Quick viewing(Text Mode)

Inhibition of Casein Kinase 1-Epsilon Induces Cancer-Cell-Selective, PERIOD2-Dependent Growth Arrest Wan Seok Yang* and Brent R Stockwell*†

Inhibition of Casein Kinase 1-Epsilon Induces Cancer-Cell-Selective, PERIOD2-Dependent Growth Arrest Wan Seok Yang* and Brent R Stockwell*†

Open Access Research2008YangVolume and 9, Stockwell Issue 6, Article R92 Inhibition of 1-epsilon induces cancer-cell-selective, PERIOD2-dependent growth arrest Wan Seok Yang* and Brent R Stockwell*†

Addresses: *Department of Biological Sciences, Columbia University, Fairchild Center, Amsterdam Avenue, New York, NY 10027, USA. †Department of Chemistry, Columbia University, New York, NY 10027, USA.

Correspondence: Brent R Stockwell. Email: [email protected]

Published: 2 June 2008 Received: 23 April 2008 Accepted: 2 June 2008 Genome Biology 2008, 9:R92 (doi:10.1186/gb-2008-9-6-r92) The electronic version of this article is the complete one and can be found online at http://genomebiology.com/2008/9/6/R92

© 2008 Yang and Stockwell; licensee BioMed Central Ltd. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Cancer-cell-selective

Casein kinase 1 epsilon drug development is identified as a potential target for developing selective anticancer reagents.

Abstract

Background: are under extensive investigation as targets for drug development. Discovering novel kinases whose inhibition induces cancer-cell-selective lethality would be of value. Recent advances in RNA interference have enabled the realization of this goal.

Results: We screened 5,760 short hairpin RNA clones targeting the kinome to detect human kinases on which cancer cells are more dependent than normal cells. We employed a two- step screening strategy using human sarcoma cell lines and human fibroblast-derived isogenic cell lines, and found that short hairpin RNAs targeting CSNK1E, a clock gene that regulates circadian rhythms, can induce selective growth inhibition in engineered tumor cells. Analysis of gene- expression data revealed that CSNK1E is overexpressed in several cancer tissue samples examined compared to non-tumorigenic normal tissue, suggesting a positive role of CSNK1E in neogenesis or maintenance. Treatment with IC261, a kinase domain inhibitor of casein kinase 1-epsilon (CK1ε), a protein product of CSNK1E, showed a similar degree of cancer-cell-selective growth inhibition. In a search for substrates of CK1ε that mediate IC261-induced growth inhibition, we discovered that knocking down PER2, another clock gene involved in control, rescues IC261- induced growth inhibition.

Conclusion: We identified CK1ε as a potential target for developing anticancer reagents with a high therapeutic index. These data support the hypothesis that circadian clock genes can control the cell cycle and cell survival signaling, and emphasize a central role of CK1ε and PERIOD2 in linking these systems.

Background tively affect cancer cells. In each case, the key is to identify Cancer can be effectively treated using targeted therapy, as target molecules that play a unique role in tumor cells. exemplified by Imatinib [1] or Sorafenib [2]. There are increasing efforts to fulfill the promise of targeted therapy, Genes encoding such target molecules can be discovered by using antibodies, peptides and small molecules that selec- either comparative or functional genomic approaches. Com- parative approaches analyze cytogenetic data, genomic

Genome Biology 2008, 9:R92 http://genomebiology.com/2008/9/6/R92 Genome Biology 2008, Volume 9, Issue 6, Article R92 Yang and Stockwell R92.2

sequences, mRNA expression profiles or proteomic profiles, Here we utilized an RNAi library consisting of shRNAs target- and select target genes or proteins based on differential ing human kinases to find kinases whose inactivation induces expression or mutation status. For example, high-throughput tumor-cell-selective lethality or growth arrest. The initial sequencing of cancer cell genomes identified BRAF [3] and screening was conducted in two sarcoma cell lines; then, a PIK3CA [4] as frequently mutated genes in multiple human series of isogenic cell lines derived from primary fibroblasts tumors. On the other hand, functional approaches involve were used for selecting tumor-cell-specific cytotoxic shRNAs. perturbing cells with agents, such as cDNAs, small RNAs, or We report that knocking down CSNK1E, a clock gene encod- small molecules, and searching for those that induce specific ing casein kinase 1-epsilon (CK1ε), induces tumor-cell-selec- phenotype changes. Subsequent target identification may tive cytotoxicity. Subsequent validation experiments showed lead to the discovery of cancer therapeutic targets. Indeed, that tumor cells depend more on the kinase activity of CK1ε the RAS oncogenes were identified using an expression clon- than normal cells do. The use of a kinase inhibitor specific to ing strategy that searched for human genes that transform the CK1ε revealed that another clock protein, PERIOD2, is a key mouse fibroblast cell line NIH3T3 [5]. of CK1ε and modulates tumor cell growth.

Among the agents used for functional genomic approaches, small RNAs are increasingly appealing, because RNA-inter- Results and discussion ference (RNAi) mediated by small RNAs enables gene silenc- Our RNAi library was made of lentivirus solutions in 384-well ing in mammalian cells. RNAi is a naturally occurring plates. Each well contains lentiviruses harboring expression phenomenon involved in the silencing of genes, which results plasmids encoding a single shRNA that is designed to target a in regulation of gene expression or activation of an antiviral single mRNA. The arrayed library targets 1,006 human genes, defense system [6]. The RNAi pathway involves DICER, including 571 kinases; most of them are protein kinases, while which processes double-stranded RNAs into small RNA other kinases acting on nucleic acids, lipids, and carbohy- duplexes (approximately 22 nucleotides). One strand of the drates are included (Figure 1a). small RNA duplex is incorporated into an effector complex known as the RNA-induced silencing complex (RISC) and We began the screen with two different sarcoma cell lines, acts as a guide molecule in translational repression or mRNA with the goal of pre-selecting shRNAs that are lethal to these cleavage, depending on the degree of base-pair match with tumor-derived cell lines. We infected U-2-OS, osteosarcoma- the target mRNA [7]. derived cells, and HT1080, fibrosarcoma-derived cells, in triplicate and incubated them for three days. This allows time The conserved RNAi pathway is also activated by experimen- for the shRNAs to be expressed, to bind to their target tally designed double-stranded RNAs or short hairpin RNAs mRNAs, and cause a reduction in expression of the encoded (shRNAs), which make it possible to knock down genes of protein, as the protein is turned over. Percent growth inhibi- interest in mammalian cells. Consequently, RNAi libraries tion was determined by adding alamar blue to the culture, targeting large numbers of mRNAs have been generated and and by measuring fluorescence. used for conducting high-throughput, loss-of-function screens in tissue culture systems. For example, RNAi libraries A number of shRNA clones displayed growth inhibitory were used to identify novel tumor suppressors [8,9], regula- effects (Figure 1b,c and Additional data file 1). Statistical anal- tors of cell death and survival [10], and novel components of ysis of primary screening data revealed 195 genes whose p53 signaling [11]. Moreover, RNAi libraries were used for knockdown inhibited growth of either cell lines more than understanding the mechanisms of action of novel compounds 25% (P < 0.01; Figure 1b and Additional data file 1). Fifty-two [12], for characterizing determinants of sensitivity to clini- genes affected cell growth in both cell lines, while other genes cally used drugs [13], and for identifying novel targets for had specific effects on each cell line, which may reflect the dif- anti-cancer therapy, using a pair of isogenic cell lines [14]. ferent tissue origin of these two sarcomas. Some of the hit genes in common between the two cell lines are well-known Isogenic cell lines are useful for discovering therapeutic regulators of the cell cycle or cell survival, but there were nine agents and probing the biology of transformation. They may genes whose functions have not been described (Additional consist of cancer cells at different stages of malignancy, or a data file 1). We were most interested in genes whose functions specific cancer gene can be deleted to create an isogenic cell are most critical to the survival of these two cancer cell lines. line counterpart. Another approach is to isolate primary cells Accordingly, we calculated the sum of the percent growth and induce transformation by sequential addition of onco- inhibition in each cell line and selected seven shRNAs whose genic elements. This system provides a series of genetically summed values were >90% (Figure 1c). Six out of these seven defined cell lines, and thereby allows for identification of genes were among the 52 common hits in Figure 1b. One tumor-cell-selective, or even genotype-selective, lethal gene, PPM1E, was not statistically significant and, as agents. The successful use of such a system has been expected, shRNAs targeting PPM1E were not active in our fol- described for identification of small molecules with poten- low-up analysis (data not shown). tially high therapeutic indices [15].

Genome Biology 2008, 9:R92 http://genomebiology.com/2008/9/6/R92 Genome Biology 2008, Volume 9, Issue 6, Article R92 Yang and Stockwell R92.3

(a) 14 11 Total genes 16 (1,006) 9 19 Protein kinases Nucleobase/side/tide kinases 24 Lipid kinases Other Kinase 29 Carbohydrate kinases genes genes Inositol/phosphoinositol kinases (435) (571) 449 Metabolic kinases Nutrient kinases Miscellaneous

(b) HT1080 U2OS

>25% G.I. (mean) 42 52 101 P < 0.01

(c)

70 60 Top 7 hits Median value of % G.I. in 50 Symbol Sum 40 HT1080 U2OS PRKD2 63.25 51.89 115.14 30 CAMK2G 54.05 55.62 109.67 20 CSNK1E 47.08 60.10 107.18 10 IKBKE % G.I. in U2OS 53.73 43.24 96.97 0 PPM1E 42.10 50.10 92.20 -20-10 0 20 40 60 80 PIK3AP1 50.61 40.58 91.18 -20 CHEK1 45.29 45.69 90.98 -30 % G.I. in HT1080

LentiviralFigure 1 RNAi library screen of human kinases identifies regulators of cancer cell growth Lentiviral RNAi library screen of human kinases identifies regulators of cancer cell growth. (a) Target genes covered by the shRNA library were classified according to gene function using Gene Ontology groups [33]. (b) Two human sarcoma cell lines, HT1080 and U-2-OS cells, were infected with lentiviruses containing shRNAs targeting human kinases in 384-well format. Genes whose knockdown inhibits growth of either cell line by more than 25% compared to control with statistical significance (P < 0.01) were considered as hits. This diagram shows the number of hits specific to each cell line and common to both cell lines. (c) Median value of percent growth inhibition (%GI) from triplicate results in each cell line. The top seven hits were selected based on the summed value of %GI in both cell lines.

Reducing expression of these six target genes causes growth these six genes may affect normal cells to the same degree. To arrest or cell death in two different sarcoma-derived cancer identify shRNA clones that have cancer cell selectivity, we cell lines; however, we were concerned that knockdown of created fresh batches of lentivirus harboring the six shRNA

Genome Biology 2008, 9:R92 http://genomebiology.com/2008/9/6/R92 Genome Biology 2008, Volume 9, Issue 6, Article R92 Yang and Stockwell R92.4

clones and retested them in a pair of nearly isogenic cell lines, level was decreased significantly. This is likely to reflect an BJ-TERT and BJ-TERT/LT/ST/RASV12. Both cell lines were off-target effect of this particular shRNA. derived from primary human BJ foreskin fibroblasts [16]. These BJ primary cells were engineered successively to The CSNK1E gene encodes the CK1ε protein, whose main express the catalytic subunit of human telomerase (hTERT), function is to regulate the circadian rhythm by phosphorylat- the SV40 large T and small T oncoproteins (LT and ST), and ing other clock gene products [18]. The role of CK1ε in cancer an oncogenic allele of HRAS (HRASG12V). We refer to these has been speculated upon, because CK1ε was shown to phos- cells lines as BJ-TERT, BJ-TERT/LT/ST, and BJ-TERT/LT/ phorylate key proteins in cancer signaling pathways, such as ST/RASV12. Only the BJ-TERT/LT/ST/RASV12 cells form p53 [19] and β-catenin [20]. However, the significance of tumors in nude mice. Therefore, testing of shRNAs in BJ- these events in carcinogenesis is not known, TERT and BJ-TERT/LT/ST/RASV12 should enable one to and the possibility of using CK1ε as a pharmacological target identify genes with a function that is essential in tumor cells, for cancer treatment has not been considered. Therefore, we but not normal cells. We measured trypan blue exclusion to analyzed the expression level of CSNK1E in human tumor evaluate the cytotoxic or growth inhibitory effect of these samples to obtain support for its involvement in human can- shRNA clones on normal cells and their isogenic engineered cer. Some genes that are specifically required for tumor main- tumor cell counterparts. tenance are overexpressed in cancer cells over normal cells. We analyzed the gene-expression database Oncomine for dif- Out of these six shRNA clones, five did not show differential ferential expression patterns in normal versus tumor in dif- activity in the two cell lines; however, the shRNA targeting ferent tissue types [21]. The Oncomine database contained CSNK1E (hereafter, shCSNK1E) had a tumorigenic-cell-line- microarray expression data for CSNK1E from ten different specific activity (Figure 2a). The activity of shCSNK1E was tissues, including brain, head and neck, renal, bladder, leuke- further tested in four different BJ-derived cell lines, namely, mia, lung, melanoma, prostate, salivary gland, and semi- BJ-TERT, BJ-TERT/LT/ST, BJ-TERT/LT/ST/RASV12, and noma. Interestingly, all tumor tissues in the database showed DRD cells. DRD cells were engineered to express hTERT, ST, upregulated CSNK1E expression compared to normal tissues, HRASG12V, dominant negative p53, and constitutively active suggesting a positive role of CK1ε in cancer maintenance or -dependent kinase (CDK)4/cyclin D, which inactivates neogenesis (Figure 3). the RB protein [17]. The p53DD/CDK4/cyclin D1 combina- tions substitute for LT. DRD cells are tumorigenic in nude The proliferation-rate-dependent action of shCSNK1E (Fig- mice, which is expected from the fact that they are also ure 2b) raises the possibility that shRNA treatment induces derived from BJ primary cells and the effects of mutations in cell cycle arrest; thus, fast growing cells have greater growth both cell lines should be similar. The growth inhibitory poten- inhibition. To test this hypothesis, we stained the DNA of tial of shCSNK1E increased as the cell doubling time shRNA-treated cells with propidium-iodide and analyzed the decreased, suggesting that the activity is proliferation-rate cell cycle distribution by flow cytometry. The cell cycle distri- dependent rather than genotype dependent (Figure 2b). bution profile indicates that, after expression of shCSNK1E, HT1080 cells were arrested in the second gap (G2) phase of Theoretically, the length of shRNA involved in base paring the cell cycle, with a concomitant increase in the population with the target mRNA is long enough to ensure specificity of of cells harboring less than the normal diploid DNA content the shRNA clone. However, mismatches between a shRNA (that is, sub-G1), implying had occurred (Figure and target mRNAs are tolerable; RISC is able to suppress 4a). expression of off-target mRNAs whose sequences do not per- fectly complement the guide strand of the shRNA [7]. In order The cell cycle is primarily regulated by the activity of to confirm our hypothesis that knocking down expression of and CDKs. Among CDK/cyclin complexes, CDK1-cyclin A CSNK1E is responsible for the observed growth inhibition, we promotes the transition from G2 to (M), while CDK1- tested multiple shRNA clones targeting the CSNK1E gene; cyclin B governs maturation of M phase [22]. We examined each shRNA clone binds to different regions of the CSNK1E whether shCSNK1E treatment affected expression of cyclin mRNA. If more than a single shRNA clone induces growth A2 and cyclin B1 in HT1080 cells. Real-time PCR analysis inhibition, CSNK1E is likely to be the relevant target, because revealed that shCSNK1E decreased mRNA levels of cyclin B1 the probability of a common off-target effect of multiple and cyclin A2 (Figure 4b). In contrast, mRNA levels of cyclin shRNA clones with unrelated sequences is low. We found that D1, whose function is important for the G1 to S transition, four shRNAs targeting CSNK1E induced strong growth inhi- were slightly increased (Figure 4b). These data are consistent bition in HT1080 cells (Figure 2c). The level of CSNK1E with the cell cycle distribution pattern after shCSNK1E treat- mRNA decreased upon expression of these shRNAs, as ment observed by flow cytometry; knocking down CSNK1E assessed by real-time quantitative PCR analysis (Figure 2d). expression caused down-regulation of cyclin B1 and cyclin Note that one of these shRNAs, clone 1838, did not display A2, which results in cell cycle arrest at the G2/M phase. stronger growth inhibition effects even though the mRNA

Genome Biology 2008, 9:R92 http://genomebiology.com/2008/9/6/R92 Genome Biology 2008, Volume 9, Issue 6, Article R92 Yang and Stockwell R92.5

(a) (b)

120 120 BJ-TERT 100 100 BJ-TERT/LT/ ST/RASV12 80

80 iability 60 60 40 40 % Cell v % Cell viability 20 20 0 0

V12 DRD (28 h) IKBKE CHEK1 PRKD2 BJ-TERT (40 h) PIK3AP1 CAMK2G CSNK1E BJ-TERT/LT/ (28 h) ST (15 h) BJ-TERT/LT/ ST/RAS

(c) (d)

1.8 120 1.6 100 1.4 1.2 80 1 60 0.8 mRNA level level mRNA 40 0.6

% Cell viability 0.4 20 quantification) (relative 0.2 0 0 HT1080 Non- 1834 1836 1837 1838 Non- 1834 1836 1837 1838 targeted targeted

CSNK1EFigure 2 is a target for developing anti-cancer drugs with a potentially high therapeutic index CSNK1E is a target for developing anti-cancer drugs with a potentially high therapeutic index. (a) Retesting of six hit shRNA clones in isogenic BJ-derived cell lines. Knocking down CSNK1E induced cancer-cell-specific growth inhibition, whereas knocking down other survival genes did not display differential activity in the two cell lines. The graph is representative of multiple experiments. (b) The activity of shCSNK1E was examined in four isogenic BJ-derived cell lines. The growth inhibitory effect of shCSNK1E was proportional to the cell proliferation rate. The doubling time of each cell line is shown in parentheses. (c) Inhibition of HT1080 cell growth by independent shRNA clones that bind to different regions of CSNK1E mRNA. (d) The knockdown efficiency of each shRNA clone targeting CSNK1E as assessed by quantitative PCR analysis. Error bars in (b-d) indicate one standard deviation of triplicate data.

In addition to G2/M phase cell cycle arrest, shCSNK1E treat- shRNA contained only full length PARP1, whereas those ment induced apoptotic cell death, as evidenced by the treated with shCSNK1E or staurosporine, a known inducer of appearance of small, fragmented cells and a sub-G1 popula- caspase-dependent apoptosis, contained a diagnostic PARP1 tion (Figure 4a). To confirm the apoptotic phenotype of fragment, indicating that apoptotic caspases were activated shCSNK1E-treated cells, we examined cleavage of poly(ADP- by these treatments (Figure 4c). Activation of apoptotic cas- ribose)polymerase-1 (PARP1), which is cleaved by caspases pases was further confirmed by western blot, which detected during apoptosis. Western blot analysis with antibodies spe- the active form of caspase-3 only in shCSNK1E or stau- cific to PARP1 showed that cells treated with a non-targeting

Genome Biology 2008, 9:R92 http://genomebiology.com/2008/9/6/R92 Genome Biology 2008, Volume 9, Issue 6, Article R92 Yang and Stockwell R92.6

CSNK1E Casein kinase 1, epsilon 2.5 2 1.5 1 0.5 0 -0.5 Bone marrow Leuke- Normal Carcino- -1 Normal Cancer Testis Semino- (6) mia(87) (6) ma (16) (14) (42) (14) ma(23) Normal Carcino- -1.5 (48) ma (109) Benign Carcino- (22) -2 Head and ma (30) neck Germ cell Bladder Leukemia Prostate Salivary gland Z-score nomalized expression level nomalized Z-score -2.5 P = 1.1e-5 P = 3.8e-7 P = 4.2e-14 P = 1.3e-8 P = 4.0e-7 P = 9.8e-7

Normalized expression level a Tissues P-value Normal Cancer Brain 0.83 (23) 1.07 (50) 8.40E-06 Head and neck -0.53 (14) -0.22 (42) 1.10E-05 Renal 0.37 (162) 0.76 (16) 6.80E-05 Bladder -0.86 (48) -0.38 (109) 4.20E-14 Leukemia -1.64 (6) 0.55 (87) 1.30E-08 Lung 1.19 (17) 1.6 (20) 1.60E-05 Melanoma 0.61 (7) 0.81 (45) 1.30E-05 Prostate -0.35 (22) 0.73 (30) 4.00E-07 Salivary gland 0.67 (6) 1.58 (16) 9.80E-07 Seminoma 0.54 (6) 0.77 (91) 1.80E-06 a Data represent median value of samples; sample size is indicated in parentheses.

GeneFigure expression 3 studies comparing normal and cancer tissues were analyzed for CSNK1E using Oncomine [21] Gene expression studies comparing normal and cancer tissues were analyzed for CSNK1E using Oncomine [21]. CSNK1E was found to be over-expressed in cancer samples over normal samples regardless of tissue origin. The graph shows representative results of CSNK1E gene expression analysis from six human tissues. The number of samples in each study is provided in parentheses. The y-axis units are based on z-score normalization and the P-value of each set is shown at the bottom of the graph. The upper and lower bands of the box represent the 75th and 25th percentiles, respectively; the upper and lower error bars represent the 90th and 10th percentiles, respectively. The table shows normalized expression levels of CSNK1E in normal and cancer samples from ten human tissues.

Genome Biology 2008, 9:R92 http://genomebiology.com/2008/9/6/R92 Genome Biology 2008, Volume 9, Issue 6, Article R92 Yang and Stockwell R92.7

(a) Non-targeting shRNA shCSNK1E

1,000 400

800 300 600 G1: 66.00% G1: 62.30% 200 Number Number 400 G2: 23.66% G2: 34.00% S: 14.03% 100 A S: 0.00% 200 A

0 0 0 50 100 150 200 250 0 50 100 150 200 250 Channels (FL2-A-FL2 area) Channels (FL2-A-FL2 area) (b)

1.4 1.2 CyclinB1 CyclinA2 2 1.2 CyclinD1 1 1 0.8 1.5 0.8 0.6 0.6 1

mRNA level mRNA 0.4 0.4 0.5 0.2 0.2

(relative quantification) 0 0 0 N.T. 1834 1837 N.T. 1834 1837 N.T. 1834 1837

(c)

M.W. M.W. (kDa) (kDa) Non-targetedshCSNK1EStaurosporine Non-targetedshCSNK1EStaurosporine 150 20

100 Cleaved Cleaved PARP 15 Casp-3 75

FigureKnocking 4 down CSNK1E induces G2/M cell cycle arrest and caspase-mediated apoptosis Knocking down CSNK1E induces G2/M cell cycle arrest and caspase-mediated apoptosis. (a) Two days after non-targeting shRNA or shCSNK1E treatment, HT1080 cells were fixed in methanol and stained with propidium iodide (Materials and methods). Flow cytometry of cells was performed on a FACSCalibur; calculation of cell cycle stages was performed using the cell cycle analysis program Modifit LT. Red area shows cell population in G1 or G2 cell cycle phase, while gray area shows dying cells. Label 'A' denotes apoptotic cell population. Insets show photographs of HT1080 cells treated with non- targeting shRNA or shCSNK1E. (b) Knocking down CSNK1E down-regulates CyclinB1 and CyclinA2. Cellular RNAs were prepared from HT1080 cells infected with either non-targeting shRNA (N.T.) or two different CSNK1E-targeting shRNAs (1834, 1837), and real-time PCR was performed with each gene-specific primer set. The expression levels of CyclinB1, CyclinA2 and CyclinD1 were first normalized to the level of an endogenous control (RPLPO), and then the relative expression level of each gene among the three cell lines was expressed as a ratio of transcripts in a cell line to those in non-targeted shRNA treated cells. Error bars indicate one standard deviation of triplicate data. (c) Knocking down CSNK1E induces caspase activation. Whole cell lysates from HT1080 cells infected with either non-targeting shRNA or shCSNK1E and cells treated with staurosporine were prepared. The cleavage of PARP1 or caspase-3 (Casp-3) in each sample was examined by western blotting using antibodies against PARP1 and cleaved caspase-3.

Genome Biology 2008, 9:R92 http://genomebiology.com/2008/9/6/R92 Genome Biology 2008, Volume 9, Issue 6, Article R92 Yang and Stockwell R92.8

rosporine-treated samples (Figure 4c). Thus, shCSNK1E changed the proliferation rate of HT1080 cells, indicating induces caspase-mediated apoptosis in sensitive cancer cells. that the protective effect of PER2 knockdown on IC261- induced growth inhibition is not caused by slowing cell These results suggest that chemotherapeutic reagents target- growth. It has been shown that a major function of CK1ε in the ing CK1ε may induce growth arrest and apoptosis with some circadian rhythm is to phosphorylate PERIOD2, which drives degree of cancer cell selectivity. To test this hypothesis, we proteosome-mediated degradation of PERIOD2 [27]. In sev- examined the effect of IC261, a kinase inhibitor of CK1ε, in eral independent reports, overexpression of PERIOD2 has cell culture. IC261 was reported to selectively inhibit casein been shown to exert anti-tumor effects in both cell culture kinase 1 compared to other protein kinases, by an ATP-com- and mouse models [26,28,29]. Therefore, treatment with petitive mechanism. Moreover, it showed an order of magni- IC261 is likely to stabilize PERIOD2, which activates the tude greater selectivity for CK1δ and CK1ε over other casein PERIOD2-mediated tumor suppression pathway. kinase 1 isoforms [23]. Treatment with IC261 started to inhibit the growth of HT1080 cells at submicromolar concen- Here, we report the identification of CK1ε as a potential target trations (Figure 5a). When we tested IC261 in BJ-TERT and for developing anticancer reagents. The mammalian CK1 BJ-TERT/LT/ST/RASV12 cells, the sensitivity of BJ-TERT/ family consists of at least seven isoforms (α, β, γ1, γ2, γ3, δ and LT/ST/RASV12 cells was greater than that of BJ-TERT cells, ε), as well as additional splice variants [18]. They share highly which was consistent with the results obtained with conserved kinase domains, but differ significantly in the shCSNK1E (Figures 5b and 2a). These data suggest that inhi- length and primary structure of their amino- and carboxy-ter- bition of the kinase activity of CK1ε is crucial for the observed minal non-catalytic domains, implying that each isoform may growth arrest and apoptosis, as opposed to other functions of play a specific role in regulating biological processes [18]. this protein, such as those mediated by protein-protein Defining isoform-specific functions will aid us in developing interactions. Moreover, as shRNAs targeting CK1δ were not agents with enhanced specificity and reduced off-target effective in suppressing cell growth during primary screening, effects. As the specificity of RNAi agents is potentially high, it the cancer-cell-selective activity of IC261 can likely be attrib- allows us to differentiate among these isoforms, which is chal- uted to its inhibition of CK1ε (Additional data file 1). lenging for some chemical inhibitors.

CK1ε is known to control the circadian rhythm by phosphor- In our screening, knocking down other isoforms of CK1 was ylating clock proteins, such as and CRYPTO- not effective at inducing growth arrest, implying that CK1ε CHROME [24]. These clock proteins are also reported to has a unique function in promoting the integrity and prolifer- regulate the cell cycle, suggesting they have a role in linking ation of tumor cells. The nature of the signaling pathway that the circadian system and the cell cycle machinery [25,26]. CK1ε uses to control cell growth remains elusive, but several Mammalian cells have three isoforms of PERIOD proteins lines of evidence support a positive role of this kinase in onco- and two isoforms of proteins, which are genesis. First, in our gene expression analysis, cancer cells encoded by PER1, PER2, PER3, CRY1 and CRY2 genes, have a high level of CK1ε compared to normal cells, regardless respectively. In order to define the role of each isoform in of the tissue origin, implying that a high level of CK1ε causes CK1ε-mediated growth regulation, we conducted counter- a growth or survival advantage during tumorigenesis (Figure screening with shRNAs targeting these genes to identify sup- 3). Second, in a recent report, enforced expression of myris- pressors of IC261-induced growth inhibition in HT1080 cells. toylated-CK1ε, but not other isoforms, induced colony forma- Knocking down expression of PER1, PER3, CRY1, or CRY2 tion in soft-agar-growing engineered human epithelial cells did not affect growth inhibition by IC261 (Additional data file [30]. Third, deletion of PERIOD2 in mice caused increased 2). However, four different shRNA clones targeting PER2 tumor development upon gamma-radiation, suggesting a suppressed IC261-induced growth inhibition, implying that tumor suppressive role of PERIOD2 [26]. CK1ε is a major PERIOD2 is the most crucial substrate of CK1ε in controlling kinase that phosphorylates and degrades the PERIOD2 pro- cell proliferation (Figure 5c,d). Note that the maximum tein through the proteasome [31]; therefore, it is likely that growth inhibition by IC261 in Figure 5c is smaller than that in CK1ε exerts its oncogenic effect by inhibiting the tumor sup- Figure 5a, though they have similar EC50 values of 0.1 μg/ml. pressive function of PERIOD2. In accordance with this This is because cells have been growing for three days before model, we showed that knocking down PER2 abrogated the being treated with IC261 in order to express shRNAs target- growth inhibitory effect of IC261, a kinase inhibitor of CK1ε ing PER2, whereas in Figure 5a, IC261 was added to culture at (Figure 5c). the time of cell seeding.

As we showed that the proliferation rate of target cells is an Conclusion important determinant of growth inhibition by CSNK1E RNAi libraries and isogenic cell lines make it possible to iden- knockdown (Figure 2b), we measured the proliferation rate of tify target genes and proteins for cancer therapeutic develop- HT1080 cells upon PER2 knockdown using the alamar blue ment. We found that CSNK1E is one such target gene upon assay (Figure 5e). None of the shRNA clones targeting PER2 which cancer cells depend more than normal cells. As kinase

Genome Biology 2008, 9:R92 http://genomebiology.com/2008/9/6/R92 Genome Biology 2008, Volume 9, Issue 6, Article R92 Yang and Stockwell R92.9

(a) (b) 60 60 BJ-TERT BJ-TERT/LT/ST/RASV12 40 40

20 20 % Growth inhibition % Growth % Growth inhibition % Growth 0 0 0.001 0.01 0.1 110 0 0.05 0.1 0.15 0.2 0.25 0.3 [IC261], µg/mL [IC261], µg/mL (c) 40 N.T. per2_538 per2_539 20 per2_541 per2_542

% Growth inhibition % Growth 0 0123 [IC261], µg/mL (d) (e)

1.4 80,000 1.2 1 70,000 0.8 0.6 60,000 0.4 mRNA level mRNA 50,000 0.2 Fluorescence value Fluorescence (relative quantification) (relative 0 (Ex535nm/Em590nm) per2 per2 per2 40,000 N.T. per2 per2 per2 per2 538 539 541 542 N.T. 538 539 541 542

PERIOD2Figure 5 is a key substrate of CK1ε that mediates IC261-induced growth inhibition PERIOD2 is a key substrate of CK1ε that mediates IC261-induced growth inhibition. (a) IC261, a kinase inhibitor of CK1ε, induces growth inhibition in HT1080 cells. (b) IC261 treatment in BJ-derived cell lines showed a similar degree of cancer cell selective growth inhibition as shCSNK1E treatment. (c) Knocking down PER2 in HT1080 cells rescues growth inhibition induced by IC261. HT1080 cells were infected with indicated lentiviruses containing different shRNA clones targeting PER2 (per2_538, per2_539, per2_541, or per2_542). After two days of infection, cells were treated with the indicated concentration of IC261 and percent growth inhibition was determined using alamar blue. Values in (a-c) represent the mean ± standard deviation of triplicate data. (d) Cellular RNAs were prepared from the same set of virus infected cells in (c), and real-time PCR was performed with a PER2-specific primer set to monitor the efficiency of knock down by shRNA clones. (e) Proliferation rate of HT1080 cells infected with the same set of viruses as in (c) was determined using alamar blue assay. Error bars in (d,e) indicate one standard deviation of triplicate data. N.T., non-targeting shRNA clone.

Genome Biology 2008, 9:R92 http://genomebiology.com/2008/9/6/R92 Genome Biology 2008, Volume 9, Issue 6, Article R92 Yang and Stockwell R92.10

inhibitors of CK1ε displayed the same phenotype as shRNA %GI = 100 × (1 - (X - N)/(P - N)) treatments, efforts to develop kinase inhibitors of CK1ε with enhanced potency and selectivity would be valuable. Future where X is values from cells infected with shRNAs, N is the work involving the screening of larger shRNA libraries might values from media only, and P is the values from cells grown reveal additional potential drug targets. without shRNAs.

All experiments were performed in triplicate and median per- Materials and methods cent growth inhibition value was taken for selecting final hits Cell lines to be analyzed. The human fibrosarcoma cell line HT1080 was maintained in Dulbecco's modified Eagle's medium (DMEM) supplemented Follow-up analysis of hit shRNA clones with non-essential amino acids and 10% calf serum. The Virus production human osteosarcoma cell line U-2-OS was grown in McCoy's We used lentiviral plasmids encoding shRNAs targeting 5A medium supplemented with 10% calf serum. BJ-fibrob- CSNK1E (catalog number SHGLY-NM_001894), PER1 (cata- last-derived cell lines were grown in a 4:1 mixture of DMEM log number SHGLY-NM_002616), PER2 (catalog number to M199 supplemented with 15% heat-inactivated fetal bovine SHGLY-NM_003894), PER3 (catalog number SHGLY- serum. Penicillin and streptomycin were used as antibiotics NM_016831), CRY1 (catalog number SHGLY-NM_004075), in all media. All cells were incubated in a tissue culture incu- or CRY2 (catalog number SHGLY-NM_021117). All shRNA ® bator at 37°C in a humidified incubator containing 5% CO2. clones were obtained from Sigma's MISSION shRNA collec- tion (Sigma, St. Louis, MO, USA). Plasmid DNA was purified Lentiviral shRNA library using a HiSpeed Plasmid Midi kit (Qiagen, Valencia, CA, We used a library targeting human kinases for our screening USA, catalog number 12643). On day one, 2 × 106 293T cells that was generated by The RNAi Consortium [32]. Our were seeded in 10 cm tissue culture dishes; on day two, 2.8 μg shRNA library consists of lentivirus solutions in 384-deep- of shRNA-plasmid construct and 2.5 μg of pDelta8.9 and 0.28 well polypropylene plates (Greiner, Monroe, NC, USA, cata- μg of pVSV-G helper plasmids were co-transfected into the log number 781270). The library targets 1,006 human genes, 293T cells using FuGENE® 6 Transfection Reagent (Roche, including kinases, those similar to kinases and some ancillary Indianapolis, IN, USA, catalog number 11-814-443-001); on proteins. The lentivirus in each well contains an expression day three, the medium was replaced with 7.5 ml of viral col- cassette (pLKO.1) encoding a single shRNA clone. On aver- lection media (VCM) that consists of DMEM supplemented age, the library contains five different shRNA clones targeting with penicillin and streptomycin (pen/strep), and 30% each gene and has a typical virus titer range from 107-108 IU Hyclone iFCS (Hyclone, Logan, UT, USA, catalog number (infection unit)/ml. We refer to these plates as virus mother 83007-198); on day four, in the morning, the supernatant plates. containing virus was harvested to empty 50 ml conical tubes and 7.5 ml of fresh VCM was added back to virus producing Primary screening 293T cell monolayer. We harvested and replaced the VCM Assay plates were prepared by seeding 400 U-2-OS or again in the evening; on day five, in the morning, we har- HT1080 cells per well in 40 μl of growth media in black, clear- vested the supernatant and bleached the 293T cell culture. bottom, 384-well plates (Corning Inc., Corning, NY, USA, cat- The collected virus supernatant was filtered through a 0.45 alog number 3712). The next day, 40 μl of virus daughter μm syringe filter (Nalgene, Rochester, NY, USA, catalog plates were prepared by transferring 2 μl of virus stock solu- number 190-9945), aliquoted in 2 ml to the cryovials, and tion from virus mother plates and 4 μl of 10× polybrene solu- stored at -80°C freezer until time of use. tion to 34 μl of cell growth media in 384-well polypropylene plates (Greiner, catalog number 781280). Whole growth Virus infection media in the assay plates were replaced with 40 μl of virus/ We seeded 200,000 target cells on 10 cm tissue culture dishes polybrene/media mixture from the virus daughter plates. and the culture was incubated at 37°C in a CO2 incubator for Then, virus infection was carried out by centrifuging the assay 24 h. The next day, frozen stocks of virus solution were plates for 1.5 h at 2,250 rpm, 37°C and the assay plates were thawed at 37°C for a couple of minutes and polybrene (Sigma, returned to a tissue culture incubator. Three days later, catalog number H9268) was added at a final concentration of alamar blue was added to the assay plates. All liquid handling 8 μg/ml. Culture media was replaced with virus/polybrene was carried out using a Biomek FX AP384 module (Beckman mix and the culture dish was incubated for 2 h with rocking Coulter, Fullerton, CA, USA). Cell viability was measured every 30 minutes. After 2 h, 10 ml of growth media was added using alamar blue (Invitrogen, Carlsbad, CA, USA, catalog to culture dish and the culture was incubated further for 2 number DAL1100); subsequently, percent growth inhibition days before harvesting or treatment of compounds. (%GI) was calculated from the following formula using fluo- rescence intensity values:

Genome Biology 2008, 9:R92 http://genomebiology.com/2008/9/6/R92 Genome Biology 2008, Volume 9, Issue 6, Article R92 Yang and Stockwell R92.11

Retesting shRNA clones in four BJ cell lines were obtained using a FACScalibur flow cytometer (BD Bio- BJ-TERT or BJ-TERT/LT/ST/RASV12 cells were seeded and sciences, San Jose, CA, USA) and CellQuest software (BD infected with lentivirus as described above. After 60 h, Biosciences). infected cells were released with trypsin/EDTA and harvested in 4 ml BJ growth medium. Aliquots of the cell suspension Real-time quantitative PCR were used for determining cell viability by trypan blue assay. Total RNA was extracted using the RNeasy kit (Qiagen, cata- A hit shRNA clone that displayed differential activity between log number 74104) as described in the manufacturer's hand- BJ-TERT and BJ-TERT/LT/ST/RASV12 cells was further book. RNA sample (1 μg) was subject to reverse transcription tested in BJ-TERT, BJ-TERT/LT/ST, BJ-TERT/LT/ST/ reaction using TaqMan® Reverse Transcription Reagents RASV12, or DRD cells using the same method. Trypan blue (Applied Biosystems, Foster City, CA, USA, catalog number staining, taking 100 images of samples, and analysis of the N8080234) according to the manufacturer's instructions. images were carried out automatically by Vi-Cell (Beckman Then, quantitative PCR was carried out using Power SYBR® Coulter). Green PCR Master Mix (Applied Biosystems, catalog number 4367659) and 7300 Real-Time PCR System (Applied Biosys- Monitoring drug sensitivity tems). The primer sequences used for quantitative PCR were: On the day of the experiment, empty 384-deep-well polypro- PER2_F, 5'-GCAAAATCTGAACACAACCC-3'; PER2_R, 5'- pylene plates (Greiner, catalog number 781270) were filled CTTTGTGTGTGTCCACTTTC-3'; CYCLINB1_F, 5'- with 50 μl growth media except for columns 5 and 13, where CTGGCTAAGAATGTAGTCATG-3'; CYCLINB1_R, 5'-GGTA- 100 μl of IC261 solution (100 μg/ml in growth media) was GAGTGCTGATCTTAGC-3'; CYCLINA2_F, 5'-CAGCAGCCT- transferred. IC261 is a kinase inhibitor of CK1ε and was pur- GCAAACTGC-3'; CYCLINA2_R, 5'- chased from Calbiochem, San Diego, CA, USA (catalog GAGGTATGGGTCAGCATC-3'; WEE1_F, 5'-GCATTTAT- number 400090). After IC261 solution transfer, 2-fold dilu- GCCATTAAGCGATC-3'; WEE1_R, 5'-GAGAATGCTGTC- tion series across columns 5-12 and columns 13-20 were done CAAGCAC-3'; CYCLIND1_F, 5'-CTTCGTTGCCCTCTGTGC- by transferring 50 μl of compound solution to the next col- 3'; CYCLIND1_R, 5'-CACCATGGAGGGCGGATTG-3'. umn successively (8-point dilution series) with mixing. We named this plate '10× IC261 plate'. Assay plates were pre- The mRNA level of human acidic ribosomal pared by seeding 1,500 shRNA-infected HT1080 cells per P0 was measured using the following primers and used as a well in 36 μl of growth media to black, clear bottom 384-well reference for quantification: RPLP0 F, 5'-ACGGGTACAAAC- plates. Cells in the assay plates were treated with IC261 in a 2- GAGTCCTG-3'; RPLP0 R, 5'-GCCTTGACCTTTTCAGCAAG- fold dilution series by transferring 4 μl solution from a 10× 3'. IC261 plate. Assay plates were returned to the culture incuba- tor and maintained for 24 h before adding alamar blue. Per- Western blotting cent growth inhibition was calculated using fluorescence Monitoring cleavage of PARP1 and caspase-3 upon shRNA treatment intensity values. We seeded 2 × 106 HT1080 cells in 10 cm dishes and treated them with 1 μM staurosporine for 16 h. Virus containing Alamar blue assay shRNAs targeting CSNK1E was used to infect HT1080 cells After 24 or 48 h of compound treatment, 10 μl of 50% alamar for 48 h. Both dying cells and live cells in each 10 cm dish were blue solution in growth medium was transferred to the assay harvested and collected in the same 15 ml tubes by centrifug- plates, which resulted in 10% final concentration alamar blue. ing cell suspensions at 1,000 rpm for 5 minutes. Cell pellets Plates were incubated further for 16 h to allow reduction of were washed three times with PBS and cells were lysed in 200 alamar blue, which results in the generation of red fluores- μl of denaturing lysis buffer (50 mM HEPES KOH (pH 7.4), cence. The fluorescence intensity was determined using a Vic- 40 mM NaCl, 2 mM EDTA, 1.5 mM Na3VO4, 50 mM NaF, 10 tor 3 plate reader (Perkin Elmer, Waltham, MA, USA) with a mM sodium pyrophosphate, 10 mM sodium β-glycerophos- 535 nm excitation filter and a 590 nm emission filter. phate, 0.5% Triton X-100, and protease inhibitor tablet (Roche, catalog number 11836170001)). Protein content was Cell cycle analysis quantified using a Bio-Rad protein assay reagent (Bio-Rad, HT1080 cells were seeded in 10 cm dishes and were infected Hercules, CA, USA, catalog number 500-00006). Equal with lentivirus-harboring shRNA targeting CSNK1E for 48 h. amounts of protein were resolved on SDS-polyacrylamide The infected cells were harvested, washed once with phos- gels. The electrophoresed proteins were transblotted onto a phate-buffered saline (PBS), and resuspended in 1 ml of ice- PVDF membrane, blocked with 5% milk, and incubated with cold PBS. We transferred 300 μl of PBS-cell suspension to rabbit primary antibodies specific to: PARP1 (Santa Cruz, pre-chilled 15 ml tubes, mixed with 5 ml of ice-cold MeOH, Santa Cruz, CA, USA, catalog number sc-7150); cleaved cas- and incubated at -20°C overnight. Fixed cells were rehy- pase-3 ( Technology, Danvers, MA, USA, cata- drated in PBS for 3 h and then pelleted by centrifugation. log number 9661) overnight at 4°C. The membrane was then Cells were reconstituted in 300 μl of PBS containing 60 μg/ml incubated in IRDye 800 goat anti-rabbit antibody (Li-cor propidium iodide and 50 μg/ml RNase A. Cell cycle profiles Bioscience, Lincoln, NE, USA, catalog number 926-32211) at

Genome Biology 2008, 9:R92 http://genomebiology.com/2008/9/6/R92 Genome Biology 2008, Volume 9, Issue 6, Article R92 Yang and Stockwell R92.12

1:3,000 dilutions for 45 minutes at room temperature. After ence to enhance cancer drug discovery. Nat Rev Drug Discov washing off the unbound antibodies, membranes were 2007, 6:556-568. 7. Kim VN: Small RNAs: classification, biogenesis, and function. scanned using the Odyssey™ Imaging System (Li-cor Mol Cells 2005, 19:1-15. Bioscience). 8. Kolfschoten IG, van Leeuwen B, Berns K, Mullenders J, Beijersbergen RL, Bernards R, Voorhoeve PM, Agami R: A genetic screen identi- fies PITX1 as a suppressor of RAS activity and tumorigenicity. Cell 2005, 121:849-858. 9. Westbrook TF, Martin ES, Schlabach MR, Leng Y, Liang AC, Feng B, Abbreviations Zhao JJ, Roberts TM, Mandel G, Hannon GJ, Depinho RA, Chin L, CDK, cyclin-dependent kinase; CK1ε, casein kinase 1-epsilon; Elledge SJ: A genetic screen for candidate tumor suppressors DMEM, Dulbecco's modified Eagle's medium; GI, growth identifies REST. Cell 2005, 121:837-848. 10. MacKeigan JP, Murphy LO, Blenis J: Sensitized RNAi screen of inhibition; hTERT, catalytic subunit of human telomerase; human kinases and identifies new regulators of LT, SV40 large T oncoprotein; PARP1, poly(ADP- apoptosis and chemoresistance. Nat Cell Biol 2005, 7:591-600. ribose)polymerase-1; PBS, phosphate-buffered saline; RISC, 11. Berns K, Hijmans EM, Mullenders J, Brummelkamp TR, Velds A, Heimerikx M, Kerkhoven RM, Madiredjo M, Nijkamp W, Weigelt B, RNA-induced silencing complex; RNAi, RNA-interference; Agami R, Ge W, Cavet G, Linsley PS, Beijersbergen RL, Bernards R: shCSNK1E, shRNA targeting CSNK1E; shRNA, short hairpin A large-scale RNAi screen in human cells identifies new com- RNA; ST, SV40 small T oncoprotein; VCM, viral collection ponents of the p53 pathway. Nature 2004, 428:431-437. 12. Brummelkamp TR, Fabius AW, Mullenders J, Madiredjo M, Velds A, media. Kerkhoven RM, Bernards R, Beijersbergen RL: An shRNA barcode screen provides insight into cancer cell vulnerability to MDM2 inhibitors. Nat Chem Biol 2006, 2:202-206. 13. Swanton C, Marani M, Pardo O, Warne PH, Kelly G, Sahai E, Authors' contributions Elustondo F, Chang J, Temple J, Ahmed AA, Brenton JD, Downward WSY and BRS conceived the study, designed the experiments, J, Nicke B: Regulators of mitotic arrest and ceramide metab- olism are determinants of sensitivity to Paclitaxel and other analyzed the data, and wrote the manuscript. WSY collected chemotherapeutic drugs. Cancer Cell 2007, 11:498-512. the data. BRS supervised the research. 14. Ngo VN, Davis RE, Lamy L, Yu X, Zhao H, Lenz G, Lam LT, Dave S, Yang L, Powell J, Staudt LM: A loss-of-function RNA interference screen for molecular targets in cancer. Nature 2006, 441:106-110. Additional data files 15. Dolma S, Lessnick SL, Hahn WC, Stockwell BR: Identification of genotype-selective antitumor agents using synthetic lethal The following additional data are available. Additional data chemical screening in engineered human tumor cells. Cancer file 1 displays the growth inhibitory activity of all shRNAs Cell 2003, 3:285-296. used in this study, statistical analysis, and list of hits. Addi- 16. Hahn WC, Counter CM, Lundberg AS, Beijersbergen RL, Brooks MW, Weinberg RA: Creation of human tumour cells with tional data file 2 is a figure showing the results of testing shR- defined genetic elements. Nature 1999, 400:464-468. NAs targeting PER1, PER3, CRY1, and CRY2 for suppressing 17. Hahn WC, Dessain SK, Brooks MW, King JE, Elenbaas B, Sabatini DM, DeCaprio JA, Weinberg RA: Enumeration of the simian virus 40 IC261-induced growth arrest. early region elements necessary for human cell AdditionalGrowthcalClickResultsfor analysis,suppressing here ofinhibitory testingfor data and file IC261-inducedfile list shRNAs activity 12 of hitshits. oftargeting all growthshRNAs PER1 arrestarrest. used, PER3 in this, CRY1 study,, and statisti- CRY2 transformation. Mol Cell Biol 2002, 22:2111-2123. 18. Knippschild U, Gocht A, Wolff S, Huber N, Löhler J, Stöter M: The casein kinase 1 family: participation in multiple cellular proc- Acknowledgements esses in eukaryotes. Cell Signal 2005, 17:675-689. This research of BRS was funded in part by a Career Award at the Scientific 19. Knippschild U, Milne DM, Campbell LE, DeMaggio AJ, Christenson E, Interface from the Burroughs Wellcome Fund, by the Arnold and Mabel Hoekstra MF, Meek DW: p53 is phosphorylated in vitro and in Beckman Foundation and by the National Cancer Institute vivo by the delta and epsilon isoforms of casein kinase 1 and (R01CA097061). We thank David Root and the RNAi Consortium for pro- enhances the level of casein kinase 1 delta in response to viding the lentiviral shRNA stocks for the primary screen. topoisomerase-directed drugs. Oncogene 1997, 15:1727-1736. 20. Amit S, Hatzubai A, Birman Y, Andersen JS, Ben-Shushan E, Mann M, Ben-Neriah Y, Alkalay I: Axin-mediated CKI phosphorylation of beta-catenin at Ser 45: a molecular switch for the Wnt References pathway. Genes Dev 2002, 16:1066-1076. 1. Capdeville R, Buchdunger E, Zimmermann J, Matter A: Glivec 21. Rhodes DR, Kalyana-Sundaram S, Mahavisno V, Varambally R, Yu J, (STI571, imatinib), a rationally developed, targeted antican- Briggs BB, Barrette TR, Anstet MJ, Kincead-Beal C, Kulkarni P, Vara- cer drug. Nat Rev Drug Discov 2002, 1:493-502. mbally S, Ghosh D, Chinnaiyan AM: Oncomine 3.0: genes, path- 2. Wilhelm S, Carter C, Lynch M, Lowinger T, Dumas J, Smith RA, ways, and networks in a collection of 18,000 cancer gene Schwartz B, Simantov R, Kelley S: Discovery and development of expression profiles. Neoplasia 2007, 9:166-180. sorafenib: a multikinase inhibitor for treating cancer. Nat Rev 22. Hunter T, Pines J: Cyclins and cancer. Cell 1991, 66:1071-1074. Drug Discov 2006, 5:835-844. 23. Mashhoon N, DeMaggio AJ, Tereshko V, Bergmeier SC, Egli M, Hoek- 3. Davies H, Bignell GR, Cox C, Stephens P, Edkins S, Clegg S, Teague J, stra MF, Kuret J: Crystal structure of a conformation-selective Woffendin H, Garnett MJ, Bottomley W, Davis N, Dicks E, Ewing R, casein kinase-1 inhibitor. J Biol Chem 2000, 275:20052-20060. Floyd Y, Gray K, Hall S, Hawes R, Hughes J, Kosmidou V, Menzies A, 24. Ko CH, Takahashi JS: Molecular components of the mammalian Mould C, Parker A, Stevens C, Watt S, Hooper S, Wilson R, Jayatilake circadian clock. Hum Mol Genet 2006, 15:R271-R277. H, Gusterson BA, Cooper C, Shipley J, et al.: Mutations of the 25. Matsuo T, Yamaguchi S, Mitsui S, Emi A, Shimoda F, Okamura H: BRAF gene in human cancer. Nature 2002, 417:949-954. Control mechanism of the circadian clock for timing of cell 4. Samuels Y, Wang Z, Bardelli A, Silliman N, Ptak J, Szabo S, Yan H, division in vivo. Science 2003, 302:255-259. Gazdar A, Powell SM, Riggins GJ, Willson JK, Markowitz S, Kinzler 26. Fu L, Pelicano H, Liu J, Huang P, Lee C: The circadian gene KW, Vogelstein B, Velculescu VE: High of mutations of Period2 plays an important role in tumor suppression and the PIK3CA gene in human cancers. Science 2004, 304:554. DNA damage response in vivo. Cell 2002, 111:41-50. 5. Parada LF, Tabin CJ, Shih C, Weinberg RA: Human EJ bladder car- 27. Lowrey PL, Takahashi JS: Mammalian circadian biology: eluci- cinoma oncogene is homologue of Harvey sarcoma virus ras dating genome-wide levels of temporal organization. Annu gene. Nature 1982, 297:474-478. Rev Genomics Hum Genet 2004, 5:407-441. 6. Iorns E, Lord CJ, Turner N, Ashworth A: Utilizing RNA interfer- 28. Hua H, Wang Y, Wan C, Liu Y, Zhu B, Yang C, Wang X, Wang Z, Cornelissen-Guillaume G, Halberg F: Circadian gene mPer2

Genome Biology 2008, 9:R92 http://genomebiology.com/2008/9/6/R92 Genome Biology 2008, Volume 9, Issue 6, Article R92 Yang and Stockwell R92.13

overexpression induces cancer cell apoptosis. Cancer Sci 2006, 97:589-596. 29. Hua H, Wang Y, Wan C, Liu Y, Zhu B, Wang X, Wang Z, Ding JM: Inhibition of tumorigenesis by intratumoral delivery of the circadian gene mPer2 in C57BL/6 mice. Cancer Gene Ther 2007, 14:815-818. 30. Boehm JS, Zhao JJ, Yao J, Kim SY, Firestein R, Dunn IF, Sjostrom SK, Garraway LA, Weremowicz S, Richardson AL, Greulich H, Stewart CJ, Mulvey LA, Shen RR, Ambrogio L, Hirozane-Kishikawa T, Hill DE, Vidal M, Meyerson M, Grenier JK, Hinkle G, Root DE, Roberts TM, Lander ES, Polyak K, Hahn WC: Integrative genomic approaches identify IKBKE as a breast cancer oncogene. Cell 2007, 129:1065-1079. 31. Eide EJ, Woolf MF, Kang H, Woolf P, Hurst W, Camacho F, Vielhaber EL, Giovanni A, Virshup DM: Control of mammalian circadian rhythm by CKIepsilon-regulated proteasome-mediated PER2 degradation. Mol Cell Biol 2005, 25:2795-2807. 32. Moffat J, Grueneberg DA, Yang X, Kim SY, Kloepfer AM, Hinkle G, Piqani B, Eisenhaure TM, Luo B, Grenier JK, Carpenter AE, Foo SY, Stewart SA, Stockwell BR, Hacohen N, Hahn WC, Lander ES, Sabatini DM, Root DE: A lentiviral RNAi library for human and mouse genes applied to an arrayed viral high-content screen. Cell 2006, 124:1283-1298. 33. Beissbarth T, Speed TP: GOstat: find statistically overrepre- sented Gene Ontologies within a group of genes. Bioinformatics 2004, 20:1464-1465.

Genome Biology 2008, 9:R92