CA2497873A1 - Novel diagnostic and therapeutic methods and reagents therefor - Google Patents

Novel diagnostic and therapeutic methods and reagents therefor Download PDF

Info

Publication number
CA2497873A1
CA2497873A1 CA002497873A CA2497873A CA2497873A1 CA 2497873 A1 CA2497873 A1 CA 2497873A1 CA 002497873 A CA002497873 A CA 002497873A CA 2497873 A CA2497873 A CA 2497873A CA 2497873 A1 CA2497873 A1 CA 2497873A1
Authority
CA
Canada
Prior art keywords
protein
edd
complex
seq
nucleic acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA002497873A
Other languages
French (fr)
Inventor
Colin Watts
Darren Saunders
Michelle Henderson
Jennifer Clancy
Susan Henshall
Robert Sutherland
Philippa O'brien
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Garvan Institute of Medical Research
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU2002951346A external-priority patent/AU2002951346A0/en
Application filed by Individual filed Critical Individual
Publication of CA2497873A1 publication Critical patent/CA2497873A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/40Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/93Ligases (6)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/112Disease subtyping, staging or classification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/118Prognosis of disease development
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/136Screening for pharmacological compounds
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/158Expression markers
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/90Enzymes; Proenzymes
    • G01N2333/9015Ligases (6)
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • Pathology (AREA)
  • Microbiology (AREA)
  • Analytical Chemistry (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Physics & Mathematics (AREA)
  • Cell Biology (AREA)
  • Hospice & Palliative Care (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • Hematology (AREA)
  • Urology & Nephrology (AREA)
  • Food Science & Technology (AREA)
  • General Physics & Mathematics (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

This invention provides novel methods of detecting or treating aberrant cell cycle regulation associated with expression of a nuclear protein encoded by a gene that is linked to map position 8q22.3 of the human genome, and to novel reagents that are useful therefor. More particularly, the invention provides novel nucleic acid and proteinaceous probes, for detecting a gene that is linked to map position 8q22.3 of the human genome or the expression products thereof, wherein expression or elevated expression of said gene is associate d with the appearance or occurrence of tumors associated with cancer, DNA dama ge and progesterone-receptor-mediated effects on cells. The invention also provides reagents and methods for detecting or modulating the expression products of the gene, such as, for example, in the diagnosis or treatment of cancer, cellular proliferation, DNA damage or progesterone receptor-mediated effects on cells.

Description

Novel diagnostic anc~ therapeutic methods and reagents therefor FIELD OF THE INVENTION
This invention relates to novel methods of detecting or treating aberrant cell cycle regulation associated with expression of a nuclear protein encoded by a gene that s -is linked to map position 8q22.3 of the human genome, and to novel reagents that are useful therefor. More particularly, the invention relates to novel nucleic acid and proteinaceous probes, including antibodies, for detecting a gene that is linked to map position 8q22.3 of the human genome or the expression products thereof, wherein expression or elevated expression of said gene is associated with the to appearance or occurrence of tumors associated with cancer, DNA damage and progesterone-receptor-mediated effects on cells. The invention also relates to reagents and methods for targeting the expression products (e.g. mRNA, protein, or protein-protein complexes) of the gene, such as, for example, in the prophylactic or therapeutic treatment of cancer or for reducing or preventing cell is proliferation (e.g. in tumors), or for modifying progesterone receptor-mediated effects. In another embodiment, the invention relates to reagents and methods for detecting or modulating the expression products (e.g. mRNA, protein, or protein-protein complexes) of the gene, such as, for example, in the diagnosis or treatment of cancer, DNA damage or progesterone receptor-mediated effects on 2o cells. In yet another embodiment, this invention relates to a variant of a gene that is linked to map position 8q22.3 of the human genome and to novel reagents and methods for detecting said variant.
2s BACKGROUND OF THE INVENTION
1. General This specification contains nucleotide and amino acid sequence information prepared using Patentln Version 3.1, presented herein after the claims. Each nucleotide sequence is identified in the sequence listing by the numeric indicator 30 <210> followed by the sequence identifier (e.g. <210>1, <210>2, <210>3, etc).
The length and type of sequence (DNA, protein (PRT), etc), and source organism for each nucleotide sequence, are indicated by information provided in the numeric indicator fields <211 >, <212> and <213>, respectively. Nucleotide sequences referred to in the specification are defined by the term "SEQ ID
NO:", followed by the sequence identifier (eg. SEQ ID NO: 1 refers to the sequence in s the sequence listing designated as <400>1 ).
The designation of nucleotide residues referred to herein are those recommended by the IUPAC-IUB Biochemical Nomenclature Commission, wherein A represents Adenine, C represents Cytosine, G represents Guanine, T represents thymine, Y
to represents a pyrimidine residue, R represents a purine residue, M
represents Adenine or Cytosine, K represents Guanine or Thymine, S represents Guanine or Cytosine, W represents Adenine or Thymine, H represents a nucleotide other than Guanine, B represents a nucleotide other than Adenine, V represents a nucleotide other than Thymine, D represents a nucleotide other than Cytosine and N
is represents any nucleotide residue.
As used herein the term "derived from" shall be taken to indicate that a specified integer may be obtained from a particular source albeit not necessarily directly from that source.
Throughout this specification, unless the context requires otherwise, the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated step or element or integer or group of steps or elements or integers but not the exclusion of any other step or element or integer 2s or group of elements or integers.
Those skilled in the art will appreciate that the invention described herein is susceptible to variations and modifications other than those specifically described.
It is to be understood that the invention includes all such variations and 3o modifications. The invention also includes all of the steps, features, compositions and compounds referred to or indicated in this specification, individually or collectively, and any and all combinations or any two or more of said steps or features.
The present invention is not to be limited in scope by the specific embodiments s described herein, which are intended for the purposes of exemplification only.
Functionally-equivalent products, compositions and methods are clearly within the scope of-the invention, as described herein.
2. Description of the related art to It is widely recognized that simple and rapid tests for hyperproliferative disorders have considerable clinical potential. Not only can such tests be used for the early diagnosis, but they also allow the detection of disease recurrence following treatment. However, the diagnosis of many hyperproliferative disorders, such as, for example, carcinoma of the ovary, is generally only possible when the disease is has progressed to a late stage of development. Whilst previously identified markers for many carcinomas, e.g. carcinomas of the lung, prostate, breast, colon, pancreas, and ovary, have facilitated efForts to diagnose and treat these serious diseases, there is a clear need for the identification of additional markers and therapeutic targets. There is a clear need for markers that will facilitate .the 2o early-stage detection and treatment of hyperproliferative disorders in humans and other mammals. The identification of factors correlating with hyperproliferative disorders is a prerequisite for the identification of diagnostic markers.
Clearly, hyperproliferative disorders involve unregulated cell division, suggesting 2s an association with aberrant cell cycle regulation. Aberrant signal transduction is associated with aberrant cell cycle regulation and the occurrence of hyperproliferative disorders such as cancer, and DNA damage. In response to DNA damage and replication blocks, cell cycle progression is halted through the control of critical cell cycle regulators. Genomic damage, if left unrepaired, can 30 lead to malignant transformation, or cell death by senescence (aging), necrosis or apoptosis.

For example, the action of steroid hormones generally involves signal transduction cascades involving translocation of hormone receptors from the cytoplasm to the nucleus, thereby effecting many cellular and tissue functions. Progesterone, a s pregnene-3,20-dione derived from cholesterol, is a critical component of the female reproductive cycle, wherein oscillations in the serum plasma levels of progesterone during each cycle of ovulation help to mediate biochemical and molecular activity in target tissues and result in anatomical and morphological changes. The molecular target of progesterone is the intracellular progesterone to receptor (PR). PR is present in the cytoplasm in a heterocomplex comprising several other proteins and factors termed the PR heterocomplex (PRC). The PR
is maintained in an inactive form by molecular chaperones, immunophillins, and heat shock proteins (hsp70, hsp90, hsp27, and p59 (hsp56), p48 and p23; Johnson et al., Mol Cell Biol 74,1956-1963, 1994). Active PR binds progesterone and is translocates to the nucleus where it binds as a transcription factor to canonical DNA transcriptional elements present in progesterone-regulated genes.
Progesterone-regulated genes have also been implicated in differentiation and in the cell cycle (Moutsatsou and Sekeris, Ann NYAcad Sci 876, 99-115, 1997) and certain cancers.
The assembly of the PRC in vitro involves an ordered interaction between PR
and at least eight components. For example, hsp70 binds to the PR and prevents interaction with its ligand; and hsp90 prevents intranuclear translocation by PR in the absence of progesterone (Kang et al., Proc Natl Acad Sci 97, 340-344, 1994).
2s Chemical modification of hsp70 and hsp90 causes release of PR. Other signals may affect the interactions of hsp90 with p23. Arrest of PRC assembly in vitro may be blocked by the selective hsp90 binding agent ~geldanamycin (GA).
Intermediate PR complexes including hsp90 and p23, but do not bind progesterone, are formed in the presence of GA (Smith et al., Mol Cell Biol 75, 6804-6812, 1995). The hsp70 protein binds the mutated tumor-suppressor gene p53 and has been associated with decreased nuclear localization of PR in tissue from node-negative breast tumors (Elledge et al, Cancer Res 54, 3752-3757, 1994).
Ubiquitin-mediated proteoloysis is required for the regulation of many key cellular s pathways including the control of cell cycle progression (King et al., Science 274, 1652-1659, 1996; Draetta, Curr, Opin. Cell Biol. 6, 842-846, 1994; Nefsky et al., EMB~ J. 75, 1301-1312, 1996), cellular signal transduction (Joanzeiro et al., Science 286, 309-312, 1999; Joanzeiro et al. Cell 702, 549-552, 2000; Zhu et al., Nature 400, 687-693, 1999), DNA damage responses of cells (Beaudenon et al., to MoLCell. Biol. 79, 6972-6979, 1999) and transcriptional control (Saleh et al., J.
Mol.Biol. 282, 933-946, 1998). Although the correct operation of the ubiquitinylation pathway in cells is most likely to play a significant role in ensuring appropriate signal transduction, thereby assisting in protecting against the development of hyperproliferative disorders, no clear mechanism of how ,this is homeostasis is maintained has emerged.
It is generally accepted that proteins having the HECT domain of E6-AP and related proteins (Huibregtse et al., Proc. Natl Acad. Sci. USA 92, 2563-2567.
1995; Schwartz ef al., J. Biol. Chem 273, 12148-12154, 1998) form a sub-class of 2o ubiquitin-protein ligases (E3 enzymes) that- are involved in the ubiquitinylation cascade that catalyzes the covalent attachment of ubiquitin to a substrate protein, thereby targeting the substrate protein for proteolytic degradation. Unlike the ubiquitin ligases that comprise a RING domain, the enzymes comprising a HECT
domain reversibly bind ubiquitin via a conserved cysteine residue within the HECT
2s domain, and directly transfer ubiquitin to the substrate protein (Scheffner et al., Nature 373, 81-83, 1995).
Based upon the presence of a carboxyl terminal HECT domain, a progestin-induced gene that is linked to map position 8q22.3 of the human genome 30 (E~ereinafter "Edd") has attracted some interest as a human E3 enzyme (Callaghan ef al., Oncogene 17, 3479-3491, 1998). However, the precise cellular function of the protein (EDD) encoded by the Edd gene has not been determined.
The biochemical properties of in vitro translated EDD protein provide evidence s . that it is a human E3 enzyme (Callaghan ef al, Oncogene 17, 3479-3491, 1998) however there are no defined cellular substrates for the protein. The sub-cellular location of the EDD protein is also unknown. Notwithstanding that the progestin-responsiveness of Edd gene expression indicates a general role for EDD protein in progestin-mediated signal transduction pathways or progestin-responsive io tumorigenesis, no clear indication of specific proteins that bind to EDD in such pathways has emerged. Nor has there been any indication of specific expression patterns of the Edd gene, or mutations at the Edd locus, that might be involved in mediating such effects. Nor has the Edd gene been implicated in non-progestin mediated cellular efFects.
~s SUMMARY OF THE INVENTION
In work leading up to the present invention, the inventors sought to elucidate the role of the EDD protein by looking at the expression patterns of the Edd gene in various tumors, and by determining substrates for the EDD protein in cells, and by 2o targeting Edd gene expression in a murine model and by using inhibitory RNA
approaches in human cell lines. Surprisingly, the inventors found that elevated Edd gene expression is associated with tumorigenesis in progestin-responsive and progestin-non-responsive tumorigenesis, and that, by reducing or inhibiting Edd gene expression, cellular is inhibited. Accompanying this effect on 2s proliferation is an effect on apoptosis. Effects on cell proliferation and apototic effects on cells as mediated by EDD are closely linked. These data indicate a general utility for the Edd gene and EDD protein in identifying aberrant cell cycle regulation, and modulating the cell cycle, such as, for example, in the treatment of hyperproliferative disorders.
The inventors have also identified many nuclear protein substrates of the EDD
.

protein using yeast two hybrid screens which are involved in progesterone-mediated signal transduction, tumor suppression or DNA damage. Given the key role of the Edd gene expression in the control of cellular proliferation, these data provide highly specific means for identifying aberrant cell cycle regulation and s downstream efFects such as hyperproliferative disorders. The protein-protein interactions also provide highly specific means for determining compounds that modulate the cell cycle.
Accordingly, one aspect of this invention provides methods for detecting a cancer io cell in a subject, said method comprising determining the level of nucleic acid that is linked to map position 8q22.3 of the human genome or an expression product thereof in a sample of said subject, wherein elevated levels of said nucleic acid or said polypeptide are indicative of cancer in the subject.
is In one embodiment, the present invention provides a method for detecting a cancer cell in a subject, said method comprising:
(i) determining the level of nucleic acid linked to map position 8q22.3 of the human genome in a test sample from said subject; and (ii) comparing the level of the nucleic acid at (i) to the level of the nucleic acid 2o in a reference sample from a healthy or normal individual, wherein a level of the nucleic acid at (i) that is enhanced in the test sample relative to the reference sample from the normal or healthy individual is indicative of the presence of a cancer cell in said subject.
2s In an alternative embodiment the invention provides a method for detecting allelic imbalance in a region of the human genome comprising hybridizing a nucleic acid probe or primer to genomic DNA and detecting the hybridization, wherein the probe or primer comprises a nucleotide sequence selected from the group consisting of:
30 (i) the sequence set forth in SEQ ID NO: 5;
(ii) the sequence set forth in SEQ ID NO: 6;

_g_ (iii) the sequence set forth in SEQ ID NO: 7;
(iv) the sequence set forth in SEQ ID NO: 24;
(v) the sequence set forth in SEQ ID NO: 25; and (vi) the sequence of a nucleic acid fragment produced by amplification using s (vi) and (vii) as amplification primers in PCR.
In another embodiment, the present invention provides a method for detecting a cancer cell in a subject, said method comprising:
(i) determining the level of mRNA encoded by nucleic acid linked to map to position 8q22.3 of the human genome that is expressed in a test sample from said subject; and (ii) comparing the level of the mRNA determined at (i) to the level of mRNA
encoded by nucleic acid linked to map position 8q22.3 of the human genome that is expressed in a reference sample from a healthy or normal Is individual, wherein a level of the mRNA at (i) that is enhanced in the test sample relative to the reference sample from the normal or healthy individual is indicative of the presence of a cancer cell in said subject.
2o In another embodiment, the present invention provides a method for diagnosing a cancer or predicting recurrence of a cancer in a subject comprising determining the level of mRNA or protein encoded by nucleic acid linked to map position 8q22.3 of the human genome in a sample of said subject, wherein an elevated level of said mRNA or protein is indicative of relapse of a cancer in said subject.
In one embodiment, the mRNA encoded by a nucleic acid linked to map position 8q22.3 of the human genome encodes an EDD protein. Preferably, the mRNA
comprises the sequence set forth in SEQ ID NO: 1 or SEQ ID NO: 3 or a fragment thereof. Even more preferably, the mRNA encodes a protein comprising the 3o sequence set forth in SEQ ID NO: 2 or SEQ ID NO: 4 or a fragement thereof.

In one embodiment the protein encoded by a nucleic acid linked to map position 8q22.3 of the human genome is an EDD protein. Preferably the protein encoded by a nucleic acid linked to map position 8q22.3 of the human genome comprises the sequence set forth in SEQ ID NO: 2 or SEQ ID NO: 4 or a fragment thereof.
s A further aspect of the present invention relates to novel nucleic acid probes for detecting a cancer in accordance with the embodiments described herein.
A further aspect of the present invention provides an isolated or recombinant to protein complex comprising:
(i) an EDD protein or a portion of an EDD protein sufficient to bind to a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting is protein, a progesterone receptor protein, and a protein associated with vascularization; and (ii) a nuclear protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, 2o a nuclear targeting protein, a progesterone receptor protein, and a protein associated with vascularization or a portion of said protein sufficient to bind to said EDD protein or said portion of an EDD
protein.
2s Additional embodiments of the present invention provides isolated peptides, polypeptides, antibodies and other ligands that bind to an EDD-containing protein complex of the invention, and kits comprising same for producing the protein complex, or for identifying a modulator of a biological interaction between EDD or a portion of EDD and one or more other polypeptides selected from the group 3o consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, a progesterone receptor protein, and a protein associated with vascularization, or a portion thereof.
Another embodiment of the present invention provides methods for isolating a s EDD binding protein or a complex comprising same from a suitable cellular source.
In another embodiment, the invention provides methods for producing a protein complex described herein by recombinant means. For expressing peptides or ~o polypeptides by recombinant means, a protein-encoding nucleotide sequence is placed in operable connection with a promoter or other regulatory sequence capable of regulating expression in a cell-free system or cellular system.
Another embodiment of the present invention provides prognostic and diagnostic is methods for determining a predisposition for disease, or a disease state, said methods comprising detecting a protein complex comprising:
(i) an EDD protein; and (ii) a nuclear protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle ao modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, a progesterone receptor protein, and a protein associated with vascularization.
A further embodiment of the present invention provides methods for determining a 2s modulator of the activity, formation or stability of an isolated or recombinant protein complex comprising:
(i) an EDD protein or a portion of an EDD protein sufficient to bind to a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a so protein associated with DNA repair or damage, a nuclear targeting protein, a progesterone receptor protein, and a protein associated with vascularization; and (ii) a nuclear protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, s a nuclear targeting protein, a progesterone receptor protein, and a protein associated with vascularization or a portion of said protein sufficient to bind to said EDD protein or said portion of an EDD
protein.
to In another embodiment, the present invention provides a method for treating a condition associated with elevated expression of an EDD protein in a cell, said method comprising administering an amount of a compound effective to reduce the level of EDD expression or the level of an EDD expression product (e.g. a protein complex comprising an EDD protein) in a cell.
is In yet another embodiment, this invention relates to a variant of a gene that is linked to map position 8q22.3 of the human genome and to novel reagents and methods for detecting said variant.
2o BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1. A. Allelic imbalance in several cancer types showing AI at the EDD
locus (8q22.3) but not extending continuously to 8q24. Key: ~ allelic imbalance, probable allelic imbalance, Oheterozygote, uninformative homozygote, gap denotes no data available. Position of polymorphic microsatellites were 2s identified from the Genome Data Base (GDB) (http://gdbwww.gdb.org/).
Microsatellites CEDD and 586F18b are encoded in introns of EDD. The EDD
gene is located at 8q22.3 (Callaghan et al, Oncogene 74, 3479-3491, 1998) and MYC at 8q24.12 (GDB).
B. Microsatellite analysis of allelic imbalance from normal hepatocellular tissue 3o and hepatocellular carcinoma from patient 8 (Figure 1A). Allelic imbalance was seen for microsatellites D8S326, CEDD, D8S545 and D8S85. Arrows indicate significant increase (>30°l°) in proportion of allele indicated.
Figure 2. Allelic imbalance in ovarian cancers on chromosome 8q22.3-23.3.
Cancers are grouped according to histopathology. Overlap indicates mixed s histology. 'Other' comprises 4 adenomas and a germ cell tumor (case 124).
Key: t allelic imbalance, Oheterozygote, uninformative homozygote, gap denotes no data available.
Figure 3. mRNA expression of EDD in breast cancers, normal breast tissue and to breast epithelial cell lines.
(A) Relative mRNA expression of EDD in 41 breast cancers and 14 normal breast samples determined by quantitative RT-PCR. Horizontal line depicts upper limit of normal tissue expression. Insert: Comparison of EDD mRNA expression in 14 breast cancer samples and matched normal breast tissue determined by is quantitative RT-PCR.
(B) Expression of EDD mRNA (solid bars) and p53 R2 mRNA (open bars) in breast epithelial cell lines determined by quantitative RT-PCR. Gene copy number (indicated above bars) was determined by quantitative PCR. EDD genomic copy number was determined in MDA-MB-436, MDA-MB-468, BT-20 and BT-483 by 2o FISH (indicated in brackets). 184 is a normal breast epithelial cell lines while all others' are breast cancer cell lines (Sutherland et al, Human Cell Culture Vol. II, 79-106, 1999).
Figure 4 EDD protein expression in breast and ovarian cancers.
2s Tissues were stained with a polyclonal EDD antibody and counterstained with haematoxylin.
(A) negative control, neural tissue from an EDD-null mouse embryo (neural epithelium [NE], neural mesenchyme [NM]).
(B) positive control, neural tissue from wildtype mouse embryo. (C) normal human 3o breast duct.
(D) human breast carcinoma with high EDD expression.

(E) human serous ovarian cancer with low EDD expression.
(F) human serous ovarian cancer with high EDD expression.
Figure 5. Structural features of the EDD sequence.
s (A) Schematic diagram of EDD and its derivatives used for mammalian expression, yeast two hybrid analysis or in vitro translation. The UBA domain, three putative nuclear localisation sequences (NLS), a HECT domain and domains with homology to N-recognin zinc finger (zf UBR1 ) or the carboxy region of polyA-binding protein (PABP-C) are indicated. The positions of potential steroid ~o receptor binding motifs (LXXLL) are indicated by asterisks. Numbers indicate amino acid positions of fragment breakpoints. The conserved cysteine within the HECT domain (Cys 2768) is mutated to alanine (X) in fragments EDDM, EDD3M
and EDDSM.
(B) Potential zinc finger in EDD protein. A cysteine-rich domain shows similarity to is D.melanogaster Calossin (dCALO) and Arabidopsis thaliana BlG proteins and has a similar arrangement of conserved cysteine and histidine residues (boxed) as the zinc finger region first identified in N-recognin. N-recognin sequences shown are from yeast (scUBR1 ) and mouse (mUBR1 ). Identical residues are designated by dark shading and conservative substitutions by lighter shading.

Figure 6. EDD interacts with importin a5 via two NLSs.
(A) Interaction of EDD with importin a5 in a yeast two-hybrid assay. The entire coding sequence of EDD was fused in-frame with the yeast GAL4 DBD. This construct or control vector pAS2.1 was co-expressed with either control vector 2s (pACT2) or the GAL4 AD-importin a constructs encoding as 1-538 (Impa) or 538 (Impa-C) in diploid yeast strain CG1945/Y187. Protein extracts were prepared from cultures of 6 independent colonies and assayed in duplicate for ~3-galactosidase activity (expressed as fold increase over pAS2.1 vector control).
(B-C) In vitro interaction of importin a, with EDD and mapping of interaction.
In 3o vitro translated 35S-labelled EDD (B) or EDD fragments (C) were incubated with a purified GST-importin a5 fusion protein or with GST alone bound to glutathione-Sepharose beads, washed and analysed by SDS-PAGE and autoradiography.
(D) Interaction of EDD with importin ,ain HEK 293 and T-47D cells. HEK 293 cells were stably transfected with a plasmid encoding full length EDD protein s (2931EDD). Extracts from these cells or T-47D cells were subjected to immunoprecipitation with anti-importin a5 antibody (middle panel) or incubated with either GST or GST-importin a5 fusion protein bound to glutathione-Sepharose beads (right panel). Bound proteins from both procedures were separated by SDS-PAGE and western blotted for EDD.
io (E) Mapping interaction between importin a and individual NLSs of EDD. In vitro translated 35S-labelled EDD derivatives from the N-terminal region were incubated with GST-importin a5 or with GST alone bound to glutathione-Sepharose beads. Bound EDD was detected by SDSPAGE and autoradiography.
(F) In vitro interaction of importin ~3 with EDD. In vitro translated 35S-labelled is EDD and derivates were incubated with GST-importin ~ fusion protein or with GST
alone bound to glutathione-Sepharose beads. Bound EDD was detected by SDS-PAGE and autoradiography. Amounts of bound EDD relative to input are indicated as percentages below.
2o Figure 7. EDD is a nuclear protein.
(A) Subcellular localisation of EDD-GFP in mammalian cell lines (x40 magnification). EDD cDNA was fused to the amino-terminus of green fluorescent protein (GFP) and transiently transfected into HEK 293 (left panel) or MCF-7 cells (right panel). Transfected cells are indicated by arrowheads on bright field images.
2s While diffuse cellular staining was observed with GFP alone, strong nuclear GFP
fluorescence was observed for EDD-GFP in both cell lines.
(B) Immunostaining of cells with EDD antibody (x40 magnification). Nuclear staining seen for endogenous EDD in HEK 293 cells (left panel) is more intense in HEK 293 cells which overexpress EDD protein (WT30, right panel).

Figure 8. EDD interacts with PR-B.
(A) Interaction of EDD with PR in T-47D cells. Extracts from T-47D cells were incubated with either GST, GST-PR(AB) or GST-PR(CDE) fusion proteins bound to glutathione-Sepharose beads. Bound proteins were separated by SDS-PAGE
s and western blotted for EDD.
(B) Mapping interaction between PR and EDD. In vitro translated 35S-labelled EDD derivatives or SRC-1 were incubated with GST-PR(AB) and GST(CDE) fusion protein or with GST alone bound to glutathione-Sepharose beads. PR-bound EDD fragments were analysed by SDSPAGE and autoradiography.
to Amounts of bound EDD or SRC-1 relative to inputs are indicated as percentages below.
(C) Fine mapping of the interaction between PR(CDE) and the N-terminal region of EDD. In vitro translated 35S-labelled EDD derivatives from the N-terminal region were incubated with GSTPR(GDE) fusion protein or with GST alone bound is to glutathione-Sepharose beads. PR(CDE)-bound EDD fragments were analysed by SDS-PAGE and autoradiography.
Figure 9. Enhancement of nuclear receptor transactivation activity by EDD.
Luciferase activity was corrected for cell number and transfection efficiency where 2o appropriate (see methods) and graphed relative to-the value for liganded receptor alone, which was set at 100%.
(A) EDD enhances PR B transactivation activity. Reporter assays were carried out using either HEK 293 (left ) or COS7 (right) cells in the presence of EDD, SRC1 or empty vector, transfection control plasmid (pGFP20) and either 1 nM of 2s the synthetic progestin ORG2058 or equivalent ethanol vehicle (EtOH).
(B) Mutation of the catalytic cysteine of EDD does not alter the effect of EDD
on PR transactivation. Reporter assays were carried out using HEK 293 cells in the presence of EDD, EDDM or empty vector and 10nM ORG2058.
(C) EDD enhances PR reporter gene expression in a dose dependent manner.
3o HEK 293 cells were transfected for a standard reporter assay along with increasing amounts of a constitutive expression vector for either EDD or empty vector (0) in the presence of 1 nM ORG2058. The amount of DNA transfected was normalised to 1.2~,g with empty vector. Cell number was monitored using proliferation assay and transfection efficiency by co-transfection with pRLTK
followed by Renilla luciferase assay.
s (D) Effect of EDD on response to the synthetic progestin ORG2058. HEK 293 cells were transfected for reporter assay along with a transfection control plasmid (pRL-TK). Cells were harvested for luciferase assay following 24h treatment with increasing concentrations of ORG2058.
(E) Enhancement of VDR reporter gene expression by EDD. HEK 293 cells to were transfected with a constitutive expression vector for VDR and a VDRE-containing luciferase reporter vector along with a constitutive expression vector for EDD or empty vector and a transfection control plasmid (pRL-TK). Cells were harvested for luciferase assay following 24h treatment with lOnM 1.25-dihydroxyvitamin D3.
~s (F) EDD does not enhance ER reporter gene expression. HEK 293 cells were transfected with a constitutive expression vector for ER and an ERE-containing luciferase reporter vector along with either a constitutive expression vector for EDD, SRC1 or empty vector and a transfection control plasmid (pGFP20). Cells were harvested for luciferase assay following 24h treatment 2o with 100nM 173-estradiol.
Figure 10. Interaction of EDD and CIB and the effect of DNA damage.
(A) Interaction of EDD with calcium-integrin binding protein (CIB) in a yeast two-hybrid assay. The entire coding sequence of EDD was fused in-frame with the 2s yeast GAL4 DBD. This construct or control vector pAS2.1 was co-expressed with either control vector (pACT2) or GAL4 AD-CIB in diploid yeast strain CG 1945/Y187.
(B) In vitro interaction of CIB with EDD. In vitro translated 35S-labelled EDD
was incubated with GST-CIB fusion protein or with GST alone bound to glutathione-3o Sepharose beads. Bound EDD was analysed by SDS-PAGE and autoradiography.

(C) Mapping interaction between CIB and EDD. In vitro translated 35S-labelled EDD derivatives were incubated with GST-CIB fusion or with GST alone bound to glutathione-Sepharose beads. Bound EDD fragments were analysed by SDS-PAGE and autoradiography.
s (D) Interaction of EDD and CIB in HEK 293 and MCF-7 cells. Left, HEK 293 cells overexpressing mutant EDD were transfected with a plasmid encoding Flag-tagged CIB or empty vector (vec). Extracts from these cells were subjected to immunoprecipitation using anti-FLAG Ab M2. Right, nuclear extracts prepared from MCF-7 cells following treatment with DNA damage agents phleomycin to (Phleo) or hydroxyurea (HU) were incubated with either GST or GST-CIB
fusion protein bound to glutathione-Sepharose beads. Bound proteins from both procedures were analysed by SDS-PAGE and western blotted for EDD and amounts bound are indicated as percentages relative to input.
(E) Potential regulation of CIB by the proteasome. HEK 293 cells were treated is with the proteasome inhibitor MG132 (20~,M) or vehicle (DMSO) for six hours and whole cell extracts analysed by SDS PAGE. Proteins were transferred to nitrocellulose and western blotted for importin a5, CIB and the proteasomal target protein, p27.
2o Figure 11 (A) Schematic diagram of EDD and chk2 and their derivatives used in binding assays. For EDD, the UBA (ubiquitin-associated) domain, three putative nuclear localisation sequences (NLS), a HECT (homologous to E6-AP carboxy terminus) domain and domains with homology to N-recognin zinc finger (zf UBR1 ) or the 2s carboxy region of polyA-binding protein (PABP-C) are indicated. The positions of potential steroid receptor binding motifs (LXXLL) are indicated by asterisks.
Numbers indicate amino acid positions of fragment breakpoints. The conserved cysteine within the HECT domain (Cys 2768) is mutated to alanine (X) in fragments EDDM and EDD3M. For chk2, the SQ/TQ domain rich in ATM family 3o kinase sites, the forkhead associated (FHA) domain and kinase domain are indicated along with key residues for chk2 function. Domain boundaries are also indicated as numbers below the diagram. Also indicated is the fragment of chk2 used in pull down assays (GSTchk2-N).
(B) EDD interacts with chk2-N in nuclear extracts. Nuclear extracts from MCF-7 cells were incubated with either GST or GSTchk2-N fusion protein bound to s glutathione-Sepharose beads. Following extensive washing, bound proteins were separated by SDS-PAGE and western blotted for EDD.
(C) EDD and chk2 associate in vivo. Cell lysates from HEK 293 or MCF-7 cells were subjected to immunoprecipitation with polyclonal chk2 antibody (N17).
Following precipitation of immune complexes and extensive washing, bound io proteins were separated by SDS-PAGE and western blotted for EDD.
Figure 12 EDD interacts with chk2 FHA domain.
(A) In vitro translated 35S-labelled EDD derivative EDDFS (aa 889-2799) was is incubated with GSTchk2-N or GSTchk2-N derivatives containing R117A or 1157T
substitutions, or with GST alone bound to glutathione-Sepharose beads. Bound EDD was detected by SDS-PAGE and autoradiography.
(B) Nuclear extracts from MCF-7 cells were incubated with GSTchk2-N or a GSTchk2-N derivative containing an R117A substitution, or with GST alone bound 2o to glutathione-Sepharose beads. Bound proteins were separated by SDS-PAGE
and western blotted for EDD.
(C) EDD is phosphorylated in vivo. HEK293 cells stably transfected with Ecdysone receptor were transfected with a vector for inducible expression of Flag-tagged EDD. Expression of EDD was induced (+) by addition of ponasterone for 2s 24 h. Medium was replaced with phosphate free medium containing 32P-labelled orthophosphate and labelling of cellular proteins allowed to proceed. Lysates were made from cells expressing (+) or not expressing (-) Flag-tagged EDD and immunoprecipitation carried out using anti-Flag antisera. Radiolabelled (ie phosphorylated) EDD was detected by SDS-PAGE and autoradiography (upper 3o panel). In the lower panel, HEK293 or MCF-7 extracts were incubated in the presence (+) or absence (-) of lambda protein phosphatase. Samples were separated by SDS-PAGE on 4.2% gels and western blotted for EDD. Removal of phosphates was indicated by a shift in the mobility of the EDD protein.
(D) The phosphopeptide-binding interface of the chk2 FHA domain is required for EDD binding. In vitro translated 35S-labelled EDD derivative EDDFS (aa 889 s 2799) was incubated with GSTchk2-N or GSTchk2-N derivatives containing R117A or 1157T substitutions, or with GST alone bound to glutathione-Sepharose beads. The incubations with GSTchk2-N or GSTchk2-N(1157T) were carried out in the presence or absence of an FHA-binding phosphopeptide at 0, 80 or 150 pM.
Bound EDD was detected by SDS-PAGE and autoradiography.
~o Figure 13 Mapping the interaction between EDD and chk2.
(A) In vitro translated 35S-labelled EDD derivatives were incubated with GSTchk2-N or with GST alone bound to glutathione-Sepharose beads. Bound Is EDD was detected by SDS-PAGE and autoradiography.
(B) In vitro translated 35S-labelled EDD derivatives of EDDF5 (EDDF6 and EDDF7) were incubated with GSTchk2-N or GSTchk2-N derivatives containing R117A or 1157T substitutions, or with GST alone bound to glutathione-Sepharose beads. Bound EDD was detected by SDS-PAGE and autoradiography. Fusion 2o protein additions were monitored by blotting for GST.
Figure 14 (A) Nuclear EDD protein levels and CHK2 T68 phosphorylation before and after irradiation. MCF-7 cells were exposed to 12Gy IR and allowed to recover for 2s 1, 4 or 12 h. Nuclear extracts were prepared from control and treated cells and analysed by western blotting for EDD, phosphorylated CHK2 (P-T68) or pRB
(loading control).
(B) Decreased association between EDD and CHK2 following radiomimetic treatment of cells. Nuclear extracts from MCF-7 cells that had been cultured in the 3o presence of the radiomimetic phleomycin (100 ~g per mf) or from control MCF-cells were immunoprecipitated with polyclonal CHK2 antibody (N17) or incubated with goat IgG as a negative control. Following precipitation of immune complexes and extensive washing, bound proteins were separated by SDS-PAGE and western blotted for EDD or CHK2.
(C) Nuclear extracts from MCF-7 cells that had been cultured in the presence s of phleomycin (100 pg per ml) or from control MCF-7 cells were incubated with GSTCHK2-N or with GST alone bound to glutathione-Sepharose beads. Bound proteins were separated by SDS-PAGE and immunoblotted for EDD and GST.
(D) Nuclear extracts from MCF-7 cells that had been cultured in the presence of phleomycin (100 pg per ml) for 1 h followed by 1 h or 3h recovery, or from control to MCF-7 cells, were incubated with CHK2 antibody (N17). Following precipitation of immune complexes and extensive washing, bound proteins were separated by SDS-PAGE and western blotted for EDD or CHK2.
(E) Dissociation of EDD and CHK2 is not dependent on ATM kinase.
Nuclear extracts from MCF-7 cells that had been cultured for 3h in the presence of is the radiomimetic phleomycin (100 Ng per ml) or from control MCF-7 cells, in the presence or absence of wortmannin (10pM), were incubated with GSTCHK2-N or with GST alone bound to glutathione-Sepharose beads. Bound proteins were separated by SDS-PAGE and immunoblotted for EDD and GST. Input lysates were immunoblotted for EDD and active CHK2 (anti P-T68).
ao (F) Immunoprecipitation with CHK2 antibody from nuclear extracts of cells deficient in ATM (FTpEBS7) and a matched ATM-complemented cell line (FTYZS). Cells were incubated in the presence or absence of phleomycin (20 pg per ml) for 4 hours, ensuring T68 phosphorylation in an ATM-dependent fashion (lower panel).
2s Figure 15 (A) MCF-7 cells were transfected with short interfering RNAs directed against GFP or EDD for 96 h. Cells were then exposed to 12 Gy ionizing radiation (IR) and harvested following 90 min recovery at 37°C. Equal amounts of protein from 3o whole cell lysates were separated by SDS-PAGE and analysed by immunoblotting for EDD, activated CHK2 (P-T68) and total CHK2.

(B) Kinase assay of CHK2 immunoprecipitated from irradiated cell lysates using GST-cdc25C subfragment as a substrate. MCF7 control cells or cells depleted of EDD (EDD siRNA) were subjected to 4 Gy ionising radiation (IR) and allowed to recover for 15 or 60 minutes before harvesting. Incorporation of 32-P
s into substrate or CHK2 was detected by autoradiography. The amounts of immunoprecipitated CHK2 and the efFiciency of phosphorylation of CHK2 on threonine 68 were monitored by western blotting.
Figure 16 to The phosphopeptide-binding interface of the CHK2 FHA domain is required for EDD binding.
(A) CHK2 FHA domain binds EDD. In vitro translated (IVT) 35S-labelled EDD
derivatives EDDF5 (aa 889-2799) and EDDF6 (aa 889-2526) (top panel) or MCF7 whole cell extracts (lower panel) were incubated with GSTCHK2-N (WT) or is GSTCHK2-N derivatives containing R117A or 1157T substitutions, or with GST
alone bound to glutathione-Sepharose beads. Bound EDD was detected by SDS-PAGE and autoradiography (top panel) or western blotting (lower panel). Fusion protein additions were monitored by Coomassie staining.
(B) Dephosphorylation of EDD prevents CHK2 association. Pull downs of IVT
2o EDD were performed as described in (A) except that where indicated IVT EDD
was incubated with lambda protein phosphatase (PPase) prior to incubation with the GSTCHK2-N fusion.
(C) In vitro translated 35S-labelled EDD derivatives EDDF5 and EDDF6 were incubated with GSTCHK2-N (WT) or GSTCHK2-N (1157T) bound to glutathione-2s Sepharose beads in the presence or absence of FHA-binding phosphopeptide at 0, 20, 50 or 100 ~M. Bound EDD was detected by SDS-PAGE and autoradiography.
3o Figure 17 (A) EDD and BRCA2 interact in cells. Extracts from HEK293 cells expressing Histagged EDD were subjected to immunoprecipitation with EDD antibody (AbPEP1 ). Following precipitation of immune complexes and extensive washing, bound proteins were separated by SDS-PAGE and western blotted for EDD (left panel) and BRCA2 (right panel).
s (B) interactions between EDD, BRCA2 and chk2 are modified in response to DNA
damaging agents. Nuclear extracts from MCF-7 cells that had been cultured in the presence of the radiomimetic phleomycin or the UV mimic hydroxyurea (HU), or from control MCF-7 cells, were subjected to immunoprecipitation with polyclonal chk2 antibody (N17). Following precipitation of immune complexes and extensive ~o washing, bound proteins were separated by SDS-PAGE and western blotted for EDD, BRCA2 or chk2.
Figure 18. Targeted disruption of mouse Edd.
is (A) Structure of the wild-type Edd locus (top), targeting vector (middle) and the mutated locus following homologous recombination (bottom). Edd exon 1 is indicated as a black box with the position of the ATG codon shown. BamH1 restriction sites are denoted by B. Genotyping was performed by PCR using primers, 1, 2 and 3, (arrowheads), and by Southern blot analysis with a 3' probe 2o as shown (probe). Expected sizes of PCR products and BamHl fragments that hybridise with the probe on Southern analysis are indicated.
(B) Southern blot and PCR analysis of genomic DNA from targeted ES cell clones.
Genomic DNA from four neomycin resistant ES cell clones, 1 B2, 3D5, 5C6 and 8E7, was digested with BamHl and hybridised with the 3' probe. The 6.Okb 2s fragment corresponding to the wild-type (WT) allele (top) and the 4.2kb fragment corresponding to the mutated (KO) allele (bottom) are indicated. Right panel:
PCR
genotype analysis of E10.5 embryos. DNA samples were subjected to PCR using the primers 1, 2 and 3. PCR amplification of the WT allele by primers 1 and 2 produces a 600bp fragment (top) and of the KO allele by primers 2 and 3 produces a 440bp fragment (bottom).
(C) Northern Blot analysis of tissues from WT and Edd+~ mice. Total RNA was s extracted and hybridised to a 32P-labelled mouse Edd cDNA probe.
Densitometry was performed and values corrected for Loading by comparison to the blot after reprobing with GAPDH cDNA.
(D) Western blot analysis and IHC showing EDD expression in wild-type (+/+), heterozygote (+/-) and knockout (-/-) tissues. EDD western blots were performed to on lysates from testes of adult WT (+/+) and Edd~~- (+l-) mice and from E10.5 WT, Edd'~~- and Edd-~- (-/-) embryos.
(E) Immunohistochemistry was performed on E9.5 wild-type (left panel) and E9.5 knockout (-/-, right panel) embryo sections with a region of neural epithelium shown. Immunohistochemstry was performed with an anti-EDD antibody.
is Figure 19. Morphology of wild-type and knockout (Eddy) embryos at E7.5 -E10.5.
Photographs showing freshly dissected WT embryos (+/+) and Edd-~ embryos (-/-). As early as E7.5, Eddy embryos display slightly delayed development while by 2o E8.5, clear growth retardation is obvious in Eddy embryos compared to WT.
From E9.5 onwards the most obvious developmental defect is the small size of Eddy embryos and the absence of turning which occurs in WT embryos around E9. In addition, many Eddy embryos display a bulbous allantois (a) indicating failure of placentation. Many Edd' embryos were also observed with pericardial effusion (p).
2s Figure 20. Edd expression in developing mouse embryo. Immunohistochemical analysis of Edd expression in wt d10.5 embryo. Inset shows predominantly nuclear expression under higher magnification.
Figure 21. Cell proliferation in WT and Eddy embryos at E8.5 - E10.5.
s (A) BrdU incorporation was used to measure cell proliferation in WT (+/+) and Edd (-/-) embryos and BrdU positive cells were detected by immunohistochemistry.
Cells positive for BrdU are darkly stained, (B) Quantification of prolioferating cells in Eddy embryos at E8.5 - E10.5.
While there are equivalent numbers of proliferating cells between Eddy and WT
embryos to at E8.5, by E9.5 the majority of cells in Eddy embryos have stopped proliferating and do not incorporate BrdU. Graph shows mean number of proliferating cells (%total ~ standard error).
Figure 22. Apoptosis in WT and Eddy embryos at E8.5 - E10.5.
is (A) Apoptosis was measured in embryo sections using the TUNEL assay with TUNEL positive nuclei stained brown. At E8.5, similar levels of staining can be seen in both WT (+/+) and Edd' (-/-) embryos. At E9.5 and 10.5, Edd' embryos show dramatically increased levels of TUNEL staining compared to WT embryos which have very few TUNEL stained cells.
20 (B) Active Caspase-3 staining was also performed on embryo sections to confirm TUNEL results. Percentage of caspase positive cells are shown in the corner of each panel. Similarly to TUNEL results, levels of staining in WT (+/+) and Eddy embryos (-/-) at E8.5 are very similar. However, by E9.5 a large increase in the level of caspase-3 mediated apoptosis is observed in Edd ~ embryos.
Figure 23. Defective vascufarisation in Edd' yolk sacs. (A) Morphology of E9.5 E10.5 WT (+/+) and Eddy (-l-) yolk sacs. Eddy yolk sacs display significantly less vascularisation than their WT littermates suggesting a defect in yolk sac circulation. In addition blood pooling can be seen within Eddy embryos at E10.5 (b);
(B) High magnification view of Eddy and WT yolk sacs at E10.5. WT yolk sacs contain large and well organised vessels (v) however, while Eddy yolk sacs do contain some vessels (v), these appear small and disorganised suggesting defective vascular development in the absence of EDD.
s Figure 24. High magnification view of histological sections from WT and Edd-~ yolk sacs at E9.5. Distinct vascular channels containing blood cells (b) are visible in WT yolk sac, whereas EDD-null yolk sacs display enlarged channels with unusual separation of mesoderm (m) and endoderm (e) and few blood cells.
io Figure 25. Western blot analysis of EDD protein expression after anti-EDD
(R) or anti-GFP (C) (control) small interfering RNA transfection in MCF-7 and HEK-293 cell lines. Time after transfection is shown in hours at the bottom of the figure.
Figure 26 Cell morphology after RNA interFerence. HMEC 184 cells are shown 5 is days after RNA interference (left) control cells and (right) cells transfected with EDD siRNA. Cell shape has altered after depletion of EDD, cells make fewer contacts and cell organisation is disturbed. (Original magnification 100x) Figure 27. Immunofluorescence microscopy of ~3-catenin in HMEC 184 cells.
2o Whereas ~i-catenin staining in control cells (left) was even along cell-cell contacts, cells transfected with EDD siRNA showed more patchy staining and decreased levels of ~-catenin (right). Furthermore, a reduction in the number of cell-cell contacts was observed in cells transfected with EDD siRNA.
2s Figure 28. Relocalization of ~i catenin after EDD RNAi transfection.
Immunofluorescence microscopy of ~3-catenin in HMEC 184 cells shows that EDD
RNAi causes ~-catenin to move from the cell periphery (left, control cells) to the cell nucleus (right).
3o Figure 29. Immunofluorescence microscopy of actin in HMEC 184 cells. Actin filaments (in control cells (left) were coordinated and continuously organised from cell to cell. Cells transfected with EDD siRNA (right) showed disorganised actin filaments, where the actin from one cell was not connected to the actin of its neighbouring cells.
DETAILED DESCRIPTION OF THE INVENTION
Nucleic acid-based diagnostics and novel nucleic acids One embodiment of this invention provides methods for detecting a cancer cell in a subject, said method comprising determining the level of nucleic acid that is to linked to map position 8q22.3 of the human genome or an expression product thereof in a sample of said subject, wherein elevated levels of said nucleic acid or said polypeptide are indicative of cancer in the subject.
In the present context, the term "cancer cell" includes any biological specimen or is sample comprising a cancer cell irrespective of its degree of isolation or purity, such as, for example, tissues, organs, cell lines, bodily fluids, or histology specimens that comprise a cell in the early stages of transformation or having been transformed. Bodily fluids shall be taken to include whole blood, serum, peripheral blood mononuclear cells (PBMC), or buffy coat fraction.
The isolated nucleic acid inked to map position 8q22.3 of the human genome or an expression product thereof is present at elevated levels in cancer cells compared to non-cancer cells.
2s The definition of "cancer cell" is not to be limited by the stage of a cancer in the subject from which said cancer cell is derived (ie. whether or not the patient is in remission or undergoing disease recurrence or whether or not the cancer is a primary tumor or the consequence of metastases). Nor is the term "cancer cell"
to be limited by the stage of the cell cycle of said cancer cell.
In one embodiment, the cancer cell is epithelial in origin (i.e. a carcinoma).

In another embodiment, the cancer cell is from a cancer selected from the group consisting of ovarian cancer, melanoma, metastatic melanoma, squamous cell carcinoma of the head and neck, squamous cell carcinoma of the tongue, s hepatocellular carcinoma, breast cancer, a metastases of ovarian cancer, a metastases of melanoma, a metastases of metastatic melanoma, a metastases of squamous cell carcinoma of the head and neck, a metastases of squamous cell carcinoma of the tongue, a metastases of hepatocellular carcinoma and a metastases of breast cancer.
io As used herein, the germ "ovarian cancer" shall be taken to refer any one or more of a number of cancers of epithelial origin, such as, for example, serous, mucinous, endometrioid, clear cell, papillary serous, Brenner cell or undifferentiated adenocarcinoma. The term "breast cancer" shall be taken to refer is to a ductal carcinoma or lobular carcinoma. The term "hepatocellular carcinoma"
will be understood to mean any carcinoma arising from the hepatocytes, as distinct from other types of hepatic cancer that may consist of liver metastases.
The terms "melanoma" and "squamous cell carcinoma" will be understood to be epithelial skin cancers of the melanocytes and squamous cells, respectively.
20 "Metastatic melanoma" is the most advanced stage of melanoma arising in the melanocytes and metastasizing in the lymph nodes and other organs of the body.
This embodiment of the invention is predicated on the finding by the present inventors that there is considerable allelic imbalance in several tumor types in the 2s vicinity of the region of the human genome that maps to position 8q22.3. In particular, allelic imbalance in this region of the human genome was found by the inventors in 14 of 37 tumors, said allelic imbalance comprising discrete regions of imbalance comprising 8q22.3 and more extensive 8q aberrations. Allelic imbalance was found by the inventors to be frequent in cases of ovarian cancer 30 (e.g. serous subtype), hepatocellular carcinoma, squamous cell carcinoma of the tongue, breast cancer and metastatic melanoma. Using quantitative RT-PCR, the inventors also found that the expression levels of EDD-encoding mRNA are frequently elevated in cancers such as breast cancer. In the case of breast cancer, elevated expression of the Edd gene was associated with amplification of the Edd locus. A closely linked gene in this region of the genome, i.e.
encoding s p53 ribonucleotide reductase (p53R2), was also shown by the inventors to be overexpressed and amplified in cancer cell lines. Accordingly, this embodiment of the invention is not limited to the detection of EDD-encoding nucleic acid.
As used herein, the term "map position 8q22.3 of the human genome" shall be io taken to refer to the region of the human genome that comprises a genomic gene encoding an EDD protein and preferably having a centromeric orientation on chromosome 8 with its first exon position at about 8q23. Those skilled in the art will be aware that a "genomic gene" includes both protein-encoding regions (i.e.
exons) and non-coding regions (i.e. 5'-upstream regulatory regions such as the is promoter and 5'-untranslated region, intervening sequences or introns, and 3'-untranslated region). Accordingly, a genomic gene encoding an EDD protein includes all such features and not merely the protein-encoding portion thereof.
By "EDD protein" is meant a polypeptide that comprises an amino acid sequence 2o having at least about 80% identity to the sequence- set forth herein as SEQ
ID
Nos: 2 or 4. For the purposes of nomenclature, the amino acid sequence set forth in SEQ ID NO: 2 relates to a first EDD protein encoded by the nucleotide sequence of SEQ ID NO: 1 as disclosed in published International Patent Application No. PCT/AU98/00280. The sequence set forth in SEQ ID NO: 4 2s relates to a novel EDD polypeptide encoded by a splice variant wherein 18 nucleotides of SEQ ID NO: 1 have been deleted. Accordingly, the amino acid sequence of SEQ ID No: 4 differs by the deletion of six amino acids comprising the sequence VLLLPL from SEQ ID NO: 2. Preferably, the percentage identity to SEQ ID NO: 2 or 4 is at least about 90%, more preferably at least about 95%, or 3o at least about 99%. In determining whether or not two amino acid sequences fall within these defined percentage identity limits, those skilled in the art will be aware that it is necessary to conduct a side-by-side comparison of amino acid sequences. In such comparisons or alignments, differences will arise in the positioning of non-identical amino acid residues depending upon the algorithm used to perform the alignment. In the present context, references to percentage s identities and similarities between two or more amino acid sequences shall be taken to refer to the number of identical and similar residues respectively, between said sequences as determined using any standard algorithm known to those skilled in the art. In particular, amino acid identities and similarities are calculated using the GAP program of the Computer Genetics Group, Inc., io University Research Park, Madison, Wisconsin, ~ United States of America (Devereaux et al, Nucl. Aeids Res. 72, 387-395,1984), which utilizes the algorithm of Needleman and Wunsch J. Mol. Biol. 48, 443-453, 1970, or alternatively, the CLUSTAL W algorithm of Thompson et al., Nucl. Acids Res. 22, 4673-4680, 1994, for multiple alignments, to maximize the number of identical/similar amino acids is and to minimize the number and/or length of sequence gaps in the alignment.
It will be apparent from the preceding description that nucleic acid that is "linked to" map position 8q22.3 of the human genome will therefore be sufficiently close to a genomic gene encoding an EDD protein for the frequency of recombination 2o between said nucleic acid and said genomic gene to indicate linkage. As will be known to those skilled in the art, the frequency of recombination between two markers on DNA will increase as the markers are spaced further apart until the association between the markers in a segregating population is random, at which point they are considered to be unlinked.
In one embodiment, the nucleic acid that is linked to map position 8q22.3 will be tightly linked such that there is only a low degree of recombination between the nucleic acid and the genomic gene encoding an EDD polypeptide. Preferably, the nucleic acid will itself map to a region of the human chromosome between 8q22.3 3o and 8q24.13, at least comprising the genomic Edd and p53R2 genes or a portion thereof.

Unless specifically stated otherwise, the expression product of any nucleic acid that is linked to map position 8q22.3 of the human genome is usefiul for the diagnosis of cancer within the context of the present invention, including the s expression products of an Edd gene, or the expression product of a p53R2 gene.
As used herein, the term "expression product" shall be taken to refer to any transcription product of a genomic gene, such as unprocessed or processed mRNA including a splice variant, or any translation product encoded by a genomic gene, such as a precursor polypeptide, processed polypeptide or a complex io involving said polypeptide. For example, a protein-protein complex or DNA-protein complex comprising an EDD polypeptide is clearly within the scope of the term "expression product of an Edd gene" or similar term as used herein.
Similarly, a protein-protein complex or DNA-protein complex comprising a p53R2 polypeptide is clearly within the scope of the term "expression product of an is p53R2 gene" or similar term as used herein.
It will be apparent from. the preceding discussion that the diagnostic methods provided by the present invention involve a degree of quantification to determine, on the one hand, the level of nucleic acid linked to map position 8q22.3 of the 2o human genome in tissue that is suspected of comprising a cancer cell, or, on the other hand, the level of an expression product of said nucleic acid. Such quantification can be readily provided by the inclusion of appropriate reference samples in the assays described below, derived from healthy or normal individuals.
Accordingly, in one embodiment, the reference sample comprises cells or tissue from the same subject taken at a time point when the individual was healthy or in remission from disease. In yet another embodiment, the reference sample comprises cells or tissues from a healthy or normal individual. In accordance with 3o both of these embodiments, the reference sample and the test sample are both processed, albeit not necessarily at the same time, and data obtained for both samples are compared.
Alternatively, if reference samples are not included in each assay conducted, the s reference sample may be derived from an established data set that has been generated from healthy or normal individuals. Accordingly, in one embodiment, the reference sample comprises data from a sample population study of healthy individuals, such as, for example, statistically significant data for the healthy range of the integer being tested. In accordance with this embodiment, the comparison io of reference and test sample is perFormed following the analysis of the test sample and comprises a comparison of data obtained for the test sample to data obtained for the sample population.
In the present context, the term "healthy individual" shall be taken to mean an is individual who is known not to suffer from cancer, such knowledge being derived from clinical data on the individual, including, but not limited to, a different cancer assay to that described herein. As the present invention is particularly useful for the early detection of cancer, it is preferred that the healthy individual is asymptomatic with respect to the early symptoms associated with a particular 2o cancer. In the case of ovarian cancer, early detection using well-known procedures is difficult, however reduced urinary frequency, rectal pressure, and abdominal bloating and swelling, are associated with the disease in its early stages, and, as a consequence, healthy individuals should not have any of these symptoms. It is also preferred for such "healthy subjects" to not have a large 2s number of risk factors associated with these diseases. Indicators of the early stages of primary breast cancer or a susceptibility for developing breast cancer include, for example, familial history, atypical hyperplasia, the occurrence of benign conditions of the breast, enhanced breast density particularly in women aged 45 years and older, radiation exposure, and abnormal breast appearance.
3o Indicators of the early stages of primary hepatocellular carcinoma or a.
susceptibility for the disease include, for example, liver cirrhosis, chronic hepatitis B or hepatitis C infection, a diet high in aflatoxins, adenomatous hyperplasia, abdominal (hepatic) pain or swelling, ascites, enlargement of the spleen, hemochromatosis, alpha-1-antitrypsin deficiency, glycogen storage disease, and tyrosinemia. . Indicators of the early stages of primary melanoma or squamous s cell carcinoma or a susceptibility for these diseases include, for example, changes to the appearance of the skin, especially in association with prolonged exposure to the sun or radiation damage, a history of smoking or chewing tobacco, actinic keratosis, and leukoplakia. Clearly, subjects suffering from later symptoms associated with any one or more of these cancers, such as, for example, to metastases in the skin, oral cavity, pharynx, omentum, abdomen, abdominal fluid, lymph nodes, lung, liver, brain, or bone, and subjects suffering from spinal cord compression, abdominal pain or swelling, elevated calcium level, elevated serum alpha-fetoprotein level, changes in BRCA1 or BRCA2 genes or gene expression, chronic pain, or pleural effusion, should also be avoided from the "healthy is individual" data set.
The term "normal individual" shall be taken to mean an individual having a normal level of an Edd gene expression product in a particular sample derived from said individual. As will be known to those skilled in the art, data obtained from a 2o sufficiently large sample of the population will normalize, allowing the generation of a data set for determining the average level of a particular parameter.
Accordingly, the level of expression of an Edd gene product can be determined for any population of individuals, and for any sample derived from said individual, for subsequent comparison to levels of the expression product determined for a 2s sample being assayed. Where such normalized data sets are relied upon, internal controls are preferably included in each assay conducted to control for variation.
In one embodiment, the present invention provides a method for detecting a cancer cell in a subject, said method comprising:
30 (i) determining the level of nucleic acid linked to map position 8q22.3 of the human genome in a test sample from said subject; and (ii) comparing the level of the nucleic acid at (i) to the level of the nucleic acid in a reference sample from a healthy or normal individual, wherein a level of the nucleic acid at (i) that is enhanced in the test sample relative to the reference sample from the normal or healthy individual is indicative s of the presence of a cancer cell in said subject.
Preferably, the sample comprises cells from a tissue or tissue selected from the group consisting of skin, an oral cavity tissue, breast, liver, spleen, ovary, prostate, kidney, uterus, placenta, cervix, omentum, rectum, brain, bone, lung, to lymph, urine, semen, blood, abdominal fluid, and serum. Cell preparations or nucleic acid preparation derived from such tissues or cells are not to be excluded.
The sample can be prepared on a solid matrix for histological analyses, or alternatively, in a suitable solution such as, for example, an extraction buffer or suspension buffer, and the present invention clearly extends to the testing of is biological solutions thus prepared.
Preferably, the nucleic acid that is determined according to this embodiment is genomic DNA. This embodiment of the invention is therefore particularly suited to determining allelic imbalance in this region of the human genome as a diagnostic 2o for cancer. -. - - - - -To determine the level of the nucleic acid, a variety protocols that are well known in the art can be utilized, such as, for example, in situ hybridization, microsatellite analysis, and microarray technology, such as, for example, using tissue 2s microarrays probed with nucleic acid probes, or nucleic acid microarrays lie.
genomic DNA microarrays or amplified DNA microarrays) probed with nucleic acid probes. All such assay formats are encompassed by the present invention. For high throughput screening of large numbers of samples, such as, for example, public health screening of subjects, particularly human subjects, having a higher 3o risk of developing cancer, microarray technology is a preferred assay format. As will be understood by those skilled in the art, nucleic acid hybridization-based or amplification-based approaches such as, for example, microsatellite analysis, are readily adaptable to microarray technology.
In a preferred embodiment, the level of nucleic acid is determined by hybridizing a s nucleic acid probe to genomic DNA encoding an EDD protein in the test sample under at least low stringency hybridization conditions and detecting the hybridization using a detection means. Similarly, the level of genomic DNA
encoding an EDD protein in the reference sample from the healthy or normal individual is preferably determined by hybridizing the probe to genomic EDD-to encoding DNA in said reference sample under at least low stringency hybridization conditions and detecting the hybridization using a detection means.
For nucleic acid hybridization-based approaches, shorter probes are hybridized at lower stringency hybridization (ie. reduced temperature and/or higher salt is concentration and/or higher detergent concentration) than longer nucleic acid probes. Generally, hybridization is carried out well below the calculated melting temperature (Tm) of a DNA duplex comprising the probe. For example, the oligonucleotide probes exemplified herein have calculated Tm values in the range of about 55°C to about 60°C, suggesting that hybridization involving such probes 2o should be carried out at a temperature in the range of ambient temperature to about 45°C, and more preferably between about 40°C to about 45°C (ie. low stringency to moderate stringency conditions). This contrasts with standard hybridization temperatures of about 65°C for nucleic acid probes of about 100 nucleotides or longer (ie. moderate to high stringency hybridization conditions).
2s For the purposes of defining the level of stringency to be used in these diagnostic assays, a low stringency is defined herein as being a hybridization and/or a wash carried out in 6xSSC buffer, 0.1 °!° (w/v) SDS at 28°C, or equivalent conditions. A
moderate stringency is defined herein as being a hybridization and/or washing 3o carried out in 2xSSC buffer, 0.1 % (w/v) SDS at a temperature in the range 45°C to 65°C, or equivalent conditions. A high stringency is defined herein as being a -3s-hybridization and/or wash carried out in 0.lxSSC buffer, 0.1 % (w/v) SDS, or lower salt concentration, and at a temperature of at least 65°C, or equivalent conditions.
Reference herein to a particular level of stringency encompasses equivalent conditions using wash/hybridization solutions other than SSC known to those s skilled in the art.
Generally, the stringency is increased by reducing the concentration of SSC
buffer, and/or increasing the concentration of SDS and/or increasing the temperature of the hybridization and/or wash. Those skilled in the art will be to aware that the conditions for hybridization and/or wash may vary depending upon the nature of the hybridization matrix used to support the sample DNA, or the type of hybridization probe used.
In one embodiment, the sample or the probe is immobilized on a solid matrix or is surface (e.g., nitrocellulose). For high throughput screening, the sample or probe will generally comprise an array of nucleic acids on glass or other solid matrix, such as, for example, as described in WO 96/17958. Techniques for producing high density arrays are described, for example, by Fodor et al., Science 767-773, 1991, and in U.S. Pat. No. 5,143,854. Typical protocols for other assay formats 2o can be found, for example in Current Protocols In Molecular Biology, Unit 2 (Northern Blotting), Unit 4 (Southern Blotting), and Unit 18 (PCR Analysis), Frederick M. Ausubul et al. (ed)., 1995.
The detection means according to this aspect of the invention may be any nucleic 2s acid-based detection means such as, for example, nucleic acid hybridization or amplification reaction (eg. PCR), a nucleic acid sequence-based amplification (NASBA) system, inverse polymerase chain reaction (iPCR), or in situ polymerase chain reaction.
3o The probe can be labelled with a reporter molecule capable of producing an identifiable signal (e.g., a radioisotope such as 32P or 35S, or a fluorescent or biotinylated molecule, or a coloured dye e.g. TAMRA, FAM, ROC, etc). According to this embodiment, those skilled in the art will be aware that the detection of said reporter molecule provides for identification of the probe and that, following the hybridization reaction, the detection of the corresponding nucleotide sequences in s the sample is facilitated. Additional probes can be used to confirm the assay results obtained using a single probe.
Wherein the detection means is an amplification reaction such as, for example, a polymerase chain reaction or a nucleic acid sequence-based amplification to (NASBA) system or a variant thereof, one or more nucleic acid probes molecules of at least about 20 contiguous nucleotides in length is hybridized to genomic DNA
and nucleic acid copies of the template are enzymically-amplified.
Those skilled in the art will be aware that there must be a sufficiently high is percentage of nucleotide sequence identity between the probes and the nucleotide sequence of the sample template molecule for hybridization to occur.
As stated previously, the stringency conditions can be selected to promote hybridization.
2o In one format, PCR provides for the hybridization of non-complementary probes to different strands of a double-stranded nucleic acid template molecule (ie. a DNA/DNA template), such that the hybridized probes are positioned to facilitate the 5'-to 3' synthesis of nucleic acid in the intervening region, under the control of a thermostable DNA polymerase enzyme. In accordance with this embodiment, 2s one sense probe and one antisense probe as described herein is used.
Variations of the embodiments described herein are described in detail by McPherson et al., PCR: A Practical Approach. (series eds, D. Rickwood and B.D.
Hames), IRL Press Limited, Oxford. pp1-253, 1991.
In one embodiment, the probe detects non-coding nucleic acid (i.e. an intron, 5'-upstream region or 3'-untranslated region). By way of exemplification, the nucleotide sequences set forth in SEQ ID NOs: 5 and 6, are used to amplify nucleic acid comprising a microsatellite designated "CEDD" that successfully detects allelic imbalance at the region of the human genome between map s positions 8q22.3 and 8q24.13 associated with ovarian cancer, hepatocellular carcinoma, breast cancer, squamous cell carcinoma and metastatic melanoma.
The microsatellite CEDD also successfully distinguishes serous ovarian cancer from other ovarian cancers of epithelial origin.
Io The nucleotide sequence of the amplification product comprising the microsatellite CEDD is set forth in SEQ ID NO: 7 and, as will be known to those skilled in the art this amplified fragment is also useful for directly hybridizing to genomic DNA
of a subject in the assay formats described herein for the purpose of diagnosing the cancers supra, particularly ovarian cancer.
is In other exemplified embodiments described herein, the present invention clearly provides nucleic acid primers and amplified probes for detecting allelic imbalance at this region of the human genome for the detection of breast cancer (e.g.
SEQ
ID Nos: 24 and 25) which amplify an intron region of the gene encoding an EDD
2o protein. As with the CEDD microsatellite, the nucleic acid that is amplified using the primers set forth in SEQ ID Nos: 24 and 25 is also useful as a hybridization probe and the present invention clearly encompasses such a use.
In one embodiment, the amplification reaction detection means described supra is 2s further coupled to a classical hybridization reaction detection means to further enhance sensitivity and specificity of the inventive method, such as by hybridizing the amplified DNA with a probe which is different from any of the probes used in the amplification reaction.
3o In another embodiment, the hybridization reaction detection means described supra is further coupled to a second hybridization step employing a probe which is different from the probe used in the first hybridization reaction.
The comparison to be performed in accordance with the present invention may be a visual comparison of the signal generated by the probe, or alternatively, a s comparison of data integrated from the signal,. such as, for example, data that have been corrected or normalized to allow for variation between samples. Such comparisons can be readily performed by those skilled in the art.
In one embodiment, the method supra further comprises isolating the test sample io and/or the reference sample from one or more suitable subjects (i.e. the individual being tested and/or one or more other subjects who are suitable to provide a reference sample). In this respect it is within the scope of the invention for the reference sample and the test sample to be isolated from the same subject at different time points, or alternatively, from different tissues of the same subject.
is Preferably, the test sample and the reference sample are from the same tissue type, or a tissue comprising cells of the same type.
In one embodiment, the test sample and/or the reference samples) has/have been obtained previously from the subject(s).
In another embodiment, the present invention provides a method for diagnosing a cancer or predicting recurrence of a cancer in a subject comprising determining the level of mRNA or protein encoded by nucleic acid linked to map position 8q22.3 of the human genome in a sample of said subject, wherein an elevated 2s level of said mRNA or protein is indicative of relapse of a cancer in said subject.
In one embodiment, the mRNA encoded by a nucleic acid linked to map position 8q22.3 of the human genome encodes an EDD protein. Preferably, the mRNA
comprises the sequence set forth in SEQ ID NO: 1 or SEQ ID NO: 3 or a fragment 3o thereof. Even more preferably, the mRNA encodes a protein comprising the sequence set forth in SEQ ID NO: 2 or SEQ ID NO: 4 or a fragment thereof.

In one embodiment the protein encoded by a nucleic acid linked to map position 8q22.3 of the human genome is an EDD protein. Preferably the protein encoded by a nucleic acid linked to map position 8q22.3 of the human genome comprises s the sequence set forth in SEQ lD NO: 2 or SEQ ID NO: 4 or a fragment thereof.
Methods of determining the amount of mRNA or protein encoded by a nucleic acid linked to map position 8q22.3 are well known in the art and/or described herein.
to As exemplified herein, the level of expression of EDD mRNA and EDD protein is ° predictive of recurrence of ovarian cancer in subjects that have received treatment for said cancer. As used herein the term "recurrence" or "relapse" shall be understood to mean that, following treatment for a cancer, a subject has developed a further cancer. The cancer that has redeveloped may be the same is form of cancer for which the patient received treatment or a different form of cancer, for example in the case of a cancer that has metastasized.
In an alternative embodiment the invention provides a method for detecting allelic imbalance in a region of the human genome comprising hybridizing a nucleic acid 2o probe or primer to genomic DNA and detecting the hybridization, wherein the probe or primer comprises a nucleotide sequence selected from the group consisting of:
(i) the sequence set forth in SEQ ID NO: 5;
(ii) the sequence set forth in SEQ ID NO: 6;
2s (iii) the sequence set forth in SEQ ID NO: 7;
(iv) the sequence set forth in SEQ ID NO: 24;
(v) the sequence set forth in SEQ ID NO: 25; and (vi) the sequence of a nucleic acid fragment produced by amplification using any one of (i) to (v) as amplification primers in PCR.

The embodiments described supra for nucleic acid detection are to be applied mutatis mutandis to this embodiment of the invention. Preferablv, the hybridization is detected by amplifying nucleic acid using said probe or primer in a PCR reaction, or alternatively, by labelling the probe with a suitable reporter s molecule and detecting the signal generated by the reporter molecule.
In another embodiment, the present invention provides a method for detecting a cancer cell in a subject, said method comprising:
(i) determining the level of mRNA encoded by nucleic acid linked to map to position 8q22.3 of the human genome that is expressed in a test sample from said subject; and (ii) comparing the level of the mRNA determined at (i) to the level of mRNA
encoded by nucleic acid linked to map position 8q22.3 of the human genome that is expressed in a reference sample from a healthy or normal is individual, wherein a level of the mRNA at (i) that is enhanced in the test sample relative to the reference sample from the normal or healthy individual is indicative of the presence of a cancer cell in said subject.
2o In one embodiment the mRNA encodes an EDD-protein. In an alternative embodiment, the mRNA encodes a p53R2 protein.
By "mRNA encoding an EDD protein" is meant mRNA encoding a EDD
polypeptide that has at least about 80% identity to SEQ ID NO: 2 or 4, and, more 2s particularly, mRNA comprising a nucleotide sequence that has at least about 80%
identity, more preferably at least about 95% identity, and still more preferably at least about 99% identity to the nucleotide sequence set forth in SEQ ID NO: 1 or 3.
3o The tissue samples, ° general hybridization and amplification methods, and detection and analysis methods described supra for detecting allelic imbalance in this region of the human genome are applied mutatis mutandis to the detection of mRNA without any undue experimentation. Alternatively, or in addition, the level of mRNA encoding an EDD protein or p53R2 protein in a patient sample is analyzed by a variations on these procedures that are well known in the art such as, for s example, in situ hybridization to mRNA, northern blotting techniques, or RT-PCR
analysis (such as, for example, performed on laser capture microdissected samples). Microarray technology is readily applied to such embodiments for high throughput screening of samples.
~o In hybridization-based approaches, the use of riboprobes is particularly preferred because RNA/RNA duplexes are more stable than RNA/DNA duplexes. As with DNA/DNA hybridizations, the conditions for RNA/DNA or RNA/RNA hybridization and/or wash will vary depending upon the nature of the hybridization matrix used to support the sample RNA, or the type of hybridization probe used.
is For RT-PCR, or a variant thereof, one or more nucleic acid probes molecules of at least about 20 contiguous nucleotides in length is hybridized to cDNA or cRNA
that has been reverse-transcribed from the target mRNA, and nucleic acid copies of the template are enzymically-amplified using nucleic acid primers. RT-PCR
is 2o particularly useful when it is desirable to determine gene expression levels. It is also known to those skilled in the art to use mRNA/DNA hybrid molecules as a template for such amplification reactions, and, as a consequence, first strand cDNA synthesis is all that is required to be performed prior to the amplification reaction. Variations of the embodiments described herein are described in detail 2s by McPherson et al., PCR: A Practical Approach. (series eds, D. Rickwood and B.D. Hames), IRL Press Limited, Oxford. pp1-253, 1991.
In one embodiment, the method supra further comprises isolating the test sample and/or the reference sample from one or more suitable subjects (i.e. the individual 3o being tested and/or one or more other subjects who are suitable to provide a reference sample). In this respect it is within the scope of the invention for the reference sample and the test sample to be isolated from the same subject at different time points, or alternatively, from different tissues of the same subject.
Preferably, the test sample and the reference sample are from the same tissue type, or a tissue comprising cells of the same type.
s In one embodiment, the test sample and/or the reference samples) has/have been obtained previously from the subject(s).
As exemplified herein, probes and primers having the nucleotide sequences set io forth in any one of SEQ ID Nos: 26 to 30, 33 or 34, 37, 38, or 40 are used to detect breast cancer cells. As with microsatellite detection, the nucleic acid primers set forth in any one of SEQ ID Nos: 26 to 30, 33 or 34, 37, 38, or 40 can also be used as a probe to detect nucleic acid directly. As with .microsatellite detection, nucleic acid that has been amplified with any one of SEQ ID Nos: 26 to is 30, 33 or 34, 37, 38, or 40 is also useful as a hybridization probe and the present invention clearly encompasses such a use.
A further embodiment of the present invention relates to nucleic acid probes for detecting a cancer in accordance with the embodiments described supra.
In one embodiment, the present invention provides an isolated nucleic acid molecule for detecting a cancer cell comprising a nucleotide sequence selected from the group consisting of:
(i) a sequence that encodes the amino acid sequence set forth in SEQ ID NO:
2s 4 wherein said amino acid sequence lacks the sequence VLLLPL;
(ii) the sequence set forth in SEQ ID NO: 3;
(iii) the sequence set forth in SEQ ID NO: 5;
(iv) the sequence set forth in SEQ ID NO: 6;
(v) the sequence set forth in SEQ ID NO: 7;
(vi) the sequence set forth in SEQ ID NO: 24;
(vii) the sequence set forth in SEQ ID NO: 25;

(viii) the sequence of a nucleic acid fragmentby amplification produced using (vi) and (vii) as amplification primers in PCR;

(ix) the sequence set forth in SEQ ID N0:26;

(x) the sequence set forth in SEQ 1D NO:
27;

s {xi) the sequence of a nucleic acid fragmentby amplification produced using (ix) and (x) as amplification primers in PCR;

(xii) the sequence set forth in SEQ ID NO:
28;

(xii) the sequence set forth in SEQ ID NO:
29;

(xiii) the sequence set forth in SEQ ID NO:
30;

io (xiv)the sequence of a nucleic acid fragmentby amplification produced using (xii) and (xiii) as amplification primers in PCR;

(xv) the sequence set forth in SEQ ID NO:
33;

(xvi) the sequence set forth in SEQ ID NO:
34;

(xvii) the sequence of a nucleic acid fragmentby amplification produced using is (xv) and (xvi) as amplification primers in PCR;

(xviii) the sequence set forth in SEQ ID NO:
37;

(xix) the sequence set forth in SEQ 1D NO:
38;

(xx) the sequence of a nucleic acid fragmentby amplification produced using (xviii) and (xix) as amplification primers in PCR;

20 (xxi)the sequence set forth in SEQ ID NO:
40; and (xxii) a sequence that is complementary to any one of (i) to (xxi).

As used herein, the term "nucleic acid" shall be taken to mean any single-stranded or double-stranded RNA, DNA, cDNA, cRNA, or synthetic oligonucleotide, or 2s alternatively, an analog of RNA, DNA, cDNA, cRNA, or a synthetic oligonucleotide. "Nucleic acid" also includes any genomic gene equivalents of a cDNA molecule.
The isolated nucleic acid of the invention will hybridize to nucleic acid from a 3o human or a non-human mammal.

Protein complexes comprising an EDD protein and diagnostic uses therefor The present inventors have also identified several expression products of a gene linked to map position 8q22.3 of the human genome wherein each of said s expression products consists of a protein-protein interaction involving the EDD
protein with a protein selected from the group consisting of a protein having tumor suppressor activity or cell cycle modulatory activity or DNA repair activity, a progesterone receptor protein, a nuclear targeting protein and a calcium/integrin binding protein (CIB). The interactions provide novel protein-protein complexes io that are useful for detecting DNA damage, progesterone-mediated effects in cells, such as for example, progestin-sensitive tumorigenesis or tumor growth, ubiquitin-mediated proteolysis in cells, or changes in vascularization in a subject. The novel protein-protein complexes are also useful for producing diagnostic reagents such as, for example, antibodies. As will be known to those skilled in the art, is antibodies are also useful for therapeutic applications. Additionally, the novel protein-protein complexes form the bases of assays for identifying modulatory compounds, including small molecule agonists or antagonists of the protein-protein interactions, such as for the treatment of hyperproliferative disorders, preventing cell proliferation or enhancing repair to damaged DNA, or for targeting ao cells having damaged DNA and/or that are tumor cells.
Accordingly, one embodiment of the present invention provides an isolated or recombinant protein complex comprising:
(i) an EDD protein or a portion of an EDD protein sufficient to bind to a 2s protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, a progesterone receptor protein and a protein associated with vascularization; and 30 (ii) a nuclear protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, a progesterone receptor protein and a protein associated with vascularization or a portion of said protein sufficient to bind to said EDD protein or said portion of an EDD
s protein.
As used herein, the term "protein having tumor suppressor activity" shall be taken to mean any protein or polypeptide that is known or thought to be involved repressing or reducing or preventing tumorigenesis or tumor growth, such as, for to example, by activating a cellular response to DNA damage, assisting DNA
repair, restoring survival after DNA damage (e.g. by interacting with and phosphorylating BRCA1 or BRCA2, thereby allowing BRCA1 or BRCA2 to restore survival after DNA damage), or preventing cellular proliferation of tumor cells (e.g. by stabilizing a tumor suppressor protein such as p53, thereby leading to cell cycle arrest in is G1). Such a functional assignment is readily determined in the art by examining the effects of mutations in genes encoding the protein having tumor suppressor activity, or alternatively, by empirical data showing a direct effect on tumorigenesis or tumor growth. Preferably, the protein having tumor suppressor activity will be a nuclear protein. Exemplary proteins having tumor suppressor activity in the 2o present context include a member of the CDS1 subfamily of serine/threonine protein kinases, p53 (TP53), TNF-alpha, HSP70, estrogen receptor, androgen receptor, progesterone receptor, HRAS1-VNTR, CHK2, BRCA1, BRCA2, AIB1, NAT1, NAT2, XRCC1, XRCC2, XRCCS, CIB, importin alpha-1, importin alpha-3, and importin alpha-5.
In one preferred embodiment, the protein having tumor suppressor activity will comprise an amino acid sequence having at least about 80% identity to a sequence set forth in any one of SEQ ID NOs: 9, 11, 13, 15, 17, 19, 21, or 23.
3o For the purposes of nomenclature, the amino acid sequence set forth in SEQ
ID
NO: 9 consists of a human importin alpha-1 protein (i.e. karyopherin alpha-2) which is encoded by nucleotides 133 to 1722 of the nucleotide sequence set forth in SEQ ID NO: 8. The amino acid sequence set forth in SEQ ID NO: 11 consists of a human importin alpha-3 protein (i.e. karyopherin alpha-4) which is encoded by nucleotides 10 to 1575 of the nucleotide sequence set forth in SEQ ID NO:
10, s The amino acid sequence set forth in SEQ lD NO: 13 consists of a human importin alpha-5 protein (i.e. karyopherin alpha-1 ) which is encoded by nucleotides 47 to 1663 of the nucleotide sequence set forth in SEQ ID NO: 12.
The amino acid sequence set forth in SEQ ID NO: 15 consists of a human progesterone receptor protein (PR) which is encoded by nucleotides 176 to 2977 ~o of the nucleotide sequence set forth in SEQ ID NO: 14. The amino acid sequence set forth in SEQ ID NO: 17 consists of a human CIB/KIP protein which is encoded by nucleotides 67 to 642 of the nucleotide sequence set forth in SEQ ID NO:
16.
The amino acid sequence set forth in SEQ ID NO: 19 consists of a human CHK2 protein (i.e. transcript variant 1 ) which is encoded by nucleotides 762 to 2393 of ~s the nucleotide sequence set forth in SEQ ID NO: 18. The amino acid sequence set forth in SEQ ID NO: 21 consists of a human CHK2 protein (i.e. transcript variant 2) which is encoded by nucleotides 762 to 2306 of the nucleotide sequence set forth in SEQ lD NO: 20. The amino acid sequence set forth in SEQ
ID NO: 23 consists of a human BRCA2 protein which is encoded by nucleotides 20 229 to 10,485 of the nucleotide sequence set forth in SEQ ID NO: 22.
More preferably, a protein having tumor suppressor activity will comprise an amino acid sequence having at least about 80% identity to a sequence set forth in any one of SEQ ID NOs: 19, 21, or 23 (i.e. at least 80% identity to a human CHK2 2s protein or a human BRCA2 protein) or a portion thereof sufficient to bind an EDD
protein.
Those skilled in the art will be aware that CHK2 is a nuclear cell cycle checkpoint regulatory protein of the CDS1 subfamily of serine/threonine protein kinases, 3o having DNA repair function. CHK2 contains a forkhead-associated protein interaction domain essential for activation in response to DNA damage and is rapidly phosphorylated in response to replication blocks and DNA damage. When activated, the encoded protein is known to inhibit CDC25C phosphatase, preventing entry into mitosis. CHK2 also interacts with BRCA1 protein, causing BRCA1 to be phosphorylated, thereby allowing BRCA1 to restore survival after s DNA damage. CHK2 also has a putative tumor suppressor activity by virtue of stabilizing the tumor suppressor protein p53 (TP53), leading to cell cycle arrest in the G1 phase.
Those skilled in the art will also be aware that BRCA2 is a tumor suppressor io protein by virtue of mutations, typically microdeletions, in the BRCA2-encoding gene being linked to an elevated risk of young onset breast cancer. BRCA2 is also associated with the activation of double-strand break repair andlor homologous recombination. The similar properties of BRCA1 and BRCA2, for example their co-localization in a biochemical complex, and similar function, suggests that these is proteins function in the same genetic pathway. The amino acid sequences of BRCA1 and BRCA2 comprise transcriptional activation protein domains.
By "protein having cell cycle modulatory activity" is meant that the protein functions either solus or in cooperation with one or more other proteins or nucleic 2o acid, to enhance cell cycle progression or to inhibit progression from one stage of the cell cycle to another. Preferred cell cycle modulatory proteins will modulate the progression of cells from the G1 into the G2 phase, or the entry of cells into mitosis. Such a functional assignment is readily determined in the art by examining the effects of mutations in genes encoding the protein having cell cycle 2s modulatory activity, or alternatively, by empirical data showing a direct effect on cell cycle progression, or by analysing protein-protein interactions with known cell cycle modulatory proteins. Preferably, the protein having cell cycle modulatory activity will be a nuclear protein. Exemplary proteins having cell cycle modulatory activity in the present context include a member of the CDS1 subfamily of 3o serine/threonine protein kinases, Cdc25, CDC2a, cyclin-dependent kinase (CDK), CDK inhibitor, a mitogenic cyclin (e.g., cyclin A, cyclin B, cyclin D, etc), p53 (TP53), and CHK2.
In one preferred embodiment, the protein having tumor suppressor activity will comprise an amino acid sequence having at least about 80% identity to a s sequence set forth in any one of SEQ ID NOs: 19 or 21.
Those skilled in the art will be aware from the preceding discussion that the Cdc25, CHK2 and TP53 proteins act as cell cycle modulatory proteins in the present context. In a particularly preferred embodiment, the protein having cell Io cycle modulatory activity is CHK2 (SEQ ID Nos: 19 or 21) or a portion thereof sufficient to bind an EDD protein.
By "protein associated with DNA repair or damage" is meant a nuclear protein that functions in modulating DNA repair, is activated by DNA damage or otherwise is activates double-strand break repair, DNA mismatch repair, homologous recombination or DNA end-ligation events in a cell or in vitro. Such a functional assignment is readily determined in the art by examining the efFects of mutations in genes encoding the protein, or alternatively, by empirical data showing a direct effect on the ability of a cell to respond to DNA damage and/or to undergo DNA
2o repair and/or to activate homologous recombination or DNA end-ligation.
Exemplary proteins associated with DNA repair or damage in the present context include a member of the CDS1 subfamily of serinelthreonine protein kinases, BRCA1, BRCA2, CIB/KIP, TP53, MLH1, MSH2, ATM, CHK2, XRCC1, XRCC2, XRCC5 and importin alpha-5.
In one preferred embodiment, the protein associated with DNA damage or repair will comprise an amino acid sequence having at least about 80% identity to a sequence set forth in any one of SEQ ID NOs: 13, 17, 19, 21, or 23.
3o In a particularly preferred embodiment, the protein associated with DNA
damage or repair is selected from the group consisting of a CHK2 protein (SEQ ID Nos:

or 21), a BRCA2 protein (SEQ ID NO: 23), a CIB protein (SEQ ID NO: 11) and an importin alpha-5 protein (SEQ ID NO: 13) or a portion thereof sufficient to bind an EDD protein.
s Those skilled in the art will be aware from the preceding discussion that the BRCA2 and CHK2 proteins are associated with DNA damage or repair, primary acting as DNA-damage checkpoint control proteins. Those skilled in the art will also be aware that the CIB protein is a member of the calcium-binding protein family that interacts with a DNA-dependent protein kinase and, as a consequence, to is in the art to play a role in kinase-phosphatase regulation of DNA end-ligation events. The CIB protein also interacts with integrin alpha(Ilb)beta(3), which may implicate this protein as a regulatory molecule for integrin alpha(Ilb)beta(3).
Those skilled in the art will also be aware that the importin alpha-5 protein is involved in ds-DNA break repair. Importin alpha-5 is also recruited by RAG1 is during V(D)J recombination, the process by which genes encoding immunoglobulins and T-cell receptors are generated.
By "nuclear targeting protein " is meant a chaperonin protein that assists with the translocation of a protein from the cytosol to the nucleus. The import of proteins 2o into the nucleus involves an energy-independent docking of the protein to the nuclear envelope and an energy-dependent translocation through the nuclear pore complex. In the present context, a nuclear targeting protein will facilitate one or both of these processes. Such a functional assignment is readily determined in the art by examining the effects of mutations in genes encoding the protein, or 2s alternatively, by empirical data showing a direct effect on the ability of a protein to facilitate nuclear localisation of a protein. Exemplary nuclear targeting proteins include importin alpha-1 (SEQ ID NO: 9), importin alpha-3 (SEQ ID NO: 11 ) and importin alpha-5 (SEQ ID NO: 13).
3o In one preferred embodiment, the nuclear targeting protein will comprise an amino acid sequence having at feast about 80% identity to a sequence set forth in any one of SEQ ID NOs: 9, 11 or 13.
Those skilled in the art are aware that importin alpha-1 and importin alpha-3 interact with the nuclear localisation sequence (NLS) of the DNA helicase Q1 s protein and the NLS of the SV40 T antigen, and dock the proteins to the nuclear envelope or nuclear pore complex in the nuclear transport of those proteins.
These importins are also considered in the art to play a role in V(D)J
recombination.
to By "nuclear localisation sequence" or "NLS" is meant a short region of basic amino acids or 2 such regions spaced about 10 amino acids apart that is required for nuclear localization of a protein, particularly transport mediated by an importin protein.
is The functional meaning of the term "progesterone receptor protein" will be apparent from the preceding description. For the purposes of defining the structure of a progesterone receptor protein, there is provided the amino acid sequence set forth herein as SEQ ID NO: 15. Variants of SEQ ID NO: 15, such as, for example, proteins having at least about 80% identity thereto are also within 2o the scope of the present invention, the only requirement being that the variant is a functional progesterone receptor or derived from a functional progesterone receptor protein. A human progesterone receptor protein has been shown by the present inventors to bind to an EDD protein.
2s By "a protein associated with vascularization" is meant that the protein functions to enhance the development of new blood vessels in a subject either directly or indirectly (eg by inducing expression of a protein that enhances vascularization or interacting with a protein that enhances or inhibits vascularization).
Preferably, the protein is associated with vasculogenic activity, whereby the term 30 "vasculogenic activity" shall be understood to mean the formation of new blood vessels de novo. The process of vasculogenesis results in the in situ differentiation of mesodermal progenitor cells to endothelial cells that organize into a primitive vascular network. Many aggressive tumors (eg melanoma tumor cells) are also capable of inducing vasculogenesis in a subject.
s In another embodiment, a protein associated with vascularization is capable of enhancing or inhibiting angiogenesis. By "angiogenesis" is meant the development of new blood vessels from previously existing blood vessels. The process of angiogenesis involves the endothelial cells of a pre-existing blood vessel secreting membranes to erode the basement membrane of the vessel.
to Endothelial cells subsequently proliferate and migrate toward an angiogenic signal, forming tight cell junctions with other endothelial cells in order to form a new blood vessel. Angiogenesis occurs during, for example, embryonic development, wound healing, and formation of the corpus luteum, endometrium and placenta. However, aberrant angiogenesis is associated with a number of is disorders, including, tumor metastasis.
A functional assignment of a protein that is associated with vascularization is readily determined in the art by examining the effects of mutations in genes encoding the protein having vascularization activity, or alternatively, by empirical 2o data showing a direct effect on vascularisation, or by analysing protein-protein interactions with known vascularization modulatory proteins. For example, the effect of a protein on vascularization may be determined any assay known in the art, such as, for example, a bovine capillary endothelial cell proliferation assay, a chick CAM assay (as described in O'Reilly et al, Cell, 79(2): 315-328 1994), a 2s mouse corneal assay or a mouse ischemic retinopathy assay (as described in Ozaki et al, Am. J. Path., 156(2): 697-707, 2000).
Preferably, the protein having vascularization activity will be a nuclear protein. For example, proteins with vascularization activity include, transforming growth factor 30 Vii, lipid phosphatase LPP3, c-Myc, dimethylarganine dimethylaminohydrolase (DDAH), cytochrome P450, Tie-2 and VE-cadherin.

In one embodiment, the protein complex is a heterodimer or hetero-multimeric protein comprising an EDD protein. Those skilled in the art will be aware that a heterodimer or heterodimeric protein complex comprises two different peptide or s polypeptide subunits. As used herein, the term "heteromultimer" shall be taken to mean a higher order protein complex comprising at least three peptide or polypeptide subunits, wherein at least two of said subunits are different. For example, a heterohexameric protein is known to comprise six peptide or pofypeptide subunits, and, in the present context, may comprise three different io homodimers, or six different monomers, or a dimer and a tetramer of different protein, etc. Accordingly, the present invention is not to be limited by the composition and size of the protein complex, the only requirement being that at least one peptide or polypeptide subunit consists of EDD or a portion of EDD, and at least one other peptide or polypeptide subunit consists of a polypeptide is selected from the group consisting of a protein having tumor suppressor activity, a protein associated with DNA damage or repair, a cell cycle modulatory protein, a nuclear targeting protein and a progesterone receptor protein, a portion of a protein having tumor suppressor activity, a portion of a protein associated with DNA damage or repair, a portion of a cell cycle modulatory protein, a portion of a 2o nuclear targeting protein and a portion of a progesterone receptor protein.
The protein subunits of the protein complex are held in physical relation by any means known to those skilled in the art. This physical relation may involve the formation of an induced magnetic field or paramagnetic field, covalent bond 2s formation such as a disulfide bridge formation between polypeptide subunits, an ionic interaction such as occur in an ionic lattice, a hydrogen bond or alternatively, a van der Waals interaction such as a dipole-dipole interaction, dipole-induced-dipole interaction, induced-dipole-induced-dipole interaction or a repulsive interaction or any combination of the above forces of attraction.
Alternatively, the 3o peptide or polypeptide subunits may be held in physical relation by expressing them as a fusion polypeptide, optionally separated by a spacer to permit their folding. Accordingly, the physical relation between the peptide or polypeptide subunits may be a consequence of their binding' capability and attraction toward one another, or alternatively, a consequence of their mode of production.
s Preferably, the peptide, polypeptide or protein partners are in direct physical relation. By "direct physical relation" is meant that the binding partners contact each other without any intervening protein moiety or non-protein moiety.
However, the protein complexes of the present invention can clearly include one or more additional protein moieties or non-protein moieties, such as, for example, to a protein or non-protein moiety that enhances or stabilizes the physical relation between EDD or a portion of EDD and the other binding partner.
The present invention further encompasses a protein complex wherein one or more of the binding partners include a post-translational modification, such as, for is example, a phosphorylated, fucosylated, myristoylated, farnesylated, or glycosylated residue. Such post-translational modifications may enhance complex formation or stabilize the complex once it is formed. Phosphorylation of one or more serine or tyrosine residues present on one or more of the binding partners including EDD or CH1~2, such as by a member of the CDS1 subfamily of 2o serine/threonine protein kinases (e.g. CHK2), is also contemplated.
Ubiquitination of one or more binding partners is also not to be excluded.
Preferably, the binding partners of the protein complex are mammalian polypeptides or proteins, and more preferably of human origin. It is not strictly 2s necessary for the binding partners to be derived from the same source, however this is preferred because the ability of the partners to associate or be maintained in physical non-covalent association with each other is generally enhanced if they are derived from the same organism.
3o Those skilled in the art will be in a position to determine a suitable portion of EDD
or other polypeptide that can form the protein complex of the invention, such as, for example a portion comprising a cysteinelhistidine rich region (e.g. a zinc-binding domain), HECT domain, RING domain, nuclear localisation sequence (NLS), UBA domain that binds to mono- or multi-ubiquitin chains, or a region comprising alpha helices (e.g. carboxy-terminal 60 amino acids of SEQ ID Nos:

s or 4). This is achieved, for example, using conventional binding assays for determining the binding between two proteins without undue experimentation.
Similarly, the skilled artisan can readily determine a portion of the other binding partner that is sufficient to bind to an EDD protein or a portion thereof.
Again, io conventional binding assays for determining the binding between two proteins may be used to assay the suitability of such portions.
Preferably, a portion of an EDD protein or other protein suitable for protein complex formation comprises at least about 5 amino acids in length, more is preferably at least about 10 amino acids in length, even more preferably at least about 15 amino acids in length and still more prefierably at least about 20 or 30 or 40 or 50 amino acids in length.
In a preferred embodiment, the portion of an EDD protein that is sufficient to bind 2o to a protein having tumor suppressor activity or having cell cycle modulatory activity will at least comprise amino acid residues from about position 1400 to about position 2550 of SEQ ID NO: 2 or the corresponding region in SEQ ID NO:
4, or more preferably, the region of SEQ ID NO: 2 from about position 1406 to about position 2526.
In another embodiment, the portion of an EDD protein that is sufficient to bind to a protein associated with DNA repair or damage will at least comprise amino acid residues in the C-terminal portion of the full-length EDD protein, such as, for example, from about position 889 to about position 2799 of SEQ ID NO: 2 or the 3o corresponding region in SEQ ID NO: 4.

In another embodiment, the portion of an EDD protein that is sufficient to bind to a nuclear targeting protein will at least comprise the nuclear localisation sequence of an EDD protein. In one embodiment, the EDD NLS comprises at least amino acid residues from about position 502 to about position 517 of SEQ ID NO: 2, or s from about position 600 to about position 605 of SEQ ID NO: 2, or from about position 1222 to about position 1241 of SEQ ID NO: 2, or the corresponding regions in SEQ ID NO: 4.
In another embodiment, the portion of an EDD protein that is sufficient to bind to a to progesterone receptor protein will at least comprise the N-terminal portion of EDD, such as, for example, amino acid residues from about position 1 to about position 889 of SEQ ID NO: 2, or the corresponding regions in SEQ ID NO: 4, or more preferably, the region of SEQ ID NO: 2 from about position 420 to about position 889 or the equivalent region in SEQ ID NO: 4.
A preferred portion of the CHK2 protein that interacts with an EDD protein will at least comprise the FHA domain of CHK2 (i.e. amino acid residues 117 to 157 of SEQ ID NO: 19 or the equivalent region in SEQ ID NO: 21, and more preferably residues 111 to 177 of SEQ ID NO: 19 or the equivalent region in SEQ ID NO:
21 ).
A preferred portion of a progesterone receptor (PR) protein that interacts with an EDD protein will comprise at least a C-terminal portion of PR, more preferably a region comprising a the hinge domain, DNA binding domain and ligand-dependent 2s activation domain-2 of the full-length receptor protein (i.e. "CDE
region").
A preferred portion of an importin protein that interacts with an EDD protein will at least comprise the C-terminal domain of importin, such as, for example, amino acids from about position 229 to about position 538 of SEQ ID NO: 13 or the 3o homologous region in SEQ ID NO: 9.

Another embodiment of the present invention provides isolated peptides, polypeptides, and kits comprising same for producing the protein complex, or for identifying a modulator of a biological interaction between EDD or a portion of EDD and one or more other polypeptides selected from the group consisting of a s protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, a progesterone receptor protein, and a protein associated with vascularization, or a portion thereof.
to In one embodiment, the kit comprises a first polypeptide consisting of EDD
or a portion thereof sufficient to bind to a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, a progesterone receptor protein and a protein associated with Is vascularization, and a second polypeptide consisting of a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, a progesterone receptor protein and a protein associated with vascularization or a portion thereof sufficient to bind to 2o said EDD protein or said portion of an EDD protein. Such kits are used to produce protein complexes comprising: EDD.
Optionally, the kit further includes a protein or a portion thereof sufficient to bind to a protein that binds EDD comprising an amino acid sequence selected from the as group consisting of SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO:
15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ 1D NO: 21, and SEQ fD NO: 23. In accordance with this embodiment, the subject kit is used to produce higher order protein complexes comprising an EDD protein and a protein that binds EDD and a protein that binds to the EDD-binding protein. Alternatively, such kits are useful 3o for analyzing competition between EDD and a protein that binds to an EDD-binding protein in the formation of a protein-protein complex. For example, the kit can be used to produce a complex comprising EDD and BRCA1 and CHK2 or to determine competition between BRCA1 and EDD for complex formation with CHK2. Similarly, the kit can be used to produce a complex comprising EDD and BRCA2 and CHK2 or to determine competition between BRCA2 and EDD for s complex formation with CHK2, or between CHK2 and EDD for complex formation with BRCA2. Similarly, the kit can be used to produce a complex comprising EDD
and BRCA2 and BRCA1 or to determine competition between BRCA1 and EDD
for complex formation with BRCA2. Similarly, the kit can be used to produce a complex comprising EDD and TP53 and CHK2 or to determine competition io between TP53 and EDD for complex formation with CHK2. Similarly, the kit can be used to produce a complex comprising EDD and CIB and integrin or to determine competition between integrin and EDD for complex formation with CIB.
Similarly, the kit can be used to produce a complex comprising EDD and importin alpha-5 and RAG1 or to determine competition between RAG1 and EDD for is complex formation with importin alpha-5. Similarly, the kit can be used to produce a complex comprising EDD and importin alpha-1/3 and DNA helicase or to determine competition between DNA helicase and EDD for complex formation with importin alpha-1/3. Similarly, the kit can be used to produce a complex comprising EDD and progesterone receptor and a heat shock protein that binds to 2o the progesterone receptor or to determine competition between a heat shock protein and EDD for complex formation with the progesterone receptor.
The kit may also include one or more antibodies or ligands that bind to the first polypeptide or the second polypeptide, or to any one or more of the protein 2s complexes supra comprising EDD, such as, for example, an antibody or ligand that specifically recognizes an assembled protein complex or the conformation of said protein complex, rather than the individual polypeptide components per se.
In another embodiment, the kit comprises:
30 (a) a first compartment comprising an EDD protein or a portion thereof sufficient to form a protein complex selected from the group consisting of:

(i) a complex comprising EDD and CHK2; (ii) a complex comprising EDD
and BRCA2; (iii) a complex comprising EDD and CIB; (iv) a complex comprising EDD and importin alpha-1; (v) a complex comprising EDD and importin alpha-3; (vi) a complex comprising EDD and importin alpha-5; and s (vii) a complex comprising EDD and progesterone receptor; and (b) a second compartment comprising an antibody or ligand that binds to a protein selected from the group consisting of (i) a CHK2 protein; (ii) a BRCA2 protein; (iii) a CIB protein; (iv) an importin alpha-1 protein; (v) an importin alpha-3 protein; (vi) an importin alpha-5 protein; and (vii) a to progesterone receptor protein, or an antibody or ligand that binds to a protein complex selected from the group consisting of: (i) a complex comprising EDD and CHK2; (ii) a complex comprising EDD and BRCA2;
(iii) a complex comprising EDD and CIB; (iv) a complex comprising EDD
and importin alpha-1; (v) a complex comprising EDD and importin alpha-3;
is (vi) a complex comprising EDD and importin alpha-5; and (vii) a complex comprising EDD and progesterone receptor.
wherein said antibody or ligand that binds to a protein complex does not bind to the individual protein binding partners.
ao In another embodiment, the kit comprises a first compartment comprising an antibody or ligand that binds to an EDD protein and a second compartment comprising a protein selected from the group consisting of (i) a CHK2 protein;
(ii) a BRCA2 protein; (iii) a ClB protein; (iv) an importin alpha-1 protein; (v) an importin alpha-3 protein; (vi) an importin alpha-5 protein; and (vii) a progesterone receptor 2s protein, or a portion thereof sufficient to bind to an EDD protein.
In another embodiment, the kit comprises:
(a) a first compartment comprising an isolated or recombinant protein complex selected from the group consisting of: (i) a complex comprising EDD and 3o CHK2; (ii) a complex comprising EDD and BRCA2; (iii) a complex comprising EDD and CIB; (iv) a complex comprising EDD and importin alpha-1; (v) a complex comprising EDD and importin alpha-3; (vi) a complex comprising EDD and importin alpha-5; and (vii) a complex comprising EDD and progesterone receptor; and (b) a second compartment comprising an (i) antibody or ligand that binds to a s polypeptide selected from the group consisting of a CHK2 protein, a BRCA2 protein, a CIB protein, an importin alpha-1 protein, an importin alpha-3 protein, an importin alpha-5 protein, a progesterone receptor protein and an EDD protein; or (ii) an antibody or ligand that binds to one or more protein complexes (a).
to As used herein, the term "antibody" refers to intact monoclonal or polyclonal antibodies, immunoglobulin (IgG, IgM, IgE) fractions, humanized antibodies, or recombinant single chain antibodies, as well as fragments thereof, such as Fab, F(ab')a, and Fv, which are capable of binding a linear or conformational epitope of is at least one binding partner of the protein complex, or to a conformational epitope of the assembled protein complex. Humanized antibodies are antibodies in which amino acids have been replaced in the non-antigen binding regions in order to more closely resemble a human antibody, while still retaining the original binding ability.
In one embodiment, antibodies are obtained from a commercial source, such as for example, Santa Cruz Biotechnology, Inc, CA 95060, USA. Other commercial sources will be well known to those skilled in the art.
2s Alternatively, antibodies are produced by conventional means. For the production of antibodies, an intact polypeptide, or a portion thereof containing a short amino acid sequence of interest is used as the immunizing antigen or immunogen. The immunogen is derived from a natural source, produced by recombinant expression means or by in vitro translation of RNA, or synthesized chemically 3o such as by Fmoc chemistry. Immunogens consisting of short peptides a preferably conjugated to a carrier protein, such as, for example bovine serum albumin (BSA), thyroglobulin, or keyhole limpet hemocyanin (KLH), prior to immunization. The coupled peptide is then used to immunize the animal. Various host animals (e.g. goats, rabbits, rats, mice, dogs, humans) are immunized by intramuscular, intraperitoneal, or intravenous injection, with immunogen, optionally s in the presence of an adjuvant to enhance the immune response to the immunogen. Preferred adjuvants include, for example, Freund's complete or incomplete adjuvant, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol. Among adjuvants used io in humans, BCG (bacilli Calmette-Guerin) and Corynebacterium parvum are preferred.
Monoclonal antibodies are prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture, such as, for is example, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique (Kohler et al. Nature 256, 495-497, 1975; Kozbor et al., J. Immunol. Methods 81, 31-42, 1985; Cote et al., Proc. Natl. Acad. Sci.
USA
80, 2026-2030, 1983; Cole ef al., Mol. Cell Biol. 62, 109-120, 1984).
2o Techniques developed for the production of chimeric antibodies are also employed. Such techniques involve splicing a mouse antibody gene to a human antibody gene to produce a molecule having the desired antigen specificity and biological activity (Morrison et al., Proc. Natl. Acad. Sci. USA 81, 6851-6855, 1984; Neuberger et al., Nature 312, 604-608, 1984; Takeda et al., Nature 314, 2s 452-454, 1985).
Alternatively, techniques described for the production of single chain antibodies are adapted, using methods known in the art, to produce single chain antibodies having the desired specificity.
Antibodies are also produced by inducing in vivo production in the lymphocyte population or by screening immunoglobulin libraries or panels of highly specific binding reagents as disclosed by Orlandi et al., Proc. Natl. Acad. Sci. USA
86, 3833-3837, 1989; Winter et al., Nature 349, 293-299, 1991 ).
s Antibody fragments, such as, for example, F(ab')a fragments, are produced by pepsin digestion of an intact antibody molecule. Fab fragments are generated by reducing the disulfide bridges of F(ab')2 fragments. Alternatively, Fab expression libraries are constructed, to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity (Huse et al., Science 254, 1275-1281, 1989).
The antibodies or ligands may assist in the subsequent isolation or detection of the complex formed between EDD and another protein. Binding of the antibody or ligand to a region of the first or second polypeptide that is not involved in complex is formation is preferred for this purpose, the only requirement being that, in use, the ligand does not disrupt the complex formed.
Preferably, the ligand is a small molecule or alternatively, a binding partner for one of the protein complexes contemplated herein. Particularly preferred ligands for 2o use in accordance with this embodiment are selected from the group consisting of a heat shock protein that binds to progesterone receptor, BRCA1, TP53, and nucleic acid (RNA or DNA).
The antibody or ligand may be labelled using a suitable reporter molecule, such 2s as, for example, a fluorophore, chromophore, or radioisotope. In the case of antibodies and small molecules, these may also be detected using antibodies in accordance with procedures known to those skilled in the art.
Optionally, the kit is packaged with instructions for use.
The kits of the invention are useful for producing and/or detecting the protein complexes of the invention in vitro or in vivo. For producing the protein complexes, one or more of the non-antibody/ligand components of the kits is added to a cellular source for a time and under conditions sufficient for complex formation to occur. The antibody components are particularly useful for isolating s the complexes) thus formed. Optionally any one of the kit components is labelled with a protein tag to facilitate subsequent isolation or purification of the protein complex.
In use, the polypeptide components are contacted for a time and under conditions to sufficient for complex formation to occur. Additional proteins may be provided from cellular or non-cellular sources to produce protein complexes other than those specifically referred to herein. When provided, the antibody or ligand is used to detect or isolate the complex formed. The ligand should be selected such that it does not disrupt the protein complex formed.
is In another embodiment of the invention there is provided an isolated antibody that binds to a protein complex comprising an EDD protein, preferably a protein complex selected from the group consisting of: (i) a complex comprising EDD
and CHK2; (ii) a complex comprising EDD and BRCA2; (iii) a complex comprising EDD
2o and CIB; (iv) a complex comprising EDD and importin alpha-1; (v) a complex comprising EDD and importin alpha-3; (vi) a complex comprising EDD and importin alpha-5; and (vii) a complex comprising EDD and progesterone receptor, subject to the proviso that said antibody does not bind to any individual protein of said complex in the absence of another protein of said complex.
Preferably, the antibody recognizes a conformational epitope of the protein complex.
Another embodiment of the present invention provides an anti-idiotypic antibody 3o that binds to an an antibody or ligand that binds to a protein complex selected from the group consisting of: (i) a complex comprising EDD and CHK2; (ii) a complex comprising EDD and BRCA2; (iii) a complex comprising EDD and CIB;
(iv) a complex comprising EDD and importin alpha-1; (v) a complex comprising EDD and importin alpha-3; (vi) a complex comprising EDD and importin alpha-5;
and (vii) a complex comprising EDD and progesterone receptor, subject to the s proviso that said anti-idiotypic antibody does not bind to an antibody that binds to an individual protein of said complex in the absence of another protein of said complex Another embodiment of the present invention provides methods for isolating a io EDD binding protein or a complex comprising same from a suitable cellular source.
Preferably, the protein or complex is provided substantially free of conspecific proteins, meaning that it is at least about 1-5% pure as determined by an analysis is of proteins by SDS/PAGE. More preferably, the protein is at least about 10%, even more preferably at least about 20% pure, even more preferably at least about 25% pure, even more preferably at least about 30% pure, and even more preferably at least about 50% pure, and still more preferably substantially pure.
2o To isolate the protein complex of the invention, one or both binding partners are separately isolated, from the same or a different cellular source that ectopically expresses or endogenously expresses at least one of the said binding partners.
The isolated binding partners are then combined in an amount and under conditions sufficient to facilitate their physical relation. Such conditions can be 2s readily determined by those skilled in protein chemistry. Selection of buffer pH, ionic strength, and temperature, sufficient to maintain the binding partners in solution are generally preferred. One or more protease inhibitors can also be included to prevent proteolytic digestion or degradation of the isolated polypeptides.
Alternatively, the protein complex per se may be isolated from a cellular source or sub-cellular source (e.g. nuclei) that contains both binding partners endogenously or ectopically. It is within the scope of this embodiment that the binding partners are expressed as a fusion protein or as distinct polypeptides.
s Preferred cellular sources of the isolated polypeptide binding partners, or the protein complex, include any mammalian cell, and preferably, a mammalian cell that is known to express EDD, and a protein selected from the group consisting of CHK2, BRCA2, CIB, importin alpha-1, importin alpha-3, importin alpha-5 and a progesterone receptor protein, or alternatively, a cell that can be engineered to io express said protein(s). Exemplary cells for such a purpose include cancer cells (e.g. carcinoma cells, breast cancer cells such as ER-negative breast cancer cells, or squamous epithelial carcinoma cells or ovarian cancer cells, or hepatocellular carcinoma cells), epithelial cells, cells of the central nervous system, kidney cells, T cells, NlH3T3 cells, murine 10T fibroblasts, MDA-MB-231 cells, MDCK cells, is COS cells, CHO cells, HeLa cells, or T-47D cells, HeLa cells, MCF-7 cells, or HEK
293 cells. The use of other cells (e.g. insect sf9 or sf21 cells, chick embryo cells and the like) is not excluded, particularly for isolation of a non-naturally occurring peptide, polypeptide or complex expressed by recombinant means.
2o Preferably, the protein complex or a binding partner thereof is isolated from cell line that endogenously expresses one or both binding partners, such as, for example, a cancer ceff selected from the group consisting of head and neck cancer, melanoma, metastatic melanoma, a squamous cell carcinoma of the tongue, breast cancer, adenocarcinoma, squamous lung cancer, gastrointestinal 2s cancer (eg. gastric, colon, or pancreatic cancer), ovarian cancer, hepatocellular carcinoma, renal cell cancer, bladder cancer, a gynecological carcinoma (eg.
ovarian cancer), prostate cancer, squamous cell carcinoma, non-squamous carcinoma, glioblastoma and medulloblastoma. More preferably, the cell will be a metastatic melanoma cell, squamous cell carcinoma cell, a breast cancer cell, a 3o hepatocellular carcinoma cell, or an ovarian cancer cell or a cell line derived from such cancers. In a particularly preferred embodiment, the protein complex or a -"65 -binding partner thereof is isolated from a carcinoma cell or carcinoma cell line, HeLa cell, MCF-7 cell, T-47D cell, or a HEK293 cell or COS cell that ectopically expresses one or more binding partners.
s Means for isolating the peptide, polypeptide, or protein binding partners, or the protein complex, include any means of protein isolation known to the skilled protein chemist, such as, for example, size exclusion chromatography, ion-exchange (anion or cation exchange) chromatography, reverse phase chromatography, or affinity chromatography. Both high pressure (e.g. HPLC, io FLPC, MALDI) and low pressure systems can be used.
Affinity methods using ligands or antibodies that bind to one or both of the binding partners to the protein-protein interaction are particularly preferred.
Antibodies against a protein domain of an EDD protein or one of its binding partners are is particularly useful for isolating EDD or a complex comprising same. In one embodiment, naturally-occurring or recombinant protein is purified free of conspecific proteins by providing a matrix comprising antibody coupled to activated chromatographic resin (eg. CNBr-activated Sepharose, Pharmacia), blocking the resin and washing to remove unbound antibody and blocking agent, 2o contacting the resin with a protein extract comprising a peptide or polypeptide to which the antibody binds under conditions sufficient to allow binding of said peptide or polypeptide (e.g., high ionic strength buffers in the presence of detergent), and eluting said peptide or polypeptide under conditions that disrupt the antibody antigen binding (eg, a buffer of pH 2-3 or a high concentration of a 2s chaotrope, such as urea or thiocyanate ion).
It will be apparent from the preceding description that small molecules, or proteins capable of binding to one of the binding partners, can also be used to isolate one or both binding partners, or the protein complex per se, by affinity means.
3o Conditions to permit such isolation can be readily determined by those skilled in protein chemistry. Selection of buffer pH, ionic strength, and temperature, sufficient to maintain the binding partners in solution are generally preferred.
Preferably, one or more protease inhibitors (e.g. papain, PMSF, leupeptin) are included to prevent proteolytic digestion or degradation of the isolated polypeptides. For example, naturally-occurring or recombinant protein is purified s free of conspecific proteins by providing a matrix comprising a small molecule or protein binding partner coupled to activated chromatographic resin (eg. CNBr-activated Sepharose, Pharmacia), blocking the resin and washing to remove unbound material and blocking agent, contacting the resin with a protein extract comprising a peptide or polypeptide to which the antibody binds under conditions io sufficient to allow binding of said peptide or polypeptide, and eluting said peptide or polypeptide under conditions that disrupt the binding.
In another embodiment, the invention provides methods for producing a protein complex described herein by recombinant means. For expressing peptides or is polypeptides by recombinant means, a protein-encoding nucleotide sequence is placed in operable connection with a promoter or other regulatory sequence capable of regulating expression in a cell-free system or cellular system.
In one embodiment of the invention, nucleic acid comprising a sequence that 2o encodes an EDD protein or a portion of an EDD protein and a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, and a progesterone receptor protein or a portion of said polypeptide sufficient to bind to said EDD protein or said portion of 2s an EDD protein, in operable connection with a suitable promoter sequence, is expressed in a suitable cell for a time and under conditions sufficient for expression to occur.
Nucleic acid encoding the binding partners is readily derived from the nucleotide 3o and amino acid sequences set forth herein, which except for SEQ ID Nos: 3-7, are publicly available. To produce a fusion polypeptide, the open reading frames are covalently linked in the same reading frame, such as, for example, using standard cloning procedures as described by Ausubel et al. (Current Protocols in Molecular Biology, Wiley Interscience, ISBN 047150338, 1992), which is herein incorporated by reference. m Optionally, a spacer is placed between the open reading frames of the binding partners to facilitate their physical relation. Preferred spacers comprise protein-encoding nucleotide sequences of at least about 15-30 nucleotides in length, preferably sequences encoding amino acids rich in proline. The spacer is io designed such that it does not interrupt the open reading frames of the partners.
Reference herein to a "promoter" is to be taken in its broadest context and includes the transcriptional regulatory sequences of a classical genomic gene, including the TATA box which is required for accurate transcription initiation, with is or without a CCAAT box sequence and additional regulatory elements (i.e., upstream activating sequences, enhancers and silencers) which alter gene expression in response to developmental and/or external stimuli, or in a tissue-specific manner. In the present context, the term "promoter" is also used to describe a recombinant, synthetic or fusion molecule, or derivative which confers, 2o activates or enhances the expression of a nucleic acid molecule to which it is operably connected, and which encodes the polypeptide or peptide fragment.
Preferred promoters can contain additional copies of one or more specific regulatory elements to further enhance expression and/or to alter the spatial expression and/or temporal expression of the said nucleic acid molecule.
Placing a nucleic acid molecule under the regulatory control of, i.e., "in operable connection with", a promoter sequence means positioning said molecule such that expression is controlled by the promoter sequence. Promoters are generally positioned 5' (upstream) to the coding sequence that they control. To construct 3o heterologous promoter/structural gene combinations, it is generally preferred to position the promoter at a distance from the gene transcription start site that is -6~-approximately the same as the distance between that promoter and the gene it controls in its natural setting, i.e., the gene from which the promoter is derived.
Furthermore, the regulatory elements comprising a promoter are usually positioned within 2 kb of the start site of transcription of the gene. As is known in s the art, some variation in this distance can be accommodated without loss of promoter function. Similarly, the preferred positioning of a regulatory sequence element with respect to a heterologous gene to be placed under its control is defined by the positioning of the element in its natural setting, i.e., the genes from which it is derived. Again, as is known in the art, some variation in this distance io can also occur.
The prerequisite for producing intact polypeptides and peptides in bacteria such as E, coli is the use of a strong promoter with an effective ribosome binding site.
Typical promoters suitable for expression in bacterial cells such as E. coli include, is but are not limited to, the lacz promoter, temperature-sensitive ~~ or AR
promoters, T7 promoter or the IPTG-inducible tac promoter. A number of other vector systems for expressing the nucleic acid molecule of the invention in E. coli are well-known in the art and are described, for example, in Ausubel et al (In:
Current Protocols in Molecular Biology. Wiley Interscience, ISBN 047150338, 1987) or 2o Sambrook et al (In: Molecular cloning, A laboratory manual, second edition, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1989). Numerous plasmids with suitable promoter sequences for expression in bacteria and efficient ribosome binding sites have been described, such as for example, pKC30 (~,~: Shimatake and Rosenberg, Nature 292, 128, 1981 ); pKK173-3 (tac: Amann and Brosius, 2s Gene 40, 183, 1985), pET-3 (T7: Studier and Moffat, J. Mol. Biol. 789, 113, 1986);
the pBAD/TOPO or pBAD/Thio-TOPO series of vectors containing an arabinose-inducible promoter (Invitrogen, Carlsbad, CA), the latter of which is designed to also produce fusion proteins with thioredoxin to enhance solubility of the expressed protein; the pFLEX series of expression vectors (Pfizer Inc., CT, USA);
30 or the pQE series of expression vectors (Qiagen, CA), amongst others.

Typical promoters suitable for expression in viruses of eukaryotic cells and eukaryotic cells include the SV40 late promoter, SV40 early promoter and cytomegalovirus (CMV) promoter, CMV IE (cytomegalovirus immediate early) promoter amongst others. Preferred vectors for expression in mammalian cells s (eg. 293, COS, CHO, 10T cells, 293T cells) include, but are not limited to, the pcDNA vector suite supplied by Invitrogen, in particular pcDNA 3.1 myc-His-tag comprising the CMV promoter and encoding a C-terminal 6xHis and MYC tag; and the retrovirus vector pSRatkneo (Muller et al., Mol. Cell. viol., 11, 1785, 1991 ).
The vector pcDNA 3.1 myc-His (Invitrogen) is particularly preferred for expressing io a secreted form of a protein in 293T cells, wherein the expressed peptide or protein can be purified free of conspecific proteins, using standard affinity techniques that employ a Nickel column to bind the protein via the His tag.
A wide range of additional hostivector systems suitable for expressing polypeptide is binding partners or immunological derivatives thereof are available publicly, and described, for example, in Sambrook et al (In: Molecular cloning, A laboratory manual, second edition, Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y., 1989).
2o Means for introducing the isolated nucleic acid molecule or a gene construct comprising same into a cell far expression are well-known to those skilled in the art. The technique used for a given organism depends on the known successful techniques. Means for introducing recombinant DNA into animal cells include microinjection, transfection mediated by DEAE-dextran, transfection mediated by 2s liposomes such as by using lipofectamine (Gibco, MD, USA) andlor cellfectin (Gibco, MD, USA), PEG-mediated DNA uptake, electroporation and microparticle bombardment such as by using DNA-coated tungsten or gold particles (Agracetus Inc., WI, USA) amongst others.
3o In another embodiment, nucleic acid comprising a sequence encoding each binding partner is placed in operable connection with a promoter sequence and expressed in a suitable cell. If the protein partners are expressed in the same cell, they may freely associate in said cell to form the protein complex of the invention. If the protein partners are produced in different cells, the cells are lysed and the cellular lysates mixed under conditions sufficient to permit the association s of the binding partners.
In accordance with this embodiment, the nucleotide sequences encoding the binding partners may be contained in the same or different nucleic acid molecules, and as a consequence, the use of single or multiple gene constructs to express io the binding partners is clearly encompassed by the invention. The requirements for expressing fusion polypeptides as described herein above are also relevant in this context, except that there is no need for a spacer. Generally, different promoters will be used to express each binding partner, such as, for example, to prevent squelching or competition between promoters or regulatory sequences for is cellular transcription factors.
Another embodiment of the present invention provides prognostic and diagnostic methods for determining a predisposition for disease, or a disease state, said methods comprising detecting a protein complex comprising:
20 (i) an EDD protein; and (ii) a nuclear protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, a progesterone receptor protein and a 2s protein associated with vascularization.
In one embodiment, the diagnostic/prognostic methods described herein detect a protein complex selected from the group consisting of (i) a complex comprising EDD and CHK2; (ii) a complex comprising EDD and BRCA2; (iii) a complex 3o comprising EDD and CIB; (iv) a complex comprising EDD and importin alpha-1;
(v) a complex comprising EDD and importin alpha-3; (vi) a complex comprising EDD and importin alpha-5; and (vii) a complex comprising EDD and progesterone rece pto r.
In one embodiment, the invention relates to reagents and methods for detecting s specific interaction between an EDD protein and a nuclear protein having tumor suppressor activity wherein the protein-protein interaction is associated with unregulated cell division or hyperproliferation of a cell or the appearance or occurrence of tumors associated with cancer, such as, for example, ovarian cancer or hepatocellular carcinoma or squamous cell carcinoma or breast cancer io or metastatic melanoma.
In another embodiment, the invention relates to reagents and methods for detecting specific interaction between an EDD protein and a nuclear protein having cell cycle modulatory activity wherein the protein-protein interaction is is associated with unregulated cell division or hyperproliferation of a cell or the appearance or occurrence of tumors associated with cancer, such as, for example, ovarian cancer or hepatocellular carcinoma or squamous cell carcinoma or breast cancer or metastatic melanoma.
2o In another embodiment, the invention relates to reagents and methods for detecting specific interaction between an EDD protein and a protein associated with DNA damage or DNA repair wherein the protein-protein interaction is associated with unregulated cell division or hyperproliferation of a cell or the appearance or occurrence of tumors associated with cancer, such as, for 2s example, ovarian cancer or hepatocellular carcinoma or squamous cell carcinoma or breast cancer or metastatic melanoma.
In another embodiment, the invention relates to reagents and methods for detecting specific interaction between an EDD protein and a progesterone 3o receptor protein, wherein the protein-protein interaction activates the receptor in a ligand-dependent manner such as, for example, to enhance progesterone-mediated tumorigenesis or tumor growth or unregulated cell division or hyperproliferation of a cell.
In another embodiment, the invention relates to reagents and methods for s detecting specific interaction between an EDD protein and a protein associated with vascularization, wherein the protein-protein interaction is associated with formation of new blood vessels in a subject. Preferably, the protein-protein interaction is associated with vasculogenesis or angiogenesis in a subject suffering from cancer, psoriasis, diabetic blindness, age related macular to degeneration or rheumatoid arthritis.
In another embodiment, the invention relates to reagents and methods for detecting specific interaction between an EDD protein and a cafcium/integrin binding protein (CIB) or DNA-dependent protein kinase interacting protein (KIP), is wherein the protein-protein interaction is reduced in cells suffering DNA
damage.
Preferred detection systems contemplated herein include any known assay for detecting a protein-protein interaction in a biological sample isolated from a human or mammalian subject, such as, for example, using one or more antibodies 2o against the complex or each binding partner, or an epitope thereof.
Alternatively, a non-antibody ligand of the protein complex may be used, such as, for example, a small molecule (e.g. a chemical compound, agonist, antagonist, allosteric modulator, competitive inhibitor, or non-competitive inhibitor, of the complex that may or may not modulate complex formation or dissociation). ' The use of antibody-based assay systems is particularly preferred. In accordance with these embodiments, the antibody or small molecule may be used in any standard solid phase or solution phase assay format amenable to the detection of protein complexes or protein-protein interactions.
Antibodies that specifically bind to the protein complex are used for the diagnosis of conditions or diseases characterized by the presence of said protein complex, or in prognostic assays to monitor disease progression in the presence of absence of treatment. Diagnostic assays for include methods which utilize the antibody and a label to detect the protein complex in human body fluids or s extracts of cells or tissues. The antibodies are used with or without modification, and may be labeled, either covalently or non-covalently, with a reporter molecule.
A wide variety of reporter molecules which are known in the art may be used, several of which are described above.
io A variety of protocols, including ELISA, RIA, and FACS, for measuring the protein complex are known in the art or described herein. Such methods provide a basis for diagnosing levels of the protein complex associated with disease. For example, a protein complex of the present invention that is associated with cancer induced by over expression of EDD and/or one of its binding partners, (e.g.
is EDD/CHK2 or EDD/BRCA2) can provide a poor prognosis of survival from the disease. Normal or standard values of the complex for a healthy individual are established by combining body fluids or cell extracts taken from normal or healthy subjects, preferably human subjects.
2o The amount of standard complex formation may be quantified by various methods, preferably by photometric means, or using antibodies in a quantitative immunoassay (e.g. ELISA), wherein the amount of protein complex is determined by comparison against known amounts of a standard peptide, such as, for example, a peptide comprising an EDD protein domain.
Quantities of the protein complex expressed in subject samples from biopsied tissues are compared with the standard values. Deviation between standard and subject values establishes the parameters for diagnosing disease or establishing a prognosis. In the case of cancerous tissues, a level of a protein complex in 3o excess of the standard level of that protein complex detected in a healthy subject, is diagnostic of disease, and indicates a poor prognosis for survival.

Optical or fluorescent detection, such as, for example, using mass spectrometry, MALDI-TOF, biosensor technology, evanescent fiber optics, or fluorescence resonance energy transfer, is clearly encompassed by the present invention.
s In biosensor diagnostic devices, the assay substrate and detector surface are integrated into a single device. One general type of biosensor employs an electrode surface in combination with current or impedance measuring elements for detecting a change in current or impedance in response to the presence of a io protein-protein binding event (e.g. U.S. Patent No. 5,567,301 ).
Gravimetric biosensors employ a piezoelectric crystal to generate a surface acoustic wave whose frequency, wavelength andlor resonance state are sensitive to surface mass on the crystal surface. The shift in acoustic wave properties is therefore indicative of a change in surface mass, such as, for example, as a consequence is of protein-protein binding (e.g. U.S. Patent Nos. 5,478,756 and 4,789,804.
Siosensors based on surface plasmon resonance (SPR) effects have also been proposed, for example, in U.S. Patent Nos. 5,485,277 and 5,492,840, which exploit the shift in SPR surface reflection angle that occurs when protein binds to the SPR interface. Finally, a variety of biosensors that utilize changes in optical 2o properties at a biosensor surface are known, (e.g., U.S. Patent No.
5,268,305).
Biosensors have a number of potential advantages over binding assay systems having separate reaction substrates and reader devices. One important advantage is the ability to manufacture small-scale, but highly reproducible, 2s biosensor units using microchip manufacturing methods, such as, for example, described in U.S. Patent Nos. 5,200,051 and 5,212,050. Another advantage is the potentially large number of different analyte detection regions that can be integrated into a single biosensor unit, allowing sensitive detection of several analytes with a very small amount of body-fluid sample. Accordingly, the 3o simultaneous detection of the individual binding partners that form the protein complex, or the simultaneous detection of one or more protein complexes of the present invention, is possible using a biosensor Evanescent biosensors are particularly preferred because they do not require separation of the protein complex from unbound material, and their use can be s coupled to standard immunoassay formats, as originally described by Hirshfield in U.S. Patent No. 4,447,546. In general, evanescent biosensors rely upon light of a particular wavelength interacting with a fluorescent molecule, such as, for example, a fluorescent antibody or small molecule attached near the probe's surface, to emit fluorescence at another wavelength, on binding of the protein to complex of the invention to the antibody or small molecule. The biosensor is protected from sensitivity degradation caused by non-specific binding of proteins to the sensor surface, by exposing the sensor surface to a solution of non-interfering proteins, so that the non-interfering proteins bind to said sensor surface to prevent the subsequent binding of the interfering proteins. Enhanced protection is of surfaces from biological proteins is also possible by completely covering surfaces with protective coatings, such as, for example, amorphous copolymers of tetrafluoroethylene and bis-2,2-trifluoromethyl-4.5-difluoro-1,2-dioxole, dissolved in a solvent containing fluorinated alkanes, and applied by deposition as a thin protective coating (US. Patent No. 5,356,668 by Paton et al.).
Assay systems suitable for use in high throughput screening of mass samples, particularly a high throughput spectroscopy resonance method (e.g. MALDI-TOF, electrospray MS or nano-electrospray MS) or a detection system facilitating determination of real time association/dissociation constants, are particularly 2s contemplated.
In an alternative embodiment, a diagnosis or prognosis is made by separately determining the levels) of expression of the binding partners. In this case, the level of expression of the binding partners is determined by standard protein-3o based detection systems, antibody-based methods, or nucleic acid-based methods. For example, a high level of expression of certain binding partners such as EDD and a progesterone receptor can be indicative of disease, or, in the case of cancerous tissues, provide a poor prognosis for survival. Without being bound by any theory or mode of action, this may be a consequence of EDD trans-activating the progesterone receptor, thereby enhancing progestin-sensitive or s progesterone-receptor mediated cell proliferation.
On the other hand, whilst low levels of CHK2 or BRCA2 coupled with high levels of EDD can be diagnostic of disease or provide a poor prognosis, low levels of EDD coupled with relatively high levels of CHK2 and/or BRCA2 are generally io indicative of a good prognosis. Without being bound by any theory or mode of action, this may be a consequence of EDD preventing normal BRCA2/CHK2 function in cells.
In one embodiment, nucleic acid encoding EDD or a binding partner thereof, such is as, for example, a synthetic oligonucleotide, complementary RNA, DNA, or protein-nucleic acid (PNA), is used to detect and quantitate gene expression in biopsied tissues in which expression of the polypeptide is correlated with disease.
The diagnostic assay may be used to distinguish between absence, presence, and over expression of the binding partner, or to monitor expression following an 2o initial diagnosis or during therapeutic intervention. As with protein detection systems, the detection of over expression of EDD and/or progesterone receptor is preferred.
Co-localization of expression of several binding partners in a particular cell, tissue 2s or organ, such as, for example, using FISH or other expression detection system, is also indicative of disease.
In one embodiment, hybridization with PCR probes capable of detecting the nucleic acid (RNA or genomic DNA) encoding a binding partner is used. The 3o specificity of the probe, is determined by its nucleotide sequence and the stringency of the hybridization or amplification (maximal, high, intermediate, or low). Generally, highly specific probes are preferred for use under more stringent conditions.
To provide a basis for the diagnosis of disease associated with expression of the s binding partners, a normal or standard profile for expression of the partners is established, such as, for example, by combining body fluids or cell extracts taken from normal subjects with nucleic acid encoding the binding partners or a portion thereof, under conditions suitable for hybridization or amplification.
Standard hybridization is then quantified by comparing the values obtained from normal io subjects with the signal obtained using a known amount of a substantially purified nucleic acid. Standard values from normal samples are then compared with values from patient samples. Deviation between standard and subject values is diagnostic of the disease. Once a diagnosis is made by this or another method, hybridization assays are carried out to evaluate expression of the binding partners is over time, or during a course of treatment.
With respect to cancer, the presence of relatively high amounts of RNA
encoding EDD and/or its cognate binding partners, particularly the progesterone receptor, in biopsied tissue from an individual indicates a predisposition for the development 20 of the disease, or is otherwise diagnostic of the disease, preferably prior to the appearance of actual clinical symptoms. Alternatively, or in addition, high levels of these transcripts can be indicative of a poor prognosis for survival.
Methods that are used to quantitate the expression include radiolabeling or 2s biotinylating nucleotides, coamplification of a control nucleic acid, and the use of standard curves onto which the experimental results are interpolated (Melby et al., J. Immunol. Methods, 159, 235-244, 1993; Duplaa et al., Anal. Biochem. 212, 229-236, 1993).
3o Screening assays for identifying modulators of profein complex formation A further embodiment of the present invention provides methods for determining a _78_ modulator of the activity, formation or stability of an isolated or recombinant protein complex comprising:
(i) an EDD protein or a portion of an EDD protein sufficient to bind to a protein selected from the group consisting of a protein having tumor s suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, a progesterone receptor protein and a protein associated with vascularization; and (ii) a nuclear protein selected from the group consisting of a protein io having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, a progesterone receptor protein and a protein associated with vascularization or a portion of said protein sufficient to bind to said EDD protein or said portion of an EDD
is protein.
In one embodiment, the protein complex is selected from the group consisting of:
(i) a complex comprising EDD and CHK2; (ii) a complex comprising EDD and BRCA2; (iii) a complex comprising EDD and CIB; (iv) a complex comprising EDD
2o and importin alpha-1; (v) a complex comprising EDD and importin alpha-3;
(vi) a complex comprising EDD and importin alpha-5; and (vii) a complex comprising EDD and progesterone receptor.
The modulator can enhance complex formation or stability or alternatively, partially 2s or completely inhibits formation of the protein complex, prevent complex formation or enhance complex turnover in cells. In one embodiment, the modulator facilitates or enhances EDD and/or progesterone receptor turnover, particularly in cancer cells.
3o In their general form, the methods of the present invention comprise determining the association or dissociation of the protein complex, or the structure of the complex, in the presence and absence of a candidate compound or a candidate antibody. In accordance with the embodiment described herein, a modified association, dissociation, or structure, of the protein complex in the presence of a candidate compound or a candidate antibody indicates that the candidate is a s modulator of the protein complex.
The association, dissociation, or structure of the complex may be determined by direct means, such as, for example, by determining ~ real time association or dissociation constants in the presence and absence of the candidate, or modified io binding of an antibody that recognizes a conformational epitope of the complex.
Biosensors used essentially as described herein above, in the presence or absence of the candidate compound or antibody, are particularly suited to such applications.
is Alternatively, the association, dissociation, or structure of the complex may be determined by indirect means, such as, for example, using a protein recruitment system, n-hybrid screen, reverse n-hybrid screen, plate agar diffusion assay, ELISA, or other well known assay format for detecting protein-protein interactions.
Such indirect means generally use a reporter system to detect formation or 2o dissociation of the protein complex.
Standard solid-phase ELISA assay formats are particularly useful for identifying antagonists of the protein-protein interaction. In accordance with this embodiment, one of the binding partners (e.g. EDD or a portion thereof) is immobilized on a Zs solid matrix, such as, for example an array of polymeric pins or a glass support.
Conveniently, the immobilized binding partner is a fusion polypeptide comprising Glutathione-S-transferase (GST; e.g. an EDD-GST fusion), wherein the GST
moiety facilitates immobilization of the protein to the solid phase support.
The second binding partner (e.g. progesterone receptor, CHK2, CIB, or BRCA2) in 3o solution is brought into physical relation with the immobilized protein to form a protein complex, which complex is detected using antibodies directed against the second binding partner. The antibodies are generally labelled with fluorescent molecules or conjugated to an enzyme (e.g. horseradish peroxidase), or alternatively, a second labelled antibody can be used that binds to the first antibody. Conveniently, the second binding partner is expressed as a fusion s polypeptide with a FLAG or oligo-histidine peptide tag, or other suitable immunogenic peptide, wherein antibodies against the peptide tag are used to detect the binding partner. Alternatively, oligo-HIS tagged protein complexes can be detected by their binding to nickel-NTA resin (Qiagen), or FLAG-labeled protein complexes detected by their binding to FLAG M2 Affinity Gel (Kodak). It will be to apparent to the skilled person that the assay format described herein is amenable to high throughput screening of samples, such as, for example, using a microarray of bound peptides or fusion proteins.
In a modification of the standard ELISA-type assay format, a binding partner is is immobilized on a solid support, such as by chemical synthesis thereon, or biotin-labelled and used in the liquid phase.
A two-hybrid assay is described in US Patent No. 6,316,223 to Payan et al., incorporated herein by reference. The basic mechanism described by Payan et 2o al. is similar to the yeast two hybrid system. In the two-hybrid system, the binding partners are expressed as two distinct fusion proteins in a mammalian host cell.
In adapting the standard two-hybrid screen to the present purpose, a first fusion protein consists of a DNA binding domain which is fused to one of the binding partners, and a second fusion protein consists of a transcriptional activation 2s domain fused to the other binding partner. The DNA binding domain binds to an operator sequence which controls expression of one or more reporter genes. The transcriptional activation domain is recruited to the promoter through the functional interaction between binding partners. Subsequently, the transcriptional activation domain interacts with the basal transcription machinery of the cell, thereby 3o activating expression of the reporter gene(s), the expression of which can be determined. Candidate bioactive agents that modulate the protein-protein interaction between the binding partners are identified by their ability to modulate transcription of the reporter genes) when incubated with the host cell.
Antagonists will prevent or reduce reporter gene expression, while agonists will enhance reporter gene expression. In the case of small molecule modulators, s these are added directly to the cell medium and reporter gene expression determined. On the other hand, peptide modulators are expressible from nucleic acid that is transfected into the host cell and reporter gene expression determined.
In fact, whole peptide libraries can be screened in transfected cells.
io Alternatively, reverse two hybrid screens, such as, for example, described by Vidal et al., Proc. Natl Acad. Sci USA 93, 10315-10320, 1996, may be employed to identify antagonist molecules. Reverse hybrid screens differ from froward screens supra in so far as they employ a counter-selectable reporter gene, such as for example, CYH2 or LYS2, to select against the protein-protein interaction. Cell Is survival or growth is reduced or prevented in the presence of a non-toxic substrate of the counter-selectable reporter gene product, which is converted by said gene product to a toxic compound. Accordingly, cells in which the protein-protein interaction of the invention does not occur, such as in the presence of an antagonist of said interaction, survive in the presence of the substrate, because it 2o will not be converted to the toxic product. For example, EDD can be expressed as a DNA binding domain fusion, such as with the DNA binding domain of GAL4, and the portion of a progesterone receptor or CHK2 or BRCA2 that binds EDD is expressed as an appropriate transcription activation domain fusion polypeptide (e.g. with the GAL4 transcription activation domain). The fusion polypeptides are 2s expressed in yeast in operable connection with the URA3 counter-selectable reporter gene, wherein expression of URA3 requires a physical relation between the GAL4 DNA binding domain and transcriptional activation domain. This physical relation is achieved, for example, by placing reporter gene expression under the control of a promoter comprising nucleotide sequences to which GAL4 3o binds. Cells in which the reporter gene is expressed do not grow in the presence of uracil and 5-fluororotic acid (5-FOA), because the 5-FOA is converted to a toxic compound. Candidate peptide inhibitors) are expressed in libraries of such cells, wherein cells that grow in the presence of uracil and 5-FOA are retained for further analysis, such as, for example, analysis of the nucleic acid encoding the candidate peptide inhibitor(s). Small molecules that antagonize the interaction are s determined by incubating the cells in the presence of the small molecules and selecting cells that grow or survive of cells in the presence of uracil and 5-FOA.
Alternatively, a protein recruitment system, such as that described in U.S.
Patent No. 5, 776, 689 to Karin et al., is used. In a standard protein recruitment system, io a protein-protein interaction is detected in a cell by the recruitment of an effector protein, which is not a transcription factor, to a specific cell compartment.
Upon translocation of the effector protein to the cell compartment, the effector protein activates a reporter molecule present in that compartment, wherein activation of the reporter molecule is detectable, for example, by cell viability, indicating the is presence of a protein-protein interaction. More specifically, the components of a protein recruitment system include a first expressible nucleic acid encoding a first fusion protein comprising the effector protein and one of the binding partners (e.g.
progesterone receptor or a portion thereof or CHK2 or a portion thereof or BRCA~
or a portion thereof), and a second expressible nucleic acid molecule encoding a 2o second protein comprising EDD with the NLS intact. The reporter molecule in this context would comprise a molecule or cellular 'event that is regulated by the effector protein. A cell line or cell strain in which the activity of an endogenous effector protein is defective or absent (e.g. a yeast cell or other non-mammalian cell), is also required, so that, in the absence of the protein-protein interaction, the as reporter molecule is not expressed. In use, a complex is formed between the binding partner moiety of the fusion polypeptide and EDD, thereby directing translocation of the complex to the nucleus mediated by an importin protein binding to the EDD NLS, wherein the effector protein then activates the reporter molecule. Such a protein recruitment system can be practiced in essentially any 3o type of cell, including, for example, mammalian, avian, insect and bacterial cells, and using various efEector protein/reporter molecule systems.

Alternatively, a yeast cell based assay can be performed in which the interaction between EDD and one or more of its binding partners results in the recruitment of a guanine nucleotide exchange factor (GEF) to the plasma membrane, wherein s GEF activates a reporter molecule, such as Ras, thereby resulting in the survival of cells that otherwise would not survive under the particular cell culture conditions. Suitable cells for this purpose include, for example, Saccharomyces cerevisiae cdc25-2 cells, which grow at 36°C only when a functional GEF
is expressed therein (Petitjean et al., Genetics 124, 797-806, 1990) Translocation of to the GEF to the plasma membrane is facilitated by a plasma membrane localization domain. Activation of Ras is detected, for example, by measuring cyclic AMP levels in the cells using commercially available assay kits and/or reagents. To detect modulators of the protein-protein interaction of the present invention, duplicate incubations are carried out in the presence and absence of a is test compound, or in the presence or absence of expression of a candidate modulatory peptide in the cell. Reduced survival or growth of cells in the presence of a candidate compound or candidate peptide indicates that the peptide or compound is an antagonist of the interaction between EDD and one or more of its binding partners.
A "reverse" protein recruitment system is also contemplated, wherein modified survival or modified growth of the cells is contingent on the disruption of the protein-protein interaction by the candidate compound or candidate peptide.
For example, NIH 3T3 cells that constitutively express activated Ras in the presence 2s of GEF can be used, wherein the absence of cell transformation is indicative of disruption of the protein complex by a candidate compound or peptide. In contrast, NIH 3T3 cells that constitutively express activated Ras in the presence of GEF
have a transformed phenotype (Aronheim et al., Cell. 78, 949-961, 1994) 3o In yet another embodiment, small molecules are tested for their ability to dissociate the protein complex of the invention, by an adaptation of plate agar diffusion assay described by Vidal and Endoh, TIBS 77, 374-331, 1999, which is incorporated herein by reference.
In a further embodiment, a modulator is determined by a process comprising:
s (i) determining the level of a protein complex selected from the group consisting of: (i) a complex comprising EDD and CHK2; (ii) a complex comprising EDD and BRCA2; (iii) a complex comprising EDD and CIB; (iv) a complex comprising EDD and importin alpha-1; (v) a complex comprising EDD and importin alpha-3; (vi) a complex comprising EDD and importin to alpha-5; and (vii) a complex comprising EDD and progesterone receptor in the absence of a candidate compound or candidate antibody; and (ii) determining the level of said protein complex in the presence of a candidate compound or in the presence of said candidate antibody wherein a difference in the level of said protein complex at (i) and (ii) indicates that is the candidate compound or candidate antibody is a modulator of said interaction.
This embodiment of the invention applies mutatis mutandis to the determination of protein complexes comprising a portion of any one or more of the protein binding partners.
It will be understood by those skilled in the art that any one or more of the assay methods for antagonists as described herein above can be adapted for this purpose. This is because the level of the protein complex in the presence or absence of a candidate compound or antibody is related to antibody binding in the 2s case of ELISAs, or to cell survival or growth, in the case of hybrid screens or protein recruitment assays. ELISA-based assay formats are particularly suitable for this purpose, because they are readily quantifiable, by calibrating the detection system against known amounts of a protein standard to which the antibody binds.
Such quantitation is well known to the skilled person.

The modulators identified using the methods described herein are useful for the therapeutic or prophylactic treatment of hyperproliferative disorders, or disorders associated with aberrant cell cycle regulation, aberrant DNA damage or repair, aberrant vascularization, progestin-sensitive disorders or progesterone receptor-s mediated disorders such as, for example, aberrant cell division, tumorigenesis, tumor metastasis, or tumor cell invasion. The modulators are preferably useful for the treatment of one or more symptoms associated with a cancer selected from the group consisting of squamous cell carcinoma, hepatocellular carcinoma, ovarian cancer, breast cancer, melanoma, head and neck cancer, to adenocarcinoma, squamous lung cancer, gastrointestinal cancer (eg. gastric, colon, or pancreatic cancer), renal cell cancer, bladder cancer, prostate cancer, non-squamous carcinoma, glioblastoma and medulloblastoma. Alternatively, or in addition, the modulators are preferably useful for the treatment of disorders or conditions associated with aberrant vascularisation, such as those disorders is associated with excessive vascularisation, eg some forms of aggressive cancer, diabetic blindness, age-related macular degeneration, rheumatoid arthritis and psoriasis, and those disorders associated with insufficient vascularisation, eg coronary artery disease, stroke, and delayed wound healing. Additionally, it is preferable that the modulators are useful for inducing wound healing, stimulating 20 organ regeneration, stimulating follicle development in the corpus luteum, stimulating placental growth during pregnancy and stimulating embryonic growth during pregnancy.
Therapeutic applications 2s In another embodiment, the present invention provides a method for treating a condition associated with elevated expression of an EDD protein in a cell, said method comprising administering an amount of a compound effective to reduce EDD expression in a cell.
3o In one embodiment, the condition associated with EDD over expression is a cancer, particularly a cancer selected from the group consisting of squamous cell carcinoma, hepatocellular carcinoma, ovarian cancer, breast cancer, melanoma, head and neck cancer, adenocarcinoma, squamous lung cancer, gastrointestinal cancer (eg. gastric, colon, or pancreatic cancer), renal cell cancer, bladder cancer, prostate cancer, non-squamous carcinoma, giioblastoma and medulloblastoma.
s However, it is to be understood that the inventive method is suitable for preventing any cell advancing into mitosis if administered at an appropriate time and in a suitable amount.
In one embodiment, the compound administered comprises nucleic acid, to Preferably, the' nucleic acid is an antagonist of EDD expression, such as, for example, an antisense nucleic acid, peptide nucleic acid (PNA), ribozyme, or interfering RNA, which is complementary, in whole or in part, to a target molecule comprising a sense strand, and can hybridize with the target molecule, in particular, EDD-encoding RNA. When introduced into a cell using suitable is methods, such a nucleic acid inhibits the expression of the EDD gene encoded by the sense strand. Antisense nucleic acid, ribozymes (eg. Cech et aL,USSN
4,987,071; Cech et al., USSN 5,116,742; Bartel and Szostak, Science 261, 1411-1418, 1993), nucleic acid capable of forming a triple helix (eg. Helene, Anticancer Drug Res. 6, 569-584, 1991 ), PNAs (Hyrup et al., Bioorganic & Med. Chem. 4, 5-20 23, 1996; O'Keefe et al., Proc. Natl Acad. Sci. USA 93, 14670-14675, 1996), interfering RNAs (Elbashir et al., Nature 411, 494-498, 2001; Sharp, Genes Devel.
15, 485-490, 2001; Lipardi et al., Cell 107, 297-307, 2001; Nishikura, Cell 107, 415-418, 2001 ) or small interfering RNAs (siRNA) may be produced by standard techniques known to the skilled artisan, based upon the sequences disclosed 2s herein.
Preferably, the antisense nucleic acid, ribozyme, PNA, interfering RNA or siRNA
comprises a sequence that is complementary to at least about 15-20 contiguous nucleotides of a sequence having at least about 80% identity to SEQ ID NO: 1 or 3o SEQ ID NO: 3 (ie, it is complementary to EDD-encoding mRNA) and can hybridize thereto. For example, such antagonistic nucleic acid can be complementary to a _g7_ target nucleic acid having the sequence of SEQ ID NOs:1 or 3 or a portion thereof sufficient to allow hybridization. Longer molecules, comprising a sequence that is complementary to at least about 25, or 30, or 35, or 40, or 45, or 50 contiguous nucleotides of EDD-encoding mRNA are also encompassed by the present s invention.
As exemplified herein, the use of interfering RNA, particularly siRNA is preferred for down-regulating EDD expression in a cell, thereby inhibiting cellular proliferation and causing cells to accumulate in the G2/M phase of the cell cycle, to or causing altered cell-cell contacts, altered cell shape and disorganisation. Such interfering RNAs generally comprise an RNA molecule having a region of self-complementarity capable of forming a double stranded RNA.
In one embodiment, a construct comprising an antisense nucleic acid, ribozyme, Is PNA, interfering RNA or siRNA, can be introduced into a suitable cell to inhibit EDD expression and/or activity therein. In another embodiment, such a construct can be introduced into some or all of the cells of a mammal. The antisense nucleic acid, ribozyme, PNA, or interfering RNA, inhibits EDD expression and the subsequent formation of deleterious protein-protein complexes involving an EDD
2o protein. Accordingly, a cancer, or a hyperproliferative process that is mediated by EDD in the cell containing the construct is inhibited.
The use of antibodies that can inhibit one or more functions characteristic of a EDD protein, such as a binding activity, a signalling activity, and/or stimulation of a 2s cellular hyperproliferative response, is also encompassed by the present invention. In one embodiment, antibodies of the present invention can inhibit binding of a ligand (i.e., one or more ligands) to a mammalian EDD protein and/or can inhibit one or more functions mediated by a mammalian EDD protein in response to ligand binding. In a particularly preferred embodiment, the antibodies 3o can inhibit (reduce or prevent) the interaction of EDD with a natural ligand such a the progesterone receptor.

_g$_ One or more agents can be administered to the host by an appropriate route, either alone or in combination with another drug. An effective amount ofi a nucleic acid or antibody agent having antagonist or agonist activity is administered.
An s effective amount is an amount sufficient to achieve the desired therapeutic or prophylactic effect, under the conditions of administration, such as an amount sufficient for inhibition or promotion of EDD function.
For the treatment of cancers it is particularly preferred to target the expression of io EDD in one or more specific cells or tissues, such as, for example, a cancer tissue or cell, thereby ensuring that the active compound is delivered to that cell/tissue and does not inhibit cell proliferation generally. Antibodies recognizing tumor-specific antigens have been used to deliver cytotoxic drugs to tumors.
Antibodies recognizing tumor-specific antigens can be conjugated to the active compound, is however in the case of solid tumors, such immunoconjugates may be less effective in penetrating tumor tissue.
Arap et al., Science, 279, 377-380, 1998 blocked tumor growth indirectly by inhibiting angiogenesis using a peptide that localized to endothelial cells 2o associated with human breast carcinoma xenografts. By conjugating the cytotoxic drug doxorubicin to the peptide, a selective destruction of blood vessels associated with tumors was observed. This, in turn, resulted in the necrosis of the tumor and an increase in the survivability of the tumor-bearing mice.
2s Hong et al., (published US Patent Publicaton No. 20020102265; USSN
09/899,376) describe the isolation of a peptide (HN-1 ) that is specifically internalized by human squamous carcinoma , and solid tumor tissue cells, such as breast cancer cells. Accordingly, an anticancer compounds that targets an Edd gene expression product, can be conjugated to HN-1 and administered 3o specifically to tumor tissue such as, for example, a squamous cell carcinoma (e.g.
of the oral cavity, pharynx, throat, paranasal sinus, nasal cavity, larynx, thyroid, -parathyroid, salivary gland, skin of the face, skin of the neck or cervical lymph node) such as a squamous cell carcinoma of the tongue or head and neck, breast cancer, glioblastoma, or astrocytoma. Such conjugates can be delivered by intravenous administration, intratumoral administration, subcutaneous s administration, intraperitoneal administration or topical administration. In an additional specific embodiment the conjugate is administered by local, regional or systemic administration.
Alternatively, nucleic acid encoding an inhibitor of an EDD expression product, to such as, for example nucleic acid encoding siRNA capable of targeting Edd gene over expression or the formation of an EDD-containing protein complex, is introduced to a subject in need of treatment and expressed therein operably under the control of a suitable tumor-specific promoter sequence. For example, an infectious recombinant viral vector expressing the RNA can be targeted to tumor is cells through cellular surface receptors by genetic or biochemical modification of the viral surface. Alternatively, cancer cells are targeted at the transcriptional level using lineage-specific promoters that restrict expression of the effector gene to a tumor cell and any related normal cell derived from the same developmental lineage. Examples of tumor types that have been targeted in this manner include 2o tumors of the colon, lung; breast, hepatocellular carcinoma and melanoma.
Tumor-specific promoters/enhancers have also been used in a therapeutic approach called "virus-directed enzyme/prodrug therapy" (VDEPT), wherein tumor-killing efficacy can be enhanced. with reduced side effects on normal cells (the so-called "bystander effect"). For example, the alpha-fetoprotein ~ (AFP) 2s promoter/enhancer cassette has been utilized to control E1 expression from an Adenoviral vector, to induce a virus-mediated oncolytic effect on hepatocellular carcinoma. Thus, a tumor specific replication competent adenoviral (TSRCA) vector comprising the alpha-fetoprotein promoter to deliver a gene encoding siRNA is particularly preferred. Alternatively, a variation of this system, the 30 "Complementary-Adenoviral Vector System" as described in US Patent Publication No. 20020142989 may be employed.

Alternatively, or in addition, the compound effective in reducing EDD
expression can be administered in the form of a liposome such as a cationic liposome.
s A variety of routes of administration are possible including, but not necessarily limited to oral, dietary, topical, parenteral (e.g., intravenous, infra-arterial, intramuscular, subcutaneous injection), and inhalation (e.g., intrabronchial, intranasal or oral inhalation, intranasal drops) routes of administration, depending on the agent and disease or condition to be treated.
io Formulation of an agent to be administered will vary according to the route of administration selected (e.g., solution, emulsion, capsule). An appropriate composition comprising the agent to be administered can be prepared in a physiologically acceptable vehicle or carrier. For solutions or emulsions, suitable is carriers include, for example, aqueous or alcoholic/aqueous solutions, emulsions or suspensions, including saline and buffered media. Parenteral vehicles can include sodium chloride solution, Ringer's dextrose, dextrose and sodium chloride, lactated Ringer's or fixed oils, for instance. Intravenous vehicles can include various additives, preservatives, or fluid, nutrient or electrolyte replenishers and 2o the like (See, generally, Remington's Pharmaceutical Sciences, 17th Edition, Mack Publishing Co., Pa., 1985). For inhalation, the agent can be solubilized and loaded into a suitable dispenser for administration (e.g., an atomizer, nebulizer or pressurized aerosol dispenser).
2s Furthermore, where the agent is a protein or peptide, the agent can be administered via in vivo expression of the recombinant protein. In vivo expression can be accomplished via somatic cell expression according to suitable methods (see, e.g. U.S. Pat. No. 5,399,346). In this embodiment, nucleic acid encoding the protein can be incorporated into a retroviral, adenoviral or other suitable vector 30 (preferably, a replication deficient infectious vector) for delivery, or can be introduced into a~transfected or transformed host cell capable of expressing the protein for delivery. In the latter embodiment, the cells can be implanted (alone or in a barrier device), injected or otherwise introduced in an amount effective to express the protein in a therapeutically effective amount.
s The present invention clearly excludes any previously disclosed isolated protein complex consisting solely of EDD protein and TopBP1.
The present invention is further described by the following non-limiting Examples.
to Example 1 Abnormalities of the EDD gene in human cancer 1.1 Materials and Methods is Tumors and DNA extraction.
Ovarian tumor tissue and matched blood or normal ovarian tissue were obtained from 98 patients and DNA extracted as described previously (Obata et al, Gancer Res. 58, 2095-2097, 1998). Metastatic melanoma tissue and matched blood or normal skin tissue were obtained from 20 patients and DNA isolated as reported 2o previously (Indsto et al, Cancer Genet Gytogenet 100, 68-71, 1998). DNA was extracted from matched normal and hepatocellular carcinoma tissue samples microdissected from 19 primary liver tumors (Macdonald et al, Hepatology 28, 97, 1998). For AI analysis, paraffin-embedded breast cancers and normal blood were obtained from 24 patients. Tumor location was determined by haemotoxylin 2s and eosin staining and cells were microdissected from 4-5 adjacent sections. DNA
was extracted in lysis buffer (0.45% Tween 20, 5mg/ml proteinase K, 0.25% BSA) at 55°C for 8 hours, then boiled for 10 min. DNA was extracted from blood using a Puregene DNA isolation kit (Gentra Systems, Minneapolis, MN).
3o For RT-PCR breast cancer samples were collected at the time of surgery.
Normal breast tissue (based on histological examination) was removed from unaffected regions of the breast at the same time as excision of the cancer.
Paraffin-embedded archival samples from 12 squamous cell carcinoma of the anterior tongue and matched lymph nodes were from a series described by Bova s et al (Bova ef al, Clin. Cancer Res. 5, 2810-2819, 1999). Areas of malignant squamous cells were identified by a pathologist from haemotoxylin and eosin stained slides and were microdissected from unstained adjacent 10pm sections under a light microscope. Normal tissue was gained from uninvolved lymph nodes and/or microdissected from areas of normal cells surrounding the squamous cell io carcinoma. Samples were digested in 250p1 of proteinase K (2mg/ml) for 5 days at 37°C with constant agitation. The digest was then extracted once with phenol, once with chloroform, and DNA precipitated in ethanol overnight and re-dissolved in 30p1 of TE bufFer ("microdissected DNA").
is Isolation and cloning of EDD-linked microsatellites CEDD and 586F~8b.
A human P1 genomic library (Incyte Genomics, CA) was screened with two cDNA
probes covering 4kb of the EDD coding sequence (Callaghan et al, Oncogene 77, 3479-3491, 1998). DNA from positive clones was extracted, digested with Hinc II, separated by gel electrophoresis and re-screened by Southern blotting using an 20 oligonucleotide probe (CA),5. Positive genomic DNA fragments were cloned into pBluescript and sequenced which identified a novel dinucleotide repeat microsatellite CEDD (CA repeat near EDD). Unique primers were designed around the microsatellite to give a product of approximately 220bp; CEDD
forward 5'-TACCCTGCAGTAAATCTCACATGTACTCCC-3' (SED ID NO: 5), CEDD
2s reverse 5'-AGAATCGCTTGAACCTAGTAGGTGAAGGTG-3' (SED ID NO: 6).
Subsequently, EDD genomic sequence became available (Genbank Accession:
AC021004) and CEDD was located in an intron between bases 2616 and 2617 of the EDD coding sequence. The minimum size of the EDD gene is 100 kb, consisting of at least 46 exons. Within the same genomic clone another EDD-3o specific dinucleotide repeat microsatellite was identified and named 586F18b.
Unique primers were designed around this microsatellite to give a product of approximately 200bp; forward 5'-GGTAGGGAACGAAACTGCCAG-3' (SEQ ID
NO: 24), reverse 5'-TGCAAAATAACAATAGCTTTGCTTAG-3' (SEQ ID NO: 25).
586F18b is located in an intron between bases 6631 and 6632 of the EDD coding sequence. Both microsatellites were between 50-55% heterozygous using human s cell line DNA (data not shown).
Microsatellite analysis Microsatellite analysis was used to determine the frequency and distribution of AI
on 8q22.3-24.13 using CEDD, 586F18b and seven other dinucleotide and to tetranucleotide repeat polymorphic markers mapped in this region (Genome Database, www.gdb.orgl) (Figure 1 ). CEDD, D8S326, D8S257, D8S300, D8S545 and D8S85, were used for analysis of ovarian cancers, hepatocellular carcinoma, metastatic melanoma and squamous cell carcinoma of the tongue. The additional markers 586F18b, MYC-PCR.3 and D8S198 were used for analysis of breast is cancers, Primer pairs were obtained from Research Genetics (Huntsville, AL) and Pacific Oligos (Lismore, Australia). Ovarian DNA was analyzed by radio-isotope based methods as previously described (Obata et al, Cancer Res. 58, 2095-2097, 1998). For DNA from other tissues, in each PCR reaction the forward primer was labeled with either 6-FAM or TET fluorescent label. Reactions were performed 2o using 40-60 ng of DNA from hepatocellular carcinoma and metastatic melanoma, 1 pl of breast cancer extracted DNA or 2-3pl microdissected DNA from squamous cell carcinoma of the tongue. The PCR reaction components were as follows: 10 mM Tris-HCI, pH 8.3; 50 mM KCI; 66 pM dNTP mix; 1.5 mM MgCl2; 5 pmoles of both forward and reverse primer and 0.8 U of Amplitaq Gold DNA polymerase 2s (Applied Biosystems, Sydney, Australia). PCR conditions for microsatellites D8S326, D8S257, D8S545, D8S85, D8S198 and MYC-PCR.3 were: 12 min hotstart 95°C; 35 cycles (40 for microdissected DNA) of (1 ) 94°C 15 s, (2) 60°C 15 s (66°C for CEDD, 52°C for 586F18b) and (3) 72°C 30 s; 5 min 72 °C; 4 °C hold.
Amplification of the CEDD microsatellite from microdissected DNA required a 30s 3o annealing step, a 1 min extension step, and 35 cycles. Amplification of the microsatellite D8S300 was performed as follows: 12 min hotstart 95 °C;
35 cycles of (1 ) 94 °G 30 s, (2) 60°C 30s and (3) 72°C 60s; 5 min 72°C; 4°C hold. No PCR
products could be obtained from DNA recovered from paraffin embedded tongue carcinoma with D8S300 primers, presumably because of DNA shearing.
s Duplicate fluorescent products were separated on an ABI 377 DNA Sequencer (Applied Biosystems) and analyzed using Genescan and Genotyper software (Applied Biosystems). Ambiguous results were resolved on separate gels. AI in hepatocellular carcinoma and breast cancer was defined by a reduction in relative fluorescence of heterozygous allele peaks by at least 30% in the tumor DNA
to compared to the matched normal DNA. A reproducible difference of 20-30% in metastatic melanoma was considered significant due to the higher level of contaminating normal DNA present in these samples. A difference of 50% or more was considered to represent Al in squamous cell carcinoma DNA due to a lower level of normal cellular DNA contamination in these samples.
Allelic imbalance (AI) is indicative of either loss or amplification of an allele and (presumably) the surrounding chromosomal region. In practise, loss of heterozygosity (LOH) is not readily distinguishable from amplification by this method, particularly when there is a significant degree of cross-contamination of 2o tumor with normal DNA. In the present study, AI was interpreted as amplification, given that chromosomal gains are much more commonly reported at 8q than losses.
FISH
as FISH analysis was carried out by Erica Woollatt at the Women's and Children's Hospital, Adelaide, Australia. Two P1 plasmids encoding approximately 100kb of EDD genomic sequence used for fluorescence in situ hybridization (FISH) were nick-translated with biotin-14-dATP and hybridized at a final concentration of ng/pl to metaphases from the breast cancer cell lines MDA-MB-436, SKBR-3, BT-30 20 and BT-483. The single copy FISH method was modified from that previously described (fallen ef al, Ann. Genet. 33, 219-221, 1990) in that chromosomes were stained before analysis with both propidium iodide (as counterstain) and DAPI (for chromosome identification). Images of metaphase preparations were captured by a CCD camera using the ChromoScan image collection and enhancement system (Applied Imaging, Newcastle, UK).
s Quantitative RT PCR - primary breast tissue.
RNA was extracted from samples using Trizol reagent (Life Technologies, Paisley Scotland, UK). Reverse transcription was performed, followed by quantitative PCR
using a Taqman based methodology as previously described (AI-Taher et al, io Yeasf 17, 201-210, 2000). PCR primers for EDD were designed within 300 bases of the polyA addition site. The sequences of EDD specific primers were:
forward EDD-407F GCTAGTCACCAACTTCTGGGTCTAA (SEQ ID NO: 26), reverse EDD-4908 CAGCAAAAAGATAAATCACAGTGTAAATT (SEQ ID NO: 27), fluorescent probe EDD-433T FAM-CCCAGCCAAAGATGACAGCAGAACAAC-ls TAMRA (SEQ ID NO: 28). Samples were also analysed for expression of several control genes; CK18 (epithelial content), GAPDH (cellular metabolism), IF2B
(general transcriptional activity) and MCM3 '(proliferation rate). Results are expressed as copies of EDDl3e9 total cDNA per sample, corrected for IF2B.
2o Quantitative PCR and RT PCR - Breast cancer cell lines.
Cells harvested from duplicate 150 cm2 flasks were pooled and RNA was extracted using the RNeasy RNA extraction kit (Qiagen, Melbourne, Australia).
cDNA was made as previously described (see EDD mRNA sequence analysis) from the normal breast epithelial cell lines 184 and the breast cancer cell lines 2s MDA-MB-468, MDA-MB-436, MDA-MB-231, MDA-MB-453, MDA-MB-175, MDA-MB-361, MDA-MB-157, BT-20, T-47D, BT-549, BT-483, MCF-7, BT-474, SK-Br-3, ZR-75-1, Hs 578T, MDA-MB-330. All PCR reactions were run on a LightCycler using LightCycler software 3.5 (Roche Diagnostics, Sydney Australia). Primers for EDD forward, TTAGGCTTTTGGTAAATGGCTGCG (SEQ ID NO: 29), reverse, 3o TGAGGGCATAGGCTGGAATCCTTC (SEQ ID NO: 30), carbonic anhydrase II , forward CCACCCCTCCTCTTCTGGAATG (SEQ ID NO: 31), reverse GCTTTGATTTGCCTGTTCTTCAGTG (SEQ ID NO: 32), p53 ribonucleotase reductase (p53R2), forward GTGACTTTGCTTGCCTGATGTTC (SEQ ID NO: 33), reverse TCTGTGGTTTCTGCCATAACTGC (SEQ ID NO: 34), GAPDH, forward GACATCAAGAAGGTGGTGAA (SEQ ID NO: 35), reverse s TGTCATACCAGGAAATGAGC (SEQ ID NO: 36). All primer pairs spanned at least one intron and products were visualized on an agarose gel to confirm product size. Relative expression of all genes was corrected for cDNA
concentration using GAPDH expression.
to DNA was extracted from the above cell lines using a DNA extraction kit (Stratagene, Sydney, Austalia). PCR reactions to determine genomic copy number utilized the reverse primer from above (unless mentioned). Primers for EDD, forward CATTGCTGACCCTATCCCTGTGTTG (SEQ ID NO: 37), reverse TAGCCCGTGAAATCCTCCCATCTC (SEQ ID NO: 38), CA ll forward Is ACCCGCCTCATGCCTCAGCCTTAC (SEQ ID NO: 39), p53R2 forward TGTCAGCCTTGAGTACCTCCAGGG (SEQ ID NO: 40), beclin forward TAGGTTTGGGGTGAGTGG (SEQ ID NO: 41 ), reverse AGTCTGTGGGCAGCAAGG (SEQ ID NO: 42). All products were visualised on an agarose gel to confirm product size. Relative DNA copy number was corrected 2o using known beclin gene copy number determined by FISH (Aita et al, Genomics 59, 59-65, 1999).
Gene expression profiling - primary ovarian tissue RNA was isolated from 66 primary ovarian cancers and borderline tumors in 2s addition to 4 normal ovary samples using Trizol reagent (Life Technologies, Rockville, MD, USA) essentially according to manufacturer's instructions. RNA
was then reverse transcribed using an oligo(dT) anchored oligonucleotide that additionally comprised a T7 promoter sequence. Isolated cDNA was then transcribed in vitro using the T7 MEGAscript kit (Ambion, Austin, TX, USA) 3o according to manufacturer's instructions. Transcription was performed with biotinylated nucleotides (Bio-11-CTP and Bio-16-UTP) to facilitate detection of the transcribed nucleic acid.
Levels of gene expression in the cancer samples was then determined by analyzing the transcribed cDNA samples using customized Affymetrix GeneChip~
s microarrays that comprise 59,618 oligonucleotide probe sets. These probe sets facilitate analysis of 46,000 gene clusters, representing over 90% of the predicted expressed genome. , Data was normalized, and changes in gene expression detected using a ranked to penalized t-statistic with p-values adjusted for multiple testing using the Holm procedure. Analysis was performed using the LIMMA package (available from Bioconductor, Biostatistics Unit of the Dana Farber Cancer Institute at the Harvard Medical SchooIlHarvard School of Public Health).
is Gene expression in 52 different tissues of the body was also determined using the previously described methods to facilitate the identification of changes in gene expression that are specific for ovarian cancer.
EDD mRNA sequence analysis.
2o Total RNA was extracted from the following human cell lines using the RNeasy Maxi Kit (Qiagen): breast cancer MDA-MB-468, MDA-MB-436, MDA-MB-231, MDA-MB-453, MDA-MB-175, MDA-MB-361, MDA-MB-134, MDA-MB-157, BT-20, T-47D, BT-549, BT-483, MCF-7, BT-474, SK-Br-3, ZR-75-1, Hs 578T; normal breast epithelium HBL-100 (SV-40 transformed), 184, HMEC 219-4; ovarian 2s cancer OVCAR-3, OVCA-420, IGROV-1, SKOV3, A2780; prostate cancer PC-3, DU-145, LnCaP. cDNA was synthesized using the Expand Reverse Transcriptase System (Boehringer Mannheim, Sydney, Australia). 2pg total RNA and 2pl oligo dT (Boehringer Mannheim) was made up to 22p1 with water, heated at 65 °C for min and cooled on ice followed by addition of 8pl Expand Reverse 3o Transcriptase buffer (5x; 250 mM Tris-HCI, 200 mM KCI, 25 mM MgCl2, 2.5 Tween 20 (v/v), pH 8.3), 1 mM each dATP, dCTP, .dGTP and dTTP, 10 mM DTT

and 2 pl (100U) Expand Reverse Transcriptase. The reverse transcriptase reaction was then performed at 42 °C for 45-60 min. Ten PCR products were designed to cover the entire 8.5 kb coding region of the EDD gene. PCR
reactions included 3pl of the reverse transcriptase reaction, 10 pmol of each primer, 1.75 s units Expand High Fidelity DNA polymerase (Boehringer) and 1.5 mM MgCl2.
Amplification was carried out using the following protocol: 2 min denature at °C; 10 cycles of 30s denature, 30s annealing and 60s extension at 72°C; 24 cycles of 30s denature, 30s annealing and 60s extension with a 5s increase per cycle; 5 min extension. Annealing temperatures depended on the primers used to (primer sequences available on request). PCR products were purified using the QIAquick PCR purification system (Qiagen) and quantitated by visualization on a gel. Sequencing reactions were performed at the Australian Genome Research Facility (Brisbane, Australia) and sequence analysis and assembly were performed using Editview and Autoassembler software, respectively (Applied is Biosystems).
SSCP
SSCP analysis was performed as previously described (Campbell et al, Hum. Mol.
Genetics 3, 589-594, 1994) on 8 exons of EDD encoding regions of potential 2o functional significance, i.e. the HECT domain, nuclear localization signal and zinc finger motif. Primers were designed within intronic sequence to generate PCR
products between 200-300 bp).
EDD immunohistochemistry 2s Immunohistochemistry (IHC) was performed on paraffin-embedded, formalin-fixed breast (9 normal breast and 46 breast cancers), general ovarian tissues (94 ovarian cancers) and 165 serous ovarian cancer tissues. Paraffin-embedded embryonic neural tissues from wild-type and EDD -(knockout) mice were used as positive and negative controls, respectively. .Paraffin-embedded cell pellets from 3o the WT-30 cell line, which overexpresses EDD (Henderson et al., 2002) were used as an additional positive control.

Sections were dewaxed and rehydrated before unmasking in target retrieval solution (high pH: DAKO Corporation, Carpenteria, CA) in a waterbath, at 100°C
for 30 min.
s Using a DAKO autostainer endogenous peroxidase activity was quenched in 3%
hydrogen peroxide in methanol, endogenous avidin and biotin were blocked with an avidin biotin block (DAKO Corporation) and non-specific binding of secondary antibody was blocked by incubation with a serum free protein block (DAKO
to Corporation). Sections were incubated for 30 min with 1:150 anti-EDD (M19) antibody (Santa Cruz Biotechnology, Santa Cruz, CA) and, subsequently for 15 min with 1:200 biotinylated horse anti-goat (Vector Laboratories, Burlingame, CA).
A streptavidin-biotin peroxidase detection system was used according to the manufacturer's instructions (Vectastain Elite kit; Vector Laboratories) with 3,3'-is diaminobenzidine as substrate. Counterstaining was performed with Mayer's hematoxylin (DAKO Corporation).
The degree of staining was assessed by two separate observers and discrepancies were resolved by conferencing. EDD expression scores were 2o determined by combining the percentage of cells expressing EDD (range of 1-for 1 %-100% cells stained) and intensity of staining (range of 0-3). A
combined score of 0 was called no expression, a score in the range of 1-5 was low expression and a score of 6 and 7 was called high expression.
2s The relative EDD expression and the proportion (percentage) of EDD positive cells in the serous ovarian cancer tissues were correlated with various clinicoathological parameters of ovarian disease (as shown in Table 2) in order to determine whether or not EDD expression is a prognostic marker of outcome of serous ovarian cancer.

1.2 Microsatellite analysis The firequency of AI surrounding the EDD locus (8q22.3) was investigated in malignant and pre-malignant ovarian tumors, breast cancers, hepatocellular carcinoma, metastatic melanoma and squamous cell carcinoma of the anterior s tongue using nine microsatellites shown with their locations in Figure 1. As CEDD
and 586F18b are located within introns of the EDD gene it can be assumed that chromosomal loss or gain involving these alleles is equivalent to loss or gain of an EDD allele.
io Within the complete cancer set (excluding non-malignant ovarian tumors) the frequency of AI in the narrow region of 8q under study was considerable: 60%
(83/139 informative cases) had AI at one or more markers. The individual frequencies (cases with AI/informative cases) were: D8S326 47/103 (46%); CEDD
38/90, (42%); D8S257 29/95 (31%);D8S300 27/69 (39%); D8S545 28/87 (32%);
is and D8S85 24/90 (27%) (Table 1 ). Notably, CEDD and the neighboring 8q22.3 marker D8S326 had the greatest frequency of AI. In addition, as can be seen from the following data, 30-40% of those cancers that have chromosomal aberrations at the CEDD microsatellite show no involvement of one or more microsatellites at the telomeric end of the region studied. This indicates that AI at the EDD
locus 20 often occurs independently of more extensive aberrations in these cancers (ie loss or gain of the whole chromosome arm or co-amplification with the MYC
oncogene).
1.3 Ovarian cancer ~s The ovarian tumor set was comprised of several cancer subtypes (predominantly mucinous, endometrioid and serous), borderline and non-malignant benign tumors. Within the cancer group 48% (34/71 ) displayed AI at one or more markers in the region (Figure 2). In marked contrast, benign and borderline tumors exhibited only 1/23 and 1/5 cases of AI, respectively, involving several 3o microsatellites but not CEDD or DS8257 (Table 1 ). Because of the low frequency of involvement of 8q22.3-8q23.3 in benign and borderline tumors they were omitted from the following analysis.
Several cancers had AI across all informative markers, consistent with large chromosomal aberrations eg cases 63, 114, 154. However, of interest are those s cancers where AI is present at the EDD locus but not at the more telomeric markers (eg cases 14, 22, 32, 211, Figure 2). This suggests that an important region of chromosomal aberration is located close to the EDD locus. Indeed a central finding of this study is the observation in serous cancers that CEDD
was the microsatellite most commonly affected by AI (16/22, 73% of informative io cases). This very high frequency of involvement differs significantly from that for the more telomeric markers D8S85 (6/18, 33%, p<0.01 ) or D8S845 (4/13, 31 %, p<0.01 ). In fact, when serous cancers are compared to other ovarian cancers, CEDD is the only microsatellite for which Al differs significantly between the two groups (p=0.0018). However, no correlation was apparent between AI at any is locus and cancer grade and stage either within the entire set of cancers or the serous subtype (data not shown).
Analysis of gene expression profiles of various ovarian cancers (both serous and epithelial) to identify those changes in gene expression that correlate with disease 20 outcome following laparotomy (ie the time at which the gene expression profile is determined), demonstrates that changes in EDD gene expression in subjects is significantly associated with the survival of a subject suffering from ovarian cancer (ie p=0.00). Accordingly, EDD is clearly a prognostic marker of recovery from ovarian cancer following a laparotomy.
1.4 Hepatocellular carcinoma, sauamous cell carcinoma of the tongue and metastatic melanoma In all three cancer types AI was common (Table 1 ) with 74%, 33% and 47% of cancers displaying AI of at least one microsatellite respectively.
In nineteen hepatocellular carcinomas four cancers had regions of imbalance that included CEDD but did not extend continuously telomeric to 8q22.3 (Figure 1A).
Similarly analysis of seventeen metastatic melanoma (Table 1 ) found three cancers where the region of AI did not involve markers telomeric to 8q22.3 (example shown in Figure 1 ), Of the twelve squamous cell tongue carcinoma s analyzed all had AI involving 8q22.3 (CEDD and/or D8S326) and in three of these cancers the region of imbalance did not extend to the most telomeric markers, D8S85 and D8S545 (example shown in Figure 1 ). Thus in these three cancer types the chromosomal region at he EDD locus is commonly and often specifically aberrant.
to 1.5 Breast cancer For microsatellite analysis of breast cancer DNA, three additional microsatellites were introduced to provide more information about AI at the EDD locus (586F18b) and at the telomeric region of 8q around MYC (8q24.12) (D8S198 and MYC-is PCR.3). As with the other cancer types studied, AI was common on 8q with (67%) breast cancers displaying AI at one or more markers (Table 1 ). CEDD or 586F18b were involved in 6/16 (38%) of informative cases. Commonly, AI
involved MYC-PCR.3 or D8S198, consistent with the frequent amplification of MYC in breast cancer, but in the majority of cancers AI was not continuous from 20 8q22.3 to 8q24 (examples shown in Figure 1 ).
1.6 EDD mRNA expression in breast cancers EDD mRNA expression levels were determined by quantitative RT-PCR in 41 breast cancers, and in matched normal breast tissue controls for 14 of these 2s cases (Figure 3A). Although the majority of cancers expressed EDD mRNA
within the normal range, a significant number 11/41 (27%) had higher expression.
Elevated expression was even more apparent when cancers were compared to their matched normal tissue controls, such that 6/14 (43%) cancers had > 4-fold increase in EDD expression, including one cancer with a 159-fold increase (Inset 3o Figure 3A). Samples were also analysed for expression of several control genes, which controlled for epithelial content (CK18), cellular metabolism (GAPDH), general transcriptional activity (IF2B) or proliferation rate (MCM3) of the tumors analysed. EDD expression was not significantly altered when normalised to the expression of these genes.
s 1.7 EDD protein expression in breast and ovarian cancers.
EDD protein levels were determined by immunohistochemistry in 9 specimens of normal breast tissue, 46 breast carcinomas and 94 serous ovarian carcinomas.
Positive controls of wild-type EDD embryonic mouse neural tissue (Figure 4B) and WT-30 cells (not shown) demonstrated intense nuclear staining while no io expression was seen in EDD null embryonic mouse neural tissue (Figure 4A).
Of 9 normal breast samples, 4 had no detectable expression and 5 had low level expression of EDD (Figure 4C). Of 46 breast carcinomas, all expressed EDD and demonstrated either low intensity (37%) or high intensity (63%) nuclear staining (Figure 4D). Among the 94 serous ovarian carcinomas, 2°t° failed to express EDD
is while 59% had low (Figure 4E) and 39% had high expression (Figure 4F).
Univariate analysis of clinicopathological parameters of 165 patients with serous ovarian cancer shows that disease stage and cancer grade, overall patient health (perFormance status) and residual disease following surgical debulking all predict 2o final patient outcome (ie whether or not a subject with serous ovarian cancer will survive) (Table 3). Analysis also showed that disease stage, presence of residual disease and EDD overexpression were predictive of disease relapse.
Accordingly, EDD expression are clearly prognostic/diagnostic of relapse of serous ovarian cancer.
Multivariate analysis of clinicopathological parameters of 165 patients with serous ovarian cancer shows that disease stage and residual disease following surgical debulking are predictive of a poor outcome (ie patient death or cancer relapse) (Table 4). Analysis also showed that disease stage, presence of residual disease 3o and EDD overexpression were predictive of disease relapse. Accordingly, EDD
expression is clearly an independent prognostic/diagnostic marker of relapse of serous ovarian cancer.
1.8 EDD, p53R2 and CA II mRNA expression and c~enomic copy number in breast cancer cell lines s Expression profiiling of 18 breast cancer cell lines with known DNA copy number showed a strong trend for EDD to be overexpressed at the mRNA level when EDD was amplified at the DNA level (Figure 3B). Almost all cell lines where EDD
was overexpressed had amplified EDD at the genomic level (7/8), whereas EDD
was rarely overexpressed when the gene copy number was two or less. This was io also true fior the expression of p53 ribonucleotide reductase (p53R2), the gene adjacent to EDD at chromosome position 8q23.1. All cell lines where p53R2 was overexpressed had increased gene copy number (Figure 3B). The relative expression of EDD and p53R2 was well correlated (R2 = 0.62) suggesting a similar mechanism of overexpression for both genes. In contrast, the expression is of carbonic anhydrase II (CA II), located at 8q22, showed no relationship with DNA copy number (data not shown), with 15/18 cell lines having less than 10%
of the expression levels of CA II mRNA of 184 normal breast epithelial cells.
1.9 Mutational analysis of EDD in cell lines and tumors 2o To assess the frequency of mutations in the EDD gene in cancer, the complete coding region of the EDD mRNA (8397 nucleotides) was sequenced using cDNA
derived from 26 breast, ovarian and prostate cancer cell lines. Three normal breast epithelial cell lines were also sequenced. A list of the sequence variants identified is shown in Table 5. Only 2/25 cancer cell lines had variations in EDD
2s mRNA that resulted in a change to the translated amino acid sequence. These putative missense mutations were confirmed in the genomic sequence. The amino acid changes were not in regions of the EDD protein having functional motifs and only the His>Asn change in the SK-Br-3 line alters amino acid polarity. In the absence of matched normal DNA from the individuals from which the cell lines 3o were derived it is not possible to determine whether these represent true somatic mutations or rare polymorphisms. Few polymorphisms or silent substitutions were observed. Of the six conservative sequence variants, at least five are likely to be polymorphisms as they are either found in RNA derived from normal cell lines or tissues, or in multiple cell lines. A splice variant was also observed in all cell lines.
The variant differs from the full length EDD mRNA by deletion of 18bp of s sequence from nt 884-901, removing the amino acid sequence VLLLPL. Although this motif is not located in any of the putative functional domains of EDD, removal of this sequence does have the potential to disrupt protein structure and might alter enzymatic function or localization of the protein.
io SSCP analysis of twenty nine ovarian cancers displaying AI (Figure 2), 37 breast cancers and 29 colon cancers confirmed the low frequency of mutation of the EDD gene, although only eight exons of EDD (covering approximately 13% of the coding sequence) were studied. These exons encode a nuclear localization signal (nt 1482-1598), a zinc finger motif (nt 3573-3812) and the majority of the HECT
is domain (nt 7602-8339) (Henderson et al , J. Biol. Chem. 277, 26468-26478, 2002). No mutations were found in the coding regions or splice junction sites of any cancer (data not shown).
1.10 DISCUSSION
2o Microsatellite allelotyping shows that a narrow region of chromosome 8q that encompasses the locus for EDD (8q22.3) is a significant and specific area of chromosomal abnormality in ovarian cancer, hepatocellular carcinoma, squamous cell carcinoma of the tongue, breast cancer and metastatic melanoma. This was particularly evident in serous carcinoma of the ovary, where AI of the EDD
locus 2s occurred in approximately 70% of cancers. Overexpression of EDD mRNA was observed in a significant proportion of breast cancers and breast cancer cell lines, suggesting the possibility that EDD may be a target for amplification in this region of 8q.
3o Evidence that the region of 8q containing EDD is a specific focus of chromosomal abnormality is particularly clear for serous carcinoma of the ovary in which the frequency of AI at the EDD gene-specific microsatellite CEDD (73%) was almost twice that of the most telomeric microsatellite examined. Ovarian carcinomas commonly show a high level of chromosomal rearrangement in comparison with other tumor types. This is subtype specific with serous tumors displaying the s greatest changes, endometrioid intermediate, and mucinous cancers the least (Pieretti et al, Int. J. Cancer 64, 434-440, 1995), as was the case for AI at 8q in our study. In at least one study the invasive potential of the serous subtype has been correlated with changes to chromosome 8 (Diebold et al, ab. Invest. 75, 485, 1996), although no correlation between clinical data (including grade and io stage) and AI at 8q was found in our study. Although the very low rates of AI at EDD in benign and borderline ovarian tumors might indicate that this region is perturbed only at late stages in tumor progression, it is debatable whether such tumors are actually precursor lesions for malignant ovarian tumors.
is Analysis of EDD expression clearly shows that EDD mRNA levels correlate with patient survival in all type of ovarian cancer studied. Furthermore, EDD
protein levels correlate significantly with disease relapse. Accordingly, EDD
expression levels (mRNA or protein) are clearly predictors of patient survival and disease relapse.
Despite the frequency of gross aberrations at the EDD locus and the common overexpression of the gene, EDD coding region mutations are apparently rare in cancer. Sequencing of the large EDD mRNA (8397 nt) revealed only 2/25 cancer cell lines with single nucleotide changes that result in amino acid changes.
These 2s alterations might represent rare polymorphisms rather than mutations as none of the changes were clearly disruptive and occurred in regions of the protein without any predicted functional significance. A splicing variant was found but this was present along with the unspliced transcript in every normal and cancer cell type examined. Similarly, SSCP analysis of a limited number of axons covering 13%
of so the coding region, and which code for putative functional domains, found no evidence of mutations in a series of breast, ovarian and colon cancers. This sequence conservation may point to a critical requirement for this gene in cell function, a conclusion reinforced by the hyd knockout lethality (Mansfield et al, Dev. Biol. 165, 507-526, 1994) and by our recent finding that targeted deletion of EDD in mice results in embryonic lethality (unpublished data).
s Nuclear Function of EDD HECT Ligase io 2.1 Experimental Procedures Plasmid constructs EDD cDNAs used for in vitro translation, transfection and yeast two-hybrid screening, are shown in Figure 5A. cDNAs encoded the full length protein EDD
(aa 1-2799), the N-terminal domain EDDF1 (aa 1-889), the central domain EDDF2 is (aa 889-1877), the carboxy domain EDDF3 (aa 1877-2799), the N-terminal plus central domains EDDF4 (aa 1-1877) and the central plus C-terminal domains EDDF5 (aa 889-2799). EDDM, EDDF3M and EDDFSM contain a mutation (Cys2768 to Ala) at the active site cysteine necessary for E3 ligase activity in HECT proteins. For mapping of the EDD N-terminus, restriction fragment cloning 2o was used to generate in vitro translation constructs expressing EDD as 1-(EDDF1 a), 578-889 (EDDF1 b), 1-419 (EDDF1 c), and 420-889 (EDDF1 d) (Figure 5A). For yeast two-hybrid screening, EDD cDNA fragments used as baits were cloned from pBluescript EDD (Callaghan et al, Oncogene 17, 3479-3491, 1998) in frame with the Gal4 DNA binding domain (DBD) of the pAS2.1 vector (Clontech 2s Laboratories, Palo Alto, CA, USA). For in vitro translation, EDD-derived cDNAs were transcribed from pBluescript (Amersham Pharmacia Biotech, UK), pSGS
(Stratagene, La Jolla, CA, USA) or pRcCMV (Invitrogen, Gronigen, Netherlands) vectors. For EDD protein expression in mammalian cells, constructs in pRcCMV
have been previously described (Callaghan et al, Oncogene 17, 3479-3491, 1998) 3o and additional constructs for expression of FLAG epitope tagged EDD were generated in the pSG5 vector. A GFP reporter vector (pGFP20, Dr S. Aota, -1~8-Osaka, Japan) was co-transfected to monitor transfection efficiency. A
bacterial plasmid expressing GST fused to amino acids 263-538 of human importin a5 (NPI-1 ) was obtained from Peter Palese (Mount Sinai School of Medicine, NY, USA). A GST fusion of mouse importin X31 (PTAC97), was expressed and purified s as described previously (Hubner et al, 1997). Full-length CIB was cloned from the pACT2 vector into the pGEX2T vector for GST-CIB fusion protein expression in bacteria and into the pCMVTag2C vector for mammalian expression of FLAG-tagged protein. For expression of GFP-tagged EDD in mammalian cells, full length EDD was cloned into pEGFP-C1 (for N-terminal EGFP tag) and pEGFP-N1 (for C-io terminal EGFP tag, Clontech Laboratories). For transient expression of progesterone receptor, phPR1 vector encoding human PR B was obtained from P.
Chambon (INSERM, France). A PRE-luciferase reporter vector (pMSGluc) was constructed by insertion of a MMTV-LTR promoter in the pGL3-Basic vector (Promega Corp.). The phPR1 vector was used to clone the PR(AB) (aa 1-546) is and PR(CDE) (aa 456-933) regions into pGEX4T2 for GST fusion protein expression. For transient expression of vitamin D receptor pCMV-VDR along with pOS2-luc reporter vector were obtained from G. Leong (Garvan Institute, Australia). Estrogen receptor was expressed from pCMV-ER (V.C. Jordan, Northwestern University Medical School, Chicago, USA) and pERE-TK-GL3 2o reporter vector was obtained from M. Parker (ICRF, UK). For in vitro translation, SRC-1 was cut from pCR3.1-SRC1A (B. O'Malley, Baylor College, Texas) and cloned into pBluescript.
Yeast two-hy,6rid assay for EDD interacting proteins 2s The cDNAs for full length EDD mutant and carboxy domain mutants (EDDM and EDDSM) were screened against a human placenta cDNA library in the pACT2 vector by the yeast two-hybrid method (Matchmaker 2, Clontech, Palo Alto, CA, USA). Stable transformants of Saccfiaromyces cerevisiae strain Y190 expressing EDD fusion protein were transformed with the library using the lithium acetate 3o method and 2-3 x 106 primary transformants selected on his-leu-trp- plates.
Following a second round of selection on the same medium, colonies were assayed for (3.galactosidase activity using a filter-based assay. Interacting plasmids that were positive for ~i.galactosidase only in the presence of the EDD
bait plasmids were transformed into E. coli DHSa cells for further analysis.
Manual sequencing was carried out using 33P end-labelled primer in conjunction with the s fmol Cycle sequencing kit (Promega Corp., Madison, WI, USA). Sequences were analysed by Blast searches of the Genbank and EMBL databases and predicted proteins analysed for motifs using the ISREC Profile Scan Server (www.isrec.isb-sib.ch).
to For semi-quantitation of protein interactions, CG1945 yeast cells containing pAS2.1-EDD constructs were mated with Y187 yeast cells harbouring pACT2-derived plasmids. Diploids were selected on leu-trp- plates and used to inoculate cultures which were grown to saturation, diluted 1:10 and grown for 16h. Yeast cells were harvested for protein and (3.galactosidase activities were determined in is a liquid chemiluminescence assay (Tropix Galacto-Light System, Applied Biosystems, CA, USA).
Recombinant protein binding assays GST-tagged fusion proteins were prepared from E, coli strain BL21 according to 2o established protocols (Pharmacia Protocol Handbook). Soluble fusion proteins were bound to glutathione agarose and quantitated via Coomassie blue staining against protein standards. 35S-labelled EDD protein and mutants or SRC-1 were synthesised in a coupled in vitro transcription/translation system (TNT Quick, Promega) and 10-20p1 reaction mixture was diluted in 1 % Triton X-100 lysis buffer 2s (Callaghan et al, Oncogene 77, 3479-3491, 1998) and incubated with 5pg of GST, GST-importin a5, GST-PR(CDE) or GST-CIB coupled to glutathione agarose beads at 4°C for 2h. Beads were collected by centrifugation, washed extensively in lysis buffer and resuspended in SDS-PAGE sample buffer. After boiling, bound protein was visualised following SDS-PAGE and autoradiography.
Cell culture and transient transfection HEK 293 and T-47D were maintained as previously described (Callaghan et al, Oncogene 17, 3479-3491, 1998). MCF-7 cells v~iere maintained in RPMI medium (Life Biotechnologies) containing 10 % serum in 5% C02. For overexpression by transient transfection, 3 x 106 HEK 293 cells were plated in HMEM containing 10%
s serum in 15cm petri dishes. The following day pRcCMV-EDD (10pg) was added to the cells along with 30p1 Fugene reagent (Roche Diagnostics, Castle Hill, NSW, Australia).
Localisation of GFP-tagged and endogenous EDD protein to HEK 293, CHO, MCF-7 or T-47D cells were seeded in 6-well plates at 1-2 x cellslwell. Cells were transfected with 2pg pEGFP-EDD or empty vector DNA and the following day split to chamber slides for 24-48h. Slides were washed in PBS, fixed in 3.7% paraformaldehyde, washed in PBS and mounted in 90% glycerol.
GFP was visualised by fluorescence microscopy. For immunostaining, HEK 293 is cells or EDD transfected HEK 293 cells (WT30) were embedded in paraffin.
Sections were de-waxed and rehydrated before unmasking in EDTA/Citrate buffer and then stained with goat anti-EDD antibody N19 (Santa Cruz, CA, USA). EDD
signal was detected using DAKO LSAB Plus Link and Label (DAKO Corporation, CA, USA) with liquid 3,3'-diaminobenzidine Plus (DAKO Corporation, CA, USA) as 2o substrate. Counterstaining was performed with hematoxylin.
Protein interactions in cell lysates For extracts of total cellular protein, cells were harvested in 1 % Triton X-100 lysis buffer as previously described (Callaghan et al, Oncogene 17, 3479-3491, 1998).
2s Extraction of nuclear proteins and cytoplasmic s100 fractions from T-47D
and MCF-7 cells was carried out according to published methods (Dignam et al, Nucleic Acids Res. 11, 1475-1489, 1983).
For GST fusion protein pull-down of endogenous or recombinant EDD from cell 30 lysates, 0.5 to 1 mg total protein was incubated with 5pg GST or GST fusion protein bound to glutathione beads for 1-2h at 4°C. Beads were washed extensively in 1 % Triton X-100 lysis buffer and bound proteins resolved by SDS-PAGE and detected by immunoblotting with EDD antisera (Callaghan et al, Oncogene 17, 3479-3491, 1998). For immunoprecipitation, 10p1 importin a5 antisera (Peter Palese, Mount Sinai School of Medicine) was incubated with 0.5 to s 1 mg cell lysate (4°C for 1 h). Antibody conjugates were captured on protein A
sepharose beads (4°C for 1 h) and washed extensively in lysis buffer.
Bound proteins were resolved by SDS-PAGE followed by immunoblotting with EDD
antisera.
io Stable HEK 293 cells overexpressing EDDM were transfected with pCMVTag2C-CIB or empty vector using Fugene 6 reagent (Ruche) for 24h. Following 6h incubation in the presence of MG132, cells were harvested and lysates prepared and one mg total protein incubated with anti-FLAG antibody M2 coupled to Sepharose (Sigma Chemical Co., St Louis, USA) for 2h at 4°C. Beads were is recovered by centrifugation and washed extensively in lysis buffer. Western blotting for EDD has been described (Callaghan et al, Oncogene 77, 3479-3491, 1998).
Nuclear receptor transactivation assays 2o HEK 293 or COS7 cells were plated in 6-well plates (2 x 105 cells/well) and the medium changed to 2% charcoal-stripped FCS the following day. Transfection was carried out using 3-4pl Fugene 6 Reagent (Ruche) with 1-2pg DNA
comprised of 90ng receptor expression vector, 450ng luciferase reporter vector and 1.2 pg EDD, EDDM or SRC-1 cDNAs in either pRcCMV or pSGS, or empty 2s vector, and 200ng GFP expression vector pGFP20. The following day cells were split into 96-well plates (7 x 103 cells/well) or 6-well plates (1.4 x 105 cells/well), and drugged 24h later. After a further 24h cells in 96-well plates were assayed for luciferase activity (Luclite reagent, Packard Bioscience, Meriden, CT, USA) and cell number (Wst-1 reagent, Ruche). In some experiments cell number was 3o monitored using the CeIITiter96~ Proliferation Assay (Promega Corp., Madison, USA). Cells in 6-weft plates were analysed for GFP expression by fluorescence microscopy to determine transfection efficiency and also used for preparation of protein lysates so that protein expression levels of various constructs could be compared. In experiments where there was significant variation in cell number or GFP expression these parameters were used to normalise luciferase activity. In s some experiments pRSV-(3-gal or pRL-TK (Promega Corp., Madison, USA) vectors were transfected in place of pGFP20 and transfection efficiency monitored by assaying for ~3-galactosidase or Renilla luciferase activity respectively.
Proteasome inhibition experiments to For proteasome inhibition experiments, HEK 293 cells were plated at 3 x 105 cells/well of a 6-well dish in miminal essential medium with Hanks' salts (HMEM) supplemented with 10 % fetal bovine serum (Life Technologies, Gaithersburg, MD, USA). After 48h, medium was replaced with medium containing 20pM
MG132 (Calbiochem, CA, USA) or DMSO vehicle for 2-6h. A monoclonal antibody is for western blotting against CIB was kindly provided by U.P. Naik (University of North Carolina, NC, USA).
Treatment of cells with DNA damaging agents MCF-7 cells were incubated in RPMI containing 0.5% foetal bovine serum for 18h ao before addition of phleomycin (Cayla, France) at 100pg/ml, hydroxyurea (Calbiochem, CA, USA) at 2mM or PBS vehicle, for 6h. Cells were harvested for total protein or nuclear protein extracts as described above.
2.2 Domain structure of EDD
2s Previous analysis of the EDD sequence showed the presence of a carboxy-terminal HECT domain, identifying EDD as a member of the HECT family of ubiquitin protein ligases (Callaghan et al, Oncogene 17, 3479-3491, 1998; and Huibregtse et al, 1995) (Figure 5A). Further examination of the central domain of EDD that is also highly conserved with HYD revealed a stretch of 68 amino acids 30 (aa 1177-1245) that is 95 % and 100 % conserved with HYD and mouse EDD, respectively (Figure 5B) and which shows a high degree of alignment with calossin (pushover), a calmodulin-binding protein important for neurotransmission and male fertility in Drosophila (Richards et al, Genetics 742, 1215-1223, 1996;
Xu et al, J. Biol. Chem. 273, 31297-31307, 1998). Contained within this region is a cysteineihistidine-rich putative zinc finger domain, zf UBR1 (Pfam PF02207, s (Bateman ef al, Nucleic Acids Res 28, 263-266, 2000)), originally identified in the N-end rule E3 ubiquitin ligase UBR1 p/N-recognin from a range of species (Bartel et al, EMBO J. 9, 3179-3189, 1990; Varshavsky, Cell 69, 725-735, 1992; Kwon et al, Proc. Natl. Acad. Sci. USA 95, 7898-7903, 1998). The consensus sequence CX~~_~6CX2CX8_~pCXzCX~5HX2HX~1-~4CXCX4_14C is reminiscent of the more common to RING domain, which is also found in a distinct region of UBR1 p. Both types of zinc-binding domains are proposed to have roles in protein-protein interaction, with the RING domain having an established role in ubiquitinylation (Freemont, Curr. Biol. ~0, 84-87, 2000; Lorick et al, Proc. Natl. Acad. Sci. USA 96, 11369, 1999). This central region of EDD also contains a potential bipartite is nuclear localisation sequence (NLS), (KLKRTSPTAYCDCWEKCKCK, as 1222-1241; SEQ ID NO: 43) while another putative NLS resides in the N-terminal region (RKKMLEKARAKNKKPK, as 502-517; SEQ ID NO: 44) upstream of a potential SV40 large T antigen-like NLS (PYKRRR, as 630-635; SEQ ID NO: 45) (Dingwall et al, Trends Biochem Sci 76, 478-781, 1991 ). Also within the N-terminal region 20 of EDD is another region conserved with HYD, designated as a "UBA domain"
(aa 188-225), which may form a protein-protein interaction interface (Hofmann et al, TIBS 27, 172-173, 1996). Amino terminal to the HECT domain (aa 2391-2455) Lies still another region that may mediate protein interactions. This 60 amino acid stretch shows 50% homology to a region within the carboxy terminus of polyA
2s binding proteins (PABP-C) from a range of species (Callaghan et al, Oncogene 7 7, 3479-3491, 1998; Kozlov et al, Proc. Natl. Acad. Sci. USA 98, 4409-4413, 2001 ). The X-ray structure of this domain in both PABP and EDD has recently been determined and forms a protein interaction interface consisting of four alpha helices (Kozlov et al, Proc. Natl. Acad. Sci. USA 98, 4409-4413, 2001; Deo et al, 3o Proc. Natl. Acad. Sci. USA 98, 4414-4419, 2001 ).

2.3 Interaction between EDD and importin a-5 Yeast two-hybrid approaches were used to identify interacting proteins that may be ubiquitinylation targets of EDD or other associating proteins with a role in EDD
function. First, yeast two-hybrid screening of a human placental cDNA
expression s library was performed against baits encoding full length EDD or fragments containing one or more potential interaction domains of EDD (see Figure 5A).
Screening with full length EDDM (C2768A mutant) identified two independent cDNAs encoding the nuclear import protein importin a5 (NPI-1, (O'Neill et al, Virology 206, 116-125, 1995; Kohler et al, Mol. Cell Biol. 19, 7782-7791, 1999)), to one full-length and the other encoding amino acid 229 to the carboxy terminus (amino acid 538). Importin a has a specific role in nuclear import, by recognising NLSs, implying both that one or more of the potential NLSs within EDD are indeed functional, and that EDD may have a role in the nucleus, with importin a involved in transporting EDD from the cytoplasm to the nucleus.
2.4 Importin a-5 interacts with a region of EDD containing- nuclear localisation signals results A strong interaction was found between NPI-liimportin a5 and full length EDD, with full length importin a5 interacting more strongly than the amino-truncated 2o protein isolated by two hybrid screening (Figure 6A). This difference might be explained if EDD, like other proteins that contain basic NLSs, is recognised by the armadillo repeats of importin a: only four of seven such repeats are present in the truncated importin a5 clone. Pull-down experiments showed that a GST importin a5 fusion protein encoding amino acids 263-538 was able to bind to in vitro 2s translated EDD and mutants encoding the N-terminal one-third of EDD but not to the central or carboxy terminal regions of EDD (Figure 6B, 6C). This suggested that the putative NLS in the central region of EDD was not functional in nuclear import. GST-importin a5 also interacted with endogenous EDD (T-47D cells) and recombinant EDD expressed in HEK 293 cells (Figure 6D), precipitating a 3o considerable proportion of the available EDD protein. Further, anti-importin a5 antisera also immunoprecipitated EDD protein from these lysates showing that EDD and importin a5 interact in vivo {Figure. 6D).
The amino terminal one-third of the EDD protein contains two potential basic NLSs, one bipartite and one simple. To determine the relative contributions of s these motifs to importin a binding, a set of constructs for in vifro translation were made that contained one, both or neither NLS. GST-importin a interacted with each NLS to some degree and no interaction was seen in the absence of both signals (Figure 6E). We therefore conclude that both N-terminal signals are required for full binding potential.
to We expected that EDD might be in a nuclear import complex with importins a and ~i so binding to the nuclear import partner importin ~i/p97 was also tested (Figure 6F). GST-importin ~i bound to in vitro translated EDD and the amino two-thirds or one-third of the EDD protein, resulting in pull-down of approximately 5% of the is available EDD protein. As the extracts used for in vitro translation contain endogenous importin a, binding of EDD and importin (3 is most likely mediated by the importin a/(3 heterodimer. Yeast two-hybrid analysis also indicated EDD
interaction with both importin a1 (Rch1) and irriportin a3 (Qip1), although the interaction between EDD and importin a5 was markedly stronger (data not 2o shown). Overall, the interaction between EDD and several importin a isoforms and importin (3, point to a role for EDD within the nucleus.
2.5 EDD is a nuclear protein To determine the cellular localisation of EDD, mammalian expression vectors 2s were made for EDD fused to the N or C terminus of green fluorescent protein (GFP). Transfection of HEK 293 cells or MCF-7 breast cancer cells with the N-terminal EDD-GFP fusion showed that fluorescence was restricted to the nucleus (Figure 7A). Identical results were obtained with the C-terminal fusion (not shown).
In contrast, when cells from either line were transfected with pEGFP vector only, a 3o diffuse pattern of staining throughout the whole cell was observed. Nuclear localisation was confirmed when EDD-specific antibodies were used to stain sections of HEK 293 cells which endogenously express EDD or WT30 derivative of HEK 293 cells, which overexpress EDD (Figure 7B). The same pattern of staining was seen for a second EDD-specific antibody (not shown).
s 2.6 EDD interacts with progesterone receptor B
Previous studies have demonstrated that the HECT-domain protein E6-AP
interacts directly with PR-B through a region containing, LXXLL motifs (Nawaz et al, Mol. Cell Biol. 79, 1182-1189, 1999). These motifs which are present in other transcriptional coactivators are potentially involved in nuclear receptor interaction to and coactivation (Heery et al, Nature 387, 733-736, 1997). Since the EDD
protein is nuclear and contains five LXXLL domains (at amino acids 248, 1102, 1255, 1398 and 2428) we tested the ability of EDD to interact with PR-B and regulate its function. First we performed GST-PR fusion protein pull-downs of EDD or in vitro synthesised EDD fragments. The N-terminal AB region of PR contains a ligand-ls independent activation function 1 while the C-terminal CDE region of PR
contains the hinge and DNA binding domains and a ligand-dependent activation function 2.
The CDE region of PR, PR(CDE), interacted with endogenously expressed EDD
from T-47D cells (Figure 8A). This interaction was mapped using in vitro translated EDD protein fragments. A strong interaction was detected between the amino 2o terminal region of EDD (EDDF1, as 1-889) and the CDE region of PR, being greater than that seen for SRC-1 (Figure 8B). In these in vitro assays, interactions between PR(CDE) and either SRC-1 or EDDF1 were not affected by the PR
ligand ORG2058 (data not shown). No significant binding was observed between PR and other fragments of EDD (Figure 8B and data not shown).
The N-terminal region of EDD contains one of the five LXXLL motifs so the interaction was mapped further to assess the involvement of this motif. EDD
fragments EDDF1 a-EDDF1 d were tested for their ability to bind GST-PR(CDE).
Although EDDF1a (aa 1-577) and EDDF1c (aa 1-419) contained the LXXLL motif, 3o the strongest binding occurred between EDDF1 b (aa 578-889) or EDDF1 d (aa 420-889) and PR(CDE) (Figure 8C), suggesting that binding is mediated by the region of EDD consisting of amino acids 420-889, which includes both NLSs but not the LXXLL motif. This also ruled out the involvement of the UBA domain in this interaction. Taken together, these data demonstrate an interaction between EDD
and PR.
2.7 EDD acts as a transcriptional coactivator for nuclear receptors The nuclear localisation of EDD and the observed interaction between EDD and PR-B, together with evidence from separate studies that other HECT-domain proteins such as yeast RspS, its human homolog hRPF1 (Imhof et al, Mol. Cell.
io Biol. 16, 2594-2605, 1996), and E6-AP (Hubner et al, J. Biol. Chem. 272, 17195, 1997) have coactivator activity for nuclear receptors, prompted an investigation of whether EDD could enhance transcriptional activation by PR-B.
To this end, HEK 293 and COS7 cells, which lack endogenous PR, were transfected with a PR expression vector (pSGS/hPRB-1 ) and the progestin-responsive MMTV-ls luciferase reporter construct together with expression vectors for EDD, or as a positive control. EDD consistently increased progestin (ORG2058)-induced luciferase activity three- to five-fold above control levels in both lines, an effect comparable to that of SRC-1 (Figure 9A). 1n the absence of added ORG2058, EDD and SRC-1 also slightly increased the basal activity of the luciferase MMTV-2o LTR promoter, an effect more apparent in the COS7 cell line. We next tested whether the observed transcriptional effect of EDD was due to the ubiquitin ligase activity of EDD. When the ligase-defective EDD mutant (EDDM) was transfected, a comparable coactivator activity was observed, suggesting that the coactivator activity of EDD is independent of its ubiquitin ligase activity (Figure 9B).
zs Importantly no effect of EDD on PR transactivation is seen in the presence of the progestin antagonist RU486 (not shown), indicating specificity for ligand-bound receptor. Also, there was no effect of EDD on reporter gene activity in the absence of PR, indicating a specific effect on transactivation by PR (not shown).
3o Transfection of increasing amounts of pRcCMV-EDD showed a clear dose response for effects on progestin-induced luciferase activity (Figure 9C), and - 11~ -ORG2058 at all concentrations between lOpM and 100nM stimulated luciferase activity to a much greater extent when EDD was co-expressed (Figure 9D). EDD
co-expression resulted in a greatly enhanced response to low concentrations of progestins such that without EDD transfection, 10nM ORG2058 gave a maximal s response, whereas this was exceeded at a 100-fold lower concentration, 100pM, with EDD overexpression.
These data reveal for the first time a cellular function for EDD as a nuclear receptor coactivator. Interestingly, EDD also enhanced transactivation by the io vitamin D receptor (VDR) three-fold (Figure 9E). However estrogen receptor (ER) activity was not enhanced by EDD, whereas in the same experiment SRC-1 acted as a coactivator (Figure 9F), demonstrating that EDD discriminates between steroid receptors. Together these data demonstrate that EDD serves as a coactivator in PR- and VDR-mediated transcription.
is 2.8 EDD interacts with CIB, a protein potentially involved in DNA damage re~onses Further yeast two-hybrid screening was aimed at identifying other proteins involved in the ubiquitinylation or coactivation functions of EDD. When full length 2o EDDM or EDDFSM (aa 889-2799) were used as baits, three clones encoding calcium- and integrin-binding protein/DNA dependent protein kinase interacting protein (CIBIKIP) were isolated: two full-length and another encoding CIB/KIP
as 5-191. ClB is a protein with possible dual roles in the cytoplasm and nucleus (Wu et al, Mutation Researeh 385, 13-20, 1997; Naik et al, J. Biol. Chem. 272, 2s 4654, 1997; Kauselmann et al, EMBO J 78, 5528-5539, 1999; Shock et al, Biochem. J. 342, 729-735, 1999). The interaction between CIB and full length EDD initially detected in the yeast two-hybrid system (Figure 10A) was confirmed by pull-down of in vitro translated EDD proteins with GST-CIB (Figure 10B).
Mapping of this interaction using in vitro translated EDD fragments showed that 3o CIB interacts with the carboxy terminal portion of the EDD protein (EDDF3, EDDF3M, Figure 10C). To obtain evidence for interaction in cells, FLAG-tagged CIB was expressed in HEK 293 cells overexpressing EDD and protein extracts were prepared. EDD protein was detected in FLAG immunoprecipitates from these lysates but not from those of vector transfected cells (Figure 10D, left panel). GST-CIB fusion protein also interacted with EDD in cell lysates prepared s from nuclei of MCF-7 cells expressing endogenous levels of EDD (Figure 10D, right panel, 'Control').
Because we observed EDD in nuclei, a possible nuclear role for CIB was investigated. As CIB was previously found to interact with the DNA damage-io sensing enzyme DNA PK (Wu et al, Mutation Research 385, 13-20, 1997), lysates from MCF-7 cells treated with the radiomimetic phleomycin, which induces double strand breaks in DNA, were incubated with GST-CIB fusion protein.
Capture of the bound protein revealed significantly less association between EDD
and CIB when cells had been treated with phleomycin, when compared to is untreated cells or cells treated with hydroxyurea, which causes DNA
crosslinking (Figure 10D, right panel). The change in binding was not due to decreases in EDD
protein levels which were unchanged (not shown).
These studies show that EDD interacts with a potential ubiquitinylation substrate, 2o CIB, and that this interaction is sensitive to DNA damage. This is the first indication of protein interactions involving eith'er~EDD or CIB being responsive to DNA damage.
2.9 The Chk2 N-terminus interacts with EDD in HeLa nuclear extracts ~s When GSTchk2-N (aa 1-225) (Figure 11A) was incubated with HeLa nuclear extract, one of the interacting proteins was found by MS sequencing to be EDD
(Steve Jackson and Brandi Williams, Wellcome/CRC, Cambridge, UK). We confirmed this by GSTchk2-N pull-downs from MCF-7 cells followed by western blotting to detect EDD protein (Figure 11 B). Immunoprecipitation of chk2 from 3o whole cell extracts prepared from either HEK 293 cells or MCF-7 cells, both of which contain endogenous levels of chk2 and EDD, followed by immunoblotting for EDD confirmed an in vivo association between EDD and chk2 (Figure 11 C).
2.10 EDD interacts with the FHA domain of Chk2 When various fragments of 35S-labelled in vitro translated EDD were tested for s binding to GSTchk2-N, the carboxy-terminal two-thirds of EDD (EDDFS, as 889 2799) was found to interact. To determine the requirement for the chk2 FHA
domain in this interaction, the binding between EDD and GSTchk2-N and two mutants was assessed. The R117A mutant contains a substitution within the FHA
domain and involves a residue directly required for phosphothreonine binding.
to This mutant did not bind EDD (Figure 12A). The 1157T Li-Fraumeni-associated mutant retains the ability to bind phosphothreonine but has been shown to be unable to bind substrates of chk2 such as p53 (Falck et al, Oncogene 20, 5506-5510, 2001 ) cdc25A (Falck et al, Nature 470, 842-847, 2001 ), BRCA1 and cdc25C (Li et al, Mol Cell 9, 1045-1054, 2002). Interestingly, the 1157T
is substitution had no effect on binding to EDD (Figure 12A), suggesting that EDD
may instead bind through a phosphothreonine residue. These data were supported by further GSTchk2-N pull-down experiments using MCF-7 nuclear extracts (Figure 12B).
2o The requirement of an intact phosphopeptide-binding motif within the FHA
domain for EDD binding to the chk2 N-terminal region suggested that the interaction is mediated by a phosphorylated threonine residue within the EDD protein.
Although the residue involved has not yet been identified, EDD is phosphorylated in cells as when Flag-tagged EDD was overexpressed in HEK-293 cells in the presence of 2s 32P-labelled orthophosphate and cell lysates prepared, the resulting Flag immunoprecipitates contained a labelled species corresponding to EDD (Figure 11 C). Further, when HEK-293 or MCF-7 extracts were incubated in the presence of lambda protein phosphatase to remove phosphates, the electrophoretic mobility of EDD was increased, indicating the presence of phosphorylated amino acid 3o residues within the EDD protein.

A phosphopeptide (RWFDT(P)YLIRR) (SEQ ID NO: 46) that binds strongly to the FHA domain was used to further investigate the affinity of the chk2 FHA domain for EDD. Pre-incubation of the GSTchk2-N fusion protein with this peptide at concentrations greater than 50 pM abolished binding between EDD and chk2-N
s (Figure 12D).
2.11 DNA damage causes disruption of the EDD-chk2 interaction The role of the EDD-chk2 interaction in the DNA damage response was investigated. MCF-7 cells were treated with the radiomimetic drug phleomycin, io which causes double strand breaks in DNA and activation of chk2 via ATM
kinase (Matsuoka et al, Science 282, 1893-1897, 1998). A phosphorylation-induced mobility shift of chk2 in response to DNA damage was observed (Figure 14A). In contrast, no change in either the mobility or levels of EDD protein in MCF-7 nuclear extracts was detectable. However when chk2 was immunoprecipitated is from these extracts the amount of EDD associating was considerably reduced following DNA damage and these results were in agreement with GSTchk2-N pull downs from these same lysates (Figure 14B).
2.12 Depletion of EDD impairs CHK2 activation 2o To examine the role of the EDD-CHK2 interaction in the DNA damage response, cells deficient in EDD were observed for CHK2 activation on DNA damage. RNA
interFerence was used to deplete cells of EDD (as described herein) which were then irradiated and allowed to recover for up to 1.5 h. In cells lacking EDD, DNA
damage-induced phosphorylation of CHK2 on T68 was considerably diminished 2s compared to control cells (Figure 15A). Next, CHK2 kinase activity towards a GST-cdc25C substrate was assayed at various times post-IR in the presence and absence of EDD. These assays showed that activation of CHK2 was delayed in cells lacking EDD and these cells were impaired in their ability to reach maximal CHK2 kinase activity compared to control cells (Figure 15B).

2.13 EDD and BRCA2 interactions with chk2 may have distinct roles in the DNA
damage response EDD also interacts with BRCA2. EDD was identified along with BRCA2 in a 2 MDa protein complex isolated from nuclear extracts of HeLa cells (R. Shiekhattar, s Wistar Institute, U. Penn, personal communication). We confirmed this association was confirmed by co-immunoprecipitation of EDD and BRCA2 from HEK-293 cell lysates (Figure 17A). Therefore we were interested to test the possibility of an interaction between chk2 and BRCA2. Indeed when chk2 was immunoprecipitated from MCF-7 nuclear extracts, both EDD and BRCA2 were detected (Figure 17B).
io While the interaction between EDD and chk2 was diminished on phleomycin treatment, there was no effect on the amount of BRCA2 associated with chk2. An opposite effect was seen when MCF-7 cells were exposed to the replication block-inducing agent hydroxyurea (HU). Replication block-induced activation of chk2 had no effect on the association with EDD, while the levels of associating is were considerably reduced (Figure 17B). It is possible that these three proteins exist in a common complex. If so, it appears that on DNA damage that results in double strand breaks, EDD tends to dissociate from the complex whereas when the replication block pathway is activated, BRCA2 dissociates. Therefore if EDD
and BRCA2 are together in complex with chk2, this complex appears to be 2o differentially affected in response to different types of DNA damage. A
complex between BRCA2 and chk2 has not previously been reported and is of great significance in the DNA damage response.
2.14 Mapping the interaction between EDD and chk2 2s The EDD sequence contains a number of threonine residues in a favourable context for FHA binding when phosphorylated (Durocher & Jackson, FEBS Lett 513, 58-66, 2002). To pinpoint regions of EDD containing actual binding sites, binding of GSTchk2-N to various 35S-labelled in vitro translated EDD deletion mutants (see Figure 13A) was examined. In initial experiments, full binding of EDD
3o to chk2 required the carboxyl two-thirds of EDD (EDDFS) - Neither EDDF2 (central third) nor EDDF3 (carboxy third) were sufficient for binding (Figure 13A).

Removal of either the Hect domain or the RING-like domain from EDDF5 (EDD6 AND EDD7 respectively) did not significantly affect binding (Figure 13B), suggesting that the region of EDD that binds to the chk2 FHA domain lies between amino acids 1406 and 2526. .
2.15 Mapping the interaction between EDD and BRCA2 To pinpoint regions of EDD containing actual binding sites for BRCA2, binding of in vitro translated BRCA2 to various 35S-labelled in vitro translated EDD
deletion mutants (see Figure 13A) is examined.
io Additionally deletion mutants of BRCA2 are generated and labelled with 35S.
The ability of the full length EDD protein is tested to determine the region of BRCA2 to which EDD binds. Such information facilitates the analysis of the BRCA2-EDD
complex and the Chk2-EDD complexes in order to determine whether these is interactions form a single complex (ie BRCA2-EDD-Chk2), or instead whether these interactions form distinct complexes.
fn order to further characterise the interactions between EDD and BRCA2, these proteins are both expressed in HCT-15 cells, that lack Chk2 expression.
2o Following expression of both proteins in HCT-15 cells, antibodies to EDD
are used in immunoprecipitation experiments. The captured protein complexes are separated using SDS-PAGE and BRCA2 levels determined using an anti-BRCA-2 antibody.
2s HCT-15 cells expressing EDD and BRCA2 are then transfected with an expression vector that expresses Chk2 and the coimmunoprecitptation experiments repeated, in order to determine the effect of Chk2 on the association of EDD and BRCA2.
30 2.16 Functional conseguences of EDD-BRCA2-CHK2 interaction Ubipuitinylation assays are be used to investigate whether EDD is activated in response to DNA damage and whether EDD targets BRCA2 for ubiquitinylation and subsequent degradation. The assay utilizes in vitro translated or immunoprecipitated EDD
with reactions performed in the presence of GST- or His-tagged ubiquitin, E1 and the E2 UbcHSb with TopBP1 as positive control substrate. EDD activity is determined in the s presence and absence of ionizing radiation. This assay is also used to test other candidate substrates for ubiquitinylation by EDD such as CIB, or the GIi1, -2 and -3 proteins.
Depletion of EDD from MCF-7 breast cancer cells using established RNAi protocols io (described herein) is used to determine the degree to which BRCA2 is targeted by EDD
for ubiquitin-mediated degradation. The effects of DNA damage and proteasome inhibitors on BRCA2 protein levels are monitored using western blotting.
In order to determine the role of EDD in BRCA2-mediated repair of double strand DNA
is lesions the frequency of radiation-induced BRCA2-RAD51 nuclear foci formation in MCF-7 or HEK-293 cells with either normal EDD levels or those transfected with EDD
short interFering RNAs (siRNAs) is determined. Complex formation between BRCA2 and RAD51 is also monitored by co-immunoprecipitation experiments.
20 2.17 The EDD-BRCA2-CHK2 interaction in the context of DNA damage.
To examine the interaction in the context of DNA damage signaling via the ATM-mediated pathway, cells are treated with ionising radiation at doses of between 4 and 12 Gy, and the cells allowed to recover for periods of 0-6h. Activation of CHIC2 and other downstream responses to DNA damage are monitored by immunoblotting to detect the levels of CHK2 2s phosphorylated at T68 and also for p53 accumulation where appropriate.
Furthermore, hydroxyurea is used to study the ATR mediated response pathway of stimulation and its effects on the EDD-BRCA2-CHK2 interactions.
30 2.18 Significance of EDD-BRCA2-chk2 interaction - a workinct model EDD dissociates from chk2 on DNA damage. Therefore EDD may normally regulate chk2 and needs to be removed for chk2 activation. Overexpression of EDD, as observed in breast cancers for example, might then inhibit the activation of chk2 in response to DNA damage, thus leading to either replication of defective DNA or continued proliferation of cells with an abnormal chromosomal s complement. These conditions could foster further mutation and lead to cancer or tumour progression. Conversely, in EDD knockout mouse embryos or cells subjected to EDD siRNAs, we propose that chk2, in the absence of the inhibitory effects of EDD, might be dysregulated and thus cause prolonged inappropriate cell cycle arrest.
io It is therefore important to understand the precise pathways impinging upon the chk2-EDD-BRCA2 interactions. In any case, the interaction of EDD with these two tumour suppressor proteins underscores a potential role for EDD in cancer development and progression. Specifically, the well established role of CHK2 and is BRCA2 in DNA damage repair and cell cycle arrest strongly suggest that EDD
may play a role in such pathways.
2.19 Discussion This study demonstrates a new functional role for the nuclear protein EDD. A
2o progestin regulated gene, EDD itself has the ability to potentiate PR
transcriptional activity. In addition, EDD may play a role in DNA damage signalling as suggested by complex formation with C1B, a DNA PK-binding protein, an interaction that is sensitive to DNA damage.
2s The present study showed that EDD is a nuclear protein, most likely arising from a direct interaction with importin a via two NLSs within the N-terminus of EDD.
The HEGT domain, which has reversible ubiquitin binding activity in EDD and other ligases (Callaghan et al, Oncogene 17, 3479-3491, 1998), is also associated with a separate role in transcriptional coactivation in related proteins:
RspS/hRPF1 and 3o E6-AP coactivate ligand-dependent nuclear receptor activity (Imhof et al, Mol. Cell Biol. 16, 2594-2605, 1996; Nawaz et al, Mol. Cell Biol 19, 1182-1189, 1999), while Tom1 p is required for transcriptional regulation of certain yeast genes (Saleh et al, J. Mol. Biol. 282, 933-946, 1998) and UREB1 enhances transcription from the rat preprodynorphin gene (Gu et al, Mol. Brain Res. 24, 77-88, 1994) but suppresses p53 transactivation of target genes (Gu et al, Oncogene 11, 2175-2178, 1995).
s EDD potentiates PR transactivation to a level comparable to that seen for the p160 coactivator SRC-1. EDD has a distinct selectivity profile, being able to coactivate PR and VDR but not ER, in a ligand-dependent manner. This is in contrast to the HECT ligase E6-AP which coactivates a range of hormone io receptors including ER, PR, AR and GR (Nawaz et al, Mol. Cell Biol 19, 1182-1189, 1999). Rsp5 also shows some selectivity, coactivating transcription by PR
and GR but not ER (Imhof et al, Mol. Cell Biol. 16, 2594-2605, 1996). EDD is unique among HECT ligases however in that enhancement of PR transactivation by EDD raises the intriguing possibility of a positive feedback loop, as EDD
itself is is a progesterone-regulated gene (Callaghan et al, Oncogene 17, 3479-3491, 1998).
Thus, overexpression of EDD seen in some breast cancers could increase the sensitivity of PR-positive tumours to lower levels of progestins.
Coactivation by EDD, like E6-AP, RspS/hRPF1 and Tom1 p, is independent of 2o ubiquitin binding ability of the HECT domain (Saleh et al, J. Mol. Biol.
282, 933-946, 1998; Imhof et al, Mol. Cell Biol. 16, 2594-2605, 1996; Nawaz et al, Mol.
Cell Biol 19, 1182-1189, 1999). These findings are somewhat surprising in the light of evidence that ubiquitinylation is intimately involved in the process of transcriptional activation. Like many other transcription factors, several nuclear receptors 2s including ER, PR and VDR are down-regulated by the 26S proteasome (Nawaz et al, Proc. Natl. Acad. Sci. USA 96, 1858-1862, 1999; Alarid et al, Mol.
Endocrinol 13, 1522-1534, 1999; Lange et al, Proc. Natl. Acad. Sci. USA 97, 1032-1037, 2000; Lonard et al, Mol. Cell 5, 939-948, 2000; Masuyama et al, Biochem 71, 440, 1998) and coactivator binding appears necessary for this degradation.
3o Inhibition of the proteasome diminishes transcriptional activity by steroid receptors ER and PR (Dennis et al, Front Biosci. 6, 954-959, 2001 ) and more general implications come from studies showing that the 19S proteasome subunit is required for transcription elongation (Ferdous et al, Mol. Cell 7, 981-991, 2001).
Furthermore, the carboxy terminal tail of RNA polymerise II itself is a target of ubiquitin-mediated proteolysis (Zhu et al, Nature 400, 687-693, 1999;
Huibregtse s ef al, Proc. Natl. Acid. Sci. USA 94, 3656-3661, 1997). It may be that EDD
and these other HECT ubiquitin ligases can still perform some function in the ubiquitinylation cascade without themselves having a catalytically active HECT
domain. EDD appears to be the only E3 ligase to possess both a RING-like zinc finger domain and a HECT domain and we cannot rule out the possibility that to coactivation by EDD is mediated through the RING-like or UBA domains.
The mechanism of coactivation by E6-AP, as for the p160 family, has been attributed to direct coactivator-receptor interaction, providing either bridging or enzymatic activities to the transcriptional complex. Many steroid receptor is coactivators possess histone acetyl transferase (HAT) activity, but when compared to p300 little or no HAT activity was associated with EDD (our unpublished data). Two regions of E6-AP contain LXXLL receptor-binding motifs and both of these regions interact with PR (Nawaz et al, Mol. Cell Biol 79, 1189, 1999). A search of the EDD sequence revealed one N-terminal, one C-2o terminal and three centrally located LXXLL domains (Figure. 5A).
Furthermore, the N-terminal and centrally located motifs lie in regions of high homology to HYD.
However, the N-terminal motif, in the region with the strongest binding to PR(CDE), was not required for the interaction. Nevertheless, direct interaction between other N-terminal sequences of EDD and PR may partially explain the 2s observed effects of EDD on PR transactivation. In their studies on the role of ubiquitinylation in transcriptional enhancement, Salghetti et al found that mono-ubiquitinylation of the VP16 transcriptional activation domain was sufficient for transcriptional activity (Salghetti et al, Science 293, 1651-1653, 2001 ).
Interestingly, another study found that the UBA domain might bind such mono-3o ubiquitinylated proteins and thus prevent the formation of multi-ubiquitin chains (Bertolaet et al, Naf. Sfruct. Biol. 8, 417-422, 2001 ), raising a possible mechanism for coactivation via PR stabilisation by EDD. However, we found that the UBA
domain was not required for interaction between EDD and PR in vitro, although we cannot rule out a separate role for the UBA domain in PR coactivation by EDD.
s In addition to the UBA domain, the zf-UBR1 domain of EDD is also likely to be involved in protein-protein interactions. The zf-UBR1 domain coincides with the type 1 site in UBR1 proteins, a binding site specific for N-end rule substrates with basic N-terminal residues (Kwon et al, Proc. Natl. Acad. Sci. USA 95, 7898-7903, 1998). This zf-UBR1 domain is critical for function of the calossin-like RING-io finger protein, BIG and it therefore may also have a role in substrate recognition and binding in EDD family members. Other HECTs have substrate interaction domains distinct from the HECT domain (eg. the WW domain (Huibregtse et al, Proc. Natl. Acad. Sci. USA 94, 3656-3661, 1997)). Unfortunately attempts to use the UBA and zf UBR1 as baits for yeast two-hybrid analysis were unsuccessful is due to autoactivation and indiscriminate binding respectively, so the precise functions of these regions in the EDD protein and their role, if any, in coactivation remain elusive.
Using cell lysates EDD and CIB were shown to interact in MCF-7 and HEK 293 2o cells. CIB is 58 % and 56 % homologous with other EF-hand proteins calmodulin and calcineurin B, respectively and may function as a calcium-dependent regulatory subunit of a kinase or phosphatase (Naik et al, J. Biol. Chem 272, 4651-4654, 1997). Interactions of CIB with five other proteins have been described: DNA PK (Wu et al, Mutation Research 385, 13-20, 1997), integrin allb 2s (Naik et al, J. Biol. Chem 272, 4651-4654, 1997), the cell cycle regulatory polo-like kinases, Snk and Fnk (Kauselmann et al, EMBO J. 18, 5528-5539, 1999; Tsuboi, J. Biol. Chem. 277, 1919-1923, 2002) and preseriilin 2 (Stabler ef al, J. Cell Biol.
145, 1277-1292, 1999). Snk and Fnk are activated by progesterone in maturing frog oocytes (Duncan et al, Exp. Cell Res. 270, 78-87, 2001 ) and have roles in 3o both G1 and mitotic phases of the cell cycle and CIB could affect the activity of these kinases. CIB is found in both the nucleus and cytoplasm and its subcellular localisation can be influenced by association with its interacting partners and by calcium levels (Kauselmann et al, EMBO J. 78, 5528-5539, 1999; Stabler et al, , J.
Cell Biol. 745, 1277-1292, 1999) but its role in the nucleus is unexplored.
Interaction with DNA PK would implicate CIB in the response to DNA double s strand break sensing and repair. We found that GST-CIB pull-down of EDD
after treatment with the radiomimetic phleomycin caused a decrease in the amount of associating EDD while the levels of EDD remained unchanged. Interestingly a recent report links EDD and ubiquitinylation of another protein involved in DNA
repair, the topoisomerase 11 associated protein, TopBP1 (Honda et al, J. Bioh to Chem 277, 3599-3605, 2002). CIB interacts with PLK in the activation of CHK2. In the light of these data and our findings that CIB is a potential target of the proteasome, experiments are currently under way to determine the possible involvement of EDD and CIB in the cellular response to DNA damage.
is These data, identifying a role for EDD in transcriptional control and DNA
damage (Honda et al, J. Biol. Chem 277, 3599-3605, 2002), together with our other data demonstrating embryonic lethality in EDD-~- mice and frequent allelic imbalance at the EDD focus in diverse human cancers, provide strong evidence that EDD plays a pivotal role in normal cellular physiology and when disregulated has important 2o consequences for development and potentially tumourigenesis.

Targeted Disruption Of Edd In Mice Causes Embryonic Lethality 2s Due To A Generalised Failure Of Cell Proliferation, Generalised Apoptosis and Defective Vascularization of the Yolk Sac 3.1 Targeted Disruption of the Mouse Edd Gene Edd-deficient (Eddy) mice were generated by homologous recombination in 3o embryonic stem (ES) cells. The Edd targeting construct was designed to delete 3.4kb of Edd genomic DNA containing 61 by of exon 1 (immediately downstream of the ATG translation start site) and 3.3kb from the following intron, and replace it with a 6.5kb ~3Gal-GFP-Neo~ expression cassette (Figure 18A). This replacement was designed to produce an effectively null Edd allele and express a (3Gal-GFP
fusion protein under control of the normal Edd gene upstream regulatory s elements. The targeting construct was electroporated into 129/SvJ ES cells and several neomycin-resistant clones were isolated and screened for disruption of the Edd gene by Southern blot analysis (Figure 18B). Restriction of wild-type genomic DNA with BamHl produced a 6kb fragment. Following homologous recombination with the targeting vector, and replacement of the 5' BamHl site, a smaller 4.2kb to hybridising fragment was generated following restriction. Therefore, a correctly targeted ES cell clone produced 6kb and 4.2kb fragments following digestion with BamH1, representing the wild type and mutated alleles respectively (Figure 18B).
Two independently targeted ES cell clones (1 B2 and 5C6) were used to generate chimeric mice by injection into blastocyst stage C57BL/6 embryos. Following is backcrossing with C57BL/6 mice, F1 animals were again analysed by Southern blotting to confirm germline transmission of the mutated Edd allele. Routine genotyping of adult tail and embryo yolk sac DNA by PCR generated an amplicon of 600bp from wild-type DNA (using primers 1 (SEQ ID NO: 50) and 2 (SEQ ID
NO: 51 )). However, the primer 1 recognition sequence is deleted following 2o insertion of the targeting vector and hence the mutated allele is detected as a 440bp amplicon (using primers 2 (SEQ ID NO: 51 ) and 3 (SEQ ID NO: 52)) (Figure 18B).
Northern blot analysis showed expression of Edd mRNA in a wide range of wild-2s type (WT) adult mouse tissues, similar to the expression pattern of human EDD.
Reduced Edd mRNA expression was observed in most tissues analysed from heterozygous (Edd+~) animals (Figure 18D). However, western blot analysis showed only slightly decreased Edd protein expression in Edd+~ adult testis and E10.5 embryonic tissue (Fig15D). Edd expression was not detected in tissue from o Eddy E10.5 embryos by either western blotting or IHC (Figure 18D and 18E), confirming the efficacy of the targeting strategy. Edd was detectable by IHC
in most cells of developing WT embryos (Figure 18E), with the exception of hematopoietic cells. Neither GFP or LacZ expression was detectable in Edd+~ or Eddy embryos, most likely due to either very low ~iGal/GFP expression, or production of a non-functional fusion protein.
3.2 Phenotype of heterozygious (Edd+~) mice Both male and female Edd+~ mice on a mixed 129SV/J x C57BL/6 background were aged up to 80 weeks and showed no apparent tumorigenic phenotype, with comparable fertility and growth rates to wild-type controls.
io 3.3 Homozygous null Edd mutation (Eddy) results in embr)ionic lethality Edd+~ mice were interbred, and genotypes of offspring determined by PCR
analysis of ear, tail or embryo yolk sac (Figure 18B). No homozygous mutant (Eddy) animals were detected from 274 offspring analysed, indicating that is complete Edd deficiency (homozygosity for the Edd null mutation) causes embryonic lethality (Table 6). To characterize the developmental stage at which Edd deficiency caused embryonic lethality we analysed embryos from heterozygous intercrosses at various stages of gestation (Table 6). Mendelian ratios of wild-type, Edd+~ and Eddy embryos were observed up to E10.5. No viable 2o Eddy embryos were observed beyond E10.5. Some Eddy embryos were present in litters at E11.5 but these were partially resorbed and scored as non-viable. A
number of empty decidua were observed at E11.5-12.5 and these may have arisen from resorption of Eddy embryos at earlier stages of gestation. These data demonstrate that homozygosity for the Edd null mutation results in embryonic 2s lethality before E11.5 and indicate that Edd is essential for post-implantation embryonic development.
Heterozygous mice and Eddy embryos generated from the two independently targeted ES cell lines showed identical phenotypes (data not shown).
3o Furthermore, no difference was observed in Eddy embryos following excision of the neomycin resistance (Neo') gene from the targeted allele by crossing Edd~+

mice with mice transgenic for Cre recombinase.
3.3 Morpholoay of Eddy embryos Eddy embryos are slightly growth retarded as early as E7.5 when compared to s wild-type and Edd+~ littermates (Figure 19). By E8.5 Eddy embryos are clearly developmentally retarded, lagging at least 0.5 days behind the development of WT and Edd+~ littermates. By E9.5 and E10.5, Eddy embryos display severe growth defects, the most apparent being the absence of turning which occurs in WT embryos around E9 (Figure 19). in addition, the head is small and embryonic io structures forming the jaw region (branchial arch) are significantly under-developed. Eddy embryos are frequently observed with a swollen pericardium, indicating osmotic imbalance within the embryo. Many Edd-~ embryos also display a bulbous allantois indicating failure of chorioallantoic fusion and placentation (Figure 19). Upon histological examination, blood cell pools can be seen in is several regions of Eddy embryos. Specifically, a pool of blood cells can be seen within the pericardial cavity, indicating pericardial effusion (data not shown). Eddy embryos also appear to contain far less neural epithelium than WT and the epithelial structure is disorganised. In,short, defects in Eddy embryonic tissue are widespread and do not appear to be restricted to a specific organ system or cell 2o type.
3.4 Failure ofproliferation in Eddf embryos To assess the cause of growth retardation of Eddy embryos, cellular proliferation in developing embryos was examined by measuring BrdU incorporation into DNA
2s during S-phase. Pregnant females were injected with 10~g1g BrdU Ihr before sacrifice, embryos collected and fixed in 4% paraformaldehyde, and IHC using anti-BrdU (Dako) antibodies was then performed on paraffin sections. Positive staining indicates incorporation of BrdU into newly synthesised DNA in proliferating cells. At E8.5, the earliest stage at which a clear defect is observed in 3o Eddy embryos, there was no significant difference in mitotic index between WT
(76% ~ 2.9) and Edd-~ embryos (73% ~ 3.8) (Figure 21 ). By E9.5 a marked decrease in proliferation was observed in Edd ~ embryos. Some proliferating cells were detected (9% ~ 2.8) but this number was dramatically lower than levels observed in WT littermates (73% ~ 3.1 ) (Figure 21 ). Interestingly, BrdU
incorporation in Edd' embryos after E9.5 was restricted almost exclusively to s hematopoietic cells. Significantly, these were the only cells found not to express EDD in wild-type embryos (Figure 20). Proliferation remained high in WT
embryos at E10.5 (67% ~ 2.9) while almost no proliferation was detected in Eddy embryos (3% ~ 1.8). Hence, complete Edd deficiency results in a widespread proliferative defect in most cells of the developing mouse embryo after E8.5 (Figure 21 ), to reflecting the widespread expression of Edd in WT embryos.
3.5 Increased apoptosis in Eddy embr)~os The observation of numerous condensed nuclei in histological sections from E10.5 Edd ~ embryos, suggested that along with a proliferative block, apoptosis is was also contributing to the embryonic lethality observed in Edd-deficient embryos. TUNEL staining was used to examine apoptosis in histological sections from embryos between E8.5 - 10.5 and no significant difference was observed in numbers of TUNEL-positive nuclei at E8.5 (Figure 22A). However, numerous condensed nuclei were observed in Edct' tissue at E9.5 and E10.5 (data not 2o shown) and significantly higher levels of TUNEL-positive cells were visible in Eddy embryos compared to WT littermates at E9.5 and E10.5 (Figure 22A).
To further characterise the apparent activation of apoptosis in Edal-~
embryos, lHC
staining using an antibody that detected the cleaved (i.e. active) form of caspase-2s 3 (R&D Systems, Minneapolis, USA) was also performed. As with BrdU and TUNEL staining, no significant difference in staining for active caspase-3 was observed between WT and Eddy embryos at E8.5 (13% and 16% respectively).
However, significantly higher staining for active caspase-3 was observed in Edd embryos compared to WT littermates at both E9.5 (44% vs 6%) and E10.5 (46%
3o vs 6%) (Figure 22B), coinciding with the increased presence of both condensed nuclei and TUNEL staining in these embryos. Hence, activation of caspase-3 mediated apoptosis coincides with the block in proliferation observed in Eddy embryos, indicating that the retarded development observed at E9.5 - E10.5 results from a combination of decreased cell proliferation and increased cell death.
s 3.6 Defective rLolk sac vascularisation in Eddy embryos The widespread defects observed in Edd-~ embryos indicate a potential failure of yolk sac vasculogenesis or placentation, leading to disrupted nutrient exchange. We therefore examined the morphology of Edd-~ and WT yolk sacs.
to From E9.5 onwards, Eddy yolk sacs are clearly less well vascularised than their Edd+~ or WT littermates, suggesting a defect in yolk sac vasculogenesis.
Numerous large vessels are visible in WT and heterozygous yolk sacs at E10.5, whereas very few, smaller vessels are visible in Edd' yolk sacs at this stage (Figure 23).
High magnification of histological sections from WT and Eddy yolk sacs at E9.5 (Figure 24) shows distinct vascular channels containing blood cells (b) are visible in WT yolk sac, whereas EDD-null yolk sacs display enlarged channels with unusual separation of mesoderm (m) and endoderm (e) and few blood cells.
2o These data indicate that the differentiation of visceral mesoderm to vascular endothelium may be disrupted by EDD deficiency.
3.7 The Eddy phenotype is p53 independent Given the potential role of Edd in DNA damage response, decreased cell 2s proliferation and increased apoptosis in Edd-~ embryos, we attempted to ascertain whether a genetic relationship exists between Edd and p53. Edd~~ mice were crossed with mice heterozygous for a truncating mutation in p53 (as described by Jacks et al, Curr Biol 4:1-7, 1994) and Eddlp53 double heterozygous animals were subsequently produced. Embryos from intercrosses of these double 3o heterozygotes were examined and no difference was observed in the survival of Edd ~ embryos on a p53 ~ background (Table 7). Edd~~lp53 ~ embryos at E10.5 showed similar morphology to Edaf~~lp53+~+ embryos and all Edd~lp53~ embryos observed at E11.5 were partially resorbed, demonstrating that decreased cell proliferation and activation of apoptosis in Eddy embryos are p53-independent.
s The Role of EDD in Mammary/ Gland Development and Tumorigenesis 4.1 Production of a conditional knock-out of the Mouse EDD gene Conditional Edd-deficient (Edd~~) mice are generated by homologous to recombination in embryonic stem (ES) cells. The Edd targeting construct is designed to flank exon 1 (which contains the ATG translation start site for EDD) of the EDD gene with Cre recombinase recognition sites (IoxP). Expression of Cre recombinase in the tissues of interest causes excision of exon 1, thereby silencing EDD in those tissues.
The targeting construct is electroporated into 129/SvJ ES cells. A portion of these cells are then electroporated with an expression vector that expresses Cre recombinase and clones that show disruption of the Edd gene by Southern blot analysis determined. A portion of the selected cells, that did not express Cre 2o recombinase (ie those cell with an intact EDD gene) are used to generate chimeric mice by injection into blastocyst stage C57BL/6 embryos. Following backcrossing with C57BL/6 mice, F1 animals are analysed by Southern blotting and/or PCR (to detect the presence of the IoxP sites) to confirm germline transmission of the mutated Edd allele.
Mice expressing Cre recombinase only in mammary tissue are then generated.
An expression construct with the nucleic acid encoding Cre recombinase (SEQ ID
NO: 49) under control of the MMTV LTR (SEQ ID NO: 43), which drives expression only in mammary tissue (Wagner ef al, Nucl. Acid Res. 25(21): 4323-4330, 1997 and Ahmed et al, Cancer Res., 62(24): 7166-7169, 2002). This gene construct is then microinjected into the pronucleus of a fertilised oocyte and the oocyte introduced into the uterus of a pseudopregnant female C57BL/6 mouse.
All mice born are screened for presence of the transgene by Southern hybridisation and/or PCR to detect the nucleic acid that encodes Cre. Those mice that are found to carry the transgene are bred with WT C57BL/6 mice, and s subsequent generations are screened for expression of Cre recombinase in mammary tissue using an anti-Cre antibody (Novagen, Madison, WI, USA).
Multiple tissues of positive mice are subsequently screened to ensure that the expression of Cre is limited to the mammary tissue.
to Transgenic mice expressing Cre and mice carrying floxed EDD are then crossed to generate mice that have had exon 1 of EDD excised in mammary tissue.
Expression of EDD in several tissues (including mammary tissue) is determined by both Western blotting and immunohistochemistry as described in Example 3.
Those mice that do not express EDD in mammary tissue, but maintain expression is of EDD in all other tissues in which EDD is known to be expressed. These mice are then analysed to establish the role of EDD in mammary cell development and tumorigenesis.
4.2 Role of EDD in mammary gland development 2o Female mice deficient for EDD in mammary tissue are analysed to determine mammary gland architecture and function. Pregnant female mice (wt and EDD-~-) are sacrificed every 5 days during pregnancy (where day 0 is considered the date on which a post-coital plug is first detected). Mammary tissue is then dissected from these animals and analysed using whole-mount microscopy to determine 2s terminal end bud density, internodal duct length, branching pattern and alveolar bud formation, and to determine the effect of EDD on these parameters (essentially as described in Hovey ef al, Mol. Endocrinol. 7 7(3): 460-473, and references therein). Additionally, the effect of EDD are analysed at weekly intervals for three weeks during lactation and at 5 day intervals for 6 weeks 3o following weaning.

At all time points mammary glands are also sectioned and stained with haemotoxylin and eosin to facilitate examination of the cellular structure of the glands. Additionally, glands are analysed for expression of EDD and EDD
interacting proteins, such as, for example, BRCA2, CHK2, TopB1 and CIB.
As recent studies have suggested that the drosophila homolog of EDD (hyd) is capable of re uq lating the expression of hedgehog and decapentaplegic, mammary tissue isolated from EDD-'- mice are analyzed for hedgehog signaling rJathway components eg ihh shh dhh GIi1, Gli2, GIi3, BMP-2, BMP-4 and to patched proteins.
In order to determine the effect of EDD in the stromal and epithelial compartments of mammary tissue epithelial tissue from an EDD-'- mice are transplanted into a fat pads of a WT mouse and e~~ithelial tissue from a wt mouse is transplanted into a is fatpad of a EDD-'- mouse (essentially as described in Naylor and Ormandy, Dev.
Dyn, 225(1): 100-105, 2002 and references cited therein).
4.3 Role of EDD in mammary tumorigenesis In order to assess carcinogenesis in mice carrying a targeted disruption of EDD in 2o the mammary gland, mice are aged for up to a period of 2 years. At various stages mice are sacrificed and analyzed for the development of spontaneous mammary tumors. The rate of tumor development in EDD-'- mice and wt mice at these stages are then compared to determine the effect of EDD on the development of mammary tumors in ageing mice.
Additionally, EDD-'- mice are crossed with the MMTV/c-myc transgenic mice (Romieu-Mourez et al, Mol. Cell 8iol, 23(16): 5738-5754, 2003). or the MMTV/wnt-1 transgenic mice (Bocchinfuso et al, Cancer Res 59(8): 1869-1876, 1999) (both of which show a high incidence of mammary tumor formation). WT, transgenic 3o and transgenic/knockout mice are then analyzed to determine the effect of EDD
on latency and frequency of tumor formation. Mammary glands are isolated from the mice at various stages of pregnancy and lactation and various ages and analyzed using whole mount analysis to determine the number of hyperplastic alveolar nodules, hyperplasias and tumors.
s 4.4 Overexpression of EDD in mammary tissue In order to determine the effect of EDD on the development of mammary tissue and mammary tumorigenesis mice overexpressing EDD in this tissue are generated. An expression construct with the nucleic acid encoding EDD (SEQ ID
NO: 1 ) under control of the MMTV LTR (SEQ ID NO: 48), which drives expression to only in mammary tissue is produced. This gene construct is then microinjected into the pronucleus of a fertilised oocyte and the oocyte introduced into the uterus of a pseudopregnant female C57BL/6 mouse. All mice born are screened for presence of the transgene by Southern hybridisation and/or PCR to detect the transgenic construct encodes mammary tissue specific EDD. Those mice that are is found to carry the transgene are bred with WT C57BL/6 mice, and subsequent generations are screened for overexpression of EDD in mammary tissue using Western Blotting as described in Example 3. Multiple tissues of positive mice are subsequently screened to ensure that the overexpression of EDD is limited to the mammary tissue.
4.5 Role of EDD in mammary development Female mice overexpressing EDD in mammary tissue are analysed to determine mammary gland architecture and function. Mammary tissue is isolated from pregnant female mice (wt and tgEDD+~-) every 5 days during pregnancy, in 2s addition to weekly intervals for three weeks during lactation and at 5 day intervals for 6 weeks following weaning Dissected mammary tissue is then analysed using whole-mount microscopy to determine terminal end bud density, internodal duct length, branching pattern and alveolar bud formation, and to determine the effect of EDD on these parameters (essentially as described in Example 4.2).
At all time points mammary glands are also sectioned and stained with haemotoxylin and eosin to facilitate examination of the cellular structure of the glands. Additionally, glands are analysed for expression of EDD and EDD
interacting proteins, such as, for example, BRCA2, CHK2, TopB1 and CIB, in addition to hedgehog siqnalin~ ap thwy components, eg ihh, shh, dhh, GIi1, GIi2, s GIi3, BMP-2, BMP-4 and patched proteins.
4.3 Role of EDD in mammary tumorigenesis In order to assess carcinogenesis in mice overexpressing EDD in the mammary gland, mice are aged for up to a period of 2 years. At various stages mice are to sacrificed and analyzed for the development of spontaneous mammary tumors.
The rate of tumor development in tgEDD+~- mice and wt mice at these stages are then compared to determine the effect of EDD on the development of mammary tumors in ageing mice.
Is Targeted downregulation of EDD expression using siRNA inhibits cell proliferation and disrupts cell-cell contacts 20 5.1 Method An siRNA was designed to inhibit the expression of EDD (sequence: sense 5'-GCAGUGUUCCUGCCUUCUUdTdT-3' (SEQ ID NO: 47), anti-sense 5'-dTdTCGUCACAAGGACGGAAGAA-3' (SEQ ID N0:48)), siRNA was synthesised, annealed and HPLC purified by Xeragon (Zurich, Switzerland). 2.2x106 MCF-7 2s cells or 2.6 x106 HEK-293 cells were plated in a 15 cm2 dish and grown overnight.
The normal breast epithelial cell lines HMEC 184 and MCF-10-A were grown in MCDB 170 media (Gibco) with 1 % pituitary extract as an additive.
For transfection of MCF-7 or HEK-293 cells were washed with serum-free medium 3o and 15 ml of serumfree medium was placed on the cells. 29 w1 of Oligofectamine (Life Sciences) was diluted in 220,1 Optimern (Life Sciences) (incubated for 5 min) and 20.5 p,l siRNA (20 ~M) was diluted in 2.5 ml Optimem. The siRNA solution and Oligofectamine solutions were gently mixed and incubated for 24 min. The combined solution was added to the 15 cm2 plate of cells in serum-free medium.
After 4 hrs 15 ml of medium containing 30% fetal calf serum was added to the s cells. After another 24 hrs cells were split into 10 cm2 plates for each time point treatment.
For transfection, HMEC 184 and MCF10-A cells were plated at 1.2 x 106 cells/15 cm plate. Transfection conditions were the same as above except HMEC 184 and to MCF10-A cells were transfected in pituitary extract-free medium and after 4 h 2%
pituitary extract-containing medium is added. 8 hrs after transfection this medium was replaced with 1 % pituitary extract-containing medium A siRNA targeted against green fluorescence protein (GFP) was used as a negative control.
5.2 Results Transfection of EDD RNAi in all cell lines resulted in substantial loss of EDD
protein as assessed by Western blotting with an anti-EDD antibody (Figure 25).
After transfection with EDD siRNA, changes in cell morphology were seen in 2o HMEC 184 cells and MCF-10A cells. This change was first observed two days after RNA interference was performed. Cells transfected with EDD siRNA showed reduced and altered cell-cell contacts, cell shape was altered and cells were disorganised, compared to control cells (Figure 26). Control cells tended to be elongated, making cell-cell contacts with neighbouring cells along the length of the 2s cells, forming a sheet. Cells transfected with EDD siRNA made fewer connections with neighbouring cells, while their more rounded shape prevents them from making connections along the length of the cells.
Cell morphology was further analysed using immunofluorescent antibodies to 3o detect proteins involved in cell-cell contacts (~i-catenin) and organisation of the cytoskeleton (actin). -catenin staining in HMEC 184 cells showed differences at cell-cell contacts after depletion of EDD. Staining in control cells was even along the cell-cell contact (Figure 27), whereas this region in cells transfected with EDD
siRNA showed reduced expression of ~i-catenin and a patchier staining (Figure 27). 67% of cell-cell contacts between 184 cells depleted of EDD had patchy s expression (compared with 36°l° in control). Further examination showed that EDD
RNAi causes a relocalization of ~i-catenin from the cell periphery to the cell nucleus (Figure 28).
Actin filaments in HMEC 184 control cells were coordinated with actin in adjacent to cells (Figure 29), whereas this organisation between neighbouring cells was not seen in cells transfected with EDD siRNA (figure 29).
Similar alterations of ~i-catenin and actin were seen in MCF-10A cells depleted of EDD.
~s Identification of Downstream Effects of EDD Silencing To ascertain biochemical pathways that are affected by EDD activity in cells, 2o transcript profiling experiments were performed using Affymetrix DNA
microarrays.
HMEC 184 and MCF-7 cells were transfected with siRNA (EDD or GFP) as described in Example 5.1. Loss of EDD was confirmed by Western or Northern blot.
2s Cells were harvested after 24 hours and 48 hours and RNA purified. RNA was then reverse transcribed using an oligo(dT) anchored oligonucleotide that additionally comprised a T7 promoter sequence. Isolated cDNA was then transcribed in vitro using the T7 MEGAscript kit (Ambion, Austin, TX, USA) according to manufacturer's instructions. Transcription was perFormed with 3o biotinylated nucleotides (Bio-11-CTP and Bio-16-UTP) to facilitate detection of the transcribed nucleic acid.

Experiments were performed in triplicate and pooled for the Affymetrix probe.
Affymetrix U133A chips were used in all experiments.
s Data analysis identified genes which had decreased or increased mRNA
expression in response to EDD depletion by RNAi.
As shown in Table 8 the expression of a variety of genes that are associated with cell cycle control (eg. Cyclin D3), oncogenes (eg R-ras) and genes associated io with cell-cell contact and cell morphology (eg Collagen IVa1 ) is modulated by the silencing of EDD. These changes in gene expression potentially explain the morphological changes observed in HMEC 184 and MCF-7 cells as a result of EDD silencing (ie. altered cell proliferation, altered cell-cell contact, and altered (3-catenin and actin localization).
is ~

r M

_ M

C) N

a ~r r ~ ~...~ ...~ ...~ ...

O M ~COO M r tl7r r 1~~f~
O

- M M MN M r N N M N ON

.N ((~V V V~IV ~I V V ~IV V~I

O

NM ~ v r r r ~

OM w -lI7(flO d' w w w Od'U
cw w w w w w Qr in~7r-N r- ~ M d'd'ON ~

a N .

=

4 ~ 'a v ~

C7 M r~ ~ .-.~ ~ 0 r ...N O

tnN M Mr r ~C O ~t!M M OM ,.O

......
47LO I~..~

~ 00CO ~

C~ ~ O ~ M O
M w ww w w w w w w WO O

N De- Cfltt~r-~-~ O ~ COd'ON O

N

O' C

O

U

O ~ ~ ~~ ~ ~ ~ ~

h O NM O .-.~ 00M h O
' O OM fitNM ~YO O M 47N M
! v~ w r v ~ ~ v ~
~

~ >>
M~ O y_'' O
?

O "'' ~M r OM ~ O) N o0r r Q~ ' ON w ww w w w w w w ~1~tn Dc- I~Nc-N O O M o0~f'ZN N tn O
L

V

O X

V

r 0 O.-.M ...~ O M ~ .~.O '.
O ' - Iwd' ~f7d'O M O O N r d OM tCf Q ~. ~ .. .r. . ~ . .
.. . . . .

L ~ ... . . . .

_ r ~ O ~ ~ ~ L

>, Oh- ~ w~ Cflw M N w I~.O)C O
w w w w w Oc~ t-'d'O N O O d'. COON W
N U

O
..' O ~
~ -O

~ ~ ~~ ~ 1 C L
00 M OM M ~ .~ ~ ~ ~ ~~ ~ ~ O

( .0 ~ ~ ~~ ~ ~ ~ M '~Yr tf7~t v ~ 'r~v D0" N ~ 'V ~

U d ~- O GOM r- C
~~. w ~a0Ln~ M r r c-d'~ ~ ,~
N COww w w w w.w so0.(6 C~N ~ d'~-r-O O COCON NM

U >

p O

~ ~ OQ

~ ~~.~ ~ v.~ ~ ~~
O

O 00r~ M ~ ~ stO 'd'hWit' tCd' d'etr M O N O 't7~ s!!v t~ O
r.

Na ....~~ ...'. ~. v . m.~rM

~ ~ O
CD

ON C'VWs ~ W ~ v ~ ~ v ~
~ C

DN r I~r N O r- f~I~I'd''d'-O
~ U C
'O
(n L
S O

(( Q. O
O_S

O O
O _ ~

O d 3 O ~ .O
(0 ~
V

c .cn E ~ c'a ' N
>
ca u~
~

ca a~~ ~ v v E _ ~

CC O O~ L R IC '~,~O OM = ~ 7 O1 ~ -a 'O O

~L ~ -OV ~ ~L ~L ~ (~ yTI~ (~ O
- O C L V O
~ O C
V
V

t3 N C~ .L..~S tC d O.- C.~.~X
> O d > G7d N
~ ~ m ~ LLI
m O f~LLI O O L Z ~ OO ~
~ ~~ ' Z

1 1 II I I 1 ~ N M
d Table 2 Clinicopathological characteristics of a serous ovarian cancer patient cohort Parameter Age 59.6 (26.4-86.4) Grade of DiseaseG1=7; G2=73; G3=80 (n=160) FIGO Stage S1=10; S2=8; S3=121; S4=26 (n=165) Length of Follow28.2 months (0-133.4) up Outcome 101 died related to malignancy 5 died unrelated to malignancy 59 alive ~°io~.~: od~-ol V7 O M V1 O ~O O
O O O O O O O O

n r~r~ s~r~n s~~
m U 0 ~ ~ r o ~ m ~

~' O y o ? y o o r N ooN

. ~ ~ '~~ ~ N M M ~ M
V
, a. N O oo ~t~ M l~00 ., ~ ~

+~C, '~
~ O O I~d~~ l M

~ riO O O .--,O ,~

a a v W o ~ ~ N

cd b~o ' N I~~ d N o0 d ' ~t , ~ d',l N M Oy-!O
c3 O ~ O ,-~N '-i,-;,-H.~(vj ..-i U ' cd t~~o O ~

, 0 o O o 0 ~ ~
, t-i m ~ ~ s~ ~ ~ r~
W ~ U o00 ~.~,,~ d~~ ov-~ v \ vON ~ ~l~lW

~., ~ , ' d ~ N r-,l d N N
~

C~. ,~

v' ~Od'~p M ~O00O d' ,~ .~o 0 0 0 ~ ~

O ~ o O ~ O ~ v O O

H ~

~ ' d ~ O O~l O ~O .-~Q~U '~td;

, ~ .-i.-iM ,-~M N '-;,-i ~

U

O

O

c~

O

U ~' .
,.., O

V ~ Q.

O

V yn ~ O N
~

C~ U

n ~~

~ ~, ~ N ~

N o > ~ urA

~

o ~ ""
o V ~

V v ~ o P;W

y a>

~
N o ~ a v~C7 'bQ, U p.~ N
~ a P~
N
~
~W

Table 4 Multivariate analysis of clinicopathological parameters and EDD
immunoreactivity to patient outcome.
Disease-specific Survival Variable Subgroup Hazards Ratio (95%P
C.L) Stage continuous 1.635 (1.165 - 0.0045 2.295) Grade 1 vs 2,3 3.745 (0.912 -15.375)0.0669 Performance 0,1 vs 2,3 2.692 (1.376 - 0.0038 status 5.268) Residual Disease<2cm vs >2cm 2.149 (1.327 - 0.0019 3.481) EDD expressionEDD negative vs EDD 1.374 (0.884 - 0.1578 positive 2.135) Relapse-free Survival Variable Subgroup Hazards Ratio (95°~o C.L) P
Stage continuous 1.494 (1.106 - 2.019) 0.0089 Residual Disease <2cm vs >2cm 1.674 (1.007 - 2.781) 0.0468 EDD expression EDD negative vs EDD positive 2.301 (1.454 - 3.643) 0.0004 Table 5 Summary of EDD sequence variants detected in 26 normal epithelial and cancer cell lines.
Nucleotide Predicted " Codon Base change aaz Cell lines) positio change 4753 1584 C-~A His->Asn SK-BR-3 6279 2093 A-aG Asn~Ser IGROV-1 886-902 296-300 Splice variantVLLPL removedDetected in all cell lines 7689 ABC No change Hs 578T

4055 CST No change BT-20 3 Human mRNA

4390 A~G No change Human mRNA3 4556 A~G No change T-47D

3956 A~G No change ZR-75-1 LnCap Hs 578T

7634 CAA No change Hs 578T

Base changes in bold confirmed at the genomic level.
' Nucleotide numbering starts at the A of the initiation codon ~ aa, amino acid 3 David Anderson (unpublished data) Table 6 Genotype analysis of embryos from Edd+~- Intercrosses. Observed and expected genotype frequencies (in brackets) at various stages of gestation.
~ -Genotype 8. 9.5 10.5 11.5 12.5 Edd+~+ 14(11 ) 3(4) 10(7) 6(4) 9(7) Edd+~- 18(22) 10(9) 12(10) 8(9) 20(15) Edd-'- 11 (11 ) 4(4) 7(7) 0(4) 0(7) Table 7.
Genotypic analysis of offspring from Edd~~°1p53+~° double heterozygous intercrosses.
AGE (days post coitum) GENOTYPE 9.5 10.5 11.5 p53+~+ Edd+~+ 1 2 3 Edd+~ 3 8 4 Edd~ 4 0 0 p53+~ Edd+~+ 4 4 3 Edd+~ 4 12 11 Edd~ 1 4 1 p53~ Edd+~+ 2 4 2 Edd+~ 2 4 2 Edd~ 1 2 6*

Total 22 40 32 * Embryos were partially resorbed and scored as non-viable.

Table 8 Gene Expression Changes in HMEC
184 and MCF-7 Cells as a Result of EDD

Silencing Gene expression Gene expression Gene expression modulated in HMEC modulated in HMEC modulated in and and MCF-7 cells cells , cells R-ras (T) Coronin (T) Id-1 (T) WAVE-2 (T) Neuregulin1 (T) paxillin (T) AP1 mu2 (T) Syndecan-4 (T) Twinfilin (T) SFRP-1 (J.) Raf1 (T) FK506BP5 (,~) Cyclin D (T)3 SERPIN H1 (,~) RhoB (T) Collagen IVa1 (,~) ARP 2/3 complex WntSa (~.) (~) Core binding factor beta (T) (T) indicates that gene expression is increased (,~) indicates that gene expression is decreased SEQUENCE LISTING
<110> Garvan Institute of Medical Research Clancy, Jennifer 0'Brien, Phillipa Saunders, Darren Henderson, Michelle Watts, Colin Sutherland, Robert Henshall, Susan <120> Novel diagnostic and therapeutic methods and reagents therefor <130> 500595/MRO
<l60> 53 <170> PatentIn version 3.1 <210> 1 <211> 8509 <212> DNA
<213> human Edd <400>

cgccctcgagtggaggacgagaaggaaagcaccatgacgtccatccatttcgtggttcac60 ccgctgccgggcaccgaggaccagctcaatgacaggttacgagaagtttctgagaagctg120 aacaaatataatttaaacagccacccccctttgaatgtattggaacaggctactattaaal80 cagtgtgtggtgggaccaaatcatgctgcctttcttcttgaggatggtagagtttgcagg240 attggtttttcagtacagccagacagattggaattgggtaaacctgataataatgatggg300 tcaaagttgaacagcaactcgggggcagggaggacgtcaaggcctggtaggacaagcgac360 tctccatggtttctctcaggttctgagactctaggcaggctggcaggcaacaccttagga420 agccgctggagttctggagtgggtggaagtggtggaggatcctctggtaggtcatcagct480 ggagctcgagattcccgccggcagactcgagttattcggacaggacgggatcgagggtct540 gggcttttgggcagtcagccccagccagttattccagcatctgtcattccagaggagctg600 atttcacaggcccaagttgttttacaaggcaaatccagaagtgtcattattcgagaactt660 cagagaacaaatcttgatgtgaaccttgctgtaaataatttacttagccgggatgatgaa720 gatggagatgatggggatgatacagccagcgaatcttatttgcctggagaggatcttatg780 tctctccttgatgccgacattcattctgcccacccaagtgtcattattgatgcagatgcc840 atgttttctgaagacattagctattttggttacccttcttttcgtcgttcatcactttcc900 aggctaggctcatctcgagttctccttcttcccttagagagagactctgagctgttgcgt960 gaacgtgaatccgttttacgtttacgtgaacgaaggtggcttgatggagcctcatttgat1020 aatgaaaggggttctaccagcaaggaaggagagccaaacttggataagaagaatacacct1080 gttcaaagtccagtatctctaggagaagatttgcagtggtggcctgataaggatggaaca1140 aaattcatctgtattggggctctgtattctgaacttctggctgtcagcagtaaaggagaa1200 ctttatcagtggaaatggagtgaatctgagccttacagaaatgcccagaatccttcatta1260 catcatccacgagcaacatttttggggttaaccaatgaaaagatagtcctcctgtctgca1320 aatagcataagagcaactgtagctacagaaaataacaaggttgctacatgggtggatgaa1380 actttaagttctgtggcttctaaattagagcacactgctcagacttactctgaacttcaa1440 ggagagcggatagtttctttacattgctgtgccctttacacctgcgctcagctggaaaac1500 agtttatattggtggggtgtagttccttttagtcaaaggaagaaaatgttagagaaagct1560 agagcaaaaaataaaaagcctaaatccagtgctggtatttcttcaatgccgaacatcact1620 gttggtacccaggtatgcttgagaaataatcetctttatcatgctggagcagttgcattt1680 tcaattagtgctgggattcctaaagttggtgtcttaatggagtcagtttggaatatgaat1740 gacagctgtagatttcaacttagatctcctgaaagcttgaaaaacatggaaaaagctagc1800 aaaactactgaagctaagcctgaaagtaagcaggagccagtgaaaacagaaatgggtcct1860 ccaccatctccagcatccacgtgtagtgatgcatcctcaattgccagcagtgcatcaatg1920 ccatacaaacgacgacggtcaacccctgcaccaaaagaagaggaaaaggtgaatgaagag1980 cagtggtctcttcgggaagtggtttttgtggaagatgtcaagaatgttcctgttggcaag2040 gtgctaaaagtagatggtgcctatgttgctgtaaaatttccaggaacctccagtaatact2100 aactgtcagaacagctctggtccagatgctgacccttcttctctcctgcaggattgtagg2160 ttacttagaattgatgaattgcaggttgtcaaaactggtggaacaccgaaggttcccgac2220 tgtttccaaaggactcctaaaaagctttgtatacctgaaaaaacagaaatattagcagtg2280 aatgtagattccaaaggtgttcatgctgttctgaagactggaaattgggtgcgatactgt2340 atctttgatcttgctacaggaaaagcagaacaggaaaataattttcctacaagcagcatt2400 gctttccttggtcagaatgagaggaatgtagccattttcactgctggacaggaatctccc2460 attattcttcgagatggaaatggtaccatctacccaatggccaaagattgcatgggagga2520 ataagggatcccgattggctggatcttccacctattagtagtcttggaatgggtgtgcat2580 tctttaataaatcttcctgccaattcaacaatcaaaaagaaagctgctgttatcatcatg2640 gctgtagagaaacaaaccttaatgcaacacattctgcgctgtgactatgaggcctgtcga2700 caatatctaatgaatcttgagcaagcggttgttttagagcagaatctacagatgctgcag2760 acattcatcagccacagatgtgatggaaatcgaaatattttgcatgcttgtgtatcagtt2820 tgctttccaaccagcaataaagaaactaaagaagaagaggaagcggagcgttctgaaaga2880 aatacatttgcagaaaggctttctgctgttgaggccattgcaaatgcaatatcagttgtt2940 tcaagtaatggcccaggtaatcgggctggatcatcaagtagccgaagtttgagattacgg3000 gaaatgatgagacgttcgttgagagcagctggtttgggtagacatgaagctggagcttca3060 tccagtgaccaccaggatccagtttcaccccccatagctccccctagttgggttcctgac3120 cctcctgcgatggatcctgatggtgacattgattttatcctggcccccgctgtgggatct3180 cttaccacagcagcaaccggtactggtcaaggaccaagcacctccactattccaggtcct3240 tccacagagccatctgtagtagaatccaaggatcgaaaggcgaatgctcattttatattg3300 aaattgttatgtgacagtgtggttctccagccctatctacgagaacttctttctgccaag3360 gatgcaagagggatgaccccatttatgtcagctgtaagtggccgagcttatcctgctgca3420 attaccatcttagaaactgctcagaaaattgcaaaagctgaaatatcctcaagtgaaaaa3480 gaggaagatgtattcatgggaatggtttgcccatcaggtaccaaccctgatgactctcct3540 ttatatgttttatgttgtaatgacacttgcagttttacatggactggagcagagcacatt3600 aaccaggatatttttgagtgtcgaacttgtggcttgctggagtcactgtgttgttgtacg3660 gaatgtgcaagggtttgtcataaaggtcatgattgcaaactcaaacggacatcaccaaca3720 gcctactgtgattgttgggagaaatgtaaatgtaaaactcttattgctggacagaaatct3780 gctcgtcttgatctactttatcgcctgctcactgctactaatctggttactctgccaaac3840 agcaggggagagcacctcttactattcttagtacagacagtcgcaaggcagacggtggag3900 cattgtcaatacaggccacctcgaatcagggaagatcgtaaccgaaaaacagccagtcct3960 gaagattcagatatgccagatcatgatttagagcctccaagatttgcccagcttgcattg4020 gagcgtgttctacaggactggaatgccttgaaatctatgattatgtttgggtcgcaggag4080 aataaagaccctcttagtgccagcagtagaataggccatcttttgccagaagagcaagta4140 tacctcaatcagcaaagtggcacaattcggctggactgtttcactcattgccttatagtt4200 aagtgtacagcagatattttgcttttagatactctactaggtacactagtgaaagaactc4260 caaaacaaatatacacctggacgtagagaagaagctattgctgtgacaatgaggtttcta4320 cgttcagtggcaagagtttttgttattctgagtgtggaaatggcttcatccaaaaagaaa4380 aacaactttattccacagccaattggaaaatgcaagcgtgtattccaagcattgctacct4440 tacgctgtggaagaattgtgcaacgtagcagagtcactgattgttcctgtcagaatgggg4500 attgctcgtccaactgcaccatttaccctggctagtactagcatagatgccatgcagggc4560 agtgaagaattattttcagtggaaccactaccaccacgaccatcatctgatcagtctagc4620 agctccagtcagtctcagtcatcctacatcatcaggaatccacagcagaggcgcatcagc4680 cagtcacagcccgttcggggcagagatgaagaacaggatgatattgtttcagcagatgtg4740 gaagaggttg aggtggtgga gggtgtggct ggagaagagg atcatcatga tgaacaggaa 4800 gaacacgggg aagaaaatgc tgaggcagag ggacaacatg atgagcatga tgaagacggg 4860 agtgatatgg agctggactt gttagcagca gctgaaacag aaagtgatag tgaaagtaac 4920 cacagcaacc aagataatgc tagtgggcgc agaagcgttg tcactgcagc aactgctggt 4980 tcagaagcag gagcaagcag tgttcctgcc ttcttttctg aagatgattc tcaatcgaat 5040 gactcaagtg attctgatag cagtagtagt cagagtgacg acatagaaca ggagaccttt 5100 atgcttgatg agccattaga aagaaccaca aatagctccc atgccaatgg tgctgcccaa 5160 gctccccgtt caatgcagtg ggctgtccgc aacacccagc atcagcgagc agccagtaca 5220 gccccttcca gtacatctac accagcagca agttcagcgg gtttgattta tattgatcct 5280 tcaaacttac gccggagtgg taccatcagt acaagtgctg cagctgcagc agctgctttg 5340 gaagctagca acgccagcag ttacctaaca tctgcaagca gtttagccag ggcttacagc 5400 attgtcatta gacaaatctc ggacttgatg ggccttattc ctaagtataa tcacctagta 5460 tactctcaga ttccagcagc tgtgaaattg acttaccaag atgcagtaaa cttacagaac 5520 tatgtagaag aaaagcttat tcccacttgg aactggatgg tcagtattat ggattctact 5580 gaagctcaat tacgttatgg ttctgcatta gcatctgctg gtgatcctgg acatccaaat 5640 catcctcttc acgcttctca gaattcagcg agaagagaga ggatgactgc gcgagaagaa 5700 gctagcttac gaacacttga aggcagacga cgtgccacct tgcttagcgc ccgtcaagga 5760 atgatgtctg cacgaggaga cttcctaaat tatgctctgt ctctaatgcg gtctcataat 5820 gatgagcatt ctgatgttct tccagttttg gatgtttgct cattgaagca tgtggcatat 5880 gtttttcaag cacttatata ctggattaag gcaatgaatc agcagacaac attggataca 5940 cctcaactag aacgcaaaag gacgcgagaa ctcttggaac tgggtattga taatgaagat 6000 tcagaacatg aaaatgatga tgacaccaat caaagtgcta ctttgaatga taaggatgat 6060 gactctcttc ctgcagaaac tggccaaaac catccatttt tccgacgttc agactccatg 6120 acattccttg ggtgtatacc cccaaatcca tttgaagtgc ctctggctga agccatcccc 6180 ttggctgatc agccacatct gttgcagcca aatgctagaa aggaggatct ttttggccgt 6240 ccaagtcagg gtctttattc ttcatctgcc agtagtggga aatgtttaat ggaggttaca 6300 gtggatagaa actgcctaga ggttcttcca acaaaaatgt cttatgctgc caatctgaaa 6360 aatgtaatga acatgcaaaa ccggcaaaaa aaagaagggg aagaacagcc cgtgctgcca 6420 gaagaaactg agagttcaaa accagggcca tctgctcatg atcttgctgc acaattaaaa 6480 agtagcttac tagcagaaat aggacttact gaaagtgaag ggccacctct cacatctttc 6540 aggccacagt gtagctttat gggaatggtt atttcccatg atatgctgct aggacgttgg 6600 cgcctttctttagaactgttcggcagggtattcatggaagatgttggagcagaacctgga6660 tcaatcctaactgaattgggtggttttgaggtaaaagaatcaaaattccgcagagaaatg6720 gaaaaactgagaaaccagcagtcaagagatttgtcactagaggttgatcgggatcgagat6780 cttctcattcagcagactatgaggcagcttaacaatcactttggtcgaagatgtgctact6840 acaccaatggctgtacacagagtaaaagtcacatttaaggatgagccaggagagggcagt6900 ggtgtagcacgaagtttttatacagccattgcacaagcatttttatcaaatgaaaaattg6960 ccaaatctag agtgtatcca aaatgccaac aaaggcaccc acacaagttt aatgcagaga 7020 ttaaggaacc gaggagagag agaccgggaa agggagagag aaagggaaat gaggaggagt 7080 agtggttt°gc gagcaggttc tcggagggac cgggatagag actttagaag acagctttcc 7140 atcgacacta ggccctttag accagcctct gaagggaatc ctagcgatga tcctgagcct 7200 ttgccagcac atcggcaggc acttggagag aggctttatc ctcgtgtaca agcaatgcaa 7260 ccagcatttg caagtaaaat cactggcatg ttgttggaat tatccccagc tcagctgctt 7320 ctccttctag caagtgagga ttctctgaga gcaagagtgg atgaggccat ggaactcatt 7380 attgcacatg gacgggaaaa tggagctgat agtatcctgg atcttggatt agtagactcc 7440 tcagaaaagg tacagcagga aaaccgaaag cgccatggct ctagtcgaag tgtagtagat 7500 atggatttag atgatacaga tgatggtgat gacaatgccc ctttgtttta ccaacctggg 7560 aaaagaggat tttatactcc aaggcctggc aagaacacag aagcaaggtt gaattgtttc 7620 agaaacattg gcaggattct tggactatgt ctgttacaga atgaactctg tcctatcaca 7680 ttgaatagac atgtaattaa agtattgctt ggtagaaaag tcaattggca tgattttgct 7740 ttttttgatc ctgtaatgta tgagagtttg cggcaactaa tcctcgcgtc tcagagttca 7800 gatgctgatg ctgttttctc agcaatggat ttggcatttg caattgacct gtgtaaagaa 7860 gaaggtggag gacaggttga actcattcct aatggtgtaa atataccagt cactccacag 7920 aatgtatatg agtatgtgcg gaaatacgca gaacacagaa tgttggtagt tgcagaacag 7980 cccttacatg caatgaggaa aggtctacta gatgtgcttc caaaaaattc attagaagat 8040 ttaacggcag aagattttag gcttttggta aatggctgcg gtgaagtcaa tgtgcaaatg 8100 ctgatcagtt ttacctcttt caatgatgaa tcaggagaaa atgctgagaa gcttctgcag 8160 ttcaagcgtt ggttctggtc aatagtagag aagatgagca tgacagaacg acaagatctt 8220 gtttactttt ggacatcaag cccatcactg ccagccagtg aagaaggatt ccagcctatg 8280 ccctcaatca caataagacc accagatgac caacatcttc ctactgcaaa tacttgcatt 8340 tctcgacttt acgtcccact ctattcctct aaacagattc tcaaacagaa attgttactc 8400 gccattaaga ccaagaattt tggttttgtg tagagtataa aaagtgtgta ttgctgtgta 8460 atattactag caaattttgt agattttttt ccatttgtct ataaaagtt 8509 <2l0> 2 <211> 2799 <212> PRT
<213> human EDD protein <400> 2 Met Thr Ser Ile His Phe Val Val His Pro Leu Pro Gly Thr Glu Asp 1 5 l0 15 Gln Leu Asn Asp Arg Leu Arg Glu Val Ser Glu Lys Leu Asn Lys Tyr Asn Leu Asn Ser His Pro Pro Leu Asn Val Leu Glu Gln Ala Thr Ile Lys Gln Cys Val Val Gly Pro Asn His Ala Ala Phe Leu Leu Glu Asp Gly Arg Val Cys Arg Ile Gly Phe Ser Val Gln Pro Asp Arg Leu Glu Leu Gly Lys Pro Asp Asn Asn Asp Gly Ser Lys Leu Asn Ser Asn Ser Gly Ala Gly Arg Thr Ser Arg Pro Gly Arg Thr Ser Asp Ser Pro Trp Phe Leu Ser Gly Ser Glu Thr Leu Gly Arg Leu Ala Gly Asn Thr Leu Gly Ser Arg Trp Ser Ser Gly Val Gly Gly Ser Gly Gly Gly Ser Ser Gly Arg Ser Ser Ala Gly Ala Arg Asp Ser Arg Arg Gln Thr Arg Val Ile Arg Thr Gly Arg Asp Arg Gly Ser Gly Leu Leu Gly Ser Gln Pro Gln Pro Val Ile Pro Ala Ser Val Ile Pro Glu Glu Leu Ile Ser Gln Ala Gln Val Val Leu Gln Gly Lys Ser Arg Ser Val Ile Ile Arg Glu Leu Gln Arg Thr Asn Leu Asp Val Asn Leu Ala Val Asn Asn Leu Leu 2l0 215 220 Ser Arg Asp Asp Glu Asp Gly Asp Asp Gly Asp Asp Thr Ala Ser Glu Ser Tyr Leu Pro Gly Glu Asp Leu Met Ser Leu Leu.Asp Ala Asp Ile His Ser Ala His Pro Ser Val Ile Ile Asp Ala Asp Ala Met Phe Ser Glu Asp Ile Ser Tyr Phe Gly Tyr Pro Ser Phe Arg Arg Ser Ser Leu Ser Arg Leu Gly Ser Ser Arg Val Leu Leu Leu Pro Leu Glu Arg Asp 5er Glu Leu Leu Arg Glu Arg Glu Ser Val Leu Arg Leu Arg Glu Arg Arg Trp Leu Asp Gly Ala Ser Phe Asp Asn Glu Arg Gly Ser Thr Ser Lys Glu Gly Glu Pro Asn Leu Asp Lys Lys Asn Thr Pro Val Gln Ser Pro Val Ser Leu Gly Glu Asp Leu Gln Trp Trp Pro Asp Lys Asp Gly Thr Lys Phe Ile Cys Ile Gly Ala Leu Tyr Ser Glu Leu Leu Ala Val Ser Ser Lys Gly Glu Leu Tyr Gln Trp Lys Trp Ser Glu Ser Glu Pro Tyr Arg Asn Ala Gln Asn Pro Ser Leu His His Pro Arg Ala Thr Phe Leu Gly Leu Thr Asn Glu Lys Ile Val Leu Leu Ser Ala Asn Ser Ile Arg Ala Thr Val Ala Thr Glu Asn Asn Lys Val Ala Thr Trp Val Asp _g_ Glu Thr Leu Ser Ser Val Ala Ser Lys Leu Glu His Thr Ala Gln Thr Tyr Ser Glu Leu Gln Gly Glu Arg Ile Val Ser Leu His Cys Cys Ala Leu Tyr Thr Cys Ala Gln Leu Glu Asn 5er Leu Tyr Trp Trp Gly Val Val Pro Phe Ser Gln Arg Lys Lys Met Leu Glu Lys Ala Arg Ala Lys Asn Lys Lys Pro Lys Ser Ser Ala Gly Ile Ser Ser Met Pro Asn Ile Thr Val Gly Thr G1n Val Cys Leu Arg Asn Asn Pro Leu Tyr His Ala Gly Ala Val Ala Phe Ser Ile Ser Ala Gly Ile Pro Lys Val Gly Val Leu Met Glu Ser Val Trp Asn Met Asn Asp Ser Cys Arg Phe Gln Leu Arg Ser Pro Glu Ser Leu Lys Asn Met Glu Lys Ala Ser Lys Thr Thr Glu Ala Lys Pro Glu Ser Lys Gln Glu Pro Val Lys Thr Glu Met Gly Pro Pro Pro Ser Pro Ala Ser Thr Cys Ser Asp Ala Ser Ser Ile Ala Ser Ser Ala Ser Met Pro Tyr Lys Arg Arg Arg Ser Thr Pro Ala Pro Lys Glu Glu Glu Lys Val Asn Glu Glu Gln Trp Ser Leu Arg Glu Val Val Phe Val Glu Asp Val Lys Asn Val Pro Val Gly Lys Val Leu Lys Val Asp Gly Ala Tyr Val Ala Val Lys Phe Pro Gly Thr Ser Ser Asn Thr Asn Cys Gln Asn Ser Ser Gly Pro Asp Ala Asp Pro Ser Ser Leu Leu Gln Asp Cys Arg Leu Leu Arg Ile Asp Glu Leu Gln Val Val Lys Thr Gly Gly Thr Pro Lys Val Pro Asp Cys Phe Gln Arg Thr Pro Lys Lys Leu Cys Ile Pro Glu Lys Thr Glu Ile Leu Ala Val Asn Val Asp Ser Lys Gly Val His Ala Val Leu Lys Thr Gly Asn Trp Val Arg Tyr Cys Ile Phe Asp Leu Ala Thr Gly Lys Ala Glu Gln Glu Asn Asn Phe Pro Thr Ser Ser Ile Ala Phe Leu Gly Gln Asn Glu Arg Asn Val Ala Ile Phe Thr Ala Gly Gln Glu Ser Pro Ile 21e Leu Arg Asp Gly Asn Gly Thr Ile Tyr Pro Met Ala Lys Asp Cys Met Gly Gly Ile Arg Asp Pro Asp Trp Leu Asp Leu Pro Pro Ile Ser Ser Leu Gly Met Gly Val His Ser Leu Ile Asn Leu Pro Ala Asn Ser Thr Tle Lys Lys Lys Ala A1a Val Ile Ile Met Ala Val Glu Lys Gln Thr Leu Met Gln His Ile Leu Arg Cys Asp Tyr Glu Ala Cys Arg Gln Tyr Leu Met Asn Leu Glu Gln Ala Val Val Leu Glu Gln Asn Leu Gln Met Leu Gln Thr Phe Ile Ser His Arg Cys Asp Gly Asn Arg Asn Ile Leu His Ala Cys Val Ser Val Cys Phe Pro Thr Ser Asn Lys Glu Thr Lys Glu Glu Glu Glu Ala Glu Arg Ser Glu Arg Asn Thr Phe Ala Glu Arg Leu Ser Ala Val Glu Ala Ile Ala Asn Ala Ile Ser Val Val Ser Ser Asn Gly Pro Gly Asn Arg Ala Gly Ser Ser Ser Ser Arg Ser Leu Arg Leu Arg Glu Met Met Arg Arg Ser Leu Arg Ala Ala Gly Leu Gly Arg His Glu Ala Gly Ala Ser Ser Ser Asp His Gln Asp Pro Val Ser Pro Pro Ile Ala Pro 1010 ~ 1015 1020 Pro Ser Trp Val Pro Asp Pro Pro Ala Met Asp Pro Asp Gly Asp Ile Asp Phe Ile Leu Ala Pro Ala Va1 Gly Ser Leu Thr Thr Ala Ala Thr Gly Thr Gly Gln Gly Pro Ser Thr Ser Thr Ile Pro Gly Pro Ser Thr Glu Pro Ser Val Val Glu Ser Lys Asp Arg Lys Ala Asn Ala His Phe Ile Leu Lys Leu Leu Cys Asp Ser Va1 Val Leu Gln Pro Tyr Leu Arg Glu Leu Leu Ser Ala Lys Asp Ala Arg Gly Met Thr Pro Phe Met Ser Ala Val Ser Gly Arg Ala Tyr Pro Ala A1a Ile Thr Ile Leu Glu Thr Ala Gln Lys Ile Ala Lys Ala Glu Ile Ser Ser Ser Glu Lys Glu Glu Asp Val Phe Met Gly Met Val Cys Pro Ser Gly Thr Asn Pro Asp Asp Ser Pro Leu Tyr Val Leu Cys Cys Asn Asp Thr Cys Ser Phe Thr Trp Thr Gly Ala Glu His Ile Asn Gln Asp Ile Phe Glu Cys Arg Thr Cys Gly Leu Leu Glu Ser Leu Cys Cys Cys Thr Glu Cys Ala Arg Val Cys His Lys Gly His Asp Cys Lys Leu Lys Arg Thr Ser Pro Thr Ala Tyr Cys Asp Cys Trp Glu Lys Cys Lys Cys Lys Thr Leu Ile Ala Gly Gln Lys Ser Ala Arg Leu Asp Leu Leu Tyr Arg Leu Leu Thr Ala Thr Asn Leu Val Thr Leu Pro Asn Ser Arg Gly Glu His Leu Leu Leu Phe Leu Val Gln Thr Val Ala Arg Gln Thr Val Glu His Cys Gln Tyr Arg Pro Pro Arg Ile Arg Glu Asp Arg Asn Arg Lys Thr Ala Ser Pro Glu Asp Ser Asp Met Pro Asp His Asp Leu Glu Pro Pro Arg Phe Ala Gln Leu Ala Leu Glu Arg Val Leu Gln Asp Trp Asn Ala Leu Lys Ser Met Ile Met Phe Gly Ser Gln Glu Asn Lys Asp Pro Leu Ser Ala Ser Ser Arg Ile Gly His Leu Leu Pro Glu Glu Gln Val Tyr Leu Asn Gln Gln Ser Gly Thr Ile Arg Leu Asp Cys Phe Thr His Cys Leu Ile Val Lys Cys Thr Ala Asp Ile Leu Leu Leu Asp Thr Leu Leu Gly Thr Leu Val Lys Glu Leu Gln Asn Lys Tyr Thr Pro Gly Arg Arg Glu Glu Ala Ile Ala Val Thr Met Arg Phe Leu Arg Ser Val Ala Arg Val Phe Val Ile Leu Ser Val Glu Met Ala Ser Ser Lys Lys Lys Asn Asn Phe Ile Pro Gln Pro Ile Gly Lys Cys Lys Arg Val Phe Gln Ala Leu Leu Pro Tyr Ala Val Glu Glu Leu Cys Asn Val Ala Glu Ser Leu Ile Val Pro Val Arg Met Gly Ile Ala Arg Pro Thr Ala Pro Phe Thr Leu Ala Ser Thr Ser Ile Asp Ala Met Gln Gly Ser Glu Glu Leu Phe Ser Val Glu Pro Leu Pro Pro Arg Pro Ser Ser Asp Gln Ser Ser Ser Ser Ser Gln Ser Gln 5er Ser Tyr Ile Ile Arg Asn Pro Gln Gln Arg Arg Ile Ser Gln Ser Gln Pro Val Arg Gly Arg Asp Glu Glu Gln Asp Asp Ile Val Ser A1a Asp Val Glu Glu Val Glu Val Val Glu Gly Val Ala Gly Glu Glu Asp His His Asp Glu Gln Glu Glu His Gly Glu Glu Asn Ala Glu Ala Glu Gly Gln His Asp Glu His Asp Glu Asp Gly Ser Asp Met Glu Leu Asp Leu Leu Ala Ala Ala Glu Thr Glu Ser Asp Ser Glu Ser Asn His Ser Asn Gln Asp Asn Ala Ser Gly Arg Arg Ser Val Val Thr Ala Ala Thr Ala Gly Ser Glu Ala Gly Ala Ser Ser Val Pro Ala Phe Phe Ser Glu Asp Asp Ser Gln Ser Asn Asp Ser Ser Asp Ser Asp Ser Ser Ser Ser Gln Ser Asp Asp Ile Glu Gln Glu Thr Phe Met Leu Asp Glu Pro Leu Glu Arg Thr Thr Asn Ser Ser His Ala Asn Gly Ala Ala Gln Ala Pro Arg Ser Met Gln Trp Ala Val Arg Asn Thr Gln His Gln Arg Ala Ala Ser Thr Ala Pro Ser Ser Thr Ser Thr Pro Ala Ala Ser Ser Ala Gly Leu Ile Tyr Ile Asp Pro Ser Asn Leu Arg Arg Ser Gly Thr Ile Ser Thr Ser Ala Ala Ala Ala Ala Ala Ala Leu Glu Ala Ser Asn Ala Ser Ser Tyr Leu Thr Ser Ala Ser Ser Leu Ala Arg Ala Tyr Ser Ile Val Ile Arg Gln Ile Ser Asp Leu Met Gly Leu Ile Pro Lys Tyr Asn His Leu Val Tyr Ser Gln Tle Pro Ala Ala Val Lys Leu Thr Tyr Gln Asp Ala Val Asn Leu Gln Asn Tyr Val Glu Glu Lys Leu Ile Pro Thr Trp Asn Trp Met Val Ser Ile Met Asp Ser Thr Glu Ala Gln Leu Arg Tyr Gly Ser Ala Leu Ala Ser Ala Gly Asp Pro Gly His Pro Asn His Pro Leu His Ala Ser Gln Asn Ser Ala Arg Arg Glu Arg Met Thr Ala Arg Glu Glu Ala Ser Leu Arg Thr Leu Glu Gly Arg Arg Arg Ala Thr Leu Leu Ser Ala Arg Gln Gly Met Met Ser Ala Arg Gly Asp Phe Leu Asn Tyr Ala Leu Ser Leu Met Arg Ser His Asn Asp Glu His Ser Asp Val Leu Pro Val Leu Asp Val Cys Ser Leu Lys His Val Ala Tyr Val Phe Gln Ala Leu Tle Tyr Trp Ile Lys Ala Met Asn G1n Gln Thr Thr Leu Asp Thr Pro Gln Leu Glu Arg Lys Arg Thr Arg Glu Leu Leu Glu Leu Gly Ile Asp Asn Glu Asp Ser Glu His Glu Asn Asp Asp Asp Thr Asn Gln Ser Ala Thr Leu Asn Asp Lys Asp Asp Asp Ser Leu Pro A1a Glu Thr Gly Gln Asn His Pro Phe Phe Arg Arg Ser Asp Ser Met Thr Phe Leu Gly Cys Ile Pro Pro Asn Pro Phe Glu Val Pro Leu Ala Glu Ala Ile Pro Leu Ala Asp Gln Pro His Leu Leu Gln Pro Asn Ala Arg Lys Glu Asp Leu Phe Gly Arg Pro Ser Gln Gly Leu Tyr Ser Ser Ser Ala Ser Ser Gly Lys Cys Leu Met Glu Val Thr Val Asp Arg Asn Cys Leu Glu Val Leu Pro Thr Lys Met Ser Tyr Ala Ala Asn Leu Lys Asn Val Met Asn Met Gln Asn Arg Gln Lys Lys Glu Gly Glu Glu Gln Pro Val Leu Pro Glu Glu Thr Glu Ser Ser Lys Pro Gly Pro Ser Ala His Asp Leu Ala Ala Gln Leu Lys Ser Ser Leu Leu Ala Glu Tle Gly Leu Thr Glu Ser Glu Gly Pro Pro Leu Thr Ser Phe Arg Pro Gln Cys Ser Phe Met Gly Met Val Tle Ser His Asp Met Leu Leu Gly Arg Trp Arg Leu Ser Leu Glu Leu Phe Gly Arg Va1 Phe Met Glu Asp Val Gly Ala Glu Pro Gly Ser Ile Leu Thr Glu Leu Gly Gly Phe Glu Val Lys Glu Ser Lys Phe Arg Arg Glu Met Glu Lys Leu Arg Asn Gln Gln Ser Arg Asp Leu Ser Leu Glu Val Asp Arg Asp Arg Asp Leu Leu I1e Gln Gln Thr Met Arg Gln Leu Asn Asn His Phe Gly Arg Arg Cys Ala Thr Thr Pro Met Ala Val His Arg Val Lys Val Thr Phe Lys Asp Glu Pro Gly Glu Gly Ser Gly Val Ala Arg Ser Phe Tyr Thr Ala Ile Ala Gln Ala Phe Leu Ser Asn Glu Lys Leu Pro Asn Leu Glu Cys Ile Gln Asn Ala Asn Lys Gly Thr His Thr Ser Leu Met Gln Arg Leu Arg Asn Arg Gly Glu Arg Asp Arg Glu Arg Glu Arg Glu Arg Glu Met Arg Arg Ser Ser Gly Leu Arg Ala Gly Ser Arg Arg Asp Arg Asp Arg Asp Phe Arg Arg Gln Leu Ser Ile Asp Thr Arg Pro Phe Arg Pro Ala Ser Glu Gly Asn Pro Ser Asp Asp Pro Glu Pro Leu Pro Ala His Arg Gln Ala Leu Gly Glu Arg Leu Tyr Pro Arg Val Gln Ala Met Gln Pro Ala Phe Ala Ser Lys Ile Thr G1y Met Leu Leu Glu Leu Ser Pro Ala Gln Leu Leu Leu Leu Leu Ala Ser Glu Asp Ser Leu Arg Ala Arg Val Asp Glu Ala Met Glu Leu Ile Ile Ala His Gly Arg Glu Asn Gly Ala Asp Ser Ile Leu Asp Leu Gly Leu Val Asp 5er Ser Glu Lys Val Gln Gln Glu Asn Arg Lys Arg His Gly Ser Ser Arg Ser Val Val Asp Met Asp Leu Asp Asp Thr Asp Asp Gly Asp Asp Asn Ala Pro Leu Phe Tyr Gln Pro Gly Lys Arg Gly Phe Tyr Thr Pro Arg Pro Gly Lys Asn Thr Glu Ala Arg Leu Asn Cys Phe Arg Asn Tle Gly Arg Ile Leu Gly Leu Cys Leu Leu Gln Asn Glu Leu Cys Pro Ile Thr Leu Asn Arg His Val Ile Lys Val Leu Leu Gly Arg Lys Val Asn Trp His Asp Phe Ala Phe Phe Asp Pro Val Met Tyr Glu Ser Leu Arg Gln Leu Ile Leu Ala Ser Gln Ser Ser Asp Ala Asp Ala Val Phe Ser Ala Met Asp Leu Ala Phe Ala Ile Asp Leu Cys Lys Glu Glu Gly Gly Gly Gln Val Glu Leu Ile Pro Asn Gly Val Asn Ile Pro Val Thr Pro Gln Asn Val Tyr Glu Tyr Val Arg Lys Tyr Ala G1u His Arg Met Leu Val Val Ala Glu Gln Pro Leu His Ala Met Arg Lys Gly Leu Leu Asp Val Leu Pro Lys Asn Ser Leu Glu Asp Leu Thr Ala Glu Asp Phe Arg Leu Leu Val Asn Gly Cys Gly Glu Val Asn Val Gln Met Leu Tle Ser Phe Thr Ser Phe Asn Asp Glu Ser Gly Glu Asn Ala Glu Lys Leu Leu Gln Phe Lys Arg Trp Phe Trp Ser Ile Val Glu Lys Met Ser Met Thr Glu Arg Gln Asp Leu Val Tyr Phe Trp Thr Ser Ser Pro Ser Leu Pro Ala Ser Glu Glu Gly Phe Gln Pro Met Pro Ser Ile Thr Ile Arg Pro Pro Asp Asp Gln His Leu Pro Thr Ala Asn Thr Cys Ile Ser Arg Leu Tyr Val Pro Leu Tyr Ser Ser Lys Gln Ile Leu Lys Gln Lys Leu Leu Leu Ala Ile Lys Thr Lys Asn Phe Gly Phe Val <210> 3 <21l> 8491 <212> DNA
<2l3> human Edd cDNA splice variant <220>
<221> CDS
<222> (34)..(8412) <223>
<400>

cgccctcgag acc tccatccatttcgtg 54 tggaggacga atg gaaggaaagc acg MetThrSerIleHisPheVal gttcacccgctgccgggcaccgaggaccagctcaatgacaggttacga 102 ValHisProLeuProGlyThrGluAspGlnLeuAsnAspArgLeuArg gaagtttctgagaagctgaacaaatataatttaaacagccacccccct l50 GluValSerGluLysLeuAsnLysTyrAsnLeuAsnSerHisProPro ttgaatgtattggaacaggetactattaaacagtgtgtggtgggacca 198 LeuAsnValLeuGluGlnAlaThrIleLysGlnCysValValG1yPro aatcatgetgcctttcttcttgaggatggtagagtttgcaggattggt 246 AsnHisAlaAlaPheLeuLeuGluAspGlyArgValCysArgIleGly ttttcagtacagccagacagattggaattgggtaaacctgataataat 294 PheSerValGlnProAspArgLeuGluLeuGlyLysProAspAsnAsn gatgggtcaaagttgaacagcaactcgggggcagggaggacgtcaagg 342 AspGlySerLysLeuAsnSerAsnSerGlyAlaGlyArgThrSerArg cctggtaggacaagcgactctccatggtttctctcaggttctgagact 390 ProGlyArgThrSerAspSerProTrpPheLeu5erGlySerGluThr ctaggcaggctggcaggcaacaccttaggaagccgctggagttctgga 438 LeuGlyArgLeuAlaGlyAsnThrLeuGlySerArgTrpSerSerGly gtgggtggaagtggtggaggatcctctggtaggtcatcagetggaget 486 ValGlyGlySerGlyGlyGlySerSerGlyArgSerSerAlaGlyAla cgagattcccgccggcagactcgagttattcggacaggacgggatcga 534 ArgAspSerArgArgGlnThrArgValIleArgThrGlyArgAspArg gggtctgggcttttgggcagtcagccccagccagttattccagcatct 582 GlySerGlyLeuLeuGlySerGlnProGlnProValIleProA1aSer gtcattccagaggagctgatttcacaggcccaagttgttttacaaggc 630 ValIleProGluGluLeuIleSerGlnAlaGlnValValLeuGlnGly aaatccagaagtgtcattattcgagaacttcagagaacaaatcttgat 678 LysSerArgSerValIleIleArgGluLeuGlnArgThrAsnLeuAsp gtgaaccttgetgtaaataatttacttagccgggatgatgaagatgga 726 ValAsnLeuAlaValAsnAsnLeuLeuSerArgAspAspGluAspGly gatgatggggatgatacagccagcgaatcttatttgcctggagaggat 774 AspAspGlyAspAspThrAlaSerGluSerTyrLeuProGlyGluAsp cttatgtctctccttgatgccgacattcattctgcccacccaagtgtc 822 LeuMetSerLeuLeuAspAlaAsp21eHisSerAlaHisProSerVal attattgatgcagatgccatgttttctgaagacattagctattttggt 870 IleIleAspA1aAspAlaMetPheSerGluAspIleSerTyrPheGly taccct.tcttttcgtcgttcatcactttccaggctaggctcatctcga 918 TyrProSerPheArgArgSerSerLeuSerArgLeuGlySerSerArg gagagagactctgagctgttgcgtgaacgtgaatccgttttacgttta 966 GluArgAspSerGluLeuLeuArgGluArgGluSerValLeuArgLeu cgtgaacgaaggtggcttgatggagcctcatttgataatgaaaggggt 1014 ArgGluArgArgTrpLeuAspGlyAlaSerPheAspAsnGluArgGly tctaccagcaaggaaggagagccaaacttggataagaagaatacacct 1062 SerThrSerLysGluGlyGluProAsnLeuAspLysLysAsnThrPro gttcaaagtccagtatctctaggagaagatttgcagtggtggcctgat 1110 ValGlnSerProValSerLeuGlyGluAspLeuGlnTrpTrpProAsp aaggatggaacaaaattcatctgtattggggetctgtattctgaactt 1158 LysAspGlyThrLysPheIleCysIleGlyAlaLeuTyrSerGluLeu ctggetgtcagcagtaaaggagaactttatcagtggaaatggagtgaa 1206 LeuAlaValSerSerLysGlyGluLeuTyrGlnTrpLysTrpSerGlu tctgagccttacagaaatgcccagaatccttcattacatcatccacga 1254 SerGluProTyrArgAsnAlaGlnAsnProSerLeuHisHisProArg gcaacatttttggggttaaccaatgaaaagatagtcctcctgtctgca 1302 AlaThrPheLeuGlyLeuThrAsnGluLysIleValLeuLeuSerAla aatagcataagagcaactgtagetacagaaaataacaaggttgetaca 1350 AsnSerIleArgAlaThrValAlaThrGluAsnAsnLysValAlaThr tgggtggatgaaactttaagttctgtggettctaaattagagcacact 1398 TrpValAspGluThrLeuSer5erValAlaSirLysLeuGluHisThr getcagacttactctgaacttcaaggagagcggatagtttctttacat 1446 AlaGlnThrTyrSerGluLeuGlnGlyGluArgIleValSerLeuHis tgctgtgccctttacacctgcgetcagctggaaaacagtttatattgg 1494 CysCysAlaLeuTyrThrCysAlaGlnLeuGluAsnSerLeuTyrTrp tggggtgtagttccttttagtcaaaggaagaaaatgttagagaaaget 1542 TrpGlyValValProPheSerGlnArgLysLysMetLeuGluLysAla agagcaaaaaataaaaagcctaaatccagtgetggtatttcttcaatg 1590 ArgAlaLysAsnLysLysProLysSerSerAlaGlyTleSerSerMet ccgaacatcactgttggtacccaggtatgcttgagaaataatcctctt 1638 ProAsnIleThrValGlyThrGlnValCysLeuArgAsnAsnProLeu tatcatgetggagcagttgcattttcaattagtgetgggattcctaaa 1686 TyrHisAlaGlyAlaValAlaPheSerIleSerAlaGlyIleProLys gttggtgtcttaatggagtcagtttggaatatgaatgacagctgtaga 1734 ValGlyValLeuMetGluSerValTrpAsnMetAsnAspSerCysArg tttcaacttagatctcctgaaagcttgaaaaacatggaaaaagetagc 1782 PheGlnLeuArgSerProGluSerLeuLysAsnMetGluLysAlaSer aaaactactgaagetaagcctgaaagtaagcaggagccagtgaaaaca 1830 LysThrThrGluAlaLysProGluSerLysGlnGluProValLysThr gaaatgggtcctccaccatctccagcatccacgtgtagtgatgcatcc 1878 GluMetGlyProProProSerProAlaSerThrCysSerAspAlaSer tcaattgccagcagtgcatcaatgccatacaaacgacgacggtcaacc 1926 5erIleAlaSerSerAlaSerMetProTyrLysArgArgArgSerThr cctgcaccaaaagaagaggaaaaggtgaatgaagagcagtggtctctt 1974 ProAlaProLysGluGluGluLysValAsnGluGluGlnTrpSerLeu cgggaagtggtttttgtggaagatgtcaagaatgttcctgttggcaag 2022 ArgGluValValPheValGluAspValLysAsnValProValGlyLys gtgctaaaagtagatggtgcctatgttgetgtaaaatttccaggaacc 2070 ValLeuLysValAspGlyAlaTyrValAlaValLysPheProGlyThr tccagtaatactaactgtcagaacagctctggtccagatgetgaccct 2118 SerSerAsnThrAsnCysGlnAsnSerSerGlyProAspAlaAspPro tcttctctcctgcaggattgtaggttacttagaattgatgaattgcag 2166 SerSerLeuLeuGlnAspCysArgLeuLeuArgIleAspGluLeuGln gttgtcaaaactggtggaacaccgaaggttcccgactgtttccaaagg 2214 ValValLysThrGlyGlyThrProLysValProAspCysPheGlnArg actcctaaaaagctttgtatacctgaaaaaacagaaatattagcagtg 2262 ThrProLysLysLeuCysIleProGluLysThrGluIleLeuAlaVal aatgtagattccaaaggtgttcatgetgttctgaagactggaaattgg 2310 AsnValAspSerLysGlyValHisAlaVa1LeuLysThrGlyAsnTrp gtgcgatactgtatctttgatcttgetacaggaaaagcagaacaggaa 2358 ValArgTyrCys21ePheAspLeuAlaThrGlyLysAlaGluGlnGlu aataattttcctacaagcagcattgetttccttggtcagaatgagagg 2406 AsnAsnPheProThrSerSerIleAlaPheLeuGlyGlnAsnGluArg aatgtagccattttcactgetggacaggaatctcccattattcttcga 2454 AsnValAlaIlePheThrAlaGlyGlnGluSerProTleIleLeuArg gatggaaatggtaccatctacccaatggccaaagattgcatgggagga 2502 AspGlyAsnGlyThrIleTyrProMetAlaLysAspCysMetGlyGly ataagggatcccgattggctggatcttccacctattagtagtcttgga 2550 IleArgAspProAspTrpLeuAspLeuProProTleSerSerLeuGly atgggtgtgcattctttaataaatcttcctgccaattcaacaatcaaa 2598 MetGlyValHisSerLeuIleAsnLeuProAlaAsnSerThrIleLys aagaaagetgetgttatcatcatggetgtagagaaacaaaccttaatg 2646 LysLysAlaAlaValIleIleMetAlaVa1GluLysGlnThrLeuMet caa cac att ctg cgc tgt gac tat gag gcc tgt cga caa tat cta atg 2694 GlnHisIleLeuArgCys Asp Tyr Glu Ala Cys Arg Met Gln Tyr Leu aatcttgagcaagcggtt gtt tta gag cag aat cta cag 2742 cag atg ctg AsnLeuGluGlnAlaVal Val Leu Glu Gln Asn Leu Gln Gln Met Leu acattcatcagccacaga tgt gat gga aat cga aat get 2790 att ttg cat ThrPheIleSerHisArg Cys Asp Gly Asn Arg Asn Ala Ile Leu His tgtgtatcagtttgcttt cca acc agc aat aaa gaa gaa 2838 act aaa gaa CysValSerValCysPhe Pro Thr Ser Asn Lys Glu Glu Thr Lys Glu gaggaagcggagcgttct gaa aga aat aca ttt gca tct 2886 gaa agg ctt GluGluAlaGluArgSer Glu Arg Asn Thr Phe Ala Ser Glu Arg Leu getgttgaggccattgca aat gca ata tca gtt gtt ggc 2934 tca agt aat AlaValGluAlaIleAla Asn Ala Ile Ser Val Val Gly Ser Ser Asn ccaggtaatcgggetgga tca tca agt agc cga agt cgg 2982 ttg aga tta ProGlyAsnArgAlaGly Ser Ser Ser Ser Arg Ser Arg Leu Arg Leu gaaatgatgagacgttcg ttg aga gca get ggt ttg gaa 3030 ggt aga cat GluMetMetArgArgSer Leu Arg Ala Ala Gly Leu Glu Gly Arg His getggagettcatccagt gac cac cag gat cca gtt ccc 3075 tca ccc AlaGlyAlaSerSerSer Asp His Gln Asp Pro Val Pro Ser Pro atagetccccctagttgg gtt cct gac cct cct gcg cct 3120 atg gat IleAlaProProSerTrp Val Pro Asp Pro Pro Ala Pro Met Asp gatggtgacattgatttt atc ctg gcc ccc get gtg ctt 3165 gga tct AspG1yAspIleAspPhe Ile Leu Ala Pro Ala Val Leu Gly Ser accacagcagcaaccggt act ggt caa gga cca agc act 3210 acc tcc ThrThrAlaAlaThrGly Thr Gly Gln Gly Pro Ser Thr Thr Ser attccaggtccttccaca gag cca tct gta gta gaa gat 3255 tcc aag IleProGlyProSerThr Glu Pro Ser Val Val Glu Asp 5er Lys cgaaaggcgaatgetcat ttt ata ttg aaa ttg tta agt 3300 tgt gac ArgLysAlaAsnAlaHis Phe Ile Leu Lys Leu Leu Ser Cys Asp gtggttctccagccctat cta cga gaa ctt ctt tct gat 3345 gcc aag ValValLeuGlnProTyr Leu Arg Glu Leu Leu Ser Asp Ala Lys gcaagagggatgacccca ttt atg tca get gta agt get 3390 ggc cga AlaArgGlyMetThrPro Phe Met Ser Ala Val Ser Ala Gly Arg tat cctgetgcaattacc atcttagaa actgetcagaaaattgca 3435 Tyr ProAlaAlaIleThr IleLeuGlu ThrAlaGlnLysIleAla aaa getgaaatatcctca agtgaaaaa gaggaagatgtattcatg 3480 Lys AlaGluIleSerSer SerGluLys GluGluAspValPheMet gga atggtttgcccatca ggtaccaac cctgatgactctccttta 3525 Gly MetValCysProSer GlyThrAsn ProAspAspSerProLeu tat gttttatgttgtaat gacacttgc agttttacatggactgga 3570 Tyr ValLeuCysCysAsn AspThrCys SerPheThrTrpThrGly gca gagcacattaaccag gatattttt gagtgtcgaacttgtggc 3615 Ala GluHisIleAsnGln AspIlePhe GluCysArgThrCysGly ttg ctggagtcactgtgt tgttgtacg gaatgtgcaagggtttgt 3660 Leu LeuGluSerLeuCys CysCysThr GluCysAlaArgValCys cat aaaggtcatgattgc aaactcaaa cggacatcaccaacagcc 3705 His LysGlyHisAspCys LysLeuLys ArgThrSerProThrAla tac tgtgattgttgggag aaatgtaaa tgtaaaactcttattget 3750 Tyr CysAspCysTrpGlu LysCysLys CysLysThrLeuIleAla gga cagaaatctgetcgt cttgatcta ctttatcgcctgctcact 3795 Gly GlnLysSerAlaArg LeuAspLeu LeuTyrArgLeuLeuThr get actaatctggttact ctgccaaac agcaggggagagcacctc 3840 Ala ThrAsnLeuValThr LeuProAsn SerArgGlyGluHisLeu tta ctattcttagtacag acagtcgca aggcagacggtggagcat 3885 Leu LeuPheLeuValGln ThrValAla ArgGlnThrValGluHis tgt caatacaggccacct cgaatcagg gaagatcgtaaccgaaaa 3930 Cys GlnTyrArgProPro ArgI1eArg GluAspArgAsnArgLys aca gccagtcctgaagat tcagatatg ccagatcatgatttagag 3975 Thr AlaSerProGluAsp SerAspMet ProAspHisAspLeuGlu cct ccaagatttgcccag cttgcattg gagcgtgttctacaggac 4020 Pro ProArgPheAlaGln LeuAlaLeu GluArgValLeuGlnAsp tgg aatgccttgaaatct atgattatg tttgggtcgcaggagaat 4065 Trp AsnAlaLeuLysSer MetIleMet PheGlySerGlnGluAsn aaagaccctcttagtgcc agcagtagaataggccatcttttgcca 4110 LysAspProLeuSerAla SerSerArgIleGlyHisLeuLeuPro gaagagcaagtatacctc aatcagcaaagtggcacaattc,ggctg 4155 GluGluGlnValTyrLeu AsnGlnGlnSerGlyThrIleArgLeu gactgtttcactcattgc cttatagttaagtgtacagcagatatt 4200 AspCysPheThrHisCys LeuIleValLysCysThrAlaAspIle ttgcttttagatactcta ctaggtacactagtgaaagaactccaa 4245 LeuLeuLeuAspThrLeu LeuGlyThrLeuValLysGluLeuGln aacaaatatacacctgga cgtagagaagaagetattgetgtgaca 4290 AsnLysTyrThrProGly ArgArgGluGluAlaIleAlaValThr atgaggtttctacgttca gtggcaagagtttttgttattctgagt 4335 MetArgPheLeuArgSer ValAlaArgValPheValIleLeuSer gtggaaatggettcatcc aaaaagaaaaacaactttattccacag 4380 ValGluMetAlaSerSer LysLysLysAsnAsnPheIleProGln ccaattggaaaatgcaag cgtgtattccaagcattgctaccttac 4425 ProIleGlyLysCysLys ArgValPheGlnAlaLeuLeuProTyr getgtggaagaattgtgc aacgtagcagagtcactgattgttcct 4470 AlaValGluGluLeuCys AsnValAlaGluSerLeuIleValPro gtcagaatggggattget cgtccaactgcaccatttaccctgget 4515 ValArgMetGlyIleAla ArgProThrAlaProPheThrLeuAla agtactagcatagatgcc atgcagggcagtgaagaattattttca 4560 SerThrSerIleAspAla MetGlnGlySerG1uGluLeuPheSer gtggaaccactaccacca cgaccatcatctgatcagtctagcagc 4605 ValGluProLeuProPro ArgProSerSerAspGlnSerSerSer tccagtcagtctcagtca tcctacatcatcaggaatccacagcag 4650 SerSerGlnSerGlnSer SerTyrIleIleArgAsnProGlnGln aggcgcatcagccagtca cagcccgttcggggcagagatgaagaa 4695 ArgArgIleSerGlnSer GlnProValArgGlyArgAspGluG1u caggatgatattgtttca gcagatgtggaagaggttgaggtggtg 4740 GlnAspAspIleValSer AlaAspValGluGluValGluValVal gagggtgtggetggagaa gaggat catcatgatgaacaggaagaa 4785 GluGlyValAlaGlyGlu GluAsp HisHisAspGluGlnGluGlu cacggggaagaaaatget gaggca gagggacaacatgatgagcat 4830 HisGlyGluGluAsnAla GluAla GluGlyGlnHisAspGluHis gatgaagacgggagtgat atggag ctggacttgttagcagcaget 4875 AspGluAspGlySerAsp MetGlu LeuAspLeuLeuAlaAlaAla gaaacagaaagtgatagt gaaagt aaccacagcaaccaagataat 4920 GluThrGluSerAspSer GluSer AsnHisSerAsnGlnAspAsn getagtgggcgcagaagc gttgtc actgcagcaactgetggttca 4965 AlaSerGlyArgArg5er ValVal ThrAlaAlaThrAlaGlySer gaagcaggagcaagcagt gttcct gccttcttttctgaagatgat 5010 GluAlaGlyAlaSerSer ValPro AlaPhePheSerGluAspAsp tctcaatcgaatgactca agtgat tctgatagcagtagtagtcag 5055 SerGlnSerAsnAspSer SerAsp SerAspSerSerSerSerGln agtgacgacatagaacag gagacc tttatgcttgatgagccatta 5100 SerAspAspIleGluGln GluThr PheMetLeuAspGluProLeu gaaagaaccacaaatagc tcccat gccaatggtgetgcccaaget 5145 GluArgThrThrAsnSer SerHis AlaAsnGlyAlaAlaGlnAla ccccgttcaatgcagtgg getgtc cgcaacacccagcatcagcga 5190 ProArgSerMetGlnTrp AlaVal ArgAsnThrGlnHisGlnArg gcagccagtacagcccct tccagt acatctacaccagcagcaagt 5235 AlaAlaSerThrAlaPro SerSer ThrSerThrProA1aAlaSer tcagcgggtttgatttat attgat ccttcaaacttacgccggagt 5280 SerAlaGlyLeuIleTyr IleAsp ProSerAsnLeuArgArgSer ggtaccatcagtacaagt getgca getgcagcagetgetttggaa 5325 GlyThrIleSerThrSer AlaAla AlaAlaAlaAlaAlaLeuGlu getagcaacgccagcagt taccta acatctgcaagcagtttagcc 5370 AlaSerAsnAlaSerSer TyrLeu ThrSerAlaSerSerLeuAla agggettacagcattgtc attaga caaatctcggacttgatgggo 5415 ArgAlaTyrSerIleVal IleArg GlnIle5erAspLeuMetGly cttattcctaagtataat caccta gtatactctcagattccagca 5460 LeuIleProLysTyrAsn HisLeuValTyrSerGlnIleProAla getgtgaaattgacttac caagatgcagtaaacttacagaactat 5505 AlaValLysLeuThrTyr GlnAspAlaValAsnLeuGlnAsnTyr gtagaagaaaagcttatt cccacttggaactggatggtcagtatt 5550 ValGluGluLysLeuIle ProThrTrpAsnTrpMetValSerIle atggattctactgaaget caattacgttatggttctgcattagca 5595 MetAspSerThrGluAla GlnLeuArgTyrGlySerAlaLeuAla tctgetggtgatcctgga catccaaatcatcetcttcacgettct 5640 SerAlaGlyAspProGly HisProAsnHisProLeuHisAlaSer cagaattcagcgagaaga gagaggatgactgcgcgagaagaaget 5685 GlnAsnSerAlaArgArg GluArgMetThrAlaArgGluGluAla agcttacgaacacttgaa ggcagacgacgtgccaccttgcttagc 5730 SerLeuArgThrLeuGlu GlyArgArgArgAlaThrLeuLeuSer gcccgtcaaggaatgatg tctgcacgaggagacttcctaaattat 5775 AlaArgGlnGlyMetMet SerAlaArgGlyAspPheLeuAsnTyr getctgtctctaatgcgg tctcataatgatgagcattctgatgtt 5820 AlaLeuSerLeuMetArg SerHisAsnAspG1uHisSerAspVal cttccagttttggatgtt tgctcattgaagcatgtggcatatgtt 5865 LeuProValLeuAspVal CysSerLeuLysHisValAlaTyrVal tttcaagcacttatatac tggattaaggcaatgaatcagcagaca 5910 PheGlnAlaLeuIleTyr TrpIleLysAlaMetAsnGlnGlnThr acattggatacacctcaa ctagaacgcaaaaggacgcgagaactc 5955 ThrLeuAspThrProGln LeuGluArgLysArgThrArgGluLeu ttggaactgggtattgat aatgaagattcagaacatgaaaatgat 6000 LeuGluLeuGlyIleAsp AsnGluAspSerGluHisGluAsnAsp gatgacaccaatcaaagt getactttgaatgataaggatgatgac 6045 AspAspThrAsnGlnSer AlaThrLeuAsnAspLysAspAspAsp tctcttcctgcagaaact ggccaaaaccatccatttttccgacgt 6090 SerLeuProAlaGluThr GlyGlnAsnHisProPhePheArgArg tcagactccatgacattc cttgggtgtatacccccaaatccattt 6135 SerAspSerMetThrPhe LeuGlyCysIleProProAsnProPhe gaagtgcctctggetgaa gccatccccttggetgatcagccacat 6180 GluValProLeuAlaGlu AlaIleProLeuAlaAspGlnProHis ctgttgcagccaaatget agaaaggaggatctttttggccgtcca 6225 LeuLeuGlnProAsnAla ArgLysGluAspLeuPheGlyArgPro agtcagggtctttattct tcatctgccagtagtgggaaatgttta 6270 SerGlnGlyLeuTyrSer SerSerAlaSerSerGlyLysCysLeu atggaggttacagtggat agaaactgcctagaggttcttccaaca 6315 MetGluValThrValAsp ArgAsnCysLeuGluValLeuProThr aaaatgtcttatgetgcc aatctgaaaaatgtaatgaacatgcaa 6360 LysMetSerTyrAlaAla AsnLeuLysAsnValMetAsnMetGln aaccggcaaaaaaaagaa ggggaagaacagcccgtgctgccagaa 6405 AsnArgGlnLysLysGlu GlyGluGluGlnProValLeuProGlu gaaactgagagttcaaaa ccagggccatctgetcatgatcttget 6450 GluThrGluSerSerLys ProGlyProSerAlaHisAspLeuA1a gcacaattaaaaagtagc ttactagcagaaataggacttactgaa 6495 AlaGlnLeuLysSerSer LeuLeuAlaGluIleGlyLeuThrGlu agtgaagggccacctctc acatctttcaggccacagtgtagcttt 6540 SerGluGlyProProLeu ThrSerPheArgProGlnCysSerPhe atgggaatggttatttcc catgatatgctgctaggacgttggcgc 6585 MetGlyMetValIleSer HisAspMetLeuLeuGlyArgTrpArg ctttctttagaactgttc ggcagggtattcatggaagatgttgga 6630 LeuSerLeuG1uLeuPhe GlyArgValPheMetGluAspValGly gcagaacctggatcaatc ctaactgaattgggtggttttgaggta 6675 AlaGluProGlySerTle LeuThrGluLeuGlyGlyPheGluVal aaagaatcaaaattccgc agagaaatggaaaaactgagaaaccag 6720 LysGluSerLysPheArg ArgGluMetGluLysLeuArgAsnGln cagtcaagagatttgtca ctagaggttgatcgggatcgagatctt 6765 GlnSerArgAspLeuSer LeuGluValAspArgAspArgAspLeu ctcattcagcagactatg aggcagcttaacaatcactttggtcga 6810 LeuIleGlnGlnThrMet ArgGlnLeuAsnAsnHisPheGlyArg _ ~8 _ agatgtgetactacacca atggetgtacacagagtaaaagtcaca 6855 ArgCysAlaThrThrPro MetAlaValHisArgValLysValThr tttaaggatgagccagga gagggcagtggtgtagcacgaagtttt 6900 PheLysAspGluProGly GluGlySerGlyValAlaArgSerPhe tatacagccattgcacaa gcatttttatcaaatgaaaaattgcca 6945 TyrThrAlaIleAlaGln AlaPheLeuSerAsnGluLysLeuPro aatctagagtgtatccaa aatgccaacaaaggcacccacacaagt 6990 AsnLeuGluCysIleGln AsnAlaAsnLysGlyThrHisThrSer ttaatgcagagattaagg aaccgaggagagagagaccgggaaagg 7035 LeuMetGlnArgLeuArg AsnArgGlyGluArgAspArgGluArg gagagagaaagggaaatg aggaggagtagtggtttgcgagcaggt 7080 GluArgGluArgGluMet ArgArgSerSerGlyLeuArgAlaGly tctcggagggaccgggat agagactttagaagacagctttccatc 7125 SerArgArgAspArgAsp ArgAspPheArgArgGlnLeuSerIle gacactaggccctttaga ccagcctctgaagggaatcctagcgat 7170 AspThrArgProPheArg ProAlaSerGluGlyAsnProSerAsp gatcctgagcctttgcca gcacatcggcaggcacttggagagagg 7215 AspProGluProLeuPro AlaHisArgGlnAlaLeuGlyGluArg ctttatcctcgtgtacaa gcaatgcaaccagcatttgcaagtaaa 7260 LeuTyrProArgValGln AlaMetGlnProAlaPheAlaSerLys atcactggcatgttgttg gaattatccccagetcagctgcttctc 7305 IleThrGlyMetLeuLeu GluLeuSerProAlaGlnLeuLeuLeu cttctagcaagtgaggat tctctgagagcaagagtggatgaggcc 7350 LeuLeuAlaSerG1uAsp SerLeuArgAlaArgValAspGluAla atggaactcattattgca catggacgggaaaatggagetgatagt 7395 MetGluLeuIleIleAla HisGlyArgGluAsnGlyAlaAspSer atcctggatcttggatta gtagactcctcagaaaaggtacagcag 7440 IleLeuAspLeuGlyLeu ValAspSerSerGluLysValGlnGln gaaaaccgaaagcgccat ggctctagtcgaagtgtagtagatatg 7485 GluAsnArgLysArgHis GlySerSerArgSerValValAspMet gatttagatgatacagat gatggtgatgacaatgcccctttgttt 7530 AspLeuAspAspThrAsp AspGlyAspAspAsnAlaProLeuPhe taccaacctgggaaaaga ggattttatactccaaggcctggcaag 7575 TyrGlnProGlyLysArg GlyPheTyrThrProArgProGlyLys aacacagaagcaaggttg aattgtttcagaaacattggcaggatt 7620 AsnThrGluAlaArgLeu AsnCysPheArgAsnIleGlyArgIle cttggactatgtctgtta cagaatgaactctgtcctatcacattg 7665 LeuGlyLeuCysLeuLeu GlnAsnG1uLeuCysProIleThrLeu aatagacatgtaattaaa gtattgcttggtagaaaagtcaattgg 7710 AsnArgHisValIleLys ValLeuLeuGlyArgLysValAsnTrp catgattttgettttttt gatcctgtaatgtatgagagtttgcgg 7755 HisAspPheAlaPhePhe AspProValMetTyrGluSerLeuArg caactaatcctcgcgtct cagagttcagatgetgatgetgttttc 7800 GlnLeuIleLeuAlaSer GlnSerSerAspAlaAspAlaValPhe tcagcaatggatttggca tttgcaattgacctgtgtaaagaagaa 7845 SerAlaMetAspLeuAla PheAlaIleAspLeuCysLysGluGlu ggtggaggacaggttgaa ctcattcctaatggtgtaaatatacca 7890 GlyGlyGlyGlnValGlu LeuIleProAsnGlyValAsnIlePro gtcactccacagaatgta tatgagtatgtgcggaaatacgcagaa 7935 ValThrProGlnAsnVal TyrGluTyrValArgLysTyrAlaGlu cacagaatgttggtagtt gcagaacagcccttacatgcaatgagg 7980 HisArgMetLeuValVal AlaGluGlnProLeuHisAlaMetArg aaaggtctactagatgtg cttccaaaaaattcattagaagattta 8025 LysGlyLeuLeuAspVal LeuProLysAsnSerLeuGluAspLeu acggcagaagattttagg cttttggtaaatggctgcggtgaagtc 8070 ThrAlaGluAspPheArg LeuLeuValAsnGlyCysGlyGluVal aatgtgcaaatgctgatc agttttacctctttcaatgatgaatca 8115 AsnValGlnMetLeuIle SerPheThrSerPheAsnAspGluSer ggagaaaatgetgagaag cttctgcagttcaagcgttggttctgg 8160 GlyGluAsnAlaGluLys LeuLeuGlnPheLysArgTrpPheTrp tca ata gta gag aag atg agc atg aca gaa cga caa gat ctt gtt 8205 SerIleValGluLysMet SerMetThr GluArgGlnAspLeu Val tacttttggacatcaagc ccatcactg ccagccagtgaagaa gga 8250 TyrPheTrpThrSerSer ProSerLeu ProAlaSerGluGlu Gly ttccagcctatgccctca atcacaata agaccaccagatgac caa 8295 PheGlnProMetProSer IleThrIle ArgProProAspAsp Gln catcttcctactgcaaat acttgcatt tctcgactttacgtc cca 8340 HisLeuProThrAlaAsn ThrCysIle SerArgLeuTyrVal Pro ctctattcctctaaacag attctcaaa cagaaattgttactc gcc 8385 LeuTyrSerSerLysGln IleLeuLys GlnLysLeuLeuLeu Ala attaagaccaagaatttt ggttttgtg tagagtataa 8432 aaagtgtgta IleLysThrLysAsnPhe GlyPheVal ttgctgtgta tgtagattttttt ataaaagtt 8491 atattactag ccatttgtct caaattt <210>

<211> 93 <212>
PRT

<213> EddcDNA variant human splice <400> 4 Met Thr Ser Ile His Phe Val Val His Pro Leu Pro Gly Thr Glu Asp 1 5 10 l5 Gln Leu Asn Asp Arg Leu Arg Glu Val Ser Glu Lys Leu Asn Lys Tyr Asn Leu Asn Ser His Pro Pro Leu Asn Val Leu Glu Gln Ala Thr Ile Lys Gln Cys Val Val Gly Pro Asn His Ala Ala Phe Leu Leu Glu Asp Gly Arg Val Cys Arg Ile Gly Phe Ser Val Gln Pro Asp Arg Leu Glu Leu Gly Lys Pro Asp Asn Asn Asp Gly Ser Lys Leu Asn Ser Asn Ser Gly Ala Gly Arg Thr Ser Arg Pro Gly Arg Thr Ser Asp Ser Pro Trp Phe Leu Ser Gly Ser Glu Thr Leu Gly Arg Leu Ala Gly Asn Thr Leu Gly Ser Arg Trp Ser Ser Gly Val Gly Gly Ser Gly Gly Gly Ser Ser Gly Arg 5er Ser Ala Gly Ala Arg Asp Ser Arg Arg Gln Thr Arg Val Ile Arg Thr Gly Arg Asp Arg Gly Ser Gly Leu Leu Gly Ser Gln Pro Gln Pro Val Tle Pro Ala Ser Val Ile Pro Glu Glu Leu Ile Ser Gln Ala Gln Val Val Leu Gln Gly Lys Ser Arg Ser Val Ile Ile Arg Glu Leu Gln Arg Thr Asn Leu Asp Val Asn Leu Ala Val Asn Asn Leu Leu Ser Arg Asp Asp Glu Asp Gly Asp Asp Gly Asp Asp Thr Ala Ser Glu Ser Tyr Leu Pro Gly Glu Asp Leu Met Ser Leu Leu Asp Ala Asp Ile His Ser Ala His Pro Ser Val Ile Ile Asp Ala Asp Ala Met Phe Ser Glu Asp Ile Ser Tyr Phe Gly Tyr Pro Ser Phe Arg Arg Ser Ser Leu Ser Arg Leu Gly Ser Ser Arg Glu Arg Asp Ser Glu Leu Leu Arg Glu Arg Glu Ser Val Leu Arg Leu Arg Glu Arg Arg Trp Leu Asp Gly Ala Ser Phe Asp Asn Glu Arg Gly Ser Thr Ser Lys Glu Gly Glu Pro Asn Leu Asp Lys Lys Asn Thr Pro Va1 Gln Ser Pro Va1 Ser Leu Gly Glu Asp Leu Gln Trp Trp Pro Asp Lys Asp Gly Thr Lys Phe Ile Cys Ile Gly Ala Leu Tyr Ser Glu Leu Leu Ala Val Ser Ser Lys Gly Glu Leu Tyr Gln Trp Lys Trp Ser Glu Ser Glu Pro Tyr Arg Asn Ala Gln Asn Pro Ser Leu His His Pro Arg Ala Thr Phe Leu Gly Leu Thr Asn Glu Lys Ile Val Leu Leu Ser Ala Asn Ser Ile Arg Ala Thr Val Ala Thr Glu Asn Asn Lys Val Ala Thr Trp Val Asp Glu Thr Leu Ser Ser Val Ala Ser Lys Leu Glu His Thr Ala Gln Thr Tyr Ser Glu Leu Gln Gly Glu Arg Ile Val Ser Leu His Cys Cys Ala Leu Tyr Thr Cys Ala Gln Leu Glu Asn Ser Leu Tyr Trp Trp Gly Val Val Pro Phe Ser Gln Arg Lys Lys Met Leu Glu Lys Ala Arg Ala Lys Asn Lys Lys Pro Lys Ser Ser Ala Gly Ile Ser Ser Met Pro Asn Ile Thr Val Gly Thr Gln Val Cys Leu Arg Asn Asn Pro Leu Tyr His Ala Gly Ala Val Ala Phe Ser Ile Ser Ala Gly Ile Pro Lys Val Gly Val Leu Met Glu Ser Val Trp Asn Met Asn Asp Ser Cys Arg Phe Gln Leu Arg Ser Pro Glu Ser Leu Lys Asn Met Glu Lys Ala Ser Lys Thr Thr Glu Ala Lys Pro Glu Ser Lys Gln Glu Pro Val Lys Thr Glu Met Gly Pro Pro Pro Ser Pro A1a Ser Thr Cys Ser Asp Ala Ser Ser Ile Ala Ser Ser Ala Ser Met Pro Tyr Lys Arg Arg Arg Ser Thr Pro Ala Pro Lys Glu Glu G1u Lys Val Asn Glu Glu Gln Trp Ser Leu Arg Glu Val Val Phe Val Glu Asp Val Lys Asn Val Pro Val Gly Lys Val Leu Lys Val Asp Gly Ala Tyr Val Ala Val Lys Phe Pro Gly Thr Ser Ser Asn Thr Asn Cys Gln Asn Ser Ser Gly Pro Asp Ala Asp Pro Ser Ser Leu Leu Gln Asp Cys Arg Leu Leu Arg Ile Asp Glu Leu Gln Val Val Lys Thr Gly Gly Thr Pro Lys Val Pro Asp Cys Phe Gln Arg Thr Pro Lys Lys Leu Cys Ile Pro Glu Lys Thr Glu Ile Leu Ala Val Asn Val Asp Ser Lys Gly Val His Ala Val Leu Lys Thr Gly Asn Trp Val Arg Tyr Cys Ile Phe Asp Leu Ala Thr Gly Lys Ala Glu Gln Glu Asn Asn Phe Pro Thr Ser Ser Ile Ala Phe Leu Gly Gln Asn Glu Arg Asn Val Ala Ile Phe Thr Ala Gly Gln Glu Ser Pro Ile Ile Leu Arg Asp Gly Asn Gly Thr Ile Tyr Pro Met Ala Lys Asp Cys Met Gly Gly Ile Arg Asp Pro Asp Trp Leu Asp Leu Pro Pro Ile Ser Ser Leu Gly Met Gly Val His Ser Leu Ile Asn Leu Pro Ala Asn Ser Thr Ile Lys Lys Lys Ala Ala Va1 Ile Tle Met Ala Val Glu Lys G1n Thr Leu Met Gln His Ile Leu Arg Cys Asp Tyr Glu Ala Cys Arg Gln Tyr Leu Met Asn Leu Glu Gln Ala Val Val Leu Glu Gln Asn Leu Gln Met Leu Gln Thr Phe Ile Ser His Arg Cys Asp Gly Asn Arg Asn Ile Leu His Ala Cys Val Ser Val Cys Phe Pro Thr Ser Asn Lys Glu Thr Lys Glu Glu Glu Glu Ala Glu Arg Ser Glu Arg Asn 930 ' 935 940 Thr Phe Ala Glu Arg Leu Ser Ala Val Glu Ala Ile Ala Asn Ala Ile Ser Val Val Ser Ser Asn Gly Pro Gly Asn Arg Ala Gly Ser Ser Ser Ser Arg Ser Leu Arg Leu Arg Glu Met Met Arg Arg Ser Leu Arg Ala Ala Gly Leu Gly Arg His Glu Ala Gly Ala Ser Ser Ser Asp His Gln Asp Pro Val Ser Pro Pro Ile Ala Pro Pro Ser Trp Val Pro Asp Pro Pro Ala Met Asp Pro Asp Gly Asp Ile Asp Phe Ile Leu Ala Pro Ala Val Gly Ser Leu Thr Thr Ala Ala Thr Gly Thr Gly Gln Gly Pro Ser Thr Ser Thr Ile Pro Gly Pro Ser Thr Glu Pro Ser Val Val Glu Ser Lys Asp Arg Lys Ala Asn Ala His Phe Ile Leu Lys~Leu Leu Cys Asp Ser Val Val Leu Gln Pro Tyr Leu Arg Glu Leu Leu Ser Ala Lys Asp Ala Arg Gly Met Thr Pro Phe Met Ser Ala Val Ser Gly Arg Ala Tyr Pro Ala Ala Ile Thr Ile Leu Glu Thr Ala Gln Lys I1e Ala Lys Ala Glu Ile Ser Ser Ser Glu Lys Glu Glu Asp Val Phe Met Gly Met Val Cys Pro Ser Gly Thr Asn Pro Asp Asp Ser Pro Leu Tyr Val Leu Cys Cys Asn Asp Thr Cys Ser Phe Thr Trp Thr Gly Ala Glu His Ile Asn Gln Asp Ile Phe Glu Cys Arg Thr Cys Gly Leu Leu Glu Ser Leu Cys Cys Cys Thr Glu Cys Ala Arg Val Cys His Lys Gly His Asp Cys Lys Leu Lys Arg Thr Ser Pro Thr Ala Tyr Cys Asp Cys Trp Glu Lys Cys Lys Cys Lys Thr Leu Ile Ala Gly Gln Lys Ser Ala Arg Leu Asp Leu Leu Tyr Arg Leu Leu Thr Ala Thr Asn Leu Val Thr Leu Pro Asn Ser Arg Gly Glu His Leu Leu Leu Phe Leu Val Gln Thr Val Ala Arg Gln Thr Val Glu His Cys Gln Tyr Arg Pro Pro Arg Ile Arg Glu Asp Arg Asn Arg Lys Thr Ala Ser Pro Glu Asp Ser Asp Met Pro Asp His Asp Leu Glu Pro Pro Arg Phe Ala Gln Leu Ala Leu G1u Arg Val Leu Gln Asp Trp Asn Ala Leu Lys Ser Met Ile Met Phe G1y Ser Gln Glu Asn Lys Asp Pro Leu Ser Ala Ser Ser Arg Ile Gly His Leu Leu Pro Glu Glu Gln Val Tyr Leu Asn Gln Gln Ser Gly Thr Ile Arg Leu Asp Cys Phe Thr His Cys Leu Ile Val Lys Cys Thr Ala Asp Ile Leu Leu Leu Asp Thr Leu Leu Gly Thr Leu Val Lys Glu Leu Gln Asn Lys Tyr Thr Pro Gly Arg Arg Glu Glu Ala Ile Ala Val Thr Met Arg Phe Leu Arg Ser Val Ala Arg Val Phe Val Ile Leu Ser Val Glu Met Ala Ser Ser Lys Lys Lys Asn Asn Phe Ile Pro Gln Pro Ile Gly Lys Cys Lys Arg Val Phe Gln A1a Leu Leu Pro Tyr Ala Val Glu Glu Leu Cys Asn Val Ala Glu Ser Leu Ile Val Pro Val Arg Met Gly Ile Ala Arg Pro Thr Ala Pro Phe Thr Leu Ala Ser Thr Ser Ile Asp Ala Met Gln Gly Ser Glu Glu Leu Phe Ser Val Glu Pro Leu Pro Pro Arg Pro Ser Ser Asp Gln Ser Ser Ser Ser Ser Gln Sex Gln Ser Ser Tyr Ile Ile Arg Asn Pro Gln Gln Arg Arg Ile Ser Gln Ser Gln Pro Val Arg Gly Arg Asp Glu Glu Gln Asp Asp Ile Val Ser Ala Asp Val Glu Glu Val Glu Val Val Glu Gly Val Ala Gly Glu Glu Asp His His Asp Glu Gln Glu Glu His Gly Glu Glu Asn Ala Glu Ala Glu Gly Gln His Asp Glu His Asp Glu Asp Gly Ser Asp Met Glu Leu Asp Leu Leu Ala Ala Ala Glu Thr Glu Ser Asp Ser Glu Ser Asn His Ser Asn Gln Asp Asn Ala Ser Gly Arg Arg Ser Val Val Thr Ala Ala Thr A1a Gly Ser Glu Ala Gly Ala Ser Ser Val Pro Ala Phe Phe Ser Glu Asp Asp Ser Gln Ser Asn Asp Ser Ser Asp Ser Asp Ser Ser Ser Ser Gln Ser Asp Asp Ile Glu Gln Glu Thr Phe Met Leu Asp Glu Pro Leu Glu Arg Thr Thr Asn Ser Ser His Ala Asn Gly Ala Ala Gln Ala Pro Arg Ser Met G1n Trp A1a Val Arg Asn Thr Gln His Gln Arg Ala Ala Ser Thr Ala Pro Ser Ser Thr Ser Thr Pro Ala Ala Ser Ser Ala Gly Leu Ile Tyr Ile Asp Pro Ser Asn Leu Arg Arg Ser Gly Thr Ile Ser Thr Ser Ala Ala Ala Ala Ala Ala Ala Leu Glu Ala Ser Asn Ala Ser Ser Tyr Leu Thr -3~-Ser Ala Ser Ser Leu Ala Arg Ala Tyr Ser Ile Val Ile Arg Gln Tle Ser Asp Leu Met Gly Leu Tle Pro Lys Tyr Asn His Leu Val Tyr Ser Gln Ile Pro Ala Ala Val Lys Leu Thr Tyr Gln Asp Ala Val Asn Leu Gln Asn Tyr Val Glu Glu Lys Leu Ile Pro Thr Trp Asn Trp Met Val Ser Ile Met Asp Ser Thr Glu Ala Gln Leu Arg Tyr Gly Ser Ala Leu Ala Ser Ala Gly Asp Pro Gly His Pro Asn His Pro Leu His Ala Ser Gln Asn Ser Ala Arg Arg Glu Arg Met Thr Ala Arg Glu Glu Ala Ser Leu Arg Thr Leu Glu Gly Arg Arg Arg Ala Thr Leu Leu Ser Ala Arg Gln Gly Met Met Ser Ala Arg Gly Asp Phe Leu Asn Tyr Ala Leu Ser Leu Met Arg Ser His Asn Asp Glu His Ser Asp Val Leu Pro Val Leu Asp Val Cys Ser Leu Lys His Val Ala Tyr Val Phe Gln Ala Leu Ile Tyr Trp Ile Lys Ala Met Asn Gln Gln Thr Thr Leu Asp Thr Pro Gln Leu Glu Arg Lys Arg Thr Arg Glu Leu Leu Glu Leu Gly Ile Asp Asn Glu Asp Ser Glu His Glu Asn Asp Asp Asp Thr Asn Gln Ser Ala Thr Leu Asn Asp Lys Asp Asp Asp Ser Leu Pro Ala Glu Thr Gly Gln Asn His Pro Phe Phe Arg Arg Ser Asp Ser Met Thr Phe Leu Gly Cys Tle Pro Pro Asn Pro Phe Glu Val Pro Leu Ala Glu Ala Ile Pro Leu Ala Asp Gln Pro His Leu Leu Gln Pro Asn Ala Arg Lys Glu Asp Leu Phe Gly Arg Pro Ser Gln Gly Leu Tyr Ser Ser Ser Ala Ser Ser Gly Lys Cys Leu Met Glu Val Thr Val Asp Arg Asn Cys Leu Glu Val Leu Pro Thr Lys Met Ser Tyr Ala Ala Asn Leu Lys Asn Val Met Asn Met Gln Asn Arg Gln Lys Lys Glu Gly Glu Glu Gln Pro Val Leu Pro Glu Glu Thr Glu Ser Ser Lys Pro G1y Pro Ser Ala His Asp Leu Ala Ala Gln Leu Lys Ser Ser Leu Leu Ala Glu Ile Gly Leu Thr Glu Ser Glu Gly Pro Pro Leu Thr Ser Phe Arg Pro Gln Cys Ser Phe Met Gly Met Val Ile Ser His Asp Met Leu Leu Gly Arg Trp Arg Leu Ser Leu Glu Leu Phe Gly Arg Val Phe Met Glu Asp Val Gly Ala Glu Pro Gly Ser Ile Leu Thr Glu Leu Gly Gly Phe Glu Val Lys Glu Ser Lys Phe Arg Arg Glu Met Glu Lys Leu Arg Asn Gln Gln Ser Arg Asp Leu Ser Leu Glu Val Asp Arg Asp Arg Asp Leu Leu Ile Gln Gln Thr Met Arg Gln Leu Asn Asn His Phe Gly Arg Arg Cys Ala Thr Thr Pro Met Ala Val His Arg Val Lys Val Thr Phe Lys Asp Glu Pro Gly Glu Gly Ser Gly Val Ala Arg Ser Phe Tyr Thr Ala Ile Ala Gln Ala Phe Leu Ser Asn Glu Lys Leu Pro Asn Leu Glu Cys Ile Gln Asn Ala Asn Lys Gly Thr His Thr Ser Leu Met Gln Arg Leu Arg Asn Arg Gly Glu Arg Asp Arg Glu Arg Glu Arg Glu Arg Glu Met Arg Arg Ser Ser Gly Leu Arg Ala Gly Ser Arg Arg Asp Arg Asp Arg Asp Phe Arg Arg Gln Leu Ser Ile Asp Thr Arg Pro Phe Arg Pro Ala Ser Glu Gly Asn Pro Ser Asp Asp Pro Glu Pro Leu Pro Ala His Arg Gln Ala Leu Gly Glu Arg Leu Tyr Pro Arg Val Gln Ala Met Gln Pro Ala Phe Ala Ser Lys Ile Thr Gly Met Leu Leu Glu Leu Ser Pro Ala Gln Leu Leu Leu Leu Leu Ala Ser Glu Asp Ser Leu Arg Ala Arg Val Asp Glu Ala Met Glu Leu Ile Ile Ala His Gly Arg Glu Asn Gly Ala Asp Ser Ile Leu Asp Leu Gly Leu Val Asp Ser Ser Glu Lys Val Gln Gln Glu Asn Arg Lys Arg His Gly Ser Ser Arg Ser Val Val Asp Met Asp Leu Asp Asp Thr Asp Asp Gly Asp Asp Asn Ala Pro Leu Phe Tyr Gln Pro Gly Lys Arg Gly Phe Tyr Thr Pro Arg Pro Gly Lys Asn Thr Glu A1a Arg Leu Asn Cys Phe Arg Asn Ile Gly Arg Ile Leu G1y Leu Cys Leu Leu Gln Asn Glu Leu Cys Pro Ile Thr Leu Asn Arg His Val Ile Lys Val Leu Leu Gly Arg Lys Val Asn Trp His Asp Phe Ala Phe Phe Asp Pro Val Met Tyr Glu Ser Leu Arg Gln Leu Ile Leu Ala Ser Gln Ser Ser Asp Ala Asp Ala Val Phe Ser Ala Met Asp Leu Ala Phe Ala Tle Asp Leu Cys Lys Glu Glu Gly Gly Gly Gln Val Glu Leu Ile Pro Asn Gly Val Asn Ile Pro Val Thr Pro Gln Asn Val Tyr G1u Tyr Val Arg Lys Tyr Ala Glu His Arg Met Leu Val Val Ala Glu G1n Pro Leu His Ala Met Arg Lys Gly Leu Leu Asp Val Leu Pro Lys Asn Ser Leu Glu Asp Leu Thr Ala Glu Asp Phe Arg Leu Leu Val Asn Gly Cys Gly Glu Val Asn Val Gln Met Leu Tle Ser Phe Thr Ser Phe Asn Asp Glu Ser Gly Glu Asn Ala Glu Lys Leu Leu Gln Phe Lys Arg Trp Phe Trp Ser Ile Val Glu Lys Met Ser Met Thr Glu Arg Gln Asp Leu Val Tyr Phe Trp Thr Ser Ser Pro Ser Leu Pro Ala Ser Glu Glu Gly Phe Gln Pro Met Pro Ser Ile Thr Ile Arg Pro Pro Asp Asp Gln His Leu Pro Thr Ala Asn Thr Cys Ile Ser Arg Leu Tyr Val Pro Leu Tyr Ser Ser Lys Gln Ile Leu Lys Glz~ Lys Leu Leu Leu Ala Ile Lys Thr Lys Asn Phe Gly Phe Val <210> 5 <211> 30 <212> DNA
<213> CEDD microsatellite primer 1 <400> 5 taccctgcag taaatctcac atgtactccc 30 <210> 6 <211> 30 <212> DNA
<213> CEDD microsatellite primer 2 <400> 6 agaatcgctt gaacctagta ggtgaaggtg 30 <210> 7 <211> 288 <212> DNA
<213> CEDD amplified microsatellite sequence <400> 7 taccctgcag taaaatctca catgtactcc caagcctaaa agtttttttt ttaaaaaaaa 60 aaaaaagaaa gaaagaaaga aatgaaaact ctgcccctcc cccccaaaaa acccttaagg 120 atataggaaa gaaagttatt ttttagatag ctacacaatg tgtgtgtgtg tgtgtgtgtg 180 tgtgtgtgtg taagacagag tctcactctg tcgcccaggc tggagtgcag tggcatgatc 240 tcagctcact gcaacctcca ccttcaccta ctaggttcaa gcgattct 288 <210> 8 <21l> 1976 <212> DNA
<213> human importin alpha-1 cDNA
<400>

gccacacggtctttgagctgagtcgaggtggaccctttgaacgcagtcgccctacagccg60 ctgattccccccgcatcgcctcccgtggaagcccaggcccgcttcgcagctttctccctt120 tgtctcataaccatgtccaccaacgagaatgctaatacaccagctgcccgtcttcacaga180 ttcaagaacaagggaaaagacagtacagaaatgaggcgtcgcagaatagaggtcaatgtg240 gagctgaggaaagctaagaaggatgaccagatgctgaagaggagaaatgtaagctcattt300 cctgatgatgctacttctccgctgcaggaaaaccgcaacaaccagggcactgtaaattgg360 tctgttgatgacattgtcaaaggcataaatagcagcaatgtggaaaatcagctccaagct420 actcaagctgccaggaaactactttccagagaaaaacagccccccatagacaacataatc480 cgggctggtttgattccgaaatttgtgtccttcttgggcagaactgattgtagtcccatt540 cagtttgaatctgcttgggcactcactaacattgcttctgggacatcagaacaaaccaag600 gctgtggtagatggaggtgccatcccagcattcatttctctgttggcatctccccatgct660 cacatcagtgaacaagctgtctgggctctaggaaacattgcaggtgatggctcagtgttc720 cgagacttggttattaagtacggtgcagttgacccactgttggctctccttgcagttcct780 gatatgtcatctttagcatgtggctacttacgtaatcttacctggacactttctaatctt840 tgccgcaacaagaatcctgcacccccgatagatgctgttgagcagattcttcctacctta900 gttcggctcctgcatcatgatgatccagaagtgttagcagatacctgctgggctatttcc960 taccttactgatggtccaaatgaacgaattggcatggtggtgaaaacaggagttgtgccc1020 caacttgtgaagcttct-aggagcttctgaattgccaattgtgactcctgccctaagagcc1080 atagggaatattgtcactggtacagatgaacagactcaggttgtgattgatgcaggagca1140 ctcgccgtctttcccagcctgctcaccaaccccaaaactaacattcagaaggaagctacg1200 tggacaatgtcaaacatcacagccggccgccaggaccagatacagcaagttgtgaatcat1260 ggattagtcccattccttgtcagtgttctctctaaggcagattttaagacacaaaaggaa1320 gctgtgtgggccgtgaccaactataccagtggtggaacagttgaacagattgtgtacctt1380 gttcactgtggcataatagaaccgttgatgaacctcttaactgcaaaagataccaagatt1440 attctggttatcctggatgccatttcaaatatctttcaggctgctgagaaactaggtgaa1500 actgagaaacttagtataatgattgaagaatgtggaggcttagacaaaattgaagctcta1560 caaaaccatgaaaatgagtctgtgtataaggcttcgttaagcttaattgagaagtatttc1620 tctgtagaggaagaggaagatcaaaacgttgtaccagaaactacctctgaaggctacact1680 ttccaagttcaggatggggctcctgggacctttaacttttagatcatgtagctgagacat1740 aaatttgttgtgtactacgtttggtattttgtcttattgtttctctactaagaactcttt1800 cttaaatgtggtttgttactgtagcactttttacactgaaactatacttgaacagttcca1860 actgtacatacatactgtatgaagcttgtcctctgactaggtttctaatttctatgtgga1920 atttcctatcttgcagcatcctgtaaataaacattcaagtccacccttaaaaaaaa 1976 <210> 9 <211> 529 <212> PRT
<213> human importin alpha-1 protein <400> 9 Met Ser Thr Asn Glu Asn Ala Asn Thr Pro Ala Ala Arg Leu His Arg Phe Lys Asn Lys Gly Lys Asp Ser Thr Glu Met Arg Arg Arg Arg Ile Glu Val Asn Val Glu Leu Arg Lys Ala Lys Lys Asp Asp Gln Met Leu Lys Arg Arg Asn Val Ser Ser Phe Pro Asp Asp Ala Thr Ser Pro Leu Gln Glu Asn Arg Asn Asn Gln Gly Thr Val Asn Trp Ser Va1 Asp Asp Ile Val Lys Gly Ile Asn Ser Ser Asn Val Glu Asn Gln Leu Gln Ala Thr Gln Ala Ala Arg Lys Leu Leu Ser Arg Glu Lys Gln Pro Pro Ile Asp Asn Ile Ile Arg Ala Gly Leu Ile Pro Lys Phe Val Ser Phe Leu 115 l20 125 Gly Arg Thr Asp Cys Ser Pro Ile Gln Phe Glu Ser Ala Trp Ala Leu Thr Asn Ile Ala Ser Gly Thr Ser Glu Gln Thr Lys Ala Val Val Asp Gly Gly Ala Ile Pro Ala Phe Ile Ser Leu Leu Ala Ser Pro His Ala His Ile Ser Glu Gln Ala Val Trp Ala Leu Gly Asn Ile Ala Gly Asp Gly Ser Val Phe Arg Asp Leu Val Ile Lys Tyr Gly Ala Val Asp Pro Leu Leu Ala Leu Leu Ala Val Pro Asp Met Ser Ser Leu Ala Cys Gly Tyr Leu Arg Asn Leu Thr Trp Thr Leu Ser Asn Leu Cys Arg Asn Lys Asn Pro Ala Pro Pro Ile Asp Ala Val Glu Gln Ile Leu Pro Thr Leu Val Arg Leu Leu His His Asp Asp Pro Glu Va1 Leu Ala Asp Thr Cys Trp Ala Ile Ser Tyr Leu Thr Asp Gly Pro Asn Glu Arg Ile Gly Met Val Val Lys Thr Gly Val Val Pro Gln Leu Val Lys Leu Leu Gly Ala Ser G1u Leu Pro Ile Val Thr Pro Ala Leu Arg Ala Ile Gly Asn Ile Val Thr Gly Thr Asp Glu Gln Thr Gln Val Val Ile Asp Ala Gly Ala Leu Ala Va1 Phe Pro Ser Leu Leu Thr Asn Pro Lys Thr Asn Ile Gln Lys Glu Ala Thr Trp Thr Met Ser Asn Ile Thr Ala Gly Arg Gln Asp Gln Ile Gln Gln Val Val Asn His Gly Leu Val Pro Phe Leu Val Ser Val Leu Ser Lys Ala Asp Phe Lys Thr Gln Lys Glu Ala Val Trp Ala Val Thr Asn Tyr Thr Ser Gly Gly Thr Val Glu Gln Ile Val Tyr Leu Val His Cys Gly Ile Ile Glu Pro Leu Met Asn Leu Leu Thr Ala Lys Asp Thr Lys Ile Ile Leu Val Ile Leu Asp Ala Ile Ser Asn Ile Phe Gln Ala Ala Glu Lys Leu Gly Glu Thr Glu Lys Leu Ser Ile Met Ile Glu Glu Cys Gly Gly Leu Asp Lys Ile Glu Ala Leu Gln Asn His Glu Asn Glu Ser Val Tyr Lys Ala Ser Leu Ser Leu Ile Glu Lys Tyr Phe Ser Val Glu Glu Glu Glu Asp Gln Asn Val Val Pro Glu Thr Thr Ser Glu Gly Tyr Thr Phe Gln Val Gln Asp Gly Ala Pro Gly Thr Phe Asn Phe <210> 10 <211> 1717 <212> DNA
<213> human importin alpha-3 cDNA
<400>

gcacgagccatggcggacaacgagaaactggacaaccaacggctcaagaatttcaagaac60 aaaggccgcgacttggagactatgagaagacaacgaaatgaagttgtagttgaattaagg120 aagaataaaagagatgaacatctcttaaagagaaggaatgtaccacatgaagatatctgt180 gaagactctgatatagatggtgattatagagtgcaaaatacctctctagaagctattgtt240 caaaatgcttcaagtgataaccaaggaattcaattaagtgcagttcaagctgctaggaag300 cttttgtccagtgatcgaaatccaccaattgatgacttaataaaatctggaatattgccc360 attttagtccattgtcttgaaagagatgacaatccttctttacagtttgaagctgcatgg420 gctttgacaaacattgcatctggaacttctgaacaaactcaagcagtagttcagtccaat480 gctgtgccacttttcctgaggcttctccattcaccccatcagaatgtctgtgagcaagca540 gtgtgggcattgggaaatatcataggtgatgggccccagtgtagagattatgtcataagt600 cttggagttgtgaaacctttactttccttcataagtccatctattcctataacattctta660 agaaatgttacttgggttatggtcaacttatgtcgccacaaagacccaccaccaccaatg720 gaaaccattcaggagattcttccagccctttgtgttttaattcatcacacagatgtaaat780 atactggtagacacagtctgggccctctcttaccttactgatgctggcaatgaacaaata840 cagatggtaatagactctggaatagttcctcatttggttcctctgctcagccaccaggaa900 gttaaagttcagactgctgcacttagagctgtgggcaacattgttactggaactgatgag960 caaacacaagtagttttgaactgtgatgctctttcacacttcccagcactcctgacacat1020 cccaaagagaaaattaataaagaagcagtgtggttcctctccaacatcactgcaggaaat1080 cagcagcaggtacaggcagtaattgatgccaatcttgtaccaatgataatacaccttttg1140 gataagggggattttggcactcaaaaagaagctgcttgggccataagtaacttaacaatt1200 agtggaaggaaagatcaagtggcttaccttatccaacaaaatgttatcccacctttttgc1260 aacttgctgactgtaaaagatgcacaagttgtgcaagtagtactcgatggactaagtaat1320 atattaaaaatggctgaagatgaggcagaaaccataggcaatcttatagaagaatgtgga1380 gggctggagaaaattgaacaacttcaaaatcatgaaaatgaagacatctacaaattggcc1440 tatgagatcattgatcagttcttctcttcagatgatattgatgaagaccctagccttgtt1500 ccagaggcaattcaaggcggaacatttggtttcaattcatctgccaatgtaccaacagaa1560 gggttccagttttagaaagatgttgtggaagttaggtacaatgcagcactgagatatata1620 tatatatatgtgtgtgtgtatatatatatatatatacatatatataaaaaggtttgatcc1680 atcaaaaaaa aaaaaaaaaa aaaaaaaaaa aaaaaaa 1717 <210> 11 <211> 521 <212> PRT
<213> human importin alpha-3 protein <400> 11 Met Ala Asp Asn Glu Lys Leu Asp Asn Gln Arg Leu Lys Asn Phe Lys Asn Lys Gly Arg Asp Leu Glu Thr Met Arg Arg Gln Arg Asn Glu Val Val Val Glu Leu Arg Lys Asn Lys Arg Asp Glu His Leu Leu Lys Arg Arg Asn Val Pro His Glu Asp Ile Cys Glu Asp Ser Asp Ile Asp Gly Asp Tyr Arg Val Gln Asn Thr Ser Leu Glu Ala Ile Val Gln Asn Ala Ser Ser Asp Asn Gln Gly Ile Gln Leu Ser Ala Val Gln Ala Ala Arg Lys Leu Leu Ser Ser Asp Arg Asn Pro Pro Ile Asp Asp Leu Ile Lys Ser Gly Ile Leu Pro Ile Leu Val His Cys Leu Glu Arg Asp Asp Asn Pro Ser Leu Gln Phe Glu Ala A1a Trp Ala Leu Thr Asn Ile Ala Ser Gly Thr Ser Glu Gln Thr Gln Ala Val Val Gln Ser Asn Ala Val Pro Leu Phe Leu Arg Leu Leu His Ser Pro His Gln Asn Val Cys Glu Gln Ala Val Trp Ala Leu Gly Asn Ile Ile Gly Asp Gly Pro Gln Cys Arg Asp Tyr Val Ile Ser Leu Gly Val Val Lys Pro Leu Leu Ser Phe Ile Ser Pro Ser Ile Pro Ile Thr Phe Leu Arg Asn Val Thr Trp Val Met Val Asn Leu Cys Arg His Lys Asp Pro Pro Pro Pro Met Glu Thr Ile Gln Glu Ile Leu Pro Ala Leu Cys Val Leu Ile His His Thr Asp Val Asn Ile Leu Val Asp Thr Val Trp Ala Leu Ser Tyr Leu Thr Asp Ala Gly Asn Glu Gln Ile Gln Met Val Ile Asp Ser Gly Ile Val Pro His Leu Val Pro Leu Leu Ser His Gln Glu Val Lys Val Gln Thr Ala Ala Leu Arg Ala Val Gly Asn Ile Val Thr Gly Thr Asp G1u Gln Thr Gln Val Val Leu Asn Cys Asp Ala Leu Ser His Phe Pro Ala Leu Leu Thr His Pro Lys G1u Lys Ile Asn Lys Glu Ala Val Trp Phe Leu Ser Asn Ile Thr Ala Gly Asn Gln Gln Gln Val Gln Ala Val Ile Asp Ala Asn Leu Val Pro Met Ile Ile His Leu Leu Asp Lys Gly Asp Phe Gly Thr Gln Lys Glu Ala Ala Trp Ala Ile Ser Asn Leu Thr Ile 5er Gly Arg Lys Asp Gln Val Ala Tyr Leu Ile Gln Gln Asn Val Ile Pro Pro Phe Cys Asn Leu Leu Thr Val Lys Asp Ala Gln Val Val Gln Val Val Leu Asp Gly Leu Ser Asn Ile Leu Lys Met Ala Glu Asp Glu Ala Glu Thr Ile Gly Asn Leu Ile Glu Glu Cys Gly Gly Leu Glu Lys Ile Glu Gln Leu Gln Asn His Glu Asn Glu Asp Ile Tyr Lys Leu Ala Tyr G1u Ile Ile Asp Gln Phe Phe Ser Ser Asp Asp Ile Asp Glu Asp Pro Ser Leu Val Pro Glu Ala Ile Gln Gly Gly Thr Phe Gly Phe Asn Ser Ser Ala Asn Val Pro Thr Glu Gly Phe Gln Phe <210> 12 <211> 2940 <212> DNA
<213> human importin alpha-5 cDNA

<400>

ctaacttcagcggtggcaccgggatcggttgccttgagcctgaaatatgaccaccccagg60 aaaagagaactttcgcctgaaaagttacaagaacaaatctctgaatcccgatgagatgcg120 caggaggagggaggaagaaggactgcagttacgaaagcagaaaagagaagagcagttatt180 caagcggagaaatgttgctacagcagaagaagaaacagaagaagaagttatgtcagatgg240 aggctttcatgaggctcagattagtaacatggagatggcaccaggtggtgtcatcacttc300 tgacatgattgagatgatattttccaaaagcccagagcaacagctttcagcaacacagaa360 attcaggaagctgctttcaaaagaacctaaccctcctattgatgaagttatcagcacacc420 aggagtagtggccaggtttgtggagttcctcaaacgaaaagagaattgttcactgcagtt480 tgaatcagcttgggtactgacaaatattgcttcaggaaattctcttcagacccgaattgt540 gattcaggcaagagctgtgcccatcttcatagagttgctcagctcagagtttgaagatgt600 ccaggaacaggcagtctgggctcttggcaacattgctggagatagtaccatgtgcaggga660 ctatgtcttagactgcaatatccttccccctcttttgcagttattttcaaagcaaaaccg720 cctgaccatgacccggaatgcagtatgggctttgtctaatctctgtagagggaaaagtcc780 acctccagaatttgcaaaggtttctccatgtctgaatgtgctttcctggttgctgtttgt840 cagtgacactgatgtactggctgatgcctgctgggccctctcatatctatcagatggacc900 caatgataaaattcaagcggtcatcgatgcgggagtatgtaggagacttgtggaactgct960 gatgcataatgattataaagtggtttctcctgctttgcgagctgtgggaaacattgtcac1020 aggggatgatattcagacacaggtaattctgaattgctcagctctgcagagtttattgca1080 tttgctgagtagcccaaaggaatctatcaaaaaggaagcatgttggacgatatctaatat1140 tacagctggaaatagggcacagatccagactgtgatagatgccaacattttcccagccct1200 cattagtattttacaaactgctgaatttcggacaagaaaagaagcagcttgggccatcac1260 aaatgcaacttctggaggatcagctgaacagatcaagtacctagtagaactgggttgtat1320 caagccgctctgtgatctcctcacggtcatggactctaagattgtacaggttgccctaaa1380 tggcttggaaaatatcctgaggcttggagaacaggaagccaaaaggaacggcactggcat1440 taacccttactgtgctttgattgaagaagcttatggtctggataaaattgagttcttaca1500 gagtcatgaaaaccaggagatctaccaaaaggcctttgatcttattgagcattacttcgg1560 gaccgaagatgaagacagcagcattgcaccccaggttgaccttaaccagcagcagtacat1620 cttccaacagtgtgaggctcctatggaaggtttccagctttgaagcaatactctgctttc1680 acgtacctgtgctcagaccaggctacccagtcgagtcctcttgtggagcccacagtcctc1740 atggagctaacttctcaaatgttttccataatactgtttgcgctcatttgcttgccttgc1800 gcacctgctctcttacacacatctggaaaacctccggctctctgtggtgggatacccttc1860 taataaaagggtaaccagaacggcccactctcttttacggaaaaatccctaggctttgga1920 gatccgcacttacattagagttatgggaatatacacatattaatgtggctccctttttct1980 tgtgggggaataaaagaggactcctcctcattccctttaacatgggggaaaaaactgaca2040 ttaaaagatgagactaaatctttatcttgaattttacacaactacttacgacaagggaga2100 tgtttagacctgttggtatacttcagagtacttttcatgagttcttccacagtgaaccct2160 tggattacctggtggctttttctagccagattgcattaatccttactgagattggatggt2220 tttctttcctctattggcgccattcttcagatattaaagttaaaccatccactccctcac2280 cttcagccttcagtgaatgtgctttctagttgtcaggaatgctgaagaattaacactttg2340 actcctaaatgtgatactggtgggtaagagcagggcacatttaatttgttcgcttttgct2400 tctctttggtctgggcacatttaatttgttcgcttttgcttctctttggtcttttcgaat2460 acttagtaatcgaaaaccatatcctgtaatttaataaaaaaaactaaggacgaaaaaacc2520 cctccaattttcccaaatgcaatcagtgtaactaggggctgtgtttctgcattaaaataa2580 atgtttcaggctttgtggtcctgatcaaggtcctcattaaaaaattggagttcaccctag2640 gcttttcccctctgtgactggcagataacacatacttttgaaagtaactttgggattttt2700 tttcttaggtgcagctcgattctaatcttttcatgctgcacacgattcctttaatcgata2760 gcatccttatctgaaagaaataaccatcttctcaacatgacctgcttaacccaaataaga2820 acagtgatcttataacctcattgtttcctaatctattttatttcatctcctgctagtact2880 gtgccgcttc cccctccccc cacacaaaat aaaaacagta tctcgcttct ggctcatttt 2940 <210> 13 <211> 538 <212> PRT
<213> importin alpha-5 protein <400> 13 Met Thr Thr Pro Gly Lys Glu Asn Phe Arg Leu Lys Ser Tyr Lys Asn Lys Ser Leu Asn Pro Asp Glu Met Arg Arg Arg Arg Glu Glu Glu Gly Leu Gln Leu Arg Lys Gln Lys Arg Glu Glu Gln Leu Phe Lys Arg Arg Asn Val Ala Thr Ala Glu Glu Glu Thr Glu Glu Glu Val Met Ser Asp Gly Gly Phe His Glu Ala Gln Tle Ser Asn Met Glu Met Ala Pro Gly Gly Val Ile Thr Ser Asp Met Ile Glu Met Ile Phe Ser Lys Ser Pro Glu Gln Gln Leu Ser Ala Thr Gln Lys Phe Arg Lys Leu Leu Ser Lys Glu Pro Asn Pro Pro Ile Asp Glu Val Ile Ser Thr Pro Gly Val Val 115 l20 125 Ala Arg Phe Val Glu Phe Leu Lys Arg Lys Glu Asn Cys Ser Leu Gln Phe Glu Ser A1a Trp Val Leu Thr Asn Ile Ala Ser Gly Asn Ser Leu Gln Thr Arg Tle Val Ile Gln Ala Arg Ala Val Pro Tle Phe Ile Glu Leu Leu Ser Ser Glu Phe Glu Asp Val Gln Glu Gln Ala Val Trp Ala 180 185 l90 Leu Gly Asn Ile Ala Gly Asp Ser Thr Met Cys Arg Asp Tyr Val Leu Asp Cys Asn Ile Leu Pro Pro Leu Leu Gln Leu Phe Ser Lys Gln Asn Arg Leu Thr Met Thr Arg Asn Ala Val Trp Ala Leu Ser Asn Leu Cys Arg Gly Lys Ser Pro Pro Pro Glu Phe Ala Lys Val Ser Pro Cys Leu Asn Val Leu Ser Trp Leu Leu Phe Va1 Ser Asp Thr Asp Val Leu Ala Asp Ala Cys Trp Ala Leu Ser Tyr Leu Ser Asp G1y Pro Asn Asp Lys Ile Gln Ala Val Ile Asp Ala Gly Val Cys Arg Arg Leu Val Glu Leu Leu Met His Asn Asp Tyr Lys Val Val Ser Pro Ala Leu Arg Ala Val Gly Asn Ile Val Thr Gly Asp Asp Ile Gln Thr Gln Val Ile Leu Asn Cys Ser Ala Leu Gln Ser Leu Leu His Leu Leu Ser Ser Pro Lys Glu Ser Ile Lys Lys Glu Ala Cys Trp Thr Ile Ser Asn Ile Thr Ala Gly Asn Arg Ala Gln Ile Gln Thr Val Ile Asp Ala Asn Ile Phe Pro Ala Leu 2le Ser 2le Leu Gln Thr A1a Glu Phe Arg Thr Arg Lys Glu Ala Ala Trp Ala 21e Thr Asn Ala Thr Ser Gly Gly Ser Ala Glu Gln Ile Lys Tyr Leu Val Glu Leu Gly Cys Tle Lys Pro Leu Cys Asp Leu Leu Thr Val Met Asp Ser Lys Ile Val Gln Val Ala Leu Asn Gly Leu Glu Asn Ile Leu Arg Leu Gly Glu Gln Glu Ala Lys Arg Asn Gly Thr Gly Ile Asn Pro Tyr Cys Ala Leu Ile Glu Glu Ala Tyr Gly Leu Asp Lys Ile Glu Phe Leu Gln Ser His Glu Asn Gln Glu Ile Tyr Gln Lys Ala Phe Asp Leu Ile Glu His Tyr Phe Gly Thr Glu Asp Glu Asp Ser Ser Ile Ala Pro Gln Val Asp Leu Asn Gln Gln Gln Tyr Ile Phe Gln Gln Cys Glu Ala Pro Met Glu Gly Phe Gln Leu <210> 14 <211> 3014 <212> DNA
<213> human progesterone receptor cDNA
<400> 14 ctgaccagcg ccgccctccc ccgcccccga cccaggaggt ggagatccct ccggtccagc 60 cacattcaac acccactttc tcctccctct gcccctatat tcccgaaacc ccctcctcct 120 tcccttttccctcctccctggagacgggggaggagaaaaggggagtccagtcgtcatgac180 tgagctgaaggcaaagggtccccgggctccccacgtggcgggcggcccgccctcccccga240 ggtcggatccccactgctgtgtcgcccagccgcaggtccgttcccggggagccagacctc300 ggacaccttgcctgaagtttcggccatacctatctccctggacgggctactcttccctcg360 gccctgccagggacaggacccctccgacgaaaagacgcaggaccagcagtcgctgtcgga420 cgtggagggcgcatattccagagctgaagctacaaggggtgctggaggcagcagttctag480 tcccccagaaaaggacagcggactgctggacagtgtcttggacactctgttggcgccctc540 aggtcccgggcagagccaacccagccctcccgcctgcgaggtcaccagctcttggtgcct600 gtttggccccgaacttcccgaagatccaccggctgcccccgccacccagcgggtgttgtc660 cccgctcatgagccggtccgggtgcaaggttggagacagctccgggacggcagctgccca720 taaagtgctgccccggggcctgtcaccagcccggcagctgctgctcccggcctctgagag780 ccctcactggtccggggccccagtgaagccgtctccgcaggccgctgcggtggaggttga840 ggaggaggatggctctgagtccgaggagtctgcgggtccgcttctgaagggcaaacctcg900 ggctctgggtggcgcggcggctggaggaggagccgcggctgtcccgccgggggcggcagc960 aggaggcgtcgccctggtccccaaggaagattcccgcttctcagcgcccagggtcgccct1020 ggtggagcaggacgcgccgatggcgcccgggcgctccccgctggccaccacggtgatgga1080 tttcatccacgtgcctatcctgcctctcaatcacgccttattggcagcccgcactcggca1140 gctgctggaagacgaaagttacgacggcggggccggggctgccagcgcctttgccccgcc1200 gcggagttcaccctgtgcctcgtccaccccggtcgctgtaggcgacttccccgactgcgc1260 gtacccgcccgacgccgagcccaaggacgacgcgtaccctctctatagcgacttccagcc1320 gcccgctctaaagataaaggaggaggaggaaggcgcggaggcctccgcgcgctccccgcg1380 ttcctaccttgtggccggtgccaaccccgcagccttcccggatttcccgttggggccacc1440 gcccccgctgccgccgcgagcgaccccatccagacccggggaagcggcggtgacggccgc1500 acccgccagtgcctcagtctcgtctgcgtcctcctcggggtcgaccctggagtgcatcct1560 gtacaaagcggagggcgcgccgccccagcagggcccgttcgcgccgccgccctgcaaggc1620 gccgggcgcgagcggctgcctgctcccgcgggacggcctgccctccacctccgcctctgc1680 cgccgccgccggggcggcccccgcgctctaccctgcactcggcctcaacgggctcccgca1740 gctcggctaccaggccgccgtgctcaaggagggcctgccgcaggtctacccgccctatct1800 caactacctgaggccggattcagaagccagccagagcccacaatacagcttcgagtcatt1860 acctcagaagatttgtttaatctgtggggatgaagcatcaggctgtcattatggtgtcct1920 tacctgtgggagctgtaaggtcttctttaagagggcaatggaagggcagcacaactactt1980 atgtgctggaagaaatgactgcatcgttgataaaatccgcagaaaaaactgcccagcatg2040 tcgccttagaaagtgctgtcaggctggcatggtccttggaggtcgaaaatttaaaaagtt2100 caataaagtcagagttgtgagagcactggatgctgttgctctcccacagccagtgggcgt2160 tccaaatgaaagccaagccctaagccagagattcactttttcaccaggtcaagacataca2220 gttgattccaccactgatcaacctgttaatgagcattgaaccagatgtgatctatgcagg2280 acatgacaacacaaaacctgacacctccagttctttgctgacaagtcttaatcaactagg2340 cgagaggcaacttctttcagtagtcaagtggtctaaatcattgccaggttttcgaaactt2400 acatattgatgaccagataactctcattcagtattcttggatgagcttaatggtgtttgg2460 tctaggatggagatcctacaaacacgtcagtgggcagatgctgtattttgcacctgatct2520 aatactaaatgaacagcggatgaaagaatcatcattctattcattatgccttaccatgtg2580 gcagatcccacaggagtttgtcaagcttcaagttagccaagaagagttcctctgtatgaa2640 agtattgttacttcttaatacaattcctttggaagggctacgaagtcaaacccagtttga2700 ggagatgaggtcaagctacattagagagctcatcaaggcaattggtttgaggcaaaaagg2760 agttgtgtcgagctcacagcgtttctatcaacttacaaaacttcttgataacttgcatga2820 tcttgtcaaacaacttcatctgtactgcttgaatacatttatccagtcccgggcactgag2880 tgttgaatttccagaaatgatgtctgaagttattgctgcacaattacccaagatattggc2940 agggatggtgaaaccccttctctttcataaaaagtgaatgtcatctttttcttttaaaga3000 attaaattttgtgg 3014 <210> 15 <211> 933 <212> PRT
<213> human progesterone receptor protein <400> 15 Met Thr Glu Leu Lys Ala Lys Gly Pro Arg Ala Pro His Val Ala Gly Gly Pro Pro Ser Pro Glu Val Gly Ser Pro Leu Leu Cys Arg Pro Ala Ala Gly Pro Phe Pro G1y Ser Gln Thr Ser Asp Thr Leu Pro Glu Val Ser Ala Ile Pro Ile Ser Leu Asp Gly Leu Leu Phe Pro Arg Pro Cys Gln Gly Gln Asp Pro Ser Asp Glu Lys Thr Gln Asp Gln Gln Ser Leu Ser Asp Val Glu Gly Ala Tyr Ser Arg Ala Glu Ala Thr Arg Gly Ala Gly Gly Ser Ser 5er Ser Pro Pro Glu Lys Asp Ser Gly Leu Leu Asp Ser Val Leu Asp Thr Leu Leu Ala Pro Ser Gly Pro Gly Gln Ser Gln l15 120 125 Pro Ser Pro Pro Ala Cys Glu Val Thr Ser Ser Trp Cys Leu Phe Gly Pro Glu Leu Pro Glu Asp Pro Pro Ala Ala Pro Ala Thr Gln Arg Val 145 150 155 l60 Leu Ser Pro Leu Met Ser Arg Ser Gly Cys Lys Val Gly Asp Ser Ser Gly Thr Ala Ala Ala His Lys Val Leu Pro Arg Gly Leu Ser Pro Ala Arg Gln Leu Leu Leu Pro Ala Ser Glu Ser Pro His Trp Ser Gly Ala Pro Val Lys Pro 5er Pro Gln A1a Ala A1a Val Glu Val Glu Glu Glu Asp Gly Ser Glu Ser Glu Glu Ser Ala Gly Pro Leu Leu Lys Gly Lys Pro Arg Ala Leu Gly Gly Ala Ala Ala Gly Gly Gly Ala Ala Ala Val Pro Pro Gly Ala Ala Ala Gly Gly Val Ala Leu Val Pro Lys Glu Asp Ser Arg Phe Ser Ala Pro Arg Val Ala Leu Val Glu Gln Asp Ala Pro Met Ala Pro Gly Arg Ser Pro Leu Ala Thr Thr Val Met Asp Phe Ile His Val Pro Ile Leu Pro Leu Asn His A1a Leu Leu Ala Ala Arg Thr Arg Gln Leu Leu Glu Asp Glu Ser Tyr Asp Gly Gly Ala Gly Ala Ala Ser Ala Phe Ala Pro Pro Arg Ser Ser Pro Cys Ala Ser Ser Thr Pro Val Ala Val Gly Asp Phe Pro Asp Cys Ala Tyr Pro Pro Asp Ala Glu Pro Lys Asp Asp Ala Tyr Pro Leu Tyr Ser Asp Phe Gln Pro Pro Ala Leu Lys Ile Lys Glu Glu Glu Glu Gly Ala Glu Ala Ser Ala Arg Ser Pro Arg Ser Tyr Leu Val Ala Gly Ala Asn Pro Ala Ala Phe Pro Asp Phe Pro Leu Gly Pro Pro Pro Pro Leu Pro Pro Arg Ala Thr Pro Ser Arg Pro Gly Glu Ala Ala Val Thr Ala Ala Pro Ala Ser Ala Ser Val Ser Ser Ala Ser Ser Ser Gly Ser Thr Leu Glu Cys Ile Leu Tyr Lys Ala Glu Gly Ala Pro Pro Gln Gln Gly Pro Phe Ala Pro Pro Pro Cys Lys Ala Pro Gly Ala Ser Gly Cys Leu Leu Pro Arg Asp Gly Leu Pro Ser Thr Ser Ala Ser Ala Ala Ala Ala Gly Ala Ala Pro Ala Leu Tyr -5~-Pro Ala Leu G1y Leu Asn Gly Leu Pro Gln Leu Gly Tyr Gln Ala Ala Val Leu Lys Glu Gly Leu Pro Gln Val Tyr Pro Pro Tyr Leu Asn Tyr Leu Arg Pro Asp Ser Glu A1a Ser Gln Ser Pro Gln Tyr Ser Phe Glu Ser Leu Pro Gln Lys Ile Cys Leu Tle Cys Gly Asp Glu Ala Ser Gly Cys His Tyr Gly Val Leu Thr Cys Gly Ser Cys Lys Val Phe Phe Lys Arg Ala Met Glu Gly Gln His Asn Tyr Leu Cys Ala Gly Arg Asn Asp Cys Ile Val Asp Lys Ile Arg Arg Lys Asn Cys Pro Ala Cys Arg Leu Arg Lys Cys Cys Gln A1a Gly Met Val Leu Gly Gly Arg Lys Phe Lys Lys Phe Asn Lys Val Arg Val Val Arg Ala Leu Asp Ala Val Ala Leu Pro Gln Pro Val G1y Val Pro Asn Glu Ser Gln Ala Leu Ser Gln Arg Phe Thr Phe Ser Pro Gly Gln Asp Ile Gln Leu Ile Pro Pro Leu Ile Asn Leu Leu Met Ser Ile Glu Pro Asp Val Ile Tyr Ala Gly His Asp Asn Thr Lys Pro Asp Thr Ser Ser Ser Leu Leu Thr Ser Leu Asn Gln Leu Gly Glu Arg Gln Leu Leu Ser Val Val Lys Trp Ser Lys Ser Leu Pro Gly Phe Arg Asn Leu His Ile Asp Asp Gln Ile Thr Leu Ile Gln Tyr Ser Trp Met Ser Leu Met Val Phe Gly Leu Gly Trp Arg Ser Tyr Lys His Val Ser Gly Gln Met Leu Tyr Phe Ala Pro Asp Leu Ile Leu Asn Glu Gln Arg Met Lys Glu Ser Ser Phe Tyr Ser Leu Cys Leu Thr Met Trp Gln Ile Pro Gln Glu Phe Val Lys Leu Gln Val Ser Gln Glu Glu Phe Leu Cys Met Lys Val Leu Leu Leu Leu Asn Thr Ile Pro Leu Glu Gly Leu Arg Ser Gln Thr Gln Phe Glu Glu Met Arg Ser Ser Tyr Ile Arg Glu Leu Ile Lys Ala Ile Gly Leu Arg Gln Lys Gly Val Val Ser Ser Ser Gln Arg Phe Tyr Gln Leu Thr Lys Leu Leu Asp Asn Leu His Asp Leu Val Lys Gln Leu His Leu Tyr Cys Leu Asn Thr Phe Ile Gln Ser Arg Ala Leu Ser Val Glu Phe Pro Glu Met Met Ser Glu Val Ile Ala A1a Gln Leu Pro Lys Ile Leu Ala Gly Met Val Lys Pro Leu 9l5 920 925 Leu Phe His Lys Lys <210> 16 <211> 905 <212> DNA
<213> human calcium and integrin binding protein (CIB1) cDNA
<400> 16 tctcccgaat tcggcacgag gcggcgtctc gaggcgagtt ggcggagctg tgcgcgcggc 60 ggggcgatgg ggggctcggg cagtcgcctg tccaaggagc tgctggccga gtaccaggac 120 ttgacgttcc tgacgaagca ggagatcctc ctagcccaca ggcggttttg tgagctgctt 180 ccccaggagc agcggaccgt ggagtcgtca cttcgggcac aagtgccctt cgagcagatt 240 ctcagccttc cagagctcaa ggccaacccc ttcaaggagc gaatctgcag ggtcttctcc 300 acatccccag ccaaagacag ccttagcttt gaggacttcc tggatctcct cagtgtgttc 360 agtgacacag ccacgccaga catcaagtcc cattatgcct tccgcatctt tgactttgat 420 gatgacggaaccttgaacagagaagacctgagccggctggtgaactgcctcacgggagag480 ggcgaggacacacggcttagtgcgtctgagatgaagcagctcatcgacaacatcctggag540 gagtctgacattgacagggatggaaccatcaacctctctgagttccagcacgtcatctcc600 cgttctccagactttgccagctcctttaagattgtcctgtgacagcagccccagcgtgtg660 tcctggcaccctgtccaagaacctttctactgctgagctgtggccaaggtcaagcctgtg720 ttgccagtgcgggccaagctggcccagcctggagctggcgctgtgcagcctcaccccggg780 caggggcggccctcgttgtcagggcctctcctcactgctgttgtcattgctccgtttgtg840 tttgtactaatcagtaataaaggtttagaagtttgaccctaaaaaaaaaaaaaaaaaaaa900 aaaaa 905 <210> 17 <211> 191 <212> PRT
<213> human CIB1 protein <400> 17 Met Gly Gly Ser Gly Ser Arg Leu Ser Lys Glu Leu Leu Ala Glu Tyr l 5 10 15 G1n Asp Leu Thr Phe Leu Thr Lys Gln Glu Ile Leu Leu Ala His Arg Arg Phe Cys Glu Leu Leu Pro Gln Glu Gln Arg Thr Val Glu Ser Ser Leu Arg Ala Gln Val Pro Phe Glu Gln Ile Leu Ser Leu Pro Glu Leu Lys Ala Asn Pro Phe Lys Glu Arg Ile Cys Arg Val Phe Ser Thr Ser Pro Ala Lys Asp Ser Leu Ser Phe Glu Asp Phe Leu Asp Leu Leu Ser Val Phe Ser Asp Thr Ala Thr Pro Asp Ile Lys Ser His Tyr Ala Phe Arg Ile Phe Asp Phe Asp Asp Asp Gly Thr Leu Asn Arg G1u Asp Leu l15 120 125 Ser Arg Leu Val Asn Cys Leu Thr Gly Glu Gly Glu Asp Thr Arg Leu Ser Ala Ser Glu Met Lys Gln Leu Ile Asp Asn Ile Leu Glu Glu Ser Asp Ile Asp Arg Asp Gly Thr Ile Asn Leu Ser Glu Phe Gln His Val 165 l70 175 Tle Ser Arg Ser Pro Asp Phe A1a Ser Ser Phe Lys Ile Val Leu <210>

<211>

<212>
DNA

<213>
human Chk2 transcript variant 1 cDNA

<400>

cttacaaggtacagtcctctgctcaggggggccaggagggtcttataggcatcattcacc60 agggtcgaatgcttctctgagaagtccttttcagtctgagacctctggctgaagaaatct120 gggtggacaagacgctgcagttgctggtacctgtgctggagcttcgctgtatcaactctg180 aaggaacggttgcagtccataaggctgaagtagtctcgagtggggtcaggtgcctgcagc240 gctcggcactgtgggcagaagaacctgtcctcccgcccggggccccatgggccgccgcag300 ttccaacagcggggataattgcttcccgcctgcgacgcagcatcgcagcttagcggtctc360 cttctgggaacccctgtcggccaaaacccccacacccggagcaaagccccggctctcccc420 cgccacatctggccggcggcctatctagccgtggtcactcgtggggaaaagcaaagagag480 cgtctaaccagactaatgttgctgattggctggggagtcgagggggcgggatcacccgag540 gggaacccgggttctaagttccgctctcccttctaaactacaactcccaggaggcattga600 ggcggcgcctgacggccacatctgctgctcctcattggtccggcggcaggggagggggtt660 ttgattggctgagggtggagtttgtatctgcaggtttagcgccactctgctggctgaggc720 tgcggagagtgtgcggctccaggtgggctcacgcggtcgtgatgtctcgggagtcggatg780 ttgaggctcagcagtctcatggcagcagtgcctgttcacagccccatggcagcgttaccc840 agtcccaaggctcctcctcacagtcccagggcatatccagctcctctaccagcacgatgc900 caaactccagccagtcctctcactccagctctgggacactgagctccttagagacagtgt960 ccactcaggaactctattctattcctgaggaccaagaacctgaggaccaagaacctgagg1020 agcctacccctgccccctgggctcgattatgggcccttcaggatggatttgccaatcttg1080 aatgtgtgaatgacaactactggtttgggagggacaaaagctgtgaatattgctttgatg1140 aaccactgctgaaaagaacagataaataccgaacatacagcaagaaacactttcggattt1200 tcagggaagtgggtcctaaaaactcttacattgcatacatagaagatcacagtggcaatg1260 gaacctttgtaaatacagagcttgtagggaaaggaaaacgccgtcctttgaataacaatt1320 ctgaaattgcactgtcactaagcagaaataaagtttttgtcttttttgatctgactgtag1380 atgatcagtcagtttatcctaaggcattaagagatgaatacatcatgtcaaaaactcttg1440 gaagtggtgcctgtggagaggtaaagctggctttcgagaggaaaacatgtaagaaagtag1500 ccataaagatcatcagcaaaaggaagtttgctattggttcagcaagagaggcagacccag1560 ctctcaatgttgaaacagaaatagaaattttgaaaaagctaaatcatccttgcatcatca1620 agattaaaaacttttttgatgcagaagattattatattgttttggaattgatggaagggg1680 gagagctgtttgacaaagtggtggggaataaacgcctgaaagaagctacctgcaagctct1740 atttttaccagatgctcttggctgtgcagtaccttcatgaaaacggtattatacaccgtg1800 acttaaagccagagaatgttttactgtcatctcaagaagaggactgtcttataaagatta1860 ctgattttgggcactccaagattttgggagagacctctctcatgagaaccttatgtggaa1920 cccccacctacttggcgcctgaagttcttgtttctgttgggactgctgggtataaccgtg1980 ctgtggactgctggagtttaggagttattctttttatctgccttagtgggtatccacctt2040 tctctgagcataggactcaagtgtcactgaaggatcagatcaccagtggaaaatacaact2100 tcattcctgaagtctgggcagaagtctcagagaaagctctggaccttgtcaagaagttgt2160 tggtagtggatccaaaggcacgttttacgacagaagaagccttaagacacccgtggcttc2220 aggatgaagacatgaagagaaagtttcaagatcttctgtctgaggaaaatgaatccacag2280 ctctaccccaggttctagcccagccttctactagtcgaaagcggccccgtgaaggggaag2340 ccgagggtgccgagaccacaaagcgcccagctgtgtgtgctgctgtgttgtgaactccgt2400 ggtttgaacacgaaagaaatgtaccttctttcactctgtcatctttcttttctttgagtc2460 tgtttttttatagtttgtattttaattatgggaataattgctttttcacagtcactgatg2520 tacaattaaa aacctgatgg aacctgg 2547 <210> 19 <211> 543 <212> PRT
<213> human Chk2 transcript variant 1 protein <400> 19 Met Ser Arg Glu Ser Asp Val Glu Ala Gln Gln Ser His Gly Ser Ser Ala Cys Ser Gln Pro His Gly Ser Val Thr Gln Ser Gln Gly Ser Ser Ser Gln Ser Gln Gly Ile Ser Ser Ser Ser Thr Ser Thr Met Pro Asn Ser Ser Gln Ser Ser His Ser Ser Ser Gly Thr Leu Ser Ser Leu Glu Thr Val Ser Thr Gln Glu Leu Tyr Ser Ile Pro Glu Asp Gln Glu Pro Glu Asp Gln Glu Pro Glu Glu Pro Thr Pro Ala Pro Trp Ala Arg Leu Trp Ala Leu Gln Asp Gly Phe Ala Asn Leu Glu Cys Val Asn Asp Asn Tyr Trp Phe Gly Arg Asp Lys Ser Cys Glu Tyr Cys Phe Asp Glu Pro Leu Leu Lys Arg Thr Asp Lys Tyr Arg Thr Tyr Ser Lys Lys His Phe Arg Ile Phe Arg Glu Val Gly Pro Lys Asn Ser Tyr Ile Ala Tyr Ile Glu Asp His Ser Gly Asn Gly Thr Phe Val Asn Thr Glu Leu Val Gly Lys Gly Lys Arg Arg Pro Leu Asn Asn Asn Ser Glu Ile Ala Leu Ser Leu Ser Arg Asn Lys Val Phe Val Phe Phe Asp Leu Thr Val Asp Asp Gln Ser Val Tyr Pro Lys Ala Leu Arg Asp Glu Tyr Ile Met Ser Lys 210 2l5 220 Thr Leu Gly Ser Gly Ala Cys Gly Glu Val Lys Leu Ala Phe Glu Arg Lys Thr Cys Lys Lys Val Ala Ile Lys Ile Ile Ser Lys Arg Lys Phe Ala Tle Gly Ser Ala Arg Glu Ala Asp Pro Ala Leu Asn Val Glu Thr Glu Ile Glu Ile Leu Lys Lys Leu Asn His Pro Cys Ile Ile Lys Ile Lys Asn Phe Phe Asp Ala Glu Asp Tyr Tyr Ile Val Leu Glu Leu Met Glu Gly Gly Glu Leu Phe Asp Lys Val Val Gly Asn Lys Arg Leu Lys 305 3l0 315 320 Glu Ala Thr Cys Lys Leu Tyr Phe Tyr Gln Met Leu Leu Ala Val Gln Tyr Leu His Glu Asn Gly Ile Ile His Arg Asp Leu Lys Pro Glu Asn Val Leu Leu Ser Ser Gln Glu Glu Asp Cys Leu Ile Lys Ile Thr Asp Phe Gly His Ser Lys Ile Leu Gly Glu Thr Ser Leu Met Arg Thr Leu Cys Gly Thr Pro Thr Tyr Leu Ala Pro Glu Val Leu Val Ser Val Gly Thr Ala Gly Tyr Asn Arg Ala Val Asp Cys Trp Ser Leu Gly Val Ile Leu Phe Ile Cys Leu Ser Gly Tyr Pro Pro Phe 5er Glu His Arg Thr Gln Val Ser Leu Lys Asp Gln Ile Thr Ser Gly Lys Tyr Asn Phe Ile Pro Glu Val Trp Ala Glu Val Ser Glu Lys Ala Leu Asp Leu Val Lys Lys Leu Leu Val Val Asp Pro Lys Ala Arg Phe Thr Thr Glu Glu Ala Leu Arg His Pro Trp Leu Gln Asp Glu Asp Met Lys Arg Lys Phe Gln Asp Leu Leu Ser Glu Glu Asn Glu Ser Thr A1a Leu Pro Gln Val Leu Ala Gln Pro Ser Thr Ser Arg Lys Arg Pro Arg Glu Gly Glu Ala Glu Gly Ala Glu Thr Thr Lys Arg Pro Ala Val Cys Ala Ala Val Leu <210> 20 <211> 2460 ' <212> DNA
<213> human Chk2 transcript variant 2 cDNA
<400>

cttacaaggtacagtcctctgctcaggggggccaggagggtcttataggcatcattcacc60 agggtcgaatgcttctctgagaagtccttttcagtctgagacctctggctgaagaaatct120 gggtggacaagacgctgcagttgctggtacctgtgctggagcttcgctgtatcaactctg180 aaggaacggttgcagtccataaggctgaagtagtctcgagtggggtcaggtgcctgcagc240 gctcggcactgtgggcagaagaacctgtcctcccgcccggggccccatgggccgccgcag300 ttccaacagcggggataattgcttcccgcctgcgacgcagcatcgcagcttagcggtctc360 cttctgggaacccctgtcggccaaaacccccacacccggagcaaagccccggctctcccc420 cgccacatctggccggcggcctatctagccgtggtcactcgtggggaaaagcaaagagag480 cgtctaaccagactaatgttgctgattggctggggagtcgagggggcgggatcacccgag540 gggaacccgggttctaagttccgctctcccttctaaactacaactcccaggaggcattga600 ggcggcgcctgacggccacatctgctgctcctcattggtccggcggcaggggagggggtt660 ttgattggctgagggtggagtttgtatctgcaggtttagcgccactctgctggctgaggc720 tgcggagagtgtgcggctccaggtgggctcacgcggtcgtgatgtctcgggagtcggatg780 ttgaggctcagcagtctcatggcagcagtgcctgttcacagccccatggcagcgttaccc840 agtcccaaggctcctcctcacagtcccagggcatatccagctcctctaccagcacgatgc900 caaactccagccagtcctctcactccagctctgggacactgagctccttagagacagtgt960 ccactcaggaactctattctattcctgaggaccaagaacctgaggaccaagaacctgagg1020 agcctacccctgccccctgggctcgattatgggcccttcaggatggatttgccaatcttg1080 aatgtgtgaatgacaactactggtttgggagggacaaaagctgtgaatattgctttgatg1140 aaccactgctgaaaagaacagataaataccgaacatacagcaagaaacactttcggattt1200 tcagggaagtgggtcctaaaaactcttacattgcatacatagaagatcacagtggcaatg1260 gaacctttgtaaatacagagcttgtagggaaaggaaaacgccgtcctttgaataacaatt1320 ctgaaattgcactgtcactaagcagaaataaagtttttgtcttttttgatctgactgtag1380 atgatcagtcagtttatcctaaggcattaagagatgaatacatcatgtcaaaaactcttg1440 gaagtggtgcctgtggagaggtaaagctggctttcgagaggaaaacatgtaagaaagtag1500 ccataaagatcatcagcaaaaggaagtttgctattggttcagcaagagaggcagacccag1560 ctctcaatgttgaaacagaaatagaaattttgaaaaagctaaatcatccttgcatcatca1620 agattaaaaacttttttgatgcagaagattattatattgttttggaattgatggaagggg1680 gagagctgtttgacaaagtggtggggaataaacgcctgaaagaagctacctgcaagctct1740 atttttaccagatgctcttggctgtgcagattactgattttgggcactccaagattttgg1800 gagagacctctctcatgagaaccttatgtggaacccccacctacttggcgcctgaagttc1860 ttgtttctgttgggactgctgggtataaccgtgctgtggactgctggagtttaggagtta1920 ttctttttatctgccttagtgggtatccacctttctctgagcataggactcaagtgtcac1980 tgaaggatcagatcaccagtggaaaatacaacttcattcctgaagtctgggcagaagtct2040 cagagaaagctctggaccttgtcaagaagttgttggtagtggatccaaaggcacgtttta2100 cgacagaagaagccttaagacacccgtggcttcaggatgaagacatgaagagaaagtttc2160 aagatcttctgtctgaggaaaatgaatccacagctctaccccaggttctagcccagcctt2220 ctactagtcgaaagcggccccgtgaaggggaagccgagggtgccgagaccacaaagcgcc2280 cagctgtgtgtgctgctgtgttgtgaactccgtggtttgaacacgaaagaaatgtacctt2340 ctttcactctgtcatctttcttttctttgagtctgtttttttatagtttgtattttaatt2400 atgggaataattgctttttcacagtcactgatgtacaattaaaaacctgatggaacctgg2460 <210> 21 <211> 514 <212> PRT
<213> human Chk2 transcript variant 2 protein <400> 21 Met Ser Arg Glu Ser Asp Val Glu Ala Gln Gln Ser His Gly Ser Ser Ala Cys Ser Gln Pro His Gly Ser Val Thr Gln Ser Gln Gly Ser Ser Ser Gln Ser Gln Gly Ile Ser Ser Ser Ser Thr Ser Thr Met Pro Asn Ser Ser Gln Ser Ser His Ser Ser 5er Gly Thr Zeu Ser Ser Leu Glu Thr Val Ser Thr Gln Glu Leu Tyr Ser Ile Pro Glu Asp Gln Glu Pro Glu Asp Gln Glu Pro Glu Glu Pro Thr Pro Ala Pro Trp Ala Arg Leu Trp Ala Leu Gln Asp Gly Phe Ala Asn Leu Glu Cys Val Asn Asp Asn Tyr Trp Phe Gly Arg Asp Lys Ser Cys Glu Tyr Cys Phe Asp Glu Pro Leu Leu Lys Arg Thr Asp Lys Tyr Arg Thr Tyr Ser Lys Lys His Phe Arg Tle Phe Arg Glu Val Gly Pro Lys Asn Ser Tyr Ile Ala Tyr Ile Glu Asp His Ser Gly Asn Gly Thr Phe Val Asn Thr Glu Leu Val Gly Lys Gly Lys Arg Arg Pro Leu Asn Asn Asn Ser Glu Ile Ala Leu Ser Leu Ser Arg Asn Lys Val Phe Val Phe Phe Asp Leu Thr Val Asp Asp Gln Ser Val Tyr Pro Lys Ala Leu Arg Asp Glu Tyr Ile Met Ser Lys Thr Leu Gly Ser Gly Ala Cys Gly Glu Val Lys Leu Ala Phe Glu Arg Lys Thr Cys Lys Lys Val Ala Ile Lys Ile Ile Ser Lys Arg Lys Phe Ala I1e Gly Ser Ala Arg Glu Ala Asp Pro Ala Leu Asn Val Glu Thr Glu Ile Glu Ile Leu Lys Lys Leu Asn His Pro Cys Ile I1e Lys Ile Lys Asn Phe Phe Asp Ala Glu Asp Tyr Tyr Ile Val Leu Glu Leu Met Glu Gly Gly Glu Leu Phe Asp Lys Val Val Gly Asn Lys Arg Leu Lys Glu Ala Thr Cys Lys Leu Tyr Phe Tyr Gln Met Leu Leu Ala Val Gln Ile Thr Asp Phe Gly His Ser Lys Ile Leu Gly Glu Thr Ser Leu Met Arg Thr Leu Cys Gly Thr Pro Thr Tyr Leu Ala Pro Glu Val Leu Val Ser Val Gly Thr Ala Gly Tyr Asn Arg Ala Val Asp Cys Trp Ser Leu Gly Val Ile Leu Phe Ile Cys Leu Ser Gly Tyr Pro Pro Phe Ser Glu His Arg Thr G1n Val Ser Leu Lys Asp Gln Ile Thr Ser Gly Lys Tyr Asn Phe Ile Pro Glu Val Trp Ala Glu Val Ser Glu Lys Ala Leu Asp Leu Va1 Lys Lys Leu Leu Val Val Asp Pro Lys Ala Arg Phe Thr Thr Glu Glu Ala Leu Arg His Pro Trp Leu Gln Asp Glu Asp Met Lys Arg Lys Phe Gln Asp Leu Leu Ser Glu Glu Asn Glu Ser Thr Ala Leu Pro Gln Val Leu Ala Gln Pro Ser Thr Ser Arg Lys Arg Pro Arg Glu Gly Glu Ala Glu Gly Ala Glu Thr Thr Lys Arg Pro Ala Val Cys Ala Ala Val Leu <210> 22 <211> 10987 <212> DNA
<213> human BRCA2 cDNA

<400>

ggtggcgcgagcttctgaaactaggcggcagaggcggagccgctgtggcactgctgcgcc60 tctgctgcgcctcgggtgtcttttgcggcggtgggtcgccgccgggagaagcgtgagggg120 acagatttgtgaccggcgcggtttttgtcagcttactccggccaaaaaagaactgcacct180 ctggagcggacttatttaccaagcattggaggaatatcgtaggtaaaaatgcctattgga240 tccaaagagaggccaacattttttgaaatttttaagacacgctgcaacaaagcagattta300 ggaccaataagtcttaattggtttgaagaactttcttcagaagctccaccctataattct360 gaacctgcagaagaatctgaacataaaaacaacaattacgaaccaaacctatttaaaact420 ccacaaaggaaaccatcttataatcagctggcttcaactccaataatattcaaagagcaa480 gggctgactctgccgctgtaccaatctcctgtaaaagaattagataaattcaaattagac540 ttaggaaggaatgttcccaatagtagacataaaagtcttcgcacagtgaaaactaaaatg600 gatcaagcagatgatgtttcctgtccacttctaaattcttgtcttagtgaaagtcctgtt660 gttctacaatgtacacatgtaacaccacaaagagataagtcagtggtatgtgggagtttg720 tttcatacaccaaagtttgtgaagggtcgtcagacaccaaaacatatttctgaaagtcta780 ggagctgaggtggatcctgatatgtcttggtcaagttctttagctacaccacccaccctt840 agttctactgtgctcatagtcagaaatgaagaagcatctgaaactgtatttcctcatgat900 actactgctaatgtgaaaagctatttttccaatcatgatgaaagtctgaagaaaaatgat960 agatttatcgcttctgtgacagacagtgaaaacacaaatcaaagagaagctgcaagtcat1020 ggatttggaaaaacatcagggaattcatttaaagtaaatagctgcaaagaccacattgga1080 aagtcaatgccaaatgtcctagaagatgaagtatatgaaacagttgtagatacctctgaa1140 gaagatagtttttcattatgtttttctaaatgtagaacaaaaaatctacaaaaagtaaga1200 actagcaagactaggaaaaaaattttccatgaagcaaacgctgatgaatgtgaaaaatct1260 aaaaaccaagtgaaagaaaaatactcatttgtatctgaagtggaaccaaatgatactgat1320 ccattagattcaaatgtagcacatcagaagccctttgagagtggaagtgacaaaatctcc1380 aaggaagttgtaccgtctttggcctgtgaatggtctcaactaaccctttcaggtctaaat1440 ggagcccagatggagaaaatacccctattgcatatttcttcatgtgaccaaaatatttca1500 gaaaaagacctattagacacagagaacaaaagaaagaaagattttcttacttcagagaat1560 tctttgccacgtatttctagcctaccaaaatcagagaagccattaaatgaggaaacagtg1620 gtaaataagagagatgaagagcagcatcttgaatctcatacagactgcattcttgcagta1680 aagcaggcaatatctggaacttctccagtggcttcttcatttcagggtatcaaaaagtct1740 atattcagaataagagaatcacctaaagagactttcaatgcaagtttttcaggtcatatg1800 actgatccaaactttaaaaaagaaactgaagcctctgaaagtggactggaaatacatact1860 gtttgctcacagaaggaggactccttatgtccaaatttaattgataatggaagctggcca1920 gccaccaccacacagaattctgtagctttgaagaatgcaggtttaatatccactttgaaa1980 aagaaaacaaataagtttatttatgctatacatgatgaaacattttataaaggaaaaaaa2040 ataccgaaagaccaaaaatcagaactaattaactgttcagcccagtttgaagcaaatgct2100 tttgaagcaccacttacatttgcaaatgctgattcaggtttattgcattcttctgtgaaa2160 agaagctgttcacagaatgattctgaagaaccaactttgtccttaactagctcttttggg2220 acaattctgaggaaatgttctagaaatgaaacatgttctaataatacagtaatctctcag2280 gatcttgattataaagaagcaaaatgtaataaggaaaaactacagttatttattacccca2340 gaagctgattctctgtcatgcctgcaggaaggacagtgtgaaaatgatccaaaaagcaaa2400 aaagtttcagatataaaagaagaggtcttggctgcagcatgtcacccagtacaacattca2460 aaagtggaatacagtgatactgactttcaatcccagaaaagtcttttatatgatcatgaa2520 aatgccagcactcttattttaactcctacttccaaggatgttctgtcaaacctagtcatg2580 atttctagaggcaaagaatcatacaaaatgtcagacaagctcaaaggtaacaattatgaa2640 tctgatgttgaattaaccaaaaatattcccatggaaaagaatcaagatgtatgtgcttta2700 aatgaaaattataaaaacgttgagctgttgccacctgaaaaatacatgagagtagcatca2760 ccttcaagaaaggtacaattcaaccaaaacacaaatctaagagtaatccaaaaaaatcaa2820 gaagaaactacttcaatttcaaaaataactgtcaatccagactctgaagaacttttctca2880 gacaatgagaataattttgtcttccaagtagctaatgaaaggaataatcttgctttagga2940 aatactaaggaacttcatgaaacagacttgacttgtgtaaacgaacccattttcaagaac3000 tctaccatggttttatatggagacacaggtgataaacaagcaacccaagtgtcaattaaa3060 aaagatttggtttatgttcttgcagaggagaacaaaaatagtgtaaagcagcatataaaa3120 atgactctaggtcaagatttaaaatcggacatctccttgaatatagataaaataccagaa3180 aaaaataatgattacatgaacaaatgggcaggactcttaggtccaatttcaaatcacagt3240 tttggaggtagcttcagaacagcttcaaataaggaaatcaagctctctgaacataacatt3300 aagaagagcaaaatgttcttcaaagatattgaagaacaatatcctactagtttagcttgt3360 gttgaaattgtaaataccttggcattagataatcaaaagaaactgagcaagcctcagtca3420 attaatactgtatctgcacatttacagagtagtgtagttgtttctgattgtaaaaatagt3480 catataacccctcagatgttattttccaagcaggattttaattcaaaccataatttaaca3540 cctagccaaaaggcagaaattacagaactttctactatattagaagaatcaggaagtcag3600 tttgaatttactcagtttagaaaaccaagctacatattgcagaagagtacatttgaagtg3660 cctgaaaaccagatgactatcttaaagaccacttctgaggaatgcagagatgctgatctt3720 catgtcataatgaatgccccatcgattggtcaggtagacagcagcaagcaatttgaaggt3780 acagttgaaattaaacggaagtttgctggcctgttgaaaaatgactgtaacaaaagtgct3840 tctggttatttaacagatgaaaatgaagtggggtttaggggcttttattctgctcatggc3900 acaaaactgaatgtttctactgaagctctgcaaaaagctgtgaaactgtttagtgatatt3960 gagaatattagtgaggaaacttctgcagaggtacatccaataagtttatcttcaagtaaa4020 tgtcatgattctgttgtttcaatgtttaagatagaaaatcataatgataaaactgtaagt4080 gaaaaaaataataaatgccaactgatattacaaaataatattgaaatgactactggcact4140 tttgttgaagaaattactgaaaattacaagagaaatactgaaaatgaagataacaaatat4200 actgctgccagtagaaattctcataacttagaatttgatggcagtgattcaagtaaaaat4260 gatactgtttgtattcataaagatgaaacggacttgctatttactgatcagcacaacata4320 tgtcttaaattatctggccagtttatgaaggagggaaacactcagattaaagaagatttg4380 tcagatttaacttttttggaagttgcgaaagctcaagaagcatgtcatggtaatacttca4440 aataaagaacagttaactgctactaaaacggagcaaaatataaaagattttgagacttct4500 gatacattttttcagactgcaagtgggaaaaatattagtgtcgccaaagagtcatttaat4560 aaaattgtaaatttctttgatcagaaaccagaagaattgcataacttttccttaaattct4620 gaattacattctgacataagaaagaacaaaatggacattctaagttatgaggaaacagac4680 atagttaaacacaaaatactgaaagaaagtgtcccagttggtactggaaatcaactagtg4740 accttccagggacaacccgaacgtgatgaaaagatcaaagaacctactctgttgggtttt4800 catacagctagcgggaaaaaagttaaaattgcaaaggaatctttggacaaagtgaaaaac4860 ctttttgatgaaaaagagcaaggtactagtgaaatcaccagttttagccatcaatgggca4920 aagaccctaaagtacagagaggcctgtaaagaccttgaattagcatgtgagaccattgag4980 atcacagctgccccaaagtgtaaagaaatgcagaattctctcaataatgataaaaacctt5040 gtttctattgagactgtggtgccacctaagctcttaagtgataatttatgtagacaaact5100 gaaaatctcaaaacatcaaaaagtatctttttgaaagttaaagtacatgaaaatgtagaa5160 aaagaaacagcaaaaagtcctgcaacttgttacacaaatcagtccccttattcagtcatt5220 gaaaattcagccttagctttttacacaagttgtagtagaaaaacttctgtgagtcagact5280 tcattacttgaagcaaaaaaatggcttagagaaggaatatttgatggtcaaccagaaaga5340 ataaatactgcagattatgtaggaaattatttgtatgaaaataattcaaacagtactata5400 gctgaaaatgacaaaaatcatctctccgaaaaacaagatacttatttaagtaacagtagc5460 atgtctaacagctattcctaccattctgatgaggtatataatgattcaggatatctctca5520 aaaaataaacttgattctggtattgagccagtattgaagaatgttgaagatcaaaaaaac5580 actagtttttccaaagtaatatccaatgtaaaagatgcaaatgcatacccacaaactgta5640 aatgaagatatttgcgttgaggaacttgtgactagctcttcaccctgcaaaaataaaaat5700 gcagccattaaattgtccatatctaatagtaataattttgaggtagggccacctgcattt5760 aggatagccagtggtaaaatcgtttgtgtttcacatgaaacaattaaaaaagtgaaagac5820 atatttacagacagtttcagtaaagtaattaaggaaaacaacgagaataaatcaaaaatt5880 tgccaaacgaaaattatggcaggttgttacgaggcattggatgattcagaggatattctt5940 cataactctctagataatgatgaatgtagcacgcattcacataaggtttttgctgacatt6000 cagagtgaagaaattttacaacataaccaaaatatgtctggattggagaaagtttctaaa6060 atatcaccttgtgatgttagtttggaaacttcagatatatgtaaatgtagtatagggaag6120 cttcataagtcagtctcatctgcaaatacttgtgggatttttagcacagcaagtggaaaa6180 tctgtccaggtatcagatgcttcattacaaaacgcaagacaagtgttttctgaaatagaa6240 gatagtaccaagcaagtcttttccaaagtattgtttaaaagtaacgaacattcagaccag6300 ctcacaagagaagaaaatactgctatacgtactccagaacatttaatatcccaaaaaggc6360 ttttcatataatgtggtaaattcatctgctttctctggatttagtacagcaagtggaaag6420 caagtttccattttagaaagttccttacacaaagttaagggagtgttagaggaatttgat6480 ttaatcagaactgagcatagtcttcactattcacctacgtctagacaaaatgtatcaaaa6540 atacttcctcgtgttgataagagaaacccagagcactgtgtaaactcagaaatggaaaaa6600 acctgcagtaaagaatttaaattatcaaataacttaaatgttgaaggtggttcttcagaa6660 aataatcactctattaaagtttctccatatctctctcaatttcaacaagacaaacaacag6720 ttggtattaggaaccaaagtctcacttgttgagaacattcatgttttgggaaaagaacag6780 gcttcacctaaaaacgtaaaaatggaaattggtaaaactgaaactttttctgatgttcct6840 gtgaaaacaaatatagaagtttgttctacttactccaaagattcagaaaactactttgaa6900 acagaagcag tagaaattgc taaagctttt atggaagatg atgaactgac agattctaaa 6960 ctgccaagtc atgccacaca ttctcttttt acatgtcccg aaaatgagga aatggttttg 7020 tcaaattcaagaattggaaaaagaagaggagagccccttatcttagtgggagaaccctca7080 atcaaaagaaacttattaaatgaatttgacaggataatagaaaatcaagaaaaatcctta7140 aaggcttcaaaaagcactccagatggcacaataaaagatcgaagattgtttatgcatcat7200 gtttctttagagccgattacctgtgtaccctttcgcacaactaaggaacgtcaagagata7260 cagaatccaaattttaccgcacctggtcaagaatttctgtctaaatctcatttgtatgaa7320 catctgactttggaaaaatcttcaagcaatttagcagtttcaggacatccattttatcaa7380 gtttctgctacaagaaatgaaaaaatgagacacttgattactacaggcagaccaaccaaa7440 gtctttgttccaccttttaaaactaaatcacattttcacagagttgaacagtgtgttagg7500 aatattaacttggaggaaaacagacaaaagcaaaacattgatggacatggctctgatgat7560 agtaaaaataagattaatgacaatgagattcatcagtttaacaaaaacaactccaatcaa7620 gcagcagctgtaactttcacaaagtgtgaagaagaacctttagatttaattacaagtctt7680 cagaatgccagagatatacaggatatgcgaattaagaagaaacaaaggcaacgcgtcttt7740 ccacagccaggcagtctgtatcttgcaaaaacatccactctgcctcgaatctctctgaaa7800 gcagcagtaggaggccaagttccctctgcgtgttctcataaacagctgtatacgtatggc7860 gtttctaaacattgcataaaaattaacagcaaaaatgcagagtcttttcagtttcacact7920 gaagattattttggtaaggaaagtttatggactggaaaaggaatacagttggctgatggt7980 ggatggctcataccctccaatgatggaaaggctggaaaagaagaattttatagggctctg8040 tgtgacactccaggtgtggatccaaagcttatttctagaatttgggtttataatcactat8100 agatggatcatatggaaactggcagctatggaatgtgcctttcctaaggaatttgctaat8160 agatgcctaagcccagaaagggtgcttcttcaactaaaatacagatatgatacggaaatt8220 gatagaagcagaagatcggctataaaaaagataatggaaagggatgacacagctgcaaaa8280 acacttgttctctgtgtttctgacataatttcattgagcgcaaatatatctgaaacttct8340 agcaataaaactagtagtgcagatacccaaaaagtggccattattgaacttacagatggg8400 tggtatgctgttaaggcccagttagatcctcccctcttagctgtcttaaagaatggcaga8460 ctgacagttggtcagaagattattcttcatggagcagaactggtgggctctcctgatgcc8520 tgtacacctcttgaagccccagaatctcttatgttaaagatttctgctaacagtactcgg8580 cctgctcgctggtataccaaacttggattctttcctgaccctagaccttttcctctgccc8640 ttatcatcgcttttcagtgatggaggaaatgttggttgtgttgatgtaattattcaaaga8700 gcataccctatacagtggatggagaagacatcatctggattatacatatttcgcaatgaa8760 agagaggaagaaaaggaagcagcaaaatatgtggaggcccaacaaaagagactagaagcc8820 ttattcactaaaattcaggaggaatttgaagaacatgaagaaaacacaacaaaaccatat8880 ttaccatcacgtgcactaacaagacagcaagttcgtgctttgcaagatggtgcagagctt8940 tatgaagcagtgaagaatgcagcagacccagcttaccttgagggttatttcagtgaagag9000 cagttaagagccttgaataatcacaggcaaatgttgaatgataagaaacaagctcagatc9060 cagttggaaattaggaaggccatggaatctgctgaacaaaaggaacaaggtttatcaagg9120 gatgtcacaa ccgtgtggaa gttgcgtatt gtaagctatt caaaaaaaga aaaagattca 9180 gttatactga gtatttggcg tccatcatca gatttatatt ctctgttaac agaaggaaag 9240 agatacagaa tttatcatct tgcaacttca aaatctaaaa gtaaatctga aagagctaac 9300 atacagttag cagcgacaaa aaaaactcag tatcaacaac taccggtttc agatgaaatt 9360 ttatttcaga tttaccagcc acgggagccc cttcacttca gcaaattttt agatccagac 9420 tttcagccat cttgttctga ggtggaccta ataggatttg tcgtttctgt tgtgaaaaaa 9480 acaggacttg cccctttcgt ctatttgtca gacgaatgtt acaatttact ggcaataaag 9540 ttttggatag accttaatga ggacattatt aagcctcata tgttaattgc tgcaagcaac 9600 ctccagtggc gaccagaatc caaatcaggc cttcttactt tatttgctgg agatttttct 9660 gtgttttctg ctagtccaaa agagggccac tttcaagaga cattcaacaa aatgaaaaat 9720 actgttgaga atattgacat actttgcaat gaagcagaaa acaagcttat gcatatactg 9780 catgcaaatg atcccaagtg gtccacccca actaaagact gtacttcagg gccgtacact 9840 gctcaaatca ttcctggtac aggaaacaag cttctgatgt cttctcctaa ttgtgagata 9900 tattatcaaa gtcctttatc actttgtatg gccaaaagga agtctgtttc cacacctgtc 9960 tcagcccaga tgacttcaaa gtcttgtaaa ggggagaaag agattgatga ccaaaagaac 10020 tgcaaaaaga gaagagcctt ggatttcttg agtagactgc ctttacctcc acctgttagt 10080 cccatttgta catttgtttc tccggctgca cagaaggcat ttcagccacc aaggagttgt 10140 ggcaccaaat acgaaacacc cataaagaaa aaagaactga attctcctca gatgactcca 10200 tttaaaaaat tcaatgaaat ttctcttttg gaaagtaatt caatagctga cgaagaactt 10260 gcattgataa atacccaagc tcttttgtct ggttcaacag gagaaaaaca atttatatct 10320 gtcagtgaat ccactaggac tgctcccacc agttcagaag attatctcag actgaaacga 10380 cgttgtacta catctctgat caaagaacag gagagttccc aggccagtac ggaagaatgt 10440 gagaaaaata agcaggacac aattacaact aaaaaatata tctaagcatt tgcaaaggcg 10500 acaataaatt attgacgctt aacctttcca gtttataaga ctggaatata atttcaaacc 10560 acacattagt acttatgttg cacaatgaga aaagaaatta gtttcaaatt tacctcagcg 10620 tttgtgtatc gggcaaaaat cgttttgccc gattccgtat tggtatactt ttgcttcagt 10680 tgcatatctt aaaactaaat gtaatttatt aactaatcaa gaaaaacatc tttggctgag 10740 ctcggtggct catgcctgta atcccaacac tttgagaagc tgaggtggga ggagtgcttg 10800 aggccaggag ttcaagacca gcctgggcaa catagggaga cccccatctt tacgaagaaa 10860 aaaaaaaagg ggaaaagaaa atcttttaaa tctttggatt tgatcactac aagtattatt 10920 ttacaatcaa caaaatggtc atccaaactc aaacttgaga aaatatcttg ctttcaaatt 10980 gacacta 10987 <210> 23 <21l> 3418 <212> PRT
<213> human BRCA2 protein <400> 23 Met Pro Ile Gly Ser Lys Glu Arg Pro Thr Phe Phe Glu Tle Phe Lys Thr Arg Cys Asn Lys Ala Asp Leu Gly Pro Ile Ser Leu Asn Trp Phe Glu Glu Leu Ser Ser Glu Ala Pro Pro Tyr Asn Ser Glu Pro Ala Glu Glu Ser Glu His Lys Asn Asn Asn Tyr Glu Pro Asn Leu Phe Lys Thr Pro Gln Arg Lys Pro Ser Tyr Asn Gln Leu Ala Ser Thr Pro Ile Ile Phe Lys Glu Gln Gly Leu Thr Leu Pro Leu Tyr Gln Ser Pro Val Lys Glu Leu Asp Lys Phe Lys Leu Asp Leu Gly Arg Asn Val Pro Asn Ser Arg His Lys Ser Leu Arg Thr Val Lys Thr Lys Met Asp Gln Ala Asp Asp Val Ser Cys Pro Leu Leu Asn Ser Cys Leu Ser Glu Ser Pro Val Val Leu Gln Cys Thr His Val Thr Pro Gln Arg Asp Lys Ser Val Val Cys Gly Ser Leu Phe His Thr Pro Lys Phe Val Lys Gly Arg Gln Thr Pro Lys His Ile Ser Glu Ser Leu Gly Ala Glu Val Asp Pro Asp Met Ser Trp Ser Ser Ser Leu Ala Thr Pro Pro Thr Leu Ser Ser Thr Val Leu Ile Val Arg Asn Glu Glu A1a Ser Glu Thr Val Phe Pro His Asp Thr Thr Ala Asn Val Lys Ser Tyr Phe Ser Asn His Asp Glu 5er Leu Lys Lys Asn Asp Arg Phe Ile Ala Ser Val Thr Asp Ser Glu Asn Thr Asn Gln Arg Glu Ala Ala Ser His Gly Phe Gly Lys Thr Ser Gly Asn Ser Phe Lys Val Asn Ser Cys Lys Asp His Ile Gly Lys Ser Met Pro Asn Val Leu Glu Asp Glu Val Tyr G1u Thr Val Val Asp Thr Ser Glu Glu Asp Ser Phe Ser Leu Cys Phe Ser Lys Cys Arg Thr Lys Asn Leu Gln Lys Val Arg Thr Ser Lys Thr Arg Lys Lys Ile Phe His Glu Ala Asn Ala Asp Glu Cys Glu Lys Ser Lys Asn Gln Val Lys Glu Lys Tyr Ser Phe Val Ser Glu Val Glu Pro Asn Asp Thr Asp Pro Leu Asp Ser Asn Val Ala His Gln Lys Pro Phe Glu Ser Gly Ser Asp Lys Ile Ser Lys Glu Val Val Pro Ser Leu Ala Cys Glu Trp Ser Gln Leu Thr Leu Ser Gly Leu Asn Gly Ala Gln Met Glu Lys Ile Pro Leu Leu His Ile Ser Ser Cys Asp Gln Asn Ile Ser Glu Lys Asp Leu Leu Asp Thr Glu Asn Lys Arg Lys Lys Asp Phe Leu Thr Ser Glu Asn Ser Leu Pro Arg Tle Ser Ser Leu Pro Lys Ser Glu Lys Pro Leu Asn Glu Glu Thr Val Val Asn Lys Arg Asp Glu Glu Gln His Leu Glu Ser His Thr Asp Cys Ile Leu Ala Val Lys Gln Ala Tle Ser Gly Thr Ser Pro Val Ala Ser Ser Phe Gln Gly Tle Lys Lys Ser Tle Phe Arg Ile Arg Glu Ser Pro Lys Glu Thr Phe Asn Ala Ser Phe Ser Gly His Met Thr Asp Pro Asn Phe Lys Lys G1u Thr Glu Ala Ser G1u Ser Gly Leu Glu Ile His Thr Val Cys Ser Gln Lys Glu Asp Ser Leu Cys Pro Asn Leu Ile Asp Asn Gly Ser Trp Pro Ala Thr Thr Thr Gln Asn Ser Val Ala Leu Lys Asn Ala Gly Leu Tle Ser Thr Leu Lys Lys Lys Thr Asn Lys Phe Ile Tyr Ala Ile His Asp Glu Thr Phe Tyr Lys Gly Lys Lys Ile Pro Lys Asp Gln Lys Ser Glu Leu Tle Asn Cys Ser Ala Gln Phe Glu Ala Asn A1a Phe Glu Ala Pro Leu Thr Phe Ala Asn A1a Asp Ser Gly Leu Leu His Ser Ser Val Lys Arg Ser Cys Ser Gln Asn Asp Ser Glu Glu Pro Thr Leu Ser Leu Thr Ser Ser Phe Gly Thr Ile Leu Arg Lys Cys Ser Arg Asn Glu Thr Cys Ser Asn Asn Thr Val Ile Ser Gln Asp Leu Asp Tyr _78_ Lys Glu Ala Lys Cys Asn Lys Glu Lys Leu Gln Leu Phe Ile Thr Pro Glu Ala Asp Ser Leu Ser Cys Leu Gln Glu Gly Gln Cys Glu Asn Asp Pro Lys Ser Lys Lys Val Ser Asp Ile Lys Glu Glu Val Leu Ala Ala Ala Cys His Pro Val Gln His Ser Lys Val Glu Tyr Ser Asp Thr Asp Phe Gln Ser Gln Lys Ser Leu Leu Tyr Asp His Glu Asn Ala Ser Thr Leu Ile Leu Thr Pro Thr Ser Lys Asp Val Leu Ser Asn Leu Val Met Ile Ser Arg Gly Lys Glu Ser Tyr Lys Met Ser Asp Lys Leu Lys Gly Asn Asn Tyr Glu Ser Asp Val Glu Leu Thr Lys Asn Ile Pro Met Glu Lys Asn Gln Asp Val Cys Ala Leu Asn Glu Asn Tyr Lys Asn Val Glu Leu Leu Pro Pro Glu Lys Tyr Met Arg Val Ala Ser Pro Ser Arg Lys Val Gln Phe Asn Gln Asn Thr Asn Leu Arg Val Ile Gln Lys Asn Gln Glu Glu Thr Thr Ser Ile Ser Lys Ile Thr Val Asn Pro Asp Ser Glu Glu Leu Phe Ser Asp Asn Glu Asn Asn Phe Val Phe Gln Val Ala Asn Glu Arg Asn Asn Leu Ala Leu Gly Asn Thr Lys Glu Leu His Glu Thr Asp Leu Thr Cys Val Asn Glu Pro Ile Phe Lys Asn Ser Thr Met Val Leu Tyr Gly Asp Thr Gly Asp Lys Gln Ala Thr Gln Val Ser Ile Lys Lys Asp Leu Val Tyr Val Leu Ala Glu Glu Asn Lys Asn Ser Val Lys Gln His Ile Lys Met Thr Leu Gly Gln Asp Leu Lys Ser Asp Ile Ser Leu Asn Ile Asp Lys Ile Pro Glu Lys Asn Asn Asp Tyr Met Asn Lys Trp Ala Gly Leu Leu Gly Pro Ile Ser Asn His Ser Phe Gly Gly Ser Phe Arg Thr Ala Ser Asn Lys Glu Ile Lys Leu Ser G1u His Asn Ile Lys Lys Ser Lys Met Phe Phe Lys Asp Ile Glu Glu Gln Tyr Pro Thr Ser Leu Ala Cys Val Glu Ile Val Asn Thr Leu Ala Leu Asp Asn Gln Lys Lys Leu Ser Lys Pro Gln Ser Ile Asn Thr Val Ser Ala His Leu Gln Ser Ser Val Val Val Ser Asp Cys Lys Asn Ser His Ile Thr Pro Gln Met Leu Phe Ser Lys Gln Asp Phe Asn Ser Asn His Asn Leu Thr Pro Ser Gln Lys Ala Glu Ile Thr Glu Leu Ser Thr Ile Leu Glu Glu Ser Gly Ser Gln Phe Glu Phe Thr Gln Phe Arg Lys Pro Ser Tyr Ile Leu Gln Lys Ser Thr Phe Glu Val Pro Glu Asn Gln Met Thr Ile Leu Lys Thr Thr Ser Glu Glu Cys Arg Asp Ala Asp Leu His Val Tle Met Asn Ala Pro Ser Ile Gly Gln Val Asp Ser Ser Lys Gln Phe Glu Gly Thr Val Glu Ile Lys Arg Lys Phe Ala Gly Leu Leu Lys Asn Asp Cys Asn Lys Ser Ala Ser Gly Tyr Leu Thr Asp Glu Asn Glu Val Gly Phe Arg Gly Phe Tyr Ser Ala His Gly Thr Lys Leu Asn Val Ser Thr Glu Ala Leu Gln Lys Ala Val Lys Leu Phe Ser Asp Ile Glu Asn Ile Ser Glu Glu Thr Ser Ala Glu Val His Pro Ile Ser Leu Ser Ser Ser Lys Cys His Asp Ser Val Val Ser Met Phe Lys Ile Glu Asn His Asn Asp Lys Thr Val Ser Glu Lys Asn Asn Lys Cys Gln Leu Ile Leu Gln Asn Asn Ile Glu Met Thr Thr Gly Thr Phe Val Glu Glu Ile Thr Glu Asn Tyr Lys Arg Asn Thr Glu Asn Glu Asp Asn Lys Tyr Thr Ala Ala Ser Arg Asn Ser His Asn Leu Glu Phe Asp Gly Ser Asp Ser Ser Lys Asn Asp Thr Val Cys Ile His Lys Asp Glu Thr Asp Leu Leu Phe Thr Asp Gln His Asn Ile Cys Leu Lys Leu Ser Gly Gln Phe Met Lys Glu Gly Asn Thr Gln Ile Lys Glu Asp Leu Ser Asp Leu Thr Phe Leu Glu Val A1a Lys Ala Gln Glu Ala Cys His Gly Asn Thr Ser Asn Lys Glu Gln Leu Thr Ala Thr Lys Thr Glu Gln Asn Ile Lys Asp Phe Glu Thr Ser Asp Thr Phe Phe Gln Thr Ala Ser Gly Lys Asn Ile Ser Val Ala Lys Glu Ser Phe Asn Lys Ile Val Asn Phe Phe Asp Gln Lys Pro Glu Glu Leu His Asn Phe Ser Leu Asn Ser Glu Leu His Ser Asp Ile Arg Lys Asn Lys Met Asp Ile Leu Ser Tyr Glu Glu Thr Asp Ile Val Lys His Lys Ile Leu Lys Glu Ser Val Pro Val Gly Thr Gly Asn Gln Leu Va1 Thr Phe Gln G1y Gln Pro Glu Arg Asp Glu Lys Ile Lys Glu Pro Thr Leu Leu Gly Phe His Thr Ala Ser Gly Lys Lys Val Lys Ile Ala Lys Glu Ser Leu Asp Lys Val Lys Asn Leu Phe Asp Glu Lys Glu Gln Gly Thr Ser Glu Ile Thr Ser Phe Ser His Gln Trp Ala Lys Thr Leu Lys Tyr Arg Glu Ala Cys Lys Asp Leu Glu Leu A1a Cys Glu Thr Ile Glu Ile Thr Ala Ala Pro Lys Cys Lys Glu Met Gln Asn Ser Leu Asn Asn Asp Lys Asn Leu Val Ser Ile Glu Thr Val Val Pro Pro Lys Leu Leu Ser Asp Asn Leu CyS Arg Gln Thr Glu Asn Leu Lys Thr Ser Lys Ser Ile Phe Leu Lys Val Lys Val His Glu Asn Va1 Glu Lys Glu Thr Ala Lys Ser Pro Ala Thr Cys Tyr Thr Asn Gln Ser Pro Tyr Ser Val I1e Glu Asn Ser Ala Leu Ala Phe Tyr Thr Ser Cys Ser Arg Lys Thr Ser Val Ser Gln Thr Ser Leu Leu Glu Ala Lys Lys Trp Leu Arg Glu Gly Tle Phe Asp Gly Gln Pro Glu Arg Ile Asn Thr Ala Asp Tyr Val Gly Asn Tyr Leu Tyr Glu Asn Asn Ser Asn Ser Thr Tle Ala Glu Asn Asp Lys Asn His Leu Ser Glu Lys Gln Asp Thr Tyr Leu Ser Asn Ser Ser Met Ser Asn Ser Tyr Ser Tyr His Ser Asp Glu Val Tyr Asn Asp Ser Gly Tyr Leu Ser Lys Asn Lys Leu Asp Ser Gly Ile Glu Pro Val Leu Lys Asn Val Glu Asp Gln Lys Asn Thr Ser Phe Ser Lys Val Ile Ser Asn Val Lys Asp Ala Asn Ala Tyr Pro Gln Thr Val Asn Glu Asp Ile Cys Val Glu Glu Leu Val Thr Ser Ser Ser Pro Cys Lys Asn Lys Asn Ala Ala Ile Lys Leu Ser Ile Ser Asn Ser Asn Asn Phe Glu Val Gly Pro Pro Ala Phe Arg Ile Ala Ser Gly Lys Ile Val Cys Val Ser His Glu Thr Ile Lys Lys Val Lys Asp Ile Phe Thr Asp Ser Phe Ser Lys Val Ile Lys Glu Asn Asn Glu Asn Lys Ser Lys Ile Cys Gln Thr Lys Ile Met Ala Gly Cys Tyr Glu Ala Leu Asp Asp Ser Glu Asp Ile Leu His Asn Ser Leu Asp Asn Asp Glu Cys Ser Thr His Ser His Lys Val Phe Ala Asp Ile Gln Ser Glu Glu Ile Leu Gln His Asn Gln Asn Met Ser Gly Leu Glu Lys Val Ser Lys Ile Ser Pro Cys Asp Val Ser Leu G1u Thr Ser Asp Ile Cys Lys Cys Ser Ile Gly Lys Leu His Lys Ser Val Ser Ser Ala Asn Thr Cys Gly Ile Phe Ser Thr Ala Ser Gly Lys Ser Val Gln Val Ser Asp Ala Ser Leu Gln Asn Ala Arg Gln Va1 Phe Ser Glu Ile Glu Asp Ser Thr Lys Gln Val Phe Ser Lys Val Leu Phe Lys Ser Asn Glu His Ser Asp Gln Leu Thr Arg Glu Glu Asn Thr Ala Ile Arg Thr Pro G1u His Leu Ile Ser Gln Lys Gly Phe Ser Tyr Asn Val Val Asn Ser Ser Ala Phe Ser Gly Phe Ser Thr Ala Ser Gly Lys Gln Val Ser Ile Leu Glu Ser Ser Leu His Lys Val Lys Gly Val Leu Glu Glu Phe Asp Leu Ile Arg Thr Glu His Ser Leu His Tyr Ser Pro Thr Ser Arg Gln Asn Val Ser Lys I1e Leu Pro Arg Val Asp Lys Arg Asn Pro Glu His Cys Val Asn Ser Glu Met Glu Lys Thr Cys Ser Lys Glu Phe Lys Leu Ser Asn Asn Leu Asn Va1 Glu Gly Gly Ser Ser Glu Asn Asn His Ser Tle Lys Val Ser Pro Tyr Leu Ser Gln Phe Gln Gln Asp Lys Gln Gln Leu Val Leu Gly Thr Lys Val Ser Leu Val Glu Asn Tle His Val Leu Gly Lys Glu Gln Ala Ser Pro Lys Asn Va1 Lys Met Glu Ile Gly Lys Thr Glu Thr Phe Ser Asp Val Pro Val Lys Thr Asn Ile Glu Val Cys Ser Thr Tyr Ser Lys Asp Ser Glu Asn Tyr Phe Glu Thr Glu Ala Val Glu Tle Ala Lys Ala Phe Met Glu Asp Asp Glu Leu Thr Asp Ser Lys Leu Pro Ser His A1a Thr His Ser Leu Phe Thr Cys Pro Glu Asn Glu Glu Met Val Leu Ser Asn Ser Arg Ile Gly Lys Arg Arg Gly Glu Pro Leu Ile Leu Val Gly Glu Pro Ser Ile Lys Arg Asn Leu Leu Asn Glu Phe Asp Arg Ile Tle Glu Asn Gln Glu Lys Ser Leu Lys Ala Ser Lys Ser Thr Pro Asp Gly Thr Ile Lys Asp Arg Arg Leu Phe Met His His Va1 Ser Leu Glu Pro Ile Thr Cys Val Pro Phe Arg Thr Thr Lys Glu Arg Gln Glu Tle Gln Asn Pro Asn Phe Thr Ala Pro Gly Gln Glu Phe Leu Ser Lys Ser His Leu Tyr Glu His Leu Thr Leu Glu Lys Ser Ser Ser Asn Leu Ala Val Ser Gly His Pro Phe Tyr Gln Val Ser Ala Thr Arg Asn Glu Lys Met Arg His Leu Ile Thr Thr Gly Arg Pro Thr Lys Val Phe Val Pro Pro Phe Lys Thr Lys Ser His Phe His Arg Val Glu Gln Cys Val Arg Asn Ile Asn Leu Glu Glu Asn Arg Gln Lys Gln Asn Ile Asp Gly His Gly Ser Asp Asp Ser Lys Asn Lys Ile Asn Asp Asn Glu Ile His Gln Phe Asn Lys Asn Asn Ser Asn Gln Ala Ala Ala Val Thr Phe Thr Lys Cys Glu Glu Glu Pro Leu Asp Leu Ile Thr Ser Leu Gln Asn Ala Arg Asp Ile Gln Asp Met Arg Ile Lys Lys Lys Gln Arg Gln Arg Val Phe Pro Gln Pro Gly Ser Leu Tyr Leu Ala Lys Thr Ser Thr Leu Pro Arg Ile Ser Leu Lys Ala Ala Val Gly Gly Gln Val Pro Ser Ala Cys Ser His Lys Gln Leu Tyr Thr Tyr Gly Val Ser Lys His Cys Ile Lys Ile Asn Ser Lys Asn Ala Glu Ser Phe Gln Phe His Thr Glu Asp Tyr Phe Gly Lys Glu Ser Leu Trp Thr G1y Lys Gly Ile Gln Leu Ala Asp Gly Gly Trp Leu T1e Pro Ser Asn Asp Gly Lys Ala Gly Lys Glu Glu Phe Tyr Arg Ala Leu Cys Asp Thr Pro Gly Val Asp Pro Lys Leu Ile Ser Arg Ile Trp Val Tyr Asn His Tyr Arg Trp Ile Ile Trp Lys Leu Ala Ala Met Glu Cys Ala Phe Pro Lys Glu Phe Ala Asn Arg Cys Leu Ser Pro Glu Arg Val Leu Leu Gln Leu Lys Tyr Arg Tyr Asp Thr Glu Tle Asp Arg Ser Arg Arg Ser Ala Ile Lys Lys Ile Met Glu Arg Asp Asp Thr Ala Ala Lys Thr Leu Val Leu Cys Val Ser Asp Ile Ile Ser Leu Ser Ala Asn I1e Ser Glu Thr Ser Ser Asn Lys Thr 5er Ser Ala Asp Thr Gln Lys Val Ala Ile Ile Glu Leu Thr Asp Gly Trp Tyr Ala Val Lys Ala Gln Leu Asp Pro Pro Leu Leu A1a Val Leu Lys Asn Gly Arg Leu Thr Val Gly Gln Lys Ile Ile Leu His Gly Ala Glu Leu Va1 G1y Ser Pro Asp Ala Cys Thr Pro Leu Glu Ala Pro Glu Ser Leu Met Leu Lys Ile _ g7 _ Ser Ala Asn Ser Thr Arg Pro Ala Arg Trp Tyr Thr Lys Leu Gly Phe Phe Pro Asp Pro Arg Pro Phe Pro Leu Pro Leu Ser Ser Leu Phe Ser Asp Gly Gly Asn Val Gly Cys Val Asp Val Ile Ile Gln Arg Ala Tyr Pro Ile Gln Trp Met Glu Lys Thr Ser 5er Gly Leu Tyr Ile Phe Arg Asn Glu Arg Glu Glu Glu Lys Glu Ala Ala Lys Tyr Val Glu Ala Gln Gln Lys Arg Leu Glu Ala Leu Phe Thr Lys Ile Gln Glu Glu Phe Glu Glu His Glu Glu Asn Thr Thr Lys Pro Tyr Leu Pro Ser Arg Ala Leu Thr Arg Gln Gln Va1 Arg Ala Leu Gln Asp Gly Ala Glu Leu Tyr Glu Ala Val Lys Asn Ala Ala Asp Pro Ala Tyr Leu Glu Gly Tyr Phe Ser Glu Glu Gln Leu Arg Ala Leu Asn Asn His Arg Gln Met Leu Asn Asp Lys Lys Gln Ala Gln Ile Gln Leu Glu Ile Arg Lys Ala Met Glu Ser Ala Glu Gln Lys Glu Gln Gly Leu Ser Arg Asp Val Thr Thr Val Trp Lys Leu Arg Ile Val Ser Tyr Ser Lys Lys Glu Lys Asp Ser Val Ile Leu Ser Ile Trp Arg Pro Ser Ser Asp Leu Tyr Ser Leu Leu Thr Glu Gly _88_ Lys Arg Tyr Arg Ile Tyr His Leu Ala Thr Ser Lys Ser Lys Ser Lys Ser Glu Arg Ala Asn Ile Gln Leu Ala Ala Thr Lys Lys Thr Gln Tyr Gln Gln Leu Pro Val Ser Asp Glu Ile Leu Phe Gln Ile Tyr Gln Pro Arg Glu Pro Leu His Phe Ser Lys Phe Leu Asp Pro Asp Phe Gln Pro Ser Cys Ser Glu Val Asp Leu Tle Gly Phe Val Val Ser Val Val Lys Lys Thr Gly Leu Ala Pro Phe Val Tyr Leu Ser Asp Glu Cys Tyr Asn Leu Leu Ala Ile Lys Phe Trp Ile Asp Leu Asn Glu Asp Ile Ile Lys Pro His Met Leu Ile Ala Ala Ser Asn Leu Gln Trp Arg Pro Glu Ser Lys Ser Gly Leu Leu Thr Leu Phe Ala Gly Asp Phe Ser Val Phe Ser Ala Ser Pro Lys Glu Gly His Phe Gln Glu Thr Phe Asn Lys Met Lys Asn Thr Val Glu Asn Ile Asp Ile Leu Cys Asn Glu Ala Glu Asn Lys Leu Met His Ile Leu His Ala Asn Asp Pro Lys Trp Ser Thr Pro Thr Lys Asp Cys Thr Ser Gly Pro Tyr Thr Ala Gln Ile Ile Pro Gly Thr Gly Asn Lys Leu Leu Met Ser Ser Pro Asn Cys Glu Ile Tyr Tyr Gln Ser Pro Leu Ser Leu Cys Met Ala Lys Arg Lys Ser Val Ser Thr Pro Val Ser Ala Gln Met Thr Ser Lys 5er Cys Lys Gly Glu Lys Glu I1e Asp Asp Gln Lys Asn Cys Lys Lys Arg Arg Ala Leu Asp Phe Leu Ser Arg Leu Pro Leu Pro Pro Pro Val Ser Pro Ile Cys Thr Phe Val Ser Pro Ala Ala G1n Lys Ala Phe Gln Pro Pro Arg Ser Cys Gly Thr Lys Tyr Glu Thr Pro Ile Lys Lys Lys Glu Leu Asn Ser Pro Gln Met Thr Pro Phe Lys Lys Phe Asn Glu Ile Ser Leu Leu Glu Ser Asn Ser Ile Ala Asp Glu Glu Leu Ala Leu Tle Asn Thr Gln Ala Leu Leu Ser Gly Ser Thr Gly Glu Lys Gln Phe I1e Ser Val Ser Glu Ser Thr Arg Thr Ala Pro Thr Ser Ser G1u Asp Tyr Leu Arg Leu Lys Arg Arg Cys Thr Thr Ser Leu Ile Lys Glu Gln Glu Ser 5er Gln A1a Ser Thr Glu Glu Cys Glu Lys Asn Lys Gln Asp Thr Ile Thr Thr Lys Lys Tyr Ile <210> 24 <211> 21 <212> DNA
<213> 586F18b microsatellite primer 1 <400> 24 gctagggaac caaactgcca g 21 <210> 25 <211> 26 <212> DNA
<213> 586F18b microsatellite primer 2 <400> 25 tgcaaaataa caatagcttt gcttag 26 <210> 26 <211> 25 <212> DNA
<213> primer EDD 407F
<400> 26 gctagtcacc aacttctggg tctaa 25 <210> 27 <211> 29 <212> DNA
<213> primer EDD-4908 -<400> 27 cagcaaaaag ataaatcaca gtgtaaatt 29 <210> 28 <211> 27 <212> DNA
<2l3> primer EDD-433T
<220>
<221> misc_feature <222> (1)..(1) <223> modified by attachment of FAM dye <220>
<221> misc_feature <222> (27) .(27) <223> modified by attachment of TAMRA dye <400> 28 cccagccaaa gatgacagca gaacaac 27 <210> 29 <211> 24 <212> DNA
<213> EDD forward primer <400> 29 ttaggctttt ggtaaatggc tgcg 24 <210> 30 <211> 24 <212> DNA

<213> EDD reverse primer <400> 30 tgagggcata ggctggaatc cttc 24 <210> 31 <211> 22 <212> DNA
<213> Carbonic anhydrase II forward primer <400> 31 ccacccctcc tcttctggaa tg , 22 <210> 32 <211> 25 <212> DNA
<213> Carbonic anhydrase II reverse primer <400> 32 gctttgattt gcctgttctt cagtg 25 <210> 33 <211> 23 <212> DNA
<213> p53 ribonucleotide reductase (p53R2) forward primer <400> 33 gtgactttgc ttgcctgatg ttc 23 <210> 34 <211> 23 <212> DNA
<213> p53 ribonucleotide reductase (p53R2) reverse primer <400> 34 tctgtggttt ctgccataac tgc 23 <210> 35 <211> 20 <2l2> DNA
<213> GAPDH forward primer <400> 35 gacatcaaga aggtggtgaa 20 <210> 36 <211> 20 <212> DNA
<213> GAPDH reverse primer <400> 36 tgtcatacca ggaaatgagc 20 <210> 37 <211> 25 <212> DNA
<213> EDD forward primer <400> 37 cattgctgac cctatccctg tgttg 25 <2l0> 38 <211> 24 <212> DNA
<213> EDD reverse primer <400> 38 tagcccgtga aatcctccca tctc 24 <210> 39 <211> 24 <212> DNA
<213> carbonic anhydrase II forward primer <400> 39 acccgcctca tgcctcagcc ttac 24 <210> 40 <211> 24 <212> DNA
<213> p53R2 forward primer <400> 40 tgtcagcctt gagtacctcc aggg 24 <210> 41 <211> 18 <212> DNA
<213> beclin forward primer <400> 41 taggtttggg gtgagtgg 18 <210> 42 <21l> 18 <212> DNA
<213> reverse primer <400> 42 agtctgtggg cagcaagg 18 <210> 43 <211> 20 <212> PRT
<213> synthetic bipartite nuclear localization sequence (NZS) from amino acid residues 1222 to 1241 of EDD
<400> 43 Lys Leu Lys Arg Thr Ser Pro Thr Ala Tyr Cys Asp Cys Trp Glu Lys Cys Lys Cys Lys <210> 44 <211> 16 <212> PRT
<213> synthetic bipartite nuclear localization sequence (NLS) from amino acid residues 502 to 517 of EDD
<400> 44 Arg Lys Lys Met Leu Glu Lys Ala Arg Ala Lys Asn Lys Lys Pro Lys l 5 10 15 <210> 45 <211> 6 <212> PRT
<213> synthetic monopartite nuclear localization sequence (NLS) from amino acid residues 630 to 635 of EDD
<400> 45 Pro Tyr Lys Arg Arg Arg <210> 46 <211> 10 <212> PRT
<213> FHA domain-binding Phosphopeptide <220>
<221> MOD_RES
<222> (6) .(6) <223> PHOSPHORYLATION
<400> 46 Arg Trp Phe Asp Thr Tyr Leu Ile Arg Arg <210> 47 <211> 21 <212> DNA
<213> synthetic siRNA strand <400> 47 gcaguguucc ugccuucuut t 21 <210> 48 <211> 21 <212> DNA

<213> synthetic siRNA strand <400> 48 ttcgucacaa ggacggaaga a 21 <210> 49 <21l> 216 <212> DNA
<213> MMTV LTR
<400> 49 gtttaaataa gtttatggtt acaaactgtt cttaaaacga ggatgtgaga caagtggttt 60 cctgacttgg tttggtatca aatgttctga tctgagctct tagtgttcta ttttcctatg 120 ttcttttgga atctatccaa gtcttatgta aatgcttatg taaaccataa tataaaagag 180 tgctgatttt ttgagtaaac ttgcaacagt cctaac 216 <210> 50 <211> 1032 <212> DNA
<2l3> Cre recombinase <400>

atgtccaatttactgaccgtacaccaaaatttgcctgcattaccggtcgatgcaacgagt60 gatgaggttcgcaagaacctgatggacatgttcagggatcgccaggcgttttctgagcat120 acctggaaaatgcttctgtccgtttgccggtcgtgggcggcatggtgcaagttgaataac180 cggaaatggtttcccgcagaacctgaagatgttcgcgattatcttctatatcttcaggcg240 cgcggtctggcagtaaaaactatccagcaacatttgggccagctaaacatgcttcatcgt300 cggtccgggctgccacgaccaagtgacagcaatgctgtttcactggttatgcggcggatc360 cgaaaagaaaacgttgatgccggtgaacgtgcaaaacaggctctagcgttcgaacgcact420 gatttcgaccaggttcgttcactcatggaaaatagcgatcgctgccaggatatacgtaat480 ctggcatttctggggattgcttataacaccctgttacgtatagccgaaattgccaggatc540 agggttaaagatatctcacgtactgacggtgggagaatgttaatccatattggcagaacg600 aaaacgctggttagcaccgcaggtgtagagaaggcacttagcctgggggtaactaaactg660 gtcgagcgatggatttccgtctctggtgtagctgatgatccgaataactacctgttttgc720 cgggtcagaaaaaatggtgttgccgcgccatctgccaccagccagctatcaactcgcgcc780 ctggaagggatttttgaagcaactcatcgattgatttacggcgctaaggatgactctggt840 cagagatacctggcctggtctggacacagtgcccgtgtcggagccgcgcgagatatggcc900 cgcgctggagtttcaataccggagatcatgcaagctggtggctggaccaatgtaaatatt960 gtcatgaactatatccgtaacctggatagtgaaacaggggcaatggtgcgcctgctggaa1020 gatggcgatt ag 1032 <210> 51 <211> 22 <212> DNA
<213> knockout genotyping primer 1 <400> 51 ctagggaagt gcattaggta ag 22 <2l0> 52 <211> 20 <212> DNA
<213> knockout genotyping primer 2 <400> 52 taagggcagg tgttcctctg 20 <210> 53 <211> 25 <212> DNA
<2l3> knockout genotyping primer 3 <400> 53 gtactgtggt ttccaaatgt gtcag 25

Claims (75)

WE CLAIM:
1. A method for detecting a cancer cell in a subject, said method comprising determining the level of nucleic acid that is linked to map position 8q22.3 of the human genome or an expression product thereof in a sample of said subject, wherein an elevated level of said nucleic acid or said polypeptide is indicative of cancer in the subject.
2. The method according to claim 1 wherein the cancer cell is epithelial in origin.
3. The method according to claim 1 or claim 2 wherein the cancer cell is from a cancer selected from the group consisting of ovarian cancer, melanoma, metastatic melanoma, squamous cell carcinoma of the head and neck, squamous cell carcinoma of the tongue, hepatocellular carcinoma, breast cancer, a metastases of ovarian cancer, a metastases of melanoma, a metastases of metastatic melanoma, a metastases of squamous cell carcinoma of the head and neck, a metastases of squamous cell carcinoma of the tongue, a metastases of hepatocellular carcinoma and a metastases of breast cancer.
4. The method according to any one of claims 1 to 3 wherein the nucleic acid that is linked to map position 8q22.3 of the human genome comprises the genomic Edd and p53R2 genes or a portion thereof.
5. The method according to any one of claims 1 to 4 wherein the nucleic acid that is linked to map position 8q22.3 of the human genome comprises a genomic gene encoding an EDD protein.
6. The method of claim 5 wherein the EDD protein is a polypeptide that comprises an amino acid sequence having at least 80% identity to the sequence set forth in SEQ ID Nos: 2 or 4.
7. The method of any one of claims 1 to 6, said method comprising:
(i) determining the level of nucleic acid linked to map position 8q22.3 of the human genome in a test sample from said subject; and (ii) comparing the level of the nucleic acid at (i) to the level of the nucleic acid in a reference sample from a healthy or normal individual, wherein a level of the nucleic acid at (ii) that is enhanced in the test sample relative to the reference sample from the normal or healthy individual is indicative of the presence of a cancer cell in said subject.
8. The method of claim 7 wherein the test sample and the reference sample comprise a cell from a tissue selected from the group consisting of skin, an oral cavity tissue, breast, liver, spleen, ovary, prostate, kidney, uterus, placenta, cervix, omentum, rectum, brain, bone, lung, lymph, urine, semen, blood, abdominal fluid, and serum.
9. The method of any one of claims 1 to 8 wherein the level of nucleic acid linked to map position 8q22.3 of the human genome is determined by hybridizing a nucleic acid probe to genomic DNA encoding an EDD
protein in the sample under stringency hybridization conditions and detecting the hybridization using a detection means.
10. The method of claim 9 wherein the detection means is nucleic acid hybridization or amplification reaction.
11. The method of any one of claims 1 to 10 wherein the level of nucleic acid that is linked to map position 8q22.3 of the human genome is determined by hybridizing a nucleic acid probe or primer to genomic DNA and detecting the hybridization, wherein the probe or primer comprises a nucleotide sequence selected from the group consisting of:
(i) the sequence set forth in SEQ ID NO: 5;
(ii) the sequence set forth in SEQ ID NO: 6;

(iii) the sequence set forth in SEQ ID NO: 7;
(iv)the sequence set forth in SEQ ID NO: 24;
(v) the sequence set forth in SEQ ID NO: 25; and the sequence of a nucleic acid fragment produced by amplification using any one of (i) to (v) as amplification primers in PCR.
12. The method of any one of claims 1 to 11 wherein the sample has been obtained previously from the subject.
13. The method of any one of claims 1 to 10 wherein the level of nucleic acid that is linked to map position 8q22.3 of the human genome is determined by hybridizing a nucleic acid probe or primer to genomic DNA and detecting the hybridization, wherein the probe or primer comprises a nucleotide sequence selected from the group consisting of:
(i) the sequence set forth in SEQ ID NO: 3;
(ii) the sequence set forth in SEQ ID NO: 5;
(iii) the sequence set forth in SEQ ID NO: 6;
(iv) the sequence set forth in SEQ ID NO: 7;
(v) the sequence set forth in SEQ ID NO: 24;
(vi) the sequence set forth in SEQ ID NO: 25;
(vii) the sequence of a nucleic acid fragment produced by amplification using (vi) and (vii) as amplification primers in PCR;
(viii) the sequence set forth in SEQ ID NO:26;
(ix) the sequence set forth in SEQ ID NO: 27;
(x) the sequence of a nucleic acid fragment produced by amplification using (ix) and (x) as amplification primers in PCR;
(xi) the sequence set forth in SEQ ID NO: 28;
(xii) the sequence set forth in SEQ ID NO: 29;
(xiii) the sequence set forth in SEQ ID NO: 30;
(xiv) the sequence of a nucleic acid fragment produced by amplification using (xii) and (xiii) as amplification primers in PCR;
(xv) the sequence set forth in SEQ ID NO: 33;

(xvi) the sequence set forth in SEQ ID NO: 34;
(xvii) the sequence of a nucleic acid fragment produced by amplification using (xv) and (xvi) as amplification primers in PCR;
(xviii) the sequence set forth in SEQ ID NO: 37;
(xix) the sequence set forth in SEQ ID NO: 38;
(xx) the sequence of a nucleic acid fragment produced by amplification using (xviii) and (xix) as amplification primers in PCR;
(xxi) the sequence set forth in SEQ ID NO: 40; and (xxii) a sequence that is complementary to any one of (i) to (xxi).
14. A method for detecting a cancer cell in a subject, said method comprising:
(i) determining the level of mRNA encoded by nucleic acid linked to map position 8q22.3 of the human genome that is expressed in a test sample from said subject; and (ii) comparing the level of the mRNA determined at (i) to the level of mRNA encoded by nucleic acid linked to map position 8q22.3 of the human genome that is expressed in a reference sample from a healthy or normal individual, wherein a level of the mRNA at (i) that is enhanced in the test sample relative to the reference sample from the normal or healthy individual is indicative of the presence of a cancer cell in said subject.
15. The method of claim 14 wherein the mRNA encodes an EDD protein.
16. The method of claim 14 or claim 15 wherein the mRNA encodes an EDD
polypeptide that has at least about 80% identity to SEQ ID NO: 2 or 4.
17. The method of claim 15 or claim 16 wherein the mRNA has 80% identity to the nucleotide sequence set forth in SEQ ID NO: 1 or 3.
18. The method of claim 14 wherein the mRNA encodes a p53R2 protein.
19. A method for diagnosing a cancer or predicting recurrence of a cancer in a subject comprising determining the level of mRNA or protein encoded by nucleic acid linked to map position 8q22.3 of the human genome in a sample of said subject, wherein an elevated level of said mRNA or protein is indicative of relapse of a cancer in said subject.
20. The method according to claim 19 wherein the cancer cell is epithelial in origin.
21. The method according to claim 19 or claim 20 wherein the cancer cell is from a an ovarian cancer.
22. The method of any one of claims 19 to 21 wherein the mRNA encodes an EDD protein.
23. The method of any one of claims 19 to 22 wherein the mRNA encodes an EDD protein that has at least 80% homology to SEQ ID NO: 2 or SEQ ID NO: 4.
24. The method of any one of claims 19 to 21 wherein the protein is an EDD
protein.
25. The method of claim 24 wherein the EDD protein has at least 80%
homology to SEQ ID NO: 2 or SEQ ID NO: 4.
26. An isolated nucleic acid molecule for detecting a cancer cell comprising a nucleotide sequence selected from the group consisting of:
a sequence that encodes the amino acid sequence set forth in SEQ ID
NO: 4 wherein said amino acid sequence lacks the sequence VLLLPL;
(i) the sequence set forth in SEQ ID NO: 3;

(ii) the sequence set forth in SEQ ID NO: 5;
(iii) the sequence set forth in SEQ ID NO: 6;
(iv) the sequence set forth in SEQ ID NO: 7;
(v) the sequence set forth in SEQ ID NO: 24;
(vi) the sequence set forth in SEQ ID NO: 25;
(vii) the sequence of a nucleic acid fragment produced by amplification using (vi) and (vii) as amplification primers in PCR;
(viii) the sequence set forth in SEQ ID NO:26;
(ix) the sequence set forth in SEQ ID NO: 27;
(x) the sequence of a nucleic acid fragment produced by amplification using (ix) and (x) as amplification primers in PCR;
(xi) the sequence set forth in SEQ ID NO: 28;
(xii) the sequence set forth in SEQ ID NO: 29;
(xiii) the sequence set forth in SEQ ID NO: 30;
(xiv) the sequence of a nucleic acid fragment produced by amplification using (xii) and (xiii) as amplification primers in PCR;
(xv) the sequence set forth in SEQ ID NO: 33;
(xvi) the sequence set forth in SEQ ID NO: 34;
(xvii) the sequence of a nucleic acid fragment produced by amplification using (xv) and (xvi) as amplification primers in PCR;
(xviii) the sequence set forth in SEQ ID NO: 37;
(xix) the sequence set forth in SEQ ID NO: 38;
(xx) the sequence of a nucleic acid fragment produced by amplification using (xviii) and (xix) as amplification primers in PCR;
(xxi) the sequence set forth in SEQ ID NO: 40; and (xxii) a sequence that is complementary to any one of (i) to (xxi).
27. An isolated or recombinant protein complex comprising:
(i) an EDD protein or a portion of an EDD protein sufficient to bind to a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, and a progesterone receptor protein;
and (ii) a nuclear protein selected from the group consisting of a protein having tumor suppresser activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein and a progesterone receptor protein or a portion of said protein sufficient to bind to said EDD
protein or said portion of an EDD protein.
28. The isolated or recombinant protein complex of claim 27 wherein the protein having tumor suppresser activity is a protein selected from the group consisting of a member of the CDS1 subfamily of serine/threonine protein kinases, p53 (TP53), TNF-alpha, HSP70, estrogen receptor, androgen receptor, progesterone receptor, HRAS1-VNTR, CHK2, BRCA1, BRCA2, AIB1, NAT1, NAT2, XRCC1, XRCC2, XRCC5, CIB, importin alpha-1, importin alpha-3, and importin alpha-5.
29. The isolated or recombinant protein complex of claim 27 or claim 28 wherein the protein having tumor suppresser activity comprises an amino acid sequence having at least 80% identity to a sequence selected from the group consisting of SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ
ID NO: 21, and SEQ ID NO: 23.
30. The isolated or recombinant protein complex of claim 27 wherein the protein having cell cycle modulatory activity is selected from the group consisting of a member of the CDS1 subfamily of serine/threonine protein kinases, Cdc25, CDC2a, cyclin-dependent kinase (CDK), CDK
inhibitor, a mitogenic cyclin (e.g., cyclin A, cyclin B, cyclin D, etc), p53 (TP53), and CHK2.
31. The isolated or recombinant protein complex of claim 27 or claim 30 wherein the protein having cell cycle modulatory activity comprises an amino acid sequence having at least 80% identity to a sequence set forth selected from the group consisting of SEQ ID NO: 19 and SEQ ID
NO:21.
32. The isolated or recombinant protein complex of claim 27 wherein the protein associated with DNA repair or damage is a protein selected from the group consisting of a member of the CDS1 subfamily of serine/threonine protein kinases, BRCA1, BRCA2, CIB/KIP, TP53, MLH1, MSH2, ATM, CHK2, XRCC1, XRCC2, XRCC5 and importin alpha-5.
33. The isolated or recombinant protein complex of claim 27 or claim 32 wherein the protein associated with DNA damage or repair comprises an amino acid sequence having at least 80% identity to a sequence selected from the group consisting of SEQ ID NO: 13, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, and SEQ ID NO: 23.
34. The isolated or recombinant protein complex of claim 27 wherein the nuclear targeting protein is selected from the group consisting of importin alpha-1, importin alpha-3 and importin alpha-5.
35. The isolated or recombinant protein complex of claim 27 or claim 34 wherein the nuclear targeting protein comprises an amino acid sequence having at least 80% identity to a sequence selected from the group SEQ
ID NOs: 9, 11 or 13.
36. The isolated or recombinant protein complex of claim 27 wherein the progesterone receptor protein comprises an amino acid sequence having at least 80% sequence homology to SEQ ID NO: 15.
37. An isolated antibody that binds to a protein complex comprising an EDD
protein.
38. The isolated antibody of claim 37 wherein the antibody binds to a protein complex comprising an EDD protein or a portion of an EDD protein sufficient to bind to a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, and a progesterone receptor protein
39. The isolated antibody of claim 37 or claim 38 wherein the antibody binds to a protein complex selected from the group consisting of: (i) a complex comprising EDD and CHK2; (ii) a complex comprising EDD and BRCA2;
(iii) a complex comprising EDD and CIB; (iv) a complex comprising EDD
and importin alpha-1; (v) a complex comprising EDD and importin alpha-3; (vi) a complex comprising EDD and importin alpha-5; and (vii) a complex comprising EDD and progesterone receptor,
40. The isolated antibody of any one of claims 37 to 39 wherein the antibody does not bind to any individual protein of said complex in the absence of another protein of said complex.
41. The isolated antibody of any one of claims 37 to 40 wherein the antibody recognises a conformational epitope of the protein complex.
42. An isolated antibody that binds to the antibody of any one of claims 37 to 41.
43. A kit for detecting or producing a protein complex, said kit comprising an EDD polypeptide or a portion of an EDD polypeptide and a second polypeptides selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, and a progesterone receptor protein or a portion thereof, wherein the portion of the second polypeptide is sufficient to bind to said EDD polypeptide or said portion of an EDD polypeptide.
44. The kit of claim 43 additionally comprising a protein that binds to the EDD-binding protein.
45. The kit of claim 44 wherein the protein that binds to the EDD-binding protein comprises an amino acid sequence selected from the group consisting of SEQ ID NO: 9, SEQ ID NO: 11, SEQ ID NO: 13, SEQ ID
NO: 15, SEQ ID NO: 17, SEQ ID NO: 19, SEQ ID NO: 21, and SEQ ID
NO: 23.
46. The kit of any one of claims 43 to 45 further comprising an antibody or ligand that binds to the first or second polypeptide or to the complex formed between said first and said second polypeptide.
47. A kit for detecting or producing a protein complex comprising:
(i) a first compartment comprising an EDD protein or a portion thereof sufficient to form a protein complex selected from the group consisting of: a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, and a progesterone receptor protein or a portion thereof, wherein the portion of the second polypeptide is sufficient to bind to said EDD
polypeptide or said portion of an EDD polypeptide; and (ii) a second compartment comprising an antibody or ligand that binds to a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, and a progesterone receptor protein or a portion thereof.
wherein said antibody or ligand that binds to a protein complex does not bind to the individual protein binding partners.
48. A kit for detecting or producing a protein complex comprising:
(i) a first compartment comprising an EDD protein or a portion thereof sufficient to form a protein complex selected from the group consisting of: (i) a complex comprising EDD and CHK2; (ii) a complex comprising EDD and BRCA2; (iii) a complex comprising EDD and CIB; (iv) a complex comprising EDD and importin alpha-1; (v) a complex comprising EDD and importin alpha-3; (vi) a complex comprising EDD and importin alpha-5; and (vii) a complex comprising EDD and progesterone receptor; and (ii) a second compartment comprising an antibody or ligand that binds to a protein selected from the group consisting of (i) a CHK2 protein; (ii) a BRCA2 protein; (iii) a CIBCIB protein; (iv) an importin alpha-1 protein; (v) an importin alpha-3 protein; (vi) an importin alpha-5 protein; and (vii) a progesterone receptor protein, or an antibody or ligand that binds to a protein complex selected from the group consisting of: (i) a complex comprising EDD and CHK2; (ii) a complex comprising EDD and BRCA2; (iii) a complex comprising EDD and CIB; (iv) a complex comprising EDD and importin alpha-1; (v) a complex comprising EDD and importin alpha-3; (vi) a complex comprising EDD and importin alpha-5; and (vii) a complex comprising EDD and progesterone receptor.

wherein said antibody or ligand that binds to a protein complex does not bind to the individual protein binding partners.
49. A kit for detecting or producing a protein complex comprising:
(i) a first compartment comprising an antibody or ligand that binds to an EDD protein; and (ii) a second compartment comprising a protein selected from the group consisting of (i) a CHK2 protein; (ii) a BRCA2 protein; (iii) a CIB protein; (iv) an importin alpha-1 protein; (v) an importin alpha-3 protein; (vi) an importin alpha-5 protein; and (vii) a progesterone receptor protein, or a portion thereof sufficient to bind to an EDD
protein.
50. A kit for detecting or producing a protein complex comprising:
(i) a first compartment comprising an isolated or recombinant protein complex selected from the group consisting of: (i) a complex comprising EDD and CHK2; (ii) a complex comprising EDD and BRCA2; (iii) a complex comprising EDD and CIB; (iv) a complex comprising EDD and importin alpha-1; (v) a complex comprising EDD and importin alpha-3; (vi) a complex comprising EDD and importin alpha-5; and (vii) a complex comprising EDD and progesterone receptor; and (ii) a second compartment comprising an (i) antibody or ligand that binds to a polypeptide selected from the group consisting of a CHK2 protein, a BRCA2 protein, a CIB protein, an importin alpha-1 protein, an importin alpha-3 protein, an importin alpha-5 protein, a progesterone receptor protein and an EDD protein; or (ii) an antibody or ligand that binds to one or more protein complexes (a).
51. A method for isolating a protein complex comprising an EDD protein or a protein of an EDD protein and a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, and a progesterone receptor protein from a suitable cellular source, said method comprising contacting an extract of said cell with an EDD polypeptide or a portion thereof that binds to said protein for a time and under conditions sufficient for a protein complex to form and then isolating the protein complex formed.
52. A method of isolating the protein complex comprising an EDD protein or a protein of an EDD protein and a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, and a progesterone receptor protein comprising:
(i) isolating a protein that is a component of the protein complex from a cell that expresses said protein;
(ii) isolating another protein that is a component of the protein complex from a cell that expresses said other protein; and (iii) combining the proteins isolated at (i) and (ii) in an amount and under conditions sufficient to facilitate the formation of a protein complex.
53. The method of claim 51 or claim 52 wherein the protein is endogenously expressed by the cell.
54. The method of claim claim 51 or claim 52 wherein the protein is ectopically expressed in the cell.
55. A method for determining a predisposition for disease, or a disease state, said method comprising detecting a protein complex comprising:
(i) an EDD protein; and (ii) a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, a progesterone receptor protein and a protein associated with vascularization, wherein an elevated level of said protein complex is indicative of a predisposition for disease, or a disease state in said subject.
56. The method of claim 55 wherein the protein complex is selected from the group consisting of (i) a complex comprising EDD and CHK2; (ii) a complex comprising EDD and BRCA2; (iii) a complex comprising EDD
and CIB; (iv) a complex comprising EDD and importin alpha-1; (v) a complex comprising EDD and importin alpha-3; (vi) a complex comprising EDD and importin alpha-5; and (vii) a complex comprising EDD and progesterone receptor.
57. The method of claim 55 or claim 56 wherein the protein complex is detected using an antibody or ligand that binds to the protein complex.
58. A method for determining a modulator of the activity, formation or stability of an isolated or recombinant protein complex comprising:
(i) determining the activity, formation or stability of a protein complex comprising (a) an EDD protein or a portion of an EDD protein; and (b) a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, and a progesterone receptor protein or a portion thereof, wherein the portion of the second polypeptide is sufficient to bind to said EDD polypeptide or said portion of an EDD polypeptide, in the absence of a candidate compound or candidate antibody; and (ii) determining the level of said protein complex in the presence of a candidate compound or in the presence of said candidate antibody wherein a difference in the level of said protein complex at (i) and (ii) indicates that the candidate compound or candidate antibody is a modulator of said interaction.
59. A method for determining a modulator of the level of protein complex formation comprising:
(i) determining the level of a protein complex comprising (a) an EDD
protein or a portion of an EDD protein; and (b) a protein selected from the group consisting of a protein having tumor suppressor activity, a protein having cell cycle modulatory activity, a protein associated with DNA repair or damage, a nuclear targeting protein, and a progesterone receptor protein or a portion thereof, wherein the portion of the second polypeptide is sufficient to bind to said EDD polypeptide or said portion of an EDD polypeptide, in the absence of a candidate compound or candidate antibody; and (ii) determining the level of said protein complex in the presence of a candidate compound or in the presence of said candidate antibody wherein a difference in the level of said protein complex at (i) and (ii) indicates that the candidate compound or candidate antibody is a modulator of said interaction.
60. The method of claim 58 or claim 59 wherein the protein complex is selected from the group consisting of: (i) a complex comprising EDD and CHK2; (ii) a complex comprising EDD and BRCA2; (iii) a complex comprising EDD and CIB; (iv) a complex comprising EDD and importin alpha-1; (v) a complex comprising EDD and importin alpha-3; (vi) a complex comprising EDD and importin alpha-5; and (vii) a complex comprising EDD and progesterone receptor.
61. The method of any one of claims 58 to 60 wherein the modulator enhances complex activity, formation or stability.
62. The method of any one of claims 58 to 60 wherein the modulator partially or completely inhibits complex activity, formation or stability.
63. A method for treating a condition associated with elevated expression of an EDD protein in a cell, said method comprising administering an amount of a compound effective to reduce EDD expression in a cell.
64. The method of claim 63 wherein the condition associated with EDD over expression is a cancer,
65. The method of claim 64 wherein the cancer is a cancer selected from the group consisting of squamous cell carcinoma, hepatocellular carcinoma, ovarian cancer, breast cancer, melanoma, head and neck cancer, adenocarcinoma, squamous lung cancer, gastrointestinal cancer (eg.
gastric, colon, or pancreatic cancer), renal cell cancer, bladder cancer, prostate cancer, non-squamous carcinoma, glioblastoma and medulloblastoma.
66. The method of any one of claims 61 to 65 wherein the compound administered comprises nucleic acid.
67. The method of claim 66 wherein the nucleic acid is an antisense nucleic acid, peptide nucleic acid (PNA), ribozyme, or interfering RNA, which is complementary; in whole or in part, to EDD-encoding RNA.
68. The method of claim 67 wherein the antisense nucleic acid, ribozyme, PNA, interfering RNA or siRNA comprises a sequence that is complementary to at least about 15-20 contiguous nucleotides of a sequence having at least 80% identity to SEQ ID NO: 1 or SEQ ID NO:
3.
69. An antisense nucleic acid, ribozyme, PNA, interfering RNA or siRNA
comprising a sequence having at least 80% homology to SEQ ID NO:
47.
70. The use of the antisense nucleic acid, ribozyme, PNA, interfering RNA or siRNA of claim 69 to reduce the expression of EDD in a cell.
71. The use of the antisense nucleic acid, ribozyme, PNA, interfering RNA or siRNA of claim 69 to inhibit cellular proliferation.
72. A pharmaceutical composition comprising the antisense nucleic acid, ribozyme, PNA, interfering RNA or siRNA of claim 69.
73. The use of the composition of claim,72 to reduce the expression of EDD
in a cell.
74. The use of the composition of claim 72 to inhibit cellular proliferation.
75. A method for determining the ability of a cell to phosphorylate CHK2 in response to a DNA damaging agent comprising determining the level of expression of EDD in said cell, wherein reduced or suppressed EDD
expression indicates that the cell has reduced ability to phosphorylate CHK2 in response to a DNA damaging agent.
CA002497873A 2002-09-05 2003-09-05 Novel diagnostic and therapeutic methods and reagents therefor Abandoned CA2497873A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
AU2002951346 2002-09-05
AU2002951346A AU2002951346A0 (en) 2002-09-05 2002-09-05 Diagnosis of ovarian cancer
US42521802P 2002-11-07 2002-11-07
US60/425,218 2002-11-07
PCT/AU2003/001164 WO2004022750A1 (en) 2002-09-05 2003-09-05 Novel diagnostic and therapeutic methods and reagents therefor

Publications (1)

Publication Number Publication Date
CA2497873A1 true CA2497873A1 (en) 2004-03-18

Family

ID=31979115

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002497873A Abandoned CA2497873A1 (en) 2002-09-05 2003-09-05 Novel diagnostic and therapeutic methods and reagents therefor

Country Status (4)

Country Link
EP (1) EP1539957A4 (en)
JP (1) JP2005537790A (en)
CA (1) CA2497873A1 (en)
WO (1) WO2004022750A1 (en)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW200639252A (en) * 2005-02-01 2006-11-16 Alcon Inc RNAi-mediated inhibition of ocular hypertension targets
IL177006A0 (en) * 2005-08-02 2006-12-10 Veridex Llc Predicting bone relapse of breast cancer
JP2008297257A (en) * 2007-05-31 2008-12-11 St Marianna Univ School Of Medicine New curative agent for digestive cancer
CN101429540B (en) * 2007-11-09 2013-07-10 北京雅康博生物科技有限公司 Method for real time quantitative PCR detection of mitotic cycle protein B2 gene expression
CN102559601B (en) * 2012-01-13 2014-09-03 清华大学 CREPT (Cell-cycle Related and Expression-elevated Protein in Tumor) antibody for identifying tumor cells or tumor tissues
JP6343560B2 (en) * 2012-09-05 2018-06-13 富士フイルム和光純薬株式会社 Breast cancer determination method

Also Published As

Publication number Publication date
JP2005537790A (en) 2005-12-15
EP1539957A1 (en) 2005-06-15
EP1539957A4 (en) 2006-10-11
WO2004022750A1 (en) 2004-03-18

Similar Documents

Publication Publication Date Title
US20060147922A1 (en) Novel diagnostic and therapeutic methods and reagents therefor
EP2518163B1 (en) Prostate cancer specific alterations in erg gene expression and detection methods based on those alterations
Wang et al. Sprouty4, a suppressor of tumor cell motility, is downregulated by DNA methylation in human prostate cancer
US6596488B2 (en) Tumor suppressor gene
US6512102B1 (en) Compositions and methods of diagnosis and treatment using casein kinase I
US20050208486A1 (en) Brca-1 regulators and methods of use
US20040018525A1 (en) Methods and compositions for the prediction, diagnosis, prognosis, prevention and treatment of malignant neoplasma
JP2004159640A5 (en) Methods and compositions for predicting, diagnosing, prognosing, preventing and treating malignant tumors
WO2013141266A1 (en) Method for detecting novel ret fused body
JP2007512807A (en) Methods and compositions for predicting response to treatment of malignant tumors
JP2008505307A (en) HAUSP-Mdm2 interaction and use thereof
EP1654383A1 (en) Materials and methods for colorectal cancer screening, diagnosis, and therapy
JP2009165473A (en) Cancer
US8029989B2 (en) Method using snail transcriptional repressor
CA2497873A1 (en) Novel diagnostic and therapeutic methods and reagents therefor
Le Roux et al. Genetically manipulating endogenous Kras levels and oncogenic mutations in vivo influences tissue patterning of murine tumorigenesis
JP2006524492A (en) Methods for identifying nucleic acid molecules associated with angiogenesis
WO2013113942A2 (en) Novel fusion genes in lung cancer
AU2003257275A1 (en) Novel diagnostic and therapeutic methods and reagents therefor
US7964369B2 (en) Polypeptide complex that regulates cell cycle and anergy
US8084434B2 (en) Runx2 isoforms in angiogenesis
WO2001055450A2 (en) AMPLIFIED GENES AT 17q23
WO2008075713A1 (en) Marker for diagnosis of cancer, and target molecule for therapy
US8241846B1 (en) Hedgehog pathway modulation and uses thereof for treating, preventing and/or diagnosing cancer
US20140234321A1 (en) Hunk, a snf1-related kinase essential for mammary tumor metastasis

Legal Events

Date Code Title Description
FZDE Dead