CELL POPULATIONS FOR POLYPEPTIDE ANALYSIS AND USES OF SAME

Abstract
Nucleic acid construct systems are disclosed. The constructs comprise: (i) a first nucleic acid construct comprising a first nucleic acid sequence encoding a first reporter polypeptide linked to an additional nucleic acid sequence capable of inserting the first nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to the first reporter polypeptide is expressed in the cell; and(ii) a second nucleic acid construct comprising a second nucleic acid sequence encoding a second reporter polypeptide, linked to an additional nucleic acid sequence capable of inserting in a non-directed manner the second nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to the second reporter polypeptide is expressed in the cell, wherein the first reporter polypeptide and the second reporter polypeptide are distinguishable.
Description
FIELD AND BACKGROUND OF THE INVENTION

The present invention, in some embodiments thereof, relates to cells comprising endogenous polypeptides attached to reporter polypeptides and uses thereof.


Genomic technology has advanced to a point at which, in principle, it has become possible to determine complete genomic sequences and to quantitatively measure the mRNA levels for each gene expressed in cell populations. Comparative cDNA array analysis and related technologies have been used to determine induced changes in gene expression at the mRNA level by concurrently monitoring the expression level of a large number of genes (in some cases all the genes) expressed by the investigated cell population/culture or tissue. Furthermore, biological and computational techniques have been used to correlate specific function with gene sequences.


These methods are highly effective for analyzing homogeneous populations of cells but loose their differentiation power when applied to heterogeneous populations due to large variability and averaging effects. Accordingly, the interpretation of the data obtained by these techniques in the context of the structure, control and mechanism of biological systems has been recognized as a considerable challenge. In particular, it has been extremely difficult to explain the mechanism of biological processes by genomic analysis alone.


Proteins are essential for the control and execution of virtually every biological process. Their rate of synthesis and half-life are controlled post-transcriptionally. Their level of expression is therefore not directly apparent from the gene sequence or even the expression level of the corresponding mRNA transcript. It is therefore essential that a complete description of a biological system includes measurements that indicate the identity, quantity and location of the proteins which constitute the system. An ideal measurement system would: (a) work at the level of individual cells, because experiments that average over cell populations can miss events that occur in only a subset of cells. Furthermore, averaging can miss all-or-none effects, and cell-cell variability; (b) follow cells over extended periods of time to reveal phenomena such as oscillations and temporal programs and (c) make minimal perturbations to the state of the cells.


At present no protein analytical technology approaches the throughput and level of automation of genomic technology. The most common implementation of proteome analysis is based on the separation of complex protein samples most commonly by two-dimensional gel electrophoresis (2DE) and the subsequent sequential identification of the separated protein species. This approach has been assisted by the development of powerful mass spectrometric techniques and the development of computer algorithms which correlate protein and peptide mass spectral data with sequence databases and thus rapidly identify proteins. This technology (two-dimensional mass spectrometry) has reached a level of sensitivity which now permits the identification of essentially any protein which is detectable by conventional protein staining methods including silver staining. However, the sequential manner in which samples are processed limits the sample throughput. In addition, the most sensitive methods have been difficult to automate and low abundance proteins, such as regulatory proteins, escape detection without prior enrichment, thus effectively limiting the dynamic range of the technique. In the 2DE/(MS)n method, proteins are quantified by densitometry of stained spots in the 2DE gels.


The development of methods and instrumentation for automated, data-dependent electrospray ionization (ESI) tandem mass spectrometry (MS)n in conjunction with microcapillary liquid chromatography (μLC) and database searching has significantly increased the sensitivity and speed of the identification of gel-separated proteins. As an alternative to the 2DE/(MS)n approach to proteome analysis, the direct analysis by tandem mass spectrometry of peptide mixtures generated by the digestion of complex protein mixtures has been proposed [Dongr'e et al., Trends Biotechnol 15:418-425 (1997)]. μLC-MS/MS has also been used successfully for the large-scale identification of individual proteins directly from mixtures without gel electrophoretic separation [Link et al., Nat Biotech, 17:676-682 (1999); Opitek et al., Anal Chem 69:1518-1524 (1997)]. While these approaches accelerate protein identification and assay protein modifications, they usually average over many cells and do not allow quantification of dynamics in individual cells.


There have also been advances in high-throughput quantification of protein levels and localizations at the single-cell level using antibody staining and microscopy. However, as staining of internal proteins requires the killing of the cell, it is not possible to follow protein dynamics in the same cell over time. A dynamic proteomics method in individual cells can complement antibody and mass spectrometry-based approaches.


Dynamic measurements in living cells are made possible by the use of fluorescent proteins as genetic tags. Labeling with fluorescent tags often leaves the wild-type localization intact. A library of cells containing GFP-labeled cDNAs, expressed under an exogenous promoter, has been created to investigate protein localization on the scale of the proteome [Bannasch, D. et al. Nucleic Acids Res. 32 Database issue, D505-D508 (2004); Simpson, J. C., et al EMBO Rep. 1, 287-292 (2000)]. A disadvantage of this approach is that exogenous expression gives no information about the transcriptional regulation of the gene, and potentially leads to non-physiological levels of expression. To follow wild-type regulation, homologous recombination can be used to integrate sequences of fluorescent proteins into the genome at the wild-type locus. This approach was made high throughput in yeast [Huh, W. K. et al. Nature, 425, 686-691 (2003)]. High-throughput homologous recombination is also being developed in mouse embryonic stem (ES) cells in the KOMP, EUCOMM and N or COMM initiatives. However, as yet, high-throughput homologous recombination has not been achieved in human cells.


Another tagging approach for analyzing proteins is known as central dogma (CD) tagging. This method labels proteins in their native chromosomal locations without the need for homologous recombination [Sigal et al., Nature Protocols, Vol 2, No. 6, 2007; Sigal et al., Nature Methods, Vol 3, No. 7, 2006; Sigal et al., Nature 444, October 2006, p. 643-646, Jarvik J, Biotechniques. 2002 October; 33(4):852-4, 856, 858-60 passim]. CD tagging labels genes by integrating a DNA sequence coding for a fluorescent tag into the genome. The tag is inserted in a non-directed manner using a retrovirus. It is marked as an exon by flanking splice acceptor and donor sequences. If the tag integrates within an expressed gene, it is then spliced into the gene's mRNA and a fusion protein is translated. The identity of the labeled gene is then determined by rapid amplification of cDNA end (RACE).


SUMMARY OF THE INVENTION

According to an aspect of some embodiments of the present invention there is provided a nucleic acid construct system comprising:


(i) a first nucleic acid construct comprising a first nucleic acid sequence encoding a first reporter polypeptide linked to an additional nucleic acid sequence capable of inserting the first nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to the first reporter polypeptide is expressed in the cell; and


(ii) a second nucleic acid construct comprising a second nucleic acid sequence encoding a second reporter polypeptide, linked to an additional nucleic acid sequence capable of inserting in a non-directed manner the second nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to the second reporter polypeptide is expressed in the cell, wherein the first reporter polypeptide and the second reporter polypeptide are distinguishable.


According to some embodiments of the invention, the nucleic acid construct system further comprises a third nucleic acid construct comprising a third nucleic acid sequence encoding the first reporter polypeptide linked to an additional nucleic acid sequence capable of inserting the third nucleic acid construct into a genome of a host cell such that an additional endogenous polypeptide covalently attached to the first reporter polypeptide is expressed in the cell.


According to some embodiments of the invention, the additional nucleic acid sequence of the first nucleic acid construct directs insertion of the first nucleic acid construct into the host cell in a directed manner.


According to some embodiments of the invention, the additional nucleic acid sequence of the first nucleic acid construct directs insertion of the first nucleic acid construct into the host cell in a non-directed manner.


According to some embodiments of the invention, the host cell is a mammalian cell.


According to some embodiments of the invention, the first nucleic acid construct comprises a retroviral sequence.


According to some embodiments of the invention, the second nucleic acid construct comprises a retroviral sequence.


According to some embodiments of the invention, the first nucleic acid construct comprises a transposon sequence.


According to some embodiments of the invention, the second nucleic acid construct comprises a transposon sequence.


According to some embodiments of the invention, a 3′ end of the first and the second reporter is flanked by a splice acceptor sequence and a 5′ end of the first and the second reporter is flanked by a splice donor sequence.


According to some embodiments of the invention, the first reporter and the second reporter are fluorescent polypeptides that fluoresce at a distinguishable wave length.


According to another aspect of some embodiments of the present invention there is provided a cell expressing at least two endogenous polypeptides, each covalently attached to a distinguishable reporter polypeptide.


According to some embodiments of the invention, at least one of the at least two endogenou polypeptides has a higher nuclear:cytoplasm expression ratio.


According to some embodiments of the invention, the cell expresses an additional endogenous polypeptide attached to a reporter polypeptide, the reporter polypeptide being identical to one of the two distinguishable reporter polypeptides.


According to some embodiments of the invention, the at least one of the at least two endogenous polypeptides is constitutive.


According to some embodiments of the invention, the cell comprises the nucleic acid construct system of the present invention.


According to some embodiments of the invention, the cell is a diseased cell.


According to some embodiments of the invention, the cell is a cancer cell.


According to some embodiments of the invention, the cell is viable.


According to an aspect of some embodiments of the present invention there is provided a cell population, wherein each cell of the population expresses at least two endogenous polypeptides, each covalently attached to a distinguishable reporter polypeptide, wherein at least one of the at least two endogenous polypeptides is identical in each cell of the cell population.


According to some embodiments of the invention, the cell population expresses an additional endogenous polypeptide attached to a reporter polypeptide, the reporter polypeptide being identical to one of the two distinguishable reporter polypeptides.


According to some embodiments of the invention, both of the at least two endogenous polypeptides are identical in each cell of the cell population.


According to some embodiments of the invention, the cell population is viable.


According to some embodiments of the invention, at least one of the at least two endogenous polypeptides comprises a sequence as set forth in SEQ ID NOs: 1-164.


According to some embodiments of the invention, the cell population comprises diseased cells.


According to an aspect of some embodiments of the present invention there is provided an isolated polypeptide comprising an amino acid sequence as set forth in SEQ ID NOs: 1-164.


According to an aspect of some embodiments of the present invention there is provided a method of generating a cell population, the method comprising:


(a) introducing a first nucleic acid construct into the cell population, the first nucleic acid construct comprising a first nucleic acid sequence encoding a first reporter polypeptide linked to an additional nucleic acid sequence capable of inserting the first nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to the first reporter polypeptide is expressed in the cell; and subsequently


(b) introducing a second nucleic acid construct into the cell population, the second nucleic acid construct comprising a second nucleic acid sequence encoding a second reporter polypeptide, linked to an additional nucleic acid sequence capable of inserting in a non-directed manner the second nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to the second reporter polypeptide is expressed in the cell, wherein the first reporter polypeptide and the second reporter polypeptide are distinguishable,


thereby generating the cell population.


According to some embodiments of the invention, the method further comprises introducing a third nucleic acid construct into the cell population prior to introducing the second nucleic acid construct, the third nucleic acid construct comprising a third nucleic acid sequence encoding the first reporter polypeptide linked to an additional nucleic acid sequence capable of inserting the third nucleic acid construct into a genome of a host cell such that an additional endogenous polypeptide covalently attached to the first reporter polypeptide is expressed in the cell.


According to some embodiments of the invention, the method further comprises:


(a) selecting a cell following administration of the first nucleic acid construct, wherein the first reporter comprises a higher nuclear:cytoplasm expression ratio;


(b) propagating the cell to generate a second population of cells; and


(c) introducing into the second population of cells the second nucleic acid construct.


According to some embodiments of the invention, the method further comprises identifying at least one of the endogenous polypeptides.


According to another aspect of some embodiments of the present invention there is provided a method of identifying a target of an agent, the method comprising:


(a) contacting the cell population of the present invention with the agent;


(b) analyzing a localization or amount of at least one of the endogenous polypeptides, wherein a change in the amount or localization is indicative of a target of the agent.


According to some embodiments of the invention, the analyzing is effected in real-time.


According to some embodiments of the invention, the agent is a therapeutic agent.


According to an aspect of some embodiments of the present invention there is provided a method of identifying an agent capable of affecting a cell state, the method comprising,


(a) contacting the cell population of the present invention, with an agent; wherein at least one of the endogenous polypeptides is a marker for the cell state; and


(b) measuring a localization or amount of the marker, wherein a change in the amount or localization of the marker is indicative of an agent capable of affecting the cell state.


According to some embodiments of the invention, the cell state is a disease state.


According to some embodiments of the invention, the marker is a therapeutic target.


According to an aspect of some embodiments of the present invention there is provided a method of identifying a marker for disease prognosis, the method comprising:


(a) contacting the cell population of the present invention with a therapeutic agent;


(b) comparing a localization or amount of the at least one endogenous polypeptide in responsive cells of the cell population with non-responsive cells of the cell population; wherein a difference in expression or localization of the at least one endogenous polypeptide in responsive and non-responsive cells is indicative that the endogenous polypeptide is the marker for disease prognosis.


According to an aspect of some embodiments of the present invention there is provided a method of isolating a polypeptide, the method comprising contacting a cell population expressing an endogenous polypeptide covalently attached to a reporter polypeptide with an antibody under conditions that allow specific binding between the antibody and the reporter polypeptide, thereby isolating the polypeptide.


According to an aspect of some embodiments of the present invention there is provided a method of analyzing a localization of a first and second endogenous polypeptide in a cell, the method comprising detecting a localization of the first and second endogenous polypeptide in the cell, wherein the first and second polypeptide are each covalently attached to a distinguishable reporter polypeptide, thereby analyzing localization of a first and second polypeptide.


According to an aspect of some embodiments of the present invention there is provided a method of treating a cancer comprising co-administering to a subject in need thereof a therapeutically effective amount of Camptothecin and an agent capable of downregulating DNA helicase DDX5 as set forth in SEQ ID NO: 165 or replication factor C activator 1 (RFC1) as set forth in SEQ ID NO: 166, thereby treating the cancer.


According to some embodiments of the invention, the agent is a silencing oligonucleotide.


According to some embodiments of the invention, the cancer is ovarian or colon cancer.


According to an aspect of some embodiments of the present invention there is provided a pharmaceutical composition comprising as an active ingredient camptothecin and an agent capable of downregulating DNA helicase DDX5 of SEQ ID NO: 165 or replication factor C activator 1 (RFC1) of SEQ ID NO: 166 and a pharmaceutically acceptable carrier.


Unless otherwise defined, all technical and/or scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the invention pertains. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of embodiments of the invention, exemplary methods and/or materials are described below. In case of conflict, the patent specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and are not intended to be necessarily limiting.





BRIEF DESCRIPTION OF THE DRAWINGS

Some embodiments of the invention are herein described, by way of example only, with reference to the accompanying drawings and images. With specific reference now to the drawings in detail, it is stressed that the particulars shown are by way of example and for purposes of illustrative discussion of embodiments of the invention. In this regard, the description taken with the drawings makes apparent to those skilled in the art how embodiments of the invention may be practiced.


In the drawings:



FIGS. 1A-E are photographs and schemes illustrating how the library of tagged proteins was generated. Cell clones in the library were created in two steps: First a red fluorescent tag flanked by splice signals (mCherry) was introduced on a retrovirus into the genome of H1299 cells, resulting in cells that express proteins with an internal mCherry exon. After two rounds of tagging, a cell clone was selected with a red labeling pattern that is suitable for image analysis, bright in the nucleus and weaker in the cytoplasm. This clone formed the basis for an additional round of tagging, with a yellow fluorescent tag (eYFP or Venus) as an internal exon. Individual YFP tagged cells were sorted, expanded into clones, and the tagged protein in each clone was identified.



FIGS. 2A-D are photographs illustrating image analysis of the library of the present invention. Image analysis used the red fluorescent images to automatically detect cell and nuclear boundaries and to quantitate the yellow fluorescent protein intensity in each compartment at each time-point.



FIGS. 3A-D are cell images in the presence and absence of the drug Camptothecin (CPT). Cells were grown in an incubated microscope for 24 hours, and then for an additional 48 hours in the presence of 10 μM CPT. Cells were imaged every 20 minutes, and fluorescent intensity in each cell was automatically tracked. Cell divisions and morphological changes associated with cell death were automatically detected. FIGS. 3B-D show a schematic of two daughter cells of the cell in 3A. The cell labeled with the blue track shows blebbing and fragmentation typical of apoptosis.



FIGS. 4A-C are pie charts comparing protein localizations on LARC (Library of Annotated Reporter Clones) database vs. all proteins in GO (Gene Ontology Consortium). Distributions of protein localizations for: FIG. 4A—proteins in LARC with published localization; FIG. 4B—all proteins in GO; FIG. 4C—“uknown” proteins in LARC based on manual inspection. (These proteins include hypothetical proteins and proteins encoded from regions in the genome denoted as ESTs and mRNA. These proteins have no published localization).



FIGS. 5A-S are graphs illustrating the results of immunoblots against 19 selected proteins. For each protein: blue line consists of 141 fluorescent measurements taken at a 20 minute resolution for 47 hours, red line denotes quantification of immunoblotting analysis (measurement taken at 0, 8.5, 17, 24, 36, 40 and 45 hours following drug (CPT) addition. Average correlation between the two measurements across all proteins is R=0.6. Error bars denote standard errors.



FIG. 6 is a graph illustrating the rate of cell death following addition of CPT. Red line denotes the fraction of dead cells at each time point following CPT addition for over 60 hours (time resolution—20 minutes). Error bars denote standard errors.



FIGS. 7A-I are graphs illustrating examples of day to day repeats of experiment for several clones. Experiment was repeated between 2 to 8 times for 9 different clones of 9 unique proteins. Thin blue lines denote normalized total fluorescence averaged over many cells in one experiment, bold line denotes average over all days, error bars denote standard error. Mean Coefficient of variance (std/mean) over all clones and all time points of all proteins is 0.13 (mean correlation between experiments at different dates is R=0.8).



FIGS. 8A-D are graphs and plots illustrating the broad temporal patterns of protein fluorescence intensity in response to drug. FIG. 8A: Examples of YFP-tagged protein intensities of individual cells, over 48 hours after drug addition. One example is show from each of the five profiles i-v. Thin lines—individual cells, bold black lines—population averages. FIG. 8B: Normalized fluorescence shows widespread waves of accumulation and decrease in intensity. Each row corresponds to one protein averaged over all cells in the movie at each time-point (at least 30 cells). Proteins were clustered according to their dynamics. TOP1 is indicated by an arrow. FIG. 8C: Ribosomal proteins show correlated dynamics (P<10−3). Cytoskeleton-related proteins show behaviors either correlated or anti-correlated to cell motility. FIG. 8D: Cell motility (mean velocity of cell center of mass) declines 10 hours following drug addition.



FIGS. 9A-D are plots illustrating clusters of proteins from the same GO annotation with similar dynamics. Each plot represents a different cluster of proteins with the same GO annotation. Each line denotes the average fluorescence measured for at least 30 individual cells normalized between zero (blue) and one (red).



FIG. 10 is a graph illustrating rapid translocations in response to the drug CPT. Nucleolar levels of tagged TOP1 (the drug target) decreased in less than 2 minutes following CPT addition. Each line corresponds to a different cell.



FIGS. 11A-F are photographs and graphs illustrating TOP1 drug and dose dependency. FIG. 11AD illustrate that nuclear exit of tagged TOP1 does not occur with an equivalently lethal dose of etoposide, a topoisomerase-2 inhibitor drug. FIG. 11E is a graph illustrating that tagged TOP1 exits from the nucleus to the cytoplasm in a CPT dose dependent manner (full lines). A control nuclear protein expressed in the same cells (XRCC5-mCherry) does not exit the nucleus at all CPT doses (dashed lines). Each line is the mean of all cells at each time-point. FIG. 11F shows immunoblots with anti-TOP1 and anti-GFP showing that most TOP1 is degraded within 4 hours. In this degradation process fragments of TOP1 linked with YFP are created. These fragments are the source of fluorescence measured in the cytoplasm following CPT addition.



FIGS. 12A-B are graphs illustrating rapid translocation in response to the drug CPT. FIG. 12A illustrates tagged proteins that show a rapid decrease in nucleolar intensity and FIG. 12B illustrates tagged proteins that show a rapid increase in nucleolar/nucleoplasm ratio followed by a decrease back to basal levels.



FIGS. 13A-B are graphs illustrating localization changes in proteins in response to actinomycin-D. Localization changes of proteins in response to addition of 1 μg/ml of actinomycin-D (a transcription inhibitor). FIG. 13A: Tagged proteins that show a rapid increase in nucleolar/nucleoplasm ratio followed in some cases by a decrease back to basal levels. FIG. 13B: Tagged proteins that show a rapid decrease in nucleolar intensity.



FIGS. 14A-C are plots and graphs illustrating slower translocations in response to the drug CPT. Localization of fluorescence (nuclear intensity divided by total intensity) for all tagged proteins over time following drug addition is illustrated in FIG. 14A, and examples of two tagged proteins that show changes in nuclear (red line) and cytoplasmic (blue line) intensity (chaperon PFDN5 and thirodoxin reductase TXNRD1) are illustrated in FIGS. 14B and C respectively.



FIG. 15 is a graph illustrating that nuclear to cytoplamic ratio of TXNRD1 increases following CPT addition. Each line denotes the nuclear to cytoplamic ratio measured for an individual cell tracked over 50 hours. Bold green line denotes the average nuclear to cytoplasmic ratio.



FIG. 16 is a graph illustrating measurement of cell-cell viability over time. CV (Coefficient of variance=std/mean) of 400 proteins. In red all proteins that show CV of over 3 standard deviations from the average normalized CV of all proteins. Each line denotes CV of a different protein. Average CV of all 400 proteins is bold black and that of the 30 “bimodal” proteins is bold brown.



FIGS. 17A-F are graphs illustrating the proteins displaying bimodal response at the single cell level in response to CPT. FIGS. 17A-B are examples of proteins that show unimodal distributions, with similarly shaped profiles in each individual cell. All cells rise with time (red lines) or decrease with time (blue lines). The CV (std/mean of cell-cell distribution at each timepoint) increases slightly over time, and the distribution of slopes of fluorescence levels show a uniform behavior, all rising or all decreasing. FIGS. 17C-F are examples of proteins that show bimodal behavior. The dynamics after about 20 hours are different in different cells: some cells show increase in fluorescence levels (red) and other cells how a decrease (blue). This results in bi-modal distributions of fluorescent intensity slopes. Slopes are defined as median time derivative of the fluorescence levels, in the interval between 24 hours following drug addition to 48 hours (or time of cell death).



FIGS. 18A-B are graphs and plots illustrating that a tagged protein with a bimodal behavior correlates with the fate of individual cells. FIG. 18A: The RNA helicase DDX5 shows an increase in intensity in cells that survive the drug after 48 hours, and a decrease in cells that show the morphological changes associated with cell death. Heavy colored lines are cells that die, with darker colors corresponding to earlier cell death. Blue lines are cells that do not die during the movie. FIG. 18B: Cells that show the morphological correlates of cell death have significantly higher slopes of DDX5 fluorescence accumulation than cells that do not (T-test P<10̂-13). Slopes are defined as in FIGS. 17A-F.



FIGS. 19A-F are graphs illustrating that DDX5 shows different dynamics in response to other drugs. Response of DDX5 to Camptothecin 0.33 μM, Cis-platinum 40 μM and Etoposide 33.3 μM. Each line denotes total fluorescence measured for a single cell. Coefficient of variance (CV) is denoted for each measurement.



FIGS. 20A-B are plots illustrating that arbitrary fluorescence units can be converted to scalable units. FIG. 20A: Each dot is the measurement of the total fluorescent levels of a specific clone on two different dates. Each measurement is averaged over many cells at the time point before drug addition. Data is corrected for exposure time and lamp intensity (R=0.97). FIG. 20B: Each dot is the measurement of the total fluorescent levels of a specific protein using two different clones. Each measurement is averaged over many cells at time point before drug addition. Data is corrected for exposure time and lamp intensity (R=0.63).



FIGS. 21A-B are graphs and plots illustrating that a tagged protein with a bimodal behavior correlates with the fate of individual cells. FIG. 21A: Thioredoxin reductase 1 (TXNRD) shows an increase in intensity in cells that survive the drug after 48 hours, and a decrease in cells that show the morphological changes associated with cell death. Heavy colored lines are cells that die, with darker colors corresponding to earlier cell death. Blue lines are cells that do not die during the movie. FIG. 21B: Cells that show the morphological correlates of cell death have significantly higher slopes of TXNRD fluorescence accumulation than cells that do not (T-test P<10̂-13). Slopes are defined as in FIGS. 17A-F.



FIG. 22 is a graph illustrating that cell death dynamics in response to CPT+DDX5 siRNA increases in phase I compared to control but decreases in phase II.





DESCRIPTION OF SPECIFIC EMBODIMENTS OF THE INVENTION

The present invention, in some embodiments thereof, relates to cells comprising endogenous polypeptides attached to reporter polypeptides. The cells may be used to analyze endogenous polypeptide localization in the cell such as in diseased and non-diseased states. Amongst a myriad of other uses, such cells may be used to test the effects of agents of interest, identify therapeutic agents as well as to determine targets of therapeutic agents and markers for disease prognosis.


Before explaining at least one embodiment of the invention in detail, it is to be understood that the invention is not necessarily limited in its application to the details set forth in the following description or exemplified by the Examples. The invention is capable of other embodiments or of being practiced or carried out in various ways.


A quantitative understanding of human protein networks requires the measurement of endogenous protein dynamics in living cells.


The present inventors have devised a novel approach for visualizing polypeptides in live cells and therefore have made it possible to analyze localizations of polypeptides and quantities thereof during a particular cell state and/or following exposure to a therapeutic agent. Their approach comprises tagging at least two polypeptides in their native chromosomal locations, where the image analysis of one of the tagged polypeptides is aided by the other tagged polypeptide.


Whilst reducing the present invention to practice, the present inventors have generated a library of more than 1000 cell lines based on the same parental clonal cell (H1299 cancer cell line), each clone expressing two tagged proteins used for image analysis of the third tagged protein. The third tagged protein is different in each of the cell lines of the library. Each of the tagged proteins was labeled at its endogenous chromosomal location, each undergoing endogenous regulation. Generation of the library was effected by three sequential rounds of random endogenous gene tagging as detailed in Example 1 herein below.


The tagged polypeptides in the library of the present invention spanned a wide range of functional categories and localization patterns including membrane, nuclear, nucleolar, cytoskeleton, Golgi, ER and other localizations (SOM) (FIGS. 4A-C). In addition, all tagged polypeptides in the library had localization patterns similar to their counterpart polypeptides without the tag. 20% of the tagged polypeptides in the library of the present invention were novel (see Table 2 in the Examples section herein below and FIG. 8B).


Using an exemplary therapeutic agent, camptothecin (CPT), the present inventors further showed that the present library of cell lines may be used to identify a drug target (FIGS. 8B and 10) and aid in determining a drug mechanism of action (FIGS. 12A-B and 13A-B).


In addition, the present inventors showed that the present system allows monitoring of cell-cell variability of a particular polypeptide over time. The present inventors identified a group of polypeptides which diverged from standard cell-cell variability following treatment with CPT (FIGS. 16 and 17A-F). The present inventors further showed that the different behaviors of some of these proteins were linked to the fate of each cell (FIGS. 18A-B and 19A-F).


These proteins are indicative of potential drug targets, since down-regualtion of same would enhance the drug effect. As such the present system allows for identification of secondary targets (FIG. 22).


Thus, according to one aspect of the present invention there is provided a cell expressing at least two endogenous polypeptides, each covalently attached to a distinguishable reporter polypeptide.


The term “cell” as used herein, refers to a biological cell, e.g. eukaryotic, such as of mammalian origin (e.g. human). The cell may be diseased (e.g. cancerous) or healthy, taken directly from a living organism or part of a cell line, immortalized or non-immortalized.


According to one embodiment, the cell is viable.


As used herein, the phrase “endogenous polypeptide” refers to a polypeptide whose polynucleotide sequence encoding same is transcribed from its native chromosomal location in the cell.


According to one embodiment, the endogenous polypeptide is full-length.


According to another embodiment, the endogenous polypeptide is tagged internally (i.e. not on the N or C terminus) with the reporter polypeptide of the present invention.


According to yet another embodiment, the endogenous polypeptide maintains wild type functionality (i.e., of non-tagged protein) and further has a similar cellular localization pattern both prior to and following attachment of the reporter polypeptide.


Exemplary endogenous polypeptides include those listed in Table 3 of Example 2 herein below including those comprising a sequence as set forth in SEQ ID NOs: 1-164.


According to one embodiment of this aspect of the present invention, one of the endogenous polypeptides serves as an aid in the determination of the localization of the second endogenous polypeptide in the cell. Such a polypeptide is referred to herein as a “helper polypeptide”. Thus for example the “helper” polypeptide may be one that allows cell structures to be identified. For example the “helper” polypeptide may be one that localizes to the nucleus, such as XRCC5—Genbank Accession No. NP066964.1, such that the nucleus may be easily identified. Alternatively, the “helper” polypeptide may be one that localizes to the entire intracellular domain, such as DAP1—Genbank Accession No. NP004385.1, such that the entire cell may be identified. Typically, the “helper” polypeptide is constitutively expressed e.g. a house keeping polypeptide i.e. is not affected by a cell state such as a disease.


According to another embodiment of this aspect of the present invention, a combination of endogenous “helper” polypeptides aid in the detection of an additional polypeptide. The combination of “helper polypeptides” may each comprise an identical reporter polypeptide or alternatively reporter polypeptides that are distinguishable one from the other. The additionally polypeptide may serve to highlight a different area of the cell—for e.g. one of the helper polypeptides may be for identifying the cell nucleus and the other for identifying a second organelle or the cell cytoplasm as a whole.


The phrase “reporter polypeptide” as used herein, refers to a polypeptide which can be detected in a cell. Preferably, the reporter polypeptide of this aspect of the present invention can be directly detected in the cell (no need for a detectable moiety with an affinity to the reporter) by exerting a detectable signal which can be viewed in living cells (e.g., using a fluorescent microscope). Non-limiting examples of reporter polypeptides include fluorescent reporter polypeptides, (e.g. those comprising an autofluorescent activity), chemiluminescent reporter polypeptides and phosphorescent reporter polypeptides. Examples of fluorescent polypeptides include those belonging to the green fluorescent protein family, including but not limited to the green fluorescent protein, the yellow fluorescent protein, the cyan fluorescent protein and the red fluorescent protein as well as their enhanced derivatives.


As mentioned, the reporter polypeptides attached to at least two endogenous polypeptides of the present invention are distinguishable from each other. Thus, fluorescent reporter polypeptides for example may be selected such that each emits light of a distinguishable wavelength and therefore color when excited by light.


The reporter polypeptides are typically attached covalently to the endogenous polypeptides directly (i.e. via peptide bonds), although indirect attachment via linker peptides is also contemplated.


Since the polypeptides of the present invention are generated by transcription of genes present in their native chromosomal location in the cell, methods of generating cells expressing same typically entail changes to the native gene sequence of the cells.


Thus, cells of the present invention are typically generated by introduction of at least two nucleic acid constructs into the cell, both of which being capable of insertion into a genome of the cell.


The nucleic acid constructs of the present invention comprise a nucleic acid sequence encoding a reporter polypeptide linked to an additional nucleic acid sequence capable of inserting the nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to the reporter polypeptide is expressed in the cell.


It will be appreciated that the nucleic acid constructs of the present invention may be inserted into the genome of the host cell in a directed fashion (e.g. by homologous recombination or site-specific recombination) or a non-directed fashion i.e. non-homologous recombination.


The phrase “directed insertion” refers to the insertion of the construct at a predetermined sequence in the genome of the cell.


The phrase “non-directed insertion” refers to the insertion of the construct at a random sequence in the genome of the cell.


As used herein, the phrase “homologous recombination” refers to the process in which nucleic acid molecules with similar nucleotide sequences associate and exchange nucleotide strands. A nucleotide sequence of a first nucleic acid molecule that is effective for engaging in homologous recombination at a predefined position of a second nucleic acid molecule will therefore have a nucleotide sequence that facilitates the exchange of nucleotide strands between the first nucleic acid molecule and a defined position of the second nucleic acid molecule. Thus, the first nucleic acid will generally have a nucleotide sequence that is sufficiently complementary to a portion of the second nucleic


As used herein, the phrase “site-specific recombinase” refers to a type of recombinase that typically has at least the following four activities (or combinations thereof): (1) recognition of specific nucleic acid sequences; (2) cleavage of said sequence or sequences; (3) topoisomerase activity involved in strand exchange; and (4) ligase activity to reseal the cleaved strands of nucleic acid (see Sauer, B., Current Opinions in Biotechnology 5:521-527 (1994)). Conservative site-specific recombination is distinguished from homologous recombination and transposition by a high degree of sequence specificity for both partners. The strand exchange mechanism involves the cleavage and rejoining of specific nucleic acid sequences in the absence of DNA synthesis (Landy, A. (1989) Ann. Rev. Biochem. 58:913-949).


Nucleic acid constructs (also referred to herein as “expression vectors”) capable of insertion in a directed manner typically comprise one or more functionally compatible recognition site for a site-specific recombination enzyme.


As used herein, the phrase “functionally compatible recognition sites for a site-specific recombination enzyme” refers to specific nucleic acid sequences which are recognized by a site-specific recombination enzyme to allow site-specific DNA recombination (i.e., a crossover event between homologous sequences). An example of a site-specific recombination enzyme is the Cre recombinase (e.g., GenBank Accession No. YP006472), which is capable of performing DNA recombination between two loxP sites. Cre recombinase can be obtained from various suppliers such as the New England BioLabs, Inc, Beverly, Mass., or it can be expressed from a nucleic acid construct in which the Cre coding sequence is under the transcriptional control of an inducible promoter (e.g., the galactose-inducible promoter) as in plasmid pSH47.


Such “directed” nucleic acid constructs typically contain other specialized elements intended to increase the level of expression of cloned nucleic acids or to facilitate the identification of cells that carry the recombinant DNA. For example, a number of animal viruses contain DNA sequences that promote extra-chromosomal replication of the viral genome in permissive cell types. Plasmids bearing these viral replicons are replicated episomally as long as the appropriate factors are provided by genes either carried on the plasmid or with the genome of the host cell.


The “directed” nucleic acid constructs of the present invention may or may not include a eukaryotic replicon. If a eukaryotic replicon is present, the vector is capable of amplification in eukaryotic cells using the appropriate selectable marker. If the vector does not comprise a eukaryotic replicon, no episomal amplification is possible. Instead, the recombinant DNA integrates into the genome of the engineered cell, where the promoter directs expression of the desired nucleic acid.


Examples of mammalian nucleic acid constructs include, but are not limited to, pcDNA3, pcDNA3.1(+/−), pGL3, pZeoSV2(+/−), pSecTag2, pDisplay, pEF/myc/cyto, pCMV/myc/cyto, pCR3.1, pSinRep5, DH26S, DHBB, pNMT1, pNMT41, and pNMT81, which are available from Invitrogen, pCI which is available from Promega, pMbac, pPbac, pBK-RSV and pBK-CMV, which are available from Strategene, pTRES which is available from Clontech, and their derivatives.


Nucleic acid constructs containing regulatory elements from eukaryotic viruses such as retroviruses can be also used. SV40 vectors include pSVT7 and pMT2, for instance. Vectors derived from bovine papilloma virus include pBV-1MTHA, and vectors derived from Epstein-Barr virus include pHEBO and p2O5. Other exemplary vectors include pMSG, pAV009/A+, pMTO10/A+, pMAMneo-5, baculovirus pDSVE, and any other vector allowing expression of proteins under the direction of the SV40 early promoter, SV40 later promoter, metallothionein promoter, murine mammary tumor virus promoter, Rous sarcoma virus promoter, polyhedrin promoter, or other promoters shown effective for expression in eukaryotic cells.


As mentioned, the nucleic acid constructs of the present invention may also be inserted into the genome of the host cell in a non-directed fashion, i.e. non-homologous recombination.


The phrase, “non-homologous recombination” as used herein refers to the joining (exchange or redistribution) of genetic material through a mechanism that does not involve homologous recombination (e.g., recombination directed by sequence homology) and that does not involve site-specific recombination (e.g., recombination directed by site-specific recombination signals and a corresponding site-specific recombinase). Examples of non-homologous recombination include integration of exogenous DNA into chromosomes at non-homologous sites, chromosomal translocations and deletions, DNA end joining, double strand break repair, bridge-break-fusion, concatemerization of transfected polynucleotides, retroviral insertion, and transposition.


Retroviral vectors integrate into eukaryotic genomes by a distinct mechanism of non-homologous recombination that is catalyzed by the action of the virally encoded integrase enzyme, and the mechanism of viral integration, replication and infection has been well described [see for example Retroviruses. Coffin, J M.; Hughes, S H.; Varmus, H E. Plainview (NY): Cold Spring Harbor Laboratory Press; c1997; Use of wildtype retroviruses as mutagens]. The mutagenic ability of retroviruses and retroviral vectors and their ability to enable the rapid identification of mutated genes through the linkage of retroviral tag sequences within the transcripts of mutagenized genes are well known in the art (Friedrich G, Soriano P. Methods Enzymol. 1993; 225:681-701; 3: Gossler A, et al., Science. Apr. 28, 1989; 244(4903):463-5; Friedrich G, Soriano P. Genes Dev. September 1991; 5(9):1513-23; 5: von Melchner H, et al Genes Dev. June 1992; 6(6):919-27].


Retroviral constructs of the present invention may contain retroviral LTRs, packaging signals, and any other sequences that facilitate creation of infectious retroviral vectors. Retroviral LTRs and packaging signals allow the reporter polypeptides of the invention to be packaged into infectious particles and delivered to the cell by viral infection. Methods for making recombinant retroviral vectors are well known in the art (see for example, Brenner et al., PNAS 86:5517-5512 (1989); Xiong et al., Developmental Dynamics 212:181-197 (1998) and references therein; each incorporated herein by reference). In preferred embodiments, the retroviral vectors used in the invention comprise splice acceptor (SA) and splice donor (SD) sequences flanking the sequence encoding the reporter polypeptide. Typically, the constructs of the present invention do not comprise a promoter, a start codon or a polyA signal. In this way, if the virus inserts into an actively transcribed gene, the reporter sequence is retained as a new exon after splicing of the mRNA. Owing to the large size of the first intron and viral preference for integration sites near the start of genes, the first intron is the most common point of insertion. The tagged mRNA translates to an internally labeled protein, with the reporter polypeptide usually near the N terminus.


Retroviral LTRs and packaging signals can be selected according to the intended host cell to be infected. Examples of retroviral sequences useful in the present invention include those derived from Murine Moloney Leukemia Virus (MMLV), Avian Leukemia Virus (ALV), Avian Sarcoma Leukosis Virus (ASLV), Feline Leukemia Virus (FLV), and Human Immunodeficiency Virus (HIV). Other viruses known in the art are also useful in the present invention and therefore will be familiar to the ordinarily skilled artisan.


Like retroviruses, transposons and transposon vectors can also be used to integrate sequences in a non-directed fashion into the chromosome of the cell. Also like retroviruses, transposons integrate by enzymatically catalyzed non-homologous recombination in which transposase enzymes catalyze the genomic integration and transposition of transposon DNA.


Numerous transposons have been characterized that function in mammals. In particular, the TC1/mariner derivative transposon, Sleeping Beauty, has been demonstrated to integrate efficiently in mammals.


The constructs of the present invention can be introduced into a cell and integrated into DNA by any method known in the art. In one embodiment, they are introduced by transfection. Methods of transfection include, but are not limited to, electroporation, particle bombardment, calcium phosphate precipitation, lipid-mediated transfection (e.g., using cationic lipids), micro-injection, DEAE-mediated transfection, polybrene mediated transfection, naked DNA uptake, and receptor mediated endocytosis.


Typically the introduction of the constructs of the present invention is effected whilst the cells are being cultured in a medium which supports well-being and propagation. The medium is typically selected according to the cell being transfected/infected.


According to one embodiment, the constructs of the present invention are introduced into the cell by viral transduction or infection. Suitable viral vectors useful in the present invention include, but are not limited to, adeno-associated virus, adenovirus vectors, alpha-herpesvirus vectors, pseudorabies virus vectors, herpes simplex virus vectors and retroviral vectors (including lentiviral vectors).


As mentioned, at least two nucleic acid constructs are introduced into the cell to generate the cells of the present invention.


According to one embodiment, the nucleic acid constructs are introduced in a non-simultaneous (i.e. consecutive) fashion into the cell. This may be particularly relevant if the nucleic acid construct is inserted into the cell in a non-directed fashion, since consecutive introduction of the nucleic acid constructs allows for selection of a particular clone following introduction of the first construct, and prior to introduction of the second construct.


For example, the present invention contemplates introduction of the first nucleic acid construct into the cell in a non-directed fashion, selection of a cell in which a particular polypeptide is tagged, propagation of that cell and subsequent introduction of the second nucleic acid construct into the cell. If the second nucleic acid construct is introduced into the cell in a directed fashion, a cell population will be generated in which both endogenously tagged polypeptides will be identical in each cell of the cell population. Alternatively, if the second nucleic acid construct is introduced into the cell in a non-directed fashion, a cell population will be generated in which only one endogenously tagged polypeptide will be identical in each cell of the cell population, whereas the other endogenously tagged polypeptide will be particular to each cell.


Other combinations contemplated by the present invention include introduction of the first nucleic acid construct into the cell in a directed fashion and simultaneous introduction of the second nucleic acid construct into the cell in a directed fashion.


Another contemplated example includes introduction of the first nucleic acid construct into the cell in a directed fashion and subsequent introduction of the second nucleic acid construct into the cell in a non-directed manner.


Following introduction of the nucleic acid constructs of the present invention the tagged reporter polypeptides may be identified, such as by 3′RACE, using a nested PCR reaction that amplifies the section between the reporter polypeptide and the polyA tail of the mRNA of the host gene. The PCR product may be sequenced directly and aligned to the genome.


Exemplary oligonucleotide primers that may be used for 3′RACE and sequencing are listed in Table 1 herein below.













TABLE 1








Alignment in



Primer name
Use
Sequence
YFP or mCherry







AP first-strand
First-strand cDNA
GGCCACGCGTCGACTAGTAC(T)17





synthesis
(SEQ ID NO: 167)





AP 92
RACE first and
GGCCACGCGTCGACTAGTAC



nested reaction 3′
(SEQ ID NO: 168)



primer





YFP 90
RACE first
GCAGAAGAACGGCATCAAGG
Bases 471-490



reaction 5′ primer
(SEQ ID NO: 169)



for YFP-tagged



genes





YFP 85
RACE-nested
CGCGATCACATGGTCCTGCTG
Bases 646-666



reaction 5′ primer
(SEQ ID NO: 170)



for YFP-tagged



genes





Cherry 45
RACE first
GTGGTGACCGTGACCCAGGA
Bases 322-341



reaction 5′ primer
(SEQ ID NO: 171)



for mCherry-



tagged genes





Cherry 46
RACE-nested
GCGGATGTACCCCGAGGACG
Bases 456-475



reaction 5′ primer
(SEQ ID NO: 172)



for mCherry-



tagged genes





Cherry 56
Sequencing of
GACTACACCATCGTGGAACA
Bases 586-605



mCherry RACE
(SEQ ID NO: 173)



product





YFP 906
Sequencing of
GGATCACTCTCGGCATGGAC
Bases 686-705



YFP RACE
(SEQ ID NO: 174)



product









In this fashion, a library of cell clones may be generated, each expressing at least two identified tagged, full-length proteins, generated by transcription of genes situated in their endogenous chromosomal location. The library may comprise any number of cell clones, such as 10, 50, 100 250, 500, 1000, 2000 or more.


The present inventors using the methods described herein generated a library of cell clones comprising about 1200 different tagged proteins, of which 80% were characterized polypeptides and 20% were novel polypeptides (comprising amino acid sequences listed in SEQ ID NOs: 1-164).


It will be appreciated that libraries generated according to the method of the present invention may be used for isolating polypeptides. Cells expressing the required tagged endogenous polypeptide may be contacted with an antibody which binds specifically to the tag (i.e. reporter polypeptide). The polypeptide may then be isolated using known techniques such as immunoprecipitation and immunoaffinity columns.


As used herein, the term “isolating” refers to removing the polypeptide from its native environment i.e. cell. According to a preferred embodiment the polypeptide is also removed from other cellular components, such as other polypeptides in the cell.


Antibodies for reporter polypeptides are known in the art. For example antibodies that bind specifically to GFP are commercially available from Abcam (e.g. Catalogue numbers ab290 and ab1218) and Cell Signalling (Catalogue No. 2555).


Alternatively antibodies for reporter polypeptides may be synthesized.


Methods of producing polyclonal and monoclonal antibodies as well as fragments thereof are well known in the art (See for example, Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, New York, 1988, incorporated herein by reference).


Using an exemplary therapeutic agent, camptothecin (CPT), the present inventors showed that the cells of the present invention may be used to identify a drug target (FIGS. 8B and 10). The novel drug targets identified using the method of the present invention are further described herein below.


Thus, according to another aspect of the present invention, there is provided a method of identifying a target of an agent, the method comprising:


(a) contacting cells of the present invention with the agent;


(b) analyzing a localization or amount of at least one of the endogenous polypeptides, wherein a change in the amount or localization is indicative of a target of the agent.


As used herein, the term “contacting” refers to direct of indirect contacting under conditions (e.g. for an appropriate time and under an appropriate temperature) such that the agent is able to cause an alteration (e.g. an up-regulation, down-regulation or change in location) in the target.


According to this aspect of the present invention, the change in the amount is by at least 1.5 fold, and more preferably by at least 2 fold or more. A change in localization may comprise a localization to a different organelle, (e.g. from mitochondria to cytoplasm or from nucleus to cell membrane) or may comprise a change in organelle expression ratio.


As used herein, the term “localization” refers to either a localization with respect to a cell compartment (e.g. nucleus, cell membrane, mitochondria etc.) or with respect to another polypeptide.


Analysis of the localization or amount of the tagged endogenous polypeptide is typically affected according to the reporter polypeptide of the present invention.


Thus, for example if the reporter polypeptide is fluorescent, a fluorescent confocal microscope may be used to analyze the localization and/or expression of tagged endogenous polypeptide. Alternatively, the expression of a tagged endogenous polypeptide may be analyzed using flow cytometry.


Preferably, the analysis does not affect the viability or function of the cell. For example the cells of the present invention may be used to monitor a change in amount or localization of endogenous polypeptide over real-time using long period time-lapse microscopy. Time-lapse movies may be obtained as described by Sigal et al. (Sigal, Milo et al. 2006, supra) with for example an automated, incubated (including humidity and CO2 control) inverted fluorescence microscope (e.g. Leica DMIRE2) and a CCD camera (e.g. ORCA ER—Hamamatsu Photonics).


It will be appreciated that if the analysis is effected in real-time, a sequence of events following a particular treatment can also be monitored. Thus for example, the camera or cameras may be capable of recording a number of cell populations at one time, each cell population comprising a different tagged endogenous polypeptide over a period of time (e.g. 24 hours). Analysis of the movies obtained following monitoring allows reconstruction of the sequence of events that occur after contact with the agent. The present inventors have shown, using the agent Camptothecin (CPT) by way of example, that typically the first polypeptide to respond is the direct target of the agent.


Agents whose targets are being determined, include therapeutic agents (such as polynucleotides, polypeptides, small molecule chemicals, carbohydrates, lipids etc.). It will be appreciated that the agent may also be a condition such as radiation. Further, the targets whose agents are being determined may be carcinogens or pollutants.


If the tagged endogenous polypeptide is a marker for a cell state, the cells of the present invention may be used to identify an agent capable of affecting that cell state.


Exemplary cell states include, but are not limited to a disease state such as cancer, an oxidative state and a hyperglycemic or hypoglycemic state etc.


According to this aspect of the present invention the cells of the present invention are contacted with a test agent and a localization or amount of the marker of the cell state is analyzed, wherein a change in the amount or localization of the marker is indicative of that the test agent is capable of affecting the cell state.


It will be appreciated that the cells of the present invention may be used to identify markers for disease prognosis. According to this aspect, diseased cells of the present invention are contacted with a therapeutic agent and the localization or amount of the tagged endogenous polypeptide in responsive cells is compared with the localization or amount of tagged endogenous polypeptide in non-responsive cells. A difference in expression or localization of the tagged endogenous polypeptide in responsive and non-responsive cells indicates that the tagged endogenous polypeptide is a marker for disease prognosis.


As used herein, the phrase “marker for disease prognosis” refers to a polypeptide whose expression or localization correlates with the severity of a disease. It will be appreciated that this method may also be used to select potential drug targets for enhancing an effect of a drug.


Detection of responsive and non-responsive cells is effected according to the cell type and the therapeutic agent. Thus, for example if the cells are cancer cells and the therapeutic agent causes a decrease in a particular marker e.g. a matrix metalloproteinase, cells may be generated that express a tagged matrix metalloproteinase, a tagged protein (or proteins) that aid in image analysis and a third tagged protein that is being analyzed. Such cells may be analyzed for other markers whose expression (or localization) correspond with the known marker of the disease.


According to another example, the cells are cancer cells and the therapeutic agent causes cell death. Individual cells may be analyzed using a microscope to see whether they show signs of cell death (e.g. cell shrinkage, nuclear fragmentation, blebbing etc.) in order to analyze if they are drug responsive or not. Comparison of the polypeptides in the responsive cell group with polypeptides in the non-responsive cell group, allows identification of potential drug targets for enhancing the effect of a drug. For example, the present inventors showed that three polypeptides were differentially up and down regulated in cells that survive the drug CPT, as opposed to cells that die. The three polypeptides were the helicase DDX5, the transport protein VPS26a and the appoptosis protein PEPP2. By targeting these proteins, together with CPT, one may be able to increase the efficacy of the drug by targeting cancer cells that would otherwise not be killed.


Since the cells of the present invention express at least two tagged endogenous polypeptides, the cells may be used to analyze localization of same.


Thus, according to yet another aspect of the present invention there is provided a method of analyzing a localization of a first and second endogenous polypeptide in a cell, the method comprising detecting a localization of the first and second endogenous polypeptide in the cell, wherein the first and second polypeptide are each covalently attached to a distinguishable reporter polypeptide, thereby analyzing localization of a first and second polypeptide.


It will be appreciated that the method of this aspect of the present invention may be used to analyze localization the two endogenous polypeptides to a particular cell compartment, or alternatively to analyze their localization with respect to one another. Accordingly, the method of this aspect of the present invention may also be used to detect a binding or interaction between the first and second endogenous polypeptide.


Accordingly, the present invention may be used as a FRET system for analyzing the interaction between two endogenous polypeptides.


As used herein, the term “FRET” refers to the process in which an excited donor fluorophore transfers energy to a lower-energy acceptor fluorophore via a short-range (e.g., less than or equal to 10 nm) dipole-dipole interaction.


As mentioned, the present invention identified novel targets for Camptothecin using the cell populations of the present invention.


As described in Example 3 herein below, the present inventors have shown that DNA helicase DDX5 and Replication factor C activator 1 (RFC1) both decrease in cells that respond to CPT treatment indicating that these proteins promote cell survival under this drug. Accordingly, inhibition of these polypeptides may increase the efficacy of CPT (FIG. 22). In addition, the present inventors have shown that inhibitors of thioredoxin and thioredoxin reductase 1 (TXNRD1) may also be used to enhance the effect of CPT.


Thus, according to another aspect of the present invention, there is provided a method of treating a cancer comprising co-administering to a subject in need thereof a therapeutically effective amount of Camptothecin and an agent capable of downregulating DNA helicase DDX5 or replication factor C activator 1 (RFC1), thereby treating the cancer.


As used herein, the term “treating” includes abrogating, substantially inhibiting, slowing or reversing the progression of a condition, substantially ameliorating clinical or aesthetical symptoms of a condition or substantially preventing the appearance of clinical or aesthetical symptoms of a condition.


As used herein the term “subject” refers to any (e.g., mammalian) subject, preferably a human subject.


As used herein, the term “camptothecin” refers to a cytotoxic quinoline alkaloid capable of inhibiting the DNA enzyme topoisomerase I. Camptothecin is widely commercially available (e.g. Sigma CPT; C9911). The camptothecin may be an analogue or a derivate of available camptothecins.


The term “DNA helicase DDX5” refers to the polypeptide whose sequence is as set forth in Genbank as NP004387.1, Swiss Prot. number P17844 and homologues and variants thereof.


The term “Replication factor C activator 1 (RFC1)” refers to the polypeptide whose sequence is as set forth in Genbank as NP002904.3, Swiss Prot. number P35251 and homologues and variants thereof.


The term “thioredoxin reductase 1 (TXNRD1)” refers to the polypeptide whose sequence is as set forth in Genbank as NP001087240.1, NP003321.3, NP877393.1, NP877419.1 or NP877420.1, Swiss Prot. number Q16881 and homologues and variants thereof.


As used herein the term “cancer” refers to the presence of cells possessing characteristics typical of cancer-causing cells, for example, uncontrolled proliferation, loss of specialized functions, immortality, significant metastatic potential, significant increase in anti-apoptotic activity, rapid growth and proliferation rate, and certain characteristic morphology and cellular markers. In some circumstances, cancer cells will be in the form of a tumor; such cells may exist locally within an animal, or circulate in the blood stream as independent cells, for example, leukemic cells.


Specific examples of cancer which can be treated using the combination of the present invention include, but are not limited to, adrenocortical carcinoma, hereditary; bladder cancer; breast cancer; breast cancer, ductal; breast cancer, invasive intraductal; breast cancer, sporadic; breast cancer, susceptibility to; breast cancer, type 4; breast cancer, type 4; breast cancer-1; breast cancer-3; breast-ovarian cancer; Burkitt's lymphoma; cervical carcinoma; colorectal adenoma; colorectal cancer; colorectal cancer, hereditary nonpolyposis, type 1; colorectal cancer, hereditary nonpolyposis, type 2; colorectal cancer, hereditary nonpolyposis, type 3; colorectal cancer, hereditary nonpolyposis, type 6; colorectal cancer, hereditary nonpolyposis, type 7; dermatofibrosarcoma protuberans; endometrial carcinoma; esophageal cancer; gastric cancer, fibrosarcoma, glioblastoma multiforme; glomus tumors, multiple; hepatoblastoma; hepatocellular cancer; hepatocellular carcinoma; leukemia, acute lymphoblastic; leukemia, acute myeloid; leukemia, acute myeloid, with eosinophilia; leukemia, acute nonlymphocytic; leukemia, chronic myeloid; Li-Fraumeni syndrome; liposarcoma, lung cancer; lung cancer, small cell; lymphoma, non-Hodgkin's; lynch cancer family syndrome II; male germ cell tumor; mast cell leukemia; medullary thyroid; medulloblastoma; melanoma, meningioma; multiple endocrine neoplasia; myeloid malignancy, predisposition to; myxosarcoma, neuroblastoma; osteosarcoma; ovarian cancer; ovarian cancer, serous; ovarian carcinoma; ovarian sex cord tumors; pancreatic cancer; pancreatic endocrine tumors; paraganglioma, familial nonchromaffin; pilomatricoma; pituitary tumor, invasive; prostate adenocarcinoma; prostate cancer; renal cell carcinoma, papillary, familial and sporadic; retinoblastoma; rhabdoid predisposition syndrome, familial; rhabdoid tumors; rhabdomyosarcoma; small-cell cancer of lung; soft tissue sarcoma, squamous cell carcinoma, head and neck; T-cell acute lymphoblastic leukemia; Turcot syndrome with glioblastoma; tylosis with esophageal cancer; uterine cervix carcinoma, Wilms' tumor, type 2; and Wilms' tumor, type 1, and the like.


According to one embodiment of this aspect of the present invention, the cancer is ovarian or colon cancer.


Down-regulating the function or expression of DNA helicase DDX5, replication factor C activator 1 (RFC1), thioredoxin or thioredoxin redutase can be effected at the RNA level or at the protein level. According to one embodiment of this aspect of the present invention the agent is an oligonucleotide capable of specifically hybridizing (e.g., in cells under physiological conditions) to a polynucleotide encoding these polypeptide. Exemplary siRNAs capable of down-regulating DDX5 are set forth in SEQ ID NO:175-178.


The prior art teaches of a number of delivery strategies which can be used to efficiently deliver oligonucleotides into a wide variety of cell types [see, for example, Luft J Mol Med 76: 75-6 (1998); Kronenwett et al., Blood 91: 852-62 (1998); Rajur et al., Bioconjug Chem 8: 935-40 (1997); Lavigne et al., Biochem Biophys Res Commun 237: 566-71 (1997) and Aoki et al., (1997) Biochem Biophys Res Commun 231: 540-5 (1997)].


According to another embodiment of this aspect of the present invention, the agent is a RNA silencing agent.


As used herein, the phrase “RNA silencing” refers to a group of regulatory mechanisms [e.g. RNA interference (RNAi), transcriptional gene silencing (TGS), post-transcriptional gene silencing (PTGS), quelling, co-suppression, and translational repression] mediated by RNA molecules which result in the inhibition or “silencing” of the expression of a corresponding protein-coding gene. RNA silencing has been observed in many types of organisms, including plants, animals, and fungi.


As used herein, the term “RNA silencing agent” refers to an RNA which is capable of inhibiting or “silencing” the expression of a target gene. In certain embodiments, the RNA silencing agent is capable of preventing complete processing (e.g, the full translation and/or expression) of an mRNA molecule through a post-transcriptional silencing mechanism. RNA silencing agents include noncoding RNA molecules, for example RNA duplexes comprising paired strands, as well as precursor RNAs from which such small non-coding RNAs can be generated. Exemplary RNA silencing agents include dsRNAs such as siRNAs, miRNAs and shRNAs. In one embodiment, the RNA silencing agent is capable of inducing RNA interference. In another embodiment, the RNA silencing agent is capable of mediating translational repression.


RNA interference refers to the process of sequence-specific post-transcriptional gene silencing in animals mediated by short interfering RNAs (siRNAs). The corresponding process in plants is commonly referred to as post-transcriptional gene silencing or RNA silencing and is also referred to as quelling in fungi. The process of post-transcriptional gene silencing is thought to be an evolutionarily-conserved cellular defense mechanism used to prevent the expression of foreign genes and is commonly shared by diverse flora and phyla. Such protection from foreign gene expression may have evolved in response to the production of double-stranded RNAs (dsRNAs) derived from viral infection or from the random integration of transposon elements into a host genome via a cellular response that specifically destroys homologous single-stranded RNA or viral genomic RNA.


The presence of long dsRNAs in cells stimulates the activity of a ribonuclease III enzyme referred to as dicer. Dicer is involved in the processing of the dsRNA into short pieces of dsRNA known as short interfering RNAs (siRNAs). Short interfering RNAs derived from dicer activity are typically about 21 to about 23 nucleotides in length and comprise about 19 base pair duplexes. The RNAi response also features an endonuclease complex, commonly referred to as an RNA-induced silencing complex (RISC), which mediates cleavage of single-stranded RNA having sequence complementary to the antisense strand of the siRNA duplex. Cleavage of the target RNA takes place in the middle of the region complementary to the antisense strand of the siRNA duplex.


Accordingly, the present invention contemplates use of dsRNA to downregulate protein expression from mRNA.


According to one embodiment, the dsRNA is greater than 30 bp. The use of long dsRNAs (i.e. dsRNA greater than 30 bp) has been very limited owing to the belief that these longer regions of double stranded RNA will result in the induction of the interferon and PKR response. However, the use of long dsRNAs can provide numerous advantages in that the cell can select the optimal silencing sequence alleviating the need to test numerous siRNAs; long dsRNAs will allow for silencing libraries to have less complexity than would be necessary for siRNAs; and, perhaps most importantly, long dsRNA could prevent viral escape mutations when used as therapeutics.


Various studies demonstrate that long dsRNAs can be used to silence gene expression without inducing the stress response or causing significant off-target effects—see for example [Strat et al., Nucleic Acids Research, 2006, Vol. 34, No. 13 3803-3810; Bhargava A et al. Brain Res. Protoc. 2004; 13:115-125; Diallo M., et al., Oligonucleotides. 2003; 13:381-392; Paddison P. J., et al., Proc. Natl. Acad. Sci. USA. 2002; 99:1443-1448; Tran N., et al., FEBS Lett. 2004; 573:127-134].


In particular, the present invention also contemplates introduction of long dsRNA (over 30 base transcripts) for gene silencing in cells where the interferon pathway is not activated (e.g. embryonic cells and oocytes) see for example Billy et al., PNAS 2001, Vol 98, pages 14428-14433 and Diallo et al, Oligonucleotides, Oct. 1, 2003, 13(5): 381-392, doi:10.1089/154545703322617069.


The present invention also contemplates introduction of long dsRNA specifically designed not to induce the interferon and PKR pathways for down-regulating gene expression. For example, Shinagwa and Ishii [Genes & Dev. 17 (11): 1340-1345, 2003] have developed a vector, named pDECAP, to express long double-strand RNA from an RNA polymerase II (Pol II) promoter. Because the transcripts from pDECAP lack both the 5′-cap structure and the 3′-poly(A) tail that facilitate ds-RNA export to the cytoplasm, long ds-RNA from pDECAP does not induce the interferon response.


Another method of evading the interferon and PKR pathways in mammalian systems is by introduction of small inhibitory RNAs (siRNAs) either via transfection or endogenous expression.


The term “siRNA” refers to small inhibitory RNA duplexes (generally between 18-30 basepairs) that induce the RNA interference (RNAi) pathway. Typically, siRNAs are chemically synthesized as 21mers with a central 19 by duplex region and symmetric 2-base 3′-overhangs on the termini, although it has been recently described that chemically synthesized RNA duplexes of 25-30 base length can have as much as a 100-fold increase in potency compared with 21mers at the same location. The observed increased potency obtained using longer RNAs in triggering RNAi is theorized to result from providing Dicer with a substrate (27mer) instead of a product (21mer) and that this improves the rate or efficiency of entry of the siRNA duplex into RISC.


It has been found that position of the 3′-overhang influences potency of an siRNA and asymmetric duplexes having a 3′-overhang on the antisense strand are generally more potent than those with the 3′-overhang on the sense strand (Rose et al., 2005). This can be attributed to asymmetrical strand loading into RISC, as the opposite efficacy patterns are observed when targeting the antisense transcript.


The strands of a double-stranded interfering RNA (e.g., an siRNA) may be connected to form a hairpin or stem-loop structure (e.g., an shRNA). Thus, as mentioned the RNA silencing agent of the present invention may also be a short hairpin RNA (shRNA).


The term “shRNA”, as used herein, refers to an RNA agent having a stem-loop structure, comprising a first and second region of complementary sequence, the degree of complementarity and orientation of the regions being sufficient such that base pairing occurs between the regions, the first and second regions being joined by a loop region, the loop resulting from a lack of base pairing between nucleotides (or nucleotide analogs) within the loop region. The number of nucleotides in the loop is a number between and including 3 to 23, or 5 to 15, or 7 to 13, or 4 to 9, or 9 to 11. Some of the nucleotides in the loop can be involved in base-pair interactions with other nucleotides in the loop. Examples of oligonucleotide sequences that can be used to form the loop include 5′-UUCAAGAGA-3′ (Brummelkamp, T. R. et al. (2002) Science 296: 550) and 5′-UUUGUGUAG-3′ (Castanotto, D. et al. (2002) RNA 8:1454). It will be recognized by one of skill in the art that the resulting single chain oligonucleotide forms a stem-loop or hairpin structure comprising a double-stranded region capable of interacting with the RNAi machinery.


According to another embodiment the RNA silencing agent may be a miRNA. miRNAs are small RNAs made from genes encoding primary transcripts of various sizes. They have been identified in both animals and plants. The primary transcript (termed the “pri-miRNA”) is processed through various nucleolytic steps to a shorter precursor miRNA, or “pre-miRNA.” The pre-miRNA is present in a folded form so that the final (mature) miRNA is present in a duplex, the two strands being referred to as the miRNA (the strand that will eventually basepair with the target) The pre-miRNA is a substrate for a form of dicer that removes the miRNA duplex from the precursor, after which, similarly to siRNAs, the duplex can be taken into the RISC complex. It has been demonstrated that miRNAs can be transgenically expressed and be effective through expression of a precursor form, rather than the entire primary form (Parizotto et al. (2004) Genes & Development 18:2237-2242 and Guo et al. (2005) Plant Cell 17:1376-1386).


Unlike, siRNAs, miRNAs bind to transcript sequences with only partial complementarity (Zeng et al., 2002, Molec. Cell 9:1327-1333) and repress translation without affecting steady-state RNA levels (Lee et al., 1993, Cell 75:843-854; Wightman et al., 1993, Cell 75:855-862). Both miRNAs and siRNAs are processed by Dicer and associate with components of the RNA-induced silencing complex (Hutvagner et al., 2001, Science 293:834-838; Grishok et al., 2001, Cell 106: 23-34; Ketting et al., 2001, Genes Dev. 15:2654-2659; Williams et al., 2002, Proc. Natl. Acad. Sci. USA 99:6889-6894; Hammond et al., 2001, Science 293:1146-1150; Mourlatos et al., 2002, Genes Dev. 16:720-728). A recent report (Hutvagner et al., 2002, Sciencexpress 297:2056-2060) hypothesizes that gene regulation through the miRNA pathway versus the sRNA pathway is determined solely by the degree of complementarity to the target transcript. It is speculated that siRNAs with only partial identity to the mRNA target will function in translational repression, similar to an miRNA, rather than triggering RNA degradation.


Synthesis of RNA silencing agents suitable for use with the present invention can be effected as follows. First, the polypeptide mRNA sequence is scanned downstream of the AUG start codon for AA dinucleotide sequences. Occurrence of each AA and the 3′ adjacent 19 nucleotides is recorded as potential sRNA target sites. Preferably, sRNA target sites are selected from the open reading frame, as untranslated regions (UTRs) are richer in regulatory protein binding sites. UTR-binding proteins and/or translation initiation complexes may interfere with binding of the sRNA endonuclease complex [Tuschl ChemBiochem. 2:239-245]. It will be appreciated though, that siRNAs directed at untranslated regions may also be effective, as demonstrated for GAPDH wherein sRNA directed at the 5′ UTR mediated about 90% decrease in cellular GAPDH mRNA and completely abolished protein level (wwwdotambiondotcom/techlib/tn/91/912dothtml).


Second, potential target sites are compared to an appropriate genomic database (e.g., human, mouse, rat etc.) using any sequence alignment software, such as the BLAST software available from the NCBI server (wwwdotncbidotnlmdotnihdotgov/BLAST/). Putative target sites which exhibit significant homology to other coding sequences are filtered out.


Qualifying target sequences are selected as template for sRNA synthesis. Preferred sequences are those including low G/C content as these have proven to be more effective in mediating gene silencing as compared to those with G/C content higher than 55%. Several target sites are preferably selected along the length of the target gene for evaluation. For better evaluation of the selected siRNAs, a negative control is preferably used in conjunction. Negative control siRNA preferably include the same nucleotide composition as the siRNAs but lack significant homology to the genome. Thus, a scrambled nucleotide sequence of the siRNA is preferably used, provided it does not display any significant homology to any other gene.


It will be appreciated that the RNA silencing agent of the present invention need not be limited to those molecules containing only RNA, but further encompasses chemically-modified nucleotides and non-nucleotides.


In some embodiments, the RNA silencing agent provided herein can be functionally associated with a cell-penetrating peptide.” As used herein, a “cell-penetrating peptide” is a peptide that comprises a short (about 12-30 residues) amino acid sequence or functional motif that confers the energy-independent (i.e., non-endocytotic) translocation properties associated with transport of the membrane-permeable complex across the plasma and/or nuclear membranes of a cell. The cell-penetrating peptide used in the membrane-permeable complex of the present invention preferably comprises at least one non-functional cysteine residue, which is either free or derivatized to form a disulfide link with a double-stranded ribonucleic acid that has been modified for such linkage. Representative amino acid motifs conferring such properties are listed in U.S. Pat. No. 6,348,185, the contents of which are expressly incorporated herein by reference. The cell-penetrating peptides of the present invention preferably include, but are not limited to, penetratin, transportan, plsl, TAT(48-60), pVEC, MTS, and MAP.


Another agent capable of downregulating the expression of the CPT modulating polypeptides of the present invention is a DNAzyme molecule capable of specifically cleaving its encoding polynucleotide. DNAzymes are single-stranded polynucleotides which are capable of cleaving both single and double stranded target sequences (Breaker, R. R. and Joyce, G. Chemistry and Biology 1995; 2:655; Santoro, S. W. & Joyce, G. F. Proc. Natl, Acad. Sci. USA 1997; 94:4262). A general model (the “10-23” model) for the DNAzyme has been proposed. “10-23” DNAzymes have a catalytic domain of 15 deoxyribonucleotides, flanked by two substrate-recognition domains of seven to nine deoxyribonucleotides each. This type of DNAzyme can effectively cleave its substrate RNA at purine:pyrimidine junctions (Santoro, S. W. & Joyce, G. F. Proc. Natl, Acad. Sci. USA 199; for rev of DNAzymes see Khachigian, L M [Curr Opin Mol Ther 4:119-21 (2002)].


Examples of construction and amplification of synthetic, engineered DNAzymes recognizing single and double-stranded target cleavage sites have been disclosed in U.S. Pat. No. 6,326,174 to Joyce et al. DNAzymes of similar design directed against the human Urokinase receptor were recently observed to inhibit Urokinase receptor expression, and successfully inhibit colon cancer cell metastasis in vivo (Itoh et al., 20002, Abstract 409, Ann Meeting Am Soc Gen Ther wwwdotasgtdotorg). In another application, DNAzymes complementary to bcr-ab1 oncogenes were successful in inhibiting the oncogenes expression in leukemia cells, and lessening relapse rates in autologous bone marrow transplant in cases of Chronic Myelogenous Leukemia (CML) and Acute Lymphocytic Leukemia (ALL).


Another agent capable of downregulating the expression of the CPT modulating polypeptides of the present invention is a ribozyme molecule capable of specifically cleaving its encoding polynucleotide. Ribozymes are being increasingly used for the sequence-specific inhibition of gene expression by the cleavage of mRNAs encoding proteins of interest [Welch et al., Curr Opin Biotechnol. 9:486-96 (1998)]. The possibility of designing ribozymes to cleave any specific target RNA has rendered them valuable tools in both basic research and therapeutic applications.


An additional method of downregulating the function of a CPT modulating polypeptide of the present invention is via triplex forming oligonucleotides (TFOs). In the last decade, studies have shown that TFOs can be designed which can recognize and bind to polypurine/polypirimidine regions in double-stranded helical DNA in a sequence-specific manner. Thus the DNA sequence encoding the polypeptide of the present invention can be targeted thereby down-regulating the polypeptide.


The recognition rules governing TFOs are outlined by Maher III, L. J., et al., Science (1989) 245:725-730; Moser, H. E., et al., Science (1987)238:645-630; Beal, P. A., et al., Science (1991) 251:1360-1363; Cooney, M., et al., Science (1988)241:456-459; and Hogan, M. E., et al., EP Publication 375408. Modification of the oligonucleotides, such as the introduction of intercalators and backbone substitutions, and optimization of binding conditions (pH and cation concentration) have aided in overcoming inherent obstacles to TFO activity such as charge repulsion and instability, and it was recently shown that synthetic oligonucleotides can be targeted to specific sequences (for a recent review see Seidman and Glazer (2003) J Clin Invest; 112:487-94).


In general, the triplex-forming oligonucleotide has the sequence correspondence:















oligo
3′--A
G
G
T







duplex
5′--A
G
C
T







duplex
3′--T
C
G
A







However, it has been shown that the A-AT and G-GC triplets have the greatest triple helical stability (Reither and Jeltsch (2002), BMC Biochem, September 12, Epub). The same authors have demonstrated that TFOs designed according to the A-AT and G-GC rule do not form non-specific triplexes, indicating that the triplex formation is indeed sequence specific.


Thus for any given sequence in the regulatory region a triplex forming sequence may be devised. Triplex-forming oligonucleotides preferably are at least 15, more preferably 25, still more preferably 30 or more nucleotides in length, up to 50 or 100 bp.


Transfection of cells (for example, via cationic liposomes) with TFOs, and subsequent formation of the triple helical structure with the target DNA, induces steric and functional changes, blocking transcription initiation and elongation, allowing the introduction of desired sequence changes in the endogenous DNA and results in the specific downregulation of gene expression. Examples of such suppression of gene expression in cells treated with TFOs include knockout of episomal supFG1 and endogenous HPRT genes in mammalian cells (Vasquez et al., Nucl Acids Res. (1999) 27:1176-81, and Puri, et al., J Biol Chem, (2001) 276:28991-98), and the sequence- and target-specific downregulation of expression of the Ets2 transcription factor, important in prostate cancer etiology (Carbone, et al., Nucl Acid Res. (2003) 31:833-43), and the pro-inflammatory ICAM-1 gene (Besch et al., J Biol Chem, (2002) 277:32473-79). In addition, Vuyisich and Beal have recently shown that sequence specific TFOs can bind to dsRNA, inhibiting activity of dsRNA-dependent enzymes such as RNA-dependent kinases (Vuyisich and Beal, Nuc. Acids Res (2000); 28:2369-74).


Additionally, TFOs designed according to the abovementioned principles can induce directed mutagenesis capable of effecting DNA repair, thus providing both downregulation and upregulation of expression of endogenous genes [Seidman and Glazer, J Clin Invest (2003) 112:487-94]. Detailed description of the design, synthesis and administration of effective TFOs can be found in U.S. Patent Application Nos. 2003 017068 and 2003 0096980 to Froehler et al., and 2002 0128218 and 2002 0123476 to Emanuele et al., and U.S. Pat. No. 5,721,138 to Lawn.


As mentioned hereinabove, down regulating the function of a CPT modulating polypeptide of the present invention can also be affected at the protein level.


Thus, another example of an agent capable of downregulating a CPT modulating polypeptide of the present invention is an antibody or antibody fragment capable of specifically binding to it, preferably to its active site, thereby preventing its function.


As used herein, the term “antibody” refers to a substantially intact antibody molecule.


As used herein, the phrase “antibody fragment” refers to a functional fragment of an antibody that is capable of binding to an antigen.


Suitable antibody fragments for practicing the present invention include, inter alia, a complementarity-determining region (CDR) of an immunoglobulin light chain (referred to herein as “light chain”), a CDR of an immunoglobulin heavy chain (referred to herein as “heavy chain”), a variable region of a light chain, a variable region of a heavy chain, a light chain, a heavy chain, an Fd fragment, and antibody fragments comprising essentially whole variable regions of both light and heavy chains such as an Fv, a single-chain Fv, an Fab, an Fab′, and an F(ab′)2.


Functional antibody fragments comprising whole or essentially whole variable regions of both light and heavy chains are defined as follows:


(i) Fv, defined as a genetically engineered fragment consisting of the variable region of the light chain and the variable region of the heavy chain expressed as two chains;


(ii) single-chain Fv (“scFv”), a genetically engineered single-chain molecule including the variable region of the light chain and the variable region of the heavy chain, linked by a suitable polypeptide linker.


(iii) Fab, a fragment of an antibody molecule containing a monovalent antigen-binding portion of an antibody molecule, obtained by treating whole antibody with the enzyme papain to yield the intact light chain and the Fd fragment of the heavy chain, which consists of the variable and CH1 domains thereof;


(iv) Fab′, a fragment of an antibody molecule containing a monovalent antigen-binding portion of an antibody molecule, obtained by treating whole antibody with the enzyme pepsin, followed by reduction (two Fab′ fragments are obtained per antibody molecule); and


(v) F(ab′)2, a fragment of an antibody molecule containing a monovalent antigen-binding portion of an antibody molecule, obtained by treating whole antibody with the enzyme pepsin (i.e., a dimer of Fab′ fragments held together by two disulfide bonds).


Methods of generating monoclonal and polyclonal antibodies are well known in the art. Antibodies may be generated via any one of several known methods, which may employ induction of in vivo production of antibody molecules, screening of immunoglobulin libraries (Orlandi, R. et al. (1989). Cloning immunoglobulin variable domains for expression by the polymerase chain reaction. Proc Natl Acad Sci USA 86, 3833-3837; and Winter, G. and Milstein, C. (1991). Man-made antibodies. Nature 349, 293-299), or generation of monoclonal antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the Epstein-Barr virus (EBV)-hybridoma technique (Kohler, G. and Milstein, C. (1975). Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 256, 495-497; Kozbor, D. et al. (1985). Specific immunoglobulin production and enhanced tumorigenicity following ascites growth of human hybridomas. J Immunol Methods 81, 31-42; Cote R J. et al. (1983). Generation of human monoclonal antibodies reactive with cellular antigens. Proc Natl Acad Sci USA 80, 2026-2030; and Cole, S. P. et al. (1984). Human monoclonal antibodies. Mol Cell Biol 62, 109-120).


It will be appreciated that for human therapy or diagnostics, humanized antibodies are preferably used. Humanized forms of non-human (e.g., murine) antibodies are genetically engineered chimeric antibodies or antibody fragments having (preferably minimal) portions derived from non-human antibodies. Humanized antibodies include antibodies in which the CDRs of a human antibody (recipient antibody) are replaced by residues from a CDR of a non-human species (donor antibody), such as mouse, rat, or rabbit, having the desired functionality. In some instances, the Fv framework residues of the human antibody are replaced by corresponding non-human residues. Humanized antibodies may also comprise residues found neither in the recipient antibody nor in the imported CDR or framework sequences. In general, the humanized antibody will comprise substantially all of at least one, and typically two, variable domains, in which all or substantially all of the CDRs correspond to those of a non-human antibody and all or substantially all of the framework regions correspond to those of a relevant human consensus sequence. Humanized antibodies optimally also include at least a portion of an antibody constant region, such as an Fc region, typically derived from a human antibody (see, for example: Jones, P. T. et al. (1986). Replacing the complementarity-determining regions in a human antibody with those from a mouse. Nature 321, 522-525; Riechmann, L. et al. (1988). Reshaping human antibodies for therapy. Nature 332, 323-327; Presta, L. G. (1992b). Curr Opin Struct Biol 2, 593-596; and Presta, L. G. (1992a). Antibody engineering. Curr Opin Biotechnol 3(4), 394-398).


Methods for humanizing non-human antibodies are well known in the art. Generally, a humanized antibody has one or more amino acid residues introduced into it from a source which is non-human. These non-human amino acid residues are often referred to as imported residues, which are typically taken from an imported variable domain. Humanization can be performed essentially as described (see, for example: Jones et al. (1986); Riechmann et al. (1988); Verhoeyen, M. et al. (1988). Reshaping human antibodies: grafting an antilysozyme activity. Science 239, 1534-1536; and U.S. Pat. No. 4,816,567), by substituting human CDRs with corresponding rodent CDRs. Accordingly, humanized antibodies are chimeric antibodies, wherein substantially less than an intact human variable domain has been substituted by the corresponding sequence from a non-human species. In practice, humanized antibodies may be typically human antibodies in which some CDR residues and possibly some framework residues are substituted by residues from analogous sites in rodent antibodies.


Human antibodies can also be produced using various additional techniques known in the art, including phage-display libraries (Hoogenboom, H. R. and Winter, G. (1991). By-passing immunisation. Human antibodies from synthetic repertoires of germline VH gene segments rearranged in vitro. J Mol Biol 227, 381-388; Marks, J. D. et al. (1991). By-passing immunization. Human antibodies from V-gene libraries displayed on phage. J Mol Biol 222, 581-597; Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96; and Boerner, P. et al. (1991). Production of antigen-specific human monoclonal antibodies from in vitro-primed human splenocytes. J Immunol 147, 86-95). Humanized antibodies can also be created by introducing sequences encoding human immunoglobulin loci into transgenic animals, e.g., into mice in which the endogenous immunoglobulin genes have been partially or completely inactivated. Upon antigenic challenge, human antibody production is observed in such animals which closely resembles that seen in humans in all respects, including gene rearrangement, chain assembly, and antibody repertoire. Ample guidance for practicing such an approach is provided in the literature of the art (for example, refer to: U.S. Pat. Nos. 5,545,807; 5,545,806; 5,569,825; 5,625,126; 5,633,425; and 5,661,016; Marks, J. D. et al. (1992). By-passing immunization: building high affinity human antibodies by chain shuffling. Biotechnology (N.Y.) 10(7), 779-783; Lonberg et al., 1994. Nature 368:856-859; Morrison, S. L. (1994). News and View: Success in Specification. Nature 368, 812-813; Fishwild, D. M. et al. (1996). High-avidity human IgG kappa monoclonal antibodies from a novel strain of minilocus transgenic mice. Nat Biotechnol 14, 845-851; Neuberger, M. (1996). Generating high-avidity human Mabs in mice. Nat Biotechnol 14, 826; and Lonberg, N. and Huszar, D. (1995). Human antibodies from transgenic mice. Int Rev Immunol 13, 65-93).


It will be appreciated that the inhibitory agents of the present invention may be administered concurrently with the CPT (e.g. by formulating them in a single composition) or may be administered prior to or following CPT administration.


The agents of the present invention can be provided to the individual per se, or as part of a pharmaceutical composition where it is mixed with a pharmaceutically acceptable carrier.


As used herein a “pharmaceutical composition” refers to a preparation of one or more of the active ingredients described herein with other chemical components such as physiologically suitable carriers and excipients. The purpose of a pharmaceutical composition is to facilitate administration of a compound to an organism.


Herein the term “active ingredient” refers to the polypeptide or polynucleotide preparation, which is accountable for the biological effect.


Hereinafter, the phrases “physiologically acceptable carrier” and “pharmaceutically acceptable carrier,” which may be used interchangeably, refer to a carrier or a diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound. An adjuvant is included under these phrases.


Herein, the term “excipient” refers to an inert substance added to a pharmaceutical composition to further facilitate administration of an active ingredient. Examples, without limitation, of excipients include calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils, and polyethylene glycols.


Techniques for formulation and administration of drugs may be found in the latest edition of “Remington's Pharmaceutical Sciences,” Mack Publishing Co., Easton, Pa., which is herein fully incorporated by reference.


Suitable routes of administration may, for example, include oral, rectal, transmucosal, especially transnasal, intestinal, or parenteral delivery, including intramuscular, subcutaneous, and intramedullary injections, as well as intrathecal, direct intraventricular, intravenous, inrtaperitoneal, intranasal, or intraocular injections.


Alternately, one may administer the pharmaceutical composition in a local rather than systemic manner, for example, via injection of the pharmaceutical composition directly into a tissue region of a patient.


Pharmaceutical compositions of the present invention may be manufactured by processes well known in the art, e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping, or lyophilizing processes.


Pharmaceutical compositions for use in accordance with the present invention thus may be formulated in conventional manner using one or more physiologically acceptable carriers comprising excipients and auxiliaries, which facilitate processing of the active ingredients into preparations that can be used pharmaceutically. Proper formulation is dependent upon the route of administration chosen.


For injection, the active ingredients of the pharmaceutical composition may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological salt buffer. For transmucosal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art.


For oral administration, the pharmaceutical composition can be formulated readily by combining the active compounds with pharmaceutically acceptable carriers well known in the art. Such carriers enable the pharmaceutical composition to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient. Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries as desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, and sodium carbomethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP). If desired, disintegrating agents, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof, such as sodium alginate, may be added.


Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active compound doses.


Pharmaceutical compositions that can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate, and, optionally, stabilizers. In soft capsules, the active ingredients may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration.


For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner.


For administration by nasal inhalation, the active ingredients for use according to the present invention are conveniently delivered in the form of an aerosol spray presentation from a pressurized pack or a nebulizer with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro-tetrafluoroethane, or carbon dioxide. In the case of a pressurized aerosol, the dosage may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of, for example, gelatin for use in a dispenser may be formulated containing a powder mix of the compound and a suitable powder base, such as lactose or starch.


The pharmaceutical composition described herein may be formulated for parenteral administration, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with, optionally, an added preservative. The compositions may be suspensions, solutions, or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing, and/or dispersing agents.


Pharmaceutical compositions for parenteral administration include aqueous solutions of the active preparation in water-soluble form. Additionally, suspensions of the active ingredients may be prepared as appropriate oily or water-based injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters such as ethyl oleate, triglycerides, or liposomes. Aqueous injection suspensions may contain substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents that increase the solubility of the active ingredients, to allow for the preparation of highly concentrated solutions.


Alternatively, the active ingredient may be in powder form for constitution with a suitable vehicle, e.g., a sterile, pyrogen-free, water-based solution, before use.


The pharmaceutical composition of the present invention may also be formulated in rectal compositions such as suppositories or retention enemas, using, for example, conventional suppository bases such as cocoa butter or other glycerides.


Pharmaceutical compositions suitable for use in the context of the present invention include compositions wherein the active ingredients are contained in an amount effective to achieve the intended purpose. More specifically, a “therapeutically effective amount” means an amount of active ingredients (e.g., a nucleic acid construct) effective to prevent, alleviate, or ameliorate symptoms of a disorder (e.g., ischemia) or prolong the survival of the subject being treated.


Determination of a therapeutically effective amount is well within the capability of those skilled in the art, especially in light of the detailed disclosure provided herein.


For any preparation used in the methods of the invention, the dosage or the therapeutically effective amount can be estimated initially from in vitro and cell culture assays. For example, a dose can be formulated in animal models to achieve a desired concentration or titer. Such information can be used to more accurately determine useful doses in humans.


Toxicity and therapeutic efficacy of the active ingredients described herein can be determined by standard pharmaceutical procedures in vitro, in cell cultures or experimental animals. The data obtained from these in vitro and cell culture assays and animal studies can be used in formulating a range of dosage for use in human. The dosage may vary depending upon the dosage form employed and the route of administration utilized. The exact formulation, route of administration, and dosage can be chosen by the individual physician in view of the patient's condition. (See, e.g., Fingl, E. et al. (1975), “The Pharmacological Basis of Therapeutics,” Ch. 1, p. 1.)


Dosage amount and administration intervals may be adjusted individually to provide sufficient plasma or brain levels of the active ingredient to induce or suppress the biological effect (i.e., minimally effective concentration, MEC). The MEC will vary for each preparation, but can be estimated from in vitro data. Dosages necessary to achieve the MEC will depend on individual characteristics and route of administration. Detection assays can be used to determine plasma concentrations.


Depending on the severity and responsiveness of the condition to be treated, dosing can be of a single or a plurality of administrations, with course of treatment lasting from several days to several weeks, or until cure is effected or diminution of the disease state is achieved.


The amount of a composition to be administered will, of course, be dependent on the subject being treated, the severity of the affliction, the manner of administration, the judgment of the prescribing physician, etc.


Compositions of the present invention may, if desired, be presented in a pack or dispenser device, such as an FDA-approved kit, which may contain one or more unit dosage forms containing the active ingredient. The pack may, for example, comprise metal or plastic foil, such as a blister pack. The pack or dispenser device may be accompanied by instructions for administration. The pack or dispenser device may also be accompanied by a notice in a form prescribed by a governmental agency regulating the manufacture, use, or sale of pharmaceuticals, which notice is reflective of approval by the agency of the form of the compositions for human or veterinary administration. Such notice, for example, may include labeling approved by the U.S. Food and Drug Administration for prescription drugs or of an approved product insert. Compositions comprising a preparation of the invention formulated in a pharmaceutically acceptable carrier may also be prepared, placed in an appropriate container, and labeled for treatment of an indicated condition, as further detailed above.


It is expected that during the life of a patent maturing from this application many relevant reporter polypeptides will be developed and the scope of the term reporter polypeptide is intended to include all such new technologies a priori.


As used herein the term “about” refers to ±10%.


The terms “comprises”, “comprising”, “includes”, “including”, “having” and their conjugates mean “including but not limited to”.


The term “consisting of means “including and limited to”.


The term “consisting essentially of” means that the composition, method or structure may include additional ingredients, steps and/or parts, but only if the additional ingredients, steps and/or parts do not materially alter the basic and novel characteristics of the claimed composition, method or structure.


As used herein, the singular form “a”, an and the include plural references unless the context clearly dictates otherwise. For example, the term “a polypeptide” or “at least one polypeptide” may include a plurality of polypeptides, including mixtures thereof.


As used herein the term “method” refers to manners, means, techniques and procedures for accomplishing a given task including, but not limited to, those manners, means, techniques and procedures either known to, or readily developed from known manners, means, techniques and procedures by practitioners of the chemical, pharmacological, biological, biochemical and medical arts.


It is appreciated that certain features of the invention, which are, for clarity, described in the context of separate embodiments, may also be provided in combination in a single embodiment. Conversely, various features of the invention, which are, for brevity, described in the context of a single embodiment, may also be provided separately or in any suitable subcombination or as suitable in any other described embodiment of the invention. Certain features described in the context of various embodiments are not to be considered essential features of those embodiments, unless the embodiment is inoperative without those elements.


Various embodiments and aspects of the present invention as delineated hereinabove and as claimed in the claims section below find experimental support in the following examples.


EXAMPLES

Reference is now made to the following examples, which together with the above descriptions illustrate some embodiments of the invention in a non limiting fashion.


Reference is now made to the following examples, which together with the above descriptions, illustrate the invention in a non limiting fashion.


Generally, the nomenclature used herein and the laboratory procedures utilized in the present invention include molecular, biochemical, microbiological and recombinant DNA techniques. Such techniques are thoroughly explained in the literature. See, for example, “Molecular Cloning: A laboratory Manual” Sambrook et al., (1989); “Current Protocols in Molecular Biology” Volumes I-III Ausubel, R. M., ed. (1994); Ausubel et al., “Current Protocols in Molecular Biology”, John Wiley and Sons, Baltimore, Md. (1989); Perbal, “A Practical Guide to Molecular Cloning”, John Wiley & Sons, New York (1988); Watson et al., “Recombinant DNA”, Scientific American Books, New York; Birren et al. (eds) “Genome Analysis: A Laboratory Manual Series”, Vols. 1-4, Cold Spring Harbor Laboratory Press, New York (1998); methodologies as set forth in U.S. Pat. Nos. 4,666,828; 4,683,202; 4,801,531; 5,192,659 and 5,272,057; “Cell Biology: A Laboratory Handbook”, Volumes I-III Cellis, J. E., ed. (1994); “Culture of Animal Cells—A Manual of Basic Technique” by Freshney, Wiley-Liss, N.Y. (1994), Third Edition; “Current Protocols in Immunology” Volumes I-III Coligan J. E., ed. (1994); Stites et al. (eds), “Basic and Clinical Immunology” (8th Edition), Appleton & Lange, Norwalk, Conn. (1994); Mishell and Shiigi (eds), “Selected Methods in Cellular Immunology”, W.H. Freeman and Co., New York (1980); available immunoassays are extensively described in the patent and scientific literature, see, for example, U.S. Pat. Nos. 3,791,932; 3,839,153; 3,850,752; 3,850,578; 3,853,987; 3,867,517; 3,879,262; 3,901,654; 3,935,074; 3,984,533; 3,996,345; 4,034,074; 4,098,876; 4,879,219; 5,011,771 and 5,281,521; “Oligonucleotide Synthesis” Gait, M. J., ed. (1984); “Nucleic Acid Hybridization” Hames, B. D., and Higgins S. J., eds. (1985); “Transcription and Translation” Hames, B. D., and Higgins S. J., eds. (1984); “Animal Cell Culture” Freshney, R. I., ed. (1986); “Immobilized Cells and Enzymes” IRL Press, (1986); “A Practical Guide to Molecular Cloning” Perbal, B., (1984) and “Methods in Enzymology” Vol. 1-317, Academic Press; “PCR Protocols: A Guide To Methods And Applications”, Academic Press, San Diego, Calif. (1990); Marshak et al., “Strategies for Protein Purification and Characterization—A Laboratory Course Manual” CSHL Press (1996); all of which are incorporated by reference as if fully set forth herein. Other general references are provided throughout this document. The procedures therein are believed to be well known in the art and are provided for the convenience of the reader. All the information contained therein is incorporated herein by reference.


Example 1
Construction of a Cherry/YFP CD-Tagged Reporter Clone Library

Gathering of quantitative information from time-lapse fluorescent movies of proteins in individual living cells is a difficult task. In order to overcome such difficulties, a system for dynamic proteomics was developed. [Perlman, Slack et al. 2004, Science 306: 1194-1198; Echeverri and Perrimon 2006, Nat Rev Genet 7: 373-384; Eggert and Mitchison 2006, Curr Opin Chem Biol 10: 232-237; Megason and Fraser 2007, Cell 130(5): 784-95)]. This system for tagging proteins in human cells, is based on a retrovirally based CD-tagging approach [Sigal et al., Nature Protocols, Vol 2, No. 6, 2007; Sigal et al., Nature Methods, Vol 3, No. 7, 2006; Sigal et al., Nature 444, October 2006, p. 643-646, all of which are incorporated herein by reference]. This allows construction of a library of cell clones, each expressing a fluorescently tagged, full-length protein from its endogenous chromosomal location.


Materials and Methods


A library of fluorescently tagged proteins was constructed in non-small cell lung carcinoma cell line (H1299) in a two stage process. In both stages a fluorescent reporter was integrated into the genome via Central Dogma tagging (CD-tagging) (Otsu 1979; Jarvik, Adler et al. 1996; Jarvik, Fisher et al. 2002; Sigal, Danon et al. 2007).


The first stage was carried out in order to produce a parental clone in which the nucleus is colored brighter than the cytoplasm and the cytoplasm is colored brighter than the medium. To achieve this, a red fluorescent protein, mCherry (Shaner, Campbell et al. 2004), was introduced in two rounds of CD-tagging. In the first round, clone H7a with tagged protein XRCC5, localized to the nucleus, was selected. In the second round (carried out on the previously selected clone H7a), clone H7 with tagged DAP1 localized to the whole intracellular domain was selected. Following these two steps, a parental clone was obtained expressing two mCherry endogenously tagged proteins (XRCC5 and DAP1), stained in the cytoplasm and brighter in the nucleus.


The second stage in the generation of the library was to use CD-tagging in order to tag different proteins with a second color EYFP or Venus (Nagai, Ibata et al. 2002) within the parental clone H1299-ul.


CD tagging described in detail by Sigal et al. [Sigal et al., Nature Protocols, Vol 2, No. 6, 2007], incorporated herein by reference. Briefly, a fluorescent protein (FP), flanked by splice acceptor and donor sequences was integrated into the genome as an artificial exon via retroviral vectors (U5000, U5001, U5002), each containing FP in one of 3 reading frames. Cells positive for relevant FP fluorescence were sorted using flow cytometry into 384 well plates and expanded into cell clones.


Results


To obtain reliable image analysis of cell movies, the parental cell (H1299 non-small cell lung carcinoma cell line) was tagged with a red fluorophore (mCherry) that colors the cytoplasm and, more strongly, the nucleus (FIG. 1C). The resulting cell clone showed no growth or morphological differences relative to the untagged parental cells. Custom software used the mCherry fluorescence to automatically distinguish the cell from its background, and to distinguish the nucleus from the cytoplasm (FIGS. 2A-D). Attempts to use transfected red proteins or exogenous dyes were unsuccessful because they led to high cell-cell variability of the tag which made it difficult to analyze the images. To avoid this variability, CD-tagging was used to introduce the red tag into endogenous proteins and a clone was selected with a fluorescence pattern suitable for image analysis. This clone was then used as a basis for the present tagged protein library: A yellow fluorescent marker was introduced into the red-tagged cells by a second round of CD-tagging, following which the yellow tagged cells were expanded into clones, and the tagged proteins were identified (FIGS. 1A-E). Thus, the red tagging is the same in all cells of the library, and is independent of the second yellow stain of the protein of interest.


Example 2
Identification of Tagged Proteins in the Library of the Present Invention

Materials and Methods


Tagged protein identities were determined by 3′RACE, using a nested PCR reaction that amplified the section between the FP and the polyA tail of the mRNA of the host gene. The PCR product was sequenced directly and aligned to the genome.


Results


The library listed herein below includes 1200 different tagged proteins, of which 80% are characterized proteins and 20% are novel proteins.


Table 2, herein below lists the novel proteins which were tagged according to the method of the present invention. The table also provides the results of measurement the ratio of total fluorescence in the cytoplasm vs. total fluorescence in the whole cell for each of these proteins, above 0.5 is denoted as nuclear localization and below 0.5 as cytoplasmic localization.














TABLE 2





SEQ


Cytoplasm/




ID


whole


NO:
GB number
Description
cell
Nucleus
Cytoplasm




















1
AA282714.1
AA282714 zt13f10.r1
0.7866
0
1




NCI_CGAP_GCB1 Homo sapiens




cDNA clone IMAGE: 713035 5′,




mRNA sequence


2
AA479512.1
AA479512 zv21f09.s1
0.779
0
1




Soares_NhHMPu_S1 Homo





sapiens cDNA clone





IMAGE: 754313 3′, mRNA




sequence


3
AA843465.1
AA843465 aj54c11.s1
0.3618
1
0




Soares_testis_NHT Homo sapiens




cDNA clone IMAGE: 1394132 3′,




mRNA sequence


4
AA928516.1
AA928516 om17h03.s1
0.4001
1
0




Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone





IMAGE: 1541333 3′, mRNA




sequence


5
AF086125.1
HUMZA79D12 Homo sapiens full
0.8349
0
1




length insert cDNA clone ZA79D12


6
AF087973.1
HUMYU79H10 Homo sapiens full
0.7233
0
1




length insert cDNA clone YU79H10


7
AI027434.1
AI027434 ow49f09.s1
0.2965
1
0




Soares_parathyroid_tumor_NbHPA





Homo sapiens cDNA clone





IMAGE: 1650185 3′ similar to




TR: Q40462 Q40462 NTGB1,




mRNA sequence


8
AI208228.1
AI208228 qg50b01.x1
0.7128
0
1




Soares_testis_NHT Homo sapiens




cDNA clone IMAGE: 1838569 3′,




mRNA sequence


9
AI434862.1
AI434862 ti13c03.x1
0.7284
0
1




NCI_CGAP_Kid11 Homo sapiens




cDNA clone IMAGE: 2130340 3′,




mRNA sequence


10
AI671392.1
AI671392 wc29g07.x1
0.3552
1
0




NCI_CGAP_Kid11 Homo sapiens




cDNA clone IMAGE: 2316636 3′,




mRNA sequence


11
AI733141.1
AI733141 ol81a03.x5
0.5479
0
1




NCI_CGAP_Kid5 Homo sapiens




cDNA clone IMAGE: 1535980 3′,




mRNA sequence


12
AI801879.1
AI801879 tx28f05.x1
0.2595
1
0




NCI_CGAP_Lu24 Homo sapiens




cDNA clone IMAGE: 2270913 3′,




mRNA sequence


13
AI870477.1
AI870477 wl74b03.x1
0.7639
0
1




NCI_CGAP_Brn25 Homo sapiens




cDNA clone IMAGE: 2430605 3′,




mRNA sequence


14
AK022356.1

Homo sapiens cDNA FLJ12294 fis,

0.6871
0
1




clone MAMMA1001817


15
AK023312.1

Homo sapiens cDNA FLJ13250 fis,

0.7707
0
1




clone OVARC1000724


16
AK023856.1

Homo sapiens cDNA FLJ13794 fis,

0.2276
1
0




clone THYRO1000092


17
AK024998.1

Homo sapiens cDNA: FLJ21345

0.6494
0
1




fis, clone COL02694


18
AK057505.1

Homo sapiens cDNA FLJ32943 fis,

0.8767
0
1




clone TESTI2007829


19
AK091021.1

Homo sapiens cDNA FLJ33702 fis,

0.7426
0
1




clone BRAWH2005533


20
AK091830.1

Homo sapiens cDNA FLJ34511 fis,

0.6938
0
1




clone HLUNG2006397


21
AK092541.1

Homo sapiens cDNA FLJ35222 fis,

0.691
0
1




clone PROST2000835


22
AK092875.1

Homo sapiens cDNA FLJ35556 fis,

0.3468
1
0




clone SPLEN2004844


23
AK095109.1

Homo sapiens cDNA FLJ37790 fis,

0.7859
0
1




clone BRHIP3000111


24
AK097658.1

Homo sapiens cDNA FLJ40339 fis,

0.3469
1
0




clone TESTI2032079


25
AK098306.1

Homo sapiens cDNA FLJ40987 fis,

0.6876
0
1




clone UTERU2015062


26
AK124927.1

Homo sapiens cDNA FLJ42937 fis,

0.1741
1
0




clone BRSSN2014556


27
AK127572.1

Homo sapiens cDNA FLJ45665 fis,

0.5898
0
1




clone CTONG2027959


28
AK127877.1

Homo sapiens cDNA FLJ45982 fis,

0.7119
0
1




clone PROST2017729


29
AK130903.1

Homo sapiens cDNA FLJ27393 fis,

0.7623
0
1




clone WMC01011


30
AK131516.1

Homo sapiens cDNA FLJ16742 fis,

0.8201
0
1




clone BRAWH2008993


31
AV741821.1
AV741821 AV741821 CB Homo
0.7017
0
1





sapiens cDNA clone CBLACB04 5′,





mRNA sequence


32
AW070221.1
AW070221 xa09d05.x1
0.6662
0
1




Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone





IMAGE: 2567817 3′ similar to




TR: O15503 O15503 INSULIN




INDUCED PROTEIN 1.;, mRNA




sequence


33
AW592040.1
AW592040 hf37f06.x1
0.8192
0
1




Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone





IMAGE: 2934083 3′, mRNA




sequence


34
AW662723.1
AW662723 hi35g04.x1
0.623
0
1




NCI_CGAP_Co14 Homo sapiens




cDNA clone IMAGE: 2974326 3′




similar to gb: M60724 RIBOSOMAL




PROTEIN S6 KINASE (HUMAN);,




mRNA sequence


35
AY054401.3

Homo sapiens non-coding

0.7634
0
1




transcript BT1C (BDNF) mRNA,




complete sequence; alternatively




spliced


36
AY176665.1

Homo sapiens nervous system

0.7225
0
1




abundant protein 11 (NSAP11)




mRNA, complete cds


37
BC033363.1
Homo sapiens, clone
0.8908
0
1




IMAGE: 4753714, mRNA


38
BC034424.1

Homo sapiens hexosaminidase A

0.6379
0
1




(alpha polypeptide), mRNA (cDNA




clone IMAGE: 4823589)


39
BC035195.2

Homo sapiens cDNA clone

0.6273
0
1




IMAGE: 5266689


40
BC035377.1

Homo sapiens cDNA clone

0.4531
1
0




IMAGE: 4826240


41
BC038752.1

Homo sapiens cDNA clone

0.7525
0
1




IMAGE: 5269351


42
BC039104.1

Homo sapiens hypothetical protein

0.8318
0
1




LOC283404, mRNA (cDNA clone




IMAGE: 4828118)


43
BC040610.1

Homo sapiens ribosomal protein

0.7936
0
1




L4, mRNA (cDNA clone




IMAGE: 3897039)


44
BC042060.1

Homo sapiens olfactory receptor,

0.7563
0
1




family 7, subfamily E, member 47




pseudogene, mRNA (cDNA clone




IMAGE: 5590288)


45
BC042816.1

Homo sapiens cDNA clone

0.7201
0
1




IMAGE: 5314175


46
BC042855.1

Homo sapiens cDNA clone

0.8326
0
1




IMAGE: 5313513, with apparent




retained intron


47
BC043574.1
Homo sapiens, clone
0.685
0
1




IMAGE: 5222953, mRNA


48
BC044257.1
Homo sapiens, clone
0.6643
0
1




IMAGE: 6063621, mRNA


49
BC044741.1

Homo sapiens cDNA clone

0.3626
1
0




IMAGE: 4828106


50
BC053955.1

Homo sapiens hypothetical protein

0.6361
0
1




LOC285548, mRNA (cDNA clone




IMAGE: 4839316)


51
BC054862.1

Homo sapiens cDNA clone

0.8227
0
1




IMAGE: 4288461, partial cds


52
BC078172.1

Homo sapiens cDNA clone

0.8116
0
1




IMAGE: 5760022, partial cds


53
BC108263.1

Homo sapiens transmembrane

0.8339
0
1




protein 56, mRNA (cDNA clone




IMAGE: 4801733), **** WARNING:




chimeric clone ****


54
BC127846.1

Homo sapiens cDNA clone

0.8948
0
1




IMAGE: 40134482


55
BE745782.1
BE745782 601579970F1
0.2625
1
0




NIH_MGC_9 Homo sapiens cDNA




clone IMAGE: 3928841 5′, mRNA




sequence


56
BE785612.1
BE785612 601475144F1
0.7293
0
1




NIH_MGC_68 Homo sapiens




cDNA clone IMAGE: 3878051 5′,




mRNA sequence


57
BE044435.1
BE044435 ho45d08.x1
0.7093
0
1




Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone





IMAGE: 3040335 3′, mRNA




sequence


58
BF062994.1
BF062994 7h73f05.x1
0.714
0
1




NCI_CGAP_Co16 Homo sapiens




cDNA clone IMAGE: 3321633 3′,




mRNA sequence


59
BF245041.1
BF245041 601864168F1
0.7327
0
1




NIH_MGC_57 Homo sapiens




cDNA clone IMAGE: 4082368 5′,




mRNA sequence


60
BF594738.1
BF594738 7o54h12.x1
0.2631
1
0




NCI_CGAP_Kid11 Homo sapiens




cDNA clone IMAGE: 3577991 3′,




mRNA sequence


61
BF688062.1
BF688062 602067272F1
0.2489
1
0




NIH_MGC_57 Homo sapiens




cDNA clone IMAGE: 4066433 5′,




mRNA sequence


62
BG189068.1
BG189068 RST8104 Athersys
0.6341
0
1




RAGE Library Homo sapiens




cDNA, mRNA sequence


63
BG201613.1
BG201613 RST20954 Athersys
0.194
1
0




RAGE Library Homo sapiens




cDNA, mRNA sequence


64
BG203790.1
BG203790 RST23181 Athersys
0.2773
1
0




RAGE Library Homo sapiens




cDNA, mRNA sequence


65
BI462136.1
BI462136 603205131F1
0.3108
1
0




NIH_MGC_97 Homo sapiens




cDNA clone IMAGE: 5270983 5′,




mRNA sequence


66
BI559775.1
BI559775 603252664F1
0.727
0
1




NIH_MGC_97 Homo sapiens




cDNA clone IMAGE: 5295231 5′,




mRNA sequence


67
BI825982.1
BI825982 603076566F1
0.7214
0
1




NIH_MGC_119 Homo sapiens




cDNA clone IMAGE: 5168225 5′,




mRNA sequence


68
BM461531.1
BM461531
0.4477
1
0




AGENCOURT_6421147




NIH_MGC_67 Homo sapiens




cDNA clone IMAGE: 5501266 5′,




mRNA sequence


69
BM690995.1
BM690995 UI-E-CI1-aba-d-08-0-
0.7291
0
1




UI.r1 UI-E-CI1 Homo sapiens




cDNA clone UI-E-CI1-aba-d-08-0-




UI 5′, mRNA sequence


70
BQ184944.1
BQ184944 UI-E-EJ1-ajo-c-04-0-
0.7141
0
1




UI.s1 UI-E-EJ1 Homo sapiens




cDNA clone UI-E-EJ1-ajo-c-04-0-




UI 3′, mRNA sequence


71
BQ233546.1
BQ233546
0.6304
0
1




AGENCOURT_7526687




NIH_MGC_70 Homo sapiens




cDNA clone IMAGE: 6018551 5′,




mRNA sequence


72
BU533525.1
BU533525
0.6682
0
1




AGENCOURT_10197749




NIH_MGC_126 Homo sapiens




cDNA clone IMAGE: 6559929 5′,




mRNA sequence


73
BU534173.1
BU534173
0.303
1
0




AGENCOURT_10240114




NIH_MGC_126 Homo sapiens




cDNA clone IMAGE: 6561006 5′,




mRNA sequence


74
BU619815.1
BU619815 UI-H-FH1-bfq-j-08-0-
0.3354
1
0




UI.s1 NCI_CGAP_FH1 Homo





sapiens cDNA clone UI-H-FH1-bfq-





j-08-0-UI 3′, mRNA sequence


75
BX089034.1
BX089034 BX089034
0.8095
0
1




Soares_parathyroid_tumor_NbHPA





Homo sapiens cDNA clone





IMAGp998M163120;




IMAGE: 1240503 5′, mRNA




sequence


76
BX090666.1
BX090666 BX090666
0.7584
0
1




Soares_testis_NHT Homo sapiens




cDNA clone IMAGp998D014412;




IMAGE: 1736400 5′, mRNA




sequence


77
BX100329.1
BX100329 BX100329
0.7407
0
1




Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone





IMAGp998H043806;




IMAGE: 1503795 5′, mRNA




sequence


78
BX100818.1
BX100818 BX100818
0.7962
0
1




Soares_fetal_lung_NbHL19W





Homo sapiens cDNA clone





IMAGp998J074430;




IMAGE: 1743462 5′, mRNA




sequence


79
BX103408.1
BX103408 BX103408 Soares
0.3196
1
0




melanocyte 2NbHM Homo sapiens




cDNA clone IMAGp998L01545;




IMAGE: 251664 5′, mRNA




sequence


80
BX103636.1
BX103636 BX103636
0.8348
0
1




Soares_testis_NHT Homo sapiens




cDNA clone IMAGp998J184112;




IMAGE: 1621361 5′, mRNA




sequence


81
BX104605.1
BX104605 BX104605
0.7985
0
1




Soares_testis_NHT Homo sapiens




cDNA clone IMAGp998B211795;




IMAGE: 731444 5′, mRNA




sequence


82
BX537644.1

Homo sapiens mRNA; cDNA

0.7389
0
1




DKFZp686M1498 (from clone




DKFZp686M1498)


83
BX537772.1

Homo sapiens mRNA; cDNA

0.8385
0
1




DKFZp781M2440 (from clone




DKFZp781M2440)


84
BX648555.1

Homo sapiens mRNA; cDNA

0.6607
0
1




DKFZp779B0135 (from clone




DKFZp779B0135)


85
BX648926.1

Homo sapiens mRNA; cDNA

0.3742
1
0




DKFZp686O0329 (from clone




DKFZp686O0329)


86
NM_022895.1

Homo sapiens chromosome 12

0.3436
1
0




open reading frame 43 (C12orf43),




mRNA


87
NM_152318.2

Homo sapiens chromosome 12

0.3186
1
0




open reading frame 45 (C12orf45),




mRNA


88
CR457199.1

Homo sapiens full open reading

0.4427
1
0




frame cDNA clone




RZPDo834G068D for gene




C14orf112, chromosome 14 open




reading frame 112; complete cds,




incl. stopcodon


89
NM_004894.2

Homo sapiens chromosome 14

0.7418
0
1




open reading frame 2 (C14orf2),




transcript variant 1, mRNA


90
BC007346.2

Homo sapiens chromosome 16

0.4108
1
0




open reading frame 14, mRNA




(cDNA clone IMAGE: 3689407),




complete cds


91
NM_033520.1

Homo sapiens chromosome 19

0.622
0
1




open reading frame 33 (C19orf33),




mRNA


92
NM_024038.2

Homo sapiens chromosome 19

0.4308
1
0




open reading frame 43 (C19orf43),




mRNA


93
NM_014047.2

Homo sapiens chromosome 19

0.7672
0
1




open reading frame 53 (C19orf53),




mRNA


94
NM_019108.2

Homo sapiens chromosome 19

0.7063
0
1




open reading frame 61 (C19orf61),




mRNA


95
NM_018840.2

Homo sapiens chromosome 20

0.7255
0
1




open reading frame 24 (C20orf24),




transcript variant 1, mRNA


96
NM_021254.1

Homo sapiens chromosome 21

0.7483
0
1




open reading frame 59 (C21orf59),




mRNA


97
NM_015702.1

Homo sapiens chromosome 2

0.7598
0
1




open reading frame 25 (C2orf25),




mRNA


98
NM_016474.4

Homo sapiens chromosome 3

0.3994
1
0




open reading frame 19 (C3orf19),




mRNA


99
NM_178335.1

Homo sapiens coiled-coil domain

0.7952
0
1




containing 50




(CCDC50), C3ORF6, transcript




variant 2, mRNA


100
NM_032302.2

Homo sapiens proteasome

0.787
0
1




(prosome, macropain) assembly




chaperone 3 (PSMG3), mRNA


101
NM_019607.1

Homo sapiens chromosome 8

0.4354
1
0




open reading frame 44 (C8orf44),




mRNA


102
NM_017998.2

Homo sapiens chromosome 9

0.7684
0
1




open reading frame 40 (C9orf40),




mRNA


103
CB045860.1
CB045860 NISC_gf01a03.x1
0.724
0
1




NCI_CGAP_Kid12 Homo sapiens




cDNA clone IMAGE: 3252364 3′,




mRNA sequence


104
CD692919.1
CD692919 EST9442 human
0.6126
0
1




nasopharynx Homo sapiens cDNA,




mRNA sequence


105
CN267986.1
CN267986 17000531863184
0.6675
0
1




GRN_EB Homo sapiens cDNA 5′,




mRNA sequence


106
CN280387.1
CN280387 17000455082974
0.7509
0
1




GRN_ES Homo sapiens cDNA 5′,




mRNA sequence


107
CN398253.1
CN398253 17000424721764
0.7986
0
1




GRN_EB Homo sapiens cDNA 5′,




mRNA sequence


108
CR593740.1
full-length cDNA clone
0.7132
0
1




CS0DF033YJ19 of Fetal brain of





Homo sapiens (human)



109
CR604408.1
full-length cDNA clone
0.8164
0
1




CS0DC001YF03 of Neuroblastoma




Cot 25-normalized of Homo





sapiens (human)



110
CR623475.1
full-length cDNA clone
0.6816
0
1




CS0DB006YA03 of Neuroblastoma




Cot 10-normalized of Homo





sapiens (human)



111
CR626360.1
full-length cDNA clone
0.7563
0
1




CS0DM014YM20 of Fetal liver of





Homo sapiens (human)



112
CR627148.1

Homo sapiens mRNA; cDNA

0.7868
0
1




DKFZp779F2127 (from clone




DKFZp779F2127)


113
CR737784.1
CR737784 CR737784 Homo
0.8232
0
1





sapiens library (Ebert L) Homo






sapiens cDNA clone





IMAGp998C154208;




IMAGE: 1658054 5′, mRNA




sequence


114
CR994463.1
CR994463 CR994463 RZPD
0.659
0
1




no. 9016 Homo sapiens cDNA




clone RZPDp9016A109 5′, mRNA




sequence


115
DB049861.1
DB049861 DB049861 TESTI2
0.8422
0
1





Homo sapiens cDNA clone





TESTI2039270 5′, mRNA




sequence


116
DB054822.1
DB054822 DB054822 TESTI2
0.7785
0
1





Homo sapiens cDNA clone





TESTI2045843 5′, mRNA




sequence


117
DB186251.1
DB186251 DB186251 TLIVE2
0.2773
1
0





Homo sapiens cDNA clone





TLIVE2006096 5′, mRNA




sequence


118
DB331110.1
DB331110 DB331110 SKMUS2
0.2272
1
0





Homo sapiens cDNA clone





SKMUS2008761 3′, mRNA




sequence


119
DB514539.1
DB514539 DB514539 RIKEN full-
0.7233
0
1




length enriched human cDNA




library, testis Homo sapiens cDNA




clone H013041M08 3′, mRNA




sequence


120
DB522524.1
DB522524 DB522524 RIKEN full-
0.7956
0
1




length enriched human cDNA




library, testis Homo sapiens cDNA




clone H013076C14 3′, mRNA




sequence


121
DC347972.1
DC347972 DC347972 CTONG3
0.6791
0
1





Homo sapiens cDNA clone





CTONG3005404 5′, mRNA




sequence


122
AL137478.1

Homo sapiens mRNA; cDNA

0.8034
0
1




DKFZp434M1123 (from clone




DKFZp434M1123)


123
EF565105.1

Homo sapiens chromosome 16

0.5012
0
1




isolate HA_003251 mRNA




sequence


124
DB089792.1
DB089792 DB089792 TESTI4
0.7495
0
1





Homo sapiens cDNA clone





TESTI4038491 5′, mRNA




sequence


125
NM_018011.3

Homo sapiens arginine and

0.3163
1
0




glutamate rich 1 (ARGLU1), mRNA


126
NM_018048.2

Homo sapiens mago-nashi

0.7617
0
1




homolog B (Drosophila)




(MAGOHB), mRNA


127
NM_017669.2

Homo sapiens excision repair

0.8155
0
1




cross-complementing rodent repair




deficiency, complementation group




6-like (ERCC6L), mRNA


128
NM_144726.1

Homo sapiens ring finger protein

0.8475
0
1




145 (RNF145), mRNA


129
XR_040666.1
PREDICTED: Homo sapiens
0.4847
1
0




misc_RNA (FLJ32065), miscRNA


130
NM_001039796.1

Homo sapiens hypothetical protein

0.752
0
1




LOC649446 (FLJ35776), mRNA


131
NM_015168.1

Homo sapiens zinc finger CCCH-

0.1932
1
0




type containing 4 (ZC3H4), mRNA


132
NM_020827.1

Homo sapiens KIAA1430

0.3263
1
0




(KIAA1430), mRNA


133
NM_001009993.2

Homo sapiens family with

0.6583
0
1




sequence similarity 168, member B




(FAM168B), mRNA


134
NM_001086521.1

Homo sapiens chromosome 17

0.6882
0
1




open reading frame 89 (C17orf89),




mRNA


135
NR_002187.2

Homo sapiens hypothetical protein

0.7608
0
1




LOC286016 (LOC286016) on




chromosome 7


136
NM_001080507.1

Homo sapiens oocyte expressed

0.6789
0
1




protein homolog (dog) (OOEP),




mRNA


137
XR_039886.1
PREDICTED: Homo sapiens
0.6685
0
1




misc_RNA (LOC541471),




miscRNA


138
NM_020314.4

Homo sapiens chromosome 16

0.7113
0
1




open reading frame 62 (C16orf62),




mRNA


139
NM_024093.1

Homo sapiens chromosome 2

0.7338
0
1




open reading frame 49 (C2orf49),




mRNA


140
NM_001004333.3

Homo sapiens ribonuclease,

0.5969
0
1




RNase K (RNASEK), mRNA


141
AK098520.1

Homo sapiens cDNA FLJ25654 fis,

0.2283
1
0




clone TST00252


142
NM_001093732.1

Homo sapiens hCG2033311

0.6534
0
1




(LOC644928), mRNA


143
NM_015681.3

Homo sapiens B9 protein domain 1

0.6197
0
1




(B9D1), mRNA


144
T85821.1
T85821 yd57b09.r1 Soares fetal
0.7951
0
1




liver spleen 1NFLS Homo sapiens




cDNA clone IMAGE: 112313 5′




similar to contains MER25




repetitive element;, mRNA




sequence


145
T85822.1
T85822 yd57b10.r1 Soares fetal
0.7259
0
1




liver spleen 1NFLS Homo sapiens




cDNA clone IMAGE: 112315 5′,




mRNA sequence


146
T85823.1
T85823 yd57b11.r1 Soares fetal
0.815
0
1




liver spleen 1NFLS Homo sapiens




cDNA clone IMAGE: 112317 5′




similar to contains LTR1 repetitive




element;, mRNA sequence


147
T85824.1
T85824 yd57b12.r1 Soares fetal
0.8146
0
1




liver spleen 1NFLS Homo sapiens




cDNA clone IMAGE: 112319 5′,




mRNA sequence


148
AI342698.1
AI342698 qo35e04.x1
0.6337
0
1




NCI_CGAP_Lu5 Homo sapiens




cDNA clone IMAGE: 1910526 3′




similar to gb: L01457




AUTOANTIGEN PM-SCL




(HUMAN);, mRNA sequence


149
AK094352.1

Homo sapiens cDNA FLJ37033 fis,

0.6052
0
1




clone BRACE2011389


150
AK094903.1

Homo sapiens cDNA FLJ37584 fis,

0.3903
1
0




clone BRCOC2004950


151
AK128457.1

Homo sapiens cDNA FLJ46600 fis,

0.3942
1
0




clone THYMU3047144


152
AW418496.1
AW418496 ha19c01.x1
0.4929
1
0




NCI_CGAP_Kid12 Homo sapiens




cDNA clone IMAGE: 2874144 3′,




mRNA sequence


153
AX748230.1
Sequence 1755 from Patent
0.7376
0
1




EP1308459


154
BC005233.1

Homo sapiens pancreatic lipase-

0.5561
0
1




related protein 1, mRNA (cDNA




clone IMAGE: 3950129), complete




cds


155
BC036259.1

Homo sapiens hypothetical gene

0.6996
0
1




supported by AK093266, mRNA




(cDNA clone IMAGE: 5271013)


156
BG221753.1
BG221753 RST41568 Athersys
0.6439
0
1




RAGE Library Homo sapiens




cDNA, mRNA sequence


157
BX648475.1

Homo sapiens mRNA; cDNA

0.795
0
1




DKFZp686P11156 (from clone




DKFZp686P11156)


158
NM_017915.2

Homo sapiens chromosome 12

0.3315
1
0




open reading frame 48 (C12orf48),




mRNA


159
BC001722.1

Homo sapiens chromosome 14

0.6383
0
1




open reading frame 166, mRNA




(cDNA clone MGC: 680




IMAGE: 3528725), complete cds


160
NM_024294.2

Homo sapiens chromosome 6

0.5592
0
1




open reading frame 106




(C6orf106), transcript variant 1,




mRNA


161
NM_138701.2

Homo sapiens chromosome 7

0.4211
1
0




open reading frame 11 (C7orf11),




mRNA


162
NG_005982.3

Homo sapiens ribosomal protein,

0.7143
0
1




large, P1 pseudogene




(LOC729416) on chromosome 5


163
N68399.1
N68399 za13b04.s1 Soares fetal
0.6699
0
1




liver spleen 1NFLS Homo sapiens




cDNA clone IMAGE: 292399 3′




similar to SW: OLF3_MOUSE




P23275 OLFACTORY RECEPTOR




OR3. [1];, mRNA sequence


164
NT_022171.14
Hs2_22327 Homo sapiens
0.6871
0
1




chromosome 2 genomic contig,




reference assembly









Table 3 lists all the proteins in the library.











TABLE 3





Clone ID
Protein name
Protein description







310505p4f1b8
08-Sep
septin 9


170407pl3E6
09-Sep
septin 10 isoform 1


200208pl2D10
10-Sep
septin 11


050707pl1E1
BE745782
heparan sulfate D-glucosaminyl


200906pl2E4
A-761H5.5
hypothetical protein LOC440350


310806pl2C10
AA033764
zk19b11.r1 Soares_pregnant_uterus_NbHPU





Homo sapiens cDNA clone IMAGE: 470973 5′,





mRNA sequence.


130207pl1D8
AA282714
zt13f10.r1 NCI_CGAP_GCB1 Homo sapiens cDNA




clone IMAGE: 713035 5′, mRNA sequence.


310806pl2E7
AA431778
zw80e04.s1 Soares_testis_NHT Homo sapiens




cDNA clone IMAGE: 782526 3′, mRNA sequence.


050707pl3H3
AA435616
zt74d10.s1 Soares_testis_NHT Homo sapiens




cDNA clone IMAGE: 728083 3′, mRNA sequence.


150506pl1F4
AA479512
zv21f09.s1 Soares_NhHMPu_S1 Homo sapiens




cDNA clone IMAGE: 754313 3′, mRNA sequence.


311007pl2C7
AA758225
ah68g10.s1 Soares_testis_NHT Homo sapiens




cDNA clone 1320834 3′, mRNA sequence.


150506pl1A5
AA843465
aj54c11.s1 Soares_testis_NHT Homo sapiens




cDNA clone IMAGE: 1394132 3′, mRNA sequence.


041206pl4C2
AA913230
ol41h07.s1 Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone IMAGE: 1526077 3′, mRNA





sequence.


041206pl7B5
AA928516
om17h03.s1 Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone IMAGE: 1541333 3′, mRNA





sequence.


310806pl3A11
AA933969
on71h05.s1 Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone IMAGE: 1562169 3′ similar to





gb: K00558 TUBULIN ALPHA-1 CHAIN (HUMAN);,




mRNA sequence.


200906pl3A5
AB051441

Homo sapiens mRNA for KIAA1654 protein, partial





cds.


200208pl2E12
ABCA4
ATP-binding cassette, sub-family A member 4


200906pl1E6
ABCF1
ATP-binding cassette, sub-family F, member 1


10704p110c8
ACOT7
acyl-CoA thioesterase 7 isoform hBACHb


171104p42c6
ACTN1
actinin, alpha 1


31104p37b6
ACTN4
actinin, alpha 4


050707pl1B4
ACTR1A
ARP1 actin-related protein 1 homolog A,


170407vpl2B6
ACTR2
actin-related protein 2 isoform a


041206pl4D12
ACTR3
ARP3 actin-related protein 3 homolog


311007pl1B8
ACYP2
muscle-type acylphosphatase 2


311007pl3G6
ADH5
class III alcohol dehydrogenase 5 chi subunit


150506pl2E6
ADK
adenosine kinase isoform b


310506pl3C9
AF086125

Homo sapiens full length insert cDNA clone





ZA79D12.


310506pl3C2
AF087973

Homo sapiens full length insert cDNA clone





YU79H10.


200906pl3G9
AF220048

Homo sapiens uncharacterized hematopoietic





stem/progenitor cells protein MDS028 mRNA,




complete cds.


201107pl2A12
AF339799

Homo sapiens clone IMAGE: 2363394, mRNA





sequence.


010806pl2C2
AHNAK
AHNAK nucleoprotein isoform 2


310506pl2A10
AI000260
ov10b02.s1 NCI_CGAP_Kid3 Homo sapiens cDNA




clone IMAGE: 1636875 3′ similar to contains




THR.b3 THR repetitive element;, mRNA




sequence.


041206pl1D9
AI001881
ot39c06.s1 Soares_testis_NHT Homo sapiens




cDNA clone IMAGE: 1619146 3′, mRNA sequence.


010806pl2A5
AI094227
qa43a12.s1 Soares_NhHMPu_S1 Homo sapiens




cDNA clone IMAGE: 1689502 3′, mRNA sequence.


310506pl1E10
AI125255
qd87h09.x1 Soares_testis_NHT Homo sapiens




cDNA clone IMAGE: 1736513 3′, mRNA sequence.


160507pl3F1
AI203131
qr34b09.x1 NCI_CGAP_GC6 Homo sapiens cDNA




clone IMAGE: 1942745 3′, mRNA sequence.


200906pl4F5
AI208228
qg50b01.x1 Soares_testis_NHT Homo sapiens




cDNA clone IMAGE: 1838569 3′, mRNA sequence.


201107pl1A1
AI215862
qm35e03.x1 NCI_CGAP_Lu5 Homo sapiens cDNA




clone IMAGE: 1883836 3′ similar to contains Alu




repetitive element; contains element MER22




repetitive element;, mRNA sequence.


050707pl3E7
AI217733
qh15h09.x1 Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone IMAGE: 1844801 3′ similar to





SW: FTCD_PIG P53603




FORMIMINOTRANSFERASE-




CYCLODEAMINASE; contains element PTR5




repetitive element;, mRNA sequence.


310506pl1G2
AI310103
qo74c04.x1 NCI_CGAP_Kid5 Homo sapiens cDNA




clone IMAGE: 1914246 3′, mRNA sequence.


201107pl3F7
AI342698
qo35e04.x1 NCI_CGAP_Lu5 Homo sapiens cDNA




clone IMAGE: 1910526 3′ similar to gb: L01457




AUTOANTIGEN PM-SCL (HUMAN);, mRNA




sequence.


010806pl2H4
AI434862
ti13c03.x1 NCI_CGAP_Kid11 Homo sapiens cDNA




clone IMAGE: 2130340 3′, mRNA sequence.


050707pl2E11
AI671392
wc29g07.x1 NCI_CGAP_Kid11 Homo sapiens




cDNA clone IMAGE: 2316636 3′, mRNA sequence.


200306f7pl1C8
AI692920
wd42h05.x1 Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone IMAGE: 2330841 3′, mRNA





sequence.


200906pl2B7
AI733141
ol81a03.x5 NCI_CGAP_Kid5 Homo sapiens cDNA




clone IMAGE: 1535980 3′, mRNA sequence.


201107pl4A11
AI769786
wj26e10.x1 NCI_CGAP_Kid12 Homo sapiens




cDNA clone IMAGE: 2403978 3′, mRNA sequence.


150506pl2E8
AI801879
tx28f05.x1 NCI_CGAP_Lu24 Homo sapiens cDNA




clone IMAGE: 2270913 3′, mRNA sequence.


170407pl3F6
AI822094
za73d07.x5 Soares_fetal_lung_NbHL19W Homo





sapiens cDNA clone IMAGE: 298189 3′ similar to





gb: X16667 HOMEOBOX PROTEIN HOX-B3




(HUMAN);, mRNA sequence.


130207pl1C12
AI869329
wl68g08.x1 NCI_CGAP_Brn25 Homo sapiens




cDNA clone IMAGE: 2430110 3′, mRNA sequence.


201107pl1G4
AI869566
wl98c09.x1 NCI_CGAP_Brn25 Homo sapiens




cDNA clone IMAGE: 2432944 3′ similar to




SW:SSRP_HUMAN Q08945 STRUCTURE-




SPECIFIC RECOGNITION PROTEIN 1;, mRNA




sequence.


041206pl5F10
AI870477
wl74b03.x1 NCI_CGAP_Brn25 Homo sapiens




cDNA clone IMAGE: 2430605 3′, mRNA sequence.


041206pl7B4
AJ412031

Homo sapiens mRNA for B-cell neoplasia





associated transcript, (BCMS gene), splice variant




D, non coding transcript.


310806pl1C11
AJ713761
AJ713761 LKPD01 Homo sapiens cDNA clone




LKPD02011, mRNA sequence.


160507pl2B5
AK000451

Homo sapiens cDNA FLJ20444 fis, clone





KAT05128.


130207pl1D5
AK022356

Homo sapiens cDNA FLJ12294 fis, clone





MAMMA1001817.


201107pl1F12
AK023018

Homo sapiens cDNA FLJ12956 fis, clone





NT2RP2005501.


010806pl1E8
AK023312

Homo sapiens cDNA FLJ13250 fis, clone





OVARC1000724.


200906pl1A1
AK023856

Homo sapiens cDNA FLJ13794 fis, clone





THYRO1000092.


311007pl3F10
AK024998

Homo sapiens cDNA: FLJ21345 fis, clone





COL02694.


200906pl2E11
AK025325

Homo sapiens cDNA: FLJ21672 fis, clone





COL09025.


200306f7pl1D8
AK055171

Homo sapiens cDNA FLJ30609 fis, clone





CTONG2000480.


050707pl2B10
AK056115

Homo sapiens cDNA FLJ31553 fis, clone





NT2RI2001178.


310506pl1A4
AK056558

Homo sapiens cDNA FLJ31996 fis, clone





NT2RP7009253.


041206pl3A1
AK057505

Homo sapiens C18orf2 isoform 1 mRNA, complete





sequence, alternatively spliced.


170407pl1G8
AK091021

Homo sapiens cDNA FLJ33702 fis, clone





BRAWH2005533.


041206pl7D6
AK091108

Homo sapiens cDNA FLJ33789 fis, clone





BRSSN2009378.


170407pl1E9
AK092541

Homo sapiens cDNA FLJ35222 fis, clone





PROST2000835.


050707pl1D5
AK092875

Homo sapiens cDNA FLJ35556 fis, clone





SPLEN2004844.


201107pl3F2
AK094352

Homo sapiens cDNA FLJ37033 fis, clone





BRACE2011389.


201107pl2A7
AK094903

Homo sapiens cDNA FLJ37584 fis, clone





BRCOC2004950.


311007pl2G12
AK095077

Homo sapiens cDNA FLJ37758 fis, clone





BRHIP2023869.


170407pl1D7
AK095109

Homo sapiens cDNA FLJ37790 fis, clone





BRHIP3000111.


041206pl1D7
AK097571

Homo sapiens cDNA FLJ40252 fis, clone





TESTI2024299.


010806pl3E4
AK097658

Homo sapiens cDNA FLJ40339 fis, clone





TESTI2032079.


200906pl2D9
AK098170

Homo sapiens cDNA FLJ40851 fis, clone





TRACH2014997, moderately similar to Rattus





norvegicus Ca2+-dependent activator protein





(CAPS) mRNA.


160507pl2G5
AK098264

Homo sapiens cDNA FLJ40945 fis, clone





UTERU2008747.


190607pl1B6
AK098306

Homo sapiens cDNA FLJ40987 fis, clone





UTERU2015062.


041206pl6H5
AK123491

Homo sapiens cDNA FLJ41497 fis, clone





BRTHA2006075.


200906pl2F6
AK123797

Homo sapiens cDNA FLJ41803 fis, clone





NHNPC2002749.


150506pl2B2
AK124927

Homo sapiens cDNA FLJ42937 fis, clone





BRSSN2014556.


200906pl5D9
AK127877

Homo sapiens cDNA FLJ45982 fis, clone





PROST2017729.


280305p1f2e12
AK128282

Homo sapiens cDNA FLJ46419 fis, clone





THYMU3012983, moderately similar to Homo





sapiens zinc finger protein 14 (KOX 6) (ZNF14).



201107pl2D4
AK128457

Homo sapiens cDNA FLJ46600 fis, clone





THYMU3047144.


310806pl1D8
AK128738

Homo sapiens cDNA FLJ16787 fis, clone





PLACE6013222.


310506pl3G7
AK130268

Homo sapiens cDNA FLJ26758 fis, clone





PRS02459.


311007pl3D4
AK130830

Homo sapiens cDNA FLJ27320 fis, clone





TMS07774.


010806pl4E5
AK130903

Homo sapiens cDNA FLJ27393 fis, clone





WMC01011.


150506pl1G6
AK131516

Homo sapiens cDNA FLJ16742 fis, clone





BRAWH2008993.


041206pl2E2
AKAP12
A-kinase anchor protein 12 isoform 1


170407pl1B12
AKAP8L
A kinase (PRKA) anchor protein 8-like


310806pl2E1
AL136790

Homo sapiens mRNA; cDNA DKFZp434F1819





(from clone DKFZp434F1819).


041206pl6H11
AL137366

Homo sapiens mRNA; cDNA DKFZp434F1626





(from clone DKFZp434F1626).


310506pl3B7
AL708335
DKFZp686L2051_r1 686 (synonym: hlcc3) Homo





sapiens cDNA clone DKFZp686L2051 5′, mRNA





sequence.


010806pl1F6
ALDH3B1

Homo sapiens mRNA for aldehyde dehydrogenase





3B1 variant protein.


311007pl1H1
ALDOA
aldolase A


170407pl1G4
ALG14
asparagine-linked glycosylation 14 homolog


180504p21c4
AMD1
S-adenosylmethionine decarboxylase 1 isoform 1


200208pl2G2
ANAPC13
anaphase promoting complex subunit 13


190607pl1C10
ANGPTL4
angiopoietin-like 4 protein isoform a precursor


280705p1f13A8
ANLN
anillin, actin binding protein (scraps homolog,


041206pl4E5
ANP32A
acidic (leucine-rich) nuclear phosphoprotein 32


280305p1f12D9
ANP32B
acidic (leucine-rich) nuclear phosphoprotein 32


160507pl3A1
ANTXR2
anthrax toxin receptor 2


200906pl5A11
ANXA1
annexin I


200906pl4A6
ANXA11
annexin A11


280305p5f2E6
ANXA2
annexin A2 isoform 1


201107pl2G6
ANXA5
annexin 5


170407vpl3H9
ANXA8L1
annexin A8-like 1


150506pl1G7
AOAH
acyloxyacyl hydrolase precursor


311007pl1H12
AOF2
amine oxidase (flavin containing) domain 2


310806pl2B6
APIP
APAF1 interacting protein


311007pl1A7
APLP2
amyloid beta (A4) precursor-like protein 2


201107pl3B8
APP
amyloid beta A4 protein precursor, isoform a


130207p2G10
ARCH

Homo sapiens archease (ARCH) mRNA, partial





cds.


010806pl2D6
ARHGAP18
Rho GTPase activating protein 18


041206pl7B1
ARID1B
AT rich interactive domain 1B (SWI1-like)


050707pl3G1
ARL3
ADP-ribosylation factor-like 3


160507pl2F5
ARL6IP1
ADP-ribosylation factor-like 6 interacting


200208pl2F6
ARMC2
armadillo repeat containing 2


010806pl4E10
ARPC1A
actin related protein 2/3 complex subunit 1A


200906pl2C10
ARPC2
actin related protein 2/3 complex subunit 2


050707pl3E10
ARPC3
actin related protein 2/3 complex subunit 3


200208pl2F12
ASNS

Homo sapiens cDNA FLJ20372 fis, clone





HEP19727, highly similar to M27396 Human




asparagine synthetase mRNA.


200906pl1B3
ATAD1
ATPase family, AAA domain containing 1


170407vpl2E12
ATF1
activating transcription factor 1


050707pl3D10
ATG3
Apg3p


200208pl2A4
ATOX1
antioxidant protein 1


27073j5
ATP1A1
Na+/K+ -ATPase alpha 1 subunit isoform a


310505p4f1c8
ATP5B
ATP synthase, H+ transporting, mitochondrial F1


311007pl1G5
ATP5C1
ATP synthase, H+ transporting, mitochondrial F1


310806pl1E1
ATP5J2
ATP synthase, H+ transporting, mitochondrial F0


170604p17c11
ATP6V1D
H(+)-transporting two-sector ATPase


310806pl1G11
AV702071
AV702071 ADB Homo sapiens cDNA clone




ADBCVC06 5′, mRNA sequence.


200906pl5G5
AV703421
AV703421 ADB Homo sapiens cDNA clone




ADBCBH03 5′, mRNA sequence.


200906pl1F1
AV741821
AV741821 CB Homo sapiens cDNA clone




CBLACB04 5′, mRNA sequence.


200306f7pl1F11
AVEN
cell death regulator aven


150506pl1A10
AW070221
xa09d05.x1 Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone IMAGE: 2567817 3′ similar to





TR: O15503 O15503 INSULIN INDUCED




PROTEIN 1.;, mRNA sequence.


041206pl6F4
AW070342
xa10d08.x1 Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone IMAGE: 2567919 3′, mRNA





sequence.


310506pl1G9
AW136353
UI-H-BI1-acn-f-11-0-UI.s1 NCI_CGAP_Sub3 Homo





sapiens cDNA clone IMAGE: 2715021 3′, mRNA





sequence.


310806pl2D6
AW241724
xn74c07.x1 Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone IMAGE: 2700204 3′, mRNA





sequence.


010806pl2B10
AW291591
UI-H-BI2-agk-g-08-0-UI.s1 NCI_CGAP_Sub4





Homo sapiens cDNA clone IMAGE: 2724686 3′,





mRNA sequence.


201107pl3E2
AW418496
ha19c01.x1 NCI_CGAP_Kid12 Homo sapiens




cDNA clone IMAGE: 2874144 3′, mRNA sequence.


160507pl3A12
AW592040
hf37f06.x1 Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone IMAGE: 2934083 3′, mRNA





sequence.


150506pl1B4
AX748015

Homo sapiens cDNA FLJ35934 fis, clone





TESTI2011315.


201107pl3D2
AX748230

Homo sapiens cDNA FLJ36305 fis, clone





THYMU2004677.


310806pl1D3
AX748388

Homo sapiens cDNA FLJ36653 fis, clone





UTERU2001176.


160507pl1A1
AY054401

Homo sapiens trapped 3′ terminal exon, clone





B2F11.


010806pl2D10
AY176665

Homo sapiens nervous system abundant protein





11 (NSAP11) mRNA, complete cds.


041206pl7C6
AY480055

Homo sapiens GKT-AML5-1 mRNA sequence;





alternatively spliced.


050707pl2G4
BAG1
BCL2-associated athanogene.


310506pl3A4
BAG2
BCL2-associated athanogene 2


170407pl3D4
BAG3
BCL2-associated athanogene 3


170407vpl2C4
BAIAP2
BAI1-associated protein 2 isoform 3


201107pl2D2
BAIAP2L1
BAI1-associated protein 2-like 1


201107pl2H3
BANK1
B-cell scaffold protein with ankyrin repeats 1


050707pl1G4
BARD1
BRCA1 associated RING domain 1


310806pl1G1
BC000085

Homo sapiens cDNA clone IMAGE: 3507983, ****





WARNING: chimeric clone ****.


200906pl3H5
BC011779

Homo sapiens cDNA clone IMAGE: 3941306,





partial cds.


050707pl2E9
BC012743

Homo sapiens cDNA clone IMAGE: 4040306, ****





WARNING: chimeric clone ****.


311007pl3C7
BC014506

Homo sapiens, clone IMAGE: 4863312, mRNA.



180504p12d6
BC014776

Homo sapiens hypothetical LOC541471, mRNA





(cDNA clone MGC: 17532 IMAGE: 3459303),




complete cds.


041206pl2G8
BC015412

Homo sapiens cDNA clone IMAGE: 4393471,





partial cds.


200306f7pl1F1
BC016972

Homo sapiens, clone IMAGE: 3896086, mRNA.



310506pl1D5
BC024924

Homo sapiens cDNA FLJ12974 fis, clone





NT2RP2006103.


041206pl4G1
BC031950

Homo sapiens cDNA clone IMAGE: 4838164.



041206pl3G3
BC033363

Homo sapiens, clone IMAGE: 4753714, mRNA.



201107pl4D10
BC033643

Homo sapiens cDNA clone MGC: 45452





IMAGE: 5562656, complete cds.


010506pl2B6
BC035195

Homo sapiens cDNA clone IMAGE: 5266689.



200306d9pl1C6
BC035377

Homo sapiens cDNA clone IMAGE: 4826240.



201107pl2G5
BC036259

Homo sapiens cDNA FLJ35947 fis, clone





TESTI2011971.


160507pl1B6
BC038752

Homo sapiens cDNA clone IMAGE: 5269351.



310506pl1D10
bc038760
hEST


150506pl1E5
BC039104

Homo sapiens hypothetical protein LOC283404,





mRNA (cDNA clone IMAGE: 4828118).


310806pl2C8
BC039429

Homo sapiens cDNA clone IMAGE: 5303182.



041206pl1C3
BC039533

Homo sapiens, clone IMAGE: 5743964, mRNA.



201107pl1G10
BC039555

Homo sapiens, clone IMAGE: 4249217, mRNA.



050707pl2F12
BC040619

Homo sapiens similar to solute carrier family 16





(monocarboxylic acid transporters), member 14,




mRNA (cDNA clone IMAGE: 5726657).


010806pl3A5
BC041444

Homo sapiens cDNA FLJ27393 fis, clone





WMC01011.


310806pl2C9
BC042816

Homo sapiens full length insert cDNA YN57B01.



160507pl1C8
BC042855

Homo sapiens mRNA; cDNA DKFZp434A0326





(from clone DKFZp434A0326).


150506pl1D7
BC044257

Homo sapiens, clone IMAGE: 6063621, mRNA.



050707pl2D12
BC044741

Homo sapiens cDNA clone IMAGE: 4828106.



310506pl3D10
BC048320

Homo sapiens, clone IMAGE: 4450067, mRNA.



200306d9pl1C11
BC048993

Homo sapiens hypothetical protein LOC285550,





mRNA (cDNA clone IMAGE: 4686377), partial cds.


130207pl2A4
BC053955

Homo sapiens hypothetical protein LOC285548,





mRNA (cDNA clone IMAGE: 5265914).


160507pl3B5
BC054862

Homo sapiens cDNA clone IMAGE: 4288461,





partial cds.


160507pl1F5
BC078172

Homo sapiens cDNA clone IMAGE: 5760022,





partial cds.


041206pl2H4
BC082260

Homo sapiens cDNA clone IMAGE: 6427299, ****





WARNING: chimeric clone ****.


170407vpl3C9
BC108263

Homo sapiens transmembrane protein 56, mRNA





(cDNA clone IMAGE: 4801733), **** WARNING:




chimeric clone ****.


041206pl5E3
BCCIP
BRCA2 and CDKN1A-interacting protein isoform C


200906pl5C5
BE043072
ho32e06.x1 NCI_CGAP_Lu24 Homo sapiens




cDNA clone IMAGE: 3039106 3′, mRNA sequence.


010506pl2D10
BE044435
ho45d08.x1 Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone IMAGE: 3040335 3′, mRNA





sequence.


041206pl7D5
BE048560
hr50f01.x1 NCI_CGAP_Kid11 Homo sapiens




cDNA clone IMAGE: 3131929 3′ similar to contains




Alu repetitive element; contains element TAR1




repetitive element;, mRNA sequence.


310506pl1G10
BE048868
hr54h09.x1 NCI_CGAP_Kid11 Homo sapiens




cDNA clone IMAGE: 3132353 3′ similar to contains




MER13.t3 MER13 repetitive element;, mRNA




sequence.


050707pl2F4
BE257831
601109413F1 NIH_MGC_16 Homo sapiens cDNA




clone IMAGE: 3350114 5′, mRNA sequence.


160507pl3D7
BE466653
hz23g02.x1 NCI_CGAP_GC6 Homo sapiens




cDNA clone IMAGE: 3208850 3′, mRNA sequence.


201107pl4A4
BE504704
hz31c02.x1 NCI_CGAP_GC6 Homo sapiens cDNA




clone IMAGE: 3209570 3′ similar to TR: P97346




P97346 NUCLEOREDOXIN;, mRNA sequence.


041206pl6G1
BE505026
hz36h06.x1 NCI_CGAP_GC6 Homo sapiens




cDNA clone IMAGE: 3210107 3′, mRNA sequence.


010806pl2A2
BE785612
601475144F1 NIH_MGC_68 Homo sapiens cDNA




clone IMAGE: 3878051 5′, mRNA sequence.


311007pl2C3
BF001694
7g91h05.x1 NCI_CGAP_Co16 Homo sapiens




cDNA clone IMAGE: 3313881 3′ similar to




TR: O60705 O60705 LIM PROTEIN.;, mRNA




sequence.


160507pl2D11
BF062994
7h73f05.x1 NCI_CGAP_Co16 Homo sapiens




cDNA clone IMAGE: 3321633 3′, mRNA sequence.


310506pl1E3
BF244436
601862730F1 NIH_MGC_57 Homo sapiens cDNA




clone IMAGE: 4080511 5′, mRNA sequence.


190607pl1C5
BF245041
601864168F1 NIH_MGC_57 Homo sapiens cDNA




clone IMAGE: 4082368 5′, mRNA sequence.


041206pl3C4
BF434856
7o74e08.x1 NCI_CGAP_Kid11 Homo sapiens




cDNA clone IMAGE: 3641967 3′, mRNA sequence.


150506pl1B11
BF509736
UI-H-BI4-apg-b-02-0-UI.s1 NCI_CGAP_Sub8





Homo sapiens cDNA clone IMAGE: 3087290 3′,





mRNA sequence.


200906pl2B2
BF594738
7o54h12.x1 NCI_CGAP_Kid11 Homo sapiens




cDNA clone IMAGE: 3577991 3′, mRNA sequence.


041206pl6A1
BF688062
602067272F1 NIH_MGC_57 Homo sapiens cDNA




clone IMAGE: 4066433 5′, mRNA sequence.


200906pl5B9
BF875734
QV3-ET0103-111100-386-a04 ET0103 Homo





sapiens cDNA, mRNA sequence.



311007pl3G12
BG189068
RST8104 Athersys RAGE Library Homo sapiens




cDNA, mRNA sequence.


041206pl3G11
BG201613
RST20954 Athersys RAGE Library Homo sapiens




cDNA, mRNA sequence.


160507pl2C7
BG203790
RST23181 Athersys RAGE Library Homo sapiens




cDNA, mRNA sequence.


201107pl3F4
BG221753
RST41568 Athersys RAGE Library Homo sapiens




cDNA, mRNA sequence.


310506pl3H3
BG426583
602493305F1 NIH_MGC_75 Homo sapiens cDNA




clone IMAGE: 4607305 5′, mRNA sequence.


311007pl3D2
BG505700
602549869F1 NIH_MGC_61 Homo sapiens cDNA




clone IMAGE: 4657624 5′, mRNA sequence.


050707pl1G10
BG716117
602677572F1 NIH_MGC_96 Homo sapiens cDNA




clone IMAGE: 4800233 5′, mRNA sequence.


310506pl2A1
BG753571
602733141F1 NIH_MGC_43 Homo sapiens cDNA




clone IMAGE: 4876330 5′, mRNA sequence.


170407pl1D3
BI462136
603205131F1 NIH_MGC_97 Homo sapiens cDNA




clone IMAGE: 5270983 5′, mRNA sequence.


150506pl1F3
BI559775
603252664F1 NIH_MGC_97 Homo sapiens cDNA




clone IMAGE: 5295231 5′, mRNA sequence.


050707pl3H8
BI762388
603049060F1 NIH_MGC_116 Homo sapiens




cDNA clone IMAGE: 5189054 5′, mRNA sequence.


311007pl3F3
BI825982
603076566F1 NIH_MGC_119 Homo sapiens




cDNA clone IMAGE: 5168225 5′, mRNA sequence.


150506pl2D3
BI838110
603083607F1 NIH_MGC_120 Homo sapiens




cDNA clone IMAGE: 5222953 5′, mRNA sequence.


130207pl2C2
BIN1
bridging integrator 1 isoform 1


010506pl1C3
BIN2
bridging integrator 2


200906pl1D2
BM461531
AGENCOURT_6421147 NIH_MGC_67 Homo





sapiens cDNA clone IMAGE: 5501266 5′, mRNA





sequence.


200906pl1E11
BM681834
UI-E-EJ0-aiq-g-07-0-UI.s1 UI-E-EJ0 Homo sapiens




cDNA clone UI-E-EJ0-aiq-g-07-0-UI 3′, mRNA




sequence.


010806pl2G8
BM684766
UI-E-EJ1-ajj-m-22-0-UI.s1 UI-E-EJ1 Homo sapiens




cDNA clone UI-E-EJ1-ajj-m-22-0-UI 3′, mRNA




sequence.


041206pl3D6
BM690995
UI-E-CI1-aba-d-08-0-UI.r1 UI-E-CI1 Homo sapiens




cDNA clone UI-E-CI1-aba-d-08-0-UI 5′, mRNA




sequence.


200906pl1D10
BM691000
UI-E-CI1-aba-e-01-0-UI.r1 UI-E-CI1 Homo sapiens




cDNA clone UI-E-CI1-aba-e-01-0-UI 5′, mRNA




sequence.


310806pl2B3
BM749023
K-EST0024086 S10SNU1 Homo sapiens cDNA




clone S10SNU1-1-F09 5′, mRNA sequence.


041206pl2D7
BM905834
AGENCOURT_6721121 NIH_MGC_71 Homo





sapiens cDNA clone IMAGE: 5556193 5′, mRNA





sequence.


170407vpl3B5
BOLA2
BolA-like protein 2 isoform b


200906pl5F8
bpl 41-16

Homo sapiens olfactory receptor, family 7,





subfamily E, member 47 pseudogene, mRNA




(cDNA clone IMAGE: 5590288).


200906pl4B10
BQ011346
UI-1-BC1p-arz-e-06-0-UI.s1 NCI_CGAP_PI3 Homo





sapiens cDNA clone UI-1-BC1p-arz-e-06-0-UI 3′,





mRNA sequence.


201107pl3E1
BQ183849
UI-H-EU0-azs-b-24-0-UI.s1 NCI_CGAP_Car1





Homo sapiens cDNA clone IMAGE: 5852855 3′,





mRNA sequence.


290307pl1A6
BQ184944
UI-E-EJ1-ajo-c-04-0-UI.s1 UI-E-EJ1 Homo sapiens




cDNA clone UI-E-EJ1-ajo-c-04-0-UI 3′, mRNA




sequence.


130207pl1D3
BQ230709
AGENCOURT_7546358 NIH_MGC_70 Homo





sapiens cDNA clone IMAGE: 6025005 5′, mRNA





sequence.


160507pl1D8
BQ233546
AGENCOURT_7526687 NIH_MGC_70 Homo





sapiens cDNA clone IMAGE: 6018551 5′, mRNA





sequence.


200208pl2B4
BRIP1
BRCA1 interacting protein C-terminal helicase 1


170407pl1E10
BRMS1
breast cancer metastasis suppressor 1 isoform 2


280705p1f13D3
BSG
basigin isoform 1


170407vpl3A9
BTK

Homo sapiens Bruton's tyrosine kinase mRNA,





complete cds.


311007pl3F2
BU533525
AGENCOURT_10197749 NIH_MGC_126 Homo





sapiens cDNA clone IMAGE: 6559929 5′, mRNA





sequence.


130207pl2C5
BU534173
AGENCOURT_10240114 NIH_MGC_126 Homo





sapiens cDNA clone IMAGE: 6561006 5′, mRNA





sequence.


010806pl2B5
BU568189
AGENCOURT_10404673 NIH_MGC_82 Homo





sapiens cDNA clone IMAGE: 6615135 5′, mRNA





sequence.


310806pl1F4
BU599750
AGENCOURT_8827710 NIH_MGC_142 Homo





sapiens cDNA clone IMAGE: 6458824 5′, mRNA





sequence.


050707pl2D5
BU607353
UI-CF-FN0-aeu-g-14-0-UI.s1 UI-CF-FN0 Homo





sapiens cDNA clone UI-CF-FN0-aeu-g-14-0-UI 3′,





mRNA sequence.


150506pl1G1
BU619815
UI-H-FH1-bfq-j-08-0-UI.s1 NCI_CGAP_FH1 Homo





sapiens cDNA clone UI-H-FH1-bfq-j-08-0-UI 3′,





mRNA sequence.


200906pl4F9
BU621210
UI-H-FL1-bfz-e-02-0-UI.s1 NCI_CGAP_FL1 Homo





sapiens cDNA clone UI-H-FL1-bfz-e-02-0-UI 3′,





mRNA sequence.


041206pl2A2
BU630466
UI-H-FL0-bdk-a-10-0-UI.s1 NCI_CGAP_FL0 Homo





sapiens cDNA clone UI-H-FL0-bdk-a-10-0-UI 3′,





mRNA sequence.


310506pl1G6
BU753850
UI-1-BC1p-alh-b-11-0-UI.s1 NCI_CGAP_PI3 Homo





sapiens cDNA clone UI-1-BC1p-alh-b-11-0-UI 3′,





mRNA sequence.


041206pl6G3
BU930695
AGENCOURT_10425457 NIH_MGC_83 Homo





sapiens cDNA clone IMAGE: 6668795 5′, mRNA





sequence.


010806pl4B8
BX090666
BX090666 Soares_testis_NHT Homo sapiens




cDNA clone IMAGp998D014412; IMAGE: 1736400




5′, mRNA sequence.


041206pl4F4
BX096972
BX096972 Soares fetal liver spleen 1NFLS Homo





sapiens cDNA clone IMAGp998A01130;





IMAGE: 127368 5′, mRNA sequence.


290307pl1D1
BX100329
BX100329 Soares_NFL_T_GBC_S1 Homo





sapiens cDNA clone IMAGp998H043806;





IMAGE: 1503795 5′, mRNA sequence.


050707pl2D8
BX100818
BX100818 Soares_fetal_lung_NbHL19W Homo





sapiens cDNA clone IMAGp998J074430;





IMAGE: 1743462 5′, mRNA sequence.


180504p11c2
BX101084
hEST


311007pl3D7
BX103408
BX103408 Soares melanocyte 2NbHM Homo





sapiens cDNA clone IMAGp998L01545;





IMAGE: 251664 5′, mRNA sequence.


160507pl1E5
BX103636
BX103636 Soares_testis_NHT Homo sapiens




cDNA clone IMAGp998J184112; IMAGE: 1621361




5′, mRNA sequence.


200906pl2H6
BX104605
BX104605 Soares_testis_NHT Homo sapiens




cDNA clone IMAGp998B211795; IMAGE: 731444




5′, mRNA sequence.


130207pl2E11
BX108181
BX108181 Soares_testis_NHT Homo sapiens




cDNA clone IMAGp998A194412; IMAGE: 1736346




5′, mRNA sequence.


200906pl5B4
BX364993
BX364993 Homo sapiens PLACENTA COT 25-




NORMALIZED Homo sapiens cDNA clone




CS0DI038YA06 5-PRIME, mRNA sequence.


311007pl1D12
BX537644

Homo sapiens cDNA: FLJ23130 fis, clone





LNG08419.


010806pl4E8
BX537772

Homo sapiens mRNA; cDNA DKFZp781M2440





(from clone DKFZp781M2440).


201107pl1B3
BX538309

Homo sapiens mRNA; cDNA DKFZp686C09130





(from clone DKFZp686C09130).


201107pl2C1
BX648475

Homo sapiens mRNA; cDNA DKFZp686p11156





(from clone DKFZp686p11156).


130207pl2D4
BX648555

Homo sapiens mRNA; cDNA DKFZp779B0135





(from clone DKFZp779B0135).


150506pl2G3
BX648926

Homo sapiens mRNA; cDNA DKFZp686O0329





(from clone DKFZp686O0329).


310806pl1F9
BXDC1
brix domain containing 1


041206pl1F7
C10orf129

Homo sapiens cDNA FLJ44146 fis, clone





THYMU2027734, weakly similar to Homo sapiens




SA hypertension-associated homolog (rat) (SAH).


150506pl2F2
C12orf43
hypothetical protein LOC64897


311007pl2D5
C12orf45
hypothetical protein LOC121053


201107pl1B10
C14orf102
hypothetical protein LOC55051 isoform 1


160507pl2A3
C14orf112
hypothetical protein LOC51241


041206pl2A8
C14orf140
chromosome 14 open reading frame 140 isoform a


190607pl1A8
C14orf2
hypothetical protein LOC9556


310506pl1G11
C16orf14
hypothetical protein LOC84331


041206pl6G12
C17orf49
hypothetical protein LOC124944


311007pl2A6
C19orf33
HAI-2 related small protein


160507pl1A2
C19orf43
hypothetical protein MGC2803


200906pl2D8
C19orf61
hypothetical protein LOC56006


050707pl3D7
C1orf121
hypothetical protein LOC51029


180504p13e3
C1orf149
hypothetical protein LOC64769


310506pl1F5
C1orf62
hypothetical protein LOC254268


010806pl1H5
C1QBP
complement component 1, q subcomponent




binding


200906pl2E6
C20orf24
hEST


160507pl3H5
C20orf52
reactive oxygen species modulator 1


160507pl2B10
C21orf59

Homo sapiens T-complex protein 10A-2 mRNA,





complete cds.


041206pl1H7
C22orf16
chromosome 22 open reading frame 16


311007pl1C5
C2orf25
hypothetical protein LOC27249


201107pl4B1
C2orf27
hypothetical protein LOC29798


170407pl3F1
C2orf49
hypothetical protein LOC79074


010506pl1E8
C3orf19
hypothetical protein LOC51244


201107pl3B1
C3orf26
hypothetical protein LOC84319


201107pl2C3
C6orf106
chromosome 6 open reading frame 106 isoform a


310806pl1E10
C6orf51
hypothetical protein LOC112495


200208pl2B5
C6orf64
hypothetical protein LOC55776


201107pl3G8
C7orf11
chromosome 7 open reading frame 11


041206pl3H11
C7orf24

Homo sapiens cDNA FLJ11717 fis, clone





HEMBA1005241.


160507pl3A4
C7orf48
hypothetical protein LOC84262


190607pl1A2
C8orf44
hypothetical protein LOC56260


050707pl3H2
C8orf53
hypothetical protein LOC84294


041206pl6D9
C8orf59

Homo sapiens cDNA FLJ20407 fis, clone





KAT01658.


170407vpl3B12
C9orf30
hypothetical protein LOC91283


130207pl1E1
C9orf40
hypothetical protein LOC55071


200906pl5G7
CA418524
UI-H-EZ1-bbd-m-02-0-UI.s1 NCI_CGAP_Ch2





Homo sapiens cDNA clone UI-H-EZ1-bbd-m-02-0-





UI 3′, mRNA sequence.


050707pl2A3
CA430002
UI-H-FH1-bfp-h-24-0-UI.s1 NCI_CGAP_FH1 Homo





sapiens cDNA clone UI-H-FH1-bfp-h-24-0-UI 3′,





mRNA sequence.


200906pl5F2
CA444589
UI-H-DT1-awl-m-08-0-UI.s1 NCI_CGAP_DT1





Homo sapiens cDNA clone UI-H-DT1-awl-m-08-0-





UI 3′, mRNA sequence.


010806pl4G11
CA453297
AGENCOURT_10577997 NIH_MGC_127 Homo





sapiens cDNA clone IMAGE: 6717046 5′, mRNA





sequence.


200906pl3H12
CA943566
ir29h04.x1 HR85 islet Homo sapiens cDNA clone




IMAGE: 6546848 3′, mRNA sequence.


041206pl7D1
CACNA2D1
calcium channel, voltage-dependent, alpha


130207pl2A9
CACYBP
calcyclin binding protein isoform 2


201107pl1H8
CALCOCO2
calcium binding and coiled-coil domain 2


200306d9pl1E8
CALD1
NAG22 protein.


130207pl1A4
CALM1
calmodulin 1


310506pl3B1
CALM2
calmodulin 2


150506pl1E2
CALM3
calmodulin 2


200208pl2B12
CAPRIN1
membrane component chromosome 11 surface




marker


170407vpl3B10
CAPZA2

Homo sapiens mRNA for capping protein (actin





filament) muscle Z-line, alpha 2 variant, clone:




HSI05568.


041206pl7A11
CASP8AP2
CASP8 associated protein 2


010806pl1A3
CAST
calpastatin isoform a


170407pl1C2
CAV1
caveolin 1


150506pl2F10
CB045860
NISC_gf01a03.x1 NCI_CGAP_Kid12 Homo





sapiens cDNA clone IMAGE: 3252364 3′, mRNA





sequence.


200906pl1D12
CB046508
NISC_gf05a01.x1 NCI_CGAP_Kid12 Homo





sapiens cDNA clone IMAGE: 3252744 3′, mRNA





sequence.


310806pl2A3
CB049395
NISC_gj10f03.x1 NCI_CGAP_Pr28 Homo sapiens




cDNA clone IMAGE: 3271421 3′, mRNA sequence.


050707pl2A6
CB155900
K-EST0214495 L17N670205n1 Homo sapiens




cDNA clone L17N670205n1-1-A03 5′, mRNA




sequence.


200906pl5B5
CB985912
AGENCOURT_13640469 NIH_MGC_184 Homo





sapiens cDNA clone IMAGE: 30328716 5′, mRNA





sequence.


041206pl1F3
CBWD2
COBW domain-containing protein 2


310806pl1C12
CBX5
chromobox homolog 5 (HP1 alpha homolog,


050707pl2D9
CCDC12
coiled-coil domain containing 12


310506pl2C3
CCDC23
coiled-coil domain containing 23


010506pl1D3
CCDC50
Ymer protein long isoform


010506pl2C10
CCDC72
coiled-coil domain containing 72


190607pl1G10
CCDC74A
coiled-coil domain containing 74A


041206pl3F4
CCDC84
coiled-coil domain containing 84


160507pl3F11
CCT5
chaperonin containing TCP1, subunit 5 (epsilon)


290307pl1F1
CCT6A
chaperonin containing TCP1, subunit 6A isoform


200208pl2F4
CCT7
chaperonin containing TCP1, subunit 7 isoform a


310506pl3H8
CCT8
CCT8 protein.


31104p47c11
CD164
CD164 antigen, sialomucin


041206pl3D11
CD44
CD44 antigen isoform 1 precursor


160507pl3D3
CD63
CD63 antigen isoform A


041206pl1C8
CD641745
AGENCOURT_14537497 NIH_MGC_191 Homo





sapiens cDNA clone IMAGE: 30416477 5′, mRNA





sequence.


050707pl1C3
CD692919
EST9442 human nasopharynx Homo sapiens




cDNA, mRNA sequence.


311007pl3H5
CD9
CD9 antigen


010806pl3D4
CDADC1
cytidine and dCMP deaminase domain containing 1


311007pl3D9
CDC37
Synthetic construct Homo sapiens mRNA for




hypothetical protein (CDC37 gene), clone




IMAGE: 3505011.1E3.


041206pl6F10
CDK3
cyclin-dependent kinase 3


050707pl3C12
CDKN3
cyclin-dependent kinase inhibitor 3


310506pl3A8
CECR4

Homo sapiens Cat eye syndrome critical region





candidate gene number 4 (CECR4) mRNA, partial




cds.


160507pl2A12
CENTB1
centaurin beta1


041206pl5B7
CFL2
cofilin 2


160507pl1D6
CFLAR
CASP8 and FADD-like apoptosis regulator


170604p17c4
CHCHD2
coiled-coil-helix-coiled-coil-helix domain


150506pl2F11
CHCHD6
coiled-coil-helix-coiled-coil-helix domain


041206pl6B6
CHCHD8
coiled-coil-helix-coiled-coil-helix domain


310506pl2E5
CHORDC1
cysteine and histidine-rich domain


041206pl1A9
CHURC1
churchill domain containing 1


311007pl3D3
CICK0721Q.1
hypothetical protein LOC729727


050707pl3A12
CIP29

Homo sapiens HSPC316 mRNA, partial cds.



280305p1f12d10
CIRBP
cold inducible RNA binding protein


201107pl3D4
CIRH1A
cirhin


010806pl2F10
CK126027
AGENCOURT_16510969 NIH_MGC_239 Homo





sapiens cDNA clone IMAGE: 30710070 5′, mRNA





sequence.


010806pl4A1
CKS2
CDC28 protein kinase 2


200306d9pl1D7
CLCN3
chloride channel 3 isoform e


050707pl2H5
CLEC2D
osteoclast inhibitory lectin isoform 1


10704p110c1
CLIC1
chloride intracellular channel 1


311007pl3A11
CLIC4
chloride intracellular channel 4


010806pl1B6
CLINT1
epsin 4


170407vpl3B2
CLPTM1
cleft lip and palate associated transmembrane


200208pl2F7
CLTC
clathrin heavy chain 1


310506pl3D11
CMTM3
chemokine-like factor superfamily 3


041206pl7A8
CN267986
17000531863184 GRN_EB Homo sapiens cDNA




5′, mRNA sequence.


200906pl5G6
CN277269
17000600176551 GRN_PREHEP Homo sapiens




cDNA 5′, mRNA sequence.


290307pl1D5
CN280387
17000455082974 GRN_ES Homo sapiens cDNA




5′, mRNA sequence.


041206pl2B2
CN290177
17000600005140 GRN_PRENEU Homo sapiens




cDNA 5′, mRNA sequence.


170407pl1E12
CN398253
17000424721764 GRN_EB Homo sapiens cDNA




5′, mRNA sequence.


010806pl3C12
CNN3
calponin 3


010806pl1F8
COPS6
COP9 signalosome subunit 6


050707pl1C8
COPZ1
coatomer protein complex, subunit zeta 1


041206pl3H8
COTL1
coactosin-like 1


311007pl2A1
COX17
COX17 homolog, cytochrome c oxidase assembly


160507pl1D1
COX4NB
neighbor of COX4


310506pl2A5
COX7C
cytochrome c oxidase subunit VIIc precursor


170407vpl3G10
COX8A
cytochrome c oxidase subunit 8A


041206pl6F11
CR593740

Homo sapiens cDNA clone IMAGE: 4823412.



200906pl1H3
CR599716

Homo sapiens Shwachman-Bodian-Diamond





syndrome pseudogene, mRNA (cDNA clone




IMAGE: 4329436).


050707pl3B3
CR604262
full-length cDNA clone CS0DC003YA14 of




Neuroblastoma Cot 25-normalized of Homo





sapiens (human).



130207pl2B12
CR604408

Homo sapiens, clone IMAGE: 5190399, mRNA.



200906pl2B3
CR623475

Homo sapiens cDNA: FLJ21942 fis, clone





HEP04527.


200306f7pl1A9
CR624523

Homo sapiens hypothetical gene , mRNA



041206pl6H12
CR625980
full-length cDNA clone CS0DC026YN07 of




Neuroblastoma Cot 25-normalized of Homo





sapiens (human).



010506pl2A12
CR626360
full-length cDNA clone CS0DM014YM20 of Fetal




liver of Homo sapiens (human).


160507pl1A9
CR627148

Homo sapiens, clone IMAGE: 5213378, mRNA.



160507pl1D7
CR737784
CR737784 Homo sapiens library (Ebert L) Homo





sapiens cDNA clone IMAGp998C154208;





IMAGE: 1658054 5′, mRNA sequence.


190607pl1B9
CR994463
CR994463 RZPD no. 9016 Homo sapiens cDNA




clone RZPDp9016A109 5′, mRNA sequence.


170407pl3E4
CRKL
v-crk sarcoma virus CT10 oncogene homolog


310505p4f1c4
CSDA
cold shock domain protein A


041206pl3B4
CSDE1
upstream of NRAS isoform 1


160507pl2F7
CSNK1A1
casein kinase 1, alpha 1 isoform 2


200208pl2D1
CXorf26

Homo sapiens HSPC245 mRNA, complete cds.



010806pl2E2
DA336829
DA336829 BRHIP3 Homo sapiens cDNA clone




BRHIP3037522 5′, mRNA sequence.


041206pl6A7
DA438551
DA438551 CTONG2 Homo sapiens cDNA clone




CTONG2006372 5′, mRNA sequence.


150506pl2A8
DA691808
DA691808 NT2NE2 Homo sapiens cDNA clone




NT2NE2011571 5′, mRNA sequence.


200906pl2F8
DA697821
DA697821 NT2NE2 Homo sapiens cDNA clone




NT2NE2019092 5′, mRNA sequence.


041206pl3H1g
DA963983
DA963983 STOMA2 Homo sapiens cDNA clone




STOMA2001983 5′, mRNA sequence.


010806pl2F11
DAP
death-associated protein


150506pl1B12
DAZAP2
DAZ associated protein 2


200306f7pl1C3
DB040854
DB040854 TESTI2 Homo sapiens cDNA clone




TESTI2027763 5′, mRNA sequence.


311007pl2C1
DB049861
DB049861 TESTI2 Homo sapiens cDNA clone




TESTI2039270 5′, mRNA sequence.


310806pl2E8
DB054822
DB054822 TESTI2 Homo sapiens cDNA clone




TESTI2045843 5′, mRNA sequence.


200906pl4C12
DB095008
DB095008 TESTI4 Homo sapiens cDNA clone




TESTI4045539 5′, mRNA sequence.


201107pl3E12
DB136282
DB136282 THYMU3 Homo sapiens cDNA clone




THYMU3007538 5′, mRNA sequence.


160507pl1B10
DB331110
DB331110 SKMUS2 Homo sapiens cDNA clone




SKMUS2008761 3′, mRNA sequence.


200906pl1G4
DB337826
DB337826 TESTI2 Homo sapiens cDNA clone




TESTI2027763 3′, mRNA sequence.


310506pl3F2
DB339365
hEST


050707pl2A9
DB344099
DB344099 THYMU2 Homo sapiens cDNA clone




THYMU2032116 3′, mRNA sequence.


041206pl7C8
DB478885
DB478885 RIKEN full-length enriched human




cDNA library, hippocampus Homo sapiens cDNA




clone H023080L11 5′, mRNA sequence.


190607pl1F10
DB499813
DB499813 RIKEN full-length enriched human




cDNA library, hypothalamus Homo sapiens cDNA




clone H033074L02 5′, mRNA sequence.


041206pl2A6
DB504537
DB504537 RIKEN full-length enriched human




cDNA library, hypothalamus Homo sapiens cDNA




clone H033091O18 5′, mRNA sequence.


160507pl3E2
DB514539
DB514539 RIKEN full-length enriched human




cDNA library, testis Homo sapiens cDNA clone




H013041M08 3′, mRNA sequence.


130207pl1H2
DB522524
DB522524 RIKEN full-length enriched human




cDNA library, testis Homo sapiens cDNA clone




H013076C14 3′, mRNA sequence.


200906pl1D3
DB566909
DB566909 RIKEN full-length enriched human




cDNA library, hypothalamus Homo sapiens cDNA




clone H033059N21 3′, mRNA sequence.


310806pl1H4
DB571782
DB571782 RIKEN full-length enriched human




cDNA library, hypothalamus Homo sapiens cDNA




clone H033077H09 3′, mRNA sequence.


310505p4f1c5
DBN1
drebrin 1 isoform a


200906pl1A9
DC347972
DC347972 CTONG3 Homo sapiens cDNA clone




CTONG3005404 5′, mRNA sequence.


190607pl1F8
DCBLD2
discoidin, CUB and LCCL domain containing 2


010806pl3A8
DCC
deleted in colorectal carcinoma


200306f7pl1G12
DDT
D-dopachrome tautomerase


311007pl1G6
DDX10
DEAD (Asp-Glu-Ala-Asp) box polypeptide 10


010806pl2C5
DDX18
DEAD (Asp-Glu-Ala-Asp) box polypeptide 18


311007pl1A12
DDX43
DEAD (Asp-Glu-Ala-Asp) box polypeptide 43


310505p7f1b3
DDX46
DEAD (Asp-Glu-Ala-Asp) box polypeptide 46


090505p3f12d6
DDX5
DEAD (Asp-Glu-Ala-Asp) box polypeptide 5


150506pl2F8
DEK
DEK oncogene


210206pl1C6
DHX15
DEAN (Asp-Glu-Ala-His) box polypeptide 15


200306f7pl1B10
DHX16
DEAN (Asp-Glu-Ala-His) box polypeptide 16


160507pl1B11
DKFZp434M1123

Homo sapiens NY-REN-50 antigen mRNA, partial





cds.


310506pl1C9
DKFZp451B1418

Homo sapiens HSPC308 mRNA, partial cds.



010806pl1H2
DKFZp686B0790

Homo sapiens clone alpha1 mRNA sequence.



010806pl1G2
DKFZp686N1150

Homo sapiens cDNA FLJ37790 fis, clone





BRHIP3000111.


160507pl1B4
DKKL1
dickkopf-like 1 (soggy) precursor


310506pl2C1
DLGAP1
discs large homolog-associated protein 1 isoform


041206pl6D1
DLGAP4
disks large-associated protein 4 isoform a


170407pl3F3
DMTF1
cyclin D binding myb-like transcription factor


041206pl7A2
DNAJA1
DnaJ (Hsp40) homolog, subfamily A, member 1


170604pl7c1
DNAJC7
DnaJ (Hsp40) homolog, subfamily C, member 7


050707pl1D3
DNAPTP6
hypothetical protein LOC26010


171104P31B6
DNMT1
DNA (cytosine-5-)-methyltransferase 1


311007pl2B12
DPH1
diptheria toxin resistance protein required for


041206pl6F8
DQ343132

Homo sapiens urothelial cancer associated 1





(UCA1) mRNA, complete sequence.


170407pl3D12
DQ578159
full-length cDNA clone CS0DA009YE19 of




Neuroblastoma of Homo sapiens (human).


130207pl1E12
DSTN
destrin isoform a


200906pl5F4
DY654337
ucsc5_1.5.1.L1.1.A06.R.1 NIH_MGC_331 Homo





sapiens cDNA clone ucsc5_1.5.1.L1.1.A06, mRNA





sequence.


041206pl5E4
DYNC1H1
dynein, cytoplasmic, heavy polypeptide 1


311007pl3F5
DYNLRB1
Roadblock-1


041206pl6E1
EAPP
E2F-associated phosphoprotein


200208pl2B1
ece-1d

Homo sapiens mRNA for endothelin-converting





enzyme-1c, complete cds.


010506pl2D4
ECM29
KIAA0368 protein


201107pl2D5
EEA1
early endosome antigen 1, 162 kD


311007pl1G11
EED
embryonic ectoderm development isoform a


050707pl2B5
EEF1A1
eukaryotic translation elongation factor 1 alpha


041206pl1A2
EEF1E1
eukaryotic translation elongation factor 1


041206pl3D5
EEF1G
eukaryotic translation elongation factor 1


190607pl1E7
EEF2
eukaryotic translation elongation factor 2


190607pl1F3
EF565105

Homo sapiens chromosome 16 isolate HA_003251





mRNA sequence.


041206pl3B8
EFHC1
EF-hand domain (C-terminal) containing 1


310505p4f1d1
EIF1AX
X-linked eukaryotic translation initiation


201107pl4B9
EIF2S2
eukaryotic translation initiation factor 2 beta


311007pl2C9
EIF2S3
eukaryotic translation initiation factor 2,


310806pl1H5
EIF3S10
eukaryotic translation initiation factor 3,


041206pl1C1
EIF3S12
eukaryotic translation initiation factor 3,


210206pl1C3
EIF4A1
eukaryotic translation initiation factor 4A


310506pl4B9
EIF4E2
eukaryotic translation initiation factor 4E


180504p21e4
EIF4EBP1
eukaryotic translation initiation factor 4E


050707pl1G11
EIF4G3
eukaryotic translation initiation factor 4


150506pl1C2
EIF4H
eukaryotic translation initiation factor 4H


150506pl1D4
EIF5B
eukaryotic translation initiation factor 5B


200906pl5E10
EMP3
epithelial membrane protein 3


150506pl2F1
ENO1
enolase 1


160507pl1A11
ENSA
endosulfine alpha isoform 5


050707pl3B8
ENY2
enhancer of yellow 2 homolog


010806pl4E2
EPRS
glutamyl-prolyl tRNA synthetase


280705p1f13C12
ERCC1
excision repair cross-complementing 1 isoform 1


170407pl1A1
ERH
enhancer of rudimentary homolog


050707pl1G7
ETFB
electron-transfer-flavoprotein, beta polypeptide


200906pl1B6
FABP5
fatty acid binding protein 5


130207pl1G3
FAM128A

Homo sapiens family with sequence similarity 128,





member A, mRNA (cDNA clone MGC: 8772




IMAGE: 3862861), complete cds.


200306d9pl1B9
FAM128B
hypothetical protein LOC80097


201107pl1C10
FAM18B2
hypothetical protein LOC201158


160507pl3E12
FAM36A
family with sequence similarity 36, member A


201107pl2H12
FAM44A
hypothetical protein LOC259282


201107pl4D5
FAM82B
hypothetical protein LOC51115


041206pl1A11
FAM86A
hypothetical protein LOC196483 isoform 1


200906pl1D8
FAU
ubiquitin-like protein fubi and ribosomal


27073i1
FBL
fibrillarin


310506pl2B1
FBXO9
F-box only protein 9 isoform 3


201107pl1E8
FC170787
1106908754941 BABEVPN-C-01-1-7KB Papio




anubis cDNA clone 1061041899735 5′ similar to H. sapiens




UQCC (UniProtKB/Swiss-Prot: Q9NVA1),




mRNA sequence.


210206pl1D3
FER1L3
myoferlin isoform a


190607pl1A3
FEZ2
zygin 2 isoform 2


190607pl1F1
FHL3
four and a half LIM domains 3


310506pl1E5
FIGN
fidgetin


310506pl2E4
FLAD1
flavin adenine dinucleotide synthetase isoform


010506pl2D7
FLJ10154
hypothetical protein LOC55082


311007pl2G6
FLJ10292
mago-nashi homolog 2


041206pl5H11
FLJ10986

Homo sapiens cDNA FLJ10986 fis, clone





PLACE1001869, weakly similar to L-




RIBULOKINASE (EC 2.7.1.16).


010506pl1A8
FLJ20105
hypothetical protein LOC54821 isoform a


010806pl1D11
FLJ20674
hypothetical protein LOC54621


050707pl3A4
FLJ21908
hypothetical protein LOC79657


041206pl6G11
FLJ31951
hypothetical protein LOC153830


050707pl1D1
FLJ32065

Homo sapiens cDNA FLJ32065 fis, clone





OCBBF1000086.


050707pl1E3
FLJ35776
hypothetical protein LOC649446


010704p19b8
FLNB
filamin B, beta (actin binding protein 278)


170407vpl2C6
FNBP1
formin binding protein 1


130207pl1F5
FOSL1
FOS-like antigen 1


010506pl1C10
FSCN1
fascin 1


010806pl4E4
FUBP1
far upstream element-binding protein


180504p1ab2
FUS
fusion (involved in t(12; 16) in malignant


200906pl5F9
FXR1
fragile X mental retardation-related protein 1


041206pl5C4
FXYD5
FXYD domain-containing ion transport regulator


310806pl1C6
FYTTD1
forty-two-three domain containing 1 isoform 1


041206pl4H8
G36884
SHGC-56440 Human Homo sapiens STS cDNA,




sequence tagged site.


010806pl2B6
GABARAP
GABA(A) receptor-associated protein


160507pl2B2
GAGE2
G antigen 2


130207pl2D12
GAGE4
G antigen 4


170407vpl2D8
GALNT2
polypeptide N-acetylgalactosaminyltransferase 2


311007pl1E7
GAP43
growth associated protein 43


010806pl2G3
GAPDH
glyceraldehyde-3-phosphate dehydrogenase


130207pl1C6
GARS
glycyl-tRNA synthetase


150506pl1A4
GCHFR
GTP cyclohydrolase I feedback regulatory


311007pl1F11
GCNT2
glucosaminyl (N-acetyl) transferase 2,


160507pl3H2
GKN1
18 kDa antrum mucosa protein


201107pl2G2
GLO1
glyoxalase I


311007pl1C9
GLRX
glutaredoxin (thioltransferase)


150506pl1D2
GNB2L1
guanine nucleotide binding protein (G protein),


010806pl2F9
GNG11
guanine nucleotide binding protein gamma 11


201107pl1B5
GNG7
guanine nucleotide binding protein (G protein),


200906pl5F3
GPR113
G-protein coupled receptor 113


010806pl2E7
GRPEL1
GrpE-like 1, mitochondrial


201107pl1B7
GRSF1
G-rich RNA sequence binding factor 1


280305p5f2E4
GSPT1
G1 to S phase transition 1


280305p1f12D4
GTF2F2
general transcription factor IIF, polypeptide 2


130207pl2C3
H2AFV
H2A histone family, member V isoform 2


311007pl1C10
HABP4
hyaluronan binding protein 4


050707pl3F9
HAT1
histone acetyltransferase 1 isoform a


041206pl5H2
HCST
hematopoietic cell signal transducer isoform 1


041206pl1E4
HDAC2
histone deacetylase 2


200208pl2C5
HGD
homogentisate 1,2-dioxygenase


310506pl2B8
HHLA3
HERV-H LTR-associating 3 isoform 2


200906pl2C2
HIST1H2BH
H2B histone family, member J


010806pl2B2
HMG2L1
high-mobility group protein 2-like 1 isoform b


031104p47c9
HMGA1
high mobility group AT-hook 1 isoform a


27073c11
HMGA2
high mobility group AT-hook 2 isoform a


150506pl1A11
HMGN2
high-mobility group nucleosomal binding domain


311007pl3E9
HMGN3
high mobility group nucleosomal binding domain 3


290307pl1E4
HMMR
hyaluronan-mediated motility receptor isoform a


310506pl1F8
HN1
hematological and neurological expressed 1


190607pl1E2
HNRPA1
heterogeneous nuclear ribonucleoprotein A1


201107pl2F6
HNRPA2B1
heterogeneous nuclear ribonucleoprotein A2/B1


210206pl1E2
HNRPA3
heterogeneous nuclear ribonucleoprotein A3


050707pl1G6
HNRPAB
heterogeneous nuclear ribonucleoprotein AB


310506pl3H12
HNRPC
heterogeneous nuclear ribonucleoprotein C


210206pl1D2
HNRPD
heterogeneous nuclear ribonucleoprotein D


210206pl1G8
HNRPM
heterogeneous nuclear ribonucleoprotein M


311007pl3E5
HSP90AA1
heat shock protein 90 kDa alpha (cytosolic),


050707pl3D4
HSP90AB1
heat shock 90 kDa protein 1, beta


310506pl2C10
HSPB1
heat shock 27 kDa protein 1


310506pl1B9
HSPCA
heat shock protein 90 kDa alpha (cytosolic),


201107pl2D3
HSPH1
heat shock 105 kD


160507pl3G7
HYPA
Hypothetical protein (Fragment).


311007pl1A1
HYPK
Huntingtin interacting protein K


200906pl3E9
IFNGR2
interferon-gamma receptor beta chain precursor


311007pl3B11
IFT20
intraflagellar transport protein IFT20


310506pl3G10
IKIP
IKK interacting protein isoform 2


010506pl2A4
IL3RA
interleukin 3 receptor, alpha precursor


010806pl2F6
ILF2
interleukin enhancer binding factor 2


311007pl1C11
INPP4B
inositol polyphosphate-4-phosphatase, type II,


130207pl1B8
IQCK
IQ motif containing K


200208pl2C11
IRAK2
interleukin-1 receptor-associated kinase 2


311007pl1B3
ISOC1
isochorismatase domain containing 1


041206pl6B11
ITIH5
inter-alpha trypsin inhibitor heavy chain


041206pl2H6
JAGN1
jagunal homolog 1


200906pl3G10
KATNA1
katanin p60 subunit A 1


310806pl1D6
KBTBD2
kelch repeat and BTB (POZ) domain containing 2


160507pl2E5
KIAA0355
hypothetical protein LOC9710


210206pl1G5
KIAA0802
hypothetical protein LOC23255


200906pl2A2
KIAA1064

Homo sapiens mRNA for KIAA1064 protein, partial





cds.


010806pl2D1
KIAA1186

Homo sapiens mRNA for KIAA1186 protein, partial





cds.


200208pl2E11
KIAA1303
raptor


041206pl1H2
KIAA1430
KIAA1430 protein (Fragment).


130207pl2C1
KIAA1783

Homo sapiens mRNA for KIAA1783 protein, partial





cds.


311007pl1G2
KIAA1949
Protein KIAA1949.


010806pl4E11
KLHDC8A
kelch domain containing 8A


170407pl1E5
KLHL31
kelch repeat and BTB (POZ) domain containing 1


201107pl2H7
KPNA1
karyopherin alpha 1


200906pl2H3
KRT18
keratin 18


190607pl1C12
KRT8
keratin 8


010506pl1E9
Kua-UEV
ubiquitin-conjugating enzyme E2 Kua-UEV isoform


170407pl1D4
LAP3
leucine aminopeptidase 3


010806pl2C12
LARP1
la related protein isoform 2


290307pl1E10
LARP4
c-Mpl binding protein isoform a


10704p19b7
LASP1
LIM and SH3 protein 1


200208pl2G6
LDHA
lactate dehydrogenase A


200306f7pl1E6
LETM2
leucine zipper-EF-hand containing transmembrane


010306d9pl1C2
LGALS1
beta-galactoside-binding lectin precursor


010806pl4F6
LGALS3
galectin 3


311007pl2F8
LHB
luteinizing hormone beta subunit precursor


170407vpl3C6
LIMA1
epithelial protein lost in neoplasm beta


041206pl6E7
LIN7B
lin-7 homolog B


27073d13
LMNA
lamin A/C isoform 1 precursor


310131d13
LMNB1
lamin B1


010506pl2C12
LOC130074
hypothetical protein LOC130074


310806pl3B11
LOC134145
hypothetical protein LOC134145


311007pl1G12
LOC283551
hypothetical protein LOC283551


311007pl2G4
LOC284184

Homo sapiens full length insert cDNA clone





ZD54C08.


190607pl1E6
LOC286016

Homo sapiens cDNA FLJ37575 fis, clone





BRCOC2003125, moderately similar to




TRIOSEPHOSPHATE ISOMERASE (EC 5.3.1.1).


200906pl2G9
LOC389072
hypothetical protein LOC389072


050707pl2C4
LOC441161
hypothetical LOC441161


310506pl1D7
LOC541471

Homo sapiens hypothetical LOC541471, mRNA





(cDNA clone MGC: 17532 IMAGE: 3459303),




complete cds.


050707pl3H6
LOC728776
hypothetical protein LOC728776


201107pl2D11
LOC729416
hypothetical protein LOC729416


311007pl2D11
LOC751071
hypothetical protein LOC751071


200306d9pl1B4
LONRF3
LON peptidase N-terminal domain and ring finger


311007pl3C8
LOXL2
lysyl oxidase-like 2 precursor


170407pl1B6
LPIN2
lipin 2


150506pl1H3
LRRC50
leucine rich repeat containing 50


311007pl2C6
LRRC59
leucine rich repeat containing 59


010806pl1G1
LRRFIP1
LRR FLI-I interacting protein 1 (Fragment).


050707pl1D10
LSM3
Lsm3 protein


041206pl2B1
LUC7L2
LUC7-like 2


041206pl6H8
LYAR
hypothetical protein FLJ20425


200306f7pl1A10
MAP2K2
mitogen-activated protein kinase kinase 2


280305p1f12C11
MAP4
microtubule-associated protein 4 isoform 1


200906pl4A2
MAPBPIP
mitogen-activated protein-binding


010604p16b2
MAPK1
mitogen-activated protein kinase 1


180504p2ab3
MAPRE2
microtubule-associated protein, RP/EB family,


130207pl1B1
MBNL2
muscleblind-like 2 isoform 1


200906pl1G2
MCEE
methylmalonyl-CoA epimerase


170407vpl2C2
MDH1
cytosolic malate dehydrogenase


160507pl2H9
ME3
malic enzyme 3, NADP(+)-dependent,


150506pl2C12
MEGF6
EGF-like-domain, multiple 3


010506pl2E1
METAP2
methionyl aminopeptidase 2


170407vpl2B2
MGC11257
hypothetical protein LOC84310


160507pl3C9
MGC16824
hypothetical protein LOC57020


041206pl2F1
MGC59937
hypothetical protein LOC375791


150506pl1D10
mimitin

Homo sapiens mimitin mRNA for Myc-induced





mitochondria protein, complete cds.


170407vpl2D2
MKI67IP
MKI67 (FHA domain) interacting nucleolar


010506pl1F4
MKRN2
makorin, ring finger protein, 2


311007pl1D5
MLLT4
myeloid/lymphoid or mixed-lineage leukemia


041206pl4E11
MMAA

Homo sapiens cDNA FLJ44706 fis, clone





BRACE3017253, weakly similar to LAO/AO




transport system kinase (EC 2.7.—.—).


050707pl2H3
MRCL3
myosin regulatory light chain MRCL3


050707pl1D12
MRLC2
myosin regulatory light chain MRCL2


310806pl2D10
MRPL37
mitochondrial ribosomal protein L37


311007pl1G9
MRPS18B
mitochondrial ribosomal protein S18B


130207pl1G10
MRTO4
ribosomal protein P0-like protein


310806pl1D11
MSH6
mutS homolog 6


27073k9
MSN
moesin


150506pl1D5
MSRA
methionine sulfoxide reductase A


010704p110d1
MT2A
metallothionein 2A


190607pl1A5
MTDH
LYRIC/3D3


311007pl1H5
MTPN
myotrophin


041206pl3C7ag
MTX1
metaxin 1 isoform 1


041206pl2H7
MYEOV
myeloma overexpressed


010506pl1B12
MYH9
myosin, heavy polypeptide 9, non-muscle


310506pl1H5
MYLE
dexamethasone-induced protein


200208pl2C3
MYO1D
myosin ID


200208pl2A2
MYO1E
myosin IE


200906pl3F8
N39715
yx92d05.r1 Soares melanocyte 2NbHM Homo





sapiens cDNA clone IMAGE: 269193 5′ similar to





contains element TAR1 repetitive element;, mRNA




sequence.


201107pl2A3
N68399
za13b04.s1 Soares fetal liver spleen 1NFLS Homo





sapiens cDNA clone IMAGE: 292399 3′ similar to





SW: OLF3_MOUSE P23275 OLFACTORY




RECEPTOR OR3. [1];, mRNA sequence.


200306f7pl1C7
NACA
nascent-polypeptide-associated complex alpha


010806pl1G12
NANOS3
NANOS3 protein.


010704p110d2
NASP
nuclear autoantigenic sperm protein isoform 2


210206pl1C12
NAT13
Mak3 homolog


010806pl4F4
NBEAL1
Neurobeachin-like 1 (Amyotrophic lateral sclerosis




2 chromosomal region candidate gene 17 protein).


050707pl2G10
NCBP2
nuclear cap binding protein subunit 2, 20 kDa


160507pl3B1
NCL
nucleolin


150506pl1F11
NDUFA12L
Myc-induced mitochondria protein


010806pl1A10
NDUFA7
NADH dehydrogenase (ubiquinone) 1 alpha


041206pl5H6
NDUFB1
NADH dehydrogenase (ubiquinone) 1 beta


050707pl1B10
NDUFB11
NADH dehydrogenase (ubiquinone) 1 beta


190607pl1D5
NDUFB7
NADH dehydrogenase (ubiquinone) 1 beta


200306d9pl1C8
NDUFB8
NADH dehydrogenase (ubiquinone) 1 beta


170407vpl2B5
NDUFC1
NADH dehydrogenase (ubiquinone) 1, subcomplex


041206pl6F9
NEDD4L
neural precursor cell expressed, developmentally


010806pl2G6
NEXN
Nexilin.


010806pl1D1
NFE2L2
nuclear factor (erythroid-derived 2)-like 2


200906pl5B12
NGRN
mesenchymal stem cell protein DSC92 isoform 2


010604p16c10b
NHP2L1
NHP2 non-histone chromosome protein 2-like 1


200906pl5C2
NM_001039753
CDNA FLJ16635 fis, clone TESTI4025268, weakly




similar to 77 kDa echinoderm microtubule-




associated protein.


050707pl3G6
NM_001089591

Homo sapiens hCG25371 (LOC440567), mRNA.



200906pl2H4
NM_001093732

Homo sapiens hCG2033311 (LOC644928),





mRNA.


050707pl1C11
NM_001097611

Homo sapiens kinocilin (KNCN), mRNA.



311007pl2A8
NM_015681

Homo sapiens B9 protein domain 1 (B9D1),





mRNA.


200306f7pl1F8
NME1-NME2
NME1-NME2 protein


311007pl1H6
NME4
nucleoside-diphosphate kinase 4


200306f7pl1A7
NMT1
N-myristoyltransferase 1


180504p2ab6
NOL1
nucleolar protein 1, 120 kDa


200906pl3H11
NOL7
nucleolar protein 7, 27 kDa


200906pl3C7
NPAT
nuclear protein, ataxia-telangiectasia locus


160507pl1A3
NPEPPS
aminopeptidase puromycin sensitive


200906pl2B11
NPHP3
nephronophthisis 3


010506pl1A7
NPM1
nucleophosmin 1 isoform 1


010506pl2A1
NQO1
NAD(P)H menadione oxidoreductase 1,


311007pl1B12
NSMCE4A
non-SMC element 4 homolog A


310506pl1E9
NT_006576.400
Predicted Gene


310506pl1E8
NT_007592.828
Predicted Gene


310506pl1A6
NT_030059.345
genescan prediction


200906pl1C11
nt_032977.1313
Predicted Gene


200906pl2E7
NT_033899.591
Predicted Gene


170407pl3F4
NTAN1
N-terminal Asn amidase


200906pl2F1
NUCKS1
nuclear ubiquitous casein kinase and


201107pl3A10
NUDC
nuclear distribution gene C homolog


150506pl1F7
NUDCD1
NudC domain containing 1


160507pl1D4
NUDCD2
NudC domain containing 2


170407vpl2E11
NUDT3
nudix-type motif 3


050707pl1E10
NUP153
nucleoporin 153 kDa


310506pl3H5
NUP93
nucleoporin 93 kDa


201107pl3G7
OBTP

Homo sapiens over-expressed breast tumor





protein (OBTP) mRNA, complete cds.


170407pl1G1
OSBPL8
oxysterol-binding protein-like protein 8 isoform


170407pl3E2
OSBPL9
oxysterol-binding protein-like protein 9 isoform


041206pl2A7
OTUB1
otubain 1


180504p12d4
PA2G4
ErbB3-binding protein 1


200906pl1C6
PABPN1
poly(A) binding protein, nuclear 1


050707pl3F11
PAGE1
P antigen family, member 1


200906pl4E4
PAK2
p21-activated kinase 2


200208pl2G7
PARP4
poly (ADP-ribose) polymerase family, member 4


170407vpl2C9
PAWR
PRKC, apoptosis, WT1, regulator


041206pl3C8
PBX3
pre-B-cell leukemia transcription factor 3


311007pl3B8
PCBD1
pterin-4 alpha-carbinolamine dehydratase


150506pl1C9
PCBP2
poly(rC)-binding protein 2 isoform b


010506pl2D2
PCMTD2
protein-L-isoaspartate (D-aspartate)


180504p12d10
PDCD5
programmed cell death 5


150506pl1C11
PDIA5
protein disulfide isomerase-associated 5


010506pl1B6
PDIA6
protein disulfide isomerase-associated 6


010806pl1G9
PDZD2
PDZ domain containing 2


160507pl3G6
PFDN1

Homo sapiens mRNA for prefoldin 1 variant, clone:





FCC107D06.


190607pl1G1
PFDN2
prefoldin subunit 2


041206pl4H9
PFDN5
prefoldin subunit 5 isoform alpha


050707pl2E5
PFN1
profilin 1


010806pl4B6
PGK1
phosphoglycerate kinase 1


031104p37b7
PGRMC1
progesterone receptor membrane component 1


041206pl1C9
PHF20
PHD finger protein 20


310506pl3C12
PHLDB2
pleckstrin homology-like domain, family B,


290307pl1E1
PHPT1
phosphohistidine phosphatase 1


201107pl1C3
PIAS2


201107pl2H11
PIGY
phosphatidylinositol glycan anchor biosynthesis,


010806pl1C10
PKN1
protein kinase N1 isoform 2


171104p31b1
PLAA
phospholipase A2-activating protein isoform 1


010306d9pl1B10
PLEC1
plectin 1 isoform 6


130207pl1D4
PLS3
plastin 3


310806pl2D4
PNN
pinin, desmosome associated protein


310506pl3E5
POLR1D
polymerase (RNA) 1 polypeptide D isoform 1


200906pl4C4
POLR2F
DNA directed RNA polymerase II polypeptide F


200906pl1F10
POLR2G
DNA directed RNA polymerase II polypeptide G


041206pl6H10
POLR2L
DNA directed RNA polymerase II polypeptide L


010806pl1A1
POLR3GL
polymerase (RNA) III (DNA directed) polypeptide


160507pl3E8
POMP
proteasome maturation protein


310506pl2B12
POR
cytochrome P450 reductase


170604pl8b4
PPA1
pyrophosphatase 1


200906pl4F8
PPFIBP1
PTPRF interacting protein binding protein 1


310506pl4C1
PPIA
peptidylprolyl isomerase A


050707pl1F2
PPP1R10
protein phosphatase 1, regulatory subunit 10


170407vpl3A11
PPP1R14A
protein phosphatase 1, regulatory (inhibitor)


190607pl1H2
PPP1R14B
protein phosphatase 1 regulatory subunit 14B


010806pl1G5
PPP1R2
protein phosphatase 1, regulatory (inhibitor)


200208pl2H5
PPP2R2C
gamma isoform of regulatory subunit B55, protein


010506pl2B8
PRC1
protein regulator of cytokinesis 1 isoform 1


160507pl3C7
PRDX5
peroxiredoxin 5 precursor, isoform a


150506pl1F2
Predicted gene
NT_030059.67


190607pl1H6
PREPL
prolyl endopeptidase-like isoform C


010506pl1F3
PRKAR2A
cAMP-dependent protein kinase, regulatory


170407pl1B7
PROCR

Homo sapiens protein C receptor, endothelial





(EPCR), mRNA (cDNA clone MGC: 23024




IMAGE: 4907433), complete cds.


041206pl2A11
PRPF4B
serine/threonine-protein kinase PRP4K


201107pl4B8
PRR11
proline rich 11


200306f7pl1H4
PRR13
proline rich 13 isoform 2


010806pl4G1
PRRX1
paired mesoderm homeobox 1 isoform pmx-1a


041206pl5C9
PSIP1
PC4 and SFRS1 interacting protein 1 isoform 2


050707pl3D5
PSMA1
proteasome alpha 1 subunit isoform 2


041206pl2D8
PSMA2
proteasome alpha 2 subunit


310506pl1A3
PSMA3
proteasome alpha 3 subunit isoform 1


160507pl2F8
PSMA7
proteasome alpha 7 subunit


200906pl5H10
PSMB1
proteasome beta 1 subunit


130207pl2B4
PSMB4

Homo sapiens proteasome (prosome, macropain)





subunit, beta type, 4, mRNA (cDNA clone




MGC: 8522 IMAGE: 2822513), complete cds.


201107pl2D10
PSMB6
proteasome beta 6 subunit


200306f7pl1C11
PSMB7
proteasome beta 7 subunit proprotein


290307pl1C6
PSMC1
proteasome 26S ATPase subunit 1


170407vpl3B9
PSMC4
proteasome 26S ATPase subunit 4 isoform 1


200906pl5C4
PSMD1
proteasome 26S non-ATPase subunit 1


310505p4f1e2
PSMD11
proteasome 26S non-ATPase subunit 11


310806pl2A5
PSMD12
proteasome 26S non-ATPase subunit 12


010806pl4E6
PSMD6
proteasome (prosome, macropain) 26S subunit,


201107pl2G3
PSME1
proteasome activator subunit 1 isoform 1


311007pl1D2
PSMF1
proteasome inhibitor subunit 1 isoform 1


311007pl1G10
PSPC1
paraspeckle protein 1


280705plf13C2
PTBP1
polypyrimidine tract-binding protein 1 isoform


041206pl7A12
PTCRA
pre T-cell antigen receptor alpha


160507pl2E10
PTMA
prothymosin, alpha (gene sequence 28)


310806pl2B11
PTMS
parathymosin


170407vpl3B6
PTPLAD1
butyrate-induced transcript 1


200306d9pl1E11
PTTG1IP
pituitary tumor-transforming gene 1


201107pl2B5
PXK
PX domain containing serine/threonine kinase


200306f7pl1A4
PXN
paxillin


010506pl1B3
RAB11A
Ras-related protein Rab-11A


010704pl9b1
RAB1A
RAB1A, member RAS oncogene family


010806pl3B11
RAB31
RAB31, member RAS oncogene family


050707pl3A5
RAB33A
Ras-related protein Rab-33A


280705p1f13C3
RAC1
ras-related C3 botulinum toxin substrate 1


311007pl2F1
RANBP1
RAN binding protein 1


310506pl3D4
RASIP1
CDNA FLJ20401 fis, clone KAT00901 (RASIP1




protein).


160507pl1A12
RAVER1
RAVER1


031104p47c12
RBBP7
retinoblastoma binding protein 7


010806pl1D10
RBM12B
RNA binding motif protein 12B


150506pl2D10
RBM27
RNA-binding protein 27 (RNA-binding motif protein




27).


010806pl3A12
RBM41
RNA binding motif protein 41


200906pl1F3
RBM8A
RNA binding motif protein 8A


010806pl3E10
RBMXL1
RNA binding motif protein, X-linked-like 1


050707pl3H9
RBX1
ring-box 1


041206pl2B7
RCOR1
REST corepressor 1


050707pl1B12
RFC1
replication factor C large subunit


150506pl1F10
RFXDC2
regulatory factor X domain containing 2


010506pl2A6
RGS10
regulator of G-protein signaling 10 isoform b


201107pl2A10
RP11-255A11.5-
Ankyrin repeat domain 18B.



001


170604p17c9a
RP3-467K16.1
Novel protein (Fragment).


190607pl1H11
RPA2
replication protein A2, 32 kDa


310134b13
RPL11
ribosomal protein L11


200906pl4E5
RPL12
ribosomal protein L12


180504riboa2
RPL13A
ribosomal protein L13a


041206pl4D11
RPL14
ribosomal protein L14


150506pl1C8
RPL18
ribosomal protein L18


160507pl3E4
RPL22
ribosomal protein L22 proprotein


200306f7pl1E8
RPL23
ribosomal protein L23


010806pl4D8
RPL23A
ribosomal protein L23a


041206pl2H2
RPL24
ribosomal protein L24


010506pl1D7
RPL27A
ribosomal protein L27a


200906pl4C11
RPL29
ribosomal protein L29


041206pl2G5
RPL35
ribosomal protein L35


031104p37b1
RPL35A
ribosomal protein L35a


031104p47d1
RPL36
ribosomal protein L36


200906pl1F9
RPL36A
ribosomal protein L36a


180504riboa7
RPL4
ribosomal protein L4


010806pl3E8
RPL41
ribosomal protein L41


310134c18
RPL5
ribosomal protein L5


311007pl2A9
RPL6
ribosomal protein L6


180504riboa1
RPL7
ribosomal protein L7


180504p11c7
RPL7A
ribosomal protein L7a


311007pl3G10
RPL8

Homo sapiens ribosomal protein L8, mRNA (cDNA





clone IMAGE: 3504599), partial cds.


170407vpl2D6
RPLP0
ribosomal protein P0


010806pl2A11
RPLP1
hypothetical protein LOC729416


041206pl7B3
RPLP2
ribosomal protein P2


311007pl2E1
RPP40
ribonuclease P 40 kDa subunit


310505p4f1e1
RPS11
ribosomal protein S11


150506pl1B6
RPS12
ribosomal protein S12


050707pl3G8
RPS13
ribosomal protein S13


010806pl1B2
RPS15
hypothetical protein LOC401019


010806pl2E10
RPS15A
ribosomal protein S15a


160507pl1B5
RPS16
ribosomal protein S16


010506pl1A6
RPS17
ribosomal protein S17


160507pl1F6
RPS18
ribosomal protein S18


201107pl3H11
RPS19BP1
S19 binding protein


290307pl1D12
RPS20

Homo sapiens clone FLB0708 mRNA sequence.



310506pl2B5
RPS23
ribosomal protein S23


150506pl1C1
RPS24

Homo sapiens full length insert cDNA clone





YB24C12.


170407pl3D2
RPS25
ribosomal protein S25


041206pl2B8
RPS28
ribosomal protein S28


010506pl2B11
RPS3
ribosomal protein S3


310505p4f1c2
RPS3A
ribosomal protein S3a


280305p1f12C1
RPS4X
ribosomal protein S4, X-linked X isoform


310506pl1G12
RPS7
ribosomal protein S7


010806pl2A7
RRM1
ribonucleoside-diphosphate reductase M1 chain


130207pl1E4
RRP15
ribosomal RNA processing 15 homolog


280705p1f13D4
RSL1D1
ribosomal L1 domain containing 1


010806pl2G2
RSRC2
arginine/serine-rich coiled-coil 2 isoform b


180504p12d12
RTN4
reticulon 4 isoform A


010806pl1H1
RY1
putative nucleic acid binding protein RY-1


041206pl1F11
S100A10
S100 calcium binding protein A10


010806pl3E7
S100A11
S100 calcium binding protein A11


150506pl1A1
S100A2
S100 calcium binding protein A2


280305p6f2B2
SAE1
SUMO-1 activating enzyme subunit 1


280705p1f13C10
SAFB
scaffold attachment factor B


311007pl1B2
SCAMP2
secretory carrier membrane protein 2


201107pl3D10
SEC13
SEC13 protein


201107pl2G11
SEC14L1
SEC14 (S. cerevisiae)-like 1 isoform a


041206pl1A1
SELM
selenoprotein M precursor


200906pl2D11
SERBP1
SERPINE1 mRNA binding protein 1 isoform 1


041206pl3E11
SERF2
small EDRK-rich factor 2


010806pl4H2
SERPINB6
MSTP057.


010306d9pl1B5
SESN1
sestrin 1


280305plf12D1
SET
SET translocation (myeloid leukemia-associated)


130207pl1B10
SETMAR
SET domain and mariner transposase fusion


170407pl1E2
SF3B1
splicing factor 3b, subunit 1 isoform 1


160507pl2C11
SF3B14
splicing factor 3B, 14 kDa subunit


310131f6b
SFRS10
splicing factor, arginine/serine-rich 10


200906pl4D3
SFRS7
splicing factor, arginine/serine-rich 7


041206pl1C5
SH3GLB1
SH3-containing protein SH3GLB1


310506pl3A11
SH3KBP1
SH3-domain kinase binding protein 1 isoform b


010806pl1F5
SHFM1
candidate for split hand/foot malformation type


160507pl1F9
SIVA1
CDNA FLJ46871 fis, clone UTERU3012999, highly




similar to Homo sapiens CD27-binding (Siva)




protein (SIVA).


310505p4f1f7
SKIV2L2
superkiller viralicidic activity 2-like 2


010506pl2E6
SLBP
histone stem-loop binding protein


170407pl1G5
SLC20A2
solute carrier family 20, member 2


050707pl2C2
SLC22A18AS
solute carrier family 22 (organic cation


010806pl2D3
SLC24A3
solute carrier family 24


050707pl2D3
SLC25A37
mitochondrial solute carrier protein


160507pl3B7
SLC25A5
solute carrier family 25, member 5


190607pl1E11
SLC2A3
solute carrier family 2 (facilitated glucose


180504p1ab11
SLC3A2
solute carrier family 3 (activators of dibasic


200906pl4A11
SLC4A7
solute carrier family 4, sodium bicarbonate


010806pl2C11
SLC6A7
solute carrier family 6, member 7


160507pl2E12
SLC9A3R1
solute carrier family 9 (sodium/hydrogen


050707pl1A10
SLTM
modulator of estrogen induced transcription


310806pl2E6
SMS
spermine synthase


090505p3f12d3
SNRPB
small nuclear ribonucleoprotein polypeptide B/B′


010506pl1D5
SNRPD1
small nuclear ribonucleoprotein D1 polypeptide


290307pl1B7
SNRPF
small nuclear ribonucleoprotein polypeptide F


201107pl2B11
SNX3
sorting nexin 3


200906pl4F3
SNX6
sorting nexin 6 isoform b


170407vpl3B11
SOD1
superoxide dismutase 1, soluble


200906pl3H7
SON
SON DNA-binding protein isoform F


201107pl1C5
SORCS3
VPS10 domain receptor protein SORCS 3


180504p1ab4
SPAG4
sperm associated antigen 4


311007pl3A9
SPATA12
spermatogenesis associated 12


150506pl1F1
SPATS2
spermatogenesis associated, serine-rich 2


050707pl2B12
SPCS2
signal peptidase complex subunit 2 homolog


170407pl1F11
SPG20
spartin


010806pl4E3
SPTBN1
spectrin, beta, non-erythrocytic 1 isoform 1


310806pl1H2
SPTY2D1
SPT2, Suppressor of Ty, domain containing 1


041206pl2A5
SR140
U2-associated SR140 protein


170407pl1D8
SRCAP
Snf2-related CBP activator protein


200306f7pl1A12
SRM
spermidine synthase


130207pl2A6
SRP14
signal recognition particle 14 kDa (homologous


170604p18b1
SRP19
signal recognition particle 19 kDa


010806pl4D2
SRPK1
SFRS protein kinase 1


170407pl1C6
SRRM1
serine/arginine repetitive matrix 1


200306d9pl1C7
SRRM2
splicing coactivator subunit SRm300


311007pl3B10
SSBP1
single-stranded DNA binding protein 1


310506pl1A12
STAG1 variant
stromal antigen 1



protein


201107pl1E6
STAMBP
STAM binding protein


050707pl3H10
STAU1
staufen isoform a


160507pl1F4
STK4
serine/threonine kinase 4


010806pl4F12
STMN1
stathmin 1


200208pl2D12
STXBP5L
Syntaxin-binding protein 5-like (Tomosyn-2)




(Lethal(2) giant larvae protein homolog 4).


027073l5
SUMO1
SMT3 suppressor of mif two 3 homolog 1 isoform a


160507pl1E9
SUMO2
SMT3 suppressor of mif two 3 homolog 2 isoform a


311007pl2A4
SYNCRIP
synaptotagmin binding, cytoplasmic RNA


050707pl2G3
T85821
yd57b09.r1 Soares fetal liver spleen 1NFLS Homo





sapiens cDNA clone IMAGE: 112313 5′ similar to





contains MER25 repetitive element;, mRNA




sequence.


170407pl1C1
TALDO1
transaldolase 1


290307pl1H5
TARS
threonyl-tRNA synthetase


010806pl3E2
TBCA
tubulin-specific chaperone a


200906pl3H2
TBCB
cytoskeleton associated protein 1


200208pl2D5
TCEA3
transcription elongation factor A (SII), 3


170407pl1A7
TCF25
NULP1


010506pl2B12
TCP1
T-complex protein 1 isoform a


310806pl2B5
TDG
thymine-DNA glycosylase


310505p4f1b4
TENC1
tensin like C1 domain containing phosphatase


201107pl2C6
TES
testin isoform 1


010506pl1A11
TFAM
transcription factor A, mitochondrial


310506pl1C6
TFPT
TCF3 (E2A) fusion partner (in childhood


170407vpl2B10
THAP7
THAP domain containing 7 isoform b


050707pl1D6
THOC4
THO complex 4


041206pl3C6
TIMP2
tissue inhibitor of metalloproteinase 2


050707pl1C9
TJP1
tight junction protein 1 isoform b


200906pl1D1
TLCD1
TLC domain containing 1


050707pl3D12
TLN2
talin 2


201107pl2C9
TLOC1
translocation protein 1


010806pl3C7
TMCO3
transmembrane and coiled-coil domains 3


050707pl3G11
TMEM11
transmembrane protein 11


310505p4f1d6
TMEM123
pro-oncosis receptor inducing membrane injury


201107pl3E8
TMEM132D
hypothetical protein LOC121256


010806pl2F12
TMEM49
transmembrane protein 49


200208pl2C6
TMEM56

Homo sapiens cDNA FLJ31842 fis, clone





NT2RP7000259.


041206pl4E12
TMEM75
hypothetical protein LOC641384


170407pl3E9
TMPO
thymopoietin isoform alpha


160507pl3C8
TNNC2
fast skeletal muscle troponin C


150506pl1E3
TOMM7
6.2 kd protein


170407pl3D10
TOMM70A
translocase of outer mitochondrial membrane 70


310505p4f1e11
TOP1
DNA topoisomerase 1


050707pl1F12
TPM1
tropomyosin 1 alpha chain isoform 1


160507pl3B12
TPM2
tropomyosin 2 (beta) isoform 2


160507pl1G2
TPM3
tropomyosin 3 isoform 1


310505p4f1c7
TPM4
tropomyosin 4


010806pl4D12
TPP1
tripeptidyl-peptidase I preproprotein


150506pl2G4
TR
Thioredoxin reductase 1.


190607pl1C7
TRAPPC6A
trafficking protein particle complex 6A


170407vpl3A3
TRIM25
tripartite motif-containing 25


041206pl4E2
TRIM33
tripartite motif-containing 33 protein isoform


310506pl3H6
TSNARE1
t-SNARE domain containing 1


290307pl1H7
TTC1
tetratricopeptide repeat domain 1


130207pl1F6
TTC26
tetratricopeptide repeat domain 26


130207pl2A3
TTC3
tetratricopeptide repeat domain 3


160507pl2A9
TTC9C

Homo sapiens clone pp8376 unknown mRNA.



041206pl1B9
TUBA1B
tubulin, alpha, ubiquitous


160507pl1G1
TUBA1C
tubulin alpha 6


050707pl3C9
TUBB2C
tubulin, beta, 2


200306f7pl1G9
TWF1
twinfilin 1


160507pl1F3
TXN
thioredoxin


010506pl2A3
TXNL1
thioredoxin-like 1


010506pl1A12
TXNRD1
thioredoxin reductase 1


041206pl4H10
TXNRD2
thioredoxin reductase 2 precursor


280705p1f13C6
U2AF1
U2 small nuclear RNA auxiliary factor 1 isoform


171104p31b2
UAP1
UDP-N-acteylglucosamine pyrophosphorylase 1


041206pl2C4
UBA52
ubiquitin and ribosomal protein L40 precursor


050707pl1C1
UBE2D2
ubiquitin-conjugating enzyme E2D 2 isoform 2


031104p47c7
UBE2J2
ubiquitin conjugating enzyme E2, J2 isoform 1


010506pl2A5
UBE2L3
ubiquitin-conjugating enzyme E2L 3 isoform 2


201107pl2C4
UBE2N
ubiquitin-conjugating enzyme E2N


170407vpl2B8
UBE2Q2
ubiquitin-conjugating enzyme E2Q (putative) 2


027073c5
UBE2R2
ubiquitin-conjugating enzyme UBC3B


010806pl3D5
UBE2V1
ubiquitin-conjugating enzyme E2 variant 1


310806pl1E2
UBE2V2
ubiquitin-conjugating enzyme E2 variant 2


310506pl2D9
UBL7
ubiquitin-like 7 (bone marrow stromal


201107pl1C8
UBXD4

Homo sapiens mRNA; cDNA DKFZp313K1023





(from clone DKFZp313K1023).


200208pl2F10
UBXD8
UBX domain containing 8


190607pl1A7
UGCG
ceramide glucosyltransferase


310506pl2A2
UGP2
UDP-glucose pyrophosphorylase 2 isoform b


200906pl3C11
UMPS
uridine monophosphate synthase


200208pl2H8
UNC5D
netrin receptor Unc5h4


160507pl1F2
UNC84A
Sad1/unc-84 protein-like 1 (Unc-84 homolog A).


160507pl1A10
UPF2
UPF2 regulator of nonsense transcripts homolog


041206pl6A3
UPF3A
UPF3 regulator of nonsense transcripts homolog A


200906pl2F9
UQCRB
ubiquinol-cytochrome c reductase binding


290307pl1A3
UQCRFS1
ubiquinol-cytochrome c reductase, Rieske


010806pl4F5
USP10
ubiquitin specific protease 10


010806pl1F11
USP12
ubiquitin-specific protease 12-like 1


130207pl1E5
USP14
ubiquitin specific protease 14 isoform a


310506pl1B3
USP34
ubiquitin specific protease 34


310131e18l1
USP7
ubiquitin specific protease 7 (herpes


170407vpl3B4
UTP11L
UTP11-like, U3 small nucleolar


050707pl3B6
UTRN
utrophin


280305p6f2B6
VAPA
vesicle-associated membrane protein-associated


210206pl1F1
VASP
vasodilator-stimulated phosphoprotein isoform 1


160507pl1E8
VBP1
von Hippel-Lindau binding protein 1


010806pl2B3
VCL
vinculin isoform meta-VCL


010806pl3E12
VIL2
villin 2


200906pl3E11
VKORC1
vitamin K epoxide reductase complex, subunit 1


010506pl1B1
VPS26A
vacuolar protein sorting 26 A isoform 1


290307pl1H3
VPS29
vacuolar protein sorting 29 isoform 2


290307pl1D8
WASF2
WAS protein family, member 2


010506pl2B4
WDR12
WD repeat domain 12 protein


201107pl2B10
WDR25
pre-mRNA splicing factor-like


311007pl1H10
WDR43
WD repeat protein 43.


290307pl1A5
XAGE1
G antigen, family D, 2 isoform 1c


160507pl3B4
XRCC5
ATP-dependent DNA helicase II


310506pl1E7
XRCC6
ATP-dependent DNA helicase II, 70 kDa subunit


310506pl1G5
YAF2
YY1 associated factor 2 isoform b


200906pl1G8
YAP1
Yes-associated protein 1, 65 kD


310806pl2A11
YBX1
nuclease sensitive element binding protein 1


010806pl1F2
YTHDC1
splicing factor YT521-B isoform 1


310506pl3A2
YWHAE
tyrosine 3/tryptophan 5-monooxygenase


170407vpl2D11
YWHAG
tyrosine 3-monooxygenase/tryptophan


201107pl3A9
YWHAH
tyrosine 3/tryptophan 5-monooxygenase


050707pl1C12
YWHAQ
tyrosine 3/tryptophan 5-monooxygenase


310506pl1B1
YY1
YY1 transcription factor


310506pl1G3
ZBTB25
zinc finger protein 46 (KUP)


130207pl1C10
ZBTB8OS
zinc finger and BTB domain containing 8 opposite


310506pl3A5
ZCD1
zinc finger CDGSH-type domain 1


311007pl1E10
ZFAND2A
zinc finger, AN1-type domain 2A


310806pl1A10
ZFR
zinc finger RNA binding protein


311007pl3C4
ZFYVE21
zinc finger, FYVE domain containing 21


280305p5f2E12
ZNF433
zinc finger protein 433


200208pl2A3
ZNF646
zinc finger protein 646


201107pl1C11
ZNHIT3
thyroid hormone receptor interactor 3 isoform 2


170407vpl3B1
ZP3
zona pellucida glycoprotein 3 preproprotein


200906pl1A5
ZW10
centromere/kinetochore protein zw10









The proteins span a wide range of functional categories and localization patterns including membrane, nuclear, nucleolar, cytoskeleton, Golgi, ER and other localizations (SOM) (FIGS. 4A-C). All proteins in the library have localization patterns that match previous studies, when available (mis-localized proteins were excluded from this study).


The present CD-tagging strategy tends to preserve protein functionality [Sigal, Milo et al. 2006, supra]. Note however that the present use of the library does not require proteins to be functional, but merely to act as reliable reporters for the dynamics and location of the endogenous proteins. To test this, the dynamics of endogenous protein using immunoblots on H1299-cherry cells with specific antibodies to 19 different proteins was measured. It was found that in 15/19 cases the immunoblot dynamics were correlated (R>0.5) with the fluorescence dynamics from the movies (FIGS. 5A-S). It was also found, that for all cases in which a band corresponding to the tagged protein was detected using anti-GFP immunoblotting, it indicated a full length fusion (Table 4, herein below).











TABLE 4








Size of YPF-fused protein,



Protein
kDa










name
Clone ID
Expected
Observed













CALM1
150506pl1E2
~47 (20 + 27)
~47


CKS2
010806pl4A1
~47 (10 + 27)
~48


DDX5
090505pl3D6
~95 (68 + 27)
~95



010806pl2F1


EIF3S12
041206pl1C1
~55 (28 + 27)
~55



041206pl5H5

~57


ENO1
150506pl2F1
~77 (50 + 27)
~77


FAU
170407pl2A5
~41 (14 + 27)
~45


FSCN1
010806pl1E12
~82 (55 + 27)
~85


GAPDH
310806pl2C2
 67 (40 + 27)
~66


GNB2L1
310806pl1H12
~64 (37 + 27)
~66


HSP90AA1
310506pl1B9
~120 (90 + 27) 
~120


LMNA/C
310806pl1H11
Lamin A:
~96




~96 (69 + 27)




Lamin C:
~89




~89 (62 + 27)


NPM1
010806pl2H1
~60 (33 + 27)
~67


PBX3
041206pl3C8
~67 (40 + 27)
~70


PEPP-2
010806pl2B4
~59 (32 + 27)
~58



010806pl2D11


PPIA
310506pl4C1
~47 (20 + 27)
~49



031206pl3B6

~47


RPL18
150506pl1C8
~47 (20 + 27)
~47


RPS3A
150506pl1B7
~63 (36 + 27)
~66


TJP1
050707pl1C9
~227 (200 + 27)
~227


TOP1
200906pl1C12
~120 (90 + 27) 
~120



200306pl1H1



010506pl1B1


VPS26A
050707pl1B11
~67 (40 + 27) 
~70



211007pl2A8









Example 3
Assay of Proteomic Response to Drug

Drugs are used to affect the state of the cells, but little is known about the effects of drugs on the dynamics of proteins in individual human cells. The present Example illustrates analysis of drug activity on the dynamics of the proteome in individual cells. To address this, the present inventors employed, as a model system, human cancer cells responding to an anticancer drug with a well characterized target and mechanism of action: camptothecin (CPT). This drug is a topoisomerase-1 (TOP1) inhibitor with no other known targets. It locks TOP1 in a complex with the DNA, causing DNA breaks and inhibiting transcription, eventually causing cell death.


Materials and Methods


Long period time-lapse microscopy: Time-lapse movies were obtained (at 20× magnification) as described by Sigal et al. (Sigal, Milo et al. 2006, supra) with an automated, incubated (including humidity and CO2 control) Leica DMIRE2 inverted fluorescence microscope and an ORCA ER cooled CCD camera (Hamamatsu Photonics). The system was controlled by ImagePro5 Plus (Media Cybernetics) software which integrated time-lapse acquisition, stage movement, and software based auto-focus. During the experiment, cells were grown and visualized in 12-well coverslip bottom plates (MatTek) coated with 10 μM fibronectin (Sigma). For each well time lapse movies were obtained at four fields of view. Each movie was taken at a time resolution of 20 minutes and was filmed for at least three days (over 200 time points). Each time point included three images—phase contrast, red and yellow fluorescence.


Drug Materials: Camptothecin (CPT; C9911 Sigma), was dissolved in DMSO (hybri-max, D2650 Sigma) to achieve a stock solution of 10 mM. In each experiment, drug was diluted to 10 μM in a transparent growth medium (RPMI, X PenStrep, 10% FCS, w/o riboflavin, w/o phenol red, Bet Haemek). Growth medium (2 ml) was replaced by the diluted drug (2 ml) under the microscope. The same procedure was carried out for the following drugs: Etoposide (E1383 Sigma), diluted to 33.3 μM and for Cisplatinum (P4394 Sigma) diluted to 40 μM. The stock solution for ActD (A1410 Sigma) was 1 mg/ml and was diluted to 1 μg/ml.


Image analysis of time lapse movies: A custom written image analysis tool was used developed using the Matlab image processing toolbox environment (Mathworks, Natick, Mass.). The main steps include; image correction, segmentation, tracking of the cells and automated identification of cell phenotypes (mitosis and cell death). Image background correction (flat field correction and background subtraction) was carried out as previously described (Sigal, Milo et al. 2006, supra). No significant bleaching was observed (on average less than 3% over the duration of the experiment). Cell and nuclei segmentation was based on the red fluorescent images—all clones in the library showed similar distribution of red fluorescence—bright in the cytoplasm and significantly brighter in the nuclei. The main steps of the segmentation process are: 1) Differentiation between cells and background by global image threshold using Otsu's method (Otsu 1979, IEEE Transactions on Systems, Man, and Cybernetics 9(1): 62-66); 2) Segmentation of neighboring cells by applying the seeded watershed segmentation algorithm. Seeds were obtained by smoothening the red intensity image and usage of bright nuclei as cell seeds (by identifying local maxima)—one seed per cell; 3) Nuclei segmentation following cell segmentation; each cell was independently stretched between zero and one and a fixed threshold was used to differentiate between the cytoplasm and the nuclei; 4) Tracking of cells was performed by analyzing the movie from end to start and linking each segmented cell to the cell in the previous image with the closest centroid; 5) The automated cell death identification algorithm utilizes the morphological changes correlated with dying cells: rounding followed by blebbing and an explosion of the outer membrane or its collapse. An artificial neural network (ANN) algorithm was constructed that could identify each one of these morphological patters similar to the method previously described in (Eden 2005, IEEE, Transactions on Medical Imaging 24: 1011-1024). Briefly, two sets of images were constructed: The first contained 400 cell images in different stages of cell death and the second contained 400 live cell images. For each image, a collection of high-level image features was computed. An example of such a feature is a measure of object roundness, which is relevant due to the rounding that typically occurs prior to cell death. This process transforms each image into a multi dimensional vector of features. Based on these features an ANN classifier was trained in order to distinguish between live and dead cells resulting in a 96% sensitivity and specificity on a previously unseen test set.


Protein dynamics clustering: The five average population dynamics profiles depicted in FIG. 8B were generated in the following manner: The levels of each protein were smoothed using a median filter and linearly scaled between −1 and 1. The distance between every pair of proteins was measured in terms of Pearson correlation and clustering was performed using a k-means algorithm (reproducibility of results using different seeds is >99%). To choose the number of clusters optimization was effected over the average silhouette score (Blashfield 1991), which measures the dissimilarity of a protein to its assigned cluster compared to other clusters.


GO enrichment analysis: To systematically search for functions processes and localizations common to proteins that show similar dynamics we performed a GO (Ashburner, Ball et al. 2000, Nat Genet 25(1): 25-9) enrichment analysis procedure. A distance measure was devised between a pair of proteins that exploits both the protein amount and its localization changes through time. Formally, each protein i is represented by two vectors, ci and ni, describing the amount of protein in the nucleus and cytoplasm respectively in 141 sequential time points each.


The distance between each pair of proteins i and j was computed using the following formulas:








D
1



(

i
,
j

)


=


1
-

Corr


(



n
i

+

c
i


,


n
j

+

c
j



)



2









D
2



(

i
,
j

)


=

Euc


(



n
i



n
i

+

c
i



,


n
j



n
j

+

c
j




)










D
tot



(

i
,
j

)


=



w
1

·


D
1



(

i
,
j

)



+


w
2

·


D
2



(

i
,
j

)








D1 is one minus the Pearson correlation between the total amounts of two proteins scaled between 0 and 1.


D2 is the normalized Euclidian distance between two vectors that depict the protein localization at each time point. Notice that at a given time






t



n


(
t
)




n


(
t
)


+

c


(
t
)








may range from 0 to 1 corresponding to a cytoplasmic and nuclear localization respectively.


Dtot is the weighted sum of the protein amount and protein localization distances where w1+w2=1 (we used w1=0.5 and w2=0.5). The larger w2 is, the more emphasis is put on localization and consequentially the GO terms that were identified (see next paragraph) were more related to Cellular Compartments terms.


The GO enrichment procedure was performed as following: For each protein a list was generated containing all other proteins ranked according to their distance. Each protein can be thought of as a cluster center and all the other proteins are ranked according to their distance from that center. The present inventors wanted to find whether a subset of proteins that show similar dynamics, i.e. reside near the cluster center, also share a common GO term. To this end a flexible cutoff version of the Hyper Geometric score termed mHG (Eden, Lipson et al. 2007, IEEE, Transactions on Medical Imaging 24: 1011-1024) was used. This analysis was done using GORILLA software [www.cbl-gorilladotcsdottechniondotacdotil/].


Quantitation of nucleolar translocations: To detect translocation events between the nucleoli and the nucleoplasm, a three step process was followed; first the present inventors focused on a subgroup of clones that showed initial nuclear localization of the YFP tagged protein (i.e. pixels of the nucleus were the source of over 50% of the total intensity). Then, for each of the selected clones, the present inventors calculated the ratio of fluorescence intensity between the top and bottom ten percent pixels in individual nuclei and averaged over the population. Clones with a max/min change of over 20 percent in this average during the experiment were inspected manually to verify the source of change in pixel intensity distribution and were classified as clones showing nucleolar translocation.


Finally, to quantify the extent and direction (nucleoli to nucleoplasm or vise versa) of the translocation, the present inventors calculated the ratio between mean fluorescence intensity of nucleoli vs. nucleoplasm (Rncll/nuc) at the two time points were the max/min ratio was maximized and minimized. Measurements were normalized to 0.5, 1 and 2 at time point of drug addition, based on the Rncll/nuc ratio at that time (Rncll/nuc<0.8, 0.8<Rncll/nuc<1.2 and Rncll/nuc>1.2 respectively).


Determination of ‘bimodal’ behaviors: The coefficient of variance (CV defined as the ration between the std between cells and the mean) was measured for 400 proteins for 47 hours following addition of CPT (at a 20 minute resolution) (see FIGS. 13A-B). All CVs were normalized to average 1 (CV(i,j)/mean(mean(CV)) where i is protein number (i=1 . . . 400) and j is timepoint (j=1 . . . 141)). All proteins deviating 3 standard deviations from the average normalized CV were considered as ‘bimodal’ candidates (N=59). Following manual inspection, 30 of these proteins listed in Table 4 were denoted as bimodal.


Immunoblots against 20 selected proteins: Total cell lysates were prepared with RIPA buffer (Pierce) according to manufacturer's instructions. The protein concentrations were determined by BCA protein assay kit (Thermo scientific). Equal amounts of proteins were resolved on SDS-PAGE and subjected to immunoblotting analysis by using the antibodies listed below. The intensity of protein bands was quantified by using ImageJ software.


The following commercially available primary antibodies were used in the study: Antibodies against AKAP8L (ab51342), Calmodulin (ab38590), Cyclophilin A (ab3563), DDX5 (ab21696), Enolase (ab35075 and ab49256), eIF3K (ab50736), GAPDH (ab9285 and ab9484), HSP90 (ab13492 and ab34909), Nucleophosmin (ab15440), PBX3 (ab56239), Topoisomerase1 (ab28432) and VPS26 (ab23892) were purchased from Abcam.


Anti-Calmodulin (FL-149), -HDAC2 (H-54), -RACK1 (H-187 and B-3) and -ZO1 (H-300) antibodies were from Santa-Cruz.


Antibodies against RPL37 (A01), RPS7 (A01) and RPS3 (A01) proteins were obtained from Abnova.


Anti-Myosin IIA (M8064) and anti-GFP (11814460001) antibodies were from Sigma and Roche, respectively.


Conversion of fluorescence arbitrary units to scalable units: The present CD-tagging approach introduces a fluorescent protein into an endogenous protein, as an artificial exon. Under constant conditions (i.e. same exposure time and same lamp intensity) and under the assumption that the number of photons emitted and captured by each fluorescent molecule is similar, one can use fluorescence measurements to compare protein abundances. However, in practice, exposure times and lamp intensities differ between experiments and thus have to be corrected for. Exposure times of yellow and red channel were recorded throughout the experiments. In order to correct for differences in lamp intensity the red fluorescence levels averaged over all cells in a movie were used as a signal to align all clones. The following procedure was used to transform arbitrary fluorescent units to scalable units:


Fr, Fy—measured red, yellow fluorescence


Er, Ey—exposure time for red, yellow channel


Pr, Py—number of proteins tagged with red, yellow fluorescence


L—lamp intensity

    • 1. Fluorescence is a product of exposure time, protein number and lamp intensity.






F
r
=E
r
·P
r
·L F
y
=E
y
·P
y
·L




    • 2. To estimate the lamp intensity, it can be assumed that the average expression of the red marker, Pr, is the same for all clones→Pr=Const.














1
+
2


L

=



F
r



E
r

·

P
r



=



F
r



E
r

·
Const


.





3







1
+
3



F
y


=



E
y

·

P
y

·
L

=


E
y

·

P
y

·



F
r



E
r

·
Const


.






4






4


P
y


=




E
r

·

F
y

·
Const



E
y

·

F
r



=




E
r

·

F
y




E
y

·

F
r






(

Const





omitted

)

.






5






Following this scaling procedure, correlation of yellow intensity of the same protein from the same clone at a given time point, measured in two different days (starting form frozen cells) is very high, R=0.975 p<0.001. Moreover, the correlation of fluorescence intensity of a protein in two different clones where the protein is tagged at different chromosomal locations within the gene, is high, R=0.63 p<0.005. (FIGS. 20A-B). This suggests that the scaling procedure results in fluorescence units that allow determination of relative protein levels despite variations in lamp intensity and exposure times.


Identification of a drug target that acts to increase cell death following CPT treatment: Cells were plated in 12 well plate in 2 ml medium and filmed using the microscope under incubator conditions. At the begining of the movie, 1 μM of DDX5-siRNA (SEQ ID NOs: 175-178) was added. After three days, the DDX5-siRNA was removed and 10 μM of camptothecin was added. The cells continued being filmed at a 20 minute resolution for over 96 hours (whole experiment is over 144 hours). As controls, the experiment was repeated, but the DDX5-siRNA was replaced either by non-targeted-siRNA or no siRNA at all. As a further control, the identical experiment was repeated in the absence of camptoithecin.


Results


Cells were grown in 12-well plates in an automated fluorescence microscope with temperature, CO2 and humidity control. Each well contained cells tagged for a different protein. After 24 hours of growth, the drug CPT was added (10 uM) and cells were tracked for another 48 hours (FIGS. 3A-D). Images in phase, red and yellow were taken every 20 minutes, at four positions in each well. An auto-focus system ensured that stable time-lapse movies could thus be collected, resulting in over 200 consecutive frames per protein studied, where each frame contained 10-40 different cells. Movies were stored and analyzed automatically using a computer cluster, resulting in traces of protein level and location in each cell over time.


The cells showed vigorous divisions in the first 24 hours prior to drug addition, with a cell cycle of about 20 hours. Then, after drug addition, cells showed loss of motility and growth arrest after about 10 hours, and began to show cell rounding and blebbing (morphological correlates of cell death) reaching about 15% of the cells after 36 hours (FIG. 6). Day-day repeats starting from frozen cells showed a mean error in the YFP fluorescent signals of up to 15% (FIGS. 7A-I). Thus, dynamic changes on the order of 20-30% change in tagged protein intensity in individual cells are typically significant using the present assay.


Temporal profiles of protein concentration: The total fluorescence of each YFP tagged protein was measured in each cell. Overall, about 70% of the proteins show a decrease in intensity in response to the drug, on diverse timescales. The median dynamic range of this response was a 1.3-fold change in fluorescence and the largest changes were about five-fold change in fluorescence. Proteins show distinct classes of profiles, as obtained using k-means clustering (FIGS. 8A-B). The fluorescence levels of a third of the proteins decrease in the first 24 hours after drug addition (profile i). About half of the proteins show an increase followed by a decrease (profiles ii and iii). Other proteins showed an increase early (profile iv) or late, more than a day after drug addition (v). The present data includes dynamics of about 200 proteins annotated as uncharacterized hypothetical proteins or ESTs (Table 2, hereinabove). The dynamics of these uncharacterized proteins are found throughout all of the present profiles (FIG. 8B).


Groups of functionally related proteins tended to show similar dynamics and protein localization profiles. For example, over 75% (31/40) of the ribosomal proteins tagged in the library showed highly correlated dynamics of early degradation (p<10−3) (FIG. 8C and FIGS. 9A-D). This rapid degradation was also found in immunoblots with antibodies against ribosomal proteins RPS3a and RPL7. Proteins with slower apparent degradation include cytoskeleton components and metabolic enzymes. The timing of degradation of most cytoskeleton proteins correlated with the timing of the loss of cell motility as measured by tracking of cells (FIG. 8D). Proteins that rise late in the response include some helicases implicated in DNA damage repair and apoptosis-related proteins such as the Bcl2 associated proteins BAG2, BAG3 and programmed cell death protein PDCD5.


The drug target is among the first to respond: The drug target TOP1 is found in the nucleoli and nucleus of cells prior to drug addition. Drug addition caused TOP1 levels in the nucleoli to drop within less than 2 minutes (FIG. 10). The total cellular fluorescence levels of tagged TOP1 decreased on a timescale of under an hour, preceding almost all other responses in the present study (TOP1 is in the first 1% of responding proteins, FIG. 8B, arrow). The higher the CPT dose, the larger the extent TOP1 fluorescence decrease (FIG. 11E). Such rapid degradation was also found in immuoblots with anti-TOP1 antibodies (FIG. 11F).


In addition to nucleolar exit in the TOP1 tagged clone, it was found that fluorescence accumulates in the cytoplasm on the timescale of 5 hours following CPT addition, and that this accumulation increased with drug dose. Immunostaining of H1299-cherry cells with anti-TOP1 antibodies also showed endogenous TOP1 in the cytoplasm 5 hours after CPT treatment. Immunoblots indicated that as TOP1 degraded, an approximately 40 KD fragment detectable with anti-YFP antibody accumulated. None of the other 20 proteins tested with immunoblots in this study showed such a YFP fragment (FIGS. 5A-L and 11F). Taken together, these results suggest that TOP1 may be proteolised, and that TOP1 fragments exit the nucleus following drug administration. Other drugs, including DNA damaging drugs like TOP-2 inhibitor etoposide and cisplatin, did not show any of these effects on TOP1 (FIGS. 11C-D).


Rapid localization changes suggest nucleolar stress: In addition to TOP1, almost all of the other proteins that show rapid localization changes following CPT addition were localized to the nucleoli. The nucleolus is a key organelle that coordinates the synthesis and assembly of ribosomal subunits. Nucleolar proteins were identified that showed a reduction in nucleolar intensity (FIG. 12A), whilst other nucleolar proteins were identified that showed an increase followed by a return to basal level (FIG. 12B). Corresponding changes in the nuclear intensity outside of the nucleoli were found, suggesting that these are translocation events. In addition to localization changes, rapid decrease in the total level was seen in several nucleolar proteins, including ribosomal proteins. Similar results for the dynamics of most of these proteins (4 out of 5 proteins tested) were also found in response to the transcriptional inhibitor actinomycinD (1 μg/ml) FIGS. 13A-B. Similar nucleolar changes have been previously found in a study that monitored the composition of nucleoli extracted from cells responding to actinomycinD [Andersen, Lam et al. 2005, Nature 433(7021): 77-83]. In summary, these results suggest that the immediate effect of CPT on these cells is transcription inhibition, causing nucleolar stress.


Nuclear localization changes following drug addition: The localization of each protein across the experiment was analyzed and the ratio of cytoplasmic to nuclear fluorescence was followed as a function of time. It was found that about 1% of the proteins showed significant change in nuclear localization (defined as >20% change in the cytoplasm/nuclear fluorescence ratio in an anti-correlated manner). Both rapid and slow localization changes between the cytoplasm and the nucleus were detected (FIGS. 14A-C). Among the latter are two proteins in the stress response pathway to oxidative stress: Both thyredoxin and thyredoxin reductase) showed an increase in nuclear/cytoplasmic ratio within 8 hours after drug addition (FIG. 15). As nuclear levels rise, cytoplasmic levels seem to decrease proportionally, and vise versa, suggesting that these translocations represent movement between these two compartments.


Several Proteins Show Highly Variable Behavior that Correlates with Outcome of Individual Cells:


The present system allows monitoring of the cell-cell variability of each protein over time. All proteins were found to show significant cell-cell variability in their fluorescence levels. At the time of drug addition, the level of each protein showed a standard deviation between cells that ranged between 10% and 60% of the mean. This variability is in accord with that previously found, both in microorganisms and human cells (Sigel, Milo et al. 2006, supra). Part of this variability is due to differences in the cell cycle stage of the cells. To quantify this, the cells were binned according to the time between their last division and the time of drug addition—an ‘in-silico’ synchronization approach (Sigel, Milo et al. 2006, supra). It was found that about 20% of the variability was due to cell-cycle stage difference, and the remainder was presumably due to stochastic processes.


The degree of cell-cell variability, defined as the standard deviation between cells divided by the mean, was found to show a slight increase as a function of time following drug addition for most proteins (FIG. 16) (noise increased by 30% on average). For most proteins, nearly all cells in the population showed similarly shaped profiles of fluorescence dynamics, rising and falling together (FIGS. 17A-B).


Diverging from this norm were about 30 proteins which showed a special behavior. At first, they showed the typical variability with similar dynamics in each cell. Then, at about 20 hours following drug addition, the cell population began to show dramatic cell-cell differences in the dynamics of these proteins (FIGS. 17C-F). Some cells showed an increase in the fluorescence levels, while other cells stayed constant or showed a decrease. Thus, these proteins seemed to show bimodal dynamical behavior.


Importantly, the different behaviors of some of these proteins are linked to the fate of each cell. For example, it was found that the RNA-helicase DDX5 increased markedly in cells that survive to the end of the movies (FIG. 18A). This is consistent with its suggested anti-apoptotic role (Yang, Lin et al. 2007, Oncogene 26(41): 6082-92). Its levels decrease in cells that undergo the morphological changes associated with cell death. Thus, the fluorescence dynamics of this protein were significantly correlated with the cell fate (p<10̂-13, FIG. 18B). Such effects can not be detected in assays that average over cell populations. The bimodality of DDX5 was found to be drug specific, since tagged DDX5 did not show bimodal behavior in response to other anti-cancer drugs including etoposide and cisplatin (see FIGS. 19A-F).


A second protein that shows similar behavior to DDX5 is Replicator factor C activator 1 (RFC1; FIGS. 21A-B). Replication factor C is a DNA-dependent ATPase that is required for eukaryotic DNA replication and repair. The protein acts as an activator of DNA polymerases.


A third protein that showed bimodal dynamical behavior is thioredoxin reductase 1 (TXNRD1). This protein is involved in the cellular response to oxidative stress. Following changes in NADPH levels, TXNRD1 reduces thioredoxin which translocates into the nucleus and eventually leads to the expression of stress related genes.


The present study showed that both TXNRD1 and thioredoxin enter the nucleus in response to Camptothecine. Previously it was suggested that these proteins are novel drug targets and that their inhibitors should be used together with ionizing radiation (IR) or H2O2 [Nguen et al., Cancer Letters, Volume 236, Issue 2, Pages 164-174 P].


Table 5, herein below lists the functions of the proteins with bimodal behavior, and gives reference to association of some of the proteins to cell fate.












TABLE 5








Reference to





association of





protein to cell


Protein name
Clone ID
description
death







BAG2
010806pl1C7
BCL2-associated athanogene 2



BAG3
170407pl3D4
BCL2-associated athanogene 3
P. Bonelli et al.,





Leukemia 18,





358-60 (Feb,





2004)


C9ORF40
130207pl1E1
hypothetical protein LOC55071


CALM1
150506pl1E2
calmodulin 1
O. Cohen, E. Feinstein,





A. Kimchi,





Embo J





16, 998-1008





(Mar. 3, 1997).





Y. Shirasaki, Y. Kanazawa,





Y. Morishima,





M. Makino,





Brain





Res 1083, 189-95





(Apr. 14, 2006


CALM2
310506pl3B1
calmodulin 2
O. Cohen, E. Feinstein,





A. Kimchi,





Embo J





16, 998-1008





(Mar. 3, 1997).





Y. Shirasaki, Y. Kanazawa,





Y. Morishima,





M. Makino,





Brain





Res 1083, 189-95





(Apr. 14, 2006


CAV1
170407pl1C2
caveolin 1
C. C. Ho et al.,





Lung Cancer 59,





105-10 (Jan,





2008).


CCDC23
310506pl2C3
coiled-coil domain containing 23


DDX5
010806pl2F1
p68 RNA helicase
L. Yang, C. Lin,





S. Y. Sun, S. Zhao,





Z. R. Liu,





Oncogene 26,





6082-92 (Sep. 6,





2007).


DKFZP434M1123
160507pl1B11
hypothetical protein


EIF1AX
010806pl2B11
eukaryotic translation initiation




factor 1A, X-linked


FABP5
200906pl1B6
fatty acid binding protein 5


FSCN1
010806pl1E12
fascin homolog 1, actin-bundling




protein


PCMTD2
010506pl2D2
protein-L-isoaspartate (D-




aspartate) O-methyltransferase




domain containing


PDCD5
170407pl1B5
programmed cell death 5
M. Xu et al.,





Gene 329, 39-49





(Mar. 31,





2004).


PFN1
050707pl2E5
profilin 1


NPM1
010806pl2H1
Nucleophosmin (B23)
Y. Qing, G. Yingmao,





B. Lujun,





L. Shaoling, J





Neurol Sci 266,





131-7 (Mar. 15,





2008)


PPP1R2
010806pl1G5
protein phosphatase 1,




regulatory (inhibitor) subunit 2


PTTG1
310506pl2C2
pituitary tumor-transforming 1
Y. Lai, D. Xin, J. Bai,





Z. Mao, Y. Na,





J Biochem





Mol Biol 40,





966-72 (Nov. 30,





2007).


RFC1
050707pl1B12
replication factor C (activator 1)


RPS3
150506pl2B7
ribosomal protein S3
C. Y. Jang, J. Y. Lee,





J. Kim,





FEBS Lett 560,





81-5 (Feb. 27,





2004).


SLBP
010506pl2E6
stem-loop binding protein
Y. Kodama, J. H. Rothman,





A. Sugimoto,





M. Yamamoto,





Development





129, 187-96





(Jan, 2002).


SPCS1
050707pl2F4
signal peptidase complex




subunit 1 homolog


TOMM70A
170407pl3H11
translocase of outer




mitochondrial membrane 70




homolog A


YT521
010806pl1F2
YTH domain containing 1









Identification of a drug target that acts to increase cell death following CPT treatment: As mentioned, a subgroup of proteins was found that show bimodal behavior in response to drug (Camptothecin). Of these, two (DDX5 and RFC1) showed that this behavior was correlattive to cell fate (FIGS. 18A-B and 21A-B).


The present inventors then hypothesised thatt down-regulation of DDX5 may lead to higher levels of cell death. As illustrated in FIG. 22, application of DDX5-siRNA, (thereby causing a reduction in expression levels by at least 80%), caused an increase rate (approximately double) in cell death following drug addition. This holds for at least the first 35 hours following drug addition. Addition of DDX5-siRNA did not cause cell death on its own (with OUT CPT—purple line). This suggests that the effect of downregulation of DDX5 on cell death will be observed only in cells that initially respond to CPT. All of the above suggests that a drug target has been identified that when inhibited doubles the rate of cell death following CPT administration.


Discussion


This study suggests that viewing the drug response of about 1000 proteins in human cancer cells in space and time, offers insight into the drug mechanisms of action, and uncovers proteins correlated with the fate of cell subpopulations. The present inventors found rapid and specific initial movements to and from the nucleoli of a group of proteins, including the drug target. Slower, broad patterns of protein accumulation and degradation followed, as the cells stopped moving and began cell death. Specific proteins showed high cell-cell variability that correlated with cell survival or death.


The present data is relevant to the question of diversity in the response of individual cells to a drug. The present inventors found that most proteins showed variability between cells, on the order of 10-60% in their mean levels. The drug seemed to cause a slight increase in the cell-cell variability of almost all proteins. This variability is not strongly correlated with the cell fate for most proteins. However, a small set of proteins showed variability that was highly correlated with the cell fate. These proteins may play a role in cell survival and death specific to this drug, or at least may be downstream factors associated with the molecular variability that underlies differential response. This suggests a way to begin to understand non-genetic resistance of human cell subpopulations to drugs, and may point to potential secondary targets that can enhance the effects of a given drug.


These results also suggest a separation of timescales in the response, where rapid and specific responses are mediated by translocation, and slower responses that include large sets of proteins are mediated by slower changes in expression and degradation. The translocations that occur soon after the drug is added may point to feedback mechanisms which sense the immediate effect of the drug. In the present study, CPT is found to have an almost immediate effect on nucleolar proteins. This response is typical of the nucleolar response to transcriptional inhibition. Notably, the drug target TOP1 is among the first to respond. This may suggest a strategy to understand drug mechanism of action and to detect drug targets and target-associated proteins for drugs with unknown targets.


The present library also provides dynamics and localization data for about 200 proteins that are classed as hypothetical proteins or ESTs (FIG. 8B and Table 2). The library provides a universal epitope tag (yellow fluorescent protein) that can in principle be used for biochemical assays on these novel proteins. The present approach may thus offer an opportunity to characterize new proteins.


The present library employs tagging that preserves endogenous regulation and is built to allow robust image quantification. Its reproducibility, temporal resolution and accuracy allow even small dynamical features to be reliably detected.


In summary, this first broad view of the response of the proteome of individual human cells to a drug points to aspects of the drug mode of action and to specific differences in protein expression in cell subpopulations. Rapid localization changes help to pinpoint the drug target, and slower waves of accumulation and degradation provide a picture of the way the cells respond to drug stress over time. A subset of proteins showed behavior correlated with the survival and death of differential cell subpopulations. This opens the way for viewing and potentially understanding the dynamics of the human proteome under diverse drugs and conditions in individual cells.


Although the invention has been described in conjunction with specific embodiments thereof, it is evident that many alternatives, modifications and variations will be apparent to those skilled in the art. Accordingly, it is intended to embrace all such alternatives, modifications and variations that fall within the spirit and broad scope of the appended claims.


All publications, patents and patent applications mentioned in this specification are herein incorporated in their entirety by into the specification, to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated herein by reference. In addition, citation or identification of any reference in this application shall not be construed as an admission that such reference is available as prior art to the present invention. To the extent that section headings are used, they should not be construed as necessarily limiting.

Claims
  • 1. A nucleic acid construct system comprising: (i) a first nucleic acid construct comprising a first nucleic acid sequence encoding a first reporter polypeptide linked to an additional nucleic acid sequence capable of inserting said first nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to said first reporter polypeptide is expressed in said cell, said endogenous polypeptide having a higher nuclear:cytoplasm expression ratio; and(ii) a second nucleic acid construct comprising a second nucleic acid sequence encoding a second reporter polypeptide, linked to an additional nucleic acid sequence capable of inserting in a non-directed manner said second nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to said second reporter polypeptide is expressed in said cell, wherein said first reporter polypeptide and said second reporter polypeptide are distinguishable.
  • 2. The nucleic acid construct system of claim 1, further comprising a third nucleic acid construct comprising a third nucleic acid sequence encoding said first reporter polypeptide linked to an additional nucleic acid sequence capable of inserting said third nucleic acid construct into a genome of a host cell such that an additional endogenous polypeptide covalently attached to said first reporter polypeptide is expressed in said cell.
  • 3.-10. (canceled)
  • 11. The nucleic construct system of claim 1, wherein said first reporter and said second reporter are fluorescent polypeptides that fluoresce at a distinguishable wave length.
  • 12. A cell expressing at least two endogenous polypeptides, each covalently attached to a distinguishable reporter polypeptide wherein at least one of said at least two endogenous polypeptides has a higher nuclear:cytoplasm expression ratio.
  • 13. (canceled)
  • 14. The cell of claim 12, expressing an additional endogenous polypeptide attached to a reporter polypeptide, said reporter polypeptide being identical to one of said two distinguishable reporter polypeptides.
  • 15. The cell of claim 12, wherein an expression of said at least one of said at least two endogenous polypeptides is constitutive.
  • 16. The cell of claim 12, comprising a nucleic acid construct system comprising: (i) a first nucleic acid construct comprising a first nucleic acid sequence encoding a first reporter polypeptide linked to an additional nucleic acid sequence capable of inserting said first nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to said first reporter polypeptide is expressed in said cell, said endogenous polypeptide having a higher nuclear:cytoplasm expression ratio; and(ii) a second nucleic acid construct comprising a second nucleic acid sequence encoding a second reporter polypeptide, linked to an additional nucleic acid sequence capable of inserting in a non-directed manner said second nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to said second reporter polypeptide is expressed in said cell, wherein said first reporter polypeptide and said second reporter polypeptide are distinguishable.
  • 17.-19. (canceled)
  • 20. A cell population, wherein each cell of the population expresses at least two endogenous polypeptides, each covalently attached to a distinguishable reporter polypeptide, wherein at least one of said at least two endogenous polypeptides is identical in each cell of said cell population.
  • 21. The cell population of claim 20, expressing an additional endogenous polypeptide attached to a reporter polypeptide, said reporter polypeptide being identical to one of said two distinguishable reporter polypeptides.
  • 22. The cell population of claim 20, wherein both of said at least two endogenous polypeptides are identical in each cell of said cell population.
  • 23. (canceled)
  • 24. The cell population of claim 20, wherein at least one of said at least two endogenous polypeptides comprises a sequence as set forth in SEQ ID NOs: 1-164.
  • 25.-26. (canceled)
  • 27. A method of generating a cell population, the method comprising: (a) introducing a first nucleic acid construct into a first population of cells, said first nucleic acid construct comprising a first nucleic acid sequence encoding a first reporter polypeptide linked to an additional nucleic acid sequence capable of inserting said first nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to said first reporter polypeptide is expressed in said cell;(b) selecting a cell wherein said first reporter comprises a higher nuclear:cytoplasm expression ratio;(c) propagating said cell to generate a second population of cells;(d) introducing a second nucleic acid construct into the second population of cells, said second nucleic acid construct comprising a second nucleic acid sequence encoding a second reporter polypeptide, linked to an additional nucleic acid sequence capable of inserting in a non-directed manner said second nucleic acid construct into a genome of a host cell such that an endogenous polypeptide covalently attached to said second reporter polypeptide is expressed in said cell, wherein said first reporter polypeptide and said second reporter polypeptide are distinguishable.thereby generating the cell population.
  • 28.-29. (canceled)
  • 30. The method of claim 27, further comprising identifying at least one of said endogenous polypeptides.
  • 31. A method of identifying a target of an agent, the method comprising: (a) contacting the cell population of claim 22 with the agent;(b) analyzing a localization or amount of at least one of said endogenous polypeptides, wherein a change in said amount or localization is indicative of a target of the agent.
  • 32.-34. (canceled)
  • 35. A method of identifying an agent capable of affecting a cell state, the method comprising, (a) contacting the cell population of claim 22 with an agent; wherein at least one of said endogenous polypeptides is a marker for the cell state; and(b) measuring a localization or amount of said marker, wherein a change in said amount or localization of said marker is indicative of an agent capable of affecting the cell state.
  • 36.-37. (canceled)
  • 38. A method of identifying a marker for disease prognosis, the method comprising: (a) contacting the cell population of claim 22 with a therapeutic agent, the cell population comprising diseased cells;(b) comparing a localization or amount of said at least one endogenous polypeptide in responsive cells of the cell population with non-responsive cells of the cell population; wherein a difference in expression or localization of said at least one endogenous polypeptide in responsive and non-responsive cells is indicative that said endogenous polypeptide is the marker for disease prognosis.
  • 39. (canceled)
  • 40. A method of analyzing a localization of a first and second endogenous polypeptide in a cell, the method comprising detecting a localization of said first and second endogenous polypeptide in said cell, wherein said first and second polypeptide are each covalently attached to a distinguishable reporter polypeptide, thereby analyzing localization of a first and second polypeptide.
  • 41.-44. (canceled)
PCT Information
Filing Document Filing Date Country Kind 371c Date
PCT/IL09/00089 1/22/2009 WO 00 7/22/2010
Provisional Applications (2)
Number Date Country
61006634 Jan 2008 US
61136356 Aug 2008 US