заявка
№ US 20120252028
МПК C12N5/09

TARGET GENES FOR CANCER THERAPY

Авторы:
Michael Shtulman Igor B. Roninson SHTULMAN MICHAEL
Все (6)
Номер заявки
13390454
Дата подачи заявки
16.08.2010
Опубликовано
04.10.2012
Страна
US
Дата приоритета
14.12.2025
Номер приоритета
Страна приоритета
Как управлять
интеллектуальной собственностью
Чертежи 
12
Реферат

The invention provides new gene targets for cancer chemotherapy, their use in assays for identifying new small molecule cancer chemotherapeutic agents, methods for inhibiting cancer cell growth comprising contacting a cell with a gene expression blocking agent that inhibits the expression of such genes and methods for therapeutic treatment of cancer in a mammal, comprising administering to the mammal such a gene expression blocking agent. A preferred gene target is coatomer protein zeta-1 subunit (COPZ1).

Формула изобретения

1-10. (canceled)

11. A method for selectively killing tumor cells comprising selectively inhibiting expression or function of coatomer protein zeta-1 subunit (COPZ1) gene or its encoded CopI-ζ1 protein, respectively.

12. The method according to claim 11, wherein the expression of COPZ1 is inhibited by an agent selected from an siRNA, an antisense oligonucleotide, and a ribozyme, wherein the agent selectively targets mRNA encoding CopI-ζ1 protein.

13. The method according to claim 11, wherein the expression of COPZ1 is inhibited by a small molecule that selectively inhibits COPZ1 expression.

14. The method according to claim 11, wherein the function of CopI-ζ1 protein is inhibited by a small molecule that inhibits CopI-ζ1 protein.

15. A method for treating an individual having cancer, comprising selectively inhibiting in the individual expression or function of COPZ1 gene or its encoded CopI-ζ1 protein respectively.

16. The method according to claim 15, wherein the expression of COPZ1 is inhibited by an agent selected from an siRNA, an antisense oligonucleotide, and a ribozyme, wherein the agent selectively targets mRNA encoding CopI-ζ1 protein.

17. The method according to claim 15, wherein the expression of COPZ1 is inhibited by a small molecule that selectively inhibits COPZ1 expression.

18. The method according to claim 15, wherein the function of CopI-ζ1 protein is inhibited by a small molecule that inhibits CopI-ζ1 protein.

19. (canceled)

20. A method for identifying a selective small molecule inhibitor of cancer cell growth comprising:

(a) culturing a mammalian cell comprising a recombinant DNA construct comprising a first reporter gene operatively associated with a COPZ1 promoter and a second reporter gene operatively associated with a COPZ2 promoter in the presence of a test compound;

(b) culturing the mammalian cell in the absence of the test compound;

(c) assaying the cells from (a) and (b) for the expression or activity of the first reporter gene and the second reporter gene, or their encoded proteins; and

(d) identifying the test compound as a selective small molecule inhibitor of cancer cell growth if the expression or activity of the first reporter gene or its encoded protein is inhibited to a greater extent than the expression or activity of the second reporter gene or its encoded protein in cells cultured as in (a), but not in cells cultured as in (b).

21-23. (canceled)

24. A method for identifying a selective small molecule inhibitor of cancer cell growth comprising:

(a) providing purified CopI-ζ1 protein and purified CopI-γ protein in the presence of a test compound to allow an interaction of an assayable magnitude between the purified CopI-ζ1 protein and the purified CopI-γ protein;

(b) providing purified CopI-ζ1 protein and purified CopI-γ protein in the absence of the test compound to allow an interaction of an assayable magnitude between the purified CopI-ζ1 protein and the purified CopI-γ protein;

(c) providing purified CopI-ζ2 protein and purified CopI-γ protein in the presence of the test compound to allow an interaction of an assayable magnitude between the purified CopI-ζ2 protein and the purified CopI-γ protein;

(d) providing purified CopI-ζ2 protein and purified CopI-γ protein in the absence of the test compound to allow an interaction of an assayable magnitude between the purified CopI-ζ2 protein and the purified CopI-γ protein;

(e) assaying the magnitude of the interaction between purified CopI-ζ1 protein and purified CopI-ζ protein in steps (a) and (b);

(f) assaying the magnitude of the interaction between purified CopI-ζ2 protein and purified CopI-γ protein in steps (c) and (d); and

(g) identifying the test compound as a selective inhibitor of CopI-ζ1 protein if the magnitude of the interaction is lesser in step (a) than in step (c), but the magnitude of the interaction in step (b) is not lesser than the magnitude of the interaction in step (d).

25. The method according to claim 24, wherein the purified CopI-ζ1 protein or the purified CopI-γ protein are labeled with a fluorophore suitable for fluorescence resonance energy transfer (FRET), the CopI-ζ2 protein or the purified CopI-γ protein are labeled with a fluorophore suitable for FRET, and the magnitude of the interactions are assayed by FRET.

26-27. (canceled)

Описание

BACKGROUND OF THE INVENTION

[0001]

1. Field of the Invention

[0002]

The invention relates to the discovery of new targets for cancer chemotherapy and to the discovery of new small molecule cancer chemotherapeutics effective against such targets.

[0003]

2. Summary of the Related Art

[0004]

There has been much interest in the identification of genes that are essential for cancer cell growth. Such genes can be used as targets for the treatment of cancer. One approach to identifying such genes utilizes expression selection of Transdominant Genetic Inhibitors (TGIs) that inhibit the growth of carcinoma cells in vitro. TGIs are represented by Genetic Suppressor Elements (GSEs) and small hairpin RNA (shRNA) templates. GSEs are biologically active cDNA fragments that interfere with the function of the gene from which they are derived. GSEs may encode antisense RNA molecules that inhibit gene expression or peptides that interfere with the function of the target protein as dominant inhibitors (Holzmayer et al., 1992; Roninson et al., 1995). shRNA templates are small (19-21 bp) cDNA fragments, cloned into an expression vector in the form of inverted repeats and giving rise upon transcription to shRNAs, which are processed by cellular enzymes into double-stranded RNA duplexes, short interfering RNA (siRNA) that cause degradation of their cDNA target via RNA interference (RNAi) (Boutros and Ahringer, 2008). General strategies for the isolation of biologically active TGIs involves the use of expression libraries that express GSEs or shRNAs derived from either a single gene, or several genes, or all the genes expressed in a cell. These libraries are then introduced into recipient cells, followed by selection for the desired phenotype and the recovery of biologically active GSEs, which should be enriched in the selected cells.

[0005]

Genes that are required for the growth of the recipient cells are expected to give rise to TGIs that would inhibit cell proliferation. Such TGIs can be isolated through negative selection techniques, such as bromodeoxyuridine (BrdU) suicide selection (Stetten et al., 1977). The applicability of this approach to the isolation of growth-inhibitory GSEs was demonstrated by Pestov and Lau (Pestov and Lau, 1994) and Primiano et al. (Primiano et al., 2003). Pestov et al. used an isopropyl-β-thio-galactoside (IPTG)-inducible plasmid expression vector to isolate cytostatic GSEs from a mixture of 19 cDNA clones of murine genes associated with the G0/G1transition, using the BrdU suicide selection protocol. Through this approach, Pestov and Lau found that three of the genes in their mixture gave rise to growth-inhibitory GSEs. Primiano et al. (2003) used a GSE library derived from normalized (reduced-redundance) cDNA of human MCF7 breast carcinoma cells and cloned into an IPTG-inducible retroviral vector to isolate GSEs that allow MDA-MB-231 human breast carcinoma cells to survive BrdU suicide selection. That study yielded biologically active GSEs from 57 human genes, potential targets for breast cancer therapy.

[0006]

There remains a need for the identification of new gene targets for cancer therapy.

BRIEF SUMMARY OF THE INVENTION

[0007]

The invention relates to the discovery of new gene targets for cancer chemotherapy and to the discovery of new small molecule cancer chemotherapeutics effective against such targets. The invention provides new gene targets for cancer chemotherapy, their use in assays for identifying new small molecule cancer chemotherapeutic agents, methods for inhibiting cancer cell growth comprising contacting a cell with a gene expression blocking agent that inhibits the expression of such genes and methods for therapeutic treatment of cancer in a mammal, comprising administering to the mammal such a gene expression blocking agent.

[0008]

In a first aspect, the invention provides a method for identifying a small molecule anti-cancer compound, the method comprising (a) culturing a mammalian cell in the presence of a test compound; (b) culturing the mammalian cell in the absence of the test compound; (c) assaying the cells from (a) and (b) for the expression or activity of a nucleic acid or its encoded protein selected from the group of nucleic acids identified in Table 1; and (d) identifying the test compound as an anti-cancer compound if the expression or activity of the nucleic acid or its encoded protein is greater in cells cultured as in (b) than in cells cultured as in (a). In certain preferred embodiments, the nucleic acid is selected from the nucleic acids identified in Tables 2, 4, 5 and 6. In particularly preferred embodiments, the nucleic acid is selected from the nucleic acids identified in Tables 2 and 6.

[0009]

More generally, in a second aspect, the method provides the use, in an assay for identifying a cancer chemotherapeutic small molecule compound, of a recombinant nucleic acid comprising a nucleic acid selected from the nucleic acids identified in Tables 2 and 6.

[0010]

In a third aspect, the invention provides a method for inhibiting cancer cell growth, comprising inhibiting the expression of a nucleic acid selected from the nucleic acids identified in Tables 2 and 6.

[0011]

In a fourth aspect, the invention provides a method for therapeutically treating a mammal having cancer, comprising administering to the mammal a gene expression blocking agent that inhibits the expression of a nucleic acid selected from the nucleic acids identified in Tables 2 and 6.

[0012]

In a fifth aspect, the invention provides a method for selectively inhibiting the growth of cancer cells comprising selectively inhibiting expression or function of coatomer protein zeta-1 subunit gene (COPZ1) or its encoded CopI-ζ1 protein, respectively.

[0013]

In a sixth aspect, the invention provides a method for identifying a selective small molecule inhibitor or peptide inhibitor of COPZ1 expression comprising: (a) culturing a mammalian cell comprising a recombinant DNA construct comprising a first reporter gene operatively associated with a COPZ1 promoter and a second reporter gene operatively associated with a COPZ2 promoter in the presence of a test compound; (b) culturing the mammalian cell in the absence of the test compound; (c) assaying the cells from (a) and (b) for the expression or activity of the first reporter gene and the second reporter gene, or their encoded proteins; and (d) identifying the test compound as a selective small molecule inhibitor of COPZ1 expression if the expression or activity of the first reporter gene or its encoded protein is inhibited to a greater extent than the expression or activity of the second reporter gene or its encoded protein in cells cultured as in (a), but not in cells cultured as in (b).

[0014]

In a seventh aspect, the invention provides a method for identifying a selective small molecule inhibitor or peptide inhibitor of CopI-ζ1 protein comprising: (a) providing purified CopI-ζ1 protein and purified CopI-ζ1 protein in the presence of a test compound to allow an interaction of an assayable magnitude between the purified CopI-ζ1 protein and the purified CopI-γ protein; (b) providing purified CopI-ζ2 protein and purified CopI-γ protein in the absence of the test compound to allow an interaction of an assayable magnitude between the purified CopI-ζ1 protein and the purified CopI-γ protein; (c) providing purified CopI-ζ2 protein and purified CopI-ζ1 protein in the presence of the test compound to allow an interaction of an assayable magnitude between the purified CopI-ζ2 protein and the purified CopI-γ protein; (d) providing purified CopI-ζ2 protein and purified CopI-γ protein in the absence of the test compound to allow an interaction of an assayable magnitude between the purified CopI-ζ2 protein and the purified CopI-γ protein; (e) assaying the magnitude of the interaction between purified CopI-ζ1 protein and purified CopI-γ protein in steps (a) and (b); (0 assaying the magnitude of the interaction between purified CopI-ζ2 protein and purified CopI-γ protein in steps (c) and (d); and (g) identifying the test compound as a selective inhibitor of CopI-ζ1 protein if the magnitude of the interaction is lesser in step (a) than in step (c), but the magnitude of the interaction in step (b) is not lesser than the magnitude of the interaction in step (d).

[0015]

In an eighth aspect, the invention provides a method for identifying a selective small molecule inhibitor or peptide inhibitor of cancer cell growth, the method comprising providing a computer model in the form of three-dimensional structural coordinates of CopI-ζ1 protein, providing three dimensional structural coordinates of a candidate compound, using a docking program to compare the three dimensional structural coordinates of the CopI-ζ1 protein with the three dimensional structural coordinates of the compound and calculate an energy-minimized conformation of the candidate compound in the CopI-ζ1 protein, and evaluating an interaction between the candidate compound and the CopI-ζ1 protein to determine binding affinity of the compound for the CopI-ζ1 protein, wherein the candidate compound is identified as a compound that selectively inhibits cancer cell growth if it has a binding affinity for the CopI-ζ1 protein site of at least 10 μM.

[0016]

In a ninth aspect, the invention provides a method for determining whether a cancer in an individual is responsive to treatment by selectively inhibiting expression or function of COPZ1 or CopI-ζ1 protein, respectively, comprising obtaining cancer cells from the individual, assaying the expression of COPZ2 and/or mIR-152 in the cancer cells, and determining that the cancer in an individual is responsive to treatment by selectively inhibiting expression or function of COPZ1 or CopI-ζ1 protein, respectively, if the expression of COPZ2 and/or mIR-152 in the cancer cells is lower than in normal cells.

BRIEF DESCRIPTION OF THE DRAWINGS

[0017]

FIG. 1 shows a scheme for shRNA library construction from a normalized cDNA fragment (GSE) library of MCF7 cells.

[0018]

FIG. 2 shows testing of gene targets enriched by shRNA selection for BrdU suicide. Panel A shows the analysis of 22 targets that were enriched by shRNA selection; panel B shows the analysis of 12 targets that were unaffected by BrdU suicide selection.

[0019]

FIG. 3 shows testing of gene targets enriched by GSE selection for BrdU suicide. The analysis was conducted as in FIG. 2. Growth-inhibitory activity of siRNAs was tested in HT1080 fibrosarcoma (A), T24 bladder carcinoma (B), and MDA-MB-231 breast carcinoma cells (C).

[0020]

FIG. 4 shows results of depletion of COPI subunits in PC3 cells by transfection of the corresponding siRNAs. Panel A shows GFP-LC3 localization analyzed by indirect immunofluorescence with anti-GM 130 antibodies. Scale bar 10 μM. Panel B shows GFP-LC3 electrophoretic mobility analyzed in parallel to (A) by immunoblotting with anti-GFP antibody.

[0021]

FIG. 5 shows effects of COPI protein knockdown on growth of tumor and normal cell lines transfected with siRNAs targeting the indicated COPI genes. Bars represents means of 3 independent transfections.

[0022]

FIG. 6 shows results of depletion of the indicated COPI proteins in PC3 and BJ-hTERT cells by siRNA transfection. Bars represents means of 6 independent transfections+/−SD.

[0023]

FIG. 7 shows that expression of COPZ2 gene is downregulated in transformed cell lines. Panel A shows QPCR analysis of expression of the indicated COPI genes in BJ-hTERT cells and tumor cell lines. Bars represents expression relative to BJ-hTERT. Panel B shows QPCR analysis of expression of the indicated COP1 genes in immortalized normal BJ-EN fibroblasts and their transformed derivates. Bars represent expression relative to BJ-EN.

[0024]

FIG. 8 shows expression of COPZ1 and COPZ2 genes in normal tissues and tumor cell lines analyzed by QPCR in (A) indicated normal tissues, (B) a panel of tumor cell lines, (C) melanoma cell lines and normal melanocytes.

[0025]

FIG. 9 shows that overexpression of COPZ2 protects PC3 cells from the growth-inhibitory effect of COPZ1 knockdown. Panel A shows results of immunobloting in lentivirus-transduced PC3 cells, using anti-FLAG, anti-COPZ1 and anti-COPZ2 antibodies. Panel B shows effects of the knockdown of COPI proteins expression with the indicated siRNAs on the proliferation of PC3 cells infected with control vector (PC3-Lenti6-Flag), COPZ1 (PC3-COPZ1-FL) or COPZ2 (PC3-COPZ2-FL) expressing vectors. siRNAs obtained from Qiagen or Thermo Scientific are marked as Q or DH. Bars represent means of 6 independent transfections+/−SD.

[0026]

FIG. 10 shows that simultaneous knockdown of both COPZ1 and COPZ2 inhibits growth of BJ-hTERT fibroblasts. Panel A shows analysis of knockdown efficacy by QPCR. Bars represents expression levels of the COPA, COPZ1 and COPZ2 mRNAs in cells transfected with the indicated siRNAs relative to the cells transfected with control siRNA. Panel B shows effects of the knockdown of COPI proteins expression with the indicated siRNAs on the proliferation of BJ-HTERT cells. Bars represent means of 6 independent transfections+/−SD.

[0027]

FIG. 11 shows that knockdown of COPA and simultaneous knockdown of COPZ1 and COPZ2 in BJ-hTERT cells results in accumulation of autophagosomes and dispersion of Golgi. Panel A shows GFP-LC3 localization analyzed by GFP fluorescence and Golgi analyzed by indirect immunofluorescence with anti-GM130 antibodies. Scale bar 10 μM. Panel B shows GFP-LC3 electrophoretic mobility analyzed in parallel to (A) by immunoblotting with anti-GFP antibody.

[0028]

FIG. 12 shows expression of miR-152 in the indicated tumor cell lines and BJ-HTERT cells measured by QPCR. Bars represent miR-152 expression relative to miR-152 level in BJ-hTERT cells.

DETAILED DESCRIPTION OF THE PREFERRED EMBODIMENTS

[0029]

The invention relates to the discovery of new gene targets for cancer chemotherapy and to the discovery of new small molecule cancer chemotherapeutics effective against such targets. The invention provides new gene targets for cancer chemotherapy, their use in assays for identifying new small molecule cancer chemotherapeutic agents, methods for inhibiting cancer cell growth comprising contacting a cell with a gene expression blocking agent that inhibits the expression of such genes and methods for therapeutic treatment of cancer in a mammal, comprising administering to the mammal such a gene expression blocking agent.

[0030]

The references cited herein reflect the level of knowledge in the art and are hereby incorporated by reference in their entirety. Any conflicts between the teachings of the cited references and the present specification shall be resolved in favor of the latter.

[0031]

The present inventors have used both GSE and shRNA libraries constructed in tetracycline/doxycline-inducible lentiviral vectors, to select for growth-inhibitory TGIs in several types of human tumor cells, using BrdU suicide selection. As described below, this approach has enabled the inventors to select TGIs that are enriched through BrdU suicide selection. Subsequent testing of synthetic siRNAs against a set of genes enriched by this selection confirmed that the majority of these genes are required for cell growth. Some of the selected TGIs are derived from known oncogenes or known positive regulators of cell growth. Other TGIs are derived from known genes that had not been previously implicated in cell growth regulation. Genes that give rise to the isolated TGIs are identified as positive growth regulators of tumor cells. Such genes may therefore be considered as targets for the development of new anticancer drugs.

[0032]

In a first aspect, the invention provides a method for identifying a small molecule anti-cancer compound, the method comprising (a) culturing a mammalian cell in the presence of a test compound; (b) culturing the mammalian cell in the absence of the test compound; (c) assaying the cells from (a) and (b) for the expression or activity of a nucleic acid or its encoded protein selected from the group of nucleic acids identified in Table 1; and (d) identifying the test compound as an anti-cancer compound if the expression or activity of the nucleic acid or its encoded protein is greater in cells cultured as in (b) than in cells cultured as in (a). In certain preferred embodiments, the nucleic acid is selected from the nucleic acids identified in Tables 2, 4, 5 and 6. In particularly preferred embodiments, the nucleic acid is selected from the nucleic acids identified in Tables 2 and 6. In some embodiments the expression or activity of more than one nucleic acid or its encoded protein from the tables is assayed in step (c).

[0033]

More generally, in a second aspect, the method provides the use, in an assay for identifying a cancer chemotherapeutic small molecule compound, of a recombinant nucleic acid comprising a nucleic acid selected from the nucleic acids identified in Tables 2 and 6. For purposes of this aspect of the invention, “a recombinant nucleic acid comprising a nucleic acid selected from” is intended to mean the selected nucleic acid covalently linked to other nucleic acid elements that do not occur in the normal chromosomal locus of the gene. Such other nucleic acid elements may include gene expression elements, such as heterologous promoters and/or enhancers, selectable markers, reporter genes and the like. Preferably, the other nucleic acid elements allow the selected nucleic acid to be expressed in mammalian cells. Such recombinant nucleic acids may frequently be incorporated into a chromosome of the mammalian cell.

[0034]

In a third aspect, the invention provides a method for inhibiting cancer cell growth, comprising inhibiting the expression of a nucleic acid selected from the nucleic acids identified in Tables 2 and 6. In preferred embodiments of this aspect of the invention, such inhibition of expression of the nucleic acid is achieved by contacting the cell with a gene expression blocking agent. For purposes of the invention, “a gene expression blocking agent” is an agent that prevents an RNA transcribed from the nucleic acid from carrying out its normal cellular function, such function being either regulatory, or being translated into a functional protein. Such prevention may be either steric, e.g., by the agent simply binding to the RNA, or may be through the destruction of the bound RNA by cellular enzymes. Representative gene expression blocking agents include, without limitation, antisense oligonucleotides, ribozymes, short interfering RNAs (siRNA), short hairpin RNAs (shRNA), microRNAs (miRNA) and the like.

[0035]

In a fourth aspect, the invention provides a method for therapeutically treating a mammal having cancer, comprising administering to the mammal a gene expression blocking agent that inhibits the expression of a nucleic acid selected from the nucleic acids identified in Tables 2 and 6. Such gene expression blocking agent is administered in a therapeutically effective amount. A therapeutically effective amount is an amount sufficient to reduce or ameliorate signs and symptoms of the cancer, such as cell proliferation or metastasis.

[0036]

The inventors have surprisingly discovered that COPZ1 knockdown selectively kills tumor cells relative to normal cells and the mechanism of this selectivity, which warrants the development of COPZ1-targeting drugs. Such drugs should inhibit the expression or function of COPZ1 but not COPZ2, since the inhibition of both COPZ1 and COPZ2 kills not only tumor but also normal cells. There are several approaches to selective inhibition of COPZ1 preferentially to COPZ2.

[0037]

In a fifth aspect, the invention provides a method for selectively inhibiting the growth of cancer cells comprising selectively inhibiting expression or function of coatomer protein zeta-1 subunit gene (COPZ1) or its encoded CopI-ζ1 protein, respectively. “Selective inhibition of cancer cell growth” means killing or inhibiting the growth of cancer cells without killing or inhibiting the growth of normal cells.

[0038]

In some embodiments, the expression of COPZ1 is inhibited by an agent selected from an siRNA, an antisense oligonucleotide, and a ribozyme, wherein the agent selectively targets mRNA encoding CopI-ζ1 protein. siRNAs and their chemically modified variants are being actively developed for therapeutic applications (Ashihara et al., 2010; Vaishnaw et al., 2010). Related approaches targeting RNA sequences that distinguish COPZ1 from COPZ2 include the use of antisense oligonucleotides (Bennett and Swayze, 2010) and ribozymes (Freelove and Zheng, 2002; Asif-Ullah et al., 2007). In some embodiments, the expression of COPZ1 is inhibited by a small molecule that selectively inhibits COPZ1 expression. The terms “selectively targets” and selectively inhibits” mean that expression of the COPZ1 gene is inhibited, but expression of the COPZ2 gene is not inhibited.

[0039]

In some embodiments, the function of CopI-ζ1 protein is inhibited by a small molecule or peptide that selectively inhibits CopI-ζ1 protein. The term “selectively inhibits CopI-ζ1 protein” means that the small molecule prevents CopI-ζ1 protein from forming CopI-ζ1 protein/CopI-γ protein dimers, to a greater extent than it prevents CopI-ζ2 protein from forming CopI-ζ2 protein/CopI-γ protein dimers.

[0040]

The term “small molecule” means a molecule having a molecular weight of less than about 1500 daltons. The greater extent includes at least 10-fold, at least 20-fold, at least 50-fold and at least 100-fold. A “peptide” is an oligomer of from about 3 to about 50 naturally occurring or modified amino acids, and thus also includes peptidomimetics. Such peptides may be further modified, e.g., by pegylation.

[0041]

In some embodiments, the cancer cells are in the body of an individual. Thus, the invention provides a method for treating an individual having cancer, comprising selectively inhibiting in the individual expression or function of expression or function of COPZ1 gene or its encoded CopI-ζ1 protein, respectively. The method comprises administering to the individual any of the agents discussed above in an effective amount. The term “an effective amount” means an amount sufficient to inhibit cancer cell growth in vivo.

[0042]

In a sixth aspect, the invention provides a method for identifying a selective small molecule inhibitor or peptide inhibitor of COPZ1 expression comprising: (a) culturing a mammalian cell comprising a recombinant DNA construct comprising a first reporter gene operatively associated with a COPZ1 promoter and a second reporter gene operatively associated with a COPZ2 promoter in the presence of a test compound; (b) culturing the mammalian cell in the absence of the test compound; (c) assaying the cells from (a) and (b) for the expression or activity of the first reporter gene and the second reporter gene, or their encoded proteins; and (d) identifying the test compound as a selective small molecule inhibitor of COPZ1 expression if the expression or activity of the first reporter gene or its encoded protein is inhibited to a greater extent than the expression or activity of the second reporter gene or its encoded protein in cells cultured as in (a), but not in cells cultured as in (b). The use of reporter gene/heterologous promoter systems to identify compounds that inhibit specific gene expression has been described previously, for example, in U.S. Pat. No. 7,235,403. A selective small molecule inhibitor of COPZ1 expression is a compound having a molecular weight of less than about 1500 daltons and which inhibits expression of the COPZ1 gene, but not the COPZ2 gene. A peptide is as described previously. A test compound can be a small molecule or a peptide. The term “inhibited to a greater extent” includes extents of at least 10-fold, at least 20-fold, at least 50-fold and at least 100-fold.

[0043]

The selective small molecule inhibitors or peptide inhibitor of COPZ1 expression selectively inhibit cancer cell growth. Thus, this method is also a method for identifying a selective small molecule or peptide inhibitor of cancer cell growth. “Selective inhibition of cancer cell growth” means that the compound kills or inhibits the growth of cancer cells without killing or inhibiting the growth of normal cells.

[0044]

In a seventh aspect, the invention provides a method for identifying a selective small molecule inhibitor or peptide inhibitor of CopI-ζ1 protein comprising: (a) providing purified CopI-ζ1 protein and purified CopI-γ protein in the presence of a test compound to allow an interaction of an assayable magnitude between the purified CopI-ζ1 protein and the purified CopI-γ protein; (b) providing purified CopI-ζ1 protein and purified CopI-γ protein in the absence of the test compound to allow an interaction of an assayable magnitude between the purified CopI-ζ1 protein and the purified CopI-γ protein; (c) providing purified CopI-ζ2 protein and purified CopI-γ protein in the presence of the test compound to allow an interaction of an assayable magnitude between the purified CopI-ζ2 protein and the purified CopI-γ protein; (d) providing purified CopI-ζ2 protein and purified CopI-γ protein in the absence of the test compound to allow an interaction of an assayable magnitude between the purified CopI-ζ2 protein and the purified CopI-γ protein; (e) assaying the magnitude of the interaction between purified CopI-ζ1 protein and purified CopI-γ protein in steps (a) and (b); (f) assaying the magnitude of the interaction between purified CopI-ζ2 protein and purified CopI-γ protein in steps (c) and (d); and (g) identifying the test compound as a selective inhibitor of CopI-ζ1 protein if the magnitude of the interaction is lesser in step (a) than in step (c), but the magnitude of the interaction in step (b) is not lesser than the magnitude of the interaction in step (d).

[0045]

An interaction between CopI-ζ1 protein and CopI-γ protein, or between CopI-ζ1 protein and CopI-γ protein, can involve either CopI-γ1 protein or CopI-γ2 protein. The interaction results in formation of an active coatomer protein complex.

[0046]

In some embodiments, the purified CopI-ζ1 protein or the purified CopI-γ protein are labeled with a fluorophore suitable for fluorescence resonance energy transfer (FRET), the CopI-ζ2 protein or the purified CopI-γ protein are labeled with a fluorophore suitable for FRET, and the magnitude of the interactions are assayed by FRET. In some embodiments, the CopI-ζ1 protein and the CopI-ζ2 protein are labeled with a different fluorophore, thereby allowing the assays to take place simultaneously in the same vessel. The use of FRET to assay protein-protein interactions has been described, for example, in Boute et al., 2002; Degorce et al., 2009.

[0047]

A “selective small molecule inhibitor or peptide inhibitor of CopI-ζ1 protein” is a molecule that prevents CopI-ζ1 protein from forming CopI-ζ1 protein/CopI-γ protein dimers, to a greater extent than it prevents CopI-ζ2 protein from forming CopI-ζ2 protein/CopI-γ protein dimers. The term “small molecule” means a molecule having a molecular weight of less than about 1500 daltons. A peptide is as described previously. The greater extent includes at least 10-fold, at least 20-fold, at least 50-fold and at least 100-fold.

[0048]

The selective small molecule inhibitors or peptide inhibitors of CopI-ζ1 protein selectively inhibit cancer cell growth. Thus, this method is also a method for identifying a selective small molecule inhibitor or peptide inhibitor of cancer cell growth. “Selective inhibition of cancer cell growth” means that the compound kills or inhibits the growth of cancer cells without killing or inhibiting the growth of normal cells.

[0049]

In an eighth aspect, the invention provides a method for identifying a selective small molecule inhibitor or peptide inhibitor of cancer cell growth, the method comprising providing a computer model in the form of three-dimensional structural coordinates of CopI-ζ1 protein, providing three dimensional structural coordinates of a candidate compound, using a docking program to compare the three dimensional structural coordinates of the CopI-ζ1 protein with the three dimensional structural coordinates of the compound and calculate an energy-minimized conformation of the candidate compound in the CopI-ζ1 protein, and evaluating an interaction between the candidate compound and the CopI-ζ1 protein to determine binding affinity of the compound for the CopI-ζ1 protein, wherein the candidate compound is identified as a compound that selectively inhibits cancer cell growth if it has a binding affinity for the CopI-ζ1 protein site of at least 10 μM. The solution structure of CopI-ζ1 protein has been described by Yu et al., 2009.

[0050]

siRNAs or other RNA-targeting drugs, inhibitors of COPZ1 expression, and molecules identified in cell-free assays (such as FRET) or predicted by computer modeling to be selective inhibitors of CopI-ζ1 function can be further tested for the expected biological effects in tumor cells. These effects include inhibition of cell proliferation, induction of cell death, disruption of Golgi and inhibition of autophagy. COPZ1-specific inhibitors inducing such biological effects in tumor cells can be considered as therapeutic candidates for further development.

[0051]

In a ninth aspect, the invention provides a method for determining whether a cancer in an individual is responsive to treatment by selectively inhibiting expression or function of COPZ1 or CopI-ζ1 protein, respectively, comprising, obtaining cancer cells from the individual, assaying the expression of COPZ2 and/or mIR-152 in the cancer cells, and determining that the cancer in an individual is responsive to treatment by selectively inhibiting expression or function of COPZ1 or CopI-ζ1 protein, respectively, if the expression of COPZ2 and/or mIR-152 in the cancer cells is lower than in normal cells. The expression level in normal cells may be measured from any normal cell, meaning a cell that is not neoplastically transformed. Alternatively, a standardized signal may be provided as a surrogate for normal cell expression. Such expression may be at least 10-fold greater, at least 20-fold greater, at least 50-fold greater or at least 100-fold greater.

[0052]

In the methods for treatment according to of the invention, the gene expression blocking agent may be formulated with a physiologically acceptable carrier, excipient, or diluent. Such physiologically acceptable carriers, excipients and diluents are known in the art and include any agents that are not physiologically toxic and that do not interfere with the function of the gene expression blocking agent. Representative carriers, excipients and diluents include, without limitation, lipids, salts, hydrates, buffers and the like.

[0053]

Administration of the gene expression blocking agents or formulations thereof may be by any suitable route, including, without limitation, parenteral, mucosal, transdermal and oral administration.

Tables

[0054]

[0000]

Genes giving rise to shRNA sequences enriched by BrdU selection in MDA-MB-231 cells. “Selection to infection ratio” is the
number of sequence reads for the corresponding gene in the sample from BrdU-selected cells relative to the sample from infected
unselected cells. The “enrichment factor” is the “selection to infection ratio” multiplied by the number of different
shRNA sequences for a given gene found in the BrdU-selected sample.
# ofSelection
differentto
shRNAinfectionEnrichment
Unigene_IDGene nameAnnotationsequencesratiofactor
Hs#S4268117C22orf16Chromosome 22 open reading frame 161159.76159.76
Hs#S21296015EPPB9B9 protein1130.04130.04
Hs#S29765129Transcribed locus1107.75107.75
Hs#S4616930RNF121Possibly chimeric cluster, Homo sapiens similar to335.58106.74
Tmem120b protein (LOC100133827),
mRNA.
Hs#S18251876TFCP2L1Ring finger protein 1211104.03104.03
Hs#S2334967ABCA1Transcription factor CP2-like 1194.7494.74
Hs#S546732PDSS2ATP-binding cassette, sub-family A (ABC1),192.8992.89
member 1
Hs#S2294640244.5889.17
Hs#S1642954KRASHomo sapiens v-Ki-ras2 Kirsten rat sarcoma viral243.2286.45
oncogene homolog (KRAS)
Hs#S3989858B3GNT1Prenyl (decaprenyl) diphosphate synthase, subunit 2181.7481.74
Hs#S1731268LOC93622Transcribed locus178.0278.02
Hs#S21293769CDNA FLJ31789 fis, clone NT2R12008656178.0278.02
Hs#S4401294LOC283575176.1076.10
Hs#S16818580Transcribed locus234.5969.18
Hs#S4619097HIST1H3DChromosome 20 open reading frame 3611.1266.71
Hs#S24527772DTD1CDNA FLJ40638 fis, clone THYMU2016113165.2365.23
Hs#S4367439Transcribed locus163.2563.25
Hs#S4060493UDP-GlcNAc:betaGal beta-1,3-N-161.3061.30
acetylglucosaminyltransferase 1
Hs#S4141000Hypothetical gene supported by BC047417320.1960.57
Hs#S2550927Hypothetical protein BC006130159.4559.45
Hs#S5978514ZNF33ATranscribed locus159.4559.45
Hs#S4613519SSSCA1Hypothetical protein LOC146346153.3753.37
Hs#S3521640LOC339929Hypothetical LOC401397153.3753.37
Hs#S14802153GLIS3Transcribed locus, strongly similar to148.4348.43
XP_001064372.1 similar to WW domain-containing
adapter with a coiled-coil region isoform 3 [Rattus
norvegicus]
Hs#S16884664LOC146346Adaptor-related protein complex 1, gamma 1 subunit224.1248.23
Hs#S33939805LOC41397Solute carrier family 23 (nucleobase transporters),316.0648.18
member 2
Hs#S4773912CDC42 small effector 2223.7247.44
Hs#S10817784ANTXR2Ubiquitin associated domain containing 1146.4546.45
Hs#S33760083Hypothetical protein LOC283575146.4446.44
Hs#S16818255SH3-domain binding protein 4315.4546.36
Hs#S21275669C1orf66Homo sapiens actin, beta (ACTB), mRNA315.3245.96
Hs#S18152358UBADC1Obscurin-like 1411.4845.92
Hs#S3993852Ring finger protein 187314.8344.48
Hs#S4613056SYTL1Chromosome 14 open reading frame 43314.5443.63
Hs#S3470133Niemann-Pick disease, type C1314.5043.49
Hs#S2140044KRASHistone cluster 1, H3d142.7342.73
Hs#S26336067DHX57DEAH (Asp-Glu-Ala-Asp/His) box polypeptide 57142.5042.50
Hs#S19132849LOC254571Transcribed locus, weakly similar to140.5240.52
XP_001136957.1 G protein-coupled receptor 175
isoform 1 [Pan troglodytes]
Hs#S17512792C1orf27BTB (POZ) domain containing 9220.2640.52
Hs#S16819291Ubiquitin family domain containing 1313.3740.12
Hs#S16906232PNAS-130139.5339.53
Hs#S2366249Solute carrier family 35, member E1219.6039.20
Hs#S37583284D-tyrosyl-tRNA deacylase 1 homolog (S. cerevisiae)139.0139.01
Hs#S16883204UCHL5Non-SMC condensin I complex, subunit D257.7438.71
Hs#S4284301FBXL2Cleavage and polyadenylation specific factor 4,312.7738.32
30 kDa
Hs#S48391500LOC44297BCL2-associated athanogene219.1138.22
Hs#S5931131Methylenetetrahydrofolate dehydrogenase (NADP+312.7138.12
dependent) 1, methenyltetrahydrofolate
cyclohydrolase, formyltetrahydrofolate synthetase
Hs#S31785054LOC1133827Ubiquitin carboxyl-terminal hydrolase L5138.0538.05
Hs#S18152367TMTC1F-box and leucine-rich repeat protein 2137.7637.76
Hs#S1731541C7orf1Similar to cytoplasmic beta-actin137.5637.56
Hs#S41445089Transcribed locus136.5736.57
Hs#S11046986IQ motif containing E218.2836.57
Hs#S22668594FXC1Transmembrane and tetratricopeptide repeat135.5835.58
containing 1
Hs#S2445725RHOUChromosome 7 open reading frame 10135.5835.58
Hs#S2652760MR1CDNA FLJ34848 fis, clone NT2NE2011684, weakly134.5934.59
similar to H. sapiens mRNA for plakophilin 2a and b
Hs#S5468493SEMA4GFracture callus 1 homolog (rat)134.2034.20
Hs#S39298991Ras homolog gene family, member U133.6033.60
Hs#S2294357GPATCH1G patch domain containing 1132.7832.78
Hs#S15970758LOC9661Hypothetical gene LOC96610131.9631.96
Hs#S16820105NDUFS5Cyclin M4215.9431.87
Hs#S226144LOC4443NADH dehydrogenase (ubiquinone) Fe—S protein 5,131.6331.63
15 kDa (NADH-coenzyme Q reductase)
Hs#S1732011NPSimilar to Phosphoglycerate mutase 1131.6331.63
(Phosphoglycerate mutase isozyme B) (PGAM-B)
(BPG-dependent PGAM 1)
Hs#S1728763SLC35E2Nucleoside phosphorylase131.3631.36
Hs#S15631764Solute carrier family 35, member E2130.6430.64
Hs#S34122828KRT39S-adenosylhomocysteine hydrolase47.5130.05
Hs#S4614963BRWD1129.7229.72
Hs#S34548570Zinc finger protein 33A129.7229.72
Hs#S4262094Sjogren's syndrome/scleroderma autoantigen 1129.7229.72
Hs#S3850280Transcribed locus129.6529.65
Hs#S5495022TMCO4Transmembrane and coiled-coil domains 4129.6529.65
Hs#S14802446Homo sapiens heat shock protein 90 kDa alpha214.8329.65
(cytosolic), class
B member 1 (HSP90AB1)
Hs#S2649948ICT1Nuclear cap binding protein subunit 1, 80 kDa214.8329.65
Hs#S4521257KIF2A129.1629.16
Hs#S1824400SCARNA12Homo sapiens CD9 molecule (CD9)214.5028.99
Hs#S3219330Structure specific recognition protein 147.2328.90
Hs#S1368502Homo sapiens immature colon carcinoma transcript128.7828.78
1 (ICT1)
Hs#S15644384AK3Kinesin heavy chain member 2A128.7328.73
Hs#S16885877ITFG1Small Cajal body-specific RNA 12128.6628.66
Hs#S2293257AFG3L1Transcribed locus128.6628.66
Hs#S16819731Transcribed locus128.1728.17
Hs#S17878167DLC1Adenylate kinase 3127.9227.92
Hs#S15841460Hypothetical protein LOC339929127.8727.87
Hs#S4838923GLIS family zinc finger 3127.8727.87
Hs#S16886550Integrin alpha FG-GAP repeat containing 1127.6727.67
Hs#S16886203Cyclin D155.4627.30
Hs#S4613863Similar to CG17293-PA213.5927.18
Hs#S1474084TMEM25Thyroid hormone receptor, alpha (erythroblastic38.9926.96
leukemia viral (v-erb-a) oncogene homolog, avian)
Hs#S20091283AGPSAFG3 ATPase family gene 3-like 1 (S. cerevisiae)126.8526.85
Hs#S3973098FTH1Transcribed locus126.6926.69
Hs#S4618434Deleted in liver cancer 1126.6926.69
Hs#S927697LOC729334126.0126.01
Hs#S3478912ZNF292Anthrax toxin receptor 2126.0126.01
Hs#S16819818CDNA FLJ25559 fis, clone JTH02834126.0126.01
Hs#S1615068Transmembrane protein 25125.9825.98
Hs#S1727203Glutamyl-prolyl-tRNA synthetase212.6525.30
Hs#S16884987AP1G1Alkylglycerone phosphate synthase125.1025.10
Hs#S2801073CDC42SE2Aprataxin212.4524.91
Hs#S24303272Ferritin, heavy polypeptide 1124.7124.71
Hs#S4084609TOR1ATranscribed locus, strongly similar to124.7124.71
XP_001155109.1 similar to Cks1 protein
homologue isoform 2 [Pan troglodytes]
Hs#S14863546Similar to ribosomal protein S6 kinase, polypeptide 1124.7124.71
Hs#S17512415Zinc finger protein 292124.7124.71
Hs#S29633556GPR143Myosin phosphatase-Rho interacting protein38.1524.46
Hs#S32436017BCDIN3Transcribed locus124.3824.38
Hs#S4613272MANEALZinc finger and BTB domain containing 3846.0724.29
Hs#S4283880UGT2A1124.2124.21
Hs#S3987156ZNF67Vacuolar protein sorting 11 homolog (S. cerevisiae)212.0824.16
Hs#S34550091Chromosome 1 open reading frame 66124.1524.15
Hs#S24303058ARTN124.1324.13
Hs#S4620111LOC3936CDNA FLJ43227 fis, clone HCHON2000212123.7223.72
Hs#S3282887Torsin family 1, member A (torsin A)123.7223.72
Hs#S16507499MTIF3CDNA FLJ40436 fis, clone TESTI2039613123.7223.72
Hs#S1362510ESCO2Transcribed locus123.7223.72
Hs#S2650087CNOT4Homo sapiens tripartite motif-containing 2937.9123.72
(TRIM29)
Hs#S2943708NBPF1G protein-coupled receptor 143123.6023.60
Hs#S29689535MAP3K3Cysteine-rich protein 1 (intestinal)37.8423.51
Hs#S39704889GOLGA5ATP-binding cassette, sub-family C (CFTR/MRP),37.8223.46
member 3
Hs#S16887890CSNK2A1Bin3, bicoid-interacting 3, homolog (Drosophila)122.7322.73
Hs#S1728947Transcribed locus211.3722.73
Hs#S1728579Ataxin 7-like 345.5822.33
Hs#S20091302LOC283398CDNA FLJ26579 fis, clone LNF06863122.2922.29
Hs#S4566561Synaptotagmin-like 1122.2922.29
Hs#S4189502BRD7122.2922.29
Hs#S34542736C1orf17Homo sapiens mannosidase, endo-alpha-like211.1522.29
(MANEAL)
Hs#S38981964LOC653519Neuroblastoma breakpoint family, member 1122.0722.07
Hs#S1367624RP5-86Pleckstrin homology domain containing, family G211.0422.07
F19.3(with RhoGef domain) member 2
Hs#S11057621Transportin 2 (importin 3, karyopherin beta 2b)37.3021.90
Hs#S36352307PNPOMitogen-activated protein kinase kinase kinase 3121.7421.74
Hs#S2139023IKBKBGolgi autoantigen, golgin subfamily a, 5121.7421.74
Hs#S3220225Homo sapiens casein kinase 2, alpha 1 polypeptide121.7421.74
(CSNK2A1)
Hs#S910697BTBD9Transcribed locus121.7421.74
Hs#S16887487TUBB1BCL2-like 137.2421.71
Hs#S1728391LOC427Family with sequence similarity 120A54.2821.41
Hs#S3218921Transcribed locus121.4121.41
Hs#S848381Similar to Succinyl-CoA ligase [GDP-forming] beta-121.4121.41
chain, mitochondrial precursor (Succinyl-CoA
synthetase, betaG chain) (SCS-betaG) (GTP-specific
succinyl-CoA synthetase beta subunit)
Hs#S4300247SLC35E1121.0821.08
Hs#S19675751TIAM1Bromodomain containing 7121.0821.08
Hs#S4618683OSTM1Chromosome 1 open reading frame 170120.7620.76
Hs#S1368546Similar to G protein-coupled receptor 89120.7620.76
Hs#S2544320GPR161KIAA1442 protein120.7620.76
Hs#S4283612BAG1Transcribed locus120.7620.76
Hs#S38872165LOC285359Pyridoxamine 5′-phosphate oxidase120.5820.58
Hs#S1731363LOC28598Inhibitor of kappa light polypeptide gene enhancer in120.5120.51
B-cells, kinase beta
Hs#S19862928UPF3BHypothetical protein LOC254571120.4320.43
Hs#S2233274TBKBP1Chromosome 1 open reading frame 27120.4320.43
Hs#S2330604RPS15ATranscribed locus120.4320.43
Hs#S34544442CDNA: FLJ21228 fis, clone COL00739120.4320.43
Hs#S3506802Transcribed locus120.3620.36
Hs#S4622649Tubulin, beta 1120.2120.21
Hs#S4622710TPP1Transcribed locus, weakly similar to120.1020.10
XP_001131248.1 hypothetical protein [Homo
sapiens]
Hs#S5517244ZNF319CDNA clone IMAGE: 5736961120.0620.06
Hs#S16818069UCP2T-cell lymphoma invasion and metastasis 1119.5719.57
Hs#S18928169Osteopetrosis associated transmembrane protein 1119.5719.57
Hs#S2935335NLRX1Transcribed locus119.5219.52
Hs#S19132577LOC644588G protein-coupled receptor 161119.5219.52
Hs#S27877558Ankyrin repeat and SOCS box-containing 129.7019.41
Hs#S26153400EPORCTP synthase29.6619.33
Hs#S19132894Phosducin-like 3 pseudogene119.0319.03
Hs#S1729449Lamin B244.7318.91
Hs#S4554552GPRC5CHypothetical protein LOC285908118.7818.78
Hs#S4426223IQCEUPF3 regulator of nonsense transcripts homolog B118.7818.78
(yeast)
Hs#S23881883LARP5TBK1 binding protein 1118.7818.78
Hs#S21106926Dual specificity phosphatase 1429.3618.71
Hs#S4399524Ribosomal protein S15a29.2918.58
Hs#S7110390LOC44286Transcribed locus118.5318.53
Hs#S19765522G protein-coupled receptor, family C, group 5,118.4518.45
member C
Hs#S24663340PCMT1La ribonucleoprotein domain family, member 5118.2818.28
Hs#S18601684Transcribed locus, strongly similar to118.2818.28
XP_001080976.1 similar to microfibrillar-associated
protein 1 [Rattus norvegicus]
Hs#S38688562LOC162632CDNA FLJ41419 fis, clone BRHIP2002339117.9917.99
Hs#S38981978TAF15 RNA polymerase II, TATA box binding35.9617.89
protein (TBP)-associated factor, 68 kDa
Hs#S4083130Similar to ribosomal protein L10117.7917.79
Hs#S2293359SLC25A24Cytidine deaminase35.9317.79
Hs#S24303314PYGO2Transcribed locus117.7917.79
Hs#S2011438TERF2Protein-L-isoaspartate (D-aspartate) O-117.7317.73
methyltransferase
Hs#S3547136117.7117.71
Hs#S3776639TL132 pseudogene117.6817.68
Hs#S16883107IPO13Replication initiator 135.8317.49
Hs#S1728033XRRA1117.4617.46
Hs#S2139325Transcribed locus, weakly similar to XP_514093.1117.3017.30
similar to Ladinin 1 (Lad-1) (120 kDa linear IgA
bullous dermatosis antigen) (97 kDa linear IgA
bullous dermatosis antigen) (Linear IgA disease
antigen homolog) (LadA) [Pan troglodytes]
Hs#S15556062Solute carrier family 25 (mitochondrial carrier;117.2017.20
phosphate carrier), member 24
Hs#S4616141EXPH5Pygopus homolog 2 (Drosophila)117.1717.17
Hs#S32813027PTTG1Homo sapiens TSC22 domain family, member 2,35.6416.91
mRNA
Hs#S1731625GARSPolymerase I and transcript release factor35.6316.90
Hs#S2650345ADAMTS8Prefoldin subunit 528.4516.89
Hs#S3619291KIAA828Yippee-like 5 (Drosophila)28.4016.80
Hs#S16113351NBEAL2Telomeric repeat binding factor 2116.8016.80
Hs#S4044961C7orf41Transcribed locus116.8016.80
Hs#S40597866SLC23A2116.8016.80
Hs#S2654982ATP5IChromosome 20 open reading frame 1128.4016.80
Hs#S1110179CNNM4Amino-terminal enhancer of split28.4016.80
Hs#S1268286Major histocompatibility complex, class I-related116.7216.72
Hs#S1415594ABCB4Sema domain, immunoglobulin domain (Ig),116.7216.72
transmembrane domain (TM) and short cytoplasmic
domain, (semaphorin) 4G
Hs#S1730946SH3BP4CDNA FLJ44879 fis, clone BRAMY2033895116.7216.72
Hs#S1971587C9orf119Importin 13116.6416.64
Hs#S2653601HSPD1X-ray radiation resistance associated 1116.6016.60
Hs#S2929368ACTB116.6016.60
Hs#S30131655PCLOTranscribed locus, strongly similar to116.5716.57
XP_001145773.1 similar to PI-3-kinase-related
kinase SMG-1 isoform 1homolog [Pan troglodytes]
Hs#S34541296FLJ22167Exophilin 5116.5516.55
Hs#S3990203LOC645668Pituitary tumor-transforming 1116.5216.52
Hs#S4838548ARHGEF18Glycyl-tRNA synthetase116.4816.48
Hs#S7089804Ubiquitin specific peptidase 1935.4416.31
Hs#S2961525MRPL52ADAM metallopeptidase with thrombospondin type116.2716.27
1 motif, 8
Hs#S19863275U1SNRNPBPAdenosylhomocysteinase 3116.2116.21
Hs#S21592134CPEB2Neurofibromin 2 (bilateral acoustic neuroma)28.1016.21
Hs#S4372715TGM1Neurobeachin-like 2116.1816.18
Hs#S16889291RAPGEF5SAPK substrate protein 128.0716.14
Hs#S14273019FIZ1Chromosome 7 open reading frame 41116.1016.10
Hs#S34548677ANKRD26Homo sapiens ATP synthase, H+ transporting,116.0116.01
mitochondrial F0
complex, subunit E (ATP5I), nuclear gene encoding
mitochondrial
protein
Hs#S15116077PLEKHC1RNA binding motif protein 2027.9115.81
Hs#S4324752AGGF1CDNA FLJ14366 fis, A-HEMBA1001020115.8115.81
Hs#S21286877probably fused seq27.9115.81
Hs#S21934876Homo sapiens farnesyl diphosphate synthase27.9115.81
(farnesyl pyrophosphate synthetase,
dimethylallyltranstransferase,
geranyltranstransferase) (FDPS)
Hs#S38795190Cofilin 1 (non-muscle)35.1915.57
Hs#S2138748ATP-binding cassette, sub-family B (MDR/TAP),115.4815.48
member 4
Hs#S4808710Chromosome 9 open reading frame 119115.3715.37
Hs#S4323658KRTAP17-1Homo sapiens heat shock 60 kDa protein 1115.3715.37
(chaperonin) (HSPD1),
nuclear gene encoding mitochondrial protein
Hs#S2294364Piccolo (presynaptic cytomatrix protein)115.3215.32
Hs#S5930916NKRFHypothetical protein FLJ22167115.1515.15
Hs#S5978605SLC26A9Similar to Elongation factor Tu, mitochondrial115.1515.15
precursor (EF-Tu) (P43)
Hs#S18928610FBXO8Chromosome 13 open reading frame 2327.5115.02
Hs#S4074937EIF4B eukaryotic translation initiation factor 4B27.4714.94
Hs#S2653589Rho/rac guanine nucleotide exchange factor (GEF)114.9014.90
18
Hs#S1729966Transcribed locus114.8614.86
Hs#S17529890Keratin 39114.8614.86
Hs#S1972664LOC28555Bromodomain and WD repeat domain containing 1114.8614.86
Hs#S2293860COQ1BCDNA FLJ35672 fis, clone SPLEN2018280114.8614.86
Hs#S4616288GSK3BTranscribed locus, moderately similar to114.8614.86
XP_215201.4 similar to RNA-binding protein 4
(RNA-binding motif protein 4) (Lark homolog)
(Mlark) [Rattus norvegicus]
Hs#S1263783Poly(A) binding protein, cytoplasmic 3114.8314.83
Hs#S21592183C9orf122Trafficking protein particle complex 2114.8314.83
Hs#S858884114.8314.83
Hs#S26122385Target clone is not clearly identified (homology with34.8914.67
himeric products)
Hs#S4546062Chromosome 10 open reading frame 54114.6114.61
Hs#S2483479PABPC3Methionine sulfoxide reductase B3114.5814.58
Hs#S2138664RNF187Testis specific, 14114.5314.53
Hs#S38795021TRAPPC2Transcribed locus114.5014.50
Hs#S2331695HSP9AB1CDNA FLJ20832 fis, clone ADKA03033114.2614.26
Hs#S16391281NCBP1PR domain containing 427.1214.23
Hs#S4283309DEAD (Asp-Glu-Ala-Asp) box polypeptide 3, X-27.0714.15
linked
Hs#S38981998C1orf54TRNA 5-methylaminomethyl-2-thiouridylate114.1314.13
methyltransferase
Hs#S4833432MSRB3114.1214.12
Hs#S5930818C14orf43TH1-like (Drosophila)34.6714.02
Hs#S4296148TSGA14Endo-beta-N-acetylglucosaminidase113.9413.94
Hs#S15640600CD9Deoxynucleotidyltransferase, terminal, interacting113.9013.90
protein 1
Hs#S3602940Ubiquitin-conjugating enzyme E2O26.9213.84
Hs#S16819411NPC1Transcribed locus113.8413.84
Hs#S17873428Transcribed locus113.8413.84
Hs#S4026358TRMUKringle containing transmembrane protein 2113.8413.84
Hs#S10817661Ankyrin repeat and MYND domain containing 226.9213.84
Hs#S4773419FLJ21865Chromosome X open reading frame 626.9213.84
Hs#S34544410DNTTIP1Transcribed locus113.6413.64
Hs#S21390380Homo sapiens integrin, beta 1 (fibronectin receptor,26.7713.53
beta polypeptide, antigen CD29 includes MDF2,
MSK12) (ITGB1)
Hs#S3218259Suppression of tumorigenicity 14 (colon carcinoma)26.7213.44
Hs#S34542734KREMEN2113.3413.34
Hs#S3438578IMP (inosine monophosphate) dehydrogenase 1113.3413.34
Hs#S4029085LOC441891Transcribed locus, weakly similar to NP_009956.2113.3413.34
homologue; Rhb1p [Saccharomyces cerevisiae]
Hs#S16885659UBFD1La ribonucleoprotein domain family, member 126.6113.21
Hs#S11131792Transcribed locus113.0013.00
Hs#S15846854IMPDH1CDNA FLJ11947 fis, clone HEMBB1000726113.0013.00
Hs#S15846862Transcribed locus113.0013.00
Hs#S16507498113.0013.00
Hs#S1728022113.0013.00
Hs#S1824434Transcribed locus112.9012.90
Hs#S19862888Transcribed locus, weakly similar to112.8512.85
NP_001039643.1 protein LOC514688 [Bos taurus]
Hs#S2139683CPSF4Transcribed locus112.8512.85
Hs#S21592301MTHFD1Full-length cDNA clone CS0DL005YA15 of B cells112.8512.85
(Ramos cell line) Cot 25-normalized of Homo
sapiens (human)
Hs#S2293277Autocrine motility factor receptor26.4212.85
Hs#S2650236EPRSHomo sapiens neurofibromin 1 (neurofibromatosis,26.4212.85
von Recklinghausen disese, Watson disease) (NF1)
Hs#S2654955LOC645615112.6812.68
Hs#S29440731HIGD2ASimilar to hepatocellular carcinoma-associated112.6012.60
antigen 66
Hs#S3197874APTXHomo sapiens HIG1 domain family, member 2A112.5712.57
(HIGD2A)
Hs#S32462690C9orf142Chromosome 9 open reading frame 142112.3512.35
Hs#S3333631CDNA clone IMAGE: 6106200112.3512.35
Hs#S3383648DNAJA2DnaJ (Hsp40) homolog, subfamily A, member 2112.3512.35
Hs#S37211091Full-length cDNA clone CS0DJ012YG05 of T cells112.3512.35
(Jurkat cell line) Cot 10-normalized of Homo sapiens
(human)
Hs#S3881511MAPK6Mitogen-activated protein kinase 6112.3512.35
Hs#S3940071ERCC1Excision repair cross-complementing rodent repair112.3512.35
deficiency, complementation group 1 (includes
overlapping antisense sequence)
Hs#S4268629WD repeat domain 57 (U5 snRNP specific)26.1512.30
Hs#S4324622C1q and tumor necrosis factor related protein 626.1312.26
Hs#S4341465CYP2S1112.1912.19
Hs#S4400974UBE3A112.1912.19
Hs#S4614668VPS11Cytochrome P450, family 2, subfamily S,112.1912.19
polypeptide 1
Hs#S4615142AGPAT1Homo sapiens ribosomal protein L23 (RPL23)26.0712.14
Hs#S4838504GSTZ1Ubiquitin protein ligase E3A (human papilloma virus112.1112.11
E6-associated protein, Angelman syndrome)
Hs#S5475261LOC7286571-acylglycerol-3-phosphate O-acyltransferase 1112.0012.00
(lysophosphatidic acid acyltransferase, alpha)
Hs#S5517477SFRS15Glutathione transferase zeta 1 (maleylacetoacetate111.9711.97
isomerase)
Hs#S5951324RBM15Similar to dual specificity phosphatase 8111.8611.86
Hs#S6145210SUMO2Splicing factor, arginine/serine-rich 15111.8611.86
Hs#S794414RNA binding motif protein 15111.8611.86
Hs#S16056656PLS1SMT3 suppressor of mif two 3 homolog 2 (S. cerevisiae)111.8611.86
Hs#S26643428USP28Transcribed locus111.8611.86
Hs#S23254699FAM3AG protein-coupled receptor kinase interactor 225.9311.86
Hs#S3255888AFF4Solute carrier family 35, member B425.9311.86
Hs#S3307627OBSL1Plastin 1 (I isoform)111.8611.86
Hs#S24443601TNFRSF1AUbiquitin specific peptidase 28111.8611.86
Hs#S26643702Solute carrier family 38, member 525.9311.86
Hs#S4546315Family with sequence similarity 3, member A111.8611.86
Hs#S4539222SETAF4/FMR2 family, member 4111.6711.67
Hs#S1970096SPRY2Tumor necrosis factor receptor superfamily, member111.4611.46
1A
Hs#S1824507LETM2Transcribed locus, strongly similar to XP_217277.4111.4611.46
similar to without children CG5965-PA [Rattus
norvegicus]
Hs#S1726632CAPN2Toll interacting protein25.6811.37
Hs#S2293240Adenosine deaminase, RNA-specific25.6611.32
Hs#S3792926SET translocation (myeloid leukemia-associated)111.3011.30
Hs#S16819337Homo sapiens SH3KBP1 binding protein 125.6011.20
(SHKBP1)
Hs#S1637758UBL3Homo sapiens v-maf musculoaponeurotic25.6011.20
fibrosarcoma oncogene homolog
G (avian) (MAFG)
Hs#S3110506UDP glucuronosyltransferase 2 family, polypeptide111.1511.15
A1
Hs#S3219661MINPP1Zinc finger protein 607111.1511.15
Hs#S3377338Transcribed locus111.1511.15
Hs#S4320649Artemin111.1511.15
Hs#S1970972Hypothetical LOC390306111.1511.15
Hs#S3781962111.1511.15
Hs#S16056988Mitochondrial translational initiation factor 3111.1511.15
Hs#S100711Establishment of cohesion 1 homolog 2 (S. cerevisiae)111.1511.15
Hs#S1727033ALOXE3CCR4-NOT transcription complex, subunit 4111.1511.15
Hs#S3219600Sprouty homolog 2 (Drosophila)25.5711.15
Hs#S1097393DRAP1Leucine zipper-EF-hand containing transmembrane25.5711.15
protein 2
Hs#S1184Homo sapiens calpain 2, (m/II) large subunit25.5711.15
(CAPN2)
Hs#S4413186Oxidative-stress responsive 125.5611.12
Hs#S3940065USP15Epithelial V-like antigen 1111.0711.07
Hs#S5515813CSF3RAbl interactor 2111.0711.07
Hs#S5899942VEGFAV-rel reticuloendotheliosis viral oncogene homolog110.8710.87
A, nuclear factor of kappa light polypeptide gene
enhancer in B-cells 3, p65 (avian)
Hs#S6100790Transcribed locus110.8710.87
Hs#S1726257Homo sapiens iduronidase, alpha-L-(IDUA),25.4410.87
Hs#S1177138Frizzled homolog 1 (Drosophila)110.8710.87
Hs#S16884388FGFR2KIAA1632110.8710.87
Hs#S2356728TNKS2Transcribed locus110.8710.87
Hs#S34543533CEP135Cyclin-dependent kinase 2110.8710.87
Hs#S2333982V-ets erythroblastosis virus E26 oncogene homolog110.8710.87
1 (avian)
Hs#S1336309Shroom family member 1110.8710.87
Hs#S19874976DMNRetinol saturase (all-trans-retinol 13,14-reductase)110.6210.62
Hs#S2140506ZNF728Small Cajal body-specific RNA 2110.6210.62
Hs#S4671903ATP8A1Zinc finger protein-like 125.3110.62
Hs#S1731389GK3P110.5410.54
Hs#S4619377Apolipoprotein B mRNA editing enzyme, catalytic110.5410.54
polypeptide-like 3C
Hs#S1730187DHRS4CDNA FLJ33736 fis, clone BRAWH2018514110.5410.54
Hs#S16884579LOC44456SUMO1/sentrin specific peptidase 1110.3810.38
Hs#S30194364NDEL1RAD9 homolog A (S. pombe)110.3810.38
Hs#S4296637TRAM1Fragile X mental retardation, autosomal homolog 1110.3210.32
Hs#S3438145SV2ALactate dehydrogenase A110.2810.28
Hs#S15967295Clone 23963 mRNA sequence110.2110.21
Hs#S4285059110.1310.13
Hs#S3989863Transmembrane protein 4325.0510.10
Hs#S24303044DDNCyclin B1110.0210.02
Hs#S34545639LOC643464Glucosidase, beta; acid (includes19.889.88
glucosylceramidase)
Hs#S3993862Similar to dynein, cytoplasmic, light peptide19.889.88
Hs#S11046863Homo sapiens pregnancy specific beta-1-24.949.88
glycoprotein 4 (PSG4)
Hs#S11062731C2orf3Polymerase (DNA directed) sigma24.949.88
Hs#S3618962EVA1Homo sapiens brain protein 13 (BR13)24.949.88
Hs#S5918966ABI2Vacuolar protein sorting 13 homolog A (S. cerevisiae)24.949.88
Hs#S3619207PLEKHG2Dolichyl-phosphate (UDP-N-acetylglucosamine) N-19.889.88
acetylglucosaminephosphotransferase 1 (GlcNAc-1-
P transferase)
Hs#S1727399RELAMevalonate (diphospho) decarboxylase24.949.88
Hs#S3973116Transcribed locus, moderately similar to19.889.88
NP_001072641.1 protein LOC7150097 [Xenopus
tropicalis]
Hs#S3782070FZD1ArsA arsenite transporter, ATP-binding, homolog 119.889.88
(bacterial)
Hs#S16887814KIAA1632Cholinergic receptor, muscarinic 219.889.88
Hs#S2138915Enolase superfamily member 119.889.88
Hs#S1728059CDK2Chromosome 19 open reading frame 2219.889.88
Hs#S29525962ETS1Low density lipoprotein receptor-related protein 1119.729.72
Hs#S16885581SHROOM1MAP-kinase activating death domain24.859.70
Hs#S875916RETSATGap junction protein, beta 3, 31 kDa19.629.62
Hs#S3265SCARNA2NAD synthetase 119.499.49
Hs#S15515243Solute carrier family 41, member 319.469.46
Hs#S34542796APOBEC3CMRNA; cDNA DKFZp434C0923 (from clone19.399.39
DKFZp434C0923)
Hs#S103088Upstream binding transcription factor, RNA19.299.29
polymerase I
Hs#S1101292SENP1Transcribed locus19.299.29
Hs#S11047073RAD9ATranscribed locus19.299.29
Hs#S14272999FXR1Transcribed locus19.299.29
Hs#S1503932LDHATripeptidyl peptidase I19.299.29
Hs#S1570046Zinc finger protein 31919.299.29
Hs#S1579109Uncoupling protein 2 (mitochondrial, proton carrier)19.299.29
Hs#S15974021CCNB1Transcribed locus19.299.29
Hs#S16102754GBANLR family member X119.299.29
Hs#S1638509LOC73138Similar to DnaJ homolog subfamily A member 119.299.29
(Heat shock 40 kDa protein 4) (DnaJ protein
homolog 2) (HSJ-2) (HSDJ)
Hs#S16535341DPAGT1CDNA FLJ31443 fis, clone NT2NE200080819.299.29
Hs#S16886868Erythropoietin receptor19.299.29
Hs#S16889882ASNA1CDNA clone IMAGE: 527788319.299.29
Hs#S17529224CHRM2Cas-Br-M (murine) ecotropic retroviral transforming24.619.22
sequence
Hs#S21591426ENOSF1Tropomyosin 3 pseudogene19.169.16
Hs#S2215784C19orf2219.149.14
Hs#S23099711LRP11SCO cytochrome oxidase deficient homolog 119.099.09
(yeast)
Hs#S2331861ASB1IQ motif containing GTPase activating protein 124.539.07
Hs#S24302745CTPSSema domain, immunoglobulin domain (Ig),19.059.05
transmembrane domain (TM) and short cytoplasmic
domain, (semaphorin) 4C
Hs#S26179767GJB3CDK5 regulatory subunit associated protein 119.009.00
Hs#S2649714NADSYN118.908.90
Hs#S2654228SLC41A3Transcribed locus18.908.90
Hs#S29684090Transcribed locus, strongly similar to18.908.90
XP_001058360.1 similar to Heterogeneous nuclear
ribonucleoprotein G (hnRNP G) (RNA-binding motif
protein, X chromosome) isoform 1 [Rattus
norvegicus]
Hs#S3355603DUSP14Hypothetical LOC34919618.908.90
Hs#S3438518UBTFMRNA; cDNA DKFZp686I19109 (from clone18.908.90
DKFZp686I19109)
Hs#S3438671LOC646839Dual specificity phosphatase 118.908.90
Hs#S3511371Transcribed locus18.908.90
Hs#S3914296SCO1Transcribed locus18.908.90
Hs#S39547751SEMA4CHomo sapiens actin related protein ⅔ complex,18.908.90
subunit 3, 21 kDa (ARPC3), mRNA.
Hs#S3989629CDK5RAP1Transmembrane protein induced by tumor necrosis18.908.90
factor alpha
Hs#S4046399THRAHypothetical protein LOC20354718.908.90
Hs#S41443272TRM5 tRNA methyltransferase 5 homolog (S. cerevisiae)24.458.90
Hs#S4261892Homo sapiens keratin 8 (KRT8), mRNA24.458.90
Hs#S4273435Jun B proto-oncogene18.708.70
Hs#S4622076LOC349196Eukaryotic translation elongation factor 1 alpha 224.358.70
Hs#S4622728Ribosomal protein L36a18.658.65
Hs#S4807467DUSP1Zinc finger and BTB domain containing 4618.658.65
Hs#S4832001Clone TESTIS-724 mRNA sequence18.658.65
Hs#S5940277Triggering receptor expressed on myeloid cells-like 224.328.65
Hs#S6120703ARPC3Tumor suppressor candidate 424.288.57
Hs#S6140404TMPITMitochondrial ribosomal protein L4824.288.57
Hs#S6158954LOC23547Homo sapiens DNA directed RNA polymerase II18.458.45
polypeptide J-related
(POLR2J2), mRNA
Hs#S7089884JUNBKelch domain containing 518.408.40
Hs#S793581RPL36ATranscribed locus, moderately similar to18.408.40
XP_001072116.1 similar to UPF0315 protein
[Rattus norvegicus]
Hs#S932405ZBTB46Jub, ajuba homolog (Xenopus laevis)18.408.40
Hs#S1729506Phosphate cytidylyltransferase 2, ethanolamine18.358.35
Hs#S1732276PFDN518.308.30
Hs#S1729763POLR2J2Protein kinase N124.158.30
Hs#S29722022YPEL5Mediator of RNA polymerase II transcription,18.248.24
subunit 8 homolog (S. cerevisiae)
Hs#S1728450C2orf11Similar to dishevelled 1 isoform a24.128.24
Hs#S2270359AESSP100 nuclear antigen18.248.24
Hs#S1263959KLHDC5Sidekick homolog 1 (chicken)24.118.21
Hs#S19626863Ubiquitin specific peptidase 1124.098.19
Hs#S3520094JUB18.158.15
Hs#S1726446PCYT2Phosphodiesterase 8A24.068.13
Hs#S1729627Nucleosome assembly protein 1-like 418.038.03
Hs#S4283794MED818.018.01
Hs#S19656869SP1Ribosomal protein L3518.008.00
Hs#S16888563M-RIPTranscribed locus17.917.91
Hs#S5472875Mesoderm induction early response 1 homolog17.917.91
(Xenopus laevis)
Hs#S2357370NF2Dual-specificity tyrosine-(Y)-phosphorylation17.917.91
regulated kinase 1B
Hs#S4616868LOC5135Tissue specific transplantation antigen P35B17.917.91
Hs#S4620348NAP1L4Zinc finger and BTB domain containing 617.917.91
Hs#S16820078KIAA0265 protein17.917.91
Hs#S1729231RPL35Transcribed locus, strongly similar to17.807.80
XP_001080201.1 similar to ribosomal protein L10
[Rattus norvegicus]
Hs#S24273099RBM2GRIP1 associated protein 117.587.58
Hs#S402201017.587.58
Hs#S953315MIER1Zinc finger protein 66417.557.55
Hs#S38872148TRIM29Keratin 1317.547.54
Hs#S4284382LOC28397Interferon-related developmental regulator 217.477.47
Hs#S3618391FDPSTranscribed locus17.437.43
Hs#S17853615DYRK1BMitochondrial ribosomal protein L5217.437.43
Hs#S34543862TSTA3U11/U12 snRNP 35K17.437.43
Hs#S3508192ZBTB6Cytoplasmic polyadenylation element binding17.437.43
protein 2
Hs#S1732446KIAA265Transglutaminase 1 (K polypeptide epidermal type I,17.437.43
protein-glutamine-gamma-glutamyltransferase)
Hs#S542952CRIP1Rap guanine nucleotide exchange factor (GEF) 517.437.43
Hs#S3782069ABCC3FLT3-interacting zinc finger 117.437.43
Hs#S1730888Ankyrin repeat domain 2617.437.43
Hs#S24303175NCAPD2Pleckstrin homology domain containing, family C17.437.43
(with FERM domain) member 1
Hs#S4725762GRIPAP1Angiogenic factor with G patch and FHA domains 117.437.43
Hs#S24573475CDNA FLJ12096 fis, clone HEMBB100261317.437.43
Hs#S4622791ZNF664CDNA: FLJ23530 fis, clone LNG0605517.437.43
Hs#S16885485KRT13Transcribed locus17.437.43
Hs#S4396109AHCY17.437.43
Hs#S11047217C13orf23Transcribed locus, strongly similar to17.437.43
XP_001164757.1 centromere protein C 1 isoform 2
[Pan troglodytes]
Hs#S1727134EIF4BKeratin associated protein 17-117.437.43
Hs#S29965937IFRD2Transcribed locus, moderately similar to17.437.43
XP_001055131.1 similar to Heat shock protein HSP
90-beta (HSP 84) (Tumor-specific transplantation 84 kDa
antigen) (TSTA) [Rattus norvegicus]
Hs#S4833612USP37NF-kappaB repressing factor17.437.43
Hs#S16884650SLC22A18Solute carrier family 26, member 917.437.43
Hs#S1972910F-box protein 817.437.43
Hs#S35152794Transcribed locus, strongly similar to17.437.43
XP_001069454.1 similar to Splicing factor,
arginine/serine-rich 3 (Pre-mRNA splicing factor
SRP20) (X16 protein) [Rattus norvegicus]
Hs#S1729605Transcribed locus17.437.43
Hs#S34549106WBP4Transcribed locus17.437.43
Hs#S2570282Transcribed locus17.437.43
Hs#S1263097Hypothetical protein LOC28550517.437.43
Hs#S1731172Coenzyme Q10 homolog B (S. cerevisiae)17.437.43
Hs#S2293760Glycogen synthase kinase 3 beta17.437.43
Hs#S213991617.437.43
Hs#S3990767Chromosome 9 open reading frame 12217.437.43
Hs#S15915730STXBP4Transcribed locus17.437.43
Hs#S4792633Transcribed locus17.437.43
Hs#S15918611Transcribed locus17.437.43
Hs#S16819792Ubiquitin specific peptidase 3723.727.43
Hs#S16885025C6orf113Family with sequence similarity 98, member A17.417.41
Hs#S21504143STAMBPL1Acid phosphatase 1, soluble17.417.41
Hs#S29128550KIAA423Solute carrier family 25, member 3617.417.41
Hs#S29441564GTDC1Ubiquitin-conjugating enzyme E2D 2 (UBC4/517.367.36
homolog, yeast)
Hs#S34541381THADATranscribed locus, moderately similar to16.926.92
XP_235480.4 similar to DEAD box polypeptide 17
isoform p82 [Rattus norvegicus]
Hs#S34547277Splicing factor 416.926.92
Hs#S418823016.926.92
Hs#S4621795PIGGDynein, axonemal, heavy chain like 116.926.92
Hs#S21300372Cytochrome P450, family 1, subfamily A,16.926.92
polypeptide 1
Hs#S24303340CSADSimilar to Glyceraldehyde-3-phosphate16.926.92
dehydrogenase (GAPDH)
Hs#S213002933′(2′),5′-bisphosphate nucleotidase 116.926.92
Hs#S19863003PIGCCDNA clone IMAGE: 296054016.926.92
Hs#S2651797IBSPTranscribed locus16.926.92
Hs#S26643453KLRA1Chromosome 19 open reading frame 4316.926.92
Hs#S15644477YWHAQMitogen-activated protein kinase-activated protein16.816.81
kinase 3
Hs#S2140695TRIM23Polymerase (DNA directed), alpha 2 (70 kD subunit)16.796.79
Hs#S16056802DDX19BPolymerase (RNA) III (DNA directed) polypeptide16.786.78
A, 155 kDa
Hs#S2241465816.786.78
Hs#S2450780Ribophorin I16.776.77
Hs#S3619160MGC1646Transforming growth factor beta 1 induced transcript 116.756.75
Hs#S4618103TSPAN15Protein (peptidylprolyl cis/trans isomerase) NIMA-16.706.70
interacting 1
Hs#S19921576RLFUbiquitin-like, containing PHD and RING finger16.646.64
domains, 2
Hs#S24639346Histocompatibility (minor) HA-116.596.59
Hs#S34550263FZD4Transcribed locus, strongly similar to XP_420179.216.596.59
similar to Nucleoporin 62 kDa [Gallus gallus]
Hs#S4022545LYAR16.546.54
Hs#S36352168Ring finger protein 3416.426.42
Hs#S4807559ZNF574Protein interacting with PRKCA 116.426.42
Hs#S1729352RAB12Transcribed locus, weakly similar to16.426.42
NP_001039775.1 homolog B [Bos taurus]
Hs#S3355506Integral membrane protein 2B16.426.42
Hs#S17668848CIRSimilar to ribosomal protein S15a16.426.42
Hs#S1730103C21orf6KIAA160216.356.35
Hs#S4553472SLC4A716.336.33
Hs#S21279396PCA3Cytokine-like nuclear factor n-pac16.336.33
Hs#S5890230LOC39637AKT1 substrate 1 (proline-rich)16.266.26
Hs#S5978485PPP2R5B16.266.26
Hs#S1263022BBS2PTC7 protein phosphatase homolog (S. cerevisiae)16.266.26
Hs#S4614707Transcribed locus, strongly similar to XP_509149.216.266.26
ATP synthase, H+ transporting, mitochondrial F1
complex, beta subunit isoform 2 [Pan troglodytes]
Hs#S4808034Rho-related BTB domain containing 216.216.21
Hs#S553751Transcribed locus, strongly similar to16.216.21
XP_001060296.1 similar to ribosomal protein L18a
[Rattus norvegicus]
Hs#S2567063GNB2L1Fasciculation and elongation protein zeta 2 (zygin II)16.136.13
Hs#S19755103START domain containing 4, sterol regulated16.096.09
Hs#S6131705RANBP2-like and GRIP domain containing 516.076.07
Hs#S4621282Glutamine and serine rich 116.036.03
Hs#S21310805Dynein, cytoplasmic 1, intermediate chain 215.985.98
Hs#S1637843Homo sapiens Rho GDP dissociation inhibitor (GDI)15.935.93
alpha (ARHGDIA)
Hs#S105525315.935.93
Hs#S1083575FANCF15.935.93
Hs#S11047086Transcribed locus, moderately similar to15.935.93
NP_058086.2 nuclear ribonucleoprotein A2/B1
isoform 1 [Mus musculus]
Hs#S11147198WASCDNA FLJ44682 fis, clone BRACE301043515.935.93
Hs#S1263033ZC3H12APhosphatase and tensin homolog (mutated in15.935.93
multiple advanced cancers 1)
Hs#S1281580SLC12A8Acyl-Coenzyme A dehydrogenase, C-4 to C-1215.935.93
straight chain
Hs#S1357226LOC347422Monoglyceride lipase15.935.93
Hs#S1399072LOC728919EBNA1 binding protein 215.935.93
Hs#S15500072LOC388116Anthrax toxin receptor 115.935.93
Hs#S15554729LOC461Transcribed locus15.935.93
Hs#S15589610RAB6A, member RAS oncogene family15.935.93
Hs#S15631516Polyglutamine binding protein 115.935.93
Hs#S1583106KCNG4Transcribed locus15.935.93
Hs#S16020110Transcribed locus, moderately similar to15.935.93
NP_001039332.1 [Bos taurus]
Hs#S16050244Poliovirus receptor-related 1 (herpesvirus entry15.845.84
mediator C; nectin)
Hs#S16056808MRPL47Ariadne homolog 2 (Drosophila)15.735.73
Hs#S16817554PSMA1KIAA025815.735.73
Hs#S16817928Tribbles homolog 1 (Drosophila)15.725.72
Hs#S16818981COMM domain containing 315.655.65
Hs#S16820045ACP5MRNA; cDNA DKFZp686E0389 (from clone15.605.60
DKFZp686E0389)
Hs#S16885672Transcribed locus, moderately similar to15.575.57
XP_527544.2 RNA-binding motif protein 16 [Pan
troglodytes]
Hs#S16888142Transcribed locus15.575.57
Hs#S16888167CDNA clone IMAGE: 529703215.575.57
Hs#S16888865Ubiquitin-like 315.575.57
Hs#S16890023LOC55426Transcribed locus, strongly similar to15.575.57
XP_001129953.1 similar to Iroquois-class
homeodomain protein IRX-5 (Iroquois homeobox
protein 5) (Homeodomain protein IRXB2) (IRX-2A)
[Homo sapiens]
Hs#S1724424UCA1Multiple inositol polyphosphate histidine15.575.57
phosphatase, 1
Hs#S1727355Transcribed locus15.575.57
Hs#S1729457DNM1DN8-2Transcribed locus15.575.57
Hs#S1731007MGC1276Transcribed locus15.575.57
Hs#S1731370Clone 24875 mRNA sequence15.575.57
Hs#S1731150015.575.57
Hs#S1732279CHMP4BCDNA FLJ43848 fis, clone TESTI400641215.575.57
Hs#S1760680Arachidonate lipoxygenase 315.575.57
Hs#S1788883Transcribed locus15.575.57
Hs#S18076913FANCCDR1-associated protein 1 (negative cofactor 2 alpha)15.575.57
Hs#S1824468HGSNAT15.575.57
Hs#S1824518SH3GLP115.575.57
Hs#S1845424Ubiquitin specific peptidase 1515.575.57
Hs#S1969005Colony stimulating factor 3 receptor (granulocyte)15.575.57
Hs#S1969727Vascular endothelial growth factor A15.575.57
Hs#S19741388CDNA FLJ11568 fis, clone HEMBA100327815.575.57
Hs#S19863067KIAA184CDNA: FLJ22750 fis, clone KAIA047815.575.57
Hs#S20337965Transcribed locus15.575.57
Hs#S21278248Fibroblast growth factor receptor 2 (bacteria-15.575.57
expressed kinase, keratinocyte growth factor
receptor, craniofacial dysostosis 1, Crouzon
syndrome, Pfeiffer syndrome, Jackson-Weiss
syndrome)
Hs#S21290655Tankyrase, TRF1-interacting ankyrin-related ADP-15.575.57
ribose polymerase 2
Hs#S21309634Centrosomal protein 135 kDa15.575.57
Hs#S2331146Transcribed locus, moderately similar to15.575.57
NP_001070679.2 protein LOC324254 [Danio rerio]
Hs#S2332235FAM98A15.575.57
Hs#S24298108ACP1Desmuslin15.575.57
Hs#S24303115SLC25A36Zinc finger protein 72815.575.57
Hs#S24303499UBE2D2ATPase, aminophospholipid transporter (APLT),15.575.57
Class I, type 8A, member 1
Hs#S2611802TNPO2Glycerol kinase 3 pseudogene15.575.57
Hs#S2654276BCL2L115.575.57
Hs#S2654817SSRP1Dehydrogenase/reductase (SDR family) member 415.575.57
Hs#S2706477PRDM4Similar to pleckstrin homology domain containing,15.575.57
family M (with RUN domain) member 1; adapter
protein 162
Hs#S2815585DDX3XNudE nuclear distribution gene E homolog (A. nidulans)-15.575.57
like 1
Hs#S28437704UBE2OTranslocation associated membrane protein 115.575.57
Hs#S29223348Synaptic vesicle glycoprotein 2A15.575.57
Hs#S2930469ANKMY2Transcribed locus15.575.57
Hs#S29625188CXorf6Transcribed locus15.575.57
Hs#S29705122SF4Transcribed locus15.575.57
Hs#S3219890Dendrin15.575.57
Hs#S3308636DNAHL1Hypothetical LOC64346415.575.57
Hs#S3322611CYP1A1Transcribed locus15.575.57
Hs#S3438621LOC441893Transcribed locus15.575.57
Hs#S3439001BPNT1Carboxypeptidase D15.535.53
Hs#S34541250Roundabout homolog 4, magic roundabout15.445.44
(Drosophila)
Hs#S34542798Smg-5 homolog, nonsense mediated mRNA decay15.415.41
factor (C. elegans)
Hs#S34543734C19orf43Histone deacetylase 115.395.39
Hs#S34544361MAPKAPK3Zinc finger, MYND-type containing 815.375.37
Hs#S3547697POLA2Phosphatidylinositol binding clathrin assembly15.345.34
protein
Hs#S3549795POLR3ATankyrase 1 binding protein 1, 182 kDa15.345.34
Hs#S3618502Echinoderm microtubule associated protein like 515.275.27
Hs#S3990867ITGB115.255.25
Hs#S3991248RPN1Phosphogluconate dehydrogenase15.215.21
Hs#S4019789TGFB1I1Microtubule associated serine/threonine kinase 215.195.19
Hs#S4020446ST14GTP binding protein 6 (putative)15.195.19
Hs#S4084938PIN1Hypothetical protein MGC1552315.145.14
Hs#S4186073UHRF2Phosphatidylinositol glycan anchor biosynthesis,14.944.94
class W
Hs#S4188085LARP1Ankyrin repeat and SOCS box-containing 1314.944.94
Hs#S4331927HMHA1Mitogen-activated protein kinase 914.944.94
Hs#S4332296F-box protein 3414.944.94
Hs#S4359392Transcribed locus14.944.94
Hs#S4522802RNF34Cytochrome P450, family 4, subfamily V,14.944.94
polypeptide 2
Hs#S4613584AMFRFamily with sequence similarity 89, member A14.944.94
Hs#S4619020NF1Enhancer of zeste homolog 2 (Drosophila)14.944.94
Hs#S4619920PICK1Mucolipin 114.944.94
Hs#S4620160Transcribed locus, moderately similar to14.944.94
XP_001170282.1 similar to PSMD4P2 protein
isoform 7 [Pan troglodytes]
Hs#S4620986ITM2BMethyltransferase like 514.944.94
Hs#S4622929LOC64479Nucleophosmin (nucleolar phosphoprotein B23,14.944.94
numatrin)
Hs#S4707124KIAA162S-phase kinase-associated protein 2 (p45)14.944.94
Hs#S476687814.944.94
Hs#S4797397N-PACHypothetical protein LOC14818914.944.94
Hs#S4805830AKT1S1Aldehyde dehydrogenase 16 family, member A114.944.94
Hs#S4805917Transcribed locus14.944.94
Hs#S4807843PPTC7Chromosome 6 open reading frame 4914.944.94
Hs#S4854304Hypothetical protein FLJ3848214.944.94
Hs#S5511550RHOBTB21-acylglycerol-3-phosphate O-acyltransferase 614.944.94
(lysophosphatidic acid acyltransferase, zeta)
Hs#S5887779Transcribed locus, strongly similar to XP_512748.214.944.94
similar to Protein fosB (G0/G1 switch regulatory
protein 3) [Pan troglodytes]
Hs#S5902854WDR57Intersectin 1 (SH3 domain protein)14.944.94
Hs#S5946389C1QTNF6SREBF chaperone14.884.88
Hs#S5978450FEZ2Citrate synthase14.874.87
Hs#S6109353STARD4WW domain binding protein 1114.864.86
Hs#S6128803ZBTB38TBC1 domain family, member 9B (with GRAM14.834.83
domain)
Hs#S6134648RPL23Tumor necrosis factor receptor superfamily, member14.824.82
10b
Hs#S7089897RGPD5Dystroglycan 1 (dystrophin-associated glycoprotein14.804.80
1)
Hs#S785737QSER1F-box protein 4114.804.80
Hs#S4615264DYNC1I2Collagen, type XVIII, alpha 114.744.74
Hs#S1731798TAF1514.494.49
Hs#S40833126ARHGDIAUridine-cytidine kinase 214.454.45
Hs#S16507492CDAEukaryotic translation initiation factor 2, subunit 314.454.45
gamma, 52 kDa
Hs#S4785071GIT2Kinesin family member C314.454.45
Hs#S4044799SLC35B4Ras homolog gene family, member T114.454.45
Hs#S20047532Fucosyltransferase 10 (alpha (1,3)14.454.45
fucosyltransferase)
Hs#S11046720Chromosome 10 open reading frame 11814.454.45
Hs#S1730518Tigger transposable element derived 614.454.45
Hs#S34550083Transcribed locus, strongly similar to14.454.45
XP_001174013.1 cortactin isoform 1 [Pan
troglodytes]
Hs#S1730019PTENCDNA FLJ36668 fis, clone UTERU200392614.454.45
Hs#S4840117ACADMG protein-coupled receptor 10814.314.31
Hs#S6125478MGLLChromosome 20 open reading frame 8214.314.31
Hs#S6686750EBNA1BP2NADH dehydrogenase (ubiquinone) flavoprotein 1,14.284.28
51 kDa
Hs#S19656836ANTXR1Sorting nexin 1214.284.28
Hs#S1731396Chromosome 14 open reading frame 3214.284.28
Hs#S1731499RAB6AHomo sapiens, clone IMAGE: 6016214, mRNA14.154.15
Hs#S377406PQBP1Crm, cramped-like (Drosophila)14.154.15
Hs#S1727714Laminin, beta 314.014.01
Hs#S16889729SLC38A5Solute carrier family 22 (organic cation transporter),13.723.72
member 18
Hs#S4618455Transcribed locus13.723.72
Hs#S4395994PVRL1CDNA clone IMAGE: 528669913.723.72
Hs#S18388775REPIN1Transcribed locus13.723.72
Hs#S4271114ARIH2WW domain binding protein 4 (formin binding13.723.72
protein 21)
Hs#S33737228KIAA 258Transcribed locus13.723.72
Hs#S17083357TRIB1Transcribed locus, moderately similar to13.723.72
XP_342747.2 similar to BMS1-like, ribosome
assembly protein [Rattus norvegicus]
Hs#S4618607TOLLIPTranscribed locus13.723.72
Hs#S15510173ADARTranscribed locus13.723.72
Hs#S16056502COMMD3Transcribed locus13.723.72
Hs#S31785558KIAA 664Transcribed locus13.723.72
Hs#S39299009PTRFSyntaxin binding protein 413.723.72
Hs#S16887491SHKBP113.723.72
Hs#S5979068MRNA full length insert cDNA clone EUROIMAGE13.723.72
122871
Hs#S4831691MAFGTranscribed locus13.723.72
Hs#S3334657ATXN7L3Chromosome 6 open reading frame 11313.723.72
Hs#S21592567OXSR1STAM binding protein-like 113.723.72
Hs#S2929936CPDKIAA042313.723.72
Hs#S1263776CCND1Glycosyltransferase-like domain containing 113.723.72
Hs#S4075831USP19Thyroid adenoma associated13.723.72
Hs#S33939840IDUACDNA FLJ45088 fis, clone BRAWH302931313.723.72
Hs#S4622777ROBO4CDNA FLJ32587 fis, clone SPLEN200040213.723.72
Hs#S4285062SMG5Phosphatidylinositol glycan anchor biosynthesis,13.723.72
class G
Hs#S35176164HDAC1CDNA FLJ41845 fis, clone NT2RI300309513.723.72
Hs#S3990709ZMYND8Cysteine sulfinic acid decarboxylase13.723.72
Hs#S34542802PICALMTranscribed locus13.723.72
Hs#S18600699TNKS1BP1Phosphatidylinositol glycan anchor biosynthesis,13.723.72
class C
Hs#S4364791ZFPL1Integrin-binding sialoprotein (bone sialoprotein,13.723.72
bone sialoprotein II)
Hs#S3374762EML5Killer cell lectin-like receptor subfamily A, member 113.723.72
Hs#S16886064Tyrosine 3-monooxygenase/tryptophan 5-13.723.72
monooxygenase activation protein, theta polypeptide
Hs#S1727540PGDTripartite motif-containing 2313.723.72
Hs#S1728251CFL1DEAD (Asp-Glu-Ala-As) box polypeptide 19B13.723.72
Hs#S1731690MAST2MAGOH2 mRNA, partial sequence13.723.72
Hs#S21296084GTPBP6Transcribed locus13.723.72
Hs#S2655773MGC15523Hypothetical protein MGC1064613.723.72
Hs#S2947943TMEM43Tetraspanin 1513.723.72
Hs#S3335313PSG4Rearranged L-myc fusion13.723.72
Hs#S4263754POLSTranscribed locus13.723.72
Hs#S4622277BRI3Frizzled homolog 4 (Drosophila)13.723.72
Hs#S4701569PIGWHypothetical protein FLJ2042513.723.72
Hs#S1732137ASB13Transcribed locus13.723.72
Hs#S1730207MAPK9Zinc finger protein 57413.723.72
Hs#S553744FBXO34RAB12, member RAS oncogene family13.723.72
Hs#S3438404Transcribed locus13.723.72
Hs#S16819494CYP4V2CBF1 interacting corepressor13.723.72
Hs#S16820209FAM89AChromosome 21 open reading frame 613.723.72
Hs#S2140550EZH2Solute carrier family 4, sodium bicarbonate13.723.72
cotransporter, member 7
Hs#S4284292MCOLN1Prostate cancer antigen 313.723.72
Hs#S5931028Similar to RIKEN cDNA D330012F22 gene13.723.72
Hs#S16818029METTL5Protein phosphatase 2, regulatory subunit B′, beta13.723.72
isoform
Hs#S16820269NPM1Bardet-Biedl syndrome 213.723.72
Hs#S1731666SKP2Transcribed locus13.723.72
Hs#S4396123CDNA: FLJ22799 fis, clone KAIA262513.723.72
Hs#S16849936LOC148189CDNA: FLJ23388 fis, clone HEP1700813.723.72
Hs#S38656788ALDH16A1Guanine nucleotide binding protein (G protein), beta13.723.72
polypeptide 2-like 1
Hs#S1731989Transcribed locus13.723.72
Hs#S3601738VPS13ATranscribed locus13.723.72
Hs#S1054906MVDTranscribed locus, moderately similar to13.723.72
XP_001137633.1 similar to phosphorylase kinase
[Pan troglodytes]
Hs#S29944162C6orf49Transcribed locus13.723.72
Hs#S15631509FLJ38482Transcribed locus, strongly similar to13.723.72
NP_001038577.1 protein LOC566642 [Danio rerio]
Hs#S3837755AGPAT6Transcribed locus13.723.72
Hs#S4554130Fanconi anemia, complementation group F13.723.72
Hs#S4621421ITSN1Transcribed locus, moderately similar to13.723.72
XP_577968.2 hypothetical protein [Rattus
norvegicus]
Hs#S1560223Wiskott-Aldrich syndrome (eczema-13.723.72
thrombocytopenia)
Hs#S16817921SCAPZinc finger CCCH-type containing 12A13.723.72
Hs#S21284579CSSolute carrier family 12 (potassium/chloride13.723.72
transporters), member 8
Hs#S24303509WBP11Similar to N(2),N(2)-dimethylguanosine tRNA13.723.72
methyltransferase (tRNA(guanine-26,N(2)-N(2))
methyltransferase) (tRNA 2,2-dimethylguanosine-26
methyltransferase)
(tRNA(m(2,2)G26)dimethyltransferase)
Hs#S2655303MADDSimilar to APC11 anaphase promoting complex13.723.72
subunit 11 isoform 2
Hs#S3235532TBC1D9BSimilar to LOC13739213.723.72
Hs#S4029823TNFRSF1BHypothetical gene supported by NM_01488613.723.72
Hs#S4053965DAG113.723.72
Hs#S41444297FBXO4113.723.72
Hs#S4552358COL18A1Potassium voltage-gated channel, subfamily G,13.723.72
member 4
Hs#S4617250LMNB2CDNA FLJ30770 fis, clone FEBRA200073413.723.72
Hs#S4631472TH1LCDNA FLJ30384 fis, clone BRACE200811413.723.72
Hs#S783506CBLMitochondrial ribosomal protein L4713.723.72
Hs#S1972981IQGAP1Proteasome (prosome, macropain) subunit, alpha13.723.72
type, 1
Hs#S3970490513.723.72
Hs#S15631448UCK2Transcribed locus13.723.72
Hs#S5931482EIF2S3Acid phosphatase 5, tartrate resistant13.723.72
Hs#S1263912KIFC3Transcribed locus13.723.72
Hs#S5978643RHOT1Transcribed locus, strongly similar to XP_509486.113.723.72
similar to unc-51-like kinase 1; unc-51 (C. elegans)-
like kinase 1 [Pan troglodytes]
Hs#S17821113TRMT5Transcribed locus13.723.72
Hs#S5517881KRT813.723.72
Hs#S17865332FUT1Hypothetical LOC55420613.723.72
Hs#S1731237C1orf118Urothelial cancer associated 113.723.72
Hs#S1728873TIGD6MRNA; cDNA DKFZp547K189 (from clone13.723.72
DKFZp547K189)
Hs#S36352308FKSG8813.723.72
Hs#S998390Hypothetical protein MGC1276013.723.72
Hs#S4808094EEF1A2MRNA; cDNA DKFZp434D1229 (from clone13.723.72
DKFZp434D1229)
Hs#S16819943TREML2Transcribed locus13.723.72
Hs#S1726692GPR18Chromatin modifying protein 4B13.723.72
Hs#S1729841C2orf8213.723.72
Hs#S27074688FAM12ACDNA clone IMAGE: 482232613.723.72
Hs#S18076714TUSC4Fanconi anemia, complementation group C13.723.72
Hs#S2293250MRPL48Heparan-alpha-glucosaminide N-acetyltransferase13.723.72
Hs#S3547087NDUFV1SH3-domain GRB2-like pseudogene 113.723.72
Hs#S4410230SNX12Transcribed locus13.723.72
Hs#S4617449C14orf32Transcribed locus13.723.72
Hs#S37452909Transcribed locus, strongly similar to13.723.72
XP_001156507.1 hypothetical protein [Pan
troglodytes]
Hs#S39548371PKN1Transcribed locus13.723.72
Hs#S3990331CRAMP1LMixed lineage kinase 413.723.72
Hs#S2653639LOC642469Transcribed locus13.723.72
Hs#S4546024SDK1Transcribed locus13.723.72
Hs#S11046968USP11Transcribed locus13.723.72
Hs#S19626865PDE8AHomo sapiens, clone IMAGE: 5211852, mRNA13.723.72
Hs#S24574386LAMB3Transcribed locus13.723.72

[0000]

New targets for cancer treatment identified by
shRNA selection and verified by siRNA testing.
growth
GeneinhibitionEnrichment
SymbolAnnotation(%)factor
AP1G1Adaptor-related protein complex 1, gamma 1 subunit17.648.23
BTBD9BTB (POZ) domain containing 932.940.52
FAM120AFamily with sequence similarity 120A (Ossa/C9orf10)24.121.41
FXC1Fracture callus 1 homolog (rat) (TIM9B, TIMM10B)43.768.39
LOC400027Hypothetical gene supported by BC04741720.460.57
NCAPD2Non-SMC condensin I complex, subunit D227.838.71
NPC1Niemann-Pick disease, type C115.043.49
OBSL1Obscurin-like 123.645.92
RNF187Ring finger protein 18745.144.48
UBFD1Ubiquitin family domain containing 123.140.12

[0000]

Selection of GSE library.
Enriched two or
InfectedBrdU selectedmore times by
Cell linesSubsetsubsetBrdU selection
BJ-hTERT18421622298
HT108025631014344
PC315621833430
T2418621027445
MDA-MB-23115241574181
Numbers of sequences homologous to Unigene clusters

[0000]

Genes giving rise to GSEs enriched by BrdU selection in two or more cell lines. Values representing
over 2-fold enrichment are highlighted in yellow.
Relative enrichment in selected set
MDA-# of
GeneBJ-MB-selections
Unigene IDSymbolAnnotationhTERTHT1080PC3T24321enriched
Hs#S21294316C5orf13Chromosome 5616.520.002655.341947.421905.974
open reading
frame 13
Hs#S3219417CYCSCytochrome c,0.000.004.983.6310.413
somatic
Hs#S40429150.002958.582.490.005.083
Hs#S4617512VIMVimentin3.972.0827.370.000.003
Hs#S34543572LOC64594Similar to myosin3.410.00531.070.003.233
regulatory light
chain-like
Hs#S4712196ERHEnhancer of4315.660.001062.132.420.003
rudimentary
homolog
(Drosophila)
Hs#S4407757COX7A2Cytochrome c0.001.56531.072.723.393
oxidase subunit
VIIa polypeptide 2
(liver)
Hs#S22515139CGGBP1CGG triplet repeat0.002958.580.00973.7124.213
binding protein 1
Hs#S16889222MGST1Microsomal0.000.007.47973.719.683
glutathione S-
transferase 1
Hs#S1731283GGPS1Geranylgeranyl2.84986.190.001947.420.003
diphosphate
synthase 1
Hs#S1730201PAICSPhosphoribosylaminoimidazole0.002958.582.490.00635.323
carboxylase,
phosphoribosylaminoimidazole
succinocarboxamide
synthetase
Hs#S61633132H9616.52986.191.660.00635.323
Hs#S4618585POLE3Polymerase (DNA3699.143.126372.811.810.003
directed), epsilon 3
(p17 subunit)
Hs#S36352304SR14U2-associated0.001972.39531.071.81635.323
SR140 protein
Hs#S37211072THOC2THO complex 20.003.120.001947.421905.973
Hs#S1732093RNF13Ring finger protein1233.050.00531.07973.710.003
13
Hs#S38753720PDHA1Pyruvate2466.093.120.00973.710.003
dehydrogenase
(lipoamide) alpha 1
Hs#S16883457RPL23CDNA FLJ26326 fis,0.003944.77531.071947.420.003
clone HRT01120,
highly similar to
60S ribosomal
protein L23
Hs#S2140320VPS24Vacuolar protein616.520.00531.070.001270.653
sorting 24
homolog (S.cerevisiae)
Hs#S3991088GTF2H5General616.522958.580.001947.420.003
transcription factor
IIH, polypeptide 5
Hs#S1729262SEP1515 kDa2.041.040.5510.880.542
selenoprotein
Hs#S2813090UBE2TUbiquitin-1.993.430.930.684.962
conjugating
enzyme E2T
(putative)
Hs#S1728170VAPA11.360.390.550.234.152
Hs#S6158998RTN4Reticulon 40.00986.190.000.002.902
Hs#S16886558AMZ2Archaemetzincins-20.002.340.412921.130.002
Hs#S4622514VPS29Vacuolar protein3.411972.390.000.911.612
sorting 29
homolog (S.cerevisiae)
Hs#S24303270COPS5COP9 constitutive0.003.902.490.001.292
photomorphogenic
homolog subunit 5
(Arabidopsis)
Hs#S34122629ADH5Alcohol0.570.000.00973.712.582
dehydrogenase 5
(class III), chi
polypeptide
Hs#S2294449MRPL1Mitochondrial3699.140.001062.130.000.002
ribosomal protein
L1
Hs#S4807293Transcribed locus1.142958.580.008.160.002
Hs#S1730224CCT4Chaperonin0.004.670.005.440.002
containing TCP1,
subunit 4 (delta)
Hs#S21592131SEC11ASEC11 homolog A0.000.0012.449.070.002
(S.cerevisiae)
Hs#S4279806RPL22Ribosomal protein2.270.00531.070.000.002
L22
Hs#S16819791MRNA; cDNA0.000.000.003.6317.912
DKFZp686D17123
(from clone
DKFZp686D17123)
Hs#S2821115DC2DC2 protein0.00986.190.000.004.842
Hs#S1731274CCNI616.520.000.831947.420.002
Hs#S16818021FAM6AFamily with#######0.000.00973.710.002
sequence similarity
60, member A
Hs#S4738740.000.001062.13973.710.002
Hs#S4719030Transcribed locus,1.14986.192124.270.000.002
weakly similar to
XP_001126181.1
similar to Cyclin-L2
(Paneth cell-
enhanced
expression protein)
isoform 2 [Homo
sapiens]
Hs#S2373081Transcribed locus0.000.00531.070.003.392
Hs#S27598574UBA52Ubiquitin A-52616.520.000.000.004.362
residue ribosomal
protein fusion
product 1
Hs#S6155739Transcribed locus0.000.001062.13973.710.002
Hs#S4264145Transcribed locus,1.140.002655.340.913.872
strongly similar to
NP_066953.1
isomerase A
isoform 1 [Homo
sapiens]
Hs#S1726877GSTM3Glutathione S-1.142.343.320.600.002
transferase M3
(brain)
Hs#S2654679Ndufv2CDNA fis, A-616.520.00531.070.000.972
COL04217, highly
similar to Homo
sapiens
mitochondrion,
NADH
dehydrogenase
subunit 2
Hs#S16889285ATP5J2ATP synthase, H+2.276.230.000.910.002
transporting,
mitochondrial F0
complex, subunit
F2
Hs#S1729877CALCOCO2Calcium binding0.003.12531.070.910.002
and coiled-coil
domain 2
Hs#S38795124HTATIP2HIV-1 Tat0.002958.58531.070.000.002
interactive protein
2, 30 kDa
Hs#S3988730FAM112BFamily with616.520.780.007.250.002
sequence similarity
112, member B
Hs#S1728131UAP1UDP-N-0.001972.390.8314.500.002
acteylglucosamine
pyrophosphorylase 1
Hs#S1730715MAP1B3′UTR of0.000.00531.073.630.002
hypothetical
protein (ORF1)
Hs#S7113046S1BS100 calcium0.000.00531.079.070.002
binding protein B
Hs#S3438442CYB5BCytochrome b50.00986.190.005.440.002
type B (outer
mitochondrial
membrane)
Hs#S24303189Zfp27Transcribed locus0.000.001062.130.0045.512
Hs#S40831018RPP3Ribonuclease6.811972.390.000.000.002
P/MRP 30 kDa
subunit
Hs#S21592214SDAD1SDA1 domain0.004.670.41973.710.002
containing 1
Hs#S2652741C7orf44Chromosome 76165.230.000.83973.710.002
open reading
frame 44
Hs#S5516781PLRG1Pleiotropic0.00986.190.00973.710.002
regulator 1 (PRL1
homolog,
Arabidopsis)
Hs#S16884266SF3A3Splicing factor 3a,0.00986.197.050.000.002
subunit 3, 60 kDa
Hs#S16820013NOL5ANucleolar protein0.003.122.490.000.002
5A (56 kDa with
KKE/D repeat)
Hs#S1730191GOT2Glutamic-0.002958.583.320.000.002
oxaloacetic
transaminase 2,
mitochondrial
(aspartate
aminotransferase
2)
Hs#S2949832HIGD1AHIG1 domain0.000.00531.071947.420.002
family, member 1A
Hs#S305321PPIGPeptidylprolyl616.520.000.00973.710.002
isomerase G
(cyclophilin G)
Hs#S935286Transcribed locus1.700.001062.13973.710.002
Hs#S1730674JUNJun oncogene0.000.000.00973.7146.482
Hs#S20337893IL2RGInterleukin 20.000.00531.070.003.872
receptor, gamma
(severe combined
immunodeficiency)
Hs#S16887291SERP1Stress-associated0.001.560.001947.422.902
endoplasmic
reticulum protein 1
Hs#S1731293KDELR2KDEL (Lys-Asp-Glu-7.951972.390.000.000.002
Leu) endoplasmic
reticulum protein
retention receptor 2
Hs#S21280884H3F3AH3 histone, family0.001972.391062.130.000.002
3A
Hs#S2139380DLG7Discs, large0.000.001062.131947.420.002
homolog 7
(Drosophila)
Hs#S4033407Transcribed locus616.520.00531.070.000.002
Hs#S4613263PTGES3Prostaglandin E0.001972.390.00973.710.002
synthase 3
(cytosolic)
Hs#S21305093RPL32Ribosomal protein2.270.000.003.630.002
L32
Hs#S4619982SFRS7Splicing factor,3.414930.970.000.000.002
arginine/serine-
rich 7, 35 kDa
Hs#S11046593C1orf14Chromosome 100.00986.19531.070.000.002
open reading
frame 104
Hs#S3990753CDNA FLJ30885 fis,3.410.780.002.720.002
clone
FEBRA2004987
Hs#S1731677SUPT16HSuppressor of Ty0.001972.390.003.630.002
16 homolog (S.cerevisiae)
Hs#S1732353NPEPPSAminopeptidase4.540.003.320.000.002
puromycin
sensitive
Hs#S16631302KTN1Kinectin 1 (kinesin1233.053.120.000.000.002
receptor)
Hs#S12428240.000.001593.205.440.002
Hs#S4046545TCEAL2Transcription2.270.00531.070.000.002
elongation factor A
(SII)-like 2
Hs#S1727241NRD1Nardilysin (N-616.521972.390.000.000.002
arginine dibasic
convertase)
Hs#S1728832CLTCClathrin, heavy0.00986.19531.070.000.002
chain (Hc)
Hs#S19626866YLPM1YLP motif0.000.00531.073.630.002
containing 1
Hs#S27228224USP16Ubiquitin specific616.521972.390.000.000.002
peptidase 16
Hs#S3218912CD44CD44 molecule0.000.00531.070.001270.652
(Indian blood
group)
Hs#S4192208CD63CD63 molecule616.520.002124.270.000.002
Hs#S857804616.520.000.003.630.002
Hs#S1970929WDSOF1WD repeats and1849.574.670.000.000.002
SOF1 domain
containing
Hs#S2649909FDPSFarnesyl0.00986.190.00973.710.002
diphosphate
synthase (farnesyl
pyrophosphate
synthetase,
dimethylallyltranstransferase,
geranyltranstransferase)
Hs#S4569950Transcribed locus616.520.000.000.00635.322
Hs#S16889879HMGB1High-mobility11.920.780.830.003811.942
group box 1
Hs#S16057065PDZD11PDZ domain0.0012.470.00973.710.002
containing 11
Hs#S16889002DNTTIP2Deoxynucleotidyltransferase,616.520.000.00973.710.002
terminal,
interacting protein 2
Hs#S1730362RAD21RAD21 homolog (S. pombe)0.00986.190.00973.710.002
Hs#S16975412S1A4S100 calcium0.003944.770.001947.420.002
binding protein A4
Hs#S2652251LSM5LSM5 homolog, U60.001972.392.490.000.002
small nuclear RNA
associated (S.cerevisiae)
Hs#S2816175TMEM126BTransmembrane0.001972.390.00973.710.002
protein 126B
Hs#S4616821UBE2V2Ubiquitin-0.001972.390.00973.710.002
conjugating
enzyme E2 variant 2
Hs#S15115685CSNK2A1616.521972.390.000.000.002
Hs#S16507502LOC28472Hypothetical616.520.000.00973.710.002
protein LOC284702
Hs#S1726314ATP6V1B2ATPase, H+1849.571972.390.000.000.002
transporting,
lysosomal
56/58 kDa, V1
subunit B2
Hs#S1728180VIL2Villin 2 (ezrin)616.520.000.000.00635.322
Hs#S17853742TIMM23Translocase of1233.05986.190.000.000.002
inner
mitochondrial
membrane 23
homolog (yeast)
Hs#S21592443MRPL42Mitochondrial0.000.002.491947.420.002
ribosomal protein
L42
Hs#S2293443SMU1Smu-1 suppressor616.520.000.001947.420.002
of mec-8 and unc-
52 homolog (C. elegans)
Hs#S2293771CMTM6CKLF-like MARVEL0.00986.190.00973.710.002
transmembrane
domain containing 6
Hs#S3323029PDCD1Programmed cell0.00986.190.00973.710.002
death 10
Hs#S4617593XBP1X-box binding0.000.002.491947.420.002
protein 1
Hs#S4831715FIP1L1FIP1 like 1 (S.cerevisiae)0.000.000.00973.71635.322
Hs#S4026538Transcribed locus,0.000.521062.13973.710.002
strongly similar to
XP_001130365.1
similar to S-phase
kinase-associated
protein 1A isoform
b [Homo sapiens]
Hs#S16817574CNIH4Cornichon0.000.00531.07973.710.002
homolog 4
(Drosophila)
Hs#S1974192BFARBifunctional0.001.561062.130.002541.302
apoptosis
regulator
Hs#S2434757Transcribed locus0.007.79531.070.000.002
Hs#S3220040RNF2Ring finger protein616.520.00531.070.000.002
20
Hs#S4384817RPS7Ribosomal protein0.000.00531.073894.840.002
S7
Hs#S6683006GABARAPL2GABA(A) receptor-616.520.00531.070.000.002
associated protein-
like 2
Hs#S1731150EIF3S1Eukaryotic2.840.00531.070.000.002
translation
initiation factor 3,
subunit 10 theta,
150/170 kDa
Hs#S2652974WDR61WD repeat domain0.000.00531.07973.710.002
61
Hs#S3990520ZMAT2Zinc finger, matrin0.000.00531.072921.130.002
type 2
Hs#S1394998Transcribed locus,0.001972.390.001947.420.002
strongly similar to
XP_001069671.1
similar to
peptidylprolyl
isomerase
(cyclophilin)-like 1
[Rattus norvegicus]
Hs#S19656426GARTPhosphoribosylglycinamide0.000.00531.071947.420.002
formyltransferase,
phosphoribosylglycinamide
synthetase,
phosphoribosylaminoimidazole
synthetase
Hs#S23238619Transcribed locus,2.270.000.00973.710.002
weakly similar to
XP_001165780.1
similar to
ribosomal protein
L11 [Pan
troglodytes]
Hs#S24302642YWHABTyrosine 3-0.001972.390.004868.550.002
monooxygenase/tryptophan
5-
monooxygenase
activation protein,
beta polypeptide
Hs#S2798208LOC38872Similar to ubiquitin2.271972.390.000.000.002
and ribosomal
protein S27a
precursor
Hs#S4373401ACAT2Acetyl-Coenzyme A0.000.000.00973.716988.562
acetyltransferase 2
(acetoacetyl
Coenzyme A
thiolase)
Hs#S4623136C6orf173Chromosome 60.000.00531.07973.710.002
open reading
frame 173
Hs#S4688195CKS2CDC28 protein0.000.00531.073894.840.002
kinase regulatory
subunit 2
Hs#S11154180PARNPoly(A)-specific0.000.00531.07973.710.002
ribonuclease
(deadenylation
nuclease)
Hs#S1503665Transcribed locus616.520.000.000.00635.322
Hs#S1560391Transcribed locus1849.570.000.00973.710.002
Hs#S16056991COX15COX15 homolog,0.000.001593.20973.710.002
cytochrome c
oxidase assembly
protein (yeast)
Hs#S16818161ZKSCAN5Zinc finger with8631.320.00531.070.000.002
KRAB and SCAN
domains 5
Hs#S16819564HNRPH1Heterogeneous0.003944.772124.270.000.002
nuclear
ribonucleoprotein
H1 (H)
Hs#S16885038SLC25A37Solute carrier0.000.001062.13973.710.002
family 25, member
37
Hs#S1728272AGLAmylo-1,6-1233.050.00531.070.000.002
glucosidase, 4-
alpha-
glucanotransferase
(glycogen
debranching
enzyme, glycogen
storage disease
type III)
Hs#S1729079PRDX6Peroxiredoxin 64932.18986.190.000.000.002
Hs#S1729804G3BP1GTPase activating0.000.002124.270.00635.322
protein (SH3
domain) binding
protein 1
Hs#S1729843SNUPNSnurportin 10.00986.190.000.00635.322
Hs#S1730168CA8Carbonic616.521972.390.000.000.002
anhydrase VIII
Hs#S1732156CHES1Checkpoint0.00986.190.00973.710.002
suppressor 1
Hs#S1969051NDFIP2Nedd4 family0.000.001062.13973.710.002
interacting protein 2
Hs#S20986282Transcribed locus0.00986.19531.070.000.002
Hs#S21274950SERPINB1Serpin peptidase616.520.00531.070.000.002
inhibitor, clade B
(ovalbumin),
member 1
Hs#S21292924PRPSAP1Phosphoribosyl0.001972.390.00973.710.002
pyrophosphate
synthetase-
associated protein 1
Hs#S21312161UBL5Ubiquitin-like 50.000.001062.130.003176.622
Hs#S2138838BAZ1A0.000.000.00973.716988.562
Hs#S2139186STAU2Staufen, RNA3082.610.000.00973.710.002
binding protein,
homolog 2
(Drosophila)
Hs#S2293866HIF1ANHypoxia-inducible0.002958.58531.070.000.002
factor 1, alpha
subunit inhibitor
Hs#S2294120PARLPresenilin616.520.00531.070.000.002
associated,
rhomboid-like
Hs#S23598070.001972.390.000.001905.972
Hs#S24849465NR3C1Nuclear receptor616.520.00531.070.000.002
subfamily 3, group
C, member 1
(glucocorticoid
receptor)
Hs#S26153487SC5DLSterol-C5-616.520.000.00973.710.002
desaturase (ERG3
delta-5-desaturase
homolog, S.cerevisiae)-
like
Hs#S29570898Transcribed locus0.000.000.00973.713176.622
Hs#S3293525Transcribed locus616.520.000.00973.710.002
Hs#S33939810SMC2Structural0.000.003186.40973.710.002
maintenance of
chromosomes 2
Hs#S34122865Q9H6U6616.520.00531.070.000.002
Hs#S3438886SLTMSAFB-like,0.002958.58531.070.000.002
transcription
modulator
Hs#S3488016Transcribed locus,0.000.00531.071947.420.002
moderately similar
to NP_066357.1
protein L36a
[Homo sapiens]
Hs#S3646083Transcribed locus0.000.000.00973.711270.652
Hs#S38657617Transcribed locus,616.520.001062.130.000.002
weakly similar to
NP_039502.1 b
[Schizosaccharomyces
pombe]
Hs#S4273337MRPS28Mitochondrial0.003944.770.00973.710.002
ribosomal protein
S28
Hs#S4283824USP33Ubiquitin specific0.000.001062.13973.710.002
peptidase 33
Hs#S4618631SCOCShort coiled-coil7398.270.000.001947.420.002
protein
Hs#S5516731Homo sapiens,616.524930.970.000.000.002
clone
IMAGE: 3923347,
mRNA
Hs#S5949691Transcribed locus,0.000.000.00973.71635.322
strongly similar to
XP_001064547.1
similar to
translocase of
outer
mitochondrial
membrane 7
homolog [Rattus
norvegicus]

[0000]

Genes giving rise to GSEs enriched by BrdU selection in at least one tumor cell line but
not in BJ-hTERT. Values representing over 2-fold enrichment are highlighted in yellow.
Relative enrichment in selected set# of
GeneMDA-selections
Unigene IDSymbolAnnotationHT1080PC3T24MB-321enriched
Hs#S16883457RPL23CDNA FLJ26326 fis, clone3944.77531.071947.420.003
HRT01120, highly similar to 60S
ribosomal protein L23
Hs#S40429152958.582.490.005.083
Hs#S22515139CGGBP1CGG triplet repeat binding2958.580.00973.7124.213
protein 1
Hs#S1730201PAICSPhosphoribosylaminoimidazole2958.582.490.00635.323
carboxylase,
phosphoribosylaminoimidazole
succinocarboxamide synthetase
Hs#S36352304SR14U2-associated SR140 protein1972.39531.071.81635.323
Hs#S37211072THOC2THO complex 23.120.001947.421905.973
Hs#S4407757COX7A2Cytochrome c oxidase subunit1.56531.072.723.393
VIIa polypeptide 2 (liver)
Hs#S3219417CYCSCytochrome c, somatic0.004.983.6310.413
Hs#S16889222MGST1Microsomal glutathione S-0.007.47973.719.683
transferase 1
Hs#S16975412S1A4S100 calcium binding protein A43944.770.001947.420.002
Hs#S16819564HNRPH1Heterogeneous nuclear3944.772124.270.000.002
ribonucleoprotein H1 (H)
Hs#S4273337MRPS28Mitochondrial ribosomal protein3944.770.00973.710.002
S28
Hs#S38795124HTATIP2HIV-1 Tat interactive protein 2,2958.58531.070.000.002
30 kDa
Hs#S1730191GOT2Glutamic-oxaloacetic2958.583.320.000.002
transaminase 2, mitochondrial
(aspartate aminotransferase 2)
Hs#S2293866HIF1ANHypoxia-inducible factor 1, alpha2958.58531.070.000.002
subunit inhibitor
Hs#S3438886SLTMSAFB-like, transcription2958.58531.070.000.002
modulator
Hs#S4807293Transcribed locus2958.580.008.160.002
Hs#S1728131UAP1UDP-N-acteylglucosamine1972.390.8314.500.002
pyrophosphorylase 1
Hs#S21280884H3F3AH3 histone, family 3A1972.391062.130.000.002
Hs#S4613263PTGES3Prostaglandin E synthase 31972.390.00973.710.002
(cytosolic)
Hs#S1731677SUPT16HSuppressor of Ty 16 homolog (S.cerevisiae)1972.390.003.630.002
Hs#S2652251LSM5LSM5 homolog, U6 small nuclear1972.392.490.000.002
RNA associated (S.cerevisiae)
Hs#S2816175TMEM126BTransmembrane protein 126B1972.390.00973.710.002
Hs#S4616821UBE2V2Ubiquitin-conjugating enzyme E21972.390.00973.710.002
variant 2
Hs#S1394998Transcribed locus, strongly similar1972.390.001947.420.002
to XP_001069671.1 similar to
peptidylprolyl isomerase
(cyclophilin)-like 1 [Rattus
norvegicus]
Hs#S24302642YWHABTyrosine 3-1972.390.004868.550.002
monooxygenase/tryptophan 5-
monooxygenase activation
protein, beta polypeptide
Hs#S21292924PRPSAP1Phosphoribosyl pyrophosphate1972.390.00973.710.002
synthetase-associated protein 1
Hs#S23598071972.390.000.001905.972
Hs#S6158998RTN4Reticulon 4986.190.000.002.902
Hs#S2821115DC2DC2 protein986.190.000.004.842
Hs#S3438442CYB5BCytochrome b5 type B (outer986.190.005.440.002
mitochondrial membrane)
Hs#S5516781PLRG1Pleiotropic regulator 1 (PRL1986.190.00973.710.002
homolog, Arabidopsis)
Hs#S16884266SF3A3Splicing factor 3a, subunit 3,986.197.050.000.002
60 kDa
Hs#S11046593C1orf14Chromosome 10 open reading986.19531.070.000.002
frame 104
Hs#S1728832CLTCClathrin, heavy chain (Hc)986.19531.070.000.002
Hs#S2649909FDPSFarnesyl diphosphate synthase986.190.00973.710.002
(farnesyl pyrophosphate
synthetase,
dimethylallyltranstransferase,
geranyltranstransferase)
Hs#S1730362RAD21RAD21 homolog (S. pombe)986.190.00973.710.002
Hs#S2293771CMTM6CKLF-like MARVEL986.190.00973.710.002
transmembrane domain
containing 6
Hs#S3323029PDCD1Programmed cell death 10986.190.00973.710.002
Hs#S1729843SNUPNSnurportin 1986.190.000.00635.322
Hs#S1732156CHES1Checkpoint suppressor 1986.190.00973.710.002
Hs#S20986282Transcribed locus986.19531.070.000.002
Hs#S4719030Transcribed locus, weakly similar986.192124.270.000.002
to XP_001126181.1 similar to
Cyclin-L2 (Paneth cell-enhanced
expression protein) isoform 2
[Homo sapiens]
Hs#S16057065PDZD11PDZ domain containing 1112.470.00973.710.002
Hs#S2434757Transcribed locus7.79531.070.000.002
Hs#S1730224CCT4Chaperonin containing TCP1,4.670.005.440.002
subunit 4 (delta)
Hs#S21592214SDAD1SDA1 domain containing 14.670.41973.710.002
Hs#S24303270COPS5COP9 constitutive3.902.490.001.292
photomorphogenic homolog
subunit 5 (Arabidopsis)
Hs#S2813090UBE2TUbiquitin-conjugating enzyme3.430.930.684.962
E2T (putative)
Hs#S1729877CALCOCO2Calcium binding and coiled-coil3.12531.070.910.002
domain 2
Hs#S16820013NOL5ANucleolar protein 5A (56 kDa with3.122.490.000.002
KKE/D repeat)
Hs#S16886558AMZ2Archaemetzincins-22.340.412921.130.002
Hs#S1726877GSTM3Glutathione S-transferase M32.343.320.600.002
(brain)
Hs#S16887291SERP1Stress-associated endoplasmic1.560.001947.422.902
reticulum protein 1
Hs#S1974192BFARBifunctional apoptosis regulator1.561062.130.002541.302
Hs#S4026538Transcribed locus, strongly similar0.521062.13973.710.002
to XP_001130365.1 similar to S-
phase kinase-associated protein
1A isoform b [Homo sapiens]
Hs#S21592131SEC11ASEC11 homolog A (S.cerevisiae)0.0012.449.070.002
Hs#S16819791MRNA; cDNA DKFZp686D171230.000.003.6317.912
(from clone DKFZp686D17123)
Hs#S4738740.001062.13973.710.002
Hs#S2373081Transcribed locus0.00531.070.003.392
Hs#S6155739Transcribed locus0.001062.13973.710.002
Hs#S1730715MAP1B3′UTR of hypothetical protein0.00531.073.630.002
(ORF1)
Hs#S7113046S1BS100 calcium binding protein B0.00531.079.070.002
Hs#S24303189Zfp27Transcribed locus0.001062.130.0045.512
Hs#S2949832HIGD1AHIG1 domain family, member 1A0.00531.071947.420.002
Hs#S1730674JUNJun oncogene0.000.00973.7146.482
Hs#S20337893IL2RGInterleukin 2 receptor, gamma0.00531.070.003.872
(severe combined
immunodeficiency)
Hs#S2139380DLG7Discs, large homolog 70.001062.131947.420.002
(Drosophila)
Hs#S12428240.001593.205.440.002
Hs#S19626866YLPM1YLP motif containing 10.00531.073.630.002
Hs#S3218912CD44CD44 molecule (Indian blood0.00531.070.001270.652
group)
Hs#S21592443MRPL42Mitochondrial ribosomal protein0.002.491947.420.002
L42
Hs#S4617593XBP1X-box binding protein 10.002.491947.420.002
Hs#S4831715FIP1L1FIP1 like 1 (S.cerevisiae)0.000.00973.71635.322
Hs#S16817574CNIH4Cornichon homolog 40.00531.07973.710.002
(Drosophila)
Hs#S4384817RPS7Ribosomal protein S70.00531.073894.840.002
Hs#S2652974WDR61WD repeat domain 610.00531.07973.710.002
Hs#S3990520ZMAT2Zinc finger, matrin type 20.00531.072921.130.002
Hs#S19656426GARTPhosphoribosylglycinamide0.00531.071947.420.002
formyltransferase,
phosphoribosylglycinamide
synthetase,
phosphoribosylaminoimidazole
synthetase
Hs#S4373401ACAT2Acetyl-Coenzyme A0.000.00973.716988.562
acetyltransferase 2 (acetoacetyl
Coenzyme A thiolase)
Hs#S4623136C6orf173Chromosome 6 open reading0.00531.07973.710.002
frame 173
Hs#S4688195CKS2CDC28 protein kinase regulatory0.00531.073894.840.002
subunit 2
Hs#S11154180PARNPoly(A)-specific ribonuclease0.00531.07973.710.002
(deadenylation nuclease)
Hs#S16056991COX15COX15 homolog, cytochrome c0.001593.20973.710.002
oxidase assembly protein (yeast)
Hs#S16885038SLC25A37Solute carrier family 25, member0.001062.13973.710.002
37
Hs#S1729804G3BP1GTPase activating protein (SH30.002124.270.00635.322
domain) binding protein 1
Hs#S1969051NDFIP2Nedd4 family interacting protein 20.001062.13973.710.002
Hs#S21312161UBL5Ubiquitin-like 50.001062.130.003176.622
Hs#S2138838BAZ1A0.000.00973.716988.562
Hs#S29570898Transcribed locus0.000.00973.713176.622
Hs#S33939810SMC2Structural maintenance of0.003186.40973.710.002
chromosomes 2
Hs#S3488016Transcribed locus, moderately0.00531.071947.420.002
similar to NP_066357.1 protein
L36a [Homo sapiens]
Hs#S3646083Transcribed locus0.000.00973.711270.652
Hs#S4283824USP33Ubiquitin specific peptidase 330.001062.13973.710.002
Hs#S5949691Transcribed locus, strongly similar0.000.00973.71635.322
to XP_001064547.1 similar to
translocase of outer
mitochondrial membrane 7
homolog [Rattus norvegicus]
Hs#S34122629ADH5Alcohol dehydrogenase 5 (class0.000.00973.712.582
III), chi polypeptide
Hs#S4264145Transcribed locus, strongly similar0.002655.340.913.872
to NP_066953.1 isomerase A
isoform 1 [Homo sapiens]
Hs#S935286Transcribed locus0.001062.13973.710.002

[0000]

New targets for cancer treatment identified by
GSE selection and verified by siRNA testing.
% growth inhibition in
MDA-
GeneMB-231HT1080
SymbolAnnotationcellsT24 cellscells
COPZ1Coatomer protein complex,70.4334.3193.14
subunit zeta 1
THOC2THO complex 246.637.3930.94
DPAGT1Dolichyl-phosphate51.6236.6763.20
(UDP-N-acetylglucosamine)
N-acetylglucosamine-
phosphotransferase 1
(GlcNAc-1-P transferase)
CGGBP1CGG triplet repeat25.6727.2363.45
binding protein 1
SR140U2-associated SR140 protein34.3826.7948.14

[0055]

The following examples are intended to further illustrate the invention and are not intended to be construed to limit the scope of the invention.

Example 1

Preparation of a Normalized Random Fragment shRNA Library from MCF-7 Breast Carcinoma Cells

[0056]

The shRNA library was prepared as follows. The strategy for shRNA library construction is depicted in FIG. 1. The starting material was a random-fragment (GSE) library of normalized cDNA from MCF7 breast carcinoma cells using previously described procedures (Primiano et al., 2003) and cloned in retroviral vector LmGCX (Kandel et al., 1997). cDNA inserts with their flanking 5′ and 3′ adaptors were amplified from the GSE library by PCR using adaptor-derived primers (Step 1). The primer corresponding to the 5′ adaptor was biotinylated, and the primer corresponding to the 3′ adaptor was sequence-modified to create a MmeI site at a position that allows for MmeI digestion within the cDNA sequence after random octanucleotide reverse transcription priming site. MmeI cuts within the cDNA sequence 18-20 nt away from its recognition site, thus producing a targeting sequence of a size suitable for shRNA. MmeI digestion was used to remove the adaptor and the octanucleotide-derived sequence, generating a two-nucleotide NN overhang at the 3′ end. The MmeI-digested 100-500 by fragments were gel-purified and ligated with hairpin adaptor (step 2), containing a NN overhang at the 3′ end. The ligated material was bound to Dynabeads® M-270 Streptavidin magnetic beads (Invitrogen/Dynal) and digested at the MmeI site in the hairpin adaptor (step 3), so that fragments containing the hairpin adaptor and 19 to 21 by of cDNA sequences could be separated from fragments containing the 5′ adaptor, which remained bound to the streptavidin beads. The purified fragments were then used for ligation with TA and subsequent steps of shRNA template generation, as described for the luciferase-derived library. The MmeI-generated fragments with 3′ NN overhangs were then ligated to a second adapter (the termination adaptor; TA) (step 4), which provides an internal primer for subsequent extension (step 5). TA contains a single-stranded nick that primes the extension with Klenow fragment without the need to denature the hairpin and anneal an external primer. TA also provides a Pol III termination signal and a 3′ (G/A)N overhang, which improves Pol III transcription by placing a purine at +1 position from the promoter (Goomer and Kunkel, 1992). Primer extension from the primer within TA (step 5) was performed with Klenow fragment of DNA polymerase I (Fermentas, Hanover, Md.). 139-bp to 143-bp long extended fragments were purified on an 8% TBE-polyacrylamide gel and digested with MlyI and XbaI restriction enzymes (step 6) to generate shRNA templates containing an inverted repeat followed by Pol III termination signal. The ˜78-80 by digestion product was purified on an 8% TBE-polyacrylamide gel, and then ligated into the LLCEP TU6LX expression vector (Maliyekkel et al., 2006) (step 7), which had been prepared by gel purification of plasmid digested with SrfI and XbaI to remove the CAT-ccdB cassette. The resulting library was transformed into ccdB-sensitive E. Cloni 10G Supreme (Lucigen, Middleton, Wis.), which selects for ccdB-free insert-containing clones. The shRNA library from normalized cDNA contained a total of 2.8×106clones. Sequence analysis of 676 randomly picked clones showed that 632 of them (93.5%) contained proper stem-and-loop inserts.

Example 2

Preparation of GSE Library from Normalized cDNA of Multiple Tumor Cell Lines

[0057]

Lung (A549, H69), colon (HCT116, SW480), breast (MCF-7, MDA-MB321), prostate (LNCaP, PC3), cervical (HeLa), ovarian (A2780), renal (ACHN) carcinomas cell lines, fibrosarcoma (HT1080), osteosarcoma (Saos-2) cell lines, melanoma (MALME-3M), glioblastoma (U251), chronic myelogenous leukemia (K562), promyelocytic leukemia (HL60), and acute lymphoblastic leukemia (CCRF-CEM) cell lines were obtained from ATCC. mRNA from these cell lines was used to prepare normalized cDNA, through duplex-specific nuclease (DSN) normalization (Zhulidov et al., 2004); the normalization was carried by Evrogen (Moscow, Russia) as a service. Normalization efficacy was tested by Q-PCR analysis of representation of cDNAs of seven transcripts with high (β-actin, GAPDH, EF1-α), medium (L32, PPMM) and low (Ubch5b, c-Yes) expression levels in parental cells. The representation of highly expressed transcripts decreased up to 70-fold in the normalized mixture, while the level of rare cDNAs increased up to 30-fold after normalization. Normalized cDNA mixture was fragmented by DNAse I digestion to obtain 100-500 by fragments, followed by end repair by treatment with T4 DNA polymerase and Klenow fragment as described (Gudkov and Roninson, 1997). cDNA fragments were amplified by ligation-mediated PCR. For amplification, adaptors containing translation start sites with Age I and Sph I restriction sites were used. cDNA fragments were digested with Age I and Sph I and ligated into a modified tetracycline/doxycycline-inducible vector, pLLCEm (Wiznerowicz and Trono, 2003), under the control of the CMV promoter. The ligation produced a library of approximately 260 million clones. The percent recombination in this library was assessed by direct sequencing of 192 clones. The number of clones containing an insert was >90%. The average length of the inserts was 135 bp.

Example 3

Preparation of Recipient Cell Lines for TGI Selection

[0058]

As the recipient cell lines for TGI selection, we have chosen four human cancer cell lines and human immortalized fibroblasts. The tumor cell lines are MDA-MB-231 breast carcinoma, PC3 prostate carcinoma, HT108 fibrosarcoma and T24 bladder carcinoma. The immortalized fibroblasts are BJ-hTERT. To obtain tetracycline/doxycycline-inducible cells, tTR-KRAB, a tetracycline/doxycycline-sensitive repressor was overexpressed in all the cell lines, by infecting them with a lentiviral vector expressing tTR-KRAB and dsRED fluorescent protein (Wiznerowicz and Trono, 2003), followed by two rounds of FACS selection for dsRed positive cells. To analyze the tetracycline/doxycycline-dependent regulation, tTR-KRAB expressing cell lines were infected with an EGFP-expressing tetracycline/doxycycline-inducible lentiviral vector. The level of activation of GFP expression by treatment with 100 ng/ml of doxycycline ranged from about 30-fold to 300-fold in different cell lines.

Example 4

Library Transduction and Selection for Doxycycline-Dependent Resistance to BrdU Suicide

[0059]

The shRNA library in pLLCE-TU6-LX vector described in above was transduced into MDA-MB-231 breast carcinoma cells expressing ttR-KRAB. The GSE library in pLLCEm lentiviral vector, described above, was transduced into all five cell lines. Lentiviral transduction was carried out using a pseudotype packaging system, by co-transfecting plasmid library DNA with Δ8.91 lentiviral packaging plasmid and VSV-G (pantropic receptor) plasmid into 293FT cells in DMEM with 10% FC2 using TransFectin reagent. 2.5×107recipient cells were infected with the shRNA library, and 1×108cells of each recipient cell line were infected with the GSE library. The infection rate (as determined by Q-PCR analysis of integrated provirus) was 95%. 25% of the infected cells were subjected to DNA purification, and the rest were plated at a density of 1×106cells per P150, to a total of 100 million cells. These cells were subjected to selection for Doxycycline-dependent resistance to BrdU suicide, as follows. Cells were treated with 0.1 μg/ml of doxycycline for 18 hrs, then with 0.1 μg/ml of doxycycline and 50 μM BrdU for 48 hrs. Cells were then incubated with 10 μM Hoechst 33258 for 3 hrs and illuminated with fluorescent white light for 15 min on a light box, to destroy the cells that replicated their DNA and incorporated BrdU in the presence of doxycycline. Cells were then washed twice with phosphate-buffered saline and allowed to recover in normal medium (DMEM, 10% FBS) for 7-10 days. The surviving cells were collected, followed by DNA purification. The cDNA fragments were amplified by PCR from genomic DNA extracted from the infected unselected and BrdU-selected cells using vector specific primers and subjected to ultra high throughput sequencing by 454 Life Science Inc (http://www.454.com/enabling-technology/the-process.asp).

Example 5

Enrichment and Functional Validation of Specific shRNA Sequences after BrdU Suicide Selection in MDA-MB-231 Cells

[0060]

High-throughput sequencing of shRNA sequences recovered by PCR from the genomic DNA of MDA-MB-231 cells before and after BrdU selection, followed by BLAST analysis, yielded 53201 sequences with homology to Unigene database entries before selection and 53803 sequences after selection. These sequences matched 14699 and 3316 Unigene clusters respectively. Among the genes found in the selected subset, 741 were targeted by four or more shRNA sequences (Table 1). The genes in Table 1 are sorted by the “enrichment factor” (EF), a value defined by multiplying the number of different shRNA sequences found to be enriched for each gene after BrdU suicide selection by the fold enrichment in the frequency of any shRNA sequences derived from the corresponding gene. By this criterion, which takes into account both the likelihood that the shRNA target gene has been correctly identified by being targeted by multiple shRNA sequences and the degree of enrichment, one of the most enriched genes was KRAS, a well-known oncogene that has undergone an activating, mutation in MDA-MB-231 cells (Kozma et al., 1987). This result validates the selection system as capable of identifying oncogenes, potential targets for anticancer drugs.

[0061]

To verify that genes enriched by the selection are required for MDA-MB-231 cell growth, we have selected 22 genes represented by at least two selected shRNA sequences and showing the highest EF value. We then used synthetic short interfering RNA (siRNA) targeting these genes and designed by Qiagen, Inc. according to Qiagen's siRNA design algorithms, for transfection into MDA-MB-231 cells, to determine if such siRNAs will inhibit cell growth. Four siRNAs per gene, obtained from Qiagen, were transfected into MDA-MB231 cells in 96-well plates, in triplicates, using Silentfect® transfection reagent (Biorad) and manufacturer's instructions, and 5 nM of siRNA per well. A cytotoxic mixture of siRNA derived from several essential genes (Qiagen, All-star Cell Death Hs siRNA, #1027298), was used as a positive control, and siRNAs targeting either no known genes (Qiagen, Negative Control siRNA #1022076) or the Green Fluorescent Protein (GFP) (Qiagen, GFP-22 siRNA, #1022064) were used as negative controls. Cells were cultured in DMEM media with 10% FBS serum, and the relative cell number was determined six days after siRNA transfection by staining cellular DNA with Hoechst 33342 (Polysciences Inc; #23491-52-3). As shown in FIG. 2A, 1-4 siRNAs per gene, targeting 19 of 22 tested genes (86%), inhibited cell growth to a greater degree than either of the negative controls, with KRAS targeting siRNAs showing the strongest effect. In contrast, none of siRNAs targeting 10 genes that were not enriched by selection inhibited cell growth (FIG. 2B). Hence, BrdU suicide selection enriches for genes that are required for tumor cell growth. Some of the genes tested and found to be essential fore growth in FIG. 2A have not been previously implicated in cell growth or carcinogenesis. These 12 genes, listed in Table 2, represent potential new targets for cancer treatment.

Example 6

Enrichment and Functional Validation of Specific GSE Sequences after BrdU Suicide Selection in Different Cell Lines

[0062]

Sequencing of GSE fragments recovered by PCR from the genomic DNA of five different cell lines before and after BrdU selection was followed by BLAST analysis. The numbers of cDNA fragment sequences with homology to Unigene database entries revealed by BLAST analysis in each PCR product and the number of sequences enriched two or more fold by BrdU selection are shown in Table 3. Among the selected genes, 178 were enriched in two or more different cell lines (Table 4), and 98 genes were enriched in tumor cell lines only but not in BJ-hTERT (Table 5). These genes represent potential targets for cancer treatment.

[0063]

To verify the growth-regulatory activity of 26 genes enriched by GSE selection, we have used transfection of the corresponding siRNAs from Qiagen siRNA collection, four siRNAs per gene, as described in section 5 above. In these assays we have used HT1080 fibrosarcoma cells (3 days analysis after transfection) T24 bladder carcinoma (3 days analysis after transfection), and MDA-MB-231 breast carcinoma cells (6 day analysis after transfection). The results presented in FIG. 3 show significant inhibition of cell growth by siRNAs against 81% (21 of 26) of the tested genes. Some of the genes tested and found to be essential for growth in FIG. 3 have not been previously implicated in cell growth or carcinogenesis. These genes, listed in Table 6, represent potential new targets for cancer treatment.

Example 7

Inhibition of COPZ1 is Cytotoxic for Multiple Tumor Cell Lines Expressing Low Levels of COPZ2

[0064]

Among the new potential targets listed in Table 6, we have investigated in greater detail COPZ1, which was targeted by GSEs identified in BrdU-selected populations of tumor cell lines HT1080, MDA-MB-231, T24, and PC3, but not in immortalized normal BJ-hTERT fibroblasts. COPZ1 encodes CopI-ζ1, one of the two isoforms of a coatomer of COPI secretory vesicles involved in Golgi to ER and Golgi to Golgi traffic (Beck et al., 2009). The other CopI-ζ isoform, CopI-ζ2, is encoded by the COPZ2 gene; the two CopI-ζ proteins have 75% amino acid identity (Wegmann et al., 2004). CopI-1 and CopI-ζ2 are alternative components of a dimeric complex that also includes one of the two isoforms of CopI-γ, encoded by another pair of closely related genes, COPG1 and COPG2. The CopI-ζ/CopI-γ dimers interact within COPI complexes with additional CopI proteins, which are encoded by the genes COPA, COPB1, COPB2, COPD and COPE (Wegmann et al., 2004; Moelleken et al., 2007).

[0065]

As shown in FIG. 3, siRNAs targeting COPZ1 inhibited HT1080, MDA-MB-231 and T24 cell proliferation. The target sequences of siRNAs used for COPZ1 knockdown and for the knockdown of other COPI genes analyzed herein are listed in Table 7.

[0000]

Target sequences of siRNAs used for the knockdown
of the indicated genes.
GenesiRNATarget sequence
COPZ1Qiagen AAGCGATTTAAATTGTATTGAA
COPZ1Qiagen BTTGGCTGTGGATGAAATTGTA
COPZ1Qiagen CTTGGGAATAGTTCATAGGGAA
COPZ1Qiagen DTCCCAGCATATTTAGATAATA
COPZ1Thermo ScientificGGACAAUGAUGGAGAUCGA
(pool of 4 siRNAs)CAACAAGACCCAUCGGACU
GGGAAUAGUUCAUAGGGAA
AUUGGAGCUCCUAUGAAA
COPAQiagen ATCCCACTGAGTTCAAATTCAA
COPAQiagen BCTGGATTTCAACAGCTCCAAA
COPAQiagen CCTGGCGCATGAATGAATCAAA
COPAQiagen DAAGCTTAATGACCTCATCCAA
COPAQiagen 5CACACGGGTGAAGGGCAACAA
COPAThermo ScientificACUCAGAUGGUGUGUAAUA
(pool of 4 siRNAs)GCAAUAUGCUACACUAUGA
GAACAUUCGUGUCAAGAGU
GCGGAGUGGUUCCAAGUUUU
COPB1Qiagen ACAGGATCACACTATCAAGAAA
COPB1Qiagen BCAAGGATTGGTTATAATATAA
COPB1Qiagen CCAGAATTGCTAGAACCTTTAA
COPB1Qiagen DCACCAACATGGTTGATTTAAA
COPB2Qiagen AACGATTCTTCAGAGTATGCAA
COPB2Qiagen BCAGGTTTCAAGGGTAGTGAAA
COPB2Qiagen CCAGTACGTATTTGGCATTCAA
COPB2Qiagen DCTGCTAGATCTGATCGAGTTA
COPEQiagen ACCGGAAGGAGCTGAAGAGAAT
COPEQiagen BCAGAGCTGTCAGGACCATGAA
COPEQiagen CCCCGGAAGGAGCTGAAGAGAA
COPEQiagen DATCTGTTAATAAATATCTCAA
COPGQiagen AAGGCCCGTGTATTTAATGAAA
COPGQiagen BCCGAGCCACCTTCTACCTAAA
COPGQiagen CCACCGACTCCACTATGTTGAA
COPGQiagen DTCCGTCGGATGTGCTACTTGA
COPG2Qiagen ACAGGTGACTGTCAGAAGTAAA
COPG2Qiagen BCTGCATCAAGTGATAATATTA
COPG2Qiagen CGACGCGATTGTTTCAATCTAA
COPG2Qiagen DAGGCTCGTATATTCAATGAAA
COPS8Qiagen DCTGCATTTGTTCAATAAATAT
COPZ2Qiagen ACTGGCCTTAACTCATATCTTA
COPZ2Qiagen BCAGCATTGACCTCTTCCTATA
COPZ2Qiagen CAACAAATTAAATGGTCGTTAT
COPZ2Qiagen DCCGGCTGCTGGCCAAGTATTA
COPZ2Thermo ScientificGGGCUCAUCCUACGAGAAU
(pool of 4 siRNAs)UCUUGGUGCUGGACGAGAU
CAACAAGACCAGCCGGACU
GAACAAAUUAAAUGGUCGU

[0066]

The knockdown of COPZ1 by siRNA was verified by quantitative reverse transcription-PCR (QPCR), as described (VanGuilder et al., 2008). The sequences of the primers used to amplify GAPDH and RPL13A (normalization standards), COPZ1 and other COPI component genes analyzed herein are listed in Table 8. QPCR analysis showed that COPZ1 Qiagen B and COPZ1. Qiagen D siRNAs decreased COPZ1 mRNA levels in MDA-MB-231, PC3 and BJ-hTERT cells by >95% relative to cells transfected with a control siRNA targeting no known genes (Qiagen).

[0000]

Primer sequences for QPCR of the indicated genes.
GeneSenseAntisense
GAPDHAGGTGAAGGTCGGAGTCAGGTCATTGATGGCAACAA
RPL13AAGATGGCGGAGGTGCAGGGCCCAGCAGTACCTGTTTA
COPZ1ACACTGGGGTAGGTGTCGTCAAGATGGAGGCGCTGATTTT
COPZ2CCTTCTGGATCACTTGCTGGGGTTGCTGGAGAACATGGAC
COPATATCAACCTCCCATGCCTTTACCCCACTATGCCCCTTATT
COPB1TCTGAAACTTGTGGAAAAGCACACAATTTCTGTCACTTGC
COPB2GCTCTGTAGGATGCAGATCCAGTAGCCGGTAACAAACGAGG
UniversalAACGAGACGACGACAGACTTT
miRNA
miR-152TCAGTGCATGACAGAACTTG

[0067]

The knockdown of COPA or COPB was reported to cause the collapse of endoplasmic reticulum and Golgi compartments and cellular traffic arrest (Styers et al., 2008). Disruption of intracellular traffic either by inhibition of COPI complex formation or by blocking COPI assembly on Golgi membrane by inhibition of adenosine diphosphate ribosylation factor with brefeldin A (Donaldson et al., 1991; Fujiwara et al., 1988) resulted in cell death (Citterio et al., 2008; Shao et al., 1996). Additionally COPA or COPB knockdown inhibits the maturation of the autophagosome (Razi et al., 2009), an essential step in autophagy, a process involving the degradation of cell components through lysosomes. Autophagy is a physiological program that plays a role in cell growth, development, and homeostasis (Mizushima et al., 2008), and therefore interference with autophagy may result in cell death (Platini et al., 2010; Filimonenko et al., 2007). To determine if COPZ1 knockdown, like that of COPA or COPB, interferes with autophagy and causes Golgi disruption, we have transfected COPZ1 siRNA (from Thermo Scientific; Table 7), in parallel with siRNAs targeting COPA and COPZ2, into PC3 cells expressing LC3, a protein marker of autophagosomes fused with Green Fluorescent Protein (GFP-LC3) (Fung et al., 2008). The knockdown effects on autophagosome accumulation and Golgi integrity were analyzed 72 hrs later by fluorescence microscopy analysis after staining with monoclonal antibodies against a Golgi marker GM130 (Golgi membrane protein 130 kD, BD Bioscience) and GFP-LC3 localization. Fluorescent microscopy analysis (FIG. 4A) shows that COPA and COPZ1-targeting but not control or COPZ2-targeting siRNAs cause fragmentation and disappearance of GM130 positive structures and accumulation of GFP-positive puncta. Knockdown of COPA and COPZ1 but not of COPZ2 also resulted in the accumulation of a 43 kd form of GFP-LC3 that becomes conjugated with phosphatidilethanolamine (PE) within the autophagosome, increasing its electrophoretic mobility (FIG. 4B); the accumulation of this form is indicative of accumulation of autophagosomes and inhibition of autophagic flux (Klionsky et al., 2008a; Klionsky et al., 2008b; Fass et al., 2006). These events closely resemble the previously reported effects of COPI complex disruption by COPA and COPB knockdown (Razi et al., 2009), indicating that COPZ1 siRNA, as expected, acts by disrupting the formation or function of COPI. We have also analyzed the ability of COPZ1 siRNA to induce cell death, as evidenced by membrane permeability revealed by the uptake of the fluorescent dye DAPI, as measured by flow cytometry. PC3 cells were transfected with COPZ1 siRNA (from Thermo Scientific), with negative control siRNA, and with siRNA targeting COPA (positive control). 4 days after transfection, the fractions of membrane-permeable (DAPI+) dead cells were 1.9% for cells transfected with negative control siRNA, 36.7% for cells transfected with COPA siRNA, 3.8% for cells transfected with COPZ2 siRNA and 29.7% for cells transfected with COPZ1 siRNA, indicating that COPZ1 (but not COPZ2) knockdown efficiently induces cell death. Hence, COPZ1 knockdown produces the phenotypic effects expected from COPI inhibition, and these effects—inhibition of autophagy and the disruption of Golgi—are likely to be responsible for the induction of cell death by COPZ1 knockdown.

[0068]

To determine if siRNA knockdown of the other COPI components would mimic the antiproliferative effect of COPZ1 siRNA, we have compared the effects of siRNAs targeting COPA, COPB1, COPB2, COPE, COPG1, COPG2, COPZ1 and COPZ2 on the proliferation of HT1080, MDA-MB-231, T24 and PC3 tumor cell lines and immortalized normal BJ-hTERT fibroblasts. This analysis was conducted through the same experimental setup as in the experiments shown in FIG. 2 and FIG. 3, using 4 siRNAs against each gene target (from Qiagen) and the same positive and negative siRNA controls as in FIG. 2 and FIG. 3. The results of this analysis are shown in FIG. 5. 1-4 siRNAs targeting most of the tested genes strongly inhibited the proliferation of all four tumor cell lines. The exceptions were COPG2 and COPZ2, where the corresponding siRNAs largely failed to inhibit the growth of tumor cell lines, with only a single COPG2 siRNA significantly inhibiting the growth of one cell line (PC3), and a single COPZ2 targeting siRNA (COPZ2 Qiagen B) inhibiting HT1080 and MDA-MB-231 proliferation and marginally inhibiting T24 proliferation (the latter effect was statistically insignificant, P>0.4, T-test) (FIG. 5). (As discussed below, inhibition of proliferation of some cell lines by COPZ2 Qiagen B siRNA is likely to represent an off-target effect.) 3-4 siRNAs against most of the tested genes strongly inhibited the proliferation of normal BJ-hTERT cells. The exceptions were COPG2 and COPZ2, where only 1 of 4 siRNAs had a weak growth-inhibitory effect, and COPZ1, where only 1 of 4 siRNAs showed an apparent effect, which, however, was statistically insignificant (P>0.5, T-test). The failure of COPZ1-targeting siRNAs to inhibit BJ-hTERT was in striking contrast to the effects of these siRNAs on the four tumor cell lines, all of which were strongly inhibited by at least one COPZ1-targeting siRNA (FIG. 5).

[0069]

The differential effect of COPZ1 siRNAs on tumor and normal cells was verified using an independent set of siRNAs (from Thermo Scientific; Table 7). FIG. 6 shows the effects of different siRNAs on the cell number of PC3 prostate carcinoma and BJ-hTERT normal fibroblasts (in this figure, the Y axis shows the cell number rather than % growth inhibition). COPZ1 siRNA from Thermo Scientific and two COPZ1 siRNAs from Qiagen strongly inhibited PC3 cell proliferation but had no effect on the proliferation of BJ-hTERT. BJ-hTERT proliferation, however, was inhibited by all three siRNAs targeting COPA (two from Qiagen and one from Thermo Scientific); COPA siRNA from Thermo Scientific was also tested and found to inhibit the proliferation of PC3 cells. COPZ2 siRNA (from Thermo Scientific) failed to inhibit the proliferation of either PC3 or BJ-hTERT. The results of the experiments in FIG. 5 and FIG. 6 demonstrate that COPZ1 is the only component of the COPI complex (with a possible exception for COPD that was not tested), the knockdown of which selectively inhibits the proliferation of tumor cells but not of normal fibroblasts.

[0070]

To understand why the knockdown of COPZ1 but not of the other COPI proteins selectively inhibits the proliferation of tumor cells relative to normal fibroblasts, we have measured the expression of COPZ1, COPZ2, COPA, COPB1 and COPB2 in BJ-hTERT, HT1080, MDA-MB-231, T24, and PC3 cell lines by QPCR, using primers listed in Table 8. FIG. 7A shows the results of these measurements, where the levels of the corresponding mRNAs in each cell line are displayed relative to their level in normal BJ-hTERT cells. COPZ1, COPA, COPB I and COPB2 showed comparable expression levels in all the cell lines but, strikingly, the expression of COPZ2 in the four tumor cell lines was negligible relative to its expression in BJ-hTERT (FIG. 7A). The lack of COPZ2 in tumor cell lines explains the failure of most of the tested COPZ2 siRNAs to inhibit the growth of these cell lines and suggests that moderate inhibitory effect of a single COPZ2-targeting siRNA (COPZ2 Qiagen B) in some of these cell lines most likely represents an off-target effect. FIG. 7B compares the expression of the same set of genes in three isogenic cell lines with increasing degrees of neoplastic transformation that were derived by Hahn et al. (Hahn et al., 1999) from normal BJ fibroblasts by sequential transduction with hTERT (cell line BJ-EN, similar to BJ-hTERT), early-region SV40 (cell line BJ-ELB, partially transformed) and KRAS (cell line BJ-ELR, fully transformed). Strikingly, the expression of COPZ2 was decreased 2.5-3 fold in BJ-ELB and BJ-ELR relative to BJ-EN, whereas none of the other genes showed significant changes in their expression. These results indicate that downregulation of COPZ2 but not of other COPI coatomers is associated with neoplastic transformation.

[0071]

We have expanded the QPCR analysis of COPZ2 and COPZ1 expression to a large set of different normal human tissues (from Ambion) (FIG. 8A) and additional tumor and leukemia cell lines (FIG. 8B). COPZ2 showed comparable expression levels among most of the normal tissues, except for lower expression in the ovary and spleen and very low expression in the thymus; COPZ1 expression was more uniform (FIG. 8A). Almost all the tumor and leukemia cell lines showed greatly decreased expression of COPZ2 relative to BJ-hTERT, but no similar decrease was observed for COPZ1 expression (FIG. 8B). The only COPZ2-expressing tumor cell line in FIG. 8B was WM 793 melanoma line, which was originally isolated from a superficial spreading melanoma and which displays poor tumorigenicity in nude mice (Kobayashi et al., 1994), indicating a relatively benign nature. We have also compared COPZ1 and COPZ2 mRNA levels among four melanoma cell lines and two samples of normal primary melanocytes (a gift of Dr. M. Nikiforov, Roswell Park Cancer Institute, Buffalo, N.Y.). As shown in FIG. 8C, COPZ1 levels were comparable among the normal melanocyte and melanoma cells, but COPZ2 levels were drastically decreased in all four melanoma lines relative to both normal melanocyte populations. Hence, COPZ2 downregulation is a broad and general event in different forms of cancer.

[0072]

COPZ2 downregulation in cancer cells offers an explanation for tumor-selective cytotoxicity of COPZ1-targeting siRNAs. Since COPZ1 and COPZ2 gene products are alternative components of CopI-ζ/CopI-γ dimers, it is likely that they can substitute for each other, and that COPI complexes remain functional if either COPZ1 or COPZ2 gene products are present. Therefore, COPZ1 knockdown is not toxic to normal cells that express COPZ2.

[0073]

However, COPZ2 is expressed at very low levels or not at all in tumor cells, and therefore such cells become dependent on COPZ1 for normal COPI function and survival. Therefore, COPZ1 knockdown kills COPZ2-deficient tumor cells but not COPZ2-proficient normal cells. To test this explanation, we asked if the restoration of COPZ2 expression in tumor cells would protect them from killing by COPZ1 siRNA. We have cloned full-length COPZ1 and COPZ2 cDNAs from MGC cDNA collection (distributed by Open Biosystems) into a lentiviral expression vector pLenti6-bsd-FLAG constructed in our laboratory, which expresses the cloned protein with a FLAG tag at the C-terminus. These recombinant lentiviruses (as well as the insert-free vector) were then transduced into PC3 cells. The transduced cells were selected with blasticidine and tested for the expression of COPZ1 and COPZ2 by immunoblotting, using FLAG-specific antibody (M2 Anti-FLAG, Sigma-Aldrich) and antibodies specific for COPZ1 (D20 anti-COPZ antibody, Santa-Cruz Biotechnology) and COPZ2 (a gift of Dr. F. Wieland, University of Heidelberg). The results of this analysis, shown in FIG. 9A, demonstrate the expected expression of FLAG-tagged COPZ1 and COPZ2 in cells transduced with the corresponding vectors. Notably, COPZ1-expressing vector increased cellular levels of the COPZ1 protein more than an order of magnitude relative to endogenous COPZ1 expression (FIG. 9A). Overexpression of either COPZ1 or COPZ2 had no apparent effect on PC3 cell growth.

[0074]

FIG. 9B shows the effects of siRNAs targeting COPA (three siRNAs), COPZ1 (three siRNAs) and COPZ2 (one siRNA) on cell proliferation of PC3 cells transduced with the insert-free vector or with the vectors expressing COPZ1 or COPZ2. COPA siRNAs inhibited the proliferation of all three cell populations. COPZ1 siRNAs inhibited the proliferation of cells transduced with the insert-free vector, but overexpression of either COPZ1 or COPZ2 rendered cells completely or partially resistant to COPZ1 knockdown (FIG. 9B). The protective effect of COPZ1 overexpression can be explained by a drastic increase in COPZ1 protein levels relative to the endogenous level of this protein (FIG. 9A), suggesting that COPZ1 knockdown by siRNA in COPZ1 lentivirus-transduced cells decreases COPZ1 expression to a level similar to the endogenous level in control cells and therefore sufficient for survival. On the other hand, the resistance of COPZ2-overexpressing cells to COPZ1 siRNA demonstrates that COPZ2 can substitute for COPZ1, in agreement with our hypothesis.

[0075]

COPZ2 siRNA alone had no effect on the proliferation of any of the three PC3 populations (FIG. 9), as expected since the original PC3 cells express COPZ1 but not COPZ2. We have also analyzed the effects of COPZ1 and COPZ2 knockdown on normal BJ-hTERT cells which, unlike PC3, express both COPZ1 and COPZ2. Knockdown of either COPZ1 or COPZ2 alone had no effect on BJ-hTERT proliferation, but a combination of COPZ1 and COPZ2 siRNAs drastically inhibited BJ-hTERT growth, as did COPA knockdown (FIG. 10A,B). Moreover, fluorescent microscopy and GFP-LC3 electrophoretic mobility analysis showed that the knockdown of either COPZ1 or COPZ2 alone did not affect Golgi structure and autophagy in BJ-hTERT cells, while simultaneous knockdown of both COPZ1 and COPZ2 caused Golgi disruption and inhibition of autophagy in these cells, as also did COPA knockdown (FIG. 11). The results of the experiments in FIGS. 9-11 demonstrate that the sensitivity of tumor cells to COPZ1 knockdown is the consequence of COPZ2 downregulation in such cells. These results also demonstrate that tumor selectivity of the antiproliferative effect of COPZ1 inhibition requires that such inhibition be selective for COPZ1 versus COPZ2, since the inhibition of COPZ1 and COPZ2 together would affect both tumor and normal cells.

[0076]

The reason for COPZ2 downregulation in tumor cells is presently unknown. However, COPZ2 gene contains in one of its introns a gene encoding the precursor of a microRNA (miRNA) mIR-152 (Weber, 2005; Rodriguez et al., 2004). miRNAs are pleiotropic regulators of gene expression, a number of which have been identified as playing important roles in cancer, either as oncogenes or as tumor suppressors (Ryan et al., 2010). Remarkably, mIR-152 was shown to be downregulated in clinical samples of several types of cancer, including breast cancer where mIR-152 gene is hypermethylated (Lehmann et al., 2008), endometrial serous adenocarcinoma where decreased expression of miR-152 was a statistically independent risk factor for overall survival (Hiroki et al., 2010), cholangiocarcinoma (Braconi et al., 2010) and gastric and colorectal cancers, where low expression of miR-152 was correlated with increased tumor size and advanced pT stage (Chen and Carmichael, 2010). Furthermore, mIR-152 overexpression in cholangiocarcinoma cells decreased cell proliferation (Braconi et al., 2010), and mIR-132 overexpression in a placental human choriocarcinoma cell line sensitized the cells to lysis by natural killer cells (Zhu et al., 2010). Hence, mIR-152 displays expression changes and biological activities indicative of a tumor suppressor. Many miRNAs located within protein-coding genes are transcriptionally linked to the expression of their host genes (Stuart et al., 2004), and a correlation between COPZ2 and mIR-152 expression has been noted among normal tissues (Bak et al., 2008). Therefore, COPZ2 downregulation in cancers could be a corollary of the downregulation of a tumor-suppressive miRNA mIR-152. To test this hypothesis, we have measured mIR-152 expression in a series of cell lines where COPZ2 expression has been determined, using QPCR with a combination of the universal miRNA (Hurteau et al., 2006) and miR-152 specific primers (Table 8). The results of this analysis, shown in FIG. 12, demonstrate that mIR-152, like COPZ2, was strongly downregulated in all the tumor cell lines and in in vitro transformed BJ-ELB and BJ-ELR cells, relative to normal BJ-EN fibroblasts. This result indicates that tumors susceptible to the inhibition of COPZ1 can be identified on the basis of decreased expression of either COPZ2 or mIR-152.

REFERENCES

[0000]

  • Abagyan, R. and Totrov, M. (2001). High-throughput docking for lead generation. Curr. Opin. Chem. Biol. 5, 375-382.
  • Ashihara, E., Kawata, E., and Maekawa, T. (2010). Future prospect of RNA interference for cancer therapies. Curr. Drug Targets. 11, 345-360.
  • Asif-Ullah, M., Levesque, M., Robichaud, G., and Perreault, J. P. (2007). Development of ribozyme-based gene-inactivations; the example of the hepatitis delta virus ribozyme. Curr. Gene Ther. 7, 205-216.
  • Bak, M., Silahtaroglu, A., Moller, M., Christensen, M., Rath, M. F., Skryabin, B., Tommerup, N., and Kauppinen, S. (2008). MicroRNA expression in the adult mouse central nervous system. RNA. 14, 432-444.
  • Beck, R., Rawet, M., Wieland, F. T., and Cassel, D. (2009). The COPI system: molecular mechanisms and function. FEBS Lett. 583, 2701-2709.
  • Bennett, C. F. and Swayze, E. E. (2010). RNA targeting therapeutics: molecular mechanisms of antisense oligonucleotides as a therapeutic platform. Annu. Rev. Pharmacol. Toxicol. 50, 259-293.
  • Boute, N., Jockers, R., and Issad, T. (2002). The use of resonance energy transfer in high-throughput screening: BRET versus FRET. Trends Pharmacol. Sci. 23, 351-354.
  • Boutros, M. and Ahringer, J. (2008). The art and design of genetic screens: RNA interference. Nat. Rev. Genet. 9, 554-566.
  • Braconi, C., Huang, N., and Patel, T. (2010). MicroRNA-dependent regulation of DNA methyltransferase-1 and tumor suppressor gene expression by interleukin-6 in human malignant cholangiocytes. Hepatology 51, 881-890.
  • Chen, L. L. and Carmichael, G. G. (2010). Decoding the function of nuclear long non-coding RNAs. Curr. Opin. Cell Biol.
  • Citterio, C., Vichi, A., Pacheco-Rodriguez, G., Aponte, A. M., Moss, J., and Vaughan, M. (2008). Unfolded protein response and cell death after depletion of brefeldin A-inhibited guanine nucleotide-exchange protein GBF1. Proc. Natl. Acad. Sci. U.S.A 105, 2877-2882.
  • Degorce, F., Card, A., Soh, S., Trinquet, E., Knapik, G. P., and Xie, B. (2009). HTRF: A technology tailored for drug discovery—a review of theoretical aspects and recent applications. Curr. Chem. Genomics 3, 22-32.
  • Donaldson, J. G., Kahn, R. A., Lippincott-Schwartz, J., and Klausner, R. D. (1991). Binding of ARF and beta-COP to Golgi membranes: possible regulation by a trimeric G protein. Science 254, 1197-1199.
  • Fass, E., Shvets, E., Degani, I., Hirschberg, K., and Elazar, Z. (2006). Microtubules support production of starvation-induced autophagosomes but not their targeting and fusion with lysosomes. J. Biol. Chem. 281, 36303-36316.
  • Filimonenko, M., Stuffers, S., Raiborg, C., Yamamoto, A., Malerod, L., Fisher, E. M., Isaacs, A., Brech, A., Stenmark, H., and Simonsen, A. (2007). Functional multivesicular bodies are required for autophagic clearance of protein aggregates associated with neurodegenerative disease. J. Cell Biol. 179, 485-500.
  • Freelove, A. C. and Zheng, R. (2002). The power of ribozyme technologies: the logical way ahead for molecular medicine and gene therapy? Curr. Opin. Mol. Ther. 4, 419-422.
  • Fujiwara, T., Oda, K., Yokota, S., Takatsuki, A., and Ikehara, Y. (1988). Brefeldin A causes disassembly of the Golgi complex and accumulation of secretory proteins in the endoplasmic reticulum. J. Biol. Chem. 263, 18545-18552.
  • Fung, C., Lock, R., Gao, S., Salas, E., and Debnath, J. (2008). Induction of autophagy during extracellular matrix detachment promotes cell survival. Mol. Biol. Cell 19, 797-806.
  • Goomer, R. S. and Kunkel, G. R. (1992). The transcriptional start site for a human U6 small nuclear RNA gene is dictated by a compound promoter element consisting of the PSE and the TATA box. Nucleic Acids Res 20, 4903-4912.
  • Gudkov, A. and Roninson, I. B. (1997). Isolation of genetic suppressor elements (GSEs) from random fragment cDNA libraries in retroviral vectors. In Methods in Molecular Biology: cDNA library protocols, I. G. Cowell and C. A. Austin, eds. (Totowa, N.J.: Humana Press), pp. 221-240.
  • Hahn, W. C., Counter, C. M., Lundberg, A. S., Beijersbergen, R. L., Brooks, M. W., and Weinberg, R. A. (1999). Creation of human tumour cells with defined genetic elements. Nature 400, 464-468.
  • Hiroki, E., Akahira, J., Suzuki, F., Nagase, S., Ito, K., Suzuki, T., Sasano, H., and Yaegashi, N. (2010). Changes in microRNA expression levels correlate with clinicopathological features and prognoses in endometrial serous adenocarcinomas. Cancer Sci. 101, 241-249.
  • Holzmayer, T. A., Pestov, D. G., and Roninson, I. B. (1992). Isolation of dominant negative mutants and inhibitory antisense RNA sequences by expression selection of random DNA fragments. Nucleic Acids Res. 20, 711-717.
  • Hurteau, G. J., Spivack, S. D., and Brock, G. J. (2006). Potential mRNA degradation targets of hsa-miR-200c, identified using informatics and qRT-PCR. Cell Cycle 5, 1951-1956.
  • Kandel, E. S., Chang, B. D., Schott, B., Shtil, A. A., Gudkov, A. V., and Roninson, I. B. (1997). Applications of green fluorescent protein as a marker of retroviral vectors. Somat. Cell Mol. Genet. 23, 325-340.
  • Klionsky, D. J., Abeliovich, H., Agostinis, P., Agrawal, D. K., Aliev, G., Askew, D. S., Baba, M., Baehrecke, E. H., Bahr, B. A., Ballabio, A., Bamber, B. A., Bassham, D. C., Bergamini, E., Bi, X., Biard-Piechaczyk, M., Blum, J. S., Bredesen, D. E., Brodsky, J. L., Brumell, J. H., Brunk, U. T., Bursch, W., Camougrand, N., Cebollero, E., Cecconi, F., Chen, Y., Chin, L. S., Choi, A., Chu, C. T., Chung, J., Clarke, P. G., Clark, R. S., Clarke, S. G., Clave, C., Cleveland, J. L., Codogno, P., Colombo, M. I, Coto-Montes, A., Cregg, J. M., Cuervo, A. M., Debnath, J., Demarchi, F., Dennis, P. B., Dennis, P. A., Deretic, V., Devenish, R. J., Di Sano, F., Dice, J. F., Difiglia, M., Dinesh-Kumar, S., Distelhorst, C. W., Djavaheri-Mergny, M., Dorsey, F. C., Droge, W., Dron, M., Dunn, W. A., Jr., Duszenko, M., Eissa, N. T., Elazar, Z., Esclatine, A., Eskelinen, E. L., Fesus, L., Finley, K. D., Fuentes, J. M., Fueyo, J., Fujisaki, K., Galliot, B., Gao, F. B., Gewirtz, D. A., Gibson, S. B., Gohla, A., Goldberg, A. L., Gonzalez, R., Gonzalez-Estevez, C., Gorski, S., Gottlieb, R. A., Haussinger, D., He, Y. W., Heidenreich, K., Hill, J. A., Hoyer-Hansen, M., Hu, X., Huang, W. P., Iwasaki, A., Jaattela, M., Jackson, W. T., Jiang, X., Jin, S., Johansen, T., Jung, J. U., Kadowaki, M., Kang, C., Kelekar, A., Kessel, D. H., Kiel, J. A., Kim, H. P., Kimchi, A., Kinsella, T. J., Kiselyov, K., Kitamoto, K., Knecht, E., Komatsu, M., Kominami, E., Kondo, S., Kovacs, A. L., Kroemer, G., Kuan, C. Y., Kumar, R., Kundu, M., Landry, J., Laporte, M., Le, W., Lei, H. Y., Lenardo, M. J., Levine, B., Lieberman, A., Lim, K. L., Lin, F. C., Liou, W., Liu, L. F., Lopez-Berestein, G., Lopez-Otin, C., Lu, B., Macleod, K. F., Malorni, W., Martinet, W., Matsuoka, K., Mautner, J. Meijer, A. J., Melendez, A., Michels, P., Miotto, G., Mistiaen, W. P., Mizushima, N., Mograbi, B., Monastyrska, I., Moore, M. N., Moreira, P. I., Moriyasu, Y., Motyl, T., Munz, C., Murphy, L. O., Naqvi, N. I., Neufeld, T. P., Nishino, I., Nixon, R. A., Noda, T., Nurnberg, B., Ogawa, M., Oleinick, N. L., Olsen, L. J., Ozpolat, B., Paglin, S., Palmer, G. E., Papassideri, I., Parkes, M., Perlmutter, D. H., Perry, G., Piacentini, M., Pinkas-Kramarski, R., Prescott, M., Proikas-Cezanne, T., Raben, N., Rami, A., Reggiori, F., Rohrer, B., Rubinsztein, D. C., Ryan, K. M., Sadoshima, J., Sakagami, H., Sakai, Y., Sandri, M., Sasakawa, C.; Sass, M., Schneider, C., Seglen, P. O., Seleverstov, O., Settleman, J., Shacka, J. J., Shapiro, I. M., Sibirny, A., Silva-Zacarin, E. C., Simon, H. U., Simone, C., Simonsen, A., Smith, M. A., Spanel-Borowski, K., Srinivas, V., Steeves, M., Stenmark, H., Stromhaug, P. E., Subauste, C. S., Sugimoto, S., Sulzer, D., Suzuki, T., Swanson, M. S., Tabas, I., Takeshita, F., Talbot, N. J., Talloczy, Z., Tanaka, K., Tanaka, K., Tanida, I., Taylor, G. S., Taylor, J. P., Terman, A., Tettamanti, G., Thompson, C. B., Thumm, M., Tolkovsky, A. M., Tooze, S. A., Truant, R., Tumanovska, L. V., Uchiyama, Y., Ueno, T., Uzcategui, N. L., van, d. K., I, Vaquero, E. C., Vellai, T., Vogel, M. W., Wang, H. G., Webster, P., Wiley, J. W., Xi, Z., Xiao, G., Yahalom, J., Yang, J. M., Yap, G., Yin, X. M., Yoshimori, T., Yu, L., Yue, Z., Yuzaki, M., Zabirnyk, O., Zheng, X., Zhu, X., and Deter, R. L. (2008a). Guidelines for the use and interpretation of assays for monitoring autophagy in higher eukaryotes. Autophagy. 4, 151-175.
  • Klionsky, D. J., Elazar, Z., Seglen, P. O., and Rubinsztein, D. C. (2008b). Does bafilomycin A1 block the fusion of autophagosomes with lysosomes? Autophagy. 4, 849-950.
  • Kobayashi, H., Man, S., MacDougall, J. R., Graham, C. H., Lu, C., and Kerbel, R. S. (1994). Variant sublines of early-stage human melanomas selected for tumorigenicity in nude mice express a multicytokine-resistant phenotype. Am. J. Pathol. 144, 776-786.
  • Kozma, S. C., Bogaard, M. E., Buser, K., Saurer, S. M., Bos, J. L., Groner, B., and Hynes, N. E. (1987). The human c-Kirsten ras gene is activated by a novel mutation in codon 13 in the breast carcinoma cell line MDA-MB231. Nucleic Acids Res 15, 5963-5971.
  • Lehmann, U., Hasemeier, B., Christgen, M., Muller, M., Romermann, D., Langer, F., and Kreipe, H. (2008). Epigenetic inactivation of microRNA gene hsa-mir-9-1 in human breast cancer. J. Pathol. 214, 17-24.
  • Maliyekkel, A., Davis, B. A., and Roninson I. B. (2006). Cell cycle arrest drastically extends the duration of gene silencing after transient expression of short hairpin RNA. Cell Cycle 5, 2390-2395.
  • Mizushima, N., Levine, B., Cuervo, A. M., and Klionsky, D. J. (2008). Autophagy fights disease through cellular self-digestion. Nature 451, 1069-1075.
  • Moelleken, J., Malsam, J., Betts, M. J., Movafeghi, A., Reckmann, I., Meissner, I., Hellwig, A., Russell, R. B., Sollner, T., Brugger, B., and Wieland, F. T. (2007). Differential localization of coatomer complex isoforms within the Golgi apparatus. Proc. Natl. Acad. Sci. U.S.A 104, 4425-4430.
  • Pestov, D. G. and Lau, L. F. (1994). Genetic selection of growth-inhibitory sequences in mammalian cells. Proc Natl. Acad Sci. U.S.A 91, 12549-12553.
  • Platini, F., Perez-Tomas, R., Ambrosio, S., and Tessitore, L. (2010). Understanding autophagy in cell death control. Curr. Pharm. Des 16, 101-113.
  • Primiano, T., Baig, M., Maliyekkel, A., Chang, B. D., Fellars, S., Sadhu, J., Axenovich, S. A., Holzmayer, T. A., and Roninson, I. B. (2003). Identification of potential anticancer drug targets through the selection of growth-inhibitory genetic suppressor elements. Cancer Cell 4, 41-53.
  • Razi, M., Chan, E. Y., and Tooze, S. A. (2009). Early endosomes and endosomal coatomer are required for autophagy. J. Cell Biol. 185, 305-321.
  • Rodriguez, A., Griffiths-Jones, S., Ashurst, J. L., and Bradley, A. (2004). Identification of mammalian microRNA host genes and transcription units. Genome Res. 14, 1902-1910.
  • Roninson, I. B., Gudkov, A. V., Holzmayer, T. A., Kirschling, D. J., Kazarov, A. R., Zelnick, C. R., Mazo, I. A., Axenovich, S., and Thimmapaya, R. (1995). Genetic suppressor elements: new tools for molecular oncology—thirteenth Cornelius P. Rhoads Memorial Award Lecture. Cancer Res. 55, 4023-4028.
  • Ryan, B. M., Robles, A. I., and Harris, C. C. (2010). Genetic variation in microRNA networks: the implications for cancer research. Nat. Rev. Cancer 10, 389-402.
  • Shao, R. G., Shimizu, T., and Pommier, Y. (1996). Brefeldin A is a potent inducer of apoptosis in human cancer cells independently of p53. Exp. Cell Res. 227, 190-196.
  • Song, C. M., Lim, S. J., and Tong, J. C. (2009). Recent advances in computer-aided drug design. Brief. Bioinform. 10, 579-591.
  • Stetten, G., Davidson, R. L., and Latt, S. A. (1977). 33258 Hoechst enhances the selectivity of the bromodeoxyuridine—light method of isolating conditional lethal mutants. Exp. Cell Res. 108, 447-452.
  • Stuart, R. O., Wachsman, W., Berry, C. C., Wang-Rodriguez, J., Wasserman. L., Klacansky, I., Masys, D., Arden, K., Goodison, S., McClelland, M., Wang, Y., Sawyers, A., Kalcheva, I., Tarin, D., and Mercola, D. (2004). In silico dissection of cell-type-associated patterns of gene expression in prostate cancer. Proc. Natl. Acad. Sci. U.S.A 101, 615-620.
  • Styers, M. L., O'Connor, A. K., Grabski, R., Cormet-Boyaka, E., and Sztul, E. (2008). Depletion of beta-COP reveals a role for COP-1 in compartmentalization of secretory compartments and in biosynthetic transport of caveolin-1. Am. J. Physiol Cell Physiol 294, C1485-C1498.
  • Vaishnaw, A. K., Gollob, J., Gamba-Vitalo, C., Hutabarat, R., Sah, D., Meyers, R., de Fougerolles, T., and Maraganore, J. (2010). A status report on RNAi therapeutics. Silence. 1, 14.
  • VanGuilder, H. D., Vrana, K. E., and Freeman, W. M. (2008). Twenty-five years of quantitative PCR for gene expression analysis. Biotechniques 44, 619-626.
  • Weber, M. J. (2005). New human and mouse microRNA genes found by homology search. FEBS J. 272, 59-73.
  • Wegmann, D., Hess, P., Baier, C., Wieland, F. T., and Reinhard, C. (2004). Novel isotypic gamma/zeta subunits reveal three coatomer complexes in mammals. Mol. Cell Biol. 24, 1070-1080.
  • Wiznerowicz, M. and Trono, D. (2003). Conditional suppression of cellular genes: lentivirus vector-mediated drug-inducible RNA interference. J. Virol. 77, 8957-8961.
  • Yu, W., Lin, J., Jin, C., and Xia, B. (2009). Solution structure of human zeta-COP: direct evidences for structural similarity between COP I and clathrin-adaptor coats. J. Mol. Biol. 386, 903-912.
  • Zhu, X. M., Han, T., Wang, X. H., Li, Y. H., Yang, H. G., Luo, Y. N., Yin, G. W., and Yao, Y. Q. (2010). Overexpression of miR-152 leads to reduced expression of human leukocyte antigen-G and increased natural killer cell mediated cytolysis in JEG-3 cells. Am. J. Obstet. Gynecol. 202, 592-597.
  • Zhulidov, P. A., Bogdanova, E. A., Shcheglov, A. S., Vagner, L. L., Khaspekov, G. L., Kozhemyako, V. B., Matz, M. V., Meleshkevitch, E., Moroz, L. L., Lukyanov, S. A., and Shagin, D. A. (2004). Simple cDNA normalization using kamchatka crab duplex-specific nuclease. Nucleic Acids Res. 32, e37.

Как компенсировать расходы
на инновационную разработку
Похожие патенты