Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2006 Oct 27.
Published in final edited form as: FEBS Lett. 2005 Aug 29;579(21):4757–4762. doi: 10.1016/j.febslet.2005.07.053

Krüppel-like factor 5 promotes mitosis by activating the cyclin B1/Cdc2 complex during oncogenic Ras-mediated transformation

Mandayam O Nandan a, Sengthong Chanchevalap a, W Brian Dalton a, Vincent W Yang a,b,*
PMCID: PMC1626271  NIHMSID: NIHMS12891  PMID: 16102754

Abstract

We previously showed that the zinc finger-containing transcription factor Krüppel-like factor 5 (KLF5) is important in mediating transformation by oncogenic H-Ras through induction of cyclin D1 expression and acceleration of the G1/S transition of the cell cycle. Here we present evidence of a role for KLF5 in accelerating mitotic entry in H-Ras-transformed NIH3T3 fibroblasts. When compared with non-transformed parental NIH3T3 cells, H-Ras-transformed fibroblasts exhibit an increase in mitotic index, levels of cyclin B1 and Cdc2, and cyclin B1/Cdc2 kinase activity. Inhibition of KLF5 expression in H-Ras-transformed cells with KLF5-specific small interfering RNA (siRNA) results in a decrease in each of the aforementioned parameters, with a concomitant reduction in the transforming potential of the cells. Conversely, over-expression of KLF5 in NIH3T3 cells leads to an increase in the promoter activity of the genes encoding cyclin B1 and Cdc2. These results indicate that KLF5 accelerates mitotic entry in H-Ras-transformed cells by transcriptionally activating cyclin B1 and Cdc2, which leads to an increase in cyclin B1/Cdc2 kinase activity. Extending our previous observation that KLF5 activates cyclin D1 transcription to promote G1/S transition, our current results further support a crucial function for KLF5 in mediating cellular transformation caused by oncogenic H-Ras.

Keywords: Transformation, Mitotic index, Colony formation, Proliferation, G2/M transition, Small interfering RNA

1. Introduction

Krüppel-like factors (KLFs) are Sp1-like transcription factors that bear significant homology to the Drosophila melanogaster segmentation gene, Krüppel [1-5]. Proteins of this family contain a DNA binding domain consisting of C2H2 zinc fingers and exhibit important regulatory functions [2-5]. Two related, yet functionally contrasting, members of the KLF family, KLF4 and KLF5, are differentially expressed in the intestinal epithelium [3,6]. While KLF4 is predominantly expressed in the post-mitotic, terminally differentiated epithelial cells of the intestinal villi [7,8], KLF5 is mostly expressed in the undifferentiated, proliferating cells of the crypt compartment [9]. Studies support a role for KLF4 in functioning as a tumor suppressor of the intestinal epithelial cells [10,11]. In contrast, KLF5 has been shown to accelerate cell proliferation in both cultured fibroblasts and intestinal epithelial cells [12-14].

Ras proteins are a family of small GTPases often found to be constitutively activated in tumors. For example, activating K-Ras mutations are found in approximately 90% of pancreatic and 50% of colon carcinomas [15,16]. Oncogenic Ras isoforms cause the constitutive activation of a signaling cascade involving several proteins including Raf and the mitogen-activated protein kinase (MAPK) [17-19]. Expression of these Ras isoforms can lead to a transformed phenotype in various cell lines, including fibroblasts [12,20] and epithelial cells [21]. Previous experiments demonstrate that constitutive activation of the Ras pathway accelerates the G1/S transition of cell cycle through induction of cyclin D1 expression [13,22,23]. Consequently, oncogenic Ras-mediated transformation leads to serum- and anchorage-independent growth and increased metastatic potential [12,24,25]. A recent study reveals that oncogenic Ras also accelerates the G2/M progression of the cell cycle by increasing the level of cyclin B1 protein [26]. Of note is the G2/M progression is rigorously controlled by the activity of the cyclin B1/Cdc2 kinase complex [27,28].

We recently showed that the level of KLF5 is increased in NIH3T3 fibroblasts transformed by oncogenic H-Ras [13]. This increase is the result of H-Ras-induced activation of MAPK and subsequent induction of the gene encoding early growth response 1 (Egr1), which transcriptionally activates KLF5 [13]. Activated KLF5 then leads to the transcriptional induction of the gene encoding cyclin D1 [13]. Importantly, inhibition of KLF5 expression is accompanied by a reduction in the transforming potential of the cells, including a reduced capacity for anchorage-independent growth [13]. Here we present evidence for an equally important role of KLF5 in accelerating mitotic entry of H-Ras-transformed NIH3T3 fibroblasts through activation of the cyclin B1/Cdc2 kinase complex.

2. Materials and methods

2.1. Cell lines and reagents

Culture media and fetal bovine serum (FBS) were purchased from Mediatech, Inc. (Herndon, VA). The expression construct containing the oncogenic H-Ras was generously provided by Dr. Raul Urrutia [29]. The monoclonal antibody against Cdc2 and Actin were purchased from Santa Cruz Biotechnology (Santa Cruz, CA) and EMD Biosciences (San Diego, CA), respectively. A polyclonal antibody against cyclin B1 was acquired from Rockland Immunochemicals (Gilbertsville, PA). Antibodies against phospho-histone H1 and histone H1 were obtained from EMD Biosciences (San Diego, CA) and Upstate USA (Charlottesville, VA), respectively. The mouse monoclonal antibody directed against phospho-Ser/Thr-Pro MPM2 (mitotic phosphoepitope marker-2) was purchased from Upstate USA. FITC-conjugated anti-mouse IgG (Fc-specific) secondary antibodies were purchased from Sigma (St. Louis, MO). The antibody against phosphorylated histone H3 (Serine-10) was purchased as part of Mitotic Index Hitkit from Cellomics, Inc (Pittsburgh, PA). The −287 cyclin B1 promoter luciferase reporter was generously provided by Dr. Karen Katula [30,31]. The luciferase construct with Cdc2 promoter was generously provided by Dr. Christopher Glass [32].

2.2. Cell lines

NIH3T3 mouse fibroblasts were maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented with 10% FBS and 1% penicillin–streptomycin at 37 °C in a 5% CO2 atmosphere. Stable clones of NIH3T3 transformed by oncogenic H-Ras were selected from foci formed in soft agar as described previously [12]. A representative clone, called Ras7, was used in the previous and current studies [13].

2.3. Cell cycle analysis

Fractions of cells at different phases of the cell cycle were analyzed as previously described [33]. In brief, cells were collected and resuspended in 70% ethanol overnight at −20 °C. Before analysis, cells were pelleted again and incubated with a phospho-Ser/Thr-Pro MPM2 monoclonal antibody diluted in Dulbecco's phosphate-buffered saline for 1 h and then with a FITC-conjugated anti-mouse secondary antibody for 30 min. Cells were then resuspended in a propidium iodide (PI) solution consisting of 50 μg/ml PI, 50 μg/ml RNase A, 0.1% Triton X-100 and 0.1 mM EDTA for 15 min. Cells were then sorted using a FACSCalibur flow cytometer (BD Biosciences, San Jose, CA).

2.4. Cdc2 activity assays

Cdc2 kinase activity was measured as previously described [31]. Cells were lysed using a cell lysis buffer purchased from Cell Signaling Tech. (Beverly, MA) supplemented with a protease inhibitor cocktail purchased from Roche (Indianapolis, IN). The cell extracts were then immunoprecipitated using a Cdc2 monoclonal antibody and protein G-conjugated sepharose beads (Sigma, St. Louis, MO). The pellets were then washed thrice with lysis buffer and then incubated for 30 min at 37 °C with 20 mM HEPES, pH 7.9, 5 mM MgCl2, 1 μg histone H1 (Roche), 1 mM EDTA and 100 μM ATP in a total volume of 20 μl. The samples were then examined by Western blotting using antibodies against phospho-histone H1 and histone H1.

2.5. siRNA transfection

Cells were transfected with small interfering RNA (siRNA) using a protocol similar to that described previously [13]. The KLF5-specific siRNA was designed to target against sequences corresponding to nucleotides between 875 and 895 of the coding region of mouse KLF5 (GenBank Accession No. NM_009769). Control siRNA was also described previously [13]. Cells were cultured in 100-mm culture dishes until 40% confluent and then transfected with siRNA in Opti-MEM with reduced serum content (Invitrogen, Carlsbad, CA) using the Oligofectamine transfection reagent (Invitrogen) for 4 h. The cells were then provided with DMEM containing 10% FBS and 1% penicillin–streptomycin.

2.6. Measurement of mitotic indices

Mitotic indices were measured using Hoechst 33258 stain and phospho-histone H3 (Ser-10) antibodies. Cells were first fixed in a 3% formaldehyde solution in phosphate-buffered saline (PBS) for 15 min. For Hoechst staining, cells were then incubated at room temperature for 20 min after the addition of cold 100% methanol. After rinsing with PBS, a Hoechst 33258 solution was added to a final concentration of 0.2 μg/ml and samples were incubated at room temperature for 15 min. Samples are then rinsed again with PBS and nuclei were visualized using an inverted fluorescence microscope (Nikon, Melville, NY). Mitotic index was scored as the number of mitotic cells (with condensed nuclei) among a total population of 500 stained cells. For staining with anti-phospho-histone H3, protocol described in the Mitotic Index Hitkit (Cellomics, Pittsburgh, PA) was applied. In brief, cells were permeabilized using a permeabilization buffer after fixing cells with 3.7% formaldehyde solution, for 15 min. After rinsing with blocking solution, cells were then incubated with the primary antibody followed by the staining solution (containing Alexa 488 labeled secondary antibody and Hoechst dye) for 1 h each. Cells were then washed and nuclei were visualized using a fluorescent microscope. Mitotic index was scored as the number of phospho-histone H3-positive cells among a total population of 1000 cells counted.

3. Results

3.1. H-Ras-transformed NIH3T3 fibroblasts exhibit an increase in mitotic index, which is decreased by inhibition of KLF5

A clonal derivative of H-Ras-transformed NIH3T3 fibroblasts, called Ras7 [13], was transfected with KLF5-specific or non-specific siRNA and examined daily for mitotic indices using several independent methods. The non-transformed parental NIH3T3 cells and untransfected Ras7 cells were used as controls. Fig. 1 shows that the mitotic index, as measured by either Hoechst 33258 staining (Fig. 1A) or phospho-histone H3 staining (Fig. 1B), was significantly higher in Ras7 cells when compared with the non-transformed parental NIH3T3 cells on all 3 days of culture. Transfection of Ras7 cells with KLF5-specific siRNA resulted in a significant reduction in mitotic index when compared to untransfected or control siRNA-transfected Ras7 cells.

Fig. 1.

Fig. 1

KLF5 increases mitotic activity in oncogenic H-Ras-transformed NIH3T3 cells. (A) NIH3T3 cells, untransfected Ras7 cells, and Ras 7 cells transfected with non-specific (NS) control siRNA or KLF5-specific siRNA from day 1 to day 3 were stained with the Hoechst 33258 dye to label those cells in mitosis. The mitotic index is the percent of cells in mitosis among a total of 500 cells counted. N = 4; **P < 0.01 by two-tailed Student's t test. (B) Cells were immuno-stained with a phospho-histone H3 antibody to label mitotic cells. Hoechst dye was used to stain nuclei. Mitotic index is represented as percent of phospho-histone H3-positive cells among a total of 1000 cells counted. N = 3; *P < 0.05; **P < 0.01 by two-tailed Student's t test.

To provide an additional means of measuring mitosis, we immuno-stained cells with a phospho-MPM2 antibody and sorted cells by FACS following staining with PI [33,34]. As shown in Fig. 2A, the population of MPM2-positive cells was significantly greater in Ras7 cells as compared to non-transformed NIH3T3 cells on days 2 and 3 of culturing. Again, transfection of Ras7 cells with KLF5-specific siRNA, but not non-specific siRNA, significantly reduced the proportion of MPM2-positive cells when compared to untransfected Ras7 cells. A similar finding is observed when the ratios between cells in the M and G2/M phase of the cell cycle were measured (Fig. 2B). These results indicate that KLF5 is responsible for the increase in mitotic index and acceleration in the G2/M transition of the cell cycle in Ras7 cells.

Fig. 2.

Fig. 2

KLF5 accelerates G2/M transition of the cell cycle in H-Ras-transformed NIH3T3 cells. (A) Immuno-staining was performed with a phospho-Ser/Thr-Pro MPM2 antibody to identify cells in the M phase of cell cycle. Cells were then stained with PI to measure the DNA content and subjected to FACS analysis. The percentages of MPM2-positive cells among all cells analyzed were plotted at different time points. N = 3; *P < 0.05; **P < 0.01 by two-tailed Student's t test. (B) The ratio of M-phase population to G2/M-phase population was calculated by dividing the number of MPM2-positive cells by those in the G2/M phase (i.e., cells with a DNA content of 4N). N = 3; *P < 0.05; **P < 0.01 by two-tailed Student's t test.

3.2. H-Ras-mediated transformation of NIH3T3 fibroblasts is accompanied by an increase in the levels of cyclin B1 and Cdc2, and cylinB1/Cdc2 kinase activity, all of which are decreased by inhibition of KLF5

As mitosis is controlled by cyclin B1 and Cdc2 [27,28], we measured KLF5, cyclin B1 and Cdc2 protein levels in NIH3T3 and Ras7 cells, as well as Ras7 cells treated with KLF5-specific siRNA or control siRNA, by Western blot analysis. As shown in Fig. 3A, Ras7 cells contained a higher level of KLF5, cyclin B1 and Cdc2 than NIH3T3 cells (compare lane 2 to lane 1). Transfection of Ras7 cells with KLF5-specific siRNA resulted in a reduction in the level of KLF5 and a concomitant reduction in the levels of cyclin B1 and Cdc2 (Fig. 3A, lane 4). In contrast, transfection of Ras7 cells with control non-specific siRNA did not change the levels of any of the three proteins (Fig. 3A, lane 3). Importantly, Fig. 3B shows that Ras7 cells contained a higher level of cyclin B1/Cdc2 kinase activity than NIH3T3 cells (compare lane 2 to lane 1) and that this increase was reduced in Ras7 cells treated with KLF5-speicfic siRNA but not control siRNA (compare lane 4 to lane 3). These results indicate that KLF5 is responsible for the increased cyclin B1/Cdc2 kinase activity and that such an increase leads to the observed increase in mitotic activity in Ras7 cells.

Fig. 3.

Fig. 3

Effect of KLF5 inhibition by siRNA on cyclin B1, Cdc2, and cyclin B1/Cdc2 kinase activity in H-Ras transformed cells. (A) The levels of KLF5, cyclin B1, and Cdc2, in NIH3T3 cells, untransfected Ras7 cells, and Ras7 cells transfected with non-specific (NS) control siRNA or KLF5-specific siRNA, were determined by Western blot analysis. Actin serves as a loading control. (B) The cyclin B1/Cdc2 kinase activity was measured by immunoprecipitation with a Cdc2 antibody followed by in vitro kinase reaction using histone H1 as a substrate. P-histone H1 is the phosphorylated product.

3.3. KLF5 activates cyclin B1 and Cdc2 promoter activity

To determine the mechanism by which KLF5 increases cyclin B1 and Cdc2, we conducted co-transfection experiments using a KLF5-expression vector and a luciferase reporter construct containing the cyclin B1 or Cdc2 promoter. Cells cotransfected with an empty vector served as a control. As seen in Fig. 4, cells transfected with the KLF5-expression vector exhibited significantly higher activities from both cyclin B1 and Cdc2 promoters when compared to those transfected with the empty vector. These results indicate that KLF5 is a transcriptional activator of the cyclin B1 and Cdc2 genes.

Fig. 4.

Fig. 4

KLF5 increases cyclin B1 and Cdc2 promoter activity. NIH3T3 cells were co-transfected with a luciferase plasmid containing cyclin B1 promoter (cyclin B1 promoter-luc) [30] or Cdc2 promoter (Cdc2 promoter-luc) [32] and either a control vector (pMT3 vector) or with a KLF5-expression vector (pMT3-KLF5). Luciferase activity was determined 2 days following transfection and normalized to the internal control, Renilla luciferase. N = 4; **P < 0.001 when compared to PMT3 vector-transfected cells using two-tailed Student's t test.

4. Discussion

Cellular transformation caused by oncogenic Ras is characterized by increased cell proliferation, loss of contact inhibition and anchorage-independent cell growth [35,36]. Cell proliferation is generally associated with accelerated G1/S and G2/M cell cycle transitions [37]. The response of the G1/S cell cycle machinery to Ras-induced MAPK stimulation is well established, consisting of the upregulation of cyclins D [23,38], E [39], and A, as well as their corresponding cyclin dependent kinases (Cdks) [40]. There is also evidence that the Ras signaling cascade can accelerate the G2/M transition [41,42].

Ample evidence indicates that cyclin B1 is an important protein regulating proliferation. Depletion of cyclin B1 is found to inhibit proliferation and induce apoptosis in human tumor cells [43]. Previous studies also demonstrate that cyclin B1 is crucial for mediating oncogenic H-Ras-induced transformation [26,41]. The onset of mitosis in the cell is strictly regulated by several factors, among them cyclin B1 and Cdc2 [27,44]. The results of our study, which demonstrate that H-Ras-transformed fibroblasts exhibit increased cyclin B1/Cdc2 protein levels and activity, as well as increased mitotic index, are consistent with these previous findings.

We previously reported that transformation by oncogenic H-Ras results in an increase in the rate of cell proliferation and the S-phase population of cells [13]. In H-Ras-transformed cells, KLF5 is up-regulated due to H-Ras-activated MAPK and the increased KLF5 is responsible for an increase in cyclin D1 expression at the transcriptional level [13]. This leads to accelerated proliferation. In the current study, we identified cyclin B1 and Cdc2 as two novel targets of KLF5 upon its induction by H-Ras (Figs. 3A and 4). Consequently, there is an increase in cyclin B1/Cdc2 kinase activity, which becomes inhibited upon treatment of H-Ras-transformed cells with KLF5-specific siRNA (Fig. 3B). Importantly, inhibition of KLF5 expression in H-Ras-transformed cells results in a reduction of mitotic index and the M-to-G2/M ratio of cells (Figs. 1 and 2). These results demonstrate that KLF5 serves a crucial function in mediating the biological activity of H-Ras in accelerating the G2/M transition, an event which promotes cellular transformation. A model describing the signal pathway as elicited by oncogenic H-Ras and mediated by KLF5 is illustrated in Fig. 5.

Fig. 5.

Fig. 5

A model for the role of KLF5 in mediating H-Ras-induced transformation. The induction of KLF5 by H-Ras is mediated by MAPK and Egr-1 as previously demonstrated [13]. KLF5 then activates the transcription of the genes encoding cyclin D1 [13] and cyclin B1/Cdc2 (this study). The combined effect is an acceleration of the cell cycle, which eventually results in cellular transformation.

KLF5 has previously been shown to exert a pro-proliferative effect on non-transformed NIH3T3 fibroblasts and intestinal epithelial cells including IEC-6, IEC-18, and IMCE [12,45]. Stable expression of KLF5 in NIH3T3 cells has also been shown to result in a transformed phenotype including increased proliferation and anchorage-independent growth [12]. In addition, all-trans retinoic acid (ATRA) inhibits proliferation of IEC-6 cells by inhibiting KLF5 gene expression [14]. In contrast, over-expression of KLF5 in several human cancer cells has been reported to inhibit their growth in vitro [45,46]. Thus, KLF5 may exhibit different functions at different stages of tumor formation. It is of interest to note that a reason for KLF5's pleiotropic effect may be its differential regulation by the ubiquitin–proteasome pathway in non-transformed and cancer cells [47]. Nonetheless, the exact mechanism by which KLF5 affects growth of cancer cells remain to be established.

Acknowledgments

We thank Drs. C. Glass, K. Katula, and R. Urrutia for providing plasmids. This work was in part supported by National Institutes of Health Grants DK52230, DK64399, and CA84197. V.W.Y. is the recipient of a Georgia Cancer Coalition Distinguished Cancer Clinician Scientist award. W.B.D. is a member of the Emory University Medical Scientist Training Program.

Abbreviations

DMEM

Dulbecco's modified Eagle's medium

FACS

fluorescence-activated cell sorting

FBS

fetal bovine serum

KLF5

Krüppel-like factor 5

MAPK

mitogen-activated protein kinase

MPM2

mitotic phosphoepitope marker 2

PBS

phosphate-buffered saline

PI

propidium iodide

siRNA

small interfering RNA

References

  • 1.Kadonaga JT, Carner KR, Masiarz FR, Tjian R. Isolation of cDNA encoding transcription factor Sp1 and functional analysis of the DNA binding domain. Cell. 1987;51:1079–1090. doi: 10.1016/0092-8674(87)90594-0. [DOI] [PubMed] [Google Scholar]
  • 2.Kaczynski J, Cook T, Urrutia R. Sp1- and Krüppel-like transcription factors. Genome Biol. 2003;4:206. doi: 10.1186/gb-2003-4-2-206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Dang DT, Pevsner J, Yang VW. The biology of the mammalian Krüppel-like family of transcription factors. Int. J. Biochem. Cell Biol. 2000;32:1103–1121. doi: 10.1016/s1357-2725(00)00059-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Bieker JJ. Krüppel-like factors: three fingers in many pies. J. Biol. Chem. 2001;276:34355–34358. doi: 10.1074/jbc.R100043200. [DOI] [PubMed] [Google Scholar]
  • 5.Black AR, Black JD, Azizkhan-Clifford J. Sp1 and krüppel-like factor family of transcription factors in cell growth regulation and cancer. J. Cell. Physiol. 2001;188:143–160. doi: 10.1002/jcp.1111. [DOI] [PubMed] [Google Scholar]
  • 6.Ghaleb AM, Nandan MO, Chanchevalap S, Dalton WB, Hisamuddin IM, Yang VW. Krüppel-like factors 4 and 5: the yin and yang regulators of cellular proliferation. Cell Res. 2005;15:92–96. doi: 10.1038/sj.cr.7290271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.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. doi: 10.1074/jbc.271.33.20009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Ton-That H, Kaestner KH, Shields JM, Mahatanankoon CS, Yang VW. Expression of the gut-enriched Krüppel-like factor gene during development and intestinal tumorigenesis. FEBS Lett. 1997;419:239–243. doi: 10.1016/s0014-5793(97)01465-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Conkright MD, Wani MA, Anderson KP, Lingrel JB. A gene encoding an intestinal-enriched member of the Krüppel-like factor family expressed in intestinal epithelial cells. Nucleic Acids Res. 1999;27:1263–1270. doi: 10.1093/nar/27.5.1263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Zhao W, Hisamuddin IM, Nandan MO, Babbin BA, Lamb NE, Yang VW. Identification of Krüppel-like factor 4 as a potential tumor suppressor gene in colorectal cancer. Oncogene. 2004;23:395–402. doi: 10.1038/sj.onc.1207067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.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 tumorigene-city. Oncogene. 2003;22:3424–3430. doi: 10.1038/sj.onc.1206413. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Sun R, Chen X, Yang VW. Intestinal-enriched Krüppel-like factor (Krüppel-like factor 5) is a positive regulator of cellular proliferation. J. Biol. Chem. 2001;276:6897–6900. doi: 10.1074/jbc.C000870200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Nandan MO, Yoon HS, Zhao W, Ouko LA, Chanchevalap S, Yang VW. Krüppel-like factor 5 mediates the transforming activity of oncogenic H-Ras. Oncogene. 2004;23:3404–3413. doi: 10.1038/sj.onc.1207397. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Chanchevalap S, Nandan MO, Merlin D, Yang VW. All-trans retinoic acid inhibits proliferation of intestinal epithelial cells by inhibiting expression of the gene encoding Krüppel-like factor 5. FEBS Lett. 2004;578:99–105. doi: 10.1016/j.febslet.2004.10.079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Bos JL, Verlaan-de Vries M, Marshall CJ, Veeneman GH, van Boom JH, van der Eb AJ. A human gastric carcinoma contains a single mutated and an amplified normal allele of the Ki-ras oncogene. Nucleic Acids Res. 1986;14:1209–1217. doi: 10.1093/nar/14.3.1209. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Forrester K, Almoguera C, Han K, Grizzle WE, Perucho M. Detection of high incidence of K-ras oncogenes during human colon tumorigenesis. Nature. 1987;327:298–303. doi: 10.1038/327298a0. [DOI] [PubMed] [Google Scholar]
  • 17.Schaeffer HJ, Weber MJ. Mitogen-activated protein kinases: specific messages from ubiquitous messengers. Mol. Cell. Biol. 1999;19:2435–2444. doi: 10.1128/mcb.19.4.2435. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Van Aelst L, Barr M, Marcus S, Polverino A, Wigler M. Complex formation between RAS and RAF and other protein kinases. Proc. Natl. Acad. Sci. USA. 1993;90:6213–6217. doi: 10.1073/pnas.90.13.6213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Khosravi-Far R, Der CJ. The Ras signal transduction pathway. Cancer Metast. Rev. 1994;13:67–89. doi: 10.1007/BF00690419. [DOI] [PubMed] [Google Scholar]
  • 20.Shao J, Sheng H, DuBois RN, Beauchamp RD. Oncogenic Ras-mediated cell growth arrest and apoptosis are associated with increased ubiquitin-dependent cyclin D1 degradation. J. Biol. Chem. 2000;275:22916–22924. doi: 10.1074/jbc.M002235200. [DOI] [PubMed] [Google Scholar]
  • 21.Subbaramaiah K, Telang N, Ramonetti JT, Araki R, DeVito B, Weksler BB, Dannenberg AJ. Transcription of cyclooxygenase-2 is enhanced in transformed mammary epithelial cells. Cancer Res. 1996;56:4424–4429. [PubMed] [Google Scholar]
  • 22.Lovec H, Sewing A, Lucibello FC, Muller R, Moroy T. Oncogenic activity of cyclin D1 revealed through cooperation with Ha-ras: link between cell cycle control and malignant transformation. Oncogene. 1994;9:323–326. [PubMed] [Google Scholar]
  • 23.Lavoie JN, L'Allemain G, Brunet A, Muller R, Pouyssegur J. Cyclin D1 expression is regulated positively by the p42/p44MAPK and negatively by the p38/HOGMAPK pathway. J. Biol. Chem. 1996;271:20608–20616. doi: 10.1074/jbc.271.34.20608. [DOI] [PubMed] [Google Scholar]
  • 24.Shields JM, Pruitt K, McFall A, Shaub A, Der CJ. Understanding Ras: ‘it ain't over’ ‘til it's over’. Trends Cell Biol. 2000;10:147–154. doi: 10.1016/s0962-8924(00)01740-2. [DOI] [PubMed] [Google Scholar]
  • 25.Yang JJ, Kang JS, Krauss RS. Ras signals to the cell cycle machinery via multiple pathways to induce anchorage-independent growth. Mol. Cell. Biol. 1998;18:2586–2595. doi: 10.1128/mcb.18.5.2586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Santana C, Ortega E, Garcia-Carranca A. Oncogenic H-ras induces cyclin B1 expression in a p53-independent manner. Mutat. Res. 2002;508:49–58. doi: 10.1016/s0027-5107(02)00172-0. [DOI] [PubMed] [Google Scholar]
  • 27.Nurse P. Universal control mechanism regulating onset of M-phase. Nature. 1990;344:503–508. doi: 10.1038/344503a0. [DOI] [PubMed] [Google Scholar]
  • 28.Taylor WR, Stark GR. Regulation of the G2/M transition by p53. Oncogene. 2001;20:1803–1815. doi: 10.1038/sj.onc.1204252. [DOI] [PubMed] [Google Scholar]
  • 29.Gebelein B, Fernandez-Zapico M, Imoto M, Urrutia R. KRAB-independent suppression of neoplastic cell growth by the novel zinc finger transcription factor KS1. J. Clin. Invest. 1998;102:1911–1919. doi: 10.1172/JCI1919. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Katula KS, Wright KL, Paul H, Surman DR, Nuckolls FJ, Smith JW, Ting JP, Yates J, Cogswell JP. Cyclin-dependent kinase activation and S-phase induction of the cyclin B1 gene are linked through the CCAAT elements. Cell Growth Differ. 1997;8:811–820. [PubMed] [Google Scholar]
  • 31.Yoon HS, Yang VW. Requirement of Krüppel-like factor 4 in preventing entry into mitosis following DNA damage. J. Biol. Chem. 2004;279:5035–5041. doi: 10.1074/jbc.M307631200. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Sugarman JL, Schonthal AH, Glass CK. Identification of a cell-type-specific and E2F-independent mechanism for repression of cdc2 transcription. Mol. Cell. Biol. 1995;15:3282–3290. doi: 10.1128/mcb.15.6.3282. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Andreassen PR, Lohez OD, Lacroix FB, Margolis RL. Tetraploid state induces p53-dependent arrest of nontrans-formed mammalian cells in G1. Mol. Biol. Cell. 2001;12:1315–1328. doi: 10.1091/mbc.12.5.1315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Davis FM, Tsao TY, Fowler SK, Rao PN. Monoclonal antibodies to mitotic cells. Proc. Natl. Acad. Sci. USA. 1983;80:2926–2930. doi: 10.1073/pnas.80.10.2926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Downward J. Targeting RAS signalling pathways in cancer therapy. Nat. Rev. Cancer. 2003;3:11–22. doi: 10.1038/nrc969. [DOI] [PubMed] [Google Scholar]
  • 36.Evan GI, Vousden KH. Proliferation, cell cycle and apoptosis in cancer. Nature. 2001;411:342–348. doi: 10.1038/35077213. [DOI] [PubMed] [Google Scholar]
  • 37.Stacey DW. Cyclin D1 serves as a cell cycle regulatory switch in actively proliferating cells. Curr. Opin. Cell. Biol. 2003;15:158–163. doi: 10.1016/s0955-0674(03)00008-5. [DOI] [PubMed] [Google Scholar]
  • 38.Albanese C, Johnson J, Watanabe G, Eklund N, Vu D, Arnold A, Pestell RG. Transforming p21ras mutants and c-Ets-2 activate the cyclin D1 promoter through distinguishable regions. J. Biol. Chem. 1995;270:23589–23597. doi: 10.1074/jbc.270.40.23589. [DOI] [PubMed] [Google Scholar]
  • 39.Leone G, DeGregori J, Sears R, Jakoi L, Nevins JR. Myc and Ras collaborate in inducing accumulation of active cyclin E/Cdk2 and E2F. Nature. 1997;387:422–426. doi: 10.1038/387422a0. [DOI] [PubMed] [Google Scholar]
  • 40.Winston JT, Coats SR, Wang YZ, Pledger WJ. Regulation of the cell cycle machinery by oncogenic ras. Oncogene. 1996;12:127–134. [PubMed] [Google Scholar]
  • 41.Agapova LS, Volodina JL, Chumakov PM, Kopnin BP. Activation of Ras–Ral pathway attenuates p53-independent DNA damage G2 checkpoint. J. Biol. Chem. 2004;279:36382–36389. doi: 10.1074/jbc.M405007200. [DOI] [PubMed] [Google Scholar]
  • 42.Pomerance M, Thang MN, Tocque B, Pierre M. The Ras-GTPase-activating protein SH3 domain is required for Cdc2 activation and mos induction by oncogenic Ras in Xenopus oocytes independently of mitogen-activated protein kinase activation. Mol. Cell. Biol. 1996;16:3179–3186. doi: 10.1128/mcb.16.6.3179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Yuan J, Yan R, Kramer A, Eckerdt F, Roller M, Kaufmann M, Strebhardt K. Cyclin B1 depletion inhibits proliferation and induces apoptosis in human tumor cells. Oncogene. 2004;23:5843–5852. doi: 10.1038/sj.onc.1207757. [DOI] [PubMed] [Google Scholar]
  • 44.Pines J. Cell cycle. Checkpoint on the nuclear frontier. Nature. 1999;397:104–105. doi: 10.1038/16344. [DOI] [PubMed] [Google Scholar]
  • 45.Bateman NW, Tan D, Pestell RG, Black JD, Black AR. Intestinal tumor progression is associated with altered function of KLF5. J. Biol. Chem. 2004;279:12093–12101. doi: 10.1074/jbc.M311532200. [DOI] [PubMed] [Google Scholar]
  • 46.Chen C, Bhalala HV, Vessella RL, Dong JT. KLF5 is frequently deleted and down-regulated but rarely mutated in prostate cancer. Prostate. 2003;55:81–88. doi: 10.1002/pros.10205. [DOI] [PubMed] [Google Scholar]
  • 47.Chen C, Sun X, Ran Q, Wilkinson KD, Murphy TJ, Simons JW, Dong JT. Ubiquitin–proteasome degradation of KLF5 transcription factor in cancer and untrans-formed epithelial cells. Oncogene. 2005;24:3319–3327. doi: 10.1038/sj.onc.1208497. [DOI] [PubMed] [Google Scholar]

RESOURCES