodomain proteins that enhance the DNA-binding activity of serum response factor. Science. 1992; 257: 1089–1095.

    Hautmann MB, Thompson MM, Swartz EA, Olson EN, Owens GK. Angiotensin II-induced stimulation of smooth muscle -actin expression by serum response factor and the homeodomain transcription factor MHox. Circ Res. 1997; 81: 600–610.

    Chen CY, Schwartz RJ. Recruitment of the tinman homolog Nkx-2.5 by serum response factor activates cardiac -actin gene transcription. Mol Cell Biol. 1996; 16: 6372–6384.

    Walsh K, Takahashi A. Transcriptional regulation of vascular smooth muscle cell phenotype. Z Kardiol. 2001; 90: 12–16.

    Wang D, Chang PS, Wang Z, Sutherland L, Richardson JA, Small E, Krieg PA, Olson EN. Activation of cardiac gene expression by myocardin, a transcriptional cofactor for serum response factor. Cell. 2001; 105: 851–862.

    Henderson JR, Macalma T, Brown D, Richardson JA, Olson EN, Beckerle MC. The LIM protein, CRP1, is a smooth muscle marker. Dev Dyn. 1999; 214: 229–238.(Yuko Oyama; Keiko Kawai-K)