Mitogen- and stress-activated kinase 1 (MSK1) belongs to a family of dual protein kinases that are activated by either extracellular signal-regulated kinase or p38 mitogen-activated protein kinases in response to stress or mitogenic extracellular stimuli. The physiologic role of MSK1 in malignant transformation and cancer development is not well understood. Here, we report that MSK1 is involved in 12-O-tetradecanoylphorbol-13-acetate (TPA)–induced or epidermal growth factor (EGF)–induced neoplastic transformation of JB6 Cl41 cells. H89, a potent inhibitor of MSK1, strongly suppressed TPA-induced or EGF-induced cell transformation. When cells overexpressing wild-type MSK1 were treated with TPA or EGF, colony formation increased substantially compared with untreated cells or cells that did not overexpress MSK1. In contrast, MSK1 COOH terminal or NH2 terminal dead dominant negative mutants dramatically suppressed cell transformation. Introduction of small interfering RNA-MSK1 into JB6 Cl41 cells resulted in suppressed TPA-induced or EGF-induced cell transformation. In addition, cell proliferation was inhibited in MSK1 knockdown cells compared with MSK1 wild-type cells. In wild-type MSK1-overexpressing cells, activator protein (AP-1) activation increased after TPA or EGF stimulation, whereas AP-1 activation decreased in both MSK1 dominant-negative mutants and in MSK1 knockdown cells. Moreover, TPA-induced or EGF-induced phosphorylation of histone H3 at Ser10 was increased in wild-type cells but the induced phosphorylation was abolished in MSK1 dominant-negative mutant or MSK1 knockdown cells. Thus, MSK1 is required for tumor promoter-induced cell transformation through its phosphorylation of histone H3 at Ser10 and AP-1 activation. [Cancer Res 2008;68(7):2538–47]

The mitogen-activated protein kinase (MAPK) cascades are required for many cellular functions, including growth, proliferation, differentiation, inflammation, apoptosis, and malignant transformation (13). The extracellular signal-regulated kinases 1/2 (ERK1/2) and p38 MAPK signaling pathways are among the best characterized of these intracellular protein kinase cascades (4). Mitogen-activated and stress-activated kinase 1 (MSK1) is a serine/threonine protein kinase that is activated by either ERKs or p38 MAPKs in response to stress or mitogenic extracellular stimuli. MSK1 is involved in the transcriptional activation of genes, including the inflammatory gene interleukin-6 and immediate early response genes, such as c-fos, c-jun, MAPK phosphatase-1 (mkp-1), and nurr1 (5, 6). MSK1 has also been reported to phosphorylate chromatin proteins histone H3 and high mobility group 14 (HMG-14; ref. 7). Further, MSK1 phosphorylates various transcription factors including cyclic AMP (cAMP)-response element-binding protein (cAMP-responsive element binding protein (CREB)), activating transcription factor 1 (ATF1) and the p65 subunit of nuclear factor-κB (NF-κB; refs. 810). Recently, MSK1 was shown to play a positive role in the control of cell proliferation of HaCaT keratinocytes (11) and might play a role in pathogenesis of psoriasis by regulating the production of proinflammatory cytokines in psoriatic epidermis (12). MSK1 was suggested to be activated in response to tumor promoters, such as epidermal growth factor (EGF), 12-O-tetradecanoylphorbol-13-acetate (TPA), or UVB (7, 13, 14). However, the role of MSK1 in tumorigenesis or cell transformation is not completely clear.

Here, we used MSK1 dominant-negative or small interfering RNA (siRNA)–MSK1 knockdown cells to show that MSK1 is involved in TPA-induced or EGF-induced cell transformation of JB6 Cl41 cells. Results indicated that blocking MSK1 suppressed anchorage-independent transformation of JB6 cells through the inhibition of both phosphorylation of histone H3 and AP-1 activation. Thus, we showed that MSK1 is required for tumor promoter-induced cell transformation.

Reagents and antibodies. Chemical reagents, including Eagle's MEM, l-glutamine, and gentamicin, were purchased from Invitrogen. TPA and EGF were purchased from Sigma-Aldrich and EMD Biosciences, Inc., respectively. Fetal bovine serum (FBS) was from Atlanta Biologicals and the polyvinylidene difluoride (PVDF) membrane was from Millipore. The DNA ligation kit (version 2.0) was purchased from Takara Bio, Inc. Antibodies against phosphorylated or total MSK1 were purchased from Cell Signaling Technology, Inc. and Santa Cruz Biotechnology, Inc., respectively. Antibodies to detect histone H3 or phosphorylated histone H3 were purchased from Upstate Biotechnology.

Cell lines and cell cultures. JB6 Cl41 mouse epidermal skin cells and stably transfected JB6 cells were grown in 5% FBS/MEM supplemented with penicillin/streptomycin (100 units/mL; Invitrogen) at 37°C in a humidified 5% CO2 incubator. The pCMV5-Flag vector, pCMV5-Flag-wild-type MSK1, pCMV-Flag-MSK1-A195/NH2 terminal kinase dead, and pCMV-Flag-MSK1-A565/COOH terminal kinase dead (generous gifts from Dr. D.R. Alessi, Medical Research Council Protein Phosphorylation Unit, Department of Biochemistry, University of Dundee; ref. 8) were each transfected into JB6 Cl41 cells. Stable cell lines were generated and characterized according to previous methods (13).

Construction of siRNA MSK1 and establishing stable cell lines. To construct the siRNA-MSK1 (si-MSK1), the pU6pro vector (a gift kindly provided by Dr. David L. Turner, Mental Health Research Institute, University of Michigan; si-MOCK) was digested with XbaI and BbsI. The annealed synthetic primers (primer 1, 5′-TTTGAGACCTACTTCAGCGTCTCTTCAAGAGAGAGACGCTGAAGTAGGTCTTTTTT-3′; primer 2, 5′-CTAGAAAAAAGACCTACTTCAGCGTCTCTCTCTTGAAGAGACGCTGAAGTAGGTCT-3′) were then introduced following the recommended protocols found online.3

The recombinant plasmid of psi-MSK1 was confirmed by agarose gel electrophoresis and DNA sequencing (GENEWIZ, Inc.). Stable JB6 Cl41 cells expressing pU6pro (si-MOCK) or si-MSK1 were established with pcDNA3.1 as the selection marker. Transfected cells were selected in medium containing 400 μg/mL G418, and the expression level of the MSK1 protein was confirmed by Western blot analysis.

3-(4,5-Dimethylthiazol-2-yl)-5-(3-carboxymethoxy-phenyl)-2-(4-sulfonyl)-2H-tetrazolium assay. To estimate cell proliferation, stable psi-MSK1 JB6 Cl41 cells (1 × 103) were seeded into 96-well plates in 100 μL of 5% FBS/MEM and incubated at 37°C in a 5% CO2 incubator. After culturing for various periods of time, 20 μL of the CellTiter 96 Aqueous One Solution (Promega) were added to each well and cells were then incubated for 1 h at 37°C in a 5% CO2 atmosphere. To stop the reaction, 25 μL of a 10% SDS solution were added, and absorbance was measured at 492 and 690 nm.

Anchorage-independent cell transformation assay. TPA-induced or EGF-induced cell transformation was investigated in JB6 Cl41 and MSK1 dominant-negative cells and psi-MSK1 stably transfected cells. In brief, cells (8 × 103) were exposed to TPA (20 ng/mL) or EGF (10 ng/mL) in 1 mL of 0.3% Basal Medium Eagle (BME) agar containing 10% FBS. The cultures were maintained at 37°C in 5% CO2 incubator for 10 d (EGF) or 12 d (TPA), and cell colonies were scored using a microscope and the Image-Pro PLUS (v.4) computer software program (Media Cybernetics).

Cell cycle analysis. psi-MOCK or psi-MSK1 stably transfected JB6 Cl41 cells (2 × 105) were seeded into 60-mm dishes and cultured for 24 h at 37°C in a 5% CO2 incubator. The cells were harvested with trypsin, fixed with ice-cold methanol, stained with propidium iodide (PI), and then analyzed for cell cycle phase by flow cytometry.

Reporter gene assays. For reporter gene assays, transient transfections were carried out using jetPEI (Qbiogene) and cells were cotransfected in triplicate with 250 ng of the AP-1 reporter gene and 50 ng of a β-galactosidase (β-Gal)–expressing plasmid. The β-Gal–expressing plasmid was cotransfected for normalizing the transfection efficiency. Routinely, 36 h after transfection, cells were washed and then starved 24 h in 0.1% FBS/MEM, and cell lysates were then prepared 12 h after stimulation with TPA (20 ng/mL) or EGF (10 ng/mL). Luciferase and β-Gal activities were measured using the Luminoskan Ascent (Thermo Electron) and Multiskan MS (Labsystems), respectively.

Isolation of histone proteins. For the isolation of histone proteins, cells (6 × 106) were homogenized in 0.5 mL nuclear preparation buffer [10 mmol/L Tris-HCl (pH 7.6), 150 mmol/L NaCl, 1.5 mmol/L MgCl2, 0.65% NP40, 1 mmol/L phenylmethylsulfonyl fluoride (PMSF)] in the presence of protein phosphatase inhibitors (10 mmol/L NaF, 1 mmol/L sodium orthovanadate, 25 mmol/L β-glycerophosphate). Nuclei were recovered by centrifugation at 1,500 × g for 15 min. All centrifugations were carried out at 4°C. Nuclei were resuspended in 0.3 mL resuspension buffer [10 mmol/L Tris-HCl (pH 7.6), 3 mmol/L MgCl2, 10 mmol/L NaCl, 1 mmol/L PMSF, protein phosphatase inhibitors]. Nuclei were extracted with 0.4 N H2SO4 to isolate total histone proteins. The samples were precipitated with trichloroacetic acid and then resuspended in double-distilled H2O.

In vitro kinase assay. The histone H3 protein was used for an in vitro kinase assay with active MSK1 (Upstate Biotechnology, Inc). Reactions were carried out at 30°C for 30 min in a mixture containing 50 μmol/L unlabeled ATP and then stopped by adding 6× SDS sample buffer. Samples were boiled and then separated by 15% SDS-PAGE and visualized by Western blotting.

Western blot analysis. Samples containing equal amounts of protein were resolved by SDS-PAGE and transferred onto PVDF membranes. The membranes were incubated in blocking buffer and hybridized with the appropriate primary antibodies to detect phosphorylated MSK1, total MSK1, phosphorylated histone H3 (Ser10 or Ser28), total histone H3, or β-actin. The Western blots were visualized using an enhanced chemiluminescence detection system (Amersham Biosciences Corp.) after hybridization with the appropriate horseradish peroxidase (HRP)–conjugated secondary antibody.

TPA or EGF induces MSK1 activation and cell transformation. TPA and EGF are well-known tumor promotion agents used to study malignant transformation in cells and carcinogenesis in animal models (15). By using the JB6 mouse epidermal cell system, we found that TPA or EGF was very effective in dose-dependently promoting the transformation of JB6 Cl41 cells (Fig. 1A and B). Results also indicated that TPA or EGF caused phosphorylation of MSK1 (Fig. 1C and D) at the same dose, as was effective for inducing cell transformation. These results suggested that MSK1 phosphorylation might be involved in TPA-induced or EGF-induced cell transformation.

Figure 1.

TPA or EGF induces cell transformation and MSK1 phosphorylation in JB6 Cl41 mouse epidermal skin cells. A and B, JB6 Cl41 cells were assessed for cell transformation in an anchorage-independent soft agar assay in the presence of TPA (A) or EGF (B). Cells (8 × 103) were exposed to TPA (0, 0.5, 5, or 20 ng/mL) or EGF (0, 0.1, 1, or 10 ng/mL) in 1 mL of 0.3% BME agar containing 10% FBS. The cultures were maintained at 37°C in a 5% CO2 atmosphere for 10 to 12 d, and then colonies were counted using a microscope and the Image-Pro PLUS (v.4) computer software program. The average colony number was calculated (right); plates were photographed (left) from three separate experiments (A and B) and representative plates are shown. Significant differences were evaluated using the Student's t test, and the respective asterisks indicate a significant increase in TPA-induced or EGF-induced cell transformation (*, P < 0.001; **, P < 0.0001) compared with untreated control. C and D, JB6 Cl41 cells were used to determine the protein levels of endogenous phosphorylated MSK1 in TPA-induced or EGF-induced cell transformation. Cells (2 × 105) were starved in 0.1% FBS-MEM for 24 h at 37°C in a 5% CO2 atmosphere and then stimulated with 20 ng/mL of TPA (C) or 10 ng/mL of EGF (D) and finally harvested at various time periods. Cells were disrupted and proteins extracted with NP40 cell lysis buffer and Western blotting was conducted as described in the Materials and Methods using specific antibodies as indicated. β-Actin was used to confirm equal protein loading.

Figure 1.

TPA or EGF induces cell transformation and MSK1 phosphorylation in JB6 Cl41 mouse epidermal skin cells. A and B, JB6 Cl41 cells were assessed for cell transformation in an anchorage-independent soft agar assay in the presence of TPA (A) or EGF (B). Cells (8 × 103) were exposed to TPA (0, 0.5, 5, or 20 ng/mL) or EGF (0, 0.1, 1, or 10 ng/mL) in 1 mL of 0.3% BME agar containing 10% FBS. The cultures were maintained at 37°C in a 5% CO2 atmosphere for 10 to 12 d, and then colonies were counted using a microscope and the Image-Pro PLUS (v.4) computer software program. The average colony number was calculated (right); plates were photographed (left) from three separate experiments (A and B) and representative plates are shown. Significant differences were evaluated using the Student's t test, and the respective asterisks indicate a significant increase in TPA-induced or EGF-induced cell transformation (*, P < 0.001; **, P < 0.0001) compared with untreated control. C and D, JB6 Cl41 cells were used to determine the protein levels of endogenous phosphorylated MSK1 in TPA-induced or EGF-induced cell transformation. Cells (2 × 105) were starved in 0.1% FBS-MEM for 24 h at 37°C in a 5% CO2 atmosphere and then stimulated with 20 ng/mL of TPA (C) or 10 ng/mL of EGF (D) and finally harvested at various time periods. Cells were disrupted and proteins extracted with NP40 cell lysis buffer and Western blotting was conducted as described in the Materials and Methods using specific antibodies as indicated. β-Actin was used to confirm equal protein loading.

Close modal

H89, a potent inhibitor of MSK1, suppresses TPA-induced or EGF-induced AP-1 activation and cell transformation. H89 {N-[2-(p-bromocinnamylamino)-ethyl]-5-isoquinolinesulfonaime} has been shown to have a potent and selective inhibitory action against cyclic AMP-dependent protein kinase (protein kinase A; ref. 16). H89 also was reported to strongly inhibit MSK1 activity in vitro or in vivo (17, 18). To test whether H89 could affect cell transformation, JB6 Cl41 cells were incubated with TPA (20 ng/mL) or EGF (10 ng/mL) and various concentrations of H89. Our results showed that H89 dramatically suppressed TPA-induced or EGF-induced cell transformation in JB6 Cl41 cells (Fig. 2A and B), which supports a role for MSK1.

Figure 2.

H89, a potent inhibitor of MSK1, strongly inhibits colony formation and AP-1 activation induced by TPA or EGF. A and B, H89 suppresses TPA-induced or EGF-induced anchorage-independent colony formation in soft agar. Cells (8 × 103) were exposed to TPA (20 ng/mL, A) or EGF (10 ng/mL, B) and different doses of H89 in 1 mL of 0.3% BME agar containing 10% FBS. The cultures were maintained at 37°C in a 5% CO2 atmosphere for 10 to 12 d, and then colonies were counted using a microscope and the Image-Pro PLUS (v.4) computer software program. The average colony number was calculated (right) and plates were photographed (left) from three separate experiments (A and B) and representative plates are shown. Significant differences were evaluated using the Student's t test, and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced cell transformation by H89 compared with cells treated with only TPA or EGF (*, P < 0.005; **, P < 0.0001). C, JB6 Cl41 cells were cotransfected with a plasmid mixture containing the AP-1-luciferase reporter gene (0.25 μg) and with the pCMV-β-Gal gene (0.05 μg) for normalization. At 36 h after transfection, cells were starved for 24 h by incubating in serum-deprived MEM at 37°C in a 5% CO2 atmosphere and then pretreated for 30 min with different doses of H89, followed by stimulation with 20 ng/mL of TPA (left) or 10 ng/mL of EGF (right) for an additional 12 h and then harvested. D, JB6 Cl41 cells were cotransfected with a plasmid mixture containing the c-fos-luciferase reporter gene (0.1 μg) and the pCMV-β-Gal gene (0.05 μg) for normalization purposes. At 36 h after transfection, cells were starved for 24 h by incubating in serum-deprived MEM at 37°C in a 5% CO2 atmosphere and then pretreated for 30 min with different doses of H89 followed by stimulation with 20 ng/mL of TPA (left) or 10 ng/mL of EGF (right) followed by harvesting after 12 h. The firefly luciferase activity was determined in cell lysates and normalized against β-gal activity. Significant differences were evaluated using the Student's t test and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced AP-1 activation by H89 compared with cells treated with only TPA or EGF (*, P < 0.01; **, P < 0.005; ***, P < 0.0001).

Figure 2.

H89, a potent inhibitor of MSK1, strongly inhibits colony formation and AP-1 activation induced by TPA or EGF. A and B, H89 suppresses TPA-induced or EGF-induced anchorage-independent colony formation in soft agar. Cells (8 × 103) were exposed to TPA (20 ng/mL, A) or EGF (10 ng/mL, B) and different doses of H89 in 1 mL of 0.3% BME agar containing 10% FBS. The cultures were maintained at 37°C in a 5% CO2 atmosphere for 10 to 12 d, and then colonies were counted using a microscope and the Image-Pro PLUS (v.4) computer software program. The average colony number was calculated (right) and plates were photographed (left) from three separate experiments (A and B) and representative plates are shown. Significant differences were evaluated using the Student's t test, and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced cell transformation by H89 compared with cells treated with only TPA or EGF (*, P < 0.005; **, P < 0.0001). C, JB6 Cl41 cells were cotransfected with a plasmid mixture containing the AP-1-luciferase reporter gene (0.25 μg) and with the pCMV-β-Gal gene (0.05 μg) for normalization. At 36 h after transfection, cells were starved for 24 h by incubating in serum-deprived MEM at 37°C in a 5% CO2 atmosphere and then pretreated for 30 min with different doses of H89, followed by stimulation with 20 ng/mL of TPA (left) or 10 ng/mL of EGF (right) for an additional 12 h and then harvested. D, JB6 Cl41 cells were cotransfected with a plasmid mixture containing the c-fos-luciferase reporter gene (0.1 μg) and the pCMV-β-Gal gene (0.05 μg) for normalization purposes. At 36 h after transfection, cells were starved for 24 h by incubating in serum-deprived MEM at 37°C in a 5% CO2 atmosphere and then pretreated for 30 min with different doses of H89 followed by stimulation with 20 ng/mL of TPA (left) or 10 ng/mL of EGF (right) followed by harvesting after 12 h. The firefly luciferase activity was determined in cell lysates and normalized against β-gal activity. Significant differences were evaluated using the Student's t test and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced AP-1 activation by H89 compared with cells treated with only TPA or EGF (*, P < 0.01; **, P < 0.005; ***, P < 0.0001).

Close modal

The AP-1 transcription factor is a dimeric complex that comprises members of the Jun, Fos, ATF1 (activating transcription factor 1), and musculoaponeurotic fibrosarcoma protein families (19). The regulation of cell proliferation by AP-1 is believed to be of crucial importance for the multistage development of tumors (20, 21). To test whether the inhibition of MSK1 by H89 might contribute to the AP-1 activation response to TPA or EGF, we cotransfected the AP-1 luciferase reporter plasmid and the pCMV-β-Gal gene into JB6 Cl41 cells. Our data showed that H89 effectively suppressed TPA-promoted (Fig. 2C,, left) or EGF-promoted (Fig. 2C,, right) AP-1 activation in JB6 cells. We also found that TPA-induced (Fig. 2D,, left) or EGF-induced (Fig. 2D , right) c-fos promoter activity in JB6 Cl41 cells was decreased by treatment with H89. These data indicated that H89, a potent inhibitor of MSK1, is very effective in preventing TPA-induced or EGF-induced cell transformation and AP-1 transactivation activity.

A dominant-negative mutant of MSK1 suppresses TPA-induced or EGF-induced cell transformation and AP-1 activation. The COOH terminal kinase domain of MSK1 is essential for activation of its NH2 terminal domain and an inactivation mutation either in the NH2 terminal or COOH terminal kinase domain completely abolishes MSK1 activation (8). To further investigate whether MSK1 is directly responsible for TPA-induced or EGF-induced cell transformation, we used JB6 cells transfected with plasmids containing a pCMV5-FLAG vector (pCMV-neo), pCMV5-FLAG-wild-type MSK1 (pCMV-MSK1 WT), pCMV5-FLAG-MSK1-A565 COOH terminal kinase-dead (pCMV-MSK1 CD), or pCMV-FLAG-MSK1-A195 NH2 terminal kinase-dead (pCMV-MSK1 ND; ref. 13). These cells were treated with TPA (20 ng/mL) or EGF (10 ng/mL) in soft agar and incubated at 37°C in a 5% CO2 incubator for 10 to 12 days, at which time colony numbers were determined. Our results showed a marked increase in colony numbers, as well as colony sizes in cells overexpressing wild-type MSK1 (pCMV-MSK1 WT) promoted by TPA or EGF compared with control cells (pCMV-neo; Fig. 3A and B). On the other hand, the overexpression of COOH terminal dead MSK1 caused a dramatic suppression of colony formation induced by TPA or EGF (Fig. 3A and B). Overexpression of the MSK1 NH2 terminal dead dominant-negative mutant resulted in a decrease in the number of TPA-induced colonies (Fig. 3A), but seemed to exert a lesser effect on EGF-induced colony numbers (Fig. 3B). These results strongly suggested that MSK1 mediates cell transformation stimulated by tumor promotion agents, such as TPA or EGF, and the COOH terminal kinase domain of MSK1 plays an important role.

Figure 3.

MSK1 dominant-negative mutants suppress cell transformation and AP-1 activation promoted by TPA or EGF. A and B, cells (8 × 103) transfected with pCMV-neo, pCMV-MSK1 WT, pCMV-MSK1 CD, or pCMV-MSK1 ND were subjected to a soft agar assay in the presence of TPA (20 ng/mL, A) or EGF (10 ng/mL, B) in 1 mL of 0.3% BME agar containing 10% FBS. The cultures were maintained at 37°C in a 5% CO2 atmosphere for 10 to 12 d, and then colonies were counted using a microscope and the Image-Pro PLUS (v.4) computer software program. The average colony number was calculated (right) and photographed (left) from three separate experiments (A and B, right). Representative plates are shown. Significant differences were evaluated using the Student's t test, and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced cell transformation compared with cells treated with only TPA or EGF (*, P < 0.005; **, P < 0.0005; ***, P < 0.0001). C and D, JB6 Cl41 cells were transfected with a plasmid mixture containing the AP-1-luciferase reporter gene (0.5 μg) or with the pCMV-β-Gal gene (0.05 μg) for normalization. At 36 h after transfection, cells were starved for 24 h by incubating in serum-deprived MEM at 37°C in a 5% CO2 atmosphere and then incubated with 20 ng/mL of TPA (C) or 10 ng/mL of EGF (D) for 12 h. The firefly luciferase activity was determined in cell lysates and normalized against β-gal activity. Significant differences were evaluated using the Student's t test, and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced AP-1 activation compared with cells treated with only TPA or EGF (*, P < 0.005; **, P < 0.0005; ***, P < 0.0001).

Figure 3.

MSK1 dominant-negative mutants suppress cell transformation and AP-1 activation promoted by TPA or EGF. A and B, cells (8 × 103) transfected with pCMV-neo, pCMV-MSK1 WT, pCMV-MSK1 CD, or pCMV-MSK1 ND were subjected to a soft agar assay in the presence of TPA (20 ng/mL, A) or EGF (10 ng/mL, B) in 1 mL of 0.3% BME agar containing 10% FBS. The cultures were maintained at 37°C in a 5% CO2 atmosphere for 10 to 12 d, and then colonies were counted using a microscope and the Image-Pro PLUS (v.4) computer software program. The average colony number was calculated (right) and photographed (left) from three separate experiments (A and B, right). Representative plates are shown. Significant differences were evaluated using the Student's t test, and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced cell transformation compared with cells treated with only TPA or EGF (*, P < 0.005; **, P < 0.0005; ***, P < 0.0001). C and D, JB6 Cl41 cells were transfected with a plasmid mixture containing the AP-1-luciferase reporter gene (0.5 μg) or with the pCMV-β-Gal gene (0.05 μg) for normalization. At 36 h after transfection, cells were starved for 24 h by incubating in serum-deprived MEM at 37°C in a 5% CO2 atmosphere and then incubated with 20 ng/mL of TPA (C) or 10 ng/mL of EGF (D) for 12 h. The firefly luciferase activity was determined in cell lysates and normalized against β-gal activity. Significant differences were evaluated using the Student's t test, and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced AP-1 activation compared with cells treated with only TPA or EGF (*, P < 0.005; **, P < 0.0005; ***, P < 0.0001).

Close modal

To determine whether TPA- or EGF-induced cell transformation elicited by the induction of AP-1 promoter activity mediated by MSK1, we cotransfected the AP-1 luciferase reporter plasmid and the pCMV-β-Gal gene into each of the MSK1 WT, COOH terminal or NH2 terminal dead dominant-negative mutants, or control JB6 Cl41 cell lines. The TPA-promoted or EGF-promoted AP-1 transcriptional activity response in JB6 Cl41 cells was much more pronounced in MSK1 WT overexpressing cells (pCMV-MSK1 WT) compared with mock vector control cells (Fig. 3C and D). In contrast, a significant inhibition of AP-1 transactivation was observed in COOH terminal and NH2 terminal kinase dead cells after treatment with TPA (Fig. 3C) or EGF (Fig. 3D). These data supported our idea that the induction of AP-1 promoter activity by TPA or EGF mediated by MSK1 might be one of the mechanisms explaining the increased anchorage-independent growth of JB6 Cl41 cells.

MSK1-mediated phosphorylation of histone H3 is important in cell transformation. Different covalent modifications, such as acetylation, phosphorylation, and methylation at different sites of the NH2 terminal tail of histone H3 seem to have various effects on cell function. Mitotic and stimulus-inducible phosphorylation of histone H3 at Ser10 and Ser28 has been characterized (2225). Furthermore, MAPK-mediated phosphorylation of Ser10 and Ser28 has also been shown to occur after stimulation with EGF (25), UVB (13, 26), TPA, or anisomycin (7). To investigate whether histone H3 phosphorylation at Ser10 or Ser28 specifically induces cell transformation promoted by TPA or EGF, we constructed stable transfectant histone H3 WT and various histone H3 mutants (S10A, S28A and S10/28A). The various cell lines were treated with TPA (20 ng/mL) or EGF (10 ng/mL) in a soft agar matrix and incubated at 37°C in a 5% CO2 incubator for 10 to 12 d. The results indicated that the number of colonies promoted by either TPA or EGF was decreased in S10A mutant cells compared with the overexpressing histone H3 WT cells (Fig. 4A and B). The cell transforming activity was also inhibited in H3 S28A mutant cells compared with overexpressing H3 WT cells but to a lesser degree than H3 S10A mutants (Fig. 4A and B). These finding indicated that the phosphorylation of histone H3 at Ser10 is very likely a critical site for mediating cell transformation induced by tumor promoters, such as TPA or EGF.

Figure 4.

MSK1-mediated phosphorylation of histone H3 at Ser10 regulates TPA-induced or EGF-induced cell transformation. A and B, cells (8 × 103) transfected with histone H3 WT or mutants, S10A, S28A, or S10/28A, were subjected to a soft agar assay in presence of TPA (20 ng/mL, A) or EGF (10 ng/mL, B) in 1 mL of 0.3% BME agar containing 10% FBS. The cultures were maintained at 37°C in a 5% CO2 atmosphere for 10 to 12 d and then colonies were counted using a microscope and the Image-Pro PLUS (v.4) computer software program. The average colony number was calculated (right) and photographed (left) from three separate experiments (A and B) and representative plates are shown. Significant differences were evaluated using the Student's t test, and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced cell transformation compared with overexpressed histone H3 WT cells (*, P < 0.005; **, P < 0.0001). C, MSK1 phosphorylates histone H3 at Ser10 or Ser28. Active MSK1 (50 ng) was combined with human histone H3 (1 μg) protein and 50 μmol/L ATP. The reaction was carried out at 30°C for 30 min and then stopped by adding 6× SDS sample buffer. The samples were resolved by 15% SDS-PAGE, and the protein bands were visualized by Western blotting with specific antibodies to detect total MSK1 or phosphorylation of histone H3 at Ser10 or Ser28. D, cells transfected with pCMV-neo, pCMV-MSK1 CD, or pCMV-MSK1 ND were analyzed to detect endogenous phosphorylated histone H3 levels induced by TPA or EGF. Cells (6 × 105/mL) were starved in 0.1% FBS-MEM for 24 h at 37°C in a 5% CO2 atmosphere and then stimulated with 20 ng/mL of TPA (top) or 10 ng/mL of EGF (bottom) for various periods of time. Histone proteins (1 μg) were resolved by SDS-15% PAGE and then transferred to membranes for Western blot analysis. The phosphorylation of histone H3 at Ser10 or Ser28 was detected with specific antibodies. Detection of total histone H3 was used for confirming equal loading of protein.

Figure 4.

MSK1-mediated phosphorylation of histone H3 at Ser10 regulates TPA-induced or EGF-induced cell transformation. A and B, cells (8 × 103) transfected with histone H3 WT or mutants, S10A, S28A, or S10/28A, were subjected to a soft agar assay in presence of TPA (20 ng/mL, A) or EGF (10 ng/mL, B) in 1 mL of 0.3% BME agar containing 10% FBS. The cultures were maintained at 37°C in a 5% CO2 atmosphere for 10 to 12 d and then colonies were counted using a microscope and the Image-Pro PLUS (v.4) computer software program. The average colony number was calculated (right) and photographed (left) from three separate experiments (A and B) and representative plates are shown. Significant differences were evaluated using the Student's t test, and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced cell transformation compared with overexpressed histone H3 WT cells (*, P < 0.005; **, P < 0.0001). C, MSK1 phosphorylates histone H3 at Ser10 or Ser28. Active MSK1 (50 ng) was combined with human histone H3 (1 μg) protein and 50 μmol/L ATP. The reaction was carried out at 30°C for 30 min and then stopped by adding 6× SDS sample buffer. The samples were resolved by 15% SDS-PAGE, and the protein bands were visualized by Western blotting with specific antibodies to detect total MSK1 or phosphorylation of histone H3 at Ser10 or Ser28. D, cells transfected with pCMV-neo, pCMV-MSK1 CD, or pCMV-MSK1 ND were analyzed to detect endogenous phosphorylated histone H3 levels induced by TPA or EGF. Cells (6 × 105/mL) were starved in 0.1% FBS-MEM for 24 h at 37°C in a 5% CO2 atmosphere and then stimulated with 20 ng/mL of TPA (top) or 10 ng/mL of EGF (bottom) for various periods of time. Histone proteins (1 μg) were resolved by SDS-15% PAGE and then transferred to membranes for Western blot analysis. The phosphorylation of histone H3 at Ser10 or Ser28 was detected with specific antibodies. Detection of total histone H3 was used for confirming equal loading of protein.

Close modal

MSK1 is a well known mitogen-responsive and stress-responsive histone H3 kinase that is activated by the Ras-MAPK pathway (7). To test whether MSK1 directly phosphorylates histone H3, active MSK1 was incubated with human histone H3 and ATP. Immunoblot results showed that MSK1 phosphorylated histone H3 at Ser10 and Ser28 (Fig. 4C). Furthermore, to investigate whether MSK1 is the histone H3 kinase responsible for TPA-induced or EGF-induced phosphorylation of histone H3, we assessed phosphorylation of histone H3 in control cells (pCMV-neo), MSK1 COOH terminal (pCMV-MSK1 CD) or NH2 terminal dead (pCMV-MSK1 ND) dominant-negative mutants after TPA or EGF stimulation. Our results indicated that TPA-induced or EGF-induced phosphorylation of histone H3 at Ser10 and Ser28 was decreased in both mutants compared with control mock cells (Fig. 4D). These results show that MSK1 directly phosphorylates histone H3 at Ser10 and Ser28 in TPA-induced or EGF-induced cell transformation.

Knockdown of MSK1 suppresses cell proliferation by causing accumulation of cells at G1. To determine whether siRNA-mediated knockdown of MSK1 in JB6 Cl41 could inhibit cell transforming activity, we designed siRNA against MSK1 (si-MSK1) and a general scrambled mock control (si-MOCK; Fig. 5A) for transfection into JB6 Cl41 cells. Immunoblot analysis of si-MSK1–transfected and si-MOCK–transfected JB6 Cl41 cells revealed a suppression of endogenous MSK1 protein expression of up to 80% compared with cells expressing the mock vector (pU6pro; Fig. 5B).

Figure 5.

Knockdown of MSK1 suppresses cell proliferation. A, DNA sequence of the si-MOCK and si-MSK1 primers. B, si-MSK1 efficiently suppresses the endogenous MSK1 protein level. JB6 Cl41 cells were cotransfected with pcDNA3.1-neo and pU6pro control or the si-MSK1 gene, selected with 400 μg/mL G418 for 12 d, and then pooled. The expression of MSK1 was analyzed in the stable transfectant JB6 Cl41 cells. The lysates were resolved by 10% SDS-PAGE, and the protein bands were visualized by Western blotting with a specific MSK1 primary antibody and an HRP-conjugated secondary antibody. Detection of total β-actin was used to verify equal loading of protein. C, cells stably transfected with the si-MOCK or si-MSK1 plasmid were seeded (1 × 103 per well) in 96-well plates in 100 μL of 5% FBS-MEM, and cell proliferation was estimated using the CellTiter 96 Aqueous One Solution detection kit (Promega). Cell proliferation was estimated by absorbance at 492 nm at 24-hour intervals up to 96 h. Points, means of triplicate experiments; bars, SD. Statistical differences were evaluated using the Student's t test, and the respective asterisks indicate a significant change in si-MSK1 cells compared with si-MOCK control cells (*, P < 0.001; **, P < 0.0001). D, si-MOCK or si-MSK1 stably expressing (2 × 105) cells were seeded into 60-mm dishes and cultured 24 h. The cells were harvested, fixed with methanol, stained with PI, and then analyzed for cell cycle phase. Data are expressed as the percentage of cells in G1-G0, S, or G2-M phase; columns, mean of values obtained from triplicate experiments; bars, SD. Statistical differences were evaluated using the Student's t test, and the respective asterisks indicate a significant change in cell cycle phase distribution in si-MSK1 cells compared with si-MOCK control cells (*, P < 0.01; **, P < 0.0001).

Figure 5.

Knockdown of MSK1 suppresses cell proliferation. A, DNA sequence of the si-MOCK and si-MSK1 primers. B, si-MSK1 efficiently suppresses the endogenous MSK1 protein level. JB6 Cl41 cells were cotransfected with pcDNA3.1-neo and pU6pro control or the si-MSK1 gene, selected with 400 μg/mL G418 for 12 d, and then pooled. The expression of MSK1 was analyzed in the stable transfectant JB6 Cl41 cells. The lysates were resolved by 10% SDS-PAGE, and the protein bands were visualized by Western blotting with a specific MSK1 primary antibody and an HRP-conjugated secondary antibody. Detection of total β-actin was used to verify equal loading of protein. C, cells stably transfected with the si-MOCK or si-MSK1 plasmid were seeded (1 × 103 per well) in 96-well plates in 100 μL of 5% FBS-MEM, and cell proliferation was estimated using the CellTiter 96 Aqueous One Solution detection kit (Promega). Cell proliferation was estimated by absorbance at 492 nm at 24-hour intervals up to 96 h. Points, means of triplicate experiments; bars, SD. Statistical differences were evaluated using the Student's t test, and the respective asterisks indicate a significant change in si-MSK1 cells compared with si-MOCK control cells (*, P < 0.001; **, P < 0.0001). D, si-MOCK or si-MSK1 stably expressing (2 × 105) cells were seeded into 60-mm dishes and cultured 24 h. The cells were harvested, fixed with methanol, stained with PI, and then analyzed for cell cycle phase. Data are expressed as the percentage of cells in G1-G0, S, or G2-M phase; columns, mean of values obtained from triplicate experiments; bars, SD. Statistical differences were evaluated using the Student's t test, and the respective asterisks indicate a significant change in cell cycle phase distribution in si-MSK1 cells compared with si-MOCK control cells (*, P < 0.01; **, P < 0.0001).

Close modal

MSK1 has been reported to play a positive role in the control of cell proliferation of both HaCaT keratinocytes and the A431 human epidermoid carcinoma line (11). To examine whether the knockdown of MSK1 could affect cell proliferation, the 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxy-phenyl)-2-(4-sulfonyl)-2H-tetrazolium assay was used to examine proliferation of cells stably transfected with si-MOCK or si-MSK1. Results indicated that si-MSK1 stably transfected cells showed a marked decrease in the rate of proliferation compared with si-MOCK stably transfected cells (Fig. 5C). In contrast, JB6 Cl41 cells overexpressing MSK1 exhibited an increased rate of proliferation (data not shown), confirming that MSK1 is involved in cell proliferation. Using these cell lines, we studied differences in cell cycle phases induced by TPA or EGF. Our data indicated that knockdown of MSK1 resulted in less cells occupying the S phase and a greater accumulation of cells in the G1-G0 phase of the cell cycle compared with si-MOCK control cell (Fig. 5D). These results clearly showed that the knockdown of MSK1 suppressed cell proliferation because of an impaired G1-S cell cycle transition.

TPA-induced or EGF-induced cell transformation is suppressed by knockdown of MSK1. To examine the effect of suppressing MSK1 expression on TPA-induced or EGF-induced cell transformation, we analyzed colony-forming activity in si-MSK1 and si-MOCK cells. These cell lines were stimulated with TPA (20 ng/mL) or EGF (10 ng/mL) in soft agar and incubated at 37°C in a 5% CO2 incubator for 10 to 12 days. Our results showed that, as expected, the knockdown of MSK1 suppressed colony formation induced by TPA (Fig. 6A,, top) or EGF (Fig. 6A , bottom). Overall, these data further showed that MSK1 plays a key role in cell transformation induced by tumor promoting agents.

Figure 6.

Knockdown of MSK1 suppresses TPA-induced or EGF-induced cell transformation and phosphorylation of histone H3. A, si-MOCK and si-MSK1 stably transfected cells (8 × 103) were exposed to TPA (20 ng/mL, top) or EGF (10 ng/mL, bottom) in 1 mL of 0.3% BME agar containing 10% FBS. The cultures were maintained at 37°C in a 5% CO2 atmosphere for 10 to 12 d, and then colonies were counted using a microscope and the Image-Pro PLUS (v.4) computer software program. The average colony number was calculated and photographed from three separate experiments (top right, TPA; bottom right, EGF) and representative plates are shown. Significant differences were evaluated using the Student's t test and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced cell transformation compared with si-MOCK cells (*, P < 0.0001). B, JB6 Cl41 cells were transiently transfected with si-MOCK or si-MSK and a plasmid mixture containing the AP-1-luciferase reporter gene (0.25 μg) or the pCMV-β-Gal gene (0.05 μg). At 36 h after transfection, cells were starved for 24 h by incubating in serum-deprived MEM at 37°C in a 5% CO2 atmosphere and then incubated with 20 ng/mL of TPA (top panel) or 10 ng/mL of EGF (bottom panel) for 12 h. C, JB6 Cl41 cells stably expressing si-MOCK or si-MSK1 were transiently transfected with a plasmid mixture containing the c-fos-luciferase reporter gene (0.1 μg) or the pCMV-β-Gal gene (0.05 μg) for normalizing. At 36 h after transfection, cells were starved for 24 h by incubating in serum-deprived MEM at 37°C in a 5% CO2 atmosphere and then incubated with 20 ng/mL of TPA (top) or 10 ng/mL of EGF (bottom) for 12 h. Significant differences were evaluated using the Student's t test, and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced AP-1 activation compared with si-MOCK cells (*, P < 0.01; **, P < 0.005). D, si-MOCK or si-MSK1 transfected cells (6 × 105/mL) were starved in 0.1% FBS-MEM for 24 h at 37°C in a 5% CO2 atmosphere and then stimulated with 20 ng/mL of TPA (left) or 10 ng/mL of EGF (right) for various periods of time. Histone proteins (1 μg) were prepared for Western blot analysis and the phosphorylation of histone H3 at Ser10 or Ser28 expression was detected with specific antibodies. Total histone H3 was used to verify equal protein loading.

Figure 6.

Knockdown of MSK1 suppresses TPA-induced or EGF-induced cell transformation and phosphorylation of histone H3. A, si-MOCK and si-MSK1 stably transfected cells (8 × 103) were exposed to TPA (20 ng/mL, top) or EGF (10 ng/mL, bottom) in 1 mL of 0.3% BME agar containing 10% FBS. The cultures were maintained at 37°C in a 5% CO2 atmosphere for 10 to 12 d, and then colonies were counted using a microscope and the Image-Pro PLUS (v.4) computer software program. The average colony number was calculated and photographed from three separate experiments (top right, TPA; bottom right, EGF) and representative plates are shown. Significant differences were evaluated using the Student's t test and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced cell transformation compared with si-MOCK cells (*, P < 0.0001). B, JB6 Cl41 cells were transiently transfected with si-MOCK or si-MSK and a plasmid mixture containing the AP-1-luciferase reporter gene (0.25 μg) or the pCMV-β-Gal gene (0.05 μg). At 36 h after transfection, cells were starved for 24 h by incubating in serum-deprived MEM at 37°C in a 5% CO2 atmosphere and then incubated with 20 ng/mL of TPA (top panel) or 10 ng/mL of EGF (bottom panel) for 12 h. C, JB6 Cl41 cells stably expressing si-MOCK or si-MSK1 were transiently transfected with a plasmid mixture containing the c-fos-luciferase reporter gene (0.1 μg) or the pCMV-β-Gal gene (0.05 μg) for normalizing. At 36 h after transfection, cells were starved for 24 h by incubating in serum-deprived MEM at 37°C in a 5% CO2 atmosphere and then incubated with 20 ng/mL of TPA (top) or 10 ng/mL of EGF (bottom) for 12 h. Significant differences were evaluated using the Student's t test, and the respective asterisks indicate a significant decrease in TPA-induced or EGF-induced AP-1 activation compared with si-MOCK cells (*, P < 0.01; **, P < 0.005). D, si-MOCK or si-MSK1 transfected cells (6 × 105/mL) were starved in 0.1% FBS-MEM for 24 h at 37°C in a 5% CO2 atmosphere and then stimulated with 20 ng/mL of TPA (left) or 10 ng/mL of EGF (right) for various periods of time. Histone proteins (1 μg) were prepared for Western blot analysis and the phosphorylation of histone H3 at Ser10 or Ser28 expression was detected with specific antibodies. Total histone H3 was used to verify equal protein loading.

Close modal

To determine whether the knockdown of MSK1 in JB6 Cl41 cells might affect the AP-1 activation response to TPA or EGF, we cotransfected the AP-1 luciferase reporter plasmid and the si-MOCK or si-MSK1 gene into JB6 Cl41 cells. Results showed that TPA-induced or EGF-induced AP-1 activation was suppressed in si-MSK1 stably expressing cells compared with mock control cells (Fig. 6B). Similar results were obtained for EGF-induced or TPA-induced c-fos promoter activity (Fig. 6C), confirming that MSK1 has a role in c-fos promoter and AP-1 activation induced by TPA or EGF.

We showed that the TPA-induced or EGF-induced phosphorylation of histone H3 at Ser10 and Ser28 was reduced in two different MSK1 dominant-negative mutant cell lines (Fig. 4D). We therefore next examined the effect of MSK1 knockdown on histone H3 phosphorylation (Ser10 or Ser28). Results confirmed that TPA-induced or EGF-induced histone H3 phosphorylation is effectively suppressed in MSK1 knockdown cells compared with control mock cells (Fig. 6D). This finding showed that MSK1 is essential for TPA-induced or EGF-induced phosphorylation of histone H3 at Ser10 and Ser28. Taken together, these results provided strong evidence showing that MKS1 is required for TPA-induced or EGF-induced cell transformation, which might be regulated through MSK1-mediated histone H3 phosphorylation at Ser10 and Ser28 and AP-1 activation.

MSK1 is a nuclear kinase that acts downstream of both the ERK and p38 MAPKs and has been suggested to be activated in response to EGF, TPA, UVB, and anisomycin (7, 8, 13, 14). MSK1 phosphorylates various transcription factors, including CREB, ATF1, ATF2, signal transducers and activators of transcription 3, and NF-κB (810, 2729). MSK1 plays a critical role in the survival of lipopolysaccharide-stimulated RAW 264.7 cells through the phosphorylation of CREB (30). MSK1 also activates the transcription factor ER81, which is involved in oncogenesis and breast tumor formation (31). MSK1 phosphorylation of these transcription factors may alter their ability to bind to their target DNA and/or recruitment of required coactivators. MSK1 has also been shown to be required for the full induction of several immediate early genes, such as c-fos, junB, and mkp-1, in response to various stimuli (5, 6). Moreover, MSK1 regulates the transcription of the Nur77, Nurr1, and Nor1 nuclear orphan receptor genes of the NR4A subfamily (32), the up-regulation of which has been implicated in cell transformation (33). MSK1 is also required for chromatin protein histone H3 and HMG-14 phosphorylation (7, 34). The activation of the Ras-MAPK pathway and MSK1 resulting in the elevation of phosphorylated H3 levels may contribute to the aberrant gene expression observed in the oncogene-transformed cells (35). However, the intriguing question is whether the activation of MSK1 directly affects tumor promoter-induced cell transformation.

Tumor promotion agents, such as TPA or EGF, can induce AP-1 activation and promote JB6 Cl41 cell transformation (20, 36). On the other hand, the inhibition of AP-1 transactivation activity suppresses TPA-induced or EGF-induced cell transformation (37). Our results showed the relationship of MSK1 activity and AP-1 transactivation activity in TPA-induced or EGF-induced cell transformation. Expression of c-Fos and c-Jun has been shown to be increased TPA-induced and UVB-induced cell transformation in JB6 cell and in mouse epidermis (38). Bcl-2–overexpressing JB6 cells enhanced TPA-induced cell transformation and also, bcl-2–transfected cells expressed significantly more c-Fos, but not c-Jun, after TPA treatment (39). Although overexpression of the Fos and Jun proteins was found to correlate with a positive effect on cell transformation (40), c-Fos might have a more important role than c-Jun during late stage tumorigenesis (41). The importance of c-Fos in tumor invasion has been supported in vivo, as the progression of chemically induced papillomas to invasive squamous cell carcinomas was shown to be impaired in c-Fos–deficient mice (42). Activated genes including c-fos and c-jun coupled with histone H3 phosphorylation at specific loci may contribute to a mechanism that allows them to be rapidly activated in response to external stimuli (25). The stimulation of the activity of the c-fos SRE by phosphorylation of histone H3 at Ser10 (25) might promote c-Fos expression and c-Fos/c-Jun heterodimer formation leading to expression of AP-1 regulated genes (43). In histone H3 mutant S10A cells, c-fos promoter activity and AP-1 activation was significantly decreased compared with control cells (44). Therefore, histone H3 phosphorylation by MSK1 seems to be closely linked with AP-1 transactivation potential. Another implication of the increased AP-1 transactivation activity in cell transformation induced by TPA or EGF is that MSK1 can regulate AP-1 activation through the phosphorylation of transcription factors, such as CREB and ATF1. MSK-mediated phosphorylation of CREB and ATF1 (6, 9) has been implicated in the induction of c-fos and junB transcription. The function of MSK1 is critical for interleukin-1–induced and CREB-mediated c-fos gene expression in keratinocytes and promotes the growth of both keratinocytes and human epidermoid carcinoma cell lines (11). Therefore, increased AP-1 activity induced by MSK1 is very much associated with TPA-induced or EGF-induced cell transformation.

In the present study, we found that MSK1 closely regulated cell transformation and cell proliferation of JB6 Cl41 cells through the phosphorylation of histone H3. Increased phosphorylation of histone H3 at Ser10 was found in mitogen-stimulated and oncogene-transformed mouse fibroblasts (45). The mutation of Ser10 of histone H3 significantly suppressed cell transformation promoted by EGF (44). Furthermore, we suggest that MSK1 can directly regulate cell transformation through the phosphorylation of histone H3 at Ser10. Histone H3 is a well-known substrate of MSK1 when induced by mitogenic or stress stimuli (35, 45). Our results indicated that MSK1 can directly phosphorylate histone H3 at Ser10in vivo and in vitro. Moreover, inhibition of MSK1 activation in dominant-negative mutants or knockdown of MSK1 expression using siRNA suppressed the phosphorylation of histone H3 at Ser10 and cell transformation induced by TPA or EGF. Other studies using MSK1 and MSK2 knockout murine embryonic fibroblasts revealed that MSK1 regulates phosphorylation of histone H3 at Ser10 and Ser28, as well as phosphorylation of HMG-14 (7, 46). Increased mitotic histone H3 Ser10 phosphorylation was also observed in various colorectal tumor cells with high AIM-1, a mammalian Ipl1/aurora kinase involved in histone H3 phosphorylation (47). These previous studies strongly support our finding that MSK1 might have an important role in carcinogenesis through its phosphorylation of histone H3 at Ser10.

Although many factors have been shown to interact with and promote cell transformation, we found that the MSK1 plays an important role in cell transformation induced by tumor promoting agents, such as TPA or EGF. MSK1 can induce cell transformation leading to phosphorylation of histone H3 and induction of AP-1 activity, which may include its transactivating potential and DNA binding capacity and the stability of AP-1 components. By linking MSK1 to histone H3 phosphorylation and AP-1 transactivation activity, these findings provide an attractive explanation of how MSK1 might be related to cell transformation induced by tumor promotion agents. Taken together, these results strongly suggested that MSK1 is required for cell transformation and the relationship of MSK1 and histone H3 might be a crucial target for cancer chemotherapy or gene therapy in the future.

Grant support: Hormel Foundation, NIH grants CA120388, CA77646, CA11536, and Minnesota Partnership for Biotechnology and Medical Genomics grant L9046001401.

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

1
Chang L, Karin M. Mammalian MAP kinase signalling cascades.
Nature
2001
;
410
:
37
–40.
2
Ballif BA, Blenis J. Molecular mechanisms mediating mammalian mitogen-activated protein kinase (MAPK) kinase (MEK)-MAPK cell survival signals.
Cell Growth Differ
2001
;
12
:
397
–408.
3
Papatsoris AG, Papavassiliou AG. Molecular ‘palpation’ of BPH: a tale of MAPK signalling?
Trends Mol Med
2001
;
7
:
288
–92.
4
Robinson MJ, Cobb MH. Mitogen-activated protein kinase pathways.
Curr Opin Cell Biol
1997
;
9
:
180
–6.
5
Schuck S, Soloaga A, Schratt G, Arthur JS, Nordheim A. The kinase MSK1 is required for induction of c-fos by lysophosphatidic acid in mouse embryonic stem cells.
BMC Mol Biol
2003
;
4
:
6
.
6
Arthur JS, Fong AL, Dwyer JM, et al. Mitogen- and stress-activated protein kinase 1 mediates cAMP response element-binding protein phosphorylation and activation by neurotrophins.
J Neurosci
2004
;
24
:
4324
–32.
7
Soloaga A, Thomson S, Wiggin GR, et al. MSK2 and MSK1 mediate the mitogen- and stress-induced phosphorylation of histone H3 and HMG-14.
EMBO J
2003
;
22
:
2788
–97.
8
Deak M, Clifton AD, Lucocq LM, Alessi DR. Mitogen- and stress-activated protein kinase-1 (MSK1) is directly activated by MAPK and SAPK2/p38, and may mediate activation of CREB.
EMBO J
1998
;
17
:
4426
–41.
9
Wiggin GR, Soloaga A, Foster JM, Murray-Tait V, Cohen P, Arthur JS. MSK1 and MSK2 are required for the mitogen- and stress-induced phosphorylation of CREB and ATF1 in fibroblasts.
Mol Cell Biol
2002
;
22
:
2871
–81.
10
Vermeulen L, De Wilde G, Van Damme P, Vanden Berghe W, Haegeman G. Transcriptional activation of the NF-κB p65 subunit by mitogen- and stress-activated protein kinase-1 (MSK1).
EMBO J
2003
;
22
:
1313
–24.
11
Schiller M, Bohm M, Dennler S, Ehrchen JM, Mauviel A. Mitogen- and stress-activated protein kinase 1 is critical for interleukin-1-induced, CREB-mediated, c-fos gene expression in keratinocytes.
Oncogene
2006
;
25
:
4449
–57.
12
Funding AT, Johansen C, Kragballe K, et al. Mitogen- and stress-activated protein kinase 1 is activated in lesional psoriatic epidermis and regulates the expression of pro-inflammatory cytokines.
J Invest Dermatol
2006
;
126
:
1784
–91.
13
Zhong S, Jansen C, She QB, et al. Ultraviolet B-induced phosphorylation of histone H3 at serine 28 is mediated by MSK1.
J Biol Chem
2001
;
276
:
33213
–9.
14
He Z, Cho YY, Liu G, Ma WY, Bode AM, Dong Z. p38 Mitogen-activated protein kinase regulation of JB6 Cl41 cell transformation promoted by epidermal growth factor.
J Biol Chem
2003
;
278
:
26435
–42.
15
Hunter T, Karin M. The regulation of transcription by phosphorylation.
Cell
1992
;
70
:
375
–87.
16
Chijiwa T, Mishima A, Hagiwara M, et al. Inhibition of forskolin-induced neurite outgrowth and protein phosphorylation by a newly synthesized selective inhibitor of cyclic AMP-dependent protein kinase, N-[2-(p-bromocinnamylamino)ethyl]-5-isoquinolinesulfonamide (H-89), of PC12D pheochromocytoma cells.
J Biol Chem
1990
;
265
:
5267
–72.
17
Davies SP, Reddy H, Caivano M, Cohen P. Specificity and mechanism of action of some commonly used protein kinase inhibitors.
Biochem J
2000
;
351
:
95
–105.
18
Thomson S, Clayton AL, Hazzalin CA, Rose S, Barratt MJ, Mahadevan LC. The nucleosomal response associated with immediate-early gene induction is mediated via alternative MAP kinase cascades: MSK1 as a potential histone H3/HMG-14 kinase.
EMBO J
1999
;
18
:
4779
–93.
19
Eferl R, Wagner EF. AP-1: a double-edged sword in tumorigenesis.
Nat Rev Cancer
2003
;
3
:
859
–68.
20
Liu Y, Ludes-Meyers J, Zhang Y, et al. Inhibition of AP-1 transcription factor causes blockade of multiple signal transduction pathways and inhibits breast cancer growth.
Oncogene
2002
;
21
:
7680
–9.
21
Park YG, Nesterova M, Agrawal S, Cho-Chung YS. Dual blockade of cyclic AMP response element- (CRE) and AP-1-directed transcription by CRE-transcription factor decoy oligonucleotide. gene-specific inhibition of tumor growth.
J Biol Chem
1999
;
274
:
1573
–80.
22
Nowak SJ, Corces VG. Phosphorylation of histone H3 correlates with transcriptionally active loci.
Genes Dev
2000
;
14
:
3003
–13.
23
Hendzel MJ, Wei Y, Mancini MA, et al. Mitosis-specific phosphorylation of histone H3 initiates primarily within pericentromeric heterochromatin during G2 and spreads in an ordered fashion coincident with mitotic chromosome condensation.
Chromosoma
1997
;
106
:
348
–60.
24
Cheung P, Allis CD, Sassone-Corsi P. Signaling to chromatin through histone modifications.
Cell
2000
;
103
:
263
–71.
25
Cheung P, Tanner KG, Cheung WL, Sassone-Corsi P, Denu JM, Allis CD. Synergistic coupling of histone H3 phosphorylation and acetylation in response to epidermal growth factor stimulation.
Mol Cell
2000
;
5
:
905
–15.
26
Zhong S, Zhang Y, Jansen C, Goto H, Inagaki M, Dong Z. MAP kinases mediate UVB-induced phosphorylation of histone H3 at serine 28.
J Biol Chem
2001
;
276
:
12932
–7.
27
Zhu F, Zhang Y, Bode AM, Dong Z. Involvement of ERKs and mitogen- and stress-activated protein kinase in UVC-induced phosphorylation of ATF2 in JB6 cells.
Carcinogenesis
2004
;
25
:
1847
–52.
28
Zhang Y, Liu G, Dong Z. MSK1 and JNKs mediate phosphorylation of STAT3 in UVA-irradiated mouse epidermal JB6 cells.
J Biol Chem
2001
;
276
:
42534
–42.
29
Wierenga AT, Vogelzang I, Eggen BJ, Vellenga E. Erythropoietin-induced serine 727 phosphorylation of STAT3 in erythroid cells is mediated by a MEK-, ERK-, and MSK1-dependent pathway.
Exp Hematol
2003
;
31
:
398
–405.
30
Mu MM, Koide N, Hassan F, et al. A role of mitogen and stress-activated protein kinase 1/2 in survival of lipopolysaccharide-stimulated RAW 264.7 macrophages.
FEMS Immunol Med Microbiol
2005
;
43
:
277
–86.
31
Janknecht R. Regulation of the ER81 transcription factor and its coactivators by mitogen- and stress-activated protein kinase 1 (MSK1).
Oncogene
2003
;
22
:
746
–55.
32
Darragh J, Soloaga A, Beardmore VA, et al. MSKs are required for the transcription of the nuclear orphan receptors Nur77, Nurr1 and Nor1 downstream of MAPK signalling.
Biochem J
2005
;
390
:
749
–59.
33
Ke N, Claassen G, Yu DH, et al. Nuclear hormone receptor NR4A2 is involved in cell transformation and apoptosis.
Cancer Res
2004
;
64
:
8208
–12.
34
Duncan EA, Anest V, Cogswell P, Baldwin AS. The kinases MSK1 and MSK2 are required for epidermal growth factor-induced, but not tumor necrosis factor-induced, histone H3 Ser10 phosphorylation.
J Biol Chem
2006
;
281
:
12521
–5.
35
Strelkov IS, Davie JR. Ser-10 phosphorylation of histone H3 and immediate early gene expression in oncogene-transformed mouse fibroblasts.
Cancer Res
2002
;
62
:
75
–8.
36
Liu G, Bibus DM, Bode AM, Ma WY, Holman RT, Dong Z. Omega 3 but not omega 6 fatty acids inhibit AP-1 activity and cell transformation in JB6 cells.
Proc Natl Acad Sci U S A
2001
;
98
:
7510
–5.
37
Dong Z, Birrer MJ, Watts RG, Matrisian LM, Colburn NH. Blocking of tumor promoter-induced AP-1 activity inhibits induced transformation in JB6 mouse epidermal cells.
Proc Natl Acad Sci U S A
1994
;
91
:
609
–13.
38
Lu YP, Chang RL, Lou YR, et al. Effect of curcumin on 12-O-tetradecanoylphorbol-13-acetate- and ultraviolet B light-induced expression of c-Jun and c-Fos in JB6 cells and in mouse epidermis.
Carcinogenesis
1994
;
15
:
2363
–70.
39
Amstad PA, Liu H, Ichimiya M, Chang S, Berezesky IK, Trump BF. bcl-2 enhancement of malignant transformation in mouse epidermal JB6 cells.
Mol Carcinog
1997
;
20
:
231
–9.
40
Clayton AL, Rose S, Barratt MJ, Mahadevan LC. Phosphoacetylation of histone H3 on c-fos- and c-jun-associated nucleosomes upon gene activation.
EMBO J
2000
;
19
:
3714
–26.
41
Reichmann E, Schwarz H, Deiner EM, et al. Activation of an inducible c-FosER fusion protein causes loss of epithelial polarity and triggers epithelial-fibroblastoid cell conversion.
Cell
1992
;
71
:
1103
–16.
42
Saez E, Rutberg SE, Mueller E, et al. c-fos is required for malignant progression of skin tumors.
Cell
1995
;
82
:
721
–32.
43
Chiu R, Boyle WJ, Meek J, Smeal T, Hunter T, Karin M. The c-Fos protein interacts with c-Jun/AP-1 to stimulate transcription of AP-1 responsive genes.
Cell
1988
;
54
:
541
–52.
44
Choi HS, Choi BY, Cho YY, et al. Phosphorylation of histone H3 at serine 10 is indispensable for neoplastic cell transformation.
Cancer Res
2005
;
65
:
5818
–27.
45
Chadee DN, Hendzel MJ, Tylipski CP, et al. Increased Ser-10 phosphorylation of histone H3 in mitogen-stimulated and oncogene-transformed mouse fibroblasts.
J Biol Chem
1999
;
274
:
24914
–20.
46
Dunn KL, Espino PS, Drobic B, He S, Davie JR. The Ras-MAPK signal transduction pathway, cancer and chromatin remodeling.
Biochem Cell Biol
2005
;
83
:
1
–14.
47
Ota T, Suto S, Katayama H, et al. Increased mitotic phosphorylation of histone H3 attributable to AIM-1/Aurora-B overexpression contributes to chromosome number instability.
Cancer Res
2002
;
62
:
5168
–77.