Skip to main content
  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

AACR logo

  • Register
  • Log in
  • Log out
  • My Cart
Advertisement

Main menu

  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

  • AACR Publications
    • Blood Cancer Discovery
    • Cancer Discovery
    • Cancer Epidemiology, Biomarkers & Prevention
    • Cancer Immunology Research
    • Cancer Prevention Research
    • Cancer Research
    • Clinical Cancer Research
    • Molecular Cancer Research
    • Molecular Cancer Therapeutics

User menu

  • Register
  • Log in
  • Log out
  • My Cart

Search

  • Advanced search
Cancer Research
Cancer Research
  • Home
  • About
    • The Journal
    • AACR Journals
    • Subscriptions
    • Permissions and Reprints
    • Reviewing
  • Articles
    • OnlineFirst
    • Current Issue
    • Past Issues
    • Meeting Abstracts
    • Collections
      • COVID-19 & Cancer Resource Center
      • Focus on Computer Resources
      • Highly Cited Collection
      • Editors' Picks
      • "Best of" Collection
  • For Authors
    • Information for Authors
    • Author Services
    • Early Career Award
    • Best of: Author Profiles
    • Submit
  • Alerts
    • Table of Contents
    • Editors' Picks
    • OnlineFirst
    • Citations
    • Author/Keyword
    • RSS Feeds
    • My Alert Summary & Preferences
  • News
    • Cancer Discovery News
  • COVID-19
  • Webinars
  • Search More

    Advanced Search

Carcinogenesis

Delineation of the Molecular Basis for Selenium-induced Growth Arrest in Human Prostate Cancer Cells by Oligonucleotide Array

Yan Dong, Haitao Zhang, Lesleyann Hawthorn, Howard E. Ganther and Clement Ip
Yan Dong
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Haitao Zhang
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Lesleyann Hawthorn
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Howard E. Ganther
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
Clement Ip
  • Find this author on Google Scholar
  • Find this author on PubMed
  • Search for this author on this site
DOI:  Published January 2003
  • Article
  • Info & Metrics
  • PDF
Loading

Abstract

Despite the growing interest in selenium intervention of prostate cancer in humans, scanty informationis currently available on the molecular mechanism of selenium action. Our past research indicated that methylseleninic acid (MSA) is an excellent reagent for investigating the anticancer effect of selenium in vitro. The present study was designed to examine the cellular and molecular effects of MSA in PC-3 human prostate cancer cells. After exposure to physiological concentrations of MSA, these cells exhibited a dose- and time-dependent inhibition of growth. MSA retarded cell cycle progression at multiple transition points without changing the proportion of cells in different phases of the cell cycle. Flow cytometric analysis of annexin V- and propidium iodide-labeled cells showed a marked induction of apoptosis by MSA. Array analysis with the Affymetrix human genome U95A chip was then applied to profile the gene expression changes that might mediate the effects of selenium. Gene profiling was done in a time course experiment (at 12, 24, 36, and 48 h) using synchronized cells. A large number of potential selenium-responsive genes with diverse biological functions were identified. These genes fell into 12 clusters of distinct kinetics pattern of modulation by MSA. The expression changes of 10 genes known to be critically involved in cell cycle regulation were selected for verification by Western analysis to determine the reliability of the array data. An agreement rate of 70% was obtained based on these confirmation experiments. The array data enabled us to focus on the role of potential key genes (e.g., GADD153, CHK2, p21WAF1, cyclin A, CDK1, and DHFR) that might be targets of MSA in impeding cell cycle progression. The data also provide valuable insights into novel biological effects of selenium, such as inhibition of cell invasion, DNA repair, and stimulation of transforming growth factor β signaling. The present study demonstrates the utility of a genome-wide analysis to elucidate the mechanism of selenium chemoprevention.

INTRODUCTION

Recruitment to the National Cancer Institute-sponsored SELECT 3 trial began in 2001. This is a Phase III, double-blind, placebo-controlled, 12-year trial designed to assess the effect of selenium and vitamin E, either individually or in combination, on the incidence of prostate cancer. The launching of this trial is largely driven by the milestone finding of Clark et al. (1) that selenized yeast supplementation was capable of significantly reducing the incidence of prostate (RR = 0.37), lung (RR = 0.54), and colon cancers (RR = 0.42). The SELECT protocol also provides for the establishment of a repository for prostate biopsy tissue, blood cells, and plasma. These materials will be put aside for research discoveries in the future. One of the secondary objectives of the trial is to study cellular and molecular biomarkers using the banked samples and to delineate their relevance with respect to prostate carcinogenesis and drug effects (2) . Despite the considerable public interest in the potential benefit of selenium chemoprevention of prostate cancer, scanty information is currently available on the molecular targets or the signaling mechanism underlying the anticancer action of selenium. Our present study was aimed at addressing this gap of knowledge with the use of a human prostate cancer cell line.

Se-Met is the selenium compound used in the SELECT. It is, however, not particularly suitable for mechanism studies in cell culture. The reason is that Se-Met needs to be metabolized primarily in the liver to a monomethylated intermediate for the expression of its anticancer activity (3, 4, 5, 6) , and epithelial tissues generally have a low capacity to generate a monomethylated selenium metabolite from Se-Met. Consequently, concentrations of Se-Met that are 20–100 times above physiological levels are necessary to cause growth inhibition in cultured cells. Excessively high concentrations of Se-Met could produce a spectrum of nonspecific effects that may not be related to the anticancer effect of selenium. To obviate this problem, a stable monomethylated selenium metabolite, MSA (CH3SeO2H), was developed specifically for in vitro studies (7) . We found that premalignant human breast cell lines were sensitive to growth inhibition and cell cycle block by MSA at a concentration as low as 2.5 μm (8) . In addition, Jiang et al. (9) recently reported that MSA induced apoptosis in DU-145 human prostate cancer cells at a concentration of 5 μm. Sinha et al. (10) also showed that with mouse mammary tumor cells, a 10-min exposure to 5 μm MSA was sufficient to cause a change in the expression of a handful of genes as detected by the Atlas mouse cDNA expression array. The concentration of selenium used in the above studies is within the physiological range of selenium in the circulation. As expected, MSA also has excellent anticancer activity in vivo (7) . We are, therefore, confident that the information obtained with MSA from cell culture studies would be relevant to the action of selenium.

In this study, we first examined the dose-dependent effect of MSA on the growth of the PC-3 human prostate cancer cell line. We then showed that growth inhibition by MSA was likely attributable to a combined effect on cell cycle block and apoptosis. Next we used the oligonucleotide array technology to gain further insight into the gene expression changes that might play a role in the regulation of these cellular events. Many potential selenium-responsive genes were identified by this method. These genes fell into 12 clusters of distinct kinetics pattern of modulation by MSA. The early response genes were grouped on the basis of their known functions in cell growth and tumorigenesis. From the array data, we were able to develop an integrated scheme of signaling pathways that might explain the action of selenium in blocking cell cycle progression.

MATERIALS AND METHODS

Selenium Reagents and Cell Line.

MSA was synthesized as described previously (7) . Se-Met was purchased from Sigma (St. Louis, MO). The PC-3 human prostate cancer cells were obtained from American Type Culture Collection (Manassas, VA). The cells were cultured in RPMI 1640 supplemented with 10% fetal bovine serum, 100 units/ml penicillin, 100 μg/ml streptomycin, and 2 mm glutamine, and maintained in an atmosphere of 5% CO2 in a 37°C humidified incubator.

MTT Cell Proliferation Assay.

The assay, which is based on the conversion of the yellow tetrazolium salt MTT to purple formazan crystals by metabolically active cells (11) , provides a quantitative determination of viable cells. Cells were seeded in 24-well plates at a density designed to reach 70–80% confluency at the time of assay. At 48 h after seeding, cells were treated with various concentrations of Se-Met or MSA in triplicate. After 24, 48, or 72 h of treatment, 200 μl of MTT was added to each well of cells, and the plate was incubated for 4 h at 37°C. The MTT crystals from both attached and floating cells were solubilized in isopropanol and subjected to centrifugation to pellet the cellular debris. Spectrophotometric absorbance of each sample was measured at 570 nm using a Spectra Microplate Reader (SLT-Labinstruments Ges.m.b.H., Salzburg, Austria).

Cell Cycle Analysis.

PC-3 cells were plated at a density of 104 cells/cm2 in T75 culture flasks and allowed to grow for 48 h to reach 70–80% confluency. Synchronization of cells was achieved by starving in serum-free medium for 48 h. Over 85% of cells were in G0 phase at the end of this time period. On returning to regular growth medium for 6 h, cells were exposed to 10 μm MSA. The procedure of serum-starvation and refeeding has been described previously by Sinha and Medina (12) and Sinha et al. (13) to study the effect of selenium on cell cycling. After treatment for 24, 32, or 48 h, cells were trypsinized, washed in PBS, and fixed overnight in 70% ethanol at 4°C. The ethanol solution was subsequently removed after centrifugation, and cells were resuspended in a buffer containing 10 mm Tris (pH 7.5), 125 mm sucrose, 2.5 mm MgCl2, 0.185% NP40, 0.02 mg/ml RNase A, 0.05% sodium citrate, and 25 μg/ml PI. After incubation on ice for 1 h, cells were analyzed for DNA content using a FACScan cytometer (Becton Dickinson).

BrdUrd Labeling Assay.

PC-3 cells were plated at a density of 104 cells/cm2 in T75 culture flasks and synchronized as described above. On returning to regular growth medium for 6 h, cells were exposed to 10 μm MSA for 24 or for 48 h. During the last 30 min of MSA treatment, cells were labeled with 10 μm BrdUrd (10 μl of 1 mm BrdUrd was added to each ml of culture media). BrdUrd-labeled cells were trypsinized, fixed, treated with DNase I, and stained with FITC-conjugated anti-BrdUrd antibody using the BrdUrd Flow Kit from BD Pharmigen (San Diego, CA). Stained cells were then quantified by flow cytometry, and the data were analyzed with the WinList software (Variety Software House, Topsham, ME).

Quantitation of Apoptosis by Flow Cytometry.

PC-3 cells were plated at a density of 104 cells/cm2 in T175 culture flasks. At 48 h after seeding, cells were exposed to either 5 or 10 μm MSA for 48 or 72 h. Adherent cells harvested by mild trypsinization were pooled together with detached cells. Cells were stained with biotin-conjugated Annexin V, FITC-conjugated streptavidin, and PI using the Annexin V-Biotin Apoptosis Detection kit (Oncogene Research Products, Boston, MA) as per the manufacturer’s protocol. Apoptotic cells were subsequently counted by flow cytometry, and the data were analyzed with the WinList software (Variety Software House, Topsham, ME).

Oligonucleotide Array Analysis.

PC-3 cells were plated at a density of 104 cells/cm2 in 15-cm culture dishes. Synchronization was achieved as described above. After exposure to 10 μm MSA for 12, 24, 36, or 48 h, total RNA and protein were isolated using TRIzol (Life Technologies, Inc.). The experiment was repeated, and the total RNA collected from the replicate was pooled and submitted to microarray analysis using the U95A chip from Affymetrix (Santa Clara, CA). Biotinylated cRNA probe generation, as well as array hybridization, washing, and staining, was carried out according to the standard Affymetrix GeneChip protocol. Fluorescence intensity for each chip was captured with a Hewlett-Packard laser confocal scanner. Absolute analysis of each chip and comparative analysis of MSA-treated samples with the untreated control samples were performed by using the Affymetrix Microarray Suite software. The mean hybridization signal for each sample was set as 1000 arbitrary units to normalize the signal values of all of the genes on the chip (global normalization) between different samples. A treatment/control signal ratio of ≥2 or ≤0.5 was chosen as the criterion for induction or repression, respectively. These threshold values are commonly used in the literature for microarray expression analysis (14, 15, 16) . GENECluster program (Massachusetts Institute of Technology, Boston, MA) and Affymetrix Data Mining Tool were used for clustering analysis.

Western Blot Analysis.

Western blot analysis was performed as described previously (17) using the TRIzol isolated protein. Briefly, ∼50 μg of protein was resolved over 10–15% SDS-PAGE and transferred to polyvinylidine difluoride membrane. The blot was blocked in blocking buffer [5% nonfat dry milk, 10 mm Tris (pH 7.5), 10 mm NaCl. and 0.1% Tween 20] overnight at 4°C, incubated with the primary antibody at 37°C for 1 h, followed by incubation with an antimouse antirabbit, or antisheep horseradish peroxidase-conjugated secondary antibody (Bio-Rad, Hercules, CA) at 37°C for 30 min. Individual protein bands were visualized by an enhanced chemiluminescence kit obtained from Amersham Pharmacia Biotech (Piscataway, NJ). Immunoreactive bands were quantitated by volume densitometry using the ImageQuant software (Molecular Dynamics, Sunnyvale, CA), and normalized to actin. The following monoclonal antibodies were used in this study (source): anti-actin (Sigma, St. Louis, MO); anti-DHFR, CDK1, and CDK2 (BD Transduction Laboratory, San Jose, CA); anti-PCNA (Santa Cruz Biotechnology, Santa Cruz, CA); and anti-cyclin A, cyclin E2, CDK4, p21WAF1 (NeoMarkers, Fremont, CA). Polyclonal antibodies to CHK2 and GADD153 were obtained from Calbiochem (La Jolla, CA) and Santa Cruz Biotechnology (Santa Cruz, CA), respectively.

Statistical Analysis.

The Student’s two-tailed t test was used to determine significant differences between treatment and control values, and P < 0.05 was considered statistically significant.

RESULTS

Sensitivity of Human Prostate Cancer Cells to MSA.

The inhibitory effects of MSA and Se-Met on the accumulation of PC-3 cells were assessed by the MTT assay. As shown in Table 1 ⇓ , MSA was able to significantly suppress the growth of PC-3 cells in a time- and dose-dependent manner. At 72 h of treatment, 5 μm MSA reduced cell number by ∼25%. Increasing the concentration of MSA to 10 μm resulted in a more pronounced effect, leading to a greater magnitude of growth inhibition in a shorter period of exposure. In contrast, a concentration of 200 or 400 μm Se-Met was required to produce significant decreases in cell number at 72 h or 48 h, respectively. It is thus evident that MSA is much more potent than Se-Met in inhibiting growth of these prostate cells.

View this table:
  • View inline
  • View popup
Table 1

Effect of MSA or Se-Met on the accumulation of PC-3 cells at three treatment durations

Cell Cycle Block by MSA.

To determine whether the decrease in cell number accumulation by MSA was related to cell cycle arrest, we proceeded to assess the evidence of cell cycle perturbation by flow cytometry of ethanol-permeabilized cells stained with PI. Synchronized PC-3 cells were treated with 10 μm MSA for 24, 32, or 48 h. MSA did not cause any significant change in cell cycle distribution (Fig. 1) ⇓ . However, flow cytometry of BrdUrd-labeled cells showed that MSA treatment resulted in a drastic decrease in the number of cells synthesizing DNA (Fig. 2) ⇓ . Therefore, the data suggest that MSA probably blocked cell cycle progression at multiple stages. It should be noted that flow cytometry of PI-stained cells would not be able to detect a change in the proportion of cells in different phases of the cell cycle (i.e., the percentage of cells in G0-G1, S phase, or G2-M) if there is a persistent slowdown in the transition in all phases of the cell cycle.

Fig. 1.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 1.

Cell cycle distribution in PC-3 cells treated with MSA. Results are expressed as means ± SE (n = 3). ∗, statistically significant (P < 0.05) versus untreated control.

Fig. 2.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 2.

BrdUrd (BrdU) labeling of PC-3 cells treated with MSA. Results are expressed as means ± SE (n = 3). ∗, statistically significant (P < 0.05) versus untreated control.

Induction of Apoptosis by MSA.

In an attempt to determine whether MSA might also induce cell death, we incubated exponentially growing PC-3 cells for 48 or 72 h in the presence of 5 or 10 μm MSA, and quantified the extent of apoptosis by flow cytometric analysis of cells labeled with annexin V and PI. Phosphatidylserine externalization is a characteristic of cells undergoing apoptosis. Annexin V has a strong affinity for phosphatidylserine. Staining cells simultaneously with annexin V and PI allows the resolution of intact cells (double negative), early apoptotic cells (annexin V-positive and PI-negative), and late apoptotic or necrotic cells (double positive), which can be located in the lower left, lower right, and upper right quadrants of the cytograms of Fig. 3A ⇓ , respectively. Because only cells that are annexin V-positive and PI-negative are truly representative of apoptotic cells, the percentage of this cell population was quantitated from four individual experiments and shown in a bar graph form in Fig. 3B ⇓ . MSA caused an induction of apoptosis at the 48-h time point, and the effect was maintained with longer exposure to MSA for 72 h. The increase of apoptosis, which followed the occurrence of growth arrest, appeared to maximize with 5 μm MSA, and no further enhancement was detected with 10 μm MSA.

Fig. 3.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 3.

Quantitation of apoptotic cells by flow cytometric analysis of MSA-treated PC-3 cells labeled with annexin V and PI. A, cytograms from flow cytometric analysis. Intact cells, early apoptotic cells, and late apoptotic and necrotic cells are located in the lower left, lower right, and upper right quadrants of the cytograms, respectively. B, percentages of early apoptotic cells. Data are presented as means ± SE (n = 4). ∗, statistically significant (P < 0.05) versus untreated control.

Profiling of MSA-responsive Genes by Oligonucleotide Array Analysis.

We used the Affymetrix human genome U95A Chip to profile the changes in gene expression and to characterize selenium-responsive targets that might lead to cell growth inhibition by MSA. This GeneChip contains probes to 12,000 known genes. As it would be more informative to do the profiling at a series of time points than to do replicate analysis at a single time point, we decided to commit our available resources to the former design. For each time point at 12, 24, 36, and 48 h post-MSA, three separate preparations of RNA samples were pooled and submitted to array hybridization.

Pairwise comparative analysis between MSA-treated samples and the corresponding untreated control samples at each time point was performed by using the Affymetrix Microarray Suite software. This software determines whether a given gene is differently expressed based on a decision matrix including the net change in intensity values, fold of change, and other parameters. A no-change decision call was assigned a value of “1.” Genes with expression changes of ≥2 or ≤0.5 were considered as MSA-responsive genes. The 2-fold difference limit was chosen based on our previous experience with microarray data analysis and was also in general agreement with other reported array experiments. Table 2 ⇓ shows the number of genes induced or repressed by MSA at each time point. There were significantly fewer MSA-modulated genes at the 48-h time point than at the other three early time points. This could be because growth inhibition by MSA has reached ∼50% at 48 h (Table 1) ⇓ and because the underpinning molecular changes have already peaked and receded by this time.

View this table:
  • View inline
  • View popup
Table 2

Number of genes modulated by MSA

To study in detail the kinetics of expression changes in response to MSA treatment, the genes modulated by MSA at one or more time points were subjected to clustering analysis using the SOM algorithm. Clustering analysis avails to the grouping of genes according to similarities in their expression profiles across multiple time points. The SOM algorithm is ideal for pattern discovery and has been found to be useful in the elucidation of biological pathways, because genes that are regulated in a coordinated fashion are often related. A total of 2647 genes were included in this analysis. This represents 21% of the genes on the U95A GeneChip. These genes fell into 12 clusters of distinct kinetics pattern of modulation by MSA (Fig. 4) ⇓ , with the early response genes in clusters 1, 7, and 8; the intermediate response genes in clusters 2, 3, 6, 10, and 11; and the late response genes in clusters 4, 5, 9, and 12. Because cell growth inhibition by 10 μm MSA occurred between 24 to 48 h (Table 1) ⇓ , we consider the genes modulated by MSA at the 12-h time point as the early-response genes. We also believe that the change in expression of these early-response genes is important in initiating the cascade of events leading to the action of MSA. We, therefore, classify these early-response genes according to their known functions in cell growth/tumorigenesis (Table 3 ⇓ ).

Fig. 4.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 4.

Average gene expression profiles for each SOM cluster. The fold of change for each gene in a cluster was averaged and plotted against duration of treatment. Data are presented as means ± SE. Numbers in parentheses, the number of genes included in each cluster. Twelve clusters were derived from the clustering analysis, demonstrating different kinetics of modulation by MSA.

View this table:
  • View inline
  • View popup
Table 3

Functional classification of MSA early-response genes

A value >1 represents induction; a value <1 represents repression.

There is a prominent group of genes implicated in cell cycle regulation. Many negative cell cycle regulators were induced by MSA, including checkpoint proteins RAD9 and CHK2, CDK inhibitors p19INK4d and p21WAF1, RB-binding protein 1, GADD45, and protein phosphatase 2C. On the other hand, numerous cyclins, CDKs, and genes required for DNA replication or mitosis were repressed by MSA. Because these genes control the transition of different phases of the cell cycle, the change in their expression could mediate the inhibitory effect of MSA on cell cycle progression. There is a second group of genes involved in apoptosis. Both toll-like receptor 2 and caspase 9 were up-regulated by MSA. Toll-like receptor 2 is a death receptor (18) , and caspase 9 is an upstream activator of the caspase cascade (19) . In addition, MSA down-regulated the expression of survivin, a member of the IAP (inhibitor of apoptosis) family. The reason that so few apoptosis-regulatory genes were affected by MSA at the 12-h time point is probably because the induction of apoptosis by MSA did not become evident until after 48 h. An analysis of apoptosis gene expression changes at the 24- and 36-h time points will be the subject of a separate study.

In Table 3 ⇓ is a group of genes encoding cytoskeleton components, cell membrane glycoproteins, as well as matrix metalloproteinases. The responses of cytoskeleton genes to MSA were varied. However, the up-regulation of invasion suppressors (e.g., cadherins) and the down-regulation of invasion activators (e.g., integrins, endonexin, hyaluronan receptors CD44 and RHAMM, and MMP21/22) suggest a possible role of MSA in inhibiting tumor cell invasion. The table also shows a group of signal transduction genes that are responsive to MSA. In particular, there is a cluster of small GTPases and their associated factors, such as Ras-like protein Tc10, GTPase activating factor-2, RAN-binding protein 8, G protein-coupled receptor 37, RAB31, RAB28, RAB7-like 1, regulator of G protein signaling 10, Rho E, Rho 2, and prenylated RAB acceptor 1. These genes belong to the Ras and Rho family, the members of which are known to regulate diverse cellular functions, such as cell cycle progression, actin cytoskeleton organization, malignant transformation, and MAPK signaling cascades. In addition, MSA-mediated up-regulation of several MAPK cascade genes, including MEK1, MEK3b, MEK5, and JNK1, may amplify its effect on Ras/Rho signaling. Another noteworthy observation is the repression of a key player of the survival pathway, PI3-kinase. This suggests that selenium not only activates proapoptosis signals, but may also suppress survival signals to augment the stimulus to apoptosis.

MSA was found to modulate a large group of transcription factors, especially the zinc-finger family proteins (ZNFs), the myc proteins and associated factors, the ATF/CREB proteins and their binding proteins, as well as the inhibitor of DNA synthesis (Id) family proteins. Many of these transcription factors play critical roles in the regulation of cell cycle progression, apoptosis, and malignant transformation. The change in the expression of these trans-acting factors could lead to an altered transcription of a series of other genes. To wrap up the information summarized in Table 3 ⇓ , two growth factors (bFGF and Wnt7a), an angiogenesis molecule (VEGF-C), and one translation-initiation factor gene (eIF-4γ) that is amplified in cancer cells (20) , were down-regulated by MSA. In contrast, three tumor suppressor genes (BRCA2, DLC-1, and PTEN), three TGF-β family members or receptor (TGF-β, TGF-β type III receptor, and BMP-4), and two DNA repair genes (hPMS2 and ERCC1) were up-regulated by MSA. The regulation of these genes may represent additional mechanisms by which MSA exerts its anticancer effect.

Confirmation of Array Data.

We used Western blot analysis to confirm the changes in expression of a subset of 10 cell cycle genes: CHK2, p21WAF1, GADD153, cyclin A, DHFR, CDK1, CDK2, CDK4, PCNA, and cyclin E2. As shown in Fig. 5 ⇓ and Table 4 ⇓ , the Western expression changes of 7 genes (CHK2, p21WAF1, GADD153, cyclin A, DHFR, CDK1, and CDK2) correlated well with the array data. This represents an agreement rate of 70%. The lack of complete concordance could be attributable to either false positive signals of the array data or the discrepancy between transcript and protein expression. One noteworthy finding is that although both array and Western analyses showed a down-regulation of CDK2 by MSA, the Western data additionally revealed a reduction of phosphorylated CDK2 (Fig. 5 ⇓ ; Table 4 ⇓ ). This dichotomy clearly reinforces a fundamental limitation of the array technology in that changes beyond the step of gene transcription are not detectable by this method.

Fig. 5.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 5.

Confirmation of array data by Western blot analysis. TRIzol-isolated proteins from PC-3 cells were subjected to immunoblotting using an antibody specific for CHK2, cyclin A, p21WAF1, GADD153, DHFR, CDK2, or CDK1. Signals were normalized to the ones for actin to control for loading variation. The results shown here are representative of that from three similar independent experiments.

View this table:
  • View inline
  • View popup
Table 4

Comparison of expression changes detected by array and Western analyses

The values represent treatment:control ratio.

DISCUSSION

Prostate cancer is the most common cancer diagnosed and the second leading cause of cancer-related deaths in men in the United States. However, little is known about the etiological factors for this disease. Consequently, it is not possible to institute primary intervention strategies to remove the causative agents from the environment. Secondary intervention strategies are, therefore, necessary to reduce the morbidity and mortality of prostate cancer, and selenium intervention has been championed as a viable option. The present study was undertaken to investigate the molecular targets and the signaling pathways underlying the anticancer activity of selenium in prostate.

Previous studies by Sinha and Medina and by Sinha et al. (12 , 13) showed that selenium is able to block cell cycle progression at specific checkpoints, which might be explained by a decrease in CDK2 kinase activity. These experiments were done with mouse mammary tumor cells treated with 50 μm of methylselenocysteine; this concentration of selenium is at least 10 times higher than that found in the circulation under normal physiological condition. For this reason, MSA is a more appropriate agent for in vitro studies. Our results demonstrate that MSA inhibits the growth of prostate cancer cells by cell cycle blockade and apoptosis. We used the GeneChip technology to profile selenium-mediated gene expression changes in a time course experiment. Of a total of 12,000 genes screened, over 2,500 were identified to be responsive to selenium treatment. The shear magnitude of this number is somewhat unexpected. These genes were grouped into early-, intermediate-, and late-response clusters. Because the early-response genes are likely to be more important in initiating the effects of selenium, we focused our attention on them in our follow-up analysis.

Certain key cell cycle regulators are among the early-response genes. On the basis of their altered expression, we propose a number of tentative signaling pathways (in a cartoon format) that might mediate the outcome of cell cycle blockade by selenium. As shown in Fig. 6 ⇓ , selenium treatment increases the expression of p21WAF1, which has dual functions in regulating the activity of CDK/cyclin complexes. Although p21WAF1 is a potent inhibitor of cyclin E/A-dependent CDK1/2, it promotes the assembly and the nuclear translocation of cyclin D-CDK4/6 complexes, leading to an increase in cyclin D-associated kinase activity (21) . However, the induction of p19INK4d by selenium counteracts the latter effect. The p19INK4d protein binds to and inhibits the cyclin D-CDK4/6 complexes, thus releasing p21WAF1 from CDK4 and CDK6. The cooperative action of p19INK4d and p21WAF1 leads ultimately to an inhibition of both cyclin D- and cyclin A/E-dependent kinases. The down-regulation of CDK1, CDK2 and cyclin A by selenium provides an amplified effect on this cascade of events. Complete phosphorylation/inactivation of pRB requires the sequential actions of cyclin D-CDK4/6 and cyclin E-CDK2 (22) . Thus, p19INK4d- and p21WAF1-mediated inhibition of CDK2, CDK4, and CDK6 could result in decreased phosphorylation of pRB. Hypophosphorylated pRB interacts with, and negatively regulates, the activity of E2F transcription factors. Loss of E2F activity prevents the transcription of genes, e.g., DHFR and cyclin A, required for progression into S phase. In addition, although not depicted in Fig. 6 ⇓ , p21WAF1 is able to bind to PCNA and directly inhibit its activity (23) , and interact with E2F subunits and disrupt E2F-CDK-p107 DNA binding complex (24 , 25) . These changes, collectively, are expected to result in a blockade of DNA replication.

Fig. 6.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 6.

Schematic representation of signaling pathways downstream of p19INK4d and p21WAF1 mediating MSA-induced cell cycle blockade (see “Discussion”). The numbers in parentheses indicate the treatment:control-signal ratio detected by array analysis. Up-regulation is denoted by a ratio of ≥2, and down-regulation is denoted by a ratio of ≤0.5. All changes, except the one for p19INK4d, were confirmed by Western blot analysis.

As shown in Fig. 7 ⇓ , the elevated expression of CHK2 by selenium treatment leads to increased phosphorylation of CDC25 proteins, which subsequently bind to 14-3-3 proteins and are exported to the cytoplasm. CDC25 proteins are responsible for removing the inhibitory phosphates from CDK1 and CDK2, allowing them to be activated by CDK activating kinases (26 , 27) . A deficiency in nuclear CDC25 proteins prevents the activation of CDK1 and CDK2 to facilitate cell cycle progression. PP2Cs are phosphatases known to remove the activation phosphates from CDK1 and CDK2. An increased expression of PP2Cs by selenium will essentially lead to less active CDK1 and CDK2. CDK2 causes increased phosphorylation of MCMs either directly or indirectly through CDC7. MCMs are important for transition to S phase. By decreasing the expression of CDK2, CDC7, and MCMs, selenium is able to block DNA synthesis. Selenium can also induce a G2-M block by increasing the expression of the checkpoint protein, RAD9. Furthermore, the expression of GADD153 is known to increase in response to a variety of growth arrest or DNA damage signals (28, 29, 30, 31, 32, 33, 34, 35) . GADD153 plays an essential role in cell cycle control and apoptosis (36) . We recently found that in premalignant human breast cells, selenium induced an 8-fold increase in the expression of the GADD153 gene (8) . Similarly, a 6–14-fold induction (Table 4) ⇓ was also observed in PC-3 cells. In summary, Figs. 6 ⇓ and 7 ⇓ show that different pathways modulated by selenium all converge to block cell cycle progression.

Fig. 7.
  • Download figure
  • Open in new tab
  • Download powerpoint
Fig. 7.

Schematic representation of signaling pathways downstream of CHK2, GADD153, and RAD9 mediating MSA-induced cell cycle blockade (see “Discussion”). The numbers in parentheses indicate the treatment:control-signal ratio detected by array analysis. Up-regulation is denoted by a ratio of ≥2, and down-regulation is denoted by a ratio of ≤0.5.

It is clear that selenium affects not just one key target, but a multitude of targets. In doing so, the impact of selenium is amplified. The diversity of the molecular responses also makes it difficult for transformed cells to escape the inhibitory effect of selenium. A reassuring aspect of our results is the considerable overlap of the selenium-modulated genes or signaling pathways identified in prostate cells with those previously identified in breast cells (8) . The congruency, however, is highlighted against a backdrop of certain differences in cell genotype and methodological issues. The PC-3 human prostate cancer cells are null for p53, whereas the premalignant MCF10AT human breast cells have a functional p53, suggesting that p53 is not required for the action of selenium The 12,000-gene oligonucleotide GeneChip was used in the prostate cell study, whereas the 200-gene membrane-based cDNA array was used in the breast cell study. Despite these differences, similar selenium targets were identified, including GADD153, cyclin A, CDK1, CDK2, CDK4, CDC25, E2Fs, as well as the MAPK/JNK and phosphoinositide-3 kinase pathways, thus lending confidence to the array data. Previous studies of gene expression changes in response to selenium have generally been limited to the analysis of a few or a subset of genes and, therefore, have provided only a very narrow view of the entire landscape. As far as we are aware of, this is the most comprehensive gene expression profiling study on the molecular mechanism of selenium in chemoprevention.

Venkateswaran et al. (37) recently reported that PC-3 cells are not growth inhibited by Se-Met unless they are transfected with a functional androgen receptor. Suffice it to point out that the sensitivity of PC-3 and another androgen-independent prostate cancer cell line, DU-145, to Se-Met has been documented by two other groups of investigators (38 , 39) . Furthermore, Jiang et al. (9) showed that DU-145 cells can be induced to undergo apoptosis by MSA at physiological concentrations. Together with our study, the weight of evidence seems to favor the notion that the responsiveness of prostate cancer cells to selenium is not dependent on the presence of a functional androgen receptor. Although androgen plays a critical role in prostate carcinogenesis, a significant proportion of prostate cancers eventually become androgen-unresponsive and refractory to hormonal therapy. The fact that androgen-unresponsive cells are sensitive to selenium-induced growth inhibition is encouraging because it suggests that selenium intervention may be a viable strategy for preventing prostate cancer recurrence after prostatectomy.

Acknowledgments

We are grateful to Dorothy Donovan, Tamora Loftus, Todd Parsons, Rita Pawlak, Cathy Russin, and Janice Hoffmann for their excellent technical assistance.

Footnotes

  • 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 This work was supported by Grant CA 91990 from the National Cancer Institute, AACR-Cancer Research Foundation of America Fellowship in Prevention Research, Department of Defense Postdoctoral Fellowship Award, and was partially supported by core resources of the Roswell Park Cancer Institute Cancer Center support Grant P30 CA 16056 from the National Cancer Institute.

  • ↵2 To whom requests for reprints should be addressed, at Department of Cancer Prevention, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263. Phone: (716) 845-8875; Fax: (716) 845-8100; E-mail: clement.ip{at}roswellpark.org

  • ↵3 The abbreviations used are: SELECT, Selenium and Vitamin E Chemoprevention Trial; MSA, methylseleninic acid; TUNEL, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling; PI, propidium iodide; GAPDH, glyceraldehyde-3-phosphate dehydrogenase; MTT, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide; TGF, transforming growth factor; RR, relative risk; BrdUrd, bromodeoxyuridine; Se-Met, selenomethionine; SOM, self-organizing map; MAPK, mitogen-activated protein kinase; PCNA, proliferating cell nuclear antigen.

  • Received July 5, 2002.
  • Accepted October 30, 2002.
  • ©2003 American Association for Cancer Research.

References

  1. ↵
    Clark L. C., Combs G. F., Jr., Turnbull B. W., Slate E. H., Chalker D. K., Chow J., Davis L. S., Glover R. A., Graham G. F., Gross E. G., Krongrad A., Lesher J. L., Jr., Park H. K., Sanders B. B., Jr., Smith C. L., Taylor J. R. Effects of selenium supplementation for cancer prevention in patients with carcinoma of the skin. A randomized controlled trial. Nutritional Prevention of Cancer Study Group. J. Am. Med. Assoc., 276: 1957-1963, 1996.
    OpenUrlCrossRefPubMed
  2. ↵
    Hoque A., Albanes D., Lippman S. M., Spitz M. R., Taylor P. R., Klein E. A., Thompson I. M., Goodman P., Stanford J. L., Crowley J. J., Coltman C. A., Santella R. M. Molecular epidemiologic studies within the selenium and vitamin E cancer prevention trial (SELECT). Cancer Causes Control, 12: 627-633, 2001.
    OpenUrlCrossRefPubMed
  3. ↵
    Ip C. Differential effect of dietary methionine on the biopotency of selenomethionine and selenite in cancer chemoprevention. J. Natl. Cancer Inst. (Bethesda), 80: 258-262, 1988.
    OpenUrlAbstract/FREE Full Text
  4. ↵
    Ip C., Hayes C. Tissue selenium levels in selenium-supplemented rats and their relevance in mammary cancer protection. Carcinogenesis (Lond.), 10: 921-925, 1989.
  5. ↵
    Ip C., Ganther H. E. Activity of methylated forms of selenium in cancer prevention. Cancer Res., 50: 1206-1211, 1990.
    OpenUrlAbstract/FREE Full Text
  6. ↵
    Ip C., Hayes C., Budnick R. M., Ganther H. E. Chemical form of selenium, critical metabolites, and cancer prevention. Cancer Res., 51: 595-600, 1991.
    OpenUrlAbstract/FREE Full Text
  7. ↵
    Ip C., Thompson H. J., Zhu Z., Ganther H. E. In vitro and in vivo studies of methylseleninic acid: evidence that a monomethylated selenium metabolite is critical for cancer chemoprevention. Cancer Res., 60: 2882-2886, 2000.
    OpenUrlAbstract/FREE Full Text
  8. ↵
    Dong Y., Ganther H. E., Stewart C., Ip C. Identification of molecular targets associated with selenium-induced growth inhibition in human breast cells using cDNA microarrays. Cancer Res., 62: 708-714, 2002.
    OpenUrlAbstract/FREE Full Text
  9. ↵
    Jiang C., Wang Z., Ganther H., Lu J. Caspases as key executors of methyl selenium-induced apoptosis (anoikis) of DU-145 prostate cancer cells. Cancer Res., 61: 3062-3070, 2001.
    OpenUrlAbstract/FREE Full Text
  10. ↵
    Sinha R., Unni E., Ganther H. E., Medina D. Methylseleninic acid, a potent growth inhibitor of synchronized mouse mammary epithelial tumor cells in vitro.. Biochem. Pharmacol., 61: 311-317, 2001.
    OpenUrlCrossRefPubMed
  11. ↵
    Vistica D. T., Skehan P., Scudiero D., Monks A., Pittman A., Boyd M. R. Tetrazolium-based assays for cellular viability: a critical examination of selected parameters affecting formazan production. Cancer Res., 51: 2515-2520, 1991.
    OpenUrlAbstract/FREE Full Text
  12. ↵
    Sinha R., Medina D. Inhibition of cdk2 kinase activity by methylselenocysteine in synchronized mouse mammary epithelial tumor cells. Carcinogenesis (Lond.), 18: 1541-1547, 1997.
    OpenUrlAbstract/FREE Full Text
  13. ↵
    Sinha R., Kiley S. C., Lu J. X., Thompson H. J., Moraes R., Jaken S., Medina D. Effects of methylselenocysteine on PKC activity, cdk2 phosphorylation and gadd gene expression in synchronized mouse mammary epithelial tumor cells. Cancer Lett., 146: 135-145, 1999.
    OpenUrlCrossRefPubMed
  14. ↵
    Wang Y., Rea T., Bian J., Gray S., Sun Y. Identification of the genes responsive to etoposide-induced apoptosis: application of DNA chip technology. FEBS Lett., 445: 269-273, 1999.
    OpenUrlCrossRefPubMed
  15. ↵
    Kaminski N., Allard J. D., Pittet J. F., Zuo F., Griffiths M. J. D., Morris D., Huang X., Sheppard D., Heller R. A. Global analysis of gene expression in pulmonary fibrosis reveals distinct programs regulating lung inflammation and fibrosis. Proc. Natl. Acad. Sci. USA, 97: 1778-1783, 2000.
    OpenUrlAbstract/FREE Full Text
  16. ↵
    Chen C-R., Kang Y., Massague J. Defective repression of c-myc in breast cancer cells. Proc. Natl. Acad. Sci. USA, 98: 992-999, 2001.
    OpenUrlAbstract/FREE Full Text
  17. ↵
    Dong Y., Asch H. L., Medina D., Ip C., Ip M., Guzman R., Asch B. B. Concurrent deregulation of gelsolin and cyclin D1 in the majority of human and rodent breast cancers. Int. J. Cancer, 81: 930-938, 1999.
    OpenUrlCrossRefPubMed
  18. ↵
    Aliprantis A. O., Yang R. B., Weiss D. S., Godowski P., Zychlinsky A. The apoptotic signaling pathway activated by Toll-like receptor-2. EMBO J., 19: 3325-3336, 2000.
    OpenUrlAbstract/FREE Full Text
  19. ↵
    Kuida K., Haydar T. F., Kuan C. Y., Gu Y., Taya C., Karasuyama H., Su M. S., Rakic P., Flavell R. A. Reduced apoptosis and cytochrome c-mediated caspase activation in mice lacking caspase 9. Cell, 94: 325-337, 1998.
    OpenUrlCrossRefPubMed
  20. ↵
    Brass N., Heckel D., Sahin U., Pfreundschuh M., Sybrecht G. W., Meese E. Translation initiation factor eIF-4γ is encoded by an amplified gene and induces an immune response in squamous cell lung carcinoma. Hum. Mol. Genet., 6: 33-39, 1997.
    OpenUrlAbstract/FREE Full Text
  21. ↵
    Sherr C. J., Roberts J. M. CDK inhibitors: positive and negative regulators of G1-phase progression. Genes Dev., 13: 1501-1512, 1999.
    OpenUrlFREE Full Text
  22. ↵
    Lundberg A. S., Weinberg R. A. Functional inactivation of the retinoblastoma protein requires sequential modification by at least two distinct cyclin-cdk complexes. Mol. Cell. Biol., 18: 753-761, 1998.
    OpenUrlAbstract/FREE Full Text
  23. ↵
    Waga S., Hannon G. J., Beach D., Stillman B. The p21 inhibitor of cyclin-dependent kinases controls DNA replication by interaction with PCNA. Nature (Lond.), 369: 574-578, 1994.
    OpenUrlCrossRefPubMed
  24. ↵
    Delavaine L., La Thangue N. B. Control of E2F activity by p21Waf1/Cip1. Oncogene, 18: 5381-5392, 1999.
    OpenUrlCrossRefPubMed
  25. ↵
    Dimri G. P., Nakanishi M., Desprez P. Y., Smith J. R., Campisi J. Inhibition of E2F activity by the cyclin-dependent protein kinase inhibitor p21 in cells expressing or lacking a functional retinoblastoma protein. Mol. Cell. Biol., 16: 2987-2997, 1996.
    OpenUrlAbstract/FREE Full Text
  26. ↵
    Falck J., Mailand N., Syljuasen R. G., Bartek J., Lukas J. The ATM-Chk2-Cdc25A checkpoint pathway guards against radioresistant DNA synthesis. Nature (Lond.), 410: 842-847, 2001.
    OpenUrlCrossRefPubMed
  27. ↵
    Matsuoka S., Huang M., Elledge S. J. Linkage of ATM to cell cycle regulation by the Chk2 protein kinase. Science (Wash. DC), 282: 1893-1897, 1998.
    OpenUrlAbstract/FREE Full Text
  28. ↵
    Abcouwer S. F., Schwarz C., Meguid R. A. Glutamine deprivation induces the expression of GADD45 and GADD153 primarily by mRNA stabilization. J. Biol. Chem., 274: 28645-28651, 1999.
    OpenUrlAbstract/FREE Full Text
  29. ↵
    Choi A. M., Fargnoli J., Carlson S. G., Holbrook N. J. Cell growth inhibition by prostaglandin A2 results in elevated expression of gadd153 mRNA. Exp. Cell Res., 199: 85-89, 1992.
    OpenUrlCrossRefPubMed
  30. ↵
    Eymin B., Dubrez L., Allouche M., Solary E. Increased gadd153 messenger RNA level is associated with apoptosis in human leukemic cells treated with etoposide. Cancer Res., 57: 686-695, 1997.
    OpenUrlAbstract/FREE Full Text
  31. ↵
    Guyton K. Z., Xu Q., Holbrook N. J. Induction of the mammalian stress response gene GADD153 by oxidative stress: role of AP-1 element. Biochem. J., 314(Pt2): 547-554, 1996.
  32. ↵
    Jeong J. K., Huang Q., Lau S. S., Monks T. J. The response of renal tubular epithelial cells to physiologically and chemically induced growth arrest. J. Biol. Chem., 272: 7511-7518, 1997.
    OpenUrlAbstract/FREE Full Text
  33. ↵
    Johnsson A., Strand C., Los G. Expression of GADD153 in tumor cells and stromal cells from xenografted tumors in nude mice treated with cisplatin: correlations with cisplatin- DNA adducts. Cancer Chemother. Pharmacol., 43: 348-352, 1999.
    OpenUrlCrossRefPubMed
  34. ↵
    Kim R., Ohi Y., Inoue H., Toge T. Taxotere activates transcription factor AP-1 in association with apoptotic cell death in gastric cancer cell lines. Anticancer Res., 19: 5399-5405, 1999.
    OpenUrlPubMed
  35. ↵
    Luethy J. D., Holbrook N. J. Activation of the gadd153 promoter by genotoxic agents: a rapid and specific response to DNA damage. Cancer Res., 52: 5-10, 1992.
    OpenUrlAbstract/FREE Full Text
  36. ↵
    Zinszner H., Kuroda M., Wang X., Batchvarova N., Lightfoot R. T., Remotti H., Stevens J. L., Ron D. CHOP is implicated in programmed cell death in response to impaired function of the endoplasmic reticulum. Genes Dev., 12: 982-995, 1998.
    OpenUrlAbstract/FREE Full Text
  37. ↵
    Venkateswaran V., Klotz L. H., Fleshner N. E. Selenium modulation of cell proliferation and cell cycle biomarkers in human prostate carcinoma cell lines. Cancer Res., 62: 2540-2545, 2002.
    OpenUrlAbstract/FREE Full Text
  38. ↵
    Menter D. G., Sabichi A. L., Lippman S. M. Selenium effects on prostate cell growth. Cancer Epidemiol. Biomark. Prev., 9: 1171-1182, 2000.
    OpenUrlAbstract/FREE Full Text
  39. ↵
    Redman C., Scott J. A., Baines A. T., Basye J. L., Clark L. C., Calley C., Roe D., Payne C. M., Nelson M. A. Inhibitory effect of selenomethionine on the growth of three selected human tumor cell lines. Cancer Lett., 125: 103-110, 1998.
    OpenUrlCrossRefPubMed
View Abstract
PreviousNext
Back to top
Cancer Research: 63 (1)
January 2003
Volume 63, Issue 1
  • Table of Contents
  • About the Cover

Sign up for alerts

View this article with LENS

Open full page PDF
Article Alerts
Sign In to Email Alerts with your Email Address
Email Article

Thank you for sharing this Cancer Research article.

NOTE: We request your email address only to inform the recipient that it was you who recommended this article, and that it is not junk mail. We do not retain these email addresses.

Enter multiple addresses on separate lines or separate them with commas.
Delineation of the Molecular Basis for Selenium-induced Growth Arrest in Human Prostate Cancer Cells by Oligonucleotide Array
(Your Name) has forwarded a page to you from Cancer Research
(Your Name) thought you would be interested in this article in Cancer Research.
CAPTCHA
This question is for testing whether or not you are a human visitor and to prevent automated spam submissions.
Citation Tools
Delineation of the Molecular Basis for Selenium-induced Growth Arrest in Human Prostate Cancer Cells by Oligonucleotide Array
Yan Dong, Haitao Zhang, Lesleyann Hawthorn, Howard E. Ganther and Clement Ip
Cancer Res January 1 2003 (63) (1) 52-59;

Citation Manager Formats

  • BibTeX
  • Bookends
  • EasyBib
  • EndNote (tagged)
  • EndNote 8 (xml)
  • Medlars
  • Mendeley
  • Papers
  • RefWorks Tagged
  • Ref Manager
  • RIS
  • Zotero
Share
Delineation of the Molecular Basis for Selenium-induced Growth Arrest in Human Prostate Cancer Cells by Oligonucleotide Array
Yan Dong, Haitao Zhang, Lesleyann Hawthorn, Howard E. Ganther and Clement Ip
Cancer Res January 1 2003 (63) (1) 52-59;
del.icio.us logo Digg logo Reddit logo Twitter logo CiteULike logo Facebook logo Google logo Mendeley logo
  • Tweet Widget
  • Facebook Like
  • Google Plus One

Jump to section

  • Article
    • Abstract
    • INTRODUCTION
    • MATERIALS AND METHODS
    • RESULTS
    • DISCUSSION
    • Acknowledgments
    • Footnotes
    • References
  • Info & Metrics
  • PDF
Advertisement

Related Articles

Cited By...

More in this TOC Section

  • Abstract LB-086: Genes targeted by drugs and curcumin in a breast carcinogenesis model
  • Abstract LB-094: Regorafenib and sildenafil interact to kill tumor cells
  • Abstract LB-093: Activation of the FGFR1 signaling pathway by the epstein-barr virus-encoded LMP1 promotes aerobic glycolysis and transformation of human nasopharyngeal epithelial cells
Show more Carcinogenesis
  • Home
  • Alerts
  • Feedback
  • Privacy Policy
Facebook  Twitter  LinkedIn  YouTube  RSS

Articles

  • Online First
  • Current Issue
  • Past Issues
  • Meeting Abstracts

Info for

  • Authors
  • Subscribers
  • Advertisers
  • Librarians

About Cancer Research

  • About the Journal
  • Editorial Board
  • Permissions
  • Submit a Manuscript
AACR logo

Copyright © 2021 by the American Association for Cancer Research.

Cancer Research Online ISSN: 1538-7445
Cancer Research Print ISSN: 0008-5472
Journal of Cancer Research ISSN: 0099-7013
American Journal of Cancer ISSN: 0099-7374

Advertisement