pmc logo imageJournal ListSearchpmc logo image
Logo of pnasPNAS Home page.Reference to the article.PNAS Info for AuthorsPNAS SubscriptionsPNAS About
Proc Natl Acad Sci U S A. 2005 September 6; 102(36): 12765–12770.
Published online 2005 August 29. doi: 10.1073/pnas.0505484102.
PMCID: PMC1200285
Cell Biology
Inhibition of Single Minded 2 gene expression mediates tumor-selective apoptosis and differentiation in human colon cancer cells
Mireille J. Aleman,*§ Maurice Phil DeYoung,*§ Matthew Tress,* Patricia Keating,* Gary W. Perry, and Ramaswamy Narayanan***
*Center for Molecular Biology and Biotechnology, Departments of Biology and Chemistry, and Center for Complex System and Brain Sciences, Florida Atlantic University, 777 Glades Road, Boca Raton, FL 33431
** To whom correspondence should be addressed. E-mail: rnarayan/at/fau.edu.
§M.J.A. and M.P.D. contributed equally to this work.
Communicated by Herbert Weissbach, Florida Atlantic University, Boca Raton, FL, July 21, 2005
Received April 4, 2005.
Abstract
A Down's syndrome associated gene, Single Minded 2 gene short form (SIM2-s), is specifically expressed in colon tumors but not in the normal colon. Antisense inhibition of SIM2-s in a RKO-derived colon carcinoma cell line causes growth inhibition, apoptosis, and inhibition of tumor growth in a nude mouse tumoriginicity model. The mechanism of cell death in tumor cells is unclear. In the present study, we investigated the pathways underlying apoptosis. Apoptosis was seen in a tumor cell-specific manner in RKO cells but not in normal renal epithelial cells, despite inhibition of SIM2-s expression in both of these cells by the antisense. Apoptosis was depended on WT p53 status and was caspase-dependent; it was inhibited by a pharmacological inhibitor of mitogen-activated protein kinase activity. Expression of a key stress response gene, growth arrest and DNA damage gene (GADD)45α, was up-regulated in antisense-treated tumor cells but not in normal cells. In an isogenic RKO cell line expressing stable antisense RNA to GADD45α, a significant protection of the antisense-induced apoptosis was seen. Whereas antisense-treated RKO cells did not undergo cell cycle arrest, several markers of differentiation were deregulated, including alkaline phosphatase activity, a marker of terminal differentiation. Protection of apoptosis and block of differentiation showed a correlation in the RKO model. Our results support the tumor cell-selective nature of SIM2-s gene function, provide a direct link between SIM2-s and differentiation, and may provide a model to identify SIM2-s targets.
Keywords: Down's syndrome, antisense, p53
 
The Single Minded 2 (SIM2) gene is present in chromosome 21 at the Down's syndrome critical region, which in triplication is associated with diverse phenotypic characteristics of Down's syndrome (1). Patients with Down's syndrome show various neurological symptoms and a high incidence of leukemia (1, 2). Members of the SIM family include SIM1 and SIM2, which map to 6q16.3-q21 and 21q22.2, respectively (3), and belong to a family of transcription factors containing a basic helix–loop–helix motif, two period homologue (PER)/ARNT/SIM (ARNT, aryl hydrocarbon receptor nuclear translocator) domains, and the HIF1α/SIM/TRH domains (46). In Drosophila, SIM is a master regulator of fruit-fly neurogenesis, regulating midline gene expression (6, 7). The SIM2 gene exists in two distinct forms, the long and short forms (SIM2-l and SIM2-s), due to alternative splicing (3). A putative cancer-related role of the SIM family of genes is their ability to transcriptionally regulate key metabolic enzymes to inactivate carcinogens (8). Binding of carcinogens to the aryl hydrocarbon receptor (AhR), which is kept sequestered in the cytoplasm by heat-shock protein (HSP) 90 (9), dissociates HSP 90. The ligand-bound AhR is then translocated into the nucleus, where it can dimerize with ARNT (10). This complex binds to the xenobiotic response element, present in the promoters of key oxidative enzymes, and activates gene transcription (8, 11), thus causing inactivation of the carcinogen. The SIM proteins can inhibit the dimerization of the ligand-bound AhR/ARNT complex (12) and hence prevent carcinogen metabolism, leading to cumulative DNA damage and cancer.
The growth arrest and DNA damage (GADD) family of genes was originally isolated from UV radiation-treated cells and subsequently grouped according to their coordinate regulation by growth arrest and DNA damage (13). The GADD family members include GADD34, -45α, -45β, -45γ, and -153 (14, 15). These are stress-response genes induced by both genotoxic and nongenotoxic stresses (1618). GADD45α is the most extensively studied member of the family and is regulated in both a p53-dependent and -independent manner (13, 19). GADD45α-mediated apoptosis may involve activation of JNK and/or p38 mitogen-activated protein kinase (MAPK) signaling pathways (14, 20).
We have recently demonstrated that the SIM2 short-form (SIM2-s) gene is specifically expressed in distinct solid tumors, including colon, pancreas, and prostate, but not in the corresponding normal tissues (2123). Antisense inhibition of SIM2-s expression caused apoptosis in colon and pancreatic cancer-derived cells. In an effort to understand the molecular mechanism underlying the role of SIM2-s in tumor cell growth, we have embarked on mapping the key pathways linked to SIM2-s. We show that apoptosis is tumor cell-selective and requires GADD45α function. Further, we demonstrate that key pathways, including caspase, p38 MAPK activities, and WT p53 status, are critical to apoptosis. Our results link apoptosis to induction of tumor cell-selective differentiation.
Methods
Cell Culture. Isogenic colon cancer-derived cell lines (RKO, RKO-E6-p53-knockout, and RKO-AS45.1-stable GADD45α antisense clone) and OM-1 and SW 480 colon carcinoma-derived cells were obtained from American Type Culture Collection and were maintained as recommended by American Type Culture Collection. Early-passage primary human renal epithelial (HRE) cells were obtained from Cambrex (Walkersville, MD) and were cultured following the manufacturer's instructions.
Inhibitors and Antibodies. Antisense SIM2-s was synthesized as described (22). Methylmethane sulfonate was obtained from Sigma Aldrich. Caspase-family inhibitor set IV was obtained from Biovision (Mountain View, CA). p38 kinase inhibitor (SB202190) was obtained from A. G. Scientific (San Diego). General tyrosine kinase inhibitor (Genistein) was from Sigma Aldrich. Antibodies were obtained from Santa Cruz Biotechnology.
RT-PCR. End-point RT-PCR analysis and primers used were described previously (22, 23). Quantitative real-time PCR was done by using β2 microglobulin standards (Roche Applied Sciences, Indianapolis). Exon-specific RT-PCR primers were used (see Table 1, which is published as supporting information on the PNAS web site).
Cell Cycle and FACS Analysis of GADD45α Protein. Cells were harvested after 100 nM antisense or control oligo treatment and fixed at 4°C overnight in 70% ethanol. The fixed cells were resuspended in PBS and treated with 20 μg/ml RNaseA for 30 min at 37°C, stained with 60 μg/ml propidium iodide at room temperature for 30 min, and scanned by an Epics XL-MCL Flow Cytometer (Beckman Coulter). Cell cycle distribution was calculated by using multicycle v.3.1.1 software (Phoenix Flow Systems, San Diego). GADD45α protein expression was analyzed in antisense-treated RKO or HRE cells by using either GADD45α-phycoerythrin (PE)-conjugated antibody or its IgG1 isotype control PE-conjugated antibody by using an Epics XL-MCL Flow Cytometer (Beckman Coulter).
Caspase and p38 Kinase Inhibitor Experiments. For inhibitor studies, the cells were pretreated with 20 μM caspase inhibitors, 20 μM SB202190 (p38 MAPK inhibitor), or 10 μM Genistein (general tyrosine kinase inhibitor) for 1 h before transfection with the oligonucleotides and then continuously incubated with the inhibitors. At select time points, apoptosis was measured by using the Cell Death Detection ELISAPlus (Roche Applied Science), as described (23). Each experiment was repeated two to three times.
Alkaline Phosphatase (ALP) Activity. ALP activity was measured as described (24). The assays were repeated in three to five independent experiments.
Immunohistochemistry (IHC). Antisense-treated cells in culture were analyzed by IHC by using affinity-purified rabbit antiserum to SIM2-s-specific hydrophobic peptides SHGGGWQMETEPSRF (Sigma Genosys, The Woodlands, TX), as described (22, 23).
Results
Inhibition of SIM2-s Expression Results in Tumor Cell-Selective Apoptosis. In an earlier study (22), we have shown that RKO colon carcinoma cells undergo rapid apoptosis (14–24 h) in response to inhibition of SIM2-s gene expression by antisense. To clarify whether the inhibition of SIM2-s mRNA precedes apoptosis, we have performed an early time-course analysis. RKO cells were treated with either the control or the SIM2-s antisense at 4, 6, 8, 10, and 14 h. The treated cells were analyzed for SIM2-s mRNA expression by real-time PCR and for apoptosis by ELISA (Fig. 1A). Inhibition of SIM2-s mRNA was seen as early as 8 h after transfection, whereas evidence of apoptosis was seen only at 10 h onwards. The half-life of SIM2-s has been shown to be 2 h (25), which can account for the rapidity of apoptosis induction. Whereas in the normal colon tissue, SIM2-s expression was not detected, normal human kidney is one of the few organs in which SIM2-s expression was detected (21, 22). Hence we tested the effects of the SIM2-s antisense in early-passage primary HRE cells and compared them with RKO cells (Fig. 1B). Similar levels of inhibition of SIM2-s mRNA were seen upon antisense treatment in both the tumor and normal cells (50%), but pronounced apoptosis was seen only in the RKO-derived tumor cells and not in the normal HRE cells. The antisense inhibited SIM2-s protein expression in both these cells (Fig. 1C). Over 80% of the antisense-treated RKO cells underwent apoptosis. The antisense-treated HRE cells did not show inhibition of cell proliferation, evidence of DNA laddering, morphological changes, nuclear condensation, or cytoplasmic blebbing (data not shown). These results suggest that apoptosis may be tumor cell-selective.
Fig. 1.Fig. 1.
SIMS2-s in apoptosis. (A) SIM2-s mRNA inhibition precedes apoptosis. RKO cells were treated with either the control or SIM2-s antisense oligonucleotides and, at indicated times, SIM2-s mRNA (shaded) was analyzed by real-time quantitative RT-PCR and the (more ...)
To further clarify the apoptotic mechanism, we next used specific pathway inhibitors. The RKO cells were significantly protected from apoptosis induced by SIM2-s antisense by both general (Z-VAD-FMK) and specific caspase inhibitors (caspase 9 and 10). This effect was not seen when caspase 2 or 8 inhibitors were used (Fig. 1D). Inhibitors of other caspases (caspases 3, 4, 5, 6, and 13) showed only marginal protection (10–15%) against apoptosis (data not shown). To further map the signal transduction involved in apoptosis, we next used key kinase pathway pharmacological inhibitors. Significant protection from apoptosis was seen when the RKO cells were pretreated with a p38 MAPK pathway inhibitor (SB 202190). As a control, we used Genistein, a nonspecific tyrosine kinase inhibitor, which did not cause such a protection (Fig. 1E). Treatment of RKO cells with these inhibitors alone did not cause apoptosis (data not shown).
We next addressed whether apoptosis mediated by the inhibition of SIM2-s expression depends on p53 tumor suppressor gene function. Isogenic RKO (WT p53) or RKO-E6 (p53 functional knockout by HPV-E6 oncogene) cells were treated either with the control oligo or SIM2-s antisense and at the indicated time analyzed by apoptosis ELISA (Fig. 1F). In the isogenic RKO E6 cells, SIM2-s antisense-induced apoptosis was 4- to 5-fold reduced.
Identification of GADD45α as a Critical Mediator of SIM2-s Antisense Mechanism. Up-regulation of GADD genes is often seen in tumor cells undergoing apoptosis. Hence we investigated the regulation of GADD family members in the SIM2-s antisense-treated RKO cells (Fig. 2A). The inhibition of SIM2-s by antisense in RKO cells resulted in up-regulation of the expression of three GADD family members (GADD45α, -45β, and -34). GADD45γ and -153 gene expression was not detected in the RKO cells. GADD45α protein expression was up-regulated in the RKO (Fig. 2B) but not in normal HRE cells (Fig. 2C) upon treatment with the antisense.
Fig. 2.Fig. 2.
GADD45α as a critical mediator of apoptosis. (A) Up-regulation of GADD family members by SIM2-s inhibition. Expression of GADD family members (GADD45α, GADD45β, and GADD34) and SIM2-s was analyzed in the SIM2-s antisense-treated (more ...)
We next tested whether GADD45α is functionally relevant to apoptosis by using the RKO-GADD45α knockout isogenic cell line. The RKO/RKO GADD45α antisense cells (RKO-AS45.1) were treated with 100 nM either control or SIM2-s antisense oligonucleotides and at 10, 14, 18, and 24 h, the cells were analyzed for apoptosis by ELISA (Fig. 2D). Apoptosis was significantly inhibited (>66%) in the RKO-AS45.1 cells. Because p38 MAPK is upstream of GADD45α, these results are consistent with the protection of apoptosis seen by using a p38 MAPK inhibitor, SB 202190 (Fig. 1E).
Inhibition of SIM2-s Expression Does Not Cause Cell Cycle Arrest. Enforced expression of GADD45α in diverse cell types causes growth suppression and activation of a G2 checkpoint, which depends on WT p53 status (for a review, see ref. 18). GADD45α overexpression results in growth suppression through activation of the G2 checkpoint of the cell cycle (15). Although the growth suppression is seen in a p53-independent manner, the G2 arrest is seen only in tumor and normal cells with WT p53 function (26, 27). The up-regulation of GADD45α mRNA and protein expression seen in our study in a tumor cell-selective manner raised a possibility that the observed apoptosis induced by the SIM2-s antisense may be a consequence of cell cycle arrest. Hence we performed cell cycle analysis of the antisense-treated RKO or HRE cells (Fig. 3A). Nonsynchronous cells were treated either with control or SIM2-s antisense oligonucleotides and, at different time points, the cells were analyzed by FACS. Methylmethane sulfonate, which has been shown to cause G2 arrest in RKO cells (27), was used as a positive control. For simplicity, RKO at 14 h and HRE at 24 h are shown. No discernible differences of cells at G1, S, and G2 phases were seen between the control and the antisense-treated RKO or HRE cells. The sub-G0 apoptotic peak was seen in the antisense-treated RKO but not in the HRE cells. Consistent with the lack of cell cycle block, expression of p21 mRNA and protein was not affected by the antisense treatment of RKO cells (Fig. 3B). These results suggested that apoptosis induction by inhibition of SIM2-s is not a consequence of the cell cycle block.
Fig. 3.Fig. 3.
Cell-cycle-independent differentiation by SIM2-s. (A) Cell cycle profile of RKO and HRE cells treated at 14 or 24 h, respectively, either untreated (–) or treated with methylmethane sulfonate (25 μg/ml) or 100 nM of either the control (more ...)
Induction of Differentiation in the Antisense-Treated Cancer Cells. Colon cancer cells undergo terminal differentiation and apoptosis in response to diverse differentiation stimuli (28, 29). Key pathways including caspase and p38 MAPK/ERK1/2 are critical to the initiation of differentiation and the subsequent apoptosis signals (3032). In colon cancer-derived cell lines, ALP activity is used to measure differentiation status (33). In addition, depending on the cell type, other markers, such as mucins (MUC2/MUC3), and carcinoembryonic antigen (CEACAM) family members are shown to be up-regulated upon terminal differentiation (34). Another marker, immature colon carcinoma transcript (ICT1), a colon-specific gene, has been shown in colon carcinoma-derived cell lines to be down-regulated in response to the induction of terminal differentiation (35). The appearance of these cell markers is often associated with indications of cell death, such as detachment from the culture dish (36). Hence, we investigated the differentiated status of the SIM2-s antisense-treated RKO cells for marker gene expression and ALP activity (Fig. 3 B–D). Expression of ICT1 was down-regulated in a time-dependent manner after antisense treatment (Fig. 3B). mRNA expression of ALP1, MUC2, and CEACAM7 was also up-regulated upon antisense treatment in the RKO cells but not in normal HRE cells (Fig. 3C). The up-regulation of these genes was seen 8 h after antisense treatment, a time point where no apoptosis is seen (see Fig. 1A). ALP activity in the antisense-treated RKO cells was stimulated to levels similar to those attained with a known inducer of differentiation, sodium butyrate (Fig. 3D). Consistent with the protection of antisense-mediated apoptosis seen in the RKO-E6 and -AS45.1 isogenic cell lines, the induction of ALP activity was also inhibited in these cell lines. Basal and antisense-induced ALP was significantly inhibited by the p38 MAPK inhibitor (SB202190). Antisense-induced ALP activity was also abolished by the caspase 9 inhibitor (Z-LEHD-FMK).
Discussion
Whereas the RKO cell expresses only the short isoform of SIM2 (SIM2-s), normal kidney and HRE cells express both the long and the short isoforms (SIM2-s/SIM2-l), and thus the tumor cell-selective apoptosis may be due to a redundancy mechanism. Alternatively, in normal HRE cells, the SIM2-s gene may transcriptionally regulate distinct sets of genes. A possibility remains that this could be due to differences in SIM2-s RNA/protein stability in normal and tumor cells.
We further map the pathways involved in SIM2-s antisense-mediated apoptosis as dependent on caspase activity (caspases 9 and 10). Caspases are key proteases involved in both initiating apoptotic mechanisms in response to proapoptotic signals (initiator caspases) and cell disassembly (effector caspases). Caspase 9 is a key mitochondrial-dependent initiator caspase that activates caspase 3, an effector molecule, as well as other caspases. Caspase 10 is an initiator caspase that plays a key role in receptor-mediated apoptotic signals (for reviews, see refs. 37 and 38). The significant protection of SIM2-s antisense-mediated apoptosis by caspase 9 and 10 inhibitors in the RKO cells identifies both extrinsic and intrinsic caspase-dependent pathway in tumor-selective apoptosis. Results with isogenic p53 null RKO cells (RKO-E6) further provide a hint into the mechanism of apoptosis induced by the inhibition of SIM2-s expression. Inactivation of p53 either by overexpression of E6 oncoprotein (RKO-E6) or mutation (as in SW 480 and OM-1 colon carcinoma cells) inhibits apoptosis induced by the antisense. It is tempting to speculate that targets of SIM2-s require WT p53 tumor suppressor gene function.
Our results establish GADD45α as a critical mediator of SIM2-s gene function in regulating growth and differentiation. A recent study involving overexpression of melanoma differentiation-associated gene (MDA-7/IL-24) showed induction of melanoma-selective apoptosis and up-regulation of GADD45α, and apoptosis was inhibited by a p38 MAPK inhibitor (39). MAPKs are serine–threonine kinases that are activated by phosphorylation in response to a wide array of extracellular signals (40, 41). Three distinct MAPKs (ERK, JNK, and p38) play a critical role in apoptosis and differentiation; the ERKs (ERK1/ERK2) are implicated in growth and differentiation, and JNK and p38 are stress-activated MAPKs that play a key role in apoptosis and stress response (for a review, see ref. 42).
The tumor-selective apoptosis seen in the RKO cells by inhibition of SIM2-s gene expression did not result in cell cycle arrest or alteration of the levels of p21 mRNA or protein. The GADD45α protein has been shown to associate with nuclear proteins, including the cdc2–cyclin B1 kinase complex (43), p21 (44), and proliferating cell nuclear antigen (45); however, up-regulation of GADD45α is not always associated with cell cycle arrest. For example, Jin et al. (26) demonstrated that overexpression of GADD45α in HCT116, but not in HeLa cells, resulted in G2 arrest despite growth suppression in both of these cell lines. Thus, activation of the GADD45-mediated stress response and the subsequent apoptosis we observe may be exerted at the level of p38 MAPK/JNK independent of cell cycle blockage. Inhibition of apoptosis induced by the SIM2-s antisense with the p38 MAPK inhibitor strongly supports this.
The induction of several markers of differentiation and the up-regulation of ALP activity in antisense-treated RKO cells implicate differentiation as a key mechanism. However, the SIM2-s independent mechanism must exist in leukemia-derived cell lines, because these cells do not express SIM2-s (22). Deregulation of diverse differentiation markers before apoptosis suggests apoptosis is a consequence of differentiation. The lack of induction of ALP-1, MUC2, and carcinoembroyonic antigen 7 mRNAs in the normal HRE cells by the antisense implies that, in these normal cells, the SIM2-s gene regulates a distinct set of genes. Frequently, when cells differentiate, they are withdrawn from the cell cycle, but there are several exceptions to this phenomenon. In murine keratinocytes, activation of the Raf/MAPK pathway leads to expression of differentiation markers, but the cells continue to proliferate in a p53-dependent manner (46). In the U-937 leukemia cell line, overexpression of WT 53 induces differentiation, but the cells are not arrested in the G1 phase of the cell cycle (47).
In recent years, a new model has been emerging, a continuum model whereby cells receive signals to differentiate at all stages of the cell cycle (48). Our results of induction of differentiation without cell cycle arrest are consistent with this model. Our results further link the GADD-mediated stress signal, p38 MAPK, and p53 function in the induction of differentiation and apoptosis by inhibition of SIM2-s gene expression. A model underlying this link is shown in Fig. 4. The putative role of SIM2 protein is as a transcriptional repressor (49). SIM2-s protein, either as a homodimer or a heterodimer, causes transcriptional repression of key differentiation-inducing genes, leading to proliferation. Down-regulation of SIM2-s expression causes a release from this repression, and SIM2-s-regulated proteins such as X or X/Y can cause transcriptional activation of gene(s) in a p53-dependent manner. Such a transcription activates GADD-mediated signals in a p38 MAPK-dependent manner, leading to terminal differentiation and caspase-dependent apoptosis.
Fig. 4.Fig. 4.
Proposed model for the role of SIM2-s in differentiation and apoptosis in tumor cells.
Trisomy 21 is linked to human cancer, as evidenced by a 20-fold elevated risk of leukemia in people with Down's syndrome (50). Higher risks for acute lymphoblastic leukemia, myelodysplastic syndrome, acute myeloid leukemia, and early-onset Alzheimer's degeneration are seen in Down's patients, whereas solid tumor formation is relatively rare. However, in an earlier study (22), we demonstrated that SIM2-s expression was not seen in diverse leukemia-derived cell lines, suggesting the epidemiological link between Down's patients and leukemia may not involve the function of the SIM2-s gene. It is unclear whether overexpression of SIM2-s is a cause or an effect of transformation. However, reduced exposure to environmental factors that contribute to cancer risk, tumor suppressor genes on chromosome 21, and a slower rate of replication or higher likelihood of apoptosis in Down's syndrome cells offer a few reasons for the low frequency of solid tumors seen in Down's patients (51).
We have also recently embarked on a genome-wide global gene expression study with a view toward identification of SIM2-s-regulated targets using SIM2-s antisense-treated RKO cells as a model. By using gene ontology of GeneChip (Affymetrix, Santa Clara, CA) output, various apoptosis and differentiation-related genes were found to be up-regulated in SIM2-s antisense-treated RKO cells, consistent with the data presented here (see Table 2, which is published as supporting information on the PNAS web site).
Conclusion
Our results suggest the tumor cell-selective nature of the SIM2-s gene. Key pathways, including GADD, caspase, and p53 function, are identified as critical to the function of the SIM2-s gene. Identification of differentiation as a key mechanism for the apoptosis provides a first link between SIM2-s, and differentiation and may allow for future experiments aimed at SIM2-s target discovery.
Supplementary Material
Supporting Tables
Acknowledgments
We thank J. Narayanan for editorial assistance. This paper is contribution no. P200504 from the Center of Excellence in Biomedical and Marine Biotechnology.
Notes
Author contributions: G.W.P. and R.N. designed research; M.J.A., M.P.D., M.T., and P.K. performed research; M.J.A., M.P.D., and R.N. analyzed data; and R.N. wrote the paper.
Abbreviations: ALP, alkaline phosphatase; GADD, growth arrest and DNA damage gene; MAPK, mitogen-activated protein kinase; SIM2, Single Minded 2; SIM2-s, SIM2 short form; HRE, human renal epithelial.
¶Present address: Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114.
References
1.
Kola, I. & Hertzog, P. J. (1997) Hum. Mol. Genet. 6:, 1713–1727. [PubMed].
2.
Kola, I. & Hertzog, P. J. (1998) Curr. Opin. Genet. Dev. 8:, 316–321. [PubMed].
3.
Chrast, R., Scott, H. S., Chen, H., Kudoh, J., Rossier, C., Minoshima, S., Wang, Y., Shimizu, N. & Antonarakis, S. E. (1997) Genome Res. 7:, 615–624. [PubMed].
4.
Isaac, D. D. & Andrew, D. J. (1996) Genes Dev. 10:, 103–117. [PubMed].
5.
McGuire, J., Coumailleau, P., Whitelaw, M. L., Gustafsson, J. A. & Poellinger, L. (1995) J. Biol. Chem. 270:, 31353–31357. [PubMed].
6.
Nambu, J. R., Lewis, J. O., Wharton, K. A., Jr., & Crews, S. T. (1991) Cell 67:, 1157–1167. [PubMed].
7.
Thomas, J. B., Crews, S. T. & Goodman, C. S. (1988) Cell 52:, 133–141. [PubMed].
8.
Hankinson, O. (1995) Annu. Rev. Pharmacol. Toxicol. 35:, 307–340. [PubMed].
9.
Swanson, H. I. & Bradfield, C. A. (1993) Pharmacogenetics 3:, 213–230. [PubMed].
10.
Swanson, H. I., Chan, W. K. & Bradfield, C. A. (1995) J. Biol. Chem. 270:, 26292–26302. [PubMed].
11.
Nebert, D. W., Roe, A. L., Dieter, M. Z., Solis, W. A., Yang, Y. & Dalton, T. P. (2000) Biochem. Pharmacol. 59:, 65–85. [PubMed].
12.
Lees, M. J. & Whitelaw, M. L. (1999) Mol. Cell. Biol. 19:, 5811–5822. [PubMed].
13.
Fornace, A. J., Jr., Nebert, D. W., Hollander, M. C., Luethy, J. D., Papathanasiou, M., Fargnoli, J. & Holbrook, N. J. (1989) Mol. Cell. Biol. 9:, 4196–4203. [PubMed].
14.
Takekawa, M. & Saito, H. (1998) Cell 95:, 521–530. [PubMed].
15.
Zhan, Q., Lord, K. A., Alamo, I., Jr., Hollander, M. C., Carrier, F., Ron, D., Kohn, K. W., Hoffman, B., Liebermann, D. A. & Fornace, A. J., Jr. (1994) Mol. Cell. Biol. 14:, 2361–2371. [PubMed].
16.
Hildesheim, J. & Fornace, A. J., Jr. (2002) Clin. Cancer Res. 8:, 2475–2479. [PubMed].
17.
Hollander, M. C., Sheikh, M. S., Yu, K., Zhan, Q., Iglesias, M., Woodworth, C. & Fornace, A. J., Jr. (2001) Int. J. Cancer 96:, 22–31. [PubMed].
18.
Sheikh, M. S., Hollander, M. C. & Fornance, A. J., Jr. (2000) Biochem. Pharmacol. 59:, 43–45. [PubMed].
19.
Kastan, M. B., Zhan, Q., el Deiry, W. S., Carrier, F., Jacks, T., Walsh, W. V., Plunkett, B. S., Vogelstein, B. & Fornace, A. J., Jr. (1992) Cell 71:, 587–597. [PubMed].
20.
Harkin, D. P., Bean, J. M., Miklos, D., Song, Y. H., Truong, V. B., Englert, C., Christians, F. C., Ellisen, L. W., Maheswaran, S., Oliner, J. D., et al. (1999) Cell 97:, 575–586. [PubMed].
21.
DeYoung, M. P., Scheurle, D., Damania, H., Zylberberg, Z. & Narayanan, R. (2002) Anticancer Res. 22:, 3149–3158. [PubMed].
22.
DeYoung, M. P., Tress, M. & Narayanan, R. (2003) Proc. Natl. Acad. Sci. USA 100:, 4760–4765. [PubMed].
23.
DeYoung, M. P., Tress, M. & Narayanan, R. (2003) Cancer Lett. 200:, 25–31. [PubMed].
24.
Huang, Y., Horvath, C. M. & Waxman, S. (2000) Cancer Res. 60:, 3200–3206. [PubMed].
25.
Woods, S. L. & Whitelaw, M. L. (2002) J. Biol. Chem. 277:, 10236–10243. [PubMed].
26.
Jin, S., Tong, T., Fan, W., Fan, F., Antinore, M. J., Zhu, X., Mazzacurati, L., Li, X., Petrik, K. L., Rajasekaran, B., et al. (2002) Oncogene 21:, 8696–8704. [PubMed].
27.
Wang, X. W., Zhan, Q., Coursen, J. D., Khan, M. A., Kontny, H. U., Yu, L., Hollander, M. C., O'Connor, P. M., Fornace, A. J., Jr., & Harris, C. C. (1999) Proc. Natl. Acad. Sci. USA 96:, 3706–3711. [PubMed].
28.
Phillips, T. E., Huet, C., Bilbo, P. R., Podolsky, D. K., Louvard, D. & Neutra, M. R. (1988) Gastroenterology 94:, 1390–1403. [PubMed].
29.
Zweibaum, A., Pinto, M., Chevalier, G., Dussaulx, E., Triadou, N., Lacroix, B., Haffen, K., Brun, J. L. & Rousset, M. (1985) J. Cell Physiol. 122:, 21–29. [PubMed].
30.
Cai, J., Chen, Y., Murphy, T. J., Jones, D. P. & Sartorelli, A. C. (2004) Arch. Biochem. Biophys. 424:, 119–127. [PubMed].
31.
Daniel, C., Schroder, O., Zahn, N., Gaschott, T. & Stein, J. (2004) Biochem. Biophys. Res. Commun. 324:, 1220–1226. [PubMed].
32.
Ding, Q., Wang, Q. & Evers, B. M. (2001) Biochem. Biophys. Res. Commun. 284:, 282–288. [PubMed].
33.
Augeron, C. & Laboisse, C. L. (1984) Cancer Res. 44:, 3961–3969. [PubMed].
34.
Velcich, A., Palumbo, L., Jarry, A., Laboisse, C., Racevskis, J. & Augenlicht, L. (1995) Cell Growth Differ. 6:, 749–757. [PubMed].
35.
van Belzen, N., Diesveld, M. P., van der Made, A. C., Nozawa, Y., Dinjens, W. N., Vlietstra, R., Trapman, J. & Bosman, F. T. (1995) Eur. J. Biochem. 234:, 843–848. [PubMed].
36.
Kirlin, W. G., Cai, J., DeLong, M. J., Patten, E. J. & Jones, D. P. (1999) J. Nutr. 129:, 1827–1835. [PubMed].
37.
Chen, M. & Wang, J. (2002) Apoptosis. 7:, 313–319. [PubMed].
38.
Thornberry, N. A. (1998) Chem. Biol. 5:, R97–R103. [PubMed].
39.
Sarkar, D., Su, Z. Z., Lebedeva, I. V., Sauane, M., Gopalkrishnan, R. V., Valerie, K., Dent, P. & Fisher, P. B. (2002) Proc. Natl. Acad. Sci. USA 99:, 10054–10059. [PubMed].
40.
Schaefer, A. W., Kamiguchi, H., Wong, E. V., Beach, C. M., Landreth, G. & Lemmon, V. (1999) J. Biol. Chem. 274:, 37965–37973. [PubMed].
41.
Seger, R. & Krebs, E. G. (1995) FASEB J. 9:, 726–735. [PubMed].
42.
Minden, A. & Karin, M. (1997) Biochim. Biophys. Acta 1333:, F85–F104. [PubMed].
43.
Zhan, Q., Antinore, M. J., Wang, X. W., Carrier, F., Smith, M. L., Harris, C. C. & Fornace, A. J., Jr. (1999) Oncogene 18:, 2892–2900. [PubMed].
44.
Kearsey, J. M., Coates, P. J., Prescott, A. R., Warbrick, E. & Hall, P. A. (1995) Oncogene 11:, 1675–1683. [PubMed].
45.
Smith, M. L., Chen, I. T., Zhan, Q., Bae, I., Chen, C. Y., Gilmer, T. M., Kastan, M. B., O'Connor, P. M. & Fornace, A. J., Jr. (1994) Science 266:, 1376–1380. [PubMed].
46.
Roper, E., Weinberg, W., Watt, F. M. & Land, H. (2001) EMBO Rep. 2:, 145–150. [PubMed].
47.
Ehinger, M., Bergh, G., Johnsson, E., Baldetorp, B., Olsson, I. & Gullberg, U. (1998) Exp. Hematol. 26:, 1043–1052. [PubMed].
48.
Cooper, C. S. (2001) J. Theor. Biol. 208:, 399–402. [PubMed].
49.
Ema, M., Morita, M., Ikawa, S., Tanaka, M., Matsuda, Y., Gotoh, O., Saijoh, Y., Fujii, H., Hamada, H., Kikuchi, Y., et al. (1996) Mol. Cell. Biol. 16:, 5865–5875. [PubMed].
50.
Hasle, H. (2001) Lancet Oncol. 2:, 429–436. [PubMed].
51.
Yang, Q., Rasmussen, S. A. & Friedman, J. M. (2002) Lancet 359:, 1019–1025. [PubMed].