Skip main navigation

Conditional Deletion of Krüppel-Like Factor 4 Delays Downregulation of Smooth Muscle Cell Differentiation Markers but Accelerates Neointimal Formation Following Vascular Injury

Originally publishedhttps://doi.org/10.1161/CIRCRESAHA.108.176974Circulation Research. 2008;102:1548–1557

Abstract

Phenotypic switching of smooth muscle cells (SMCs) plays a key role in vascular proliferative diseases. We previously showed that Krüppel-like factor 4 (Klf4) suppressed SMC differentiation markers in cultured SMCs. Here, we derive mice deficient for Klf4 by conditional gene ablation and analyze their vascular phenotype following carotid injury. Klf4 expression was rapidly induced in SMCs of control mice after vascular injury but not in Klf4-deficient mice. Injury-induced repression of SMC differentiation markers was transiently delayed in Klf4-deficient mice. Klf4 mutant mice exhibited enhanced neointimal formation in response to vascular injury caused by increased cellular proliferation in the media but not an altered apoptotic rate. Consistent with these findings, cultured SMCs overexpressing Klf4 showed reduced cellular proliferation, in part, through the induction of the cell cycle inhibitor, p21WAF1/Cip1 via increased binding of Klf4 and p53 to the p21WAF1/Cip1 promoter/enhancer. In vivo chromatin immunoprecipitation assays also showed increased Klf4 binding to the promoter/enhancer regions of the p21WAF1/Cip1 gene and SMC differentiation marker genes following vascular injury. Taken together, we have demonstrated that Klf4 plays a critical role in regulating expression of SMC differentiation markers and proliferation of SMCs in vivo in response to vascular injury.

Phenotypic switching of smooth muscle cells (SMCs) plays a key role in the development of vascular diseases such as atherosclerosis and restenosis after percutaneous coronary interventions.1 Differentiated SMCs in adult vessels express a unique repertoire of contractile proteins (ie, SMC differentiation markers) and exhibit an extremely low rate of proliferation. However, in association with vascular injury or disease, SMCs modulate their phenotype and participate in the formation of neointima by markedly decreasing expression of SMC differentiation markers and increasing proliferation, migration, and synthesis of extracellular matrix proteins. Therefore, elucidation of the molecular mechanisms controlling SMC phenotypic switching is likely to provide important insights toward understanding of the development of vascular disease.

A hallmark of SMC phenotypic switching is the coordinate downregulation of expression of SMC differentiation markers including smooth muscle (SM) α-actin, SM22α, and SM–myosin heavy chain (SM-MHC).1 The promoter/enhancer regions of these SMC differentiation makers contain common cis elements, including multiple CC(A/T-rich)6GG (CArG) elements and a transforming growth factor (TGF)-β control element.1 Previously, we identified Krüppel-like factor 4 (Klf4) (formerly known as gut-enriched Krüppel-like factor or GKLF) as a binding factor for the TGF-β control element located within the SM22α promoter using a yeast 1-hybrid screen.2 Of interest, Klf4 behaved as a potent repressor of SMC differentiation markers including SM α-actin, SM22α, and SM-MHC in cultured SMCs through multiple mechanisms including: (1) direct Klf4 binding to the TGF-β control element within the promoter/enhancer regions of SMC differentiation markers; (2) interaction with serum response factor (SRF), a trans-binding factor of CArG elements; and (3) repression of expression of an SRF coactivator, myocardin.3,4 Results from our previous studies also showed that Klf4 contributed to phenotypic switching of SMCs in vitro in response to platelet-derived growth factor (PDGF)-BB and oxidized phospholipids.4,5 Indeed, PDGF-BB and oxidized phospholipids activated Klf4 expression, and small interfering (si)RNA-induced knockdown of Klf4 attenuated PDGF-BB– and oxidized phospholipid–induced downregulation of SM α-actin and/or SM-MHC expression in cultured SMCs. Moreover, expression of Klf4 mRNA was transiently induced in rat carotid arteries after balloon injury.4 The preceding results provide evidence that Klf4 is a potent transcriptional repressor of SMC differentiation markers in cultured SMCs and suggest a potential role for Klf4 in control of SMC phenotypic switching in vivo. However, as yet, no studies have directly assessed the function of Klf4 following vascular injury in vivo.

Klf4 regulates cellular proliferation in multiple cell types. Overexpression of Klf4 inhibited DNA synthesis and reduced cellular growth in colon cancer cells and fibroblasts.6–8 Klf4-induced growth suppression was caused by cell cycle arrest at the G1/S boundary, and there is evidence showing that this is the result of transcriptional activation of the cyclin-dependent kinase inhibitor p21WAF1/Cip1.9,10 In response to DNA damage, p53 and Klf4 were induced, and these proteins synergistically activated p21WAF1/Cip1 gene transcription.9 Of interest, induction of Klf4 following DNA damage was abolished in p53-null cells.9 Moreover, siRNA-induced knockdown of Klf4 attenuated DNA damage–induced expression of p21WAF1/Cip1, as well as the accumulation of cells at the G1 phase.11 These results indicate that Klf4 is essential in mediating p53-induced G1/S cell cycle arrest as a consequence of DNA damage. Consistent with these findings, Klf4 expression is significantly decreased in multiple human cancers including colon cancer and gastric cancer.8,12 In addition, loss of Klf4 in mouse gastric epithelia resulted in increased proliferation and altered differentiation of the gastric epithelia.12 Results of recent studies also demonstrated that Klf4 induces p21WAF1/Cip1 expression and reduces cellular growth in cultured SMCs.13 Taken together, these studies provide compelling evidence that Klf4 inhibits cellular proliferation in multiple cell types. However, elevated Klf4 levels have been reported in mammary carcinoma and squamous cell carcinoma of the oropharynx.14,15 Indeed, Rowland et al16 found that Klf4 had a potential to switch from a growth-inhibiting tumor suppressor to a growth-promoting oncogene in response to changes in the cellular context. As such, the functions of Klf4 in vivo are poorly defined.

Klf4-null mice are born at the expected Mendelian ratio but die shortly after birth because of loss of skin barrier function,17 precluding the analysis of Klf4 function in adult animals. To overcome this limitation, we have derived conditional Klf4-deficient mice by breeding tamoxifen-inducible Cre recombinase–expressing (ERT-Cre) mice18 with mice carrying a loxP allele of Klf4 (Klf4loxP mice)19 and analyzed their phenotype in response to vascular injury. We show that Klf4 deficiency results in a delay in suppression of SMC differentiation markers and enhanced neointimal formation following vascular injury. We analyze the molecular mechanisms of Klf4 action and demonstrate that Klf4 induces p21WAF/Cip1 expression through direct binding of Klf4 and enhanced binding of p53 to the promoter/enhancer regions of the p21WAF/Cip1 gene in cultured SMCs, as well as in injured carotid arteries in vivo.

Materials and Methods

Animal protocols were approved by the University of Virginia Animal Care and Use Committee. ERT-Cre mice18 and Klf4loxP mice19 were bred to generate tamoxifen-inducible Klf4-deficient mice.

An expanded Materials and Methods section is available in the online data supplement at http://circres.ahajournals.org.

Results

Klf4 Expression Was Induced in the Medial Layer of Vessels After Vascular Injury

We derived tamoxifen-inducible Klf4-deficient mice by breeding ERT-Cre mice with Klf4loxP mice. Seven- to 8-week-old ERT-Cre+/Klf4loxP/loxP mice were injected with 4-hydroxytamoxifen (4-OHT) 10 times IP over a 14-day period to activate Cre recombinase (Figure 1A), because 1 or 3 times of 4-OHT injection did not elicit sufficient recombination of the Klf4loxP allele (data not shown). ERT-Cre/Klf4loxP/loxP mice were injected with 4-OHT and used as controls. After 4-OHT injection, a highly efficient recombination of the Klf4loxP allele was observed in the aorta of ERT-Cre+/Klf4loxP/loxP (Klf4 knockout [KO]) mice, whereas no recombination was seen in ERT-Cre/Klf4loxP/loxP (control) mice (Figure 1B). The Klf4 gene consists of 4 exons, and exon 1 only encodes a first methionine codon. Recombination of the Klf4loxP allele deletes exons 2 and 3 and causes a frameshift mutation in exon 4, which abolishes Klf4 function completely.19 In support of this, expression of Klf4 protein and Klf4 mRNA was markedly reduced in the lung and the colon of Klf4 KO mice, respectively (Figure 1C and 1D). However, there were no differences in visible appearance (data not shown) and body weight (23.0±1.3 g in control mice versus 22.2±1.2 g in Klf4 KO mice, n=10, P=0.65) between Klf4 KO and control mice after 4-OHT injection, which is in contrast to defects in skin barrier function observed in conventional Klf4-null mice, which die shortly after birth.17 We determined changes in Klf4 expression patterns in response to carotid ligation injury in wild-type mice. Klf4 mRNA expression was rapidly induced at day 1 after injury, whereas SM α-actin mRNA expression was decreased at day 3 and day 7 (Figure 1E). Injury-induced changes in Klf4 protein levels were also examined by immunohistochemistry. In agreement with previous studies,2,20 Klf4 was expressed in endothelial cells but not in SMCs of uninjured vessels in control mice (Figure 1F). Three days after injury, Klf4 expression was induced in the medial SMC layer of carotid arteries of control mice. In contrast, Klf4 induction was undetectable in Klf4 KO mice. These results clearly indicate that Klf4 is induced in the SMC layer of carotid arteries of control mice following vascular injury and that tamoxifen-inducible Klf4-deficient mice are suitable model to test the effects of Klf4 deletion on the vascular phenotype after injury.

Figure 1. Induction of Klf4 expression following vascular injury was abolished in conditional Klf4-deficient mice. A, Experimental protocol showing the timing of 4-OHT injection, ligation injury, and harvesting tissues. B, After completion of 4-OHT injection IP 10 times, recombination of the Klf4loxP allele was examined by Southern blotting in the aorta of ERT-Cre+/Klf4loxP/loxP (Klf4 KO) mice and ERT-Cre/Klf4loxP/loxP (control) mice. C, Expression of Klf4 and GAPDH protein was determined by Western blotting in the lung of Klf4 KO and control mice. *P<0.05 compared with control (n=4). D, Expression of Klf4 mRNA was determined by real-time RT-PCR in the colon of Klf4 KO and control mice. *P<0.05 compared with control (n=5). E, Injury-induced changes in expression of SM α-actin and Klf4 were determined by real-time RT-PCR in the carotid arteries of wild-type mice. The ratios of the right injured artery to the left uninjured artery are shown (n=3). *P<0.05 compared with Klf4 expression at day 0, #P<0.05 compared with SM α-actin expression at day 0. F, Klf4 expression was examined by immunohistochemistry in the carotid arteries of Klf4 KO and control mice at day 3 after injury. Representative pictures are shown from 4 mice analyzed per each genotype. Bar: 20 μm.

Klf4 Ablation Caused a Delay in Repression of SMC Differentiation Markers After Vascular Injury

We examined whether deletion of the Klf4 gene affects downregulation of SMC differentiation markers following vascular injury. As shown in Figure 2A and Figure I in the online data supplement, expression of both SM α-actin and SM22α was markedly decreased in the medial layer of carotid arteries in control mice at day 3 and day 7 after ligation injury. In Klf4 KO mice, expression of these SMC differentiation markers was decreased at day 7 after injury. However, repression of both SM α-actin and SM22α was attenuated in carotid arteries of Klf4 KO mice as compared with control mice at day 3 following injury. The results indicate that ablation of the Klf4 gene causes a transient delay in repression of SMC differentiation markers in response to vascular injury.

Figure 2. Injury-induced downregulation of SMC differentiation markers was delayed in conditional Klf4-deficient mice. A, Expression of SM α-actin and SM22α was examined by immunohistochemistry in the carotid arteries in Klf4 KO and control mice at day 3 and day 7 after injury. Representative pictures are shown from 4 to 6 mice analyzed per each genotype and each time point. Bar: 50 μm. B, In vivo ChIP assays were performed to determine the binding of Klf4 with the TGF-β control element–containing regions of the SM α-actin promoter and the SM22α promoter, as well as the first intronic region of the SM22α gene, in carotid arteries of wild-type mice at day 3 and day 7 after injury. Un indicates uninjured; Inj, injured. *P<0.05 compared with uninjured arteries (n=2).

Binding of Klf4 to the promoter regions of SMC differentiation marker genes was examined in injured carotid arteries of wild-type mice. Of major significance, in vivo chromatin immunoprecipitation (ChIP) assays showed that Klf4 bound to the TGF-β control element-containing promoter regions of the SM α-actin gene and the SM22α gene in carotid arteries at day 3 after injury, whereas no Klf4 binding was detected in uninjured arteries (Figure 2B). Consistent with the kinetics of Klf4 expression in response to carotid ligation injury (Figure 1E and 1F), Klf4 binding was not seen in carotid arteries at day 7 after injury. Association of Klf4 with the intronic 1 region of the SM22α gene, which contains no Klf4 binding sites, was undetectable in both injured and uninjured carotid arteries. These results provide evidence that Klf4 is rapidly induced in SMCs of carotid arteries following vascular injury and that it represses expression of SMC differentiation markers by binding to the promoter regions of these genes at early time points following injury.

Conditional Klf4 Mutant Mice Exhibited Enhanced Neointimal Formation Following Vascular Injury

Next, we performed morphometric analyses in carotid arteries of Klf4 KO and control mice. Of major interest, Klf4 KO mice exhibited enhanced formation of neointima as compared with control mice (Figure 3). Neointimal area was significantly larger in Klf4 KO mice than control mice by 2.8-fold at day 7, 3.0-fold at day 14, and 3.1-fold at day 21 after injury (Figure 3C). Medial areas of injured vessels were increased as compared with uninjured vessels in both Klf4 KO and control mice but not significantly different from one another although Klf4 KO mice exhibited a modest increase (Figure 3B). Medial areas of uninjured carotid arteries were not different between Klf4 KO mice and control mice at any time point. These results indicate that, in addition to regulating SMC differentiation marker expression following vascular injury, Klf4 also plays a key role in inhibiting neointimal formation.

Figure 3. Neointimal formation was accelerated in conditional Klf4-deficient mice following vascular injury. Carotid arteries were harvested from Klf4 KO and control mice at days 7, 14, and 21 after injury. Six mice were analyzed per each genotype and each time point. A, Verhoeff–van Gieson staining was performed in cross-sections from the injured and uninjured carotid arteries. Bar: 100 μm. B and C, Areas of the media (B) and the intima (C) were determined using ImagePro software. Ct indicates control; KO, Klf4 KO; Un, uninjured; Inj, injured. *P<0.05 compared with uninjured arteries, #P<0.05 compared with the arteries in corresponding control mice.

Klf4 Reduced the Proliferation Rate of SMCs Without Affecting Apoptosis

To determine the mechanisms of enhanced neointimal formation in Klf4 KO mice, proliferation and apoptotic rates in the media and the intima were examined by 5-bromodeoxyuridine (BrdUrd) and TUNEL staining, respectively. BrdUrd staining revealed that medial cells in injured carotid arteries of Klf4 KO mice exhibited enhanced proliferation as compared with control mice (15.3±2.5% versus 9.7±1.9% at day 7, 13.9±0.8% versus 5.4±2.1% at day 14, 4.7±1.0% versus 1.6±0.4% at day 21), although the fraction of BrdUrd-positive cells in the intima was not different between Klf4 KO and control mice (Figure 4A and 4C). The apoptotic rate in the media and the intima was not different between Klf4 KO mice and control mice, although it was increased in injured vessels as compared with uninjured vessels in both groups (Figure 4B and 4D). We also tested whether Klf4 ablation alters the accumulation of macrophages and T lymphocytes in carotid arteries after vascular injury by Mac2 and CD3-ε immunostaining, respectively (supplemental Figure II). Although previous studies showed the induction of Klf4 expression in cultured macrophages in response to inflammatory cytokines,21 accumulation of macrophages as well as T lymphocytes was not different between Klf4 KO and control mice. These results suggest that enhanced neointimal formation in Klf4 KO mice following vascular injury is caused by increased proliferation of medial cells, rather than an altered apoptotic rate. This is likely to be caused, at least in part, by enhanced SMC proliferation, because SMCs are a major cell type in the media and the recruitment of monocytes/macrophages and T lymphocytes to the site of vascular injury was unaltered in Klf4 KO mice.

Figure 4. Enhanced neointimal formation in conditional Klf4-deficient mice was caused by increased cellular proliferation in the media. BrdUrd staining (A and C) and TUNEL staining (B and D) were performed in the carotid arteries of Klf4 KO and control mice at days 3, 7, 14, and 21 after injury. A and B, Representative pictures of BrdUrd staining at day 7 (A) and TUNEL staining at day 3 (B) are shown. Bars: 50 μm. C and D, The ratios of BrdUrd-positive cells (C) and TUNEL-positive cells (D) in the media and the intima were calculated (n=4 to 6). Ct indicates control; KO, Klf4 KO; Un, uninjured; Inj, injured. *P<0.05 compared with uninjured arteries, #P<0.05 compared with the arteries in corresponding control mice.

To determine the role of Klf4 in regulating SMC growth, SMCs were isolated from thoracic aorta of Klf4loxP/loxP mice and infected with adenovirus-expressing Cre recombinase or empty adenovirus to generate Klf4-null SMCs and control SMCs, respectively. Recombination of the Klf4loxP allele in cultured Klf4-null SMCs was confirmed by PCR (Figure 5A). Cellular growth was assessed by cell counting over a time course of 96 hours. We observed no differences in cell number between Klf4-null and control SMCs (Figure 5B and 5C), indicating that basal expression of Klf4 is low as seen in medial SMCs in vivo (Figure 1F) and does not contribute to SMC growth under these culture conditions. However, adenovirus-mediated overexpression of Klf4 markedly reduced cell number in both Klf4-null and control SMCs, demonstrating its potent growth repressing properties for SMCs. Consistent with these results, BrdUrd-incorporation assays showed that adenovirus-mediated expression of Klf4 reduced the proliferation rate of cultured SMCs (Figure 5D). Overexpression of Klf4 did not elicit any morphological changes indicative of apoptosis in nuclei of SMCs (Figure 5E) and did not induce expression of apoptosis markers including cleaved caspase 3, Bax, and Puma (Figure 5F). Taken together, results provide evidence that induction of Klf4 expression suppresses cellular proliferation in SMCs.

Figure 5. Klf4 reduced SMC proliferation. A, SMCs were isolated from the thoracic aorta of Klf4loxP/loxP mice. Cells were infected with adenovirus-expressing Cre recombinase or empty adenovirus and designated as Klf4-null SMCs and control SMCs, respectively. Recombination of the Klf4loxP alleles was confirmed by PCR. B and C, Cell numbers were counted in control SMCs (B) and Klf4-null SMCs (C). Cells were uninfected or infected with adenovirus expressing Klf4 (Ad/Klf4) or control (Ad/CMV) 1 day after plating (arrows). *P<0.05 compared with uninfected cells (n=4). D, BrdUrd-incorporation assays were performed in mouse SMCs infected with adenovirus expressing Klf4 (Ad/Klf4) or control (Ad/CMV). *P<0.05 compared with Ad/CMV-infected SMCs (n=3). E, Left, Mouse SMCs were transfected with FLAG-tagged Klf4 expression plasmid and stained with anti-FLAG antibody. Right, Magnified image of DAPI nuclear staining from the surrounded region in the left image. F, Mouse SMCs were uninfected or infected with adenovirus expressing Klf4 (Ad/Klf4) or control (Ad/CMV), and expression of caspase 3, Bax, Puma, and GAPDH was examined by Western blotting (caspase 3: 35 kDa; cleaved caspase 3: 17 and 19 kDa).

Klf4 Induced p21WAF1/Cip1 Expression in Concert With p53

Results of previous studies in multiple cell culture systems have shown that Klf4 is a mediator of p53-induced activation of p21WAF1/Cip1 gene transcription in response to DNA damage and that p21WAF1/Cip1 is a key factor leading to cell cycle arrest at the G1/S transition.9–11 We tested whether p21WAF1/Cip1 contributes to Klf4-induced suppression of SMC proliferation. First, injury-induced changes in expression of p21WAF1/Cip1 were examined in the medial SMC layer of carotid arteries in Klf4 KO and control mice. Expression of p21WAF1/Cip1 was increased at day 3 and day 7 after injury in control mice but not in Klf4 KO mice (Figure 6A). Second, results in cultured mouse SMCs showed that adenovirus-mediated overexpression of Klf4 induced p21WAF1/Cip1 expression, whereas p53 expression was unaltered (Figure 6B). Third, adenovirus-mediated overexpression of Klf4 did not reduce cellular proliferation in p21WAF1/Cip1 KO mouse SMCs (Figure 6C). These results suggest that Klf4-induced suppression of SMC proliferation is mediated in part by activation of p21WAF1/Cip1.

Figure 6. Klf4-induced growth suppression of SMCs was dependent on p21WAF1/Cip1. A, p21WAF1/Cip1 staining was performed in the carotid arteries of Klf4 KO and control mice at days 3, 7, 14, and 21 after injury. Representative pictures at day 7 are shown. The ratio of p21WAF1/Cip1-positive cells in the media was calculated (n=4 to 6). Ct indicates control; KO, Klf4 KO; Un, uninjured; Inj, injured. Bar: 50 μm. *P<0.05 compared with uninjured arteries, #P<0.05 compared with the arteries in corresponding control mice. B, Expression of Klf4, p53, p21WAF1/Cip1, and GAPDH was examined by Western blotting in cultured mouse SMCs that were uninfected or infected with adenovirus expressing Klf4 (Ad/Klf4) or empty virus (Ad/CMV). *P<0.05 compared with control (n=3). C, Cell numbers were counted in cultured p21WAF1/Cip1 KO SMCs (n=4). Cells were uninfected or infected with adenovirus expressing Klf4 (Ad/Klf4) or control (Ad/CMV) 1 day after plating (arrows).

Previous studies showed that Klf4 activates p21WAF1/Cip1 gene transcription through the proximal Klf4 binding site in HEK293 cells and that p53 induced p21WAF1/Cip1 gene transcription via the p53 binding site and the proximal Klf4 binding site.9,22 The consensus Klf4 binding site is 5′-(G/A)(G/A)GG(C/T)G(C/T)-3′, whereas the consensus element for p53 is 2 copies of a 10-bp motif, 5′-(G/A)(G/A)(G/A)C(A/T)(A/T)G(C/T)(C/T)(C/T)-3′, separated by multiple nucleotides. Inspection of the mouse p21WAF1/Cip1 promoter/enhancer sequence revealed a novel Klf4 consensus binding site at −2611/−2605 bp, as well as the proximal Klf4 binding site (−176/−170 bp) and the p53 binding site (−2869/−2838 bp). We tested the involvement of these cis elements for Klf4-induced activation of p21WAF1/Cip1 gene transcription in cultured aortic SMCs. Klf4 induced transcriptional activity of the p21WAF1/Cip1 promoter/enhancer–luciferase construct by 2.2-fold, but the induction was reduced by mutation of either 1 of these cis elements (Figure 7A). Simultaneous mutation of all 3 of these cis elements completely abolished the response. Moreover, results of siRNA-induced knockdown experiments showed that suppression of p53 blocked Klf4-induced p21WAF1/Cip1 activation in rat aortic SMCs (Figure 7B and supplemental Figure III). These results suggest that, in addition to 2 Klf4 binding sites, p53 and its binding site are required for Klf4-induced activation of the p21WAF1/Cip1 gene.

Figure 7. Klf4 induced p21WAF1/Cip1 expression in concert with p53. A, Klf4 expression plasmid was cotransfected into rat aortic SMCs with the wild-type p21WAF1/Cip1 promoter/enhancer–luciferase construct or its mutants, and luciferase activity was measured (n=3). B, Rat aortic SMCs were transfected with siRNA duplex for p53 or EGFP and infected with adenovirus expressing Klf4 (Ad/Klf4) or empty adenovirus (Ad/CMV). Expression of Klf4, p53, p21WAF1/Cip1, and GAPDH was examined by Western blotting. *P<0.05 compared with control (n=3). C, Association of Klf4 and p53 with the proximal and the distal regions of the p21WAF1/Cip1 promoter/enhancer was determined by ChIP assays in mouse cultured SMCs infected with adenovirus expressing Klf4 (Ad/Klf4) or empty adenovirus (Ad/CMV). *P<0.05 compared with Ad/CMV-infected SMCs (n=3). D, Association of Klf4 and p53 with the proximal and the distal regions of the p21WAF1/Cip1 promoter/enhancer was determined by in vivo ChIP assays in carotid arteries of wild-type mice at day 3 after injury. Un indicates uninjured; Inj, injured. *P<0.05 compared with uninjured arteries (n=2).

ChIP assays were carried out to determine whether Klf4 alters the association of Klf4 and p53 with the p21WAF1/Cip1 promoter/enhancer in SMCs. Adenovirus-mediated expression of Klf4 induced the binding of Klf4 to the promoter/enhancer regions of the p21WAF1/Cip1 gene that contain the proximal as well as the distal Klf4 binding sites in cultured mouse SMCs (Figure 7C). Klf4 overexpression did not induce Klf4 binding to the c-fos promoter in which the consensus Klf4 binding site is absent (data not shown). Of interest, Klf4 induced the binding of p53 to the p53 binding site within the p21WAF1/Cip1 promoter/enhancer. Finally, association of Klf4 and p53 with the p21WAF1/Cip1 promoter/enhancer was examined in injured carotid arteries of wild-type mice by in vivo ChIP assays. Three days after injury, at which time Klf4 expression was induced in the SMC layer (Figure 1E and 1F), binding of both Klf4 and p53 to the p21WAF1/Cip1 promoter/enhancer was increased within injured carotid arteries (Figure 7D). Taken together, these results provide novel evidence that Klf4 induces p21WAF1/Cip1 gene transcription through Klf4 binding to the proximal and the distal Klf4 binding sites, as well as the recruitment of p53 to the p53 binding site within the p21WAF1/Cip1 promoter/enhancer.

Discussion

In the present studies, we found that injury-induced suppression of SMC differentiation markers is delayed, but neointimal formation accelerated in the carotid arteries of tamoxifen-inducible conditional Klf4-deficient mice in vivo. We also showed that enhanced neointimal formation in conditional Klf4-deficient mice was caused by increased cellular proliferation in the media rather than an altered apoptotic rate. In addition, we demonstrated that Klf4 represses SMC proliferation by inducing p21WAF1/Cip1 gene transcription through enhanced binding of Klf4 and p53 to the p21WAF1/Cip1 promoter/enhancer. As such, results of the present studies provide the first in vivo example, to our knowledge, showing that deletion of a single transcription factor alone is sufficient to delay injury-induced downregulation of SMC differentiation markers and that a single transcription factor can function as a SMC growth repressor, as well as a repressor of SMC differentiation.

Although we discovered that injury-induced downregulation of SMC differentiation markers was delayed in SMCs and injury-induced cellular proliferation was accelerated in medial SMCs in tamoxifen-inducible conditional Klf4-deficient mice, it is possible that loss of Klf4 in other cell-types may also contribute to the phenotype observed in these mice. For example, although we showed that accumulation of macrophages and T lymphocytes was unaltered in Klf4 KO mice, we cannot rule out the possibility that loss of Klf4 may alter the function of these cells and thereby alter SMC phenotypic switching, growth, and lesion formation. Indeed, Feinberg et al21 reported that Klf4 was induced in cultured macrophages in response to interferon-γ, lipopolysaccharide, and tumor necrosis factor-α and that Klf4 partially mediated effects of these factors on the induction of inducible NO synthase by binding to the promoter/enhancer region of this gene. In addition, Hamik et al20 showed that Klf4 was expressed in vascular endothelial cells in vivo and that Klf4 induced expression of antiinflammatory and antithrombotic factors including endothelial NO synthase and thrombomodulin, whereas knockdown of Klf4 led to enhancement of tumor necrosis factor-α–induced expression of vascular cell adhesion molecule-1 and tissue factor in cultured endothelial cells. Based on the results of these studies, enhanced neointimal formation in conditional Klf4-deficient mice may be caused by the combinatorial effects of multiple cell types including SMCs, macrophages, and endothelial cells. However, Klf4 function in macrophages was proinflammatory, and thus its loss in macrophages would be expected to reduce inflammation and neointimal formation, neither of which was observed in our conditional Klf4-deficient mice. The loss of antiinflammatory functions of Klf4 in endothelial cells could contribute to enhanced neointimal formation. However, of critical importance, results of in vivo ChIP assays in injured carotid arteries showed enhanced Klf4 binding to the promoter regions of SMC differentiation marker genes, as well as the p21WAF1/Cip1 gene, thus strongly suggesting that Klf4 directly regulates expression of differentiation markers and p21WAF1/Cip1 in SMCs. However, analysis of cell type–specific KO of the Klf4 gene will be required to clearly define the cell autonomous functions of Klf4 in vascular lesion development.

We have discovered that Klf4 functions as a suppressor of SMC differentiation as well as an inhibitor of SMC proliferation in response to vascular injury. These results are highly intriguing in that they provide the first example of a single transcriptional regulator that can function both as a SMC growth repressor and a repressor of SMC differentiation, a marked contrast to the belief that cellular proliferation is inversely related to cellular differentiation. Indeed, there are now several examples that differentiation and proliferation are not necessarily mutually exclusive processes in SMCs. First, during late embryogenesis and postnatal development, SMCs exhibit an extremely high rate of proliferation, yet at the same time, they undergo the most rapid rate of induction of expression of SMC differentiation markers in the rat and chicken aorta.1,23 Second, PDGF-BB–induced suppression of SMC differentiation markers has been shown to be independent of its mitogenic effect.24 Indeed, daily pulsing treatment with PDGF-BB in postconfluent culture of SMCs caused a sustained decrease in expression of SMC differentiation markers in spite of the absence of sustained mitosis. Third, phenotypically modulated SMCs within advanced human atherosclerotic lesions typically exhibit very low rates of proliferation.25 Taken together, these results support a model wherein SMC growth and differentiation are not mutually exclusive.

Results of the present studies showed that Klf4 induced p21WAF1/Cip1 gene expression through a novel distal Klf4 binding site located within close proximity of the p53 binding site, in addition to the proximal Klf4 binding site. We also found that Klf4-induced p21WAF1/Cip1 expression was mediated, in part, by increased p53 binding to the p53 binding site. The increased p53 binding to the p21WAF1/Cip1 promoter/enhancer was caused by the recruitment of p53, because Klf4 did not induce p53 expression. These results are supported by previous studies demonstrating a physical interaction between the zinc finger region of Klf4 and the N-terminal domain of p53 by coimmunoprecipitation assays.9 Our model is that vascular injury induces expression of Klf4, which counteracts against the mitogenic stimuli of SMCs by inducing p21WAF1/Cip1 expression in concert with p53. However, in conditional Klf4-deficient mice, neointimal formation is accelerated because of the lack of p21WAF1/Cip1 induction. Consistent with this model, overexpression of p21WAF1/Cip1 inhibited neointimal formation following vascular injury, whereas p21WAF1/Cip1 ablation enhanced neointimal formation in the carotid injury model.26 Moreover, overexpression of p53 decreased neointimal formation following carotid injury, whereas loss of p53 accelerated formation of neointima in a vein bypass graft model, a femoral injury model, and in ApoE KO mice.26 However, results of recent studies by Wassmann et al13 differed from ours regarding the mechanisms whereby Klf4 suppresses SMC proliferation, because those authors showed that Klf4 induced p53 expression in cultured SMCs. Although the reasons for this discrepancy are unknown, we found no differences in the apoptotic rate between conditional Klf4 mutant mice and control mice, suggesting the selective recruitment of p53 to the p21WAF1/Cip1 promoter/enhancer rather than a simple induction of p53 expression. Recently, p53 has been shown to be subjected to multiple types of posttranslational modifications, including acetylation, methylation, phosphorylation, and ubiquitylation.27 It will be interesting to determine whether Klf4 regulates posttranslational modifications of p53 that selectively enhance its binding to the p21WAF1/Cip1 promoter/enhancer.

In summary, we provide evidence that Klf4 plays a critical role in regulation of SMC phenotypic switching and neointimal formation in response to vascular injury. Indeed, Klf4 is an early suppressor of SMC differentiation markers and an inhibitor of SMC proliferation by inducing p21WAF1/Cip1 expression in concert with p53. Further studies are needed to determine the role of Klf4 in the development of vascular diseases, including atherosclerosis in experimental animal models, as well as in humans.

Original received December 27, 2007; resubmission received April 3, 2008; accepted May 1, 2008.

Sources of Funding

This study was supported by NIH grants P01HL19242, R01HL38854, and R37HL57353 (to G.K.O.) and R01DK053839 (to K.H.K.) and by American Heart Association National Scientist Development Grant 0635253N (to T.Y.).

Disclosures

None.

Footnotes

Correspondence to Tadashi Yoshida, MD, PhD, Department of Molecular Physiology and Biological Physics, University of Virginia, MR5 Room 1226, 415 Lane Rd, Charlottesville, VA 22908. E-mail

References

  • 1 Owens GK, Kumar MS, Wamhoff BR. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev. 2004; 84: 767–801.CrossrefMedlineGoogle Scholar
  • 2 Adam PJ, Regan CP, Hautmann MB, Owens GK. Positive- and negative-acting Krüppel-like transcription factors bind a transforming growth factor β control element required for expression of the smooth muscle cell differentiation marker SM22α in vivo. J Biol Chem. 2000; 275: 37798–37806.CrossrefMedlineGoogle Scholar
  • 3 Liu Y, Sinha S, Owens G. A transforming growth factor-β control element required for SM α-actin expression in vivo also partially mediates GKLF-dependent transcriptional repression. J Biol Chem. 2003; 278: 48004–48011.CrossrefMedlineGoogle Scholar
  • 4 Liu Y, Sinha S, McDonald OG, Shang Y, Hoofnagle MH, Owens GK. Kruppel-like factor 4 abrogates myocardin-induced activation of smooth muscle gene expression. J Biol Chem. 2005; 280: 9719–9727.CrossrefMedlineGoogle Scholar
  • 5 Pidkovka NA, Cherepanova OA, Yoshida T, Alexander MR, Deaton RA, Thomas JA, Leitinger N, Owens GK. Oxidized phospholipids induce phenotypic switching of vascular smooth muscle cells in vivo and in vitro. Circ Res. 2007; 101: 792–801.LinkGoogle Scholar
  • 6 Shields JM, Christy RJ, Yang VW. Identification and characterization of a gene encoding a gut-enriched Krüppel-like factor expressed during growth arrest. J Biol Chem. 1996; 271: 20009–20017.CrossrefMedlineGoogle Scholar
  • 7 Dang DT, Chen X, Feng J, Torbenson M, Dang LH, Yang VW. Overexpression of Krüppel-like factor 4 in the human colon cancer cell line RKO leads to reduced tumorigenecity. Oncogene. 2003; 22: 3424–3430.CrossrefMedlineGoogle Scholar
  • 8 Wei D, Gong W, Kanai M, Schlunk C, Wang L, Yao JC, Wu T-T, Huang S, Xie K. Drastic down-regulation of Krüppel-like factor 4 expression is critical in human gastric cancer development and progression. Cancer Res. 2005; 65: 2746–2754.CrossrefMedlineGoogle Scholar
  • 9 Zhang W, Geiman DE, Shields JM, Dang DT, Mahatan CS, Kaestner KH, Biggs JR, Kraft AS, Yang VW. The gut-enriched Krüppel-like factor (Krüppel-like factor 4) mediates the transactivating effect of p53 on the p21WAF1/Cip1 promoter. J Biol Chem. 2000; 275: 18391–18398.CrossrefMedlineGoogle Scholar
  • 10 Chen X, Johns DC, Geiman DE, Marban E, Dang DT, Hamlin G, Sun R, Yang VW. Krüppel-like factor 4 (gut-enriched Krüppel-like factor) inhibits cell proliferation by blocking G1/S progression of the cell cycle. J Biol Chem. 2001; 276: 30423–30428.CrossrefMedlineGoogle Scholar
  • 11 Yoon HS, Chen X, Yang VW. Krüppel-like factor 4 mediates p53-dependent G1/S cell cycle arrest in response to DNA damage. J Biol Chem. 2003; 278: 2101–2105.CrossrefMedlineGoogle Scholar
  • 12 Katz JP, Perreault N, Goldstein BG, Actman L, McNally SR, Silberg DG, Furth EE, Kaestner KH. Loss of Klf4 in mice causes altered proliferation and differentiation and precancerous changes in the adult stomach. Gastroenterology. 2005; 128: 935–945.CrossrefMedlineGoogle Scholar
  • 13 Wassmann S, Wassmann K, Jung A, Velten M, Knuefermann P, Petoumenos V, Becher U, Werner C, Mueller C, Nickenig G. Induction of p53 by GKLF is essential for inhibition of proliferation of vascular smooth muscle cells. J Mol Cell Cardiol. 2007; 43: 301–307.CrossrefMedlineGoogle Scholar
  • 14 Foster KW, Ren S, Louro ID, Lobo-Ruppert SM, McKie-Bell P, Grizzle W, Hayes MR, Broker TR, Chow LT, Ruppert JM. Oncogene expression cloning by retroviral transduction of adenovirus E1A-immortalized rat kidney RK3E cells: transformation of a host with epithelial features by c-MYC and the zinc finger protein GKLF. Cell Growth Diff. 1999; 10: 423–434.MedlineGoogle Scholar
  • 15 Foster KW, Frost AR, McKie-Bell P, Lin C-Y, Engler JA, Grizzle WE, Ruppert JM. Increase of GKLF messenger RNA and protein expression during progression of breast cancer. Cancer Res. 2000; 60: 6488–6495.MedlineGoogle Scholar
  • 16 Rowland BD, Bernards R, Peeper DS. The KLF4 tumour suppressor is a transcriptional repressor of p53 that acts as a context-dependent oncogene. Nat Cell Biol. 2005; 7: 1074–1082.CrossrefMedlineGoogle Scholar
  • 17 Segre JA, Bauer C, Fuchs E. Klf4 is a transcription factor required for establishing the barrier function of the skin. Nat Genet. 1999; 22: 356–360.CrossrefMedlineGoogle Scholar
  • 18 Vooijs M, Jonkers J, Berns A. A highly efficient ligand-regulated Cre recombinase mouse line shows that LoxP recombination is position dependent. EMBO Rep. 2001; 2: 292–297.CrossrefMedlineGoogle Scholar
  • 19 Katz JP, Perreault N, Goldstein BG, Lee CS, Labosky PA, Yang VW, Kaestner KH. The zinc-finger transcription factor Klf4 is required for terminal differentiation of goblet cells in the colon. Development. 2002; 129: 2619–2628.CrossrefMedlineGoogle Scholar
  • 20 Hamik A, Lin Z, Kumar A, Balcells M, Sinha S, Katz J, Feinberg MW, Gerszten RE, Edelman ER, Jain MK. Kruppel-like factor 4 regulates endothelial inflammation. J Biol Chem. 2007; 282: 13769–13779.CrossrefMedlineGoogle Scholar
  • 21 Feinberg MW, Cao Z, Wara AK, Lebedeva MA, SenBanerjee S, Jain MK. Kruppel-like factor 4 is a mediator of proinflammatory signaling in macrophages. J Biol Chem. 2005; 280: 38247–38258.CrossrefMedlineGoogle Scholar
  • 22 El-Deiry WS, Tokino T, Velculescu VE, Levy DB, Parsons R, Trent JM, Lin D, Mercer WE, Kinzler KW, Vogelstein B. WAF1, a potential mediator of p53 tumor suppression. Cell. 1993; 75: 817–825.CrossrefMedlineGoogle Scholar
  • 23 Lee SH, Hungerford JE, Little CD, Iruela-Arispe ML. Proliferation and differentiation of smooth muscle cell precursors occurs simultaneously during the development of the vessel wall. Dev Dyn. 1997; 209: 342–352.CrossrefMedlineGoogle Scholar
  • 24 Blank RS, Owens GK. Platelet-derived growth factor regulates actin isoform expression and growth state in cultured rat aortic smooth muscle cells. J Cell Physiol. 1990; 142: 635–642.CrossrefMedlineGoogle Scholar
  • 25 Gordon D, Reidy MA, Benditt EP, Schwartz SM. Cell proliferation in human coronary arteries. Proc Natl Acad Sci U S A. 1990; 87: 4600–4604.CrossrefMedlineGoogle Scholar
  • 26 Andrés V. Unexpected proatherogenic properties of p21: beyond cell cycle control? Circulation. 2004; 110: 3749–3752.LinkGoogle Scholar
  • 27 Toledo F, Wahl GM. Regulating the p53 pathway: in vitro hypotheses, in vivo veritas. Nat Rev Cancer. 2006; 6: 909–923.CrossrefMedlineGoogle Scholar

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.