WO2000040756A1 - Novel coactivator overexpressed in endocrine tumors - Google Patents

Novel coactivator overexpressed in endocrine tumors Download PDF

Info

Publication number
WO2000040756A1
WO2000040756A1 PCT/US2000/000288 US0000288W WO0040756A1 WO 2000040756 A1 WO2000040756 A1 WO 2000040756A1 US 0000288 W US0000288 W US 0000288W WO 0040756 A1 WO0040756 A1 WO 0040756A1
Authority
WO
WIPO (PCT)
Prior art keywords
coactivator
protein
ubiquitin
hormone
cells
Prior art date
Application number
PCT/US2000/000288
Other languages
French (fr)
Inventor
Bert W. O'malley
Ming-Jer Tsai
Sophia Y. Tsai
Zafar Nawaz
Original Assignee
Baylor College Of Medicine
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
Application filed by Baylor College Of Medicine filed Critical Baylor College Of Medicine
Priority to AU27207/00A priority Critical patent/AU2720700A/en
Publication of WO2000040756A1 publication Critical patent/WO2000040756A1/en

Links

Classifications

    • 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
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/86Viral vectors
    • 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
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2710/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA dsDNA viruses
    • C12N2710/00011Details
    • C12N2710/14011Baculoviridae
    • C12N2710/14041Use of virus, viral particle or viral elements as a vector
    • C12N2710/14043Use of virus, viral particle or viral elements as a vector viral genome or elements thereof as genetic vectore
    • 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
    • C12N2800/00Nucleic acids vectors
    • C12N2800/40Systems of functionally co-operating vectors
    • 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)

Definitions

  • endocrine tumors comprising assaying for a novel nuclear hormone receptor
  • agents comprising assaying treated cells for nuclear hormone coactivators. It furthermore,
  • Nuclear hormone receptors contain common structural motifs
  • HBD carboxy-terminal hormone-binding domain
  • AF-2 activation function-2
  • Nuclear hormone receptor coactivators represent a growing class of proteins
  • HAT acetyl transferase activity
  • the level of coactivator expression may contribute to
  • mice resulted in an attenuated response to steroid hormones, a finding consistent
  • ER and PR are members of a structurally related ligand-activated transcription factor superfamily. Upon ligand
  • E6-AP associated protein, E6-AP.
  • the coactivator protein, E6-AP enhances the hormone-
  • estrogen compounds can slow or prevent tumor spread. Many anti-estrogens,
  • anti-estrogen tamoxifen for example, which is established as the treatment of choice
  • tamoxifen and other anti-estrogens can have
  • E6-associated protein (E6-AP) (Huibregtse et al. (1993) Mol. Cell. Biol.
  • E6-AP is a member of a family of
  • Protein ubiquitination also involves two other classes of enzymes, namely El ubiquitin-activating enzymes and E2 ubiquitin-conjugating enzymes which
  • terminal 350 aa of E6-AP constitutes a 'hecf (homologous to the E6-AP carboxy
  • the extreme carboxyl-terminal 100 aa contains the catalytic region of E6-
  • a blood or other easily accessed tissue sample provides
  • gross deletions or deletions of the E6-AP gene can be detected with some reliability by fluorescence in situ hybridization (FISH) and
  • RFLP restriction fragment length polymorphism
  • RFLP can also be useful in detection of more limited deletions
  • PCR polymerase chain reaction
  • RFLP are generally predictive of the AS phenotype, they are capable of producing
  • FISH and RFLP may be unable to detect smaller partial deletions
  • a single point mutation can substantially alter E6-AP function without changing the
  • altered gene size such as FISH, RFLP, and PCR, would be unable to detect such a
  • a small deletion may be indicated by a PCR method which does not result in deficient E6-AP function. Such a false
  • This invention provides a method of treating or detecting endocrine tumors
  • the goal of this treatment is to reduce the hormone coactivating activity
  • nuclear coactivator proteins comprising (a) reducing the concentration of said
  • Methods for (a) above include, but are not limited to.
  • such a neutralizing protein will possess a transcription-incompetent
  • This invention further provides a method for screening tissues for endocrine
  • tumors comprising assaying for a nuclear hormone coactivator mRNA or protein.
  • samples from tissue biopsies are screened to determine if a tumor exists
  • this assay serves at least two functions. First, the assay serves
  • the assay provides
  • tumor growth may be indicated by other means but not by the above assay. In such an instance, the results
  • This invention further provides a method for the evaluation of possible anti-
  • tumor agents comprising assaying treated cells for nuclear hormone coactivators. It
  • the tumor suppressor gene product p53 is assayed for
  • tumor suppressor protein p53
  • This invention further provides a method for the evaluation of endocrine
  • tumorogenicity for possible tumor promoting agents comprising assaying treated cells
  • cultured cells expressing normal levels of coactivator mRNA or protein are exposed to an agent suspected as a promoter of
  • tumor promoting agents will increase coactivator
  • suppressor gene product p53 concentration is also measured. In the latter instance,
  • This invention further provides a method for the prenatal or postnatal
  • AS Angelman syndrome
  • tissue such as could be derived from
  • a decreased ubiquitin-ligase activity correlates with development of
  • This invention further provides a method for the detection of AS comprising
  • an assay for ubiquitin-ligase activity from a tissue or blood sample.
  • This invention is particularly useful in detecting breast tumors when the tissue sample is obtained from a mammary gland biopsy. This invention further provides a method
  • the tissue sample is obtained from a mammary gland biopsy.
  • Figure 1A shows interaction of PR with wild-type E6-AP in a yeast two-
  • Figure IB shows a Western blot analysis of E6-AP bound PR.
  • Figure 2 A shows a schematic representation of E6-AP showing the position
  • Figure 2B shows localization of the PR interaction site in E6-AP using the
  • Figure 3 A shows E6-AP coactivating the transcriptional activity of PR-B.
  • FIG. 3B shows that E6-AP coactivates the hormone-dependent
  • Figure 3C shows the effect of E6-AP expression on the transcriptional activity
  • Figure 4A shows E6-AP, but not a C-terminal mutant lacking the activation
  • FIG. 4B shows that the C-terminal fragment of E6-AP (aa 680-851) was
  • Figure 5 shows transcriptional activity of the GAL4-E6-AP fusion protein.
  • Figure 6 shows E6-AP levels in mouse mammary tumors.
  • Figure 7 shows E6-AP protein levels in mouse mammary glands.
  • This invention provides a method for treating endocrine tumors comprising
  • This invention further provides a method for screening tissues for
  • this invention provides a method for the identification of
  • AS Angelman syndrome
  • kit for the prenatal or postnatal detection of AS comprising an assay for
  • the objective of the anti-coactivator treatment is to reduce the hormone
  • E6- AP comprising of (a) reducing
  • tumor cells or (c) neutralizing the target protein(s) of said coactivator protein in
  • This embodiment further comprises the administration of a therapeutically
  • AP is capable of interacting with, and thus increasing transcriptional activity of, a
  • the preferred hormone receptor is the ER. While one of hormone receptors, the preferred hormone receptor is the ER. While one of the hormone receptors, the preferred hormone receptor is the ER. While one of the hormone receptors, the preferred hormone receptor is the ER. While one of the hormone receptors, the preferred hormone receptor is the ER. While one of the hormone receptors, the preferred hormone receptor is the ER. While one of the hormone receptors, the preferred hormone receptor is the ER. While one
  • a protein comprising the E6-AP binding domain of ER.
  • any naturally occurring protein possessing an E6-AP binding domain may be mutated
  • proteins may include, but are not limited to ER, PR, thyroid hormone receptor (TR),
  • RAR ⁇ retinoic acid receptor
  • AR androgen receptor
  • This embodiment also allows for a protein which binds the target protein E6-AP at
  • E6-AP comprises a truncated or mutant E6-AP protein which lacks the ability to enhance transcription of the ER.
  • E6-AP has been demonstrated to posses an intrinsic
  • activation domain located within amino acids 170 to 680. It is to be understood that
  • Nuclear receptors which are regulated by E6-
  • AP include, but are not limited to TR, PR, RAR ⁇ , AR, and GR.
  • the method comprises the insertion of DNA encoding the desired protein into a viral
  • Suitable viral vectors include, but are not
  • adenovirus adenovirus
  • retrovirus adenovirus
  • vaccinia virus adenovirus virus
  • viral vectors encoding partial or complete lengths of anti-sense mRNA to a
  • coactivator are used to insert complementary nucleotide sequences.
  • Suitable viral vectors are used to insert complementary nucleotide sequences.
  • vectors for this purpose include, but are not limited to adenovirus, retrovirus, and
  • Treatment of mammary tumors may also utilize any combination of some or
  • the anti-E6-AP treatment method can also be used in conjunction with other
  • the anti-E6-AP In an embodiment where mammary tumors are being treated, the anti-E6-AP
  • treatment method can be combined with an antihormone treatment method, including
  • E6- AP has been shown to directly promote degradation of the tumor suppressor gene
  • a preferred embodiment comprises assaying for increased
  • the increased level of E6-AP can also be a measured by
  • the assay for these factors may also comprise the determination of the
  • E6-AP mRNA levels of E6-AP mRNA are correspondingly increased in mammary tumor cells.
  • Such techniques may include,
  • Tumors can also be screened
  • present method is to provide a means to quickly evaluate a large number of agents as
  • a preferred embodiment comprises an
  • the tumor cell line can be selected from a group of cell lines including,
  • MCF-7 but not limited to, MCF-7, T47D, MDA-MB-231, LnCaP, PC3, and RUCA-1 cells.
  • method is to provide a means for evaluating a large number of agents for the ability to increase hormone coactivator mRNA or protein concentration when said agents are
  • a preferred embodiment comprises the measurement of E6-AP
  • a more preferred embodiment comprises the measurement of E6-AP mRNA
  • the cell type expresses E6-AP (mRNA and protein) and
  • embodiment can be selected from a group of cells including, but not limited to
  • fibroblasts e.g., fibroblasts, epithelial cells, COS cells, HeLa cells, blood cells (e.g. leukocytes,
  • tissue such as blood or cells obtained from amniocentesis, from a chorionic villus
  • Cells may
  • cell extract is added to a solution comprising ubiquitin ( ⁇ 8 KDa) and [ 35 S]methionine labeled target protein HHR23A (-80 KDa).
  • E6-AP +/- or E6-AP -/- facilitates
  • E6-AP +/+ protein standard may be
  • wild type human or animal donor cells including, but not limited to
  • tumors comprising an assay for ubiquitin-ligase activity with provided E6-AP +/+
  • ubiquitin-ligase activity is measured from a tissue sample using ubiquitin and [ 35 S]methionine
  • HHR23A labeled HHR23A as a target protein. It is to be understood that any of several
  • ubiquitin target proteins may be used. Furthermore, it is to be understood that any one of the following proteins listed in the following paragraphs:
  • the standard for comparison comprises wild type
  • comparison comprises wild type recombinant E6-AP protein.
  • AS or mutant cells, cell extract, or recombinant protein is provided as
  • the present invention provides
  • ubiquitin-ligase activity indicates that the tumor is cancerous.
  • the invention is a method that is particularly useful in detecting breast tumors when
  • the tissue sample is obtained from a mammary gland biopsy.
  • cancerous tumor comprising comparing the ubiquitin-ligase activity of tissue or blood
  • tested samples is indicative of progression or regression of said tumor.
  • One further embodiment of the method can be used to monitor the progression or regression of
  • tissue sample is obtained from a mammary gland biopsy.
  • the bait plasmid for the yeast two-hybrid system (pASl-PRLBD) (Onate et
  • reporter plasmids UAS4-TATA-LUC (LUC, luciferase) (Spencer et al.
  • pPRE/GRE-Elb-LUC and pERE-Elb-LUC were constructed by inserting PvuII-
  • E6-AP wild-type E6-AP (aal-851), 76 kDaE6-AP (aal 70-851), and C833-S mutant E6-AP (aal -851 ), the BamHI-Hindlll fragments of pGEM E6-AP (100 kDa), pGEM-E6-AP
  • AAATCC-3' lower strand
  • CTTCAACAA-3' (lower strand); (SEQ. ID. NO. 9) 5'-GCGGATCCAC
  • E6-AP forms of E6-AP were constructed by utilizing site directed mutagenesis to create
  • the BsaAI-Hindlll fragment of E6-AP was amplified by PCR with
  • GGACACTATCACCACCA-3' (lower strand) using AS patient DNA as a template.
  • E6-AP were subcloned in frame into the pABVPl ⁇ and pABGAL plasmids utilizing
  • GAL4 DNA-binding domain GAL-DBD-hPRA. Then the GAL-DBD-PRA
  • yeast cells were treated with either vehicle alone (-H), 10-6 M progesterone (+P), or 10-6 M RU486 (+RU).
  • vehicle alone (-H), 10-6 M progesterone (+P), or 10-6 M RU486 (+RU).
  • the ⁇ -galactosidase activity is expressed in Miller units and
  • the PR-B was expressed as a his-tagged
  • E6-AP was expressed in Escherichia coli cells and purified on glutathione-sepharose
  • PR-B in NETN buffer 50 mM Nacl, ImM EDTA, 20 mM Tris, pH 8.0, and 0.5%
  • E6-AP bound PR was analyzed by SDS-PAGE on a 7.5%
  • HeLa cells were maintained in DMEM supplemented with 10% fetal bovine
  • HeLa cells were transfected with 0.1 ⁇ g of PR-B expression plasmid, 1 ⁇ g of
  • (+) represents positive for ubiquitin-protein ligase activity and (-) represents negative for ubiquitin-protein ligase activity.
  • (*) represents natural mutants of E6-AP cloned
  • Figure 2 A is a schematic representation of E6-AP important for interaction with PR.
  • Wild-type E6-AP is a 100
  • KDa (aa 1-851) protein.
  • the C833S mutant represents a Cysteine 833 to Serine 833 mutant
  • the AS disease mutants are represented by the 98 kDa (deletion of
  • the I804K form of E6-AP contains lysine at position 804 instead of isoleucine
  • the F782 ⁇ contains a deletion of phenylalanine at position 782, and the 104 kDa (1-
  • E6-AP 885 ⁇ stop form of E6-AP is a read through mutant.
  • E6-AP were fused to the VP 16 activation domain and the ability of the VP 16-E6-AP
  • hybrid proteins to interact with receptor was determined in the absence or presence
  • progesterone As shown in Fig. 2B, wild-type E6-AP, N-terminal deletion fragments (aa 170-851 and aa 680-851 ) and C-terminal deletion fragments (aa 1-714
  • E6-AP cDNA and the N-terminal fragment (aa 1-240) of E6-AP did not interact
  • One site is located within the C-terminal (aa 680-851) (21 kDa) fragment of E6-AP,
  • the second PR interaction site is located within aa 240-449 and overlaps with the E6
  • PR has a minimal
  • E6-AP was able to stimulate the hormone-dependent
  • HeLa cells is not dependent on the E6 protein.
  • GR retinoic acid
  • TR thyroid hormone receptor
  • HeLa cells were transfected with
  • EXAMPLE 10 E6-AP Contains an Intrinsic Activation Domain
  • DBD DNA-binding domain
  • E6-AP full-length E6-AP (aa 1-851), N-terminal deletion fragments of E6-AP (aa 170-851 and aa 680-851) and C-terminal deletion fragment of E6-AP (aa 1-714) (shown in
  • Figure 1 A were fused to the yeast GAL4 DBD. HeLa cells were then transfected
  • AP expression plasmids (1.0 ⁇ g). The data are expressed as fold activation and each
  • a transcriptional activation domain located between aa 170-680.
  • Glycerol 1 mM PMSF, and protease inhibitors Leupeptin (1.25 g/ml) and
  • Pepstatin A (1.0 g/ml). Protein concentrations were estimated using the Bradford
  • E6-AP expression of E6-AP was associated with tumor progression because E6-AP
  • EXAMPLE 12 E6-AP Contains Two Independent Separable Functions, Coactivation and Ubiquitin-Ligase Activity
  • E6-AP is a ubiquitin-protein ligase
  • E6-AP activity of E6-AP is not required for the coactivation function of E6-AP.
  • E6-AP possesses two independent, separable functions; coactivation and
  • This mutant E6-AP was able to give different amino acids from the new reading frame.
  • HHR23A ubiquitinate a protein
  • missense mutant (I804K), was able to ubiquitinate the target protein HHR23 A, to

Abstract

This invention provides screening and treatment methods for endocrine tumors based on abnormally elevated hormone coactivator levels. In addition, the invention provides a vector delivery system for the introduction of proteins and nucleotide sequences into cells for the purpose of altering the expression or effectiveness of hormone coactivators. The invention also provides methods for the evaluation of agents as possible endocrine tumor growth regulators. Also, methods are provided for evaluating the tumor promoting capacity of environmental agents. Furthermore, methods are provided for the prenatal or postnatal detection of Angelman syndrome. These methods comprise the measurement of ubiquitin-ligase activity in a tissue sample. The invention also provides methods for the detection of coactivator, E6-AP in mammary gland biopsy samples using a ubiquitin-ligase assay.

Description

NOVEL COACTIVATOR OVEREXPRESSED IN ENDOCRINE TUMORS
This application claims the benefit of U.S. Provisional Application Serial No. 60/115,025, filed January 7, 1999.
The work herein was supported, in part, by grants from the United States
Government. The United States Government may have certain rights in the
invention.
FIELD OF THE INVENTION
This invention relates to the fields of biotechnology and medicine and
provides methods for treatment of endocrine tumors comprising interfering with a
hormone coactivator pathway. It further relates to a method for screening tissues for
endocrine tumors comprising assaying for a novel nuclear hormone receptor
coactivator. It further relates to a method for the evaluation of possible anti-tumor
agents comprising assaying treated cells for nuclear hormone coactivators. It further
relates to a method for the evaluation of tumorogenicity of environmental agents
comprising assaying exposed cells for nuclear hormone coactivators. It further
relates to a method for the prenatal or postnatal detection of Angelman syndrome
comprising a ubiquitin-ligase activity assay. It further relates to a method for the
detection of Angelman syndrome comprising a ubiquitin-ligase assay. It additionally
relates to a methods of tumor detection and monitoring comprising use of a ubiquitin-
ligase assay. BACKGROUND OF THE INVENTION
Steroids, thyroid hormones, vitamin D and retinoids regulate diverse
biological processes including growth, development, and homeostasis through their
cognate nuclear hormone receptors which make up a superfamily of structurally
related intracellular ligand-activated transcription factors. Horwitz et al. (1996),
Mol. Endocrinol. 10: 1167-1177; Perlmann et al. (1997), Cell 90: 391-397; Shibata
et al. (1997), Recent. Prog. Hormone Res. 52: 141-165; Tsai et al. (1994), Ann. Rev.
Biochem.63: 451-486. Nuclear hormone receptors contain common structural motifs
which include a less well conserved amino-terminal activation function (AF-1) that
affects transcription efficiency, a central DNA-binding domain, which mediates
receptor binding to specific DNA enhancer sequences and determines target gene
specificity, and a carboxy-terminal hormone-binding domain (HBD). The HBD
contains activation function-2 (AF-2), a region which mediates the hormone-
dependent activation function of receptors. Shibata et al. (1997), Recent. Prog.
Hormone Res. 52: 141-165. When bound to hormone, these receptors undergo a
conformational change, dissociation from heat shock proteins, receptor dimerization,
phosphorylation, DNA-binding at an enhancer element of the target gene, interaction
with coactivators and subsequent recruitment of basal transcription factors to form
a stable preinitiation complex. These events are followed by either up-regulation or
down-regulation of target gene transcription. Shibata et al. (1997), Recent. Prog.
Hormone Res. 52: 141-165. Nuclear hormone receptor coactivators represent a growing class of proteins
which interact with receptors in a ligand-specific manner and serve to enhance their
transcriptional activity. Onate et al. (1995), Science 270: 1354-1357. Prior to their
identification, coactivators were predicted to exist based upon experiments which
showed that different receptors compete for a limiting pool of factors required for
optimal transcription. Stimulation of one receptor resulted in transrepression of
another receptor, indicating the depletion of a common coactivator pool. Bocquel et
al. (1989), Nuc. Acids Res. 17: 2581-2594; Conneely et al. (1989), In A. K. Roy and
J. Clark (ed.), Gene regulation by steroid hormones IV, p. 220-223, Spring-Verlag
Press, New York; Meyer et al. (1989), Cell 57: 433-442; Shemshedini et al. (1992),
J. Biol. Chem. 261 : 1834-1839. A number of coactivators have been cloned to date,
including steroid receptor coactivator- 1 (SRC-1) Onate et al. (1995), Science 270:
1354-1357; TIF2 (GRIP1) Hong et al. (1996), Proc. Natl Acad. Sci. USA. 93:
4948-4952; Voegel et al. (1996), EMBO J. 15: 3667-3675; p/CIP
(ACTR/RAC3/AIB1/TRAM-1) Anzick et al. (1997), Science 277: 965-968; Chen
et al. (1997), Cell 90: 569-580; Li et al. (1997), Proc. Natl. Acad. USA. 94: 8479-
8484; Torchia et al. (1997), Nature 387: 677-684; Tukeshita et al. (1997), J. Biol.
Chem. 272: 27629-27634; and ARA70 Yeh et al. (1996), Proc. Natl. Acad. Sci. USA.
93: 5517-5521. Coactivators were originally envisioned to serve a bridging role,
linking the receptor to the basal transcription machinery. Pugh et al. (1992), J. Biol.
Chem. 267: 679-682; Tjian et al. (1994), Cell 11: 5-8. Recently, the functional role
of coactivators has expanded by the observation that they have been shown to possess enzymatic activities which contribute to their ability to enhance receptor
mediated transcription; SRC-1, p300/CBP, and RAC3/ACTR/AIB 1 possess histone'
acetyl transferase activity (HAT). Anzick et al. (1997), Science 277: 965-968; Chen
et al. (1997), Cell 90: 569-580; Li et al. (1997), Proc. Natl. Acad. USA. 94: 8479-
8484; Ogryzko et al. (1996), Cell 87: 953-959; Spencer et al. (1997), Nature 389:
194-198. Ligand-activated receptors are thought to bring these HAT activity
containing coactivators to the chromatin surrounding the receptor, disrupting the local
repressive chromatin structure by acetylating histones and possibly other chromatin
associated factors. Spencer et al. (1997), Nature 389: 194-198.
Due to their ability to enhance receptor mediated gene expression,
coactivators are thought to play an important role in regulating the magnitude of the
biological response to steroids, vitamin D, and retinoids in different tissues or from
individual to individual. The level of coactivator expression may contribute to
variations in hormone responsiveness seen in the population and disruption in
coactivator expression could lead to the pathologically hyper- or hypo-sensitivity to
steroid hormones. Recently, it has been shown that disruption of the SRC-1 locus in
mice resulted in an attenuated response to steroid hormones, a finding consistent
with this hypothesis. Xu et al. (1998), Science 279:1922-1924.
Steroid hormones, estrogen and progesterone, play major roles in the
development of the normal mammary gland and in breast tumor development. These
molecules mediate their signaling through intracellular receptors called estrogen
receptors (ER) and progesterone receptors (PR). ER and PR are members of a structurally related ligand-activated transcription factor superfamily. Upon ligand
binding, these factors undergo a conformational change, dissociate from heat shock
proteins and recruit a diverse group of coactivator proteins, including that of E6-
associated protein, E6-AP. The coactivator protein, E6-AP, enhances the hormone-
dependent transcriptional activity of ER, PR, and other members of the steroid
receptor superfamily. Considering the influence of E6-AP as a coactivator on
transactivation of target genes by ER, PR, and other members of the superfamily, the
expression pattern of coactivator E6-AP in mouse mammary tumors was examined.
The data indicates that coactivator E6-AP is overexpressed in 90-95% of breast
tumors in mouse tumor models, which indicates a role for E6-AP in tumor formation.
Many breast tumors require estrogens for tumor growth. Thus, treatment with anti-
estrogen compounds can slow or prevent tumor spread. Many anti-estrogens,
however, show both estrogen antagonistic and agonistic activity. The non-steroidal
anti-estrogen tamoxifen, for example, which is established as the treatment of choice
for the endocrine therapy of advanced breast cancer, shows both agonistic and
antagonistic activity. Sutherland, S.& Jackson, M. Cancer Treat. Revs. 15:183-194
(1987).
The prior art fails to provide a completely satisfactory treatment for endocrine
tumors. The agonistic activity of tamoxifen and other anti-estrogens can have
profound negative effects upon patients. Tamoxifen for example sometimes
increases endometrial tumor incidence, e.g. , lino, et al. Cancer Treat. & Res. 53 :228-
237 (1991), or switches from inhibition to stimulation of estrogen dependent growth in breast tumor progression. Parker, M.G. (Ed) Cancer Surveys 14: Growth
Regulation by Nuclear Hormone Receptors. Cold Spring Harbor Laboratory Press"
(1992). Therefore, a need for more selective treatment methods still exists.
The E6-associated protein (E6-AP) (Huibregtse et al. (1993) Mol. Cell. Biol.
13: 775-784.), a protein linked to Angelman syndrome (AS) (Kishino et al. (1993),
Nature Genet. 15: 70-73; Matsuura et al. (1997), Nature Genet. 15: 74-77; Sutcliff
et al. (1997), Genome Res. 1: 368-377) is a progesterone receptor (PR) interacting
protein which has been cloned and characterized. E6-AP was previously identified
as a protein of 100 kDa, present both in the cytoplasm and the nucleus (Hatakeyama
et al. (1997), J. Biol Chem. 272: 15085-15092.). E6-AP mediates the interaction of
human papillomaviruses type 16 and 18 E6 proteins with p53, a growth-suppressive
and tumor-suppressive protein. Hatakeyama etal. (1997), J. Biol Chem. 212: 15085-
15092; Huibregtse et al. (1993), Mol. Cell. Bio. 13: 4918-4927. Initial in vitro
studies suggested that the E6/E6-AP complex specifically interacts with p53 and
promotes the degradation of p53 via the ubiquitin-proteasome degradation pathway
but recent in vivo studies show that E6-AP can directly interact with p53 and promote
its degradation even in the absence of the papillomavirus E6 protein. Daniel et al.
(1998), J. Gen. Virol. 79: 489-499; Huibregtse et al. (1991), EMBO J. 10: 4129-
4135; Scheff er et al. (1993), Cell 75: 495-505. E6-AP is a member of a family of
proteins known as E3 ubiquitin-protein ligases which have been proposed to play a
role in defining the substrate specificity of the ubiquitin-proteasome degradation
system. Protein ubiquitination also involves two other classes of enzymes, namely El ubiquitin-activating enzymes and E2 ubiquitin-conjugating enzymes which
activate ubiquitin moities and transfer them to target proteins or E3's respectively.
Huibregtse et al. (1995), Proc. Natl. Acad. Sci. USA. 92: 2563-2567. The carboxyl-
terminal 350 aa of E6-AP constitutes a 'hecf (homologous to the E6-AP carboxy
terminus) domain which is conserved among many E3 ubiquitin protein-ligases and
E6-AP related proteins. Huibregtse et al. (1995), Proc. Natl. Acad. Sci. USA. 92:
2563-2567. The extreme carboxyl-terminal 100 aa contains the catalytic region of E6-
AP which transfers ubiquitin to the protein targeted for degradation. Huibregtse et
al. (1995), Proc. Natl. Acad. Sci. USA. 92: 2563-2567. The E6-binding domain
consists of an 18 aa region located within the central portion of the E6-AP protein
Huibregtse et al. (1993), Mol. Cell. Bio. 13: 4918-4927.
Recently, it has been shown that a genetic disorder, Angelman syndrome
(AS), is caused by the absence of a functional maternal copy of the E6-AP gene.
Kishino etal. (1993),Nαtwre Genet. 15: 70-73; Matsuura et al. (1991), Nature Genet.
15: 74-77; Sutcliff et al. (1997), Genome Res. 1: 368-377. AS is a neurological
disorder characterized by severe mental retardation, seizures, speech impairment, and
other symptoms. Beuten et al. (1996), Human Genet. 97: 294-298. Due to paternal
gene imprinting in the brain, only the defective maternally derived gene is expressed.
However, outside the brain, both the paternal and the maternal gene is expressed.
Thus, expression of the single functional paternal copy allows for partial
compensation throughout the rest of the body. While the exact mechanism by which the defective E6-AP gene causes AS
remains unknown, analysis of mutant E6-AP proteins from AS patients revealed that
the ubiquitin-protein ligase function of E6-AP was defective. The coactivator
function, however, was intact in the majority of the examined AS patients, but in the
same patients, both coactivation and ubiquitin-ligase functions are defective. It was
also demonstrated that the ubiquitin-ligase activity of E6-AP was not required for the
coactivation function of E6-AP. The data further indicated that the catalytic function
located within the hect domain of E6-AP was not necessary for the ability of E6-AP
to interact with and coactivate steroid hormone receptor function. These findings
indicate that E6-AP possesses two independent functions, as both a coactivator and
a ubiquitin-protein ligase.
Since patients with AS express either zero (brain) or one (rest of body)
functional E6-AP gene, tissues derived from these patients, except for brain samples,
possess lower ubiquitin-ligase activity compared to tissues expressing two functional
gene copies. On rare occasion, an AS patient will express zero functional E6-AP
genes peripherally. While brain tissue samples posses the greatest reduction in
ubiquitin-ligase activity, non-brain tissues posses sufficiently reduced activity such
that a lowered activity can be reliably detected compared to tissues possessing two
functional gene copies. Thus, a blood or other easily accessed tissue sample provides
adequate protein for assay.
Current methods for screening patients for AS involve screening DNA for
gross deletions or deletions of the E6-AP gene. For instance, gross deletions can be detected with some reliability by fluorescence in situ hybridization (FISH) and
restriction fragment length polymorphism (RFLP). Robson et al. (1995), Hum.
Genet., 96:345-349). RFLP can also be useful in detection of more limited deletions,
such as of the E6- AP gene. Smaller deletions within the E6-AP gene can be detected
using polymerase chain reaction (PCR) techniques to amplify DNA fragments of a
predicted size. Saitoh, S. (1993), No To Hattatsu, 25:501-507.
However, there are problems inherent to the above methods for AS detection.
While gross deletions or gene deletions detected by techniques such as FISH and
RFLP are generally predictive of the AS phenotype, they are capable of producing
false negative results (for example, see Robson et al. (1995), Hum. Genet., 96:345-
349). For instance, FISH and RFLP may be unable to detect smaller partial deletions
of critical regions of the E6-AP gene. However, while PCR techniques should
indicate most smaller DNA deletions or insertions undetected by FISH and RFLP,
even the PCR techniques cannot not readily detect point mutations within the gene.
A single point mutation can substantially alter E6-AP function without changing the
size of a predicted DNA product. As an example, changing a single cysteine residue
at position 833 to alanine or serine eliminates ubiquitin-ligase activity. Huibregtse
et al. (1995), Proc. Natl. Acad. Sci. USA. 92: 2563-2567. Methods which rely on an
altered gene size, such as FISH, RFLP, and PCR, would be unable to detect such a
change. In addition to the shortcomings in the prior art to predict AS when small
gene deletions or point mutations is the cause (false negatives), the prior art is also
susceptible to false positive results. For instance, a small deletion may be indicated by a PCR method which does not result in deficient E6-AP function. Such a false
positive diagnosis would be equally devastating as a false negative diagnosis. Given
that some parents might choose to terminate a pregnancy based on the findings of a
prenatal screening (See e.g., Toth-Fejel et al. (1995), Am. J. Med. Genet., 55:444-
452), it is crucial that the results be as accurate as possible. One significant
advantage of the ubiquitin-ligase activity assay is that the actual function of the E6-
AP protein is evaluated, thus adding certainty to the diagnosis. The ubiquitin-ligase
activity alone or in conjunction with existing DNA screening techniques would thus
be a significant improvement over the prior art.
BRIEF SUMMARY OF THE INVENTION
This invention provides a method of treating or detecting endocrine tumors
comprising interfering with a hormone coactivator pathway (anti-coactivator
method). The goal of this treatment is to reduce the hormone coactivating activity
of nuclear coactivator proteins comprising (a) reducing the concentration of said
protein in tumor cells, or (b) neutralizing said protein in tumor cells, or (c)
neutralizing the target protein(s) of said coactivator protein in tumor cells, or (d) any
combination of the above. It is to be understood that the invention is not to be limited
by the above examples. Methods for (a) above include, but are not limited to.
introducing oligonucleotides complementary to a region of selected coactivator
mRNA or introducing a chemical substance which reduces expression or stability of
selected coactivator mRNA or protein, or reducing the effective level of coactivator protein by neutralization with antibodies (see below). Methods for (b) and (c) above
include, but are not limited to, introducing a protein into the tumor cell which binds
or neutralizes a coactivator protein or a target protein for a coactivator protein. It is
preferred that such a neutralizing protein will possess a transcription-incompetent
fragment of a hormone receptor which binds the coactivator protein ('false substrate')
or a mutant of the coactivator which binds to the hormone receptor but which is
incapable of increasing transcription of the hormone receptor ('dominant negative').
It is expected that the anti-coactivator treatment method will be used in conjunction
with other endocrine tumor treatments, including but not limited to antihormones.
In doing so, it is expected that the combined treatment process will act synergistically
thereby increasing the effectiveness of the treatment and possibly reducing the dose
of one or both anti -tumor agents required.
This invention further provides a method for screening tissues for endocrine
tumors comprising assaying for a nuclear hormone coactivator mRNA or protein. In
this method, samples from tissue biopsies are screened to determine if a tumor exists
which is characterized as having high coactivator protein levels. In greater than 90%
of mouse mammary tumors evaluated, the coactivator E6-AP was found to be
overexpressed. Thus, this assay serves at least two functions. First, the assay serves
as a screening tool for a group of endocrine tumors. Second, the assay provides
information regarding the level of coactivators in the tissue. It follows that higher
than normal coactivator levels in a tissue sample indicates a tumor growth which is
a candidate for anti-coactivator treatment. In other instances, tumor growth may be indicated by other means but not by the above assay. In such an instance, the results
may suggest that an anti-coactivator treatment may not be effective, thus sparing a
patient from enduring an ineffective treatment.
This invention further provides a method for the evaluation of possible anti-
tumor agents comprising assaying treated cells for nuclear hormone coactivators. It
is an object of the present invention to provide a means of evaluating a large number
of agents for the ability to reduce the concentration, or reduce the coactivation
functions, of coactivators. In this method, cultured tumor cells are used to screen a
large number of possible anti-tumor agents for the ability to decrease coactivator
mRNA, protein concentration, or the coactivation function. Agents which
demonstrate the ability to reduce coactivator concentration, or the coactivation
function, can then be tested in vivo for the ability to inhibit tumor growth. In another
embodiment of this method, the tumor suppressor gene product p53 is assayed for,
in addition to the specific coactivator. Certain coactivators (such as E6-AP) have
been shown to directly promote degradation of p53. Thus, in the latter instance, a
promising anti-tumor agent will result in lowered levels of the coactivator and
increased levels of tumor suppressor protein, p53.
This invention further provides a method for the evaluation of endocrine
tumorogenicity for possible tumor promoting agents comprising assaying treated cells
for nuclear hormone coactivators. It is an object of the present invention to provide
a means for the evaluation of a large number of agents for the propensity to promote
endocrine tumors. In this method, cultured cells expressing normal levels of coactivator mRNA or protein are exposed to an agent suspected as a promoter of
endocrine tumors. Presumed tumor promoting agents will increase coactivator
mRNA or protein concentration. In another embodiment of this method, tumor
suppressor gene product p53 concentration is also measured. In the latter instance,
presumed tumor promoting agents will increase coactivator mRNA or protein
concentrations and decrease p53 mRNA or protein concentrations.
This invention further provides a method for the prenatal or postnatal
detection of Angelman syndrome (AS) by assaying a tissue or blood sample for
ubiquitin-ligase activity. Small amounts of tissue, such as could be derived from
amniocentesis or a blood drawing, are sufficient for the determination of ubiquitin-
ligase activity. A decreased ubiquitin-ligase activity correlates with development of
the AS phenotype. A significant advantage of this method over prior art methods is
that the protein function is actually measured rather than predicted based on DNA
alterations.
This invention further provides a method for the detection of AS comprising
an assay for ubiquitin-ligase activity from a tissue or blood sample. Such an assay
is capable of detecting reduced or lowered ubiquitin-ligase activity from a tissue
lacking expression of one or both functional E6-AP genes. The invention also
provides a method for assaying for the presence of tumors by comparing the
ubiquitin-ligase activity of a tissue or blood sample to a non-tumor control, where a
decrease in the ubiquitin-ligase activity indicates that the tumor is cancerous. The
method is particularly useful in detecting breast tumors when the tissue sample is obtained from a mammary gland biopsy. This invention further provides a method
of monitoring the progression or regression of a cancerous tumor comprising
comparing the ubiquitin-ligase activity of tissue or blood samples to a non-tumor
control and previously tested samples, wherein a change in the ubiquitin-ligase
activity over time compared to the control and the previously tested samples is
indicative of progression or regression of said tumor. One version of this latter
method can be used to monitor thr progression or regression of a breast tumor when
the tissue sample is obtained from a mammary gland biopsy.
BRIEF DESCRIPTION OF THE DRAWINGS
Figure 1A shows interaction of PR with wild-type E6-AP in a yeast two-
hybrid assay as indicated by β-Gal activity.
Figure IB shows a Western blot analysis of E6-AP bound PR.
Figure 2 A shows a schematic representation of E6-AP showing the position
of the catalytic region (solid box), hect domain, transactivation domain, and PR-
binding domains.
Figure 2B shows localization of the PR interaction site in E6-AP using the
mammalian two-hybrid assay.
Figure 3 A shows E6-AP coactivating the transcriptional activity of PR-B.
Figure 3B shows that E6-AP coactivates the hormone-dependent
transcriptional activity of nuclear hormone receptors. Figure 3C shows the effect of E6-AP expression on the transcriptional activity
of diverse transcription factors.
Figure 4A shows E6-AP, but not a C-terminal mutant lacking the activation
domain reverses the transcriptional squelching between PR and ER.
Figure 4B shows that the C-terminal fragment of E6-AP (aa 680-851) was
unable to reverse the transcriptional interference between PR and ER.
Figure 5 shows transcriptional activity of the GAL4-E6-AP fusion protein.
Figure 6 shows E6-AP levels in mouse mammary tumors.
Figure 7 shows E6-AP protein levels in mouse mammary glands.
DETAILED DESCRIPTION OF THE INVENTION
This invention provides a method for treating endocrine tumors comprising
interfering with the function of certain hormone coactivators ('anti-coactivator'
treatment). This invention further provides a method for screening tissues for
endocrine tumors based on increased hormone coactivator levels. This method is also
useful as a means of identifying tumors which might be responsive to anti-coactivator
treatment. Additionally, this invention provides a method for the identification of
agents which affect the concentration of coactivators. The above mentioned methods
can be applied to multiple endocrine tumors including, but not limited to mammary,
prostate, and endometrial tumors. In another embodiment of this invention, a
screening method is provided for the evaluation of endocrine tumorogenicity for
suspected tumor promoting agents. In yet another embodiment of this invention, a method is provided for prenatal or postnatal detection of Angelman syndrome (AS)
comprising the assay of an accessible tissue for ubiquitin-ligase activity. Finally, a
kit for the prenatal or postnatal detection of AS is provided comprising an assay for
ubiquitin-ligase activity.
The objective of the anti-coactivator treatment is to reduce the hormone
coactivating activity of nuclear coactivator protein, E6- AP comprising of (a) reducing
the concentration of said protein in tumor cells or (b) neutralizing said protein in
tumor cells or (c) neutralizing the target protein(s) of said coactivator protein in
tumor cells or (d) any combination of the above. The term "neutralizing" in this
context is meant to indicate that a protein is rendered incapable of performing a
certain function, such as binding with another protein or facilitating transcription. It
is to be understood that the treatment is not to be limited by the above examples.
One embodiment of the method comprises the administration of a therapeutically
effective concentration of oligonucleotides to the tumor cells which are
complementary to the E6-AP mRNA. A person skilled in the art will recognize an
oligomer of appropriate length and hybridizing location. This embodiment also
allows for the use of more than one oligomer coding for several regions of E6-AP
mRNA. This embodiment further comprises the administration of a therapeutically
effective concentration of a chemical substance which inhibits the formation of E6-
AP, promotes the degradation of E6-AP, or both.
Another embodiment of the method comprises the administration of a
therapeutically effective concentration of a protein which binds E6-AP, thus preventing E6-AP from effectively interacting with a hormone receptor. While E6-
AP is capable of interacting with, and thus increasing transcriptional activity of, a
number of hormone receptors, the preferred hormone receptor is the ER. While one
skilled in the art will recognize that a number of proteins can serve this purpose, the
preferred embodiment is for a protein comprising the E6-AP binding domain of ER.
Such a protein will be sufficiently truncated or mutated so as to be ineffective as an
activator of transcription. An ER mutant protein in which the DNA binding domain
has been mutated or deleted is but one example. It is to be further understood that
any naturally occurring protein possessing an E6-AP binding domain may be mutated
or truncated in order to serve as a binding site for E6-AP. Such naturally occurring
proteins may include, but are not limited to ER, PR, thyroid hormone receptor (TR),
retinoic acid receptor (RARα), androgen receptor (AR), and glucocorticoid receptor
(GR).
Another embodiment of the method comprises the administration of a
therapeutically effective concentration of a protein which competes for the binding
site on the target protein for E6-AP, thus preventing the binding of native E6-AP.
This embodiment also allows for a protein which binds the target protein E6-AP at
a different site but which prevents binding of E6-AP. The preferred embodiment
comprises the delivery of a protein which binds to the E6-AP binding site of the ER.
One skilled in the art will recognize that numerous proteins can be made which will
bind to the E6-AP binding site on the ER. The preferred ER-binding protein
comprises a truncated or mutant E6-AP protein which lacks the ability to enhance transcription of the ER. E6-AP has been demonstrated to posses an intrinsic
activation domain located within amino acids 170 to 680. It is to be understood that
other receptors can be targeted as well. Nuclear receptors which are regulated by E6-
AP include, but are not limited to TR, PR, RARα, AR, and GR. One embodiment of
the method comprises the insertion of DNA encoding the desired protein into a viral
vector and then injecting a therapeutically effective concentration directly into or into
the immediate vicinity of tumor cells. Suitable viral vectors include, but are not
limited to, adenovirus, retrovirus, and vaccinia virus.
Another embodiment comprises the delivery of a therapeutically effective
concentration of oligonucleotides to the tumor cells. One skilled in the art will
recognize that the uptake of these oligonucleotides can be affected by several means
including adding nucleotides to the oligomers which facilitate uptake into the cells
but which are not complementary to the coactivator mRNA. In another embodiment,
viral vectors encoding partial or complete lengths of anti-sense mRNA to a
coactivator are used to insert complementary nucleotide sequences. Suitable viral
vectors for this purpose include, but are not limited to adenovirus, retrovirus, and
vaccinia virus.
Another embodiment comprises the delivery of a therapeutically effective
concentration of E6-AP neutralizing antibodies to the tumors. These antibodies can
be administered by any one of a number of methods well known in the art for
administering antibodies. Treatment of mammary tumors may also utilize any combination of some or
all of the above methods.
The anti-E6-AP treatment method can also be used in conjunction with other
endocrine tumor treatments. In doing so, the combined treatment process will act
synergistically thereby increasing the effectiveness of the treatment and possibly
reducing the dose of one or both anti -tumor agents required.
In an embodiment where mammary tumors are being treated, the anti-E6-AP
treatment method can be combined with an antihormone treatment method, including
but not limited to, tamoxifen. Such a combined treatment method can increase the
effectiveness compared to individual treatments alone. Alternatively, it can reduce
the concentration requirement for one or more treating agents. This latter expectation
is especially significant given the possible tumor promoting activity of tamoxifen in
the uterus.
Another embodiment comprises assaying tissue samples for increased levels
of E6-AP mRNA or protein. In >90% of mouse mammary tumors currently
evaluated, higher than normal concentrations of E6-AP protein were found. Also,
E6- AP has been shown to directly promote degradation of the tumor suppressor gene
product, p53. Therefore, a preferred embodiment comprises assaying for increased
levels of E6-AP and decreased levels of p53. This assay can be accomplished with
specific monoclonal or polyclonal antibodies to E6-AP, or any of several methods
well known in the art. The increased level of E6-AP can also be a measured by
utilizing a ubiquitin-protein ligase assay of the biopsy samples. The assay for these factors may also comprise the determination of the
relative amount of mRNA in a given tissue sample. It has been demonstrated that
levels of E6-AP mRNA are correspondingly increased in mammary tumor cells. One
skilled in the art will recognize that a number of techniques are available which are
capable of measuring relative mRNA concentrations. Such techniques may include,
but are not limited to RT-PCR and Northern blot hybridization. It is to be understood
that several endocrine tumors can be screened for with this method, including but not
limited to mammary, endometrial, and prostate tumors. Tumors can also be screened
by studying the ubiquitin-ligase activity of E6-AP from mammary biopsy samples.
Another embodiment comprises a method for the evaluation of possible anti-
tumor agents for the ability to alter hormone coactivator levels. The object of the
present method is to provide a means to quickly evaluate a large number of agents as
possible endocrine tumor therapeutics. A preferred embodiment comprises an
endocrine tumor cell line which is contacted with a possible anti-tumor agent and
then assayed for E6-AP. The above mentioned method for assay can be used.
Another preferred embodiment comprises a tumor cell line which is contacted with
a possible anti-tumor agent and then assayed for E6-AP and p53. In this
embodiment, the tumor cell line can be selected from a group of cell lines including,
but not limited to, MCF-7, T47D, MDA-MB-231, LnCaP, PC3, and RUCA-1 cells.
Another embodiment comprises a method for the evaluation of endocrine
tumorogenicity for selected tumor promoting agents. The object of the present
method is to provide a means for evaluating a large number of agents for the ability to increase hormone coactivator mRNA or protein concentration when said agents are
contacted with a cell. A preferred embodiment comprises the measurement of E6-AP
mRNA or protein within a cell following contact with a suspected tumor promoting
agent. A more preferred embodiment comprises the measurement of E6-AP mRNA
or protein and p53 protein within a cell following contact with a suspected tumor
promoting agent. Any agent which increases levels of E6-AP (mRNA or protein) and
decreases levels of p53 (protein) will be considered to be a possible tumor promoting
factor. In this embodiment, the cell type expresses E6-AP (mRNA and protein) and
p53 (protein) within a normal concentration range. Cell types for use in this
embodiment can be selected from a group of cells including, but not limited to
fibroblasts, epithelial cells, COS cells, HeLa cells, blood cells (e.g. leukocytes,
monocytes, etc.), and mammary-derived cells which do not express high levels of
E6-AP.
Another embodiment consists of a method for prenatal or postnatal detection
of AS comprising the measurement of ubiquitin-ligase activity from a tissue sample
and comparing that value to a control sample. AS has been shown to correlate well
with decreased ubiquitin-ligase activity. A preferred embodiment of this method
comprises the evaluation of ubiquitin-ligase activity from a relatively easily accessed
tissue such as blood or cells obtained from amniocentesis, from a chorionic villus
sampling, or isolation of fetal cells present within the mother' s circulation. Cells may
also be obtained directly from the fetus through procedures including fetoscopy. In
one embodiment of this method, cell extract is added to a solution comprising ubiquitin (~8 KDa) and [35S]methionine labeled target protein HHR23A (-80 KDa).
Ubiquitin attachment occurs during the incubation period. Normal/wild type E6-AP
(E6-AP +/+) facilitates the attachment of several ubiquitin molecules in a
multiubiquitin chain whereas AS/mutant E6-AP (E6-AP +/- or E6-AP -/-) facilitates
attachment of fewer or no ubiquitin molecules. Following the incubation period, the
products are analyzed by SDS-PAGE and autoradiography. SDS-PAGE results in a
"ladder" consisting of products of approximately 80-200 KDa., reflecting the
attachment of varying numbers of ubiquitin to the labeled target protein. E6-AP +/+
results in a significantly greater proportion of larger, polyubiquinated products
compared to AS E6-AP +/- or E6-AP -/- protein. E6-AP +/+ protein standard may be
obtained from wild type human or animal donor cells including, but not limited to
blood, fibroblasts, epithelial cells, COS-7 cells, HeLa cells, or from recombinant wild
type E6-AP protein. Production of ubiquitin/target protein complexes of a
significantly smaller size is indicative of defective E6-AP which is responsible for
the AS phenotype. One skilled in the art will recognize that any of several methods
well known in the art are suitable for discriminating between protein complexes of
different sizes. It is to be understood that the present method for detecting AS can
be used alone or in combination with other methods for the detection of AS.
Yet another embodiment provides an assay kit for the prenatal or postnatal
detection of AS, and for the detection of coactivator dependent mammary gland
tumors, comprising an assay for ubiquitin-ligase activity with provided E6-AP +/+
standard for comparison. In a preferred embodiment of this product, ubiquitin-ligase activity is measured from a tissue sample using ubiquitin and [35S]methionine
labeled HHR23A as a target protein. It is to be understood that any of several
ubiquitin target proteins may be used. Furthermore, it is to be understood that any
of several means well known in the art may be used to label and/or otherwise isolate
said target protein. In another embodiment, recombinant E6-AP protein is produced
from a tissue sample and ubiquitin-ligase activity measured and compared to a
standard. In one embodiment, the standard for comparison comprises wild type
human or animal cells or cell extract. In another embodiment, the standard for
comparison comprises wild type recombinant E6-AP protein. In yet another
embodiment, AS or mutant cells, cell extract, or recombinant protein is provided as
a disease state standard.
In other embodiments of the present invention, the present invention provides
a method for assaying for the presence of tumors by comparing the ubiquitin-ligase
activity of a tissue or blood sample to a non-tumor control, where a decrease in the
ubiquitin-ligase activity indicates that the tumor is cancerous. One embodiment of
the invention is a method that is particularly useful in detecting breast tumors when
the tissue sample is obtained from a mammary gland biopsy. Still another
embodiment provides a method of monitoring the progression or regression of a
cancerous tumor comprising comparing the ubiquitin-ligase activity of tissue or blood
samples to a non-tumor control and previously tested samples, wherein a change in
the ubiquitin-ligase activity over time compared to the control and the previously
tested samples is indicative of progression or regression of said tumor. One further embodiment of the method can be used to monitor the progression or regression of
a breast tumor when the tissue sample is obtained from a mammary gland biopsy.
The following examples serve to illustrate specific embodiments of this
invention, but should not be considered as limiting the scope of the invention.
EXAMPLE 1 Plasmid Construction
The bait plasmid for the yeast two-hybrid system (pASl-PRLBD) (Onate et
al. (1995), Science 270: 1354-1357), mammalian expression plasmids for PR-B
(Allan et al. (1992), J. Biol. Chem. 267: 19513-19520), estrogen receptor (ER)
(Burris et al. (1995), Proc. Natl. Acad. Sci. USA. 922: 9525-9529), androgen receptor
(AR) (Tilley et al. (1989), Proc. Natl. Acad. Sci. USA. 86:327-331),E2F, baculovirus
expression plasmids pVLGST and pVLGST-PRA (Onate et al. (1995), Science 270:
1354-1357), reporter plasmids UAS4-TATA-LUC (LUC, luciferase) (Spencer et al.
(1997), Nature 389: 194-198), E2F, Spl, and CREB responsive reporters (Onate et
al. (1995), Science 270: 1354-1357) as evidenced in the foregoing cited references,
are all well known in the art. To construct GR expression vector, the pSTCGR vector
was digested with BamHI then the BamHI fragment containing the GR cDNA was
cloned into the corresponding sites of the pCR3.1 plasmid (Invitrogen). The
pPRE/GRE-Elb-LUC and pERE-Elb-LUC were constructed by inserting PvuII-
Smal fragments of pPRE/GRE-Elb-CAT and pERE-Elb-CAT into the Smal site of
pGL3 basic plasmid (Promega). To construct mammalian expression plasmids of
wild-type E6-AP (aal-851), 76 kDaE6-AP (aal 70-851), and C833-S mutant E6-AP (aal -851 ), the BamHI-Hindlll fragments of pGEM E6-AP (100 kDa), pGEM-E6-AP
(76 kDa), and pGEM E6-AP(C833-S) were cloned into the corresponding sites of the
pBKRS V plasmid (Stratagene). The C-terminal fragment of E6-AP (aa 680-851 ), the
truncated mutant E6-AP (aal -449) and the 98 kDa (aa 1-834) form of E6-AP, found
in AS (See e.g., Kishino et al. (1993), Nature Genet. 15: 70-73; Matsuura et al.
(\991),Nature Genet. 15: 74-77; Sutcliff et al. (1997), GenomeRes. 1: 368-377) and
the 99kDa (aa 1-845) form of E6-AP, the 86 kDa (aa 1-714) form of E6-AP, the 47
kDa (aa450-851) form of E6-AP, and the 28 kDa (aa 1-240) were amplified by
polymerase chain reaction (PCR) with the primers (SEQ. ID. NO. 1.) 5'-
GCGGATCCACCATGAGGAATTCGGCACGAGATCTAAAGGA A-3' (upper
strand) and (SEQ. ID. NO. 2) 5'-CGGAATTCAAGCTTGTTTTACAGCATGCC
AAATCC-3' (lower strand); and (SEQ.ID.NO.3) 5'-GCGGATCCAC
CATGGAAGCCTGCACGAATGAGTTTTGTG CT-3' (upper strand) and (SEQ. ID.
NO.4) 5'- CCCAAGCTTGTTTTATGTTTCTACTTTGAAAAAAGTATA-3' (lower
strand); (SEQ. ID. NO. 5) 5'-GCGGATCCACCATGAGGAATTCGGCACGAG
ATCTAAAGGA A-3' (upper strand) and (SEQ. ID. NO. 6) 5'-CCCAAGCTTG
TTTTAAAGTTTTTCTTTGCTTGAGTATTC-3' (lower strand); (SEQ. ID. NO. 7)
5'-GCGGATCCACCATGAGGAATTCGGCACGAGATCTAAAGGAA-3'(upper
strand) and (SEQ.ID.NO.8) 5'-CCCAAGCTTGTTTTAGGCATACGTGATGGC
CTTCAACAA-3' (lower strand); (SEQ. ID. NO. 9) 5'-GCGGATCCAC
CATGAGGAATTCGGCACGAGATCTAAAGGAA-3' (upper strand) and (SEQ. ID.
NO. 10) 5'-CCCAAGCTTGTTTTACATATGAAAACCTCTCCGAAAAGC (lower strand); (SEQ. ID. NO. 11) 5'-GCGGATCCACCATGTACAGTGAACGAAGAA
TCACTGTT-3' (upper strand) and (SEQ.ID.NO.12) 5'-CGGAATTCGC
GGCCGCGTTTTACAGCATGCCAAATCC-3' (lower strand); and (SEQ. ID. NO.
13) 5'-GCGGATCCACCATGGAAGCCTGCACGAATGAGTTTTGTGCT-3' (upper
strand) and (SEQ. ID. NO. 14) 5'-GAATTCAAGCTTGTTTTACAAATATA
CAAGTGCATTGAG-3' (lower strand). The PCR product was digested with BamHI-
Hindlll and cloned into the corresponding sites of the pBKRSV plasmid. Then the
BamHI-Notl fragments of pBKRSV-E6-AP plasmids were subcloned into the
corresponding sites of pCR3.1 plasmid (Invitrogen). The I804K and F782Δ mutant
forms of E6-AP were constructed by utilizing site directed mutagenesis to create
these mutations in pCR3.1 E6-AP. To reconstitute the 104kDa 1-885 Δstop mutation
into the E6-AP, the BsaAI-Hindlll fragment of E6-AP was amplified by PCR with
the primers (SEQ. ID. NO. 15) 5'-GTTGAAGGCCATCACGTATGCCAAAGG-3'
(lower strand) and (SEQ. ID. NO. 16) 5'-GAATTCAAGCTTGTTTTAGTACTG
GGACACTATCACCACCA-3' (lower strand) using AS patient DNA as a template.
Then this BsaAI-Hindlll fragment was cloned into the corresponding sites of pGEM
E6-AP. To reconstitute this mutation into the mammalian expression plasmid, the
BamHI-Hindll fragment of E6-AP was cloned into the corresponding sites of
pBK.RSV plasmid. The BamHI-Notl fragment of pBK.RSVE6-AP was subcloned
into the BamHI-Notl sites of pCR3.1 plasmid. To reconstitute the full-length E6-AP
gene into a yeast two-hybrid plasmid, the Hindlll-digested (and filled) pGEM E6-AP
(100 kDa) was redigested with BamHI. The resulting BamHI-Hindlll (filled) fragment was inserted into the BamHI-EcoRI (filled) sites of pGADIO (Clonetech).
To reconstitute the PR-A gene into the yeast pASl plasmid, the Ncol-Sall fragment"
of PRA was ligated into the corresponding sites of the vector. To fuse E6-AP with
the VP 16 activation domain and GAL DNA-binding domain (DBD) (residues 1 - 147),
the BamHI-Hindlll fragment of full-length E6-AP and several deletion fragments of
E6-AP were subcloned in frame into the pABVPlό and pABGAL plasmids utilizing
mehtods well known in the art, such as those contained in Baniahmad et al. (1993),
Proc. Natl. Acad. Sci. USA. 90: 8832-8836 and Baniahmad et al. (1995), Mol. Cell.
Biol. 15, 76-86). To fuse E6-AP with the glutathione-S-transferase (GST), the
BamHI-Notl fragments of full-length E6-AP and various mutant forms of E6-AP
were subcloned in frame with GST into the pGEX4T plasmid (Pharmacia).
EXAMPLE 2 In Vivo Interaction Assays
The interaction of PR with wild-type E6-AP was evaluated in a yeast two-
hybrid assay. The entire coding sequence of PR-A was fused in frame with the yeast
GAL4 DNA-binding domain (GAL-DBD-hPRA). Then the GAL-DBD-PRA
construct was coexpressed with either control vector or the GAL-AD-E6-AP
construct (GAL activation domain fused in frame with wild-type E6-AP) along with
a reporter plasmid in yeast strain B J2186. The transformants were propagated, and
the β-galactosidase activities from three independent colonies were determined. The
yeast cells were treated with either vehicle alone (-H), 10-6 M progesterone (+P), or 10-6 M RU486 (+RU). The β-galactosidase activity is expressed in Miller units and
each bar depicts the average of three assays, i.e. n=3.
EXAMPLE 3 In Vitro Interaction Assay
For the in vitro interaction assay, the PR-B was expressed as a his-tagged
fusion protein in a baculovirus expression system in the presence or absence of
progesterone, and purified using a nickel affinity column (Pharmacia). GST-tagged
E6-AP was expressed in Escherichia coli cells and purified on glutathione-sepharose
beads. The purified and glutathione-bound E6-AP was incubated with the purified
PR-B in NETN buffer (50 mM Nacl, ImM EDTA, 20 mM Tris, pH 8.0, and 0.5%
NP-40) for 1 hour at room temperature. Subsequently, the beads were incubated with
8 μl of in vitro transcribed and translated [35S]methionine labeled E6-AP and
interactions were allowed to occur at 4 °C overnight. Beads were then washed five
times with NETN buffer. E6-AP bound PR was analyzed by SDS-PAGE on a 7.5%
gel followed by Western blot analysis using antibodies which specifically recognize
PR.
EXAMPLE 4 Transfections
HeLa cells were maintained in DMEM supplemented with 10% fetal bovine
serum. Twenty- four hours before transfection, 3X105 cells were plated per well in
Falcon six-well dishes in DMEM containing 5% dextran-coated charcoal-stripped serum. Cells were transfected with the indicated DNAs using Superfect reagent
(Qiagen) or Lipofectin (Gibco BRL) according to the manufacturer's guidelines."
Cells were washed, and fed with DMEM containing 5% stripped serum and
subsequently treated with the indicated hormones. Cells were harvested twenty-four
hours thereafter. Cell extracts were assayed for luciferase activity using the
Luciferase Assay System (Promega), and values are corrected for either protein
concentration or β-Gal activity. Data is analyzed as the mean of triplicate values
obtained from representative experiments.
EXAMPLE 5
Coactivation Ubiquitin-Protein Ligase Activity
To study the coactivation function of wild- type and various mutant forms of
E6-AP, HeLa cells were transfected with 0.1 μg of PR-B expression plasmid, 1 μg of
pPRE/GRE.Elb.LUC and 0.25 μg of wild-type E6-AP expression plasmid (aa 1-851 )
or 0.25 μg of various mutant forms of E6-AP expression plasmids (aa 450-851, aa 1-
845, aa 1-834, aa 1-714, R417X, C833S, I804K, F782Δ, and Δstop aa 1-885). The
cells were incubated with 10"7 M progesterone. The coactivation by wild-type E6-AP
is represented by (++++) signs. The coactivation by various mutant forms of E6-AP
is presented as relative activity compared to that of wild-type E6-AP.
To study the ubiquitin-protein ligase activity of wild-type E6-AP and various
mutant forms of E6-AP, the wild-type E6-AP and various mutant forms of E6-AP
(shown in Table 1) were expressed and purified from E.coli as GST-fusion proteins.
(+) represents positive for ubiquitin-protein ligase activity and (-) represents negative for ubiquitin-protein ligase activity. (*) represents natural mutants of E6-AP cloned
from AS patients. The E6 independent substrate of E6-AP, HHR23 A, was expressed
in vitro in TNT-coupled wheat germ extracts in the presence of [35S]methionine as
per manufacturer's instructions (Promega). Ubiquitin was obtained from commercial
sources (Sigma). The ubiquitin-protein ligase activity of these proteins was measured
by incubating the labeled HHR23 A target protein (5 μl aliquot), ubiquitin (4 μg), and
cell extract at 30 °C in a reaction buffer comprising 20 mM Tris-HCl, pH 7.6, 50 mM
NaCl, 4mM ATP, 10 mM MgCl2, and 0.2 mM dithiothreitol. Reactions were
terminated at 90 to 120 minutes by the addition of SDS-sample buffer. Samples were
then boiled for 5 minutes prior to size fractionation by SDS-PAGE and visualization
by autoradiography. Other suitable methods are known in the art, such as Kumar et
al. (1997), J. Biol. Chem. 272: 13548-13554.
EXAMPLE 6 Isolation and Characterization of E6-AP as a PR Interacting Protein
To identify novel proteins which selectively modulate the transactivation
functions of members of the nuclear receptor superfamily, a HeLa cDNA library was
screened using the ligand binding domain (LBD) of PR as a bait in a yeast two-hybrid
screening assay. Thirteen colonies were isolated which strongly interacted with the
LBD of PR. These colonies contained cDNAs with identical sequences. A sequence
similarity search in the Gene Bank data base revealed that all colonies encoded the
carboxy-terminal amino acids, aa 680-851 region of the E6-AP protein. EXAMPLE 7 Regions of E6-AP required for interaction with PR
Since E6-AP interacts with PR in a hormone-dependent manner, the regions
of E6-AP important for interaction with PR were defined. Figure 2 A is a schematic
representation of E6-AP showing the position of the catalytic region (solid box), hect
domain, transactivation domain, and PR-binding domains. Wild-type E6-AP is a 100
KDa (aa 1-851) protein. The 76 KDa (aa 170-851) and the 47 KDa (aa 450-851)
represent E6-AP which contain deletions at the N-terminus. The 21 kDa form (aa
680-851 ) represents the carboxy-terminus of the E6-AP protein identified in the yeast
two-hybrid screen. The 99 kDa (aa 1-845), the 98 kDa (aa 1-834), the 86 kDa (aa 1-
714), the 53 kDa (aa 1-449), and the 28 kDa (aa 1-240) represent E6-AP C-terminus
deletion mutants. The C833S mutant represents a Cysteine 833 to Serine 833 mutant
form of E6-AP. The AS disease mutants are represented by the 98 kDa (deletion of
17 aa from the C-terminus), the 53 kDa C-terminally truncated E6-AP protein (aa 1 -
449), the I804K form of E6-AP contains lysine at position 804 instead of isoleucine,
the F782Δ contains a deletion of phenylalanine at position 782, and the 104 kDa (1-
885 Δstop) form of E6-AP is a read through mutant.
In order to locate the PR interaction site in E6-AP, an in vivo mammalian two-
hybrid interaction assay system was utilized. See e.g., Spencer et al. (1997), Nature
389: 194-198). In this assay, full-length E6-AP and various deletion fragments of
E6-AP were fused to the VP 16 activation domain and the ability of the VP 16-E6-AP
hybrid proteins to interact with receptor was determined in the absence or presence
of progesterone. As shown in Fig. 2B, wild-type E6-AP, N-terminal deletion fragments (aa 170-851 and aa 680-851 ) and C-terminal deletion fragments (aa 1-714
and aa 1 -449) of E6-AP were able to interact with PR while the control vector lacking
E6-AP cDNA and the N-terminal fragment (aa 1-240) of E6-AP did not interact,
indicating that at least two PR interaction sites are located within the E6-AP protein.
One site is located within the C-terminal (aa 680-851) (21 kDa) fragment of E6-AP,
the fragment of E6-AP which was originally isolated in the yeast two-hybrid screen.
The second PR interaction site is located within aa 240-449 and overlaps with the E6
binding site.
EXAMPLE 8
E6-AP as a Coactivator for Nuclear Hormone Receptor Superfamily
To investigate the possible role of E6-AP in receptor-dependent activation of
target gene expression, co-transfection assays in HeLa cells were performed. HeLa
cells were transiently transfected with 0.2 μg of pPR-B expression plasmid and 1 μg
of pPRE/GRE.Elb.LUC (a reporter plasmid containing a progesterone response
element) in the absence or presence of E6-AP expression plasmid, pBK.RSV-E6-AP
(0.250 μg). The cells were treated with either vehicle only (-H), 10"7 M progesterone
(+P), or 10"7 M RU486 (+RU). The data are expressed as fold activation and each bar
depicts the average of at least three wells. In the absence of ligand, PR has a minimal
effect on reporter gene expression either in the absence or in the presence of E6-AP
(Fig 3 A). Addition of the hormone yielded an eight- fold increase in PR activity in the
absence of E6-AP; when E6-AP was coexpressed with PR, the activity of PR was
further stimulated by ~5-fold, a total of 40-fold over basal. In contrast, coexpression of E6-AP with PR had no significant effect on the transcription of the reporter gene
when receptor was bound to antihormone-RU486 (Fig. 3A). These data are
consistent with data which indicate that the antihormone RU486 induces a distinct
conformational change in the receptor molecule that has reduced affinity for
coactivators (See e.g., Allan et al. (1992), J. Biol. Chem. 267: 19513-19520; Onate
et al. (1995), Science 270: 1354-1357; Vegeto et al. (1992), Cell 69: 703-713; Xu
et al. (1996), Proc. Natl. Acad. Sci. USA. 93: 12195-12199). Since, HeLa cells are
derived from a papillomavirus type 18 positive cervical carcinoma patient and thus
express the E6 protein and because E6-AP was originally cloned as an E6 interacting
protein, it was necessary to rule out the possibility that the E6 protein influences the
coactivation function of E6-AP. E6-AP was able to stimulate the hormone-dependent
transcriptional activity of steroid hormone receptors in the E6-negative HepG2 and
SK-N-SH cell lines (data not shown), suggesting that the coactivation observed in
HeLa cells is not dependent on the E6 protein.
The data show that E6-AP stimulates the hormone-dependent transcriptional
activity of PR by acting as a coactivator. In order to determine if E6-AP functions
as a coactivator for of the nuclear receptor superfamily, the effect of E6-AP
expression on the ligand-dependent transcriptional activity of different nuclear
hormone receptors and on several other transcription factors was examined (Figures
3B and C). E6-AP significantly enhanced the hormone-dependent transcriptional
activity of PR, estrogen receptor (ER), androgen receptor (AR), and glucocorticoid
receptor (GR). It also enhanced the transcriptional activity of retinoic acid (RARa), and thyroid hormone receptors (TR) (data not shown). E6-AP had a minimal or no
effect on the transcriptional activity of E2F and cyclic AMP response element
binding protein (CREB). Coexpression of E6-AP had only a moderate effect on the
activation function of Spl (Figure 3C). These data show that E6-AP preferentially
coactivates the hormone-dependent transcriptional activity of nuclear hormone
receptors, but is not uniquely specific for them as is the case for other coactivators
such as SRC-1. Onate et al. (1995), Science 270: 1354-1357.
EXAMPLE 9
E6-AP Relieves Squelching Between ER and PR
It has been shown previously that ER and PR share common coactivators
since hormone bound-ER can sequester limited pools of coactivators from PR, a
phenomenon known as squelching. See Conneely et al. (1989),. In A. K. Roy and J.
Clark (ed.), Gene regulation by steroid hormones IV, p. 220-223, Spring-Verlag
Press, New York., Meyer et al. (1989), Cell 57: 433-442; Shemshedini et al. (1992),
J. Biol. Chem. 261 : 1834-1839. It was determined whether coexpression of E6-AP
was able to reverse this squelching phenomenon. HeLa cells were transfected with
0.2 μg of PR expression plasmid, 0.3 μg of ER expression plasmid together with 1.0
μg of pPRE/GRE.Elb.LUC and increasing concentrations of wild-type E6-AP (0,
0.1, 0.5. and 1.0 μg). Cells were then treated with progesterone (Prog) or
progesterone and estradiol (E2) together (each at 10~8 M). The data are expressed as
fold activation and each bar depicts the average of at least three wells. The hormone- induced transcriptional activity mediated by PR was reduced by 91% upon
coexpression of estradiol-bound ER (Figure 4A, compare lanes 2 and 3). Addition
of E6-AP reversed this squelching by as much as 9.6-fold (Figure 4A, compare lanes
3 and 6) in a dose-dependent manner. At the highest concentration of E6-AP used
in this reverse squelching experiment, the PR activity was enhanced by only 2.6 fold
(compare lanes 2 and 7). These data demonstrate that E6-AP is a limiting factor
which is necessary for efficient PR and ER transactivation. The fold coactivation by
E6-AP is lower in this experiment compared to that shown in Figure 3B, due to
different experimental conditions used in these experiments. In Figure 4 (lane 7), the
coactivation function of E6- AP on the transcriptional activity of PR was observed in
the presence of ER expression plasmid whereas in Figure 3B, only a single receptor
was transfected. As expected, no significant reverse squelching was observed (Figure.
4B, compare lanes 3 and 6) with the C-terminal fragment of E6-AP (aa 680-851)
(Figure 2A) which weakly interacts with ER and has no activation function (Figure
5). Western blot analysis confirmed that the C-terminal fragment of E6-AP (aa 680-
851) and full-length E6-AP are equally expressed.
EXAMPLE 10 E6-AP Contains an Intrinsic Activation Domain
To ascertain whether E6-AP possesses an intrinsic, transferable activation
domain, wild-type and deletion fragments of E6-AP were recruited to DNA by
linking them to the DNA-binding domain (DBD) of GAL4 in a fusion protein. The
full-length E6-AP (aa 1-851), N-terminal deletion fragments of E6-AP (aa 170-851 and aa 680-851) and C-terminal deletion fragment of E6-AP (aa 1-714) (shown in
Figure 1 A) were fused to the yeast GAL4 DBD. HeLa cells were then transfected
with 0.5 μg of UAS4-TATA-luciferase reporter DNA and GAL4DBD or GAL4-E6-
AP expression plasmids (1.0 μg). The data are expressed as fold activation and each
bar depicts the average of at least three wells. The wild-type E6-AP (aa 1-851), N-
terminal deletion fragment (aa 170-851, 76 kDa), and C-terminal deletion fragment
(aa 1-714, 86 kDa) stimulated the transcriptional activity of the reporter gene
compared to that of the control vector containing only GAL4DBD (Figure 5) while
the 21 kDa fragment (aa 680-851) did not. This suggests that E6-AP itself contains
a transcriptional activation domain located between aa 170-680.
EXAMPLE 11 Expression Pattern of E6-AP in Mouse Mammary Glands
Changes in the expression levels of coactivator proteins such as E6-AP can
contribute to the etiology of steroidal cancers. The altered expression pattern of
E6-AP in mouse mammary gland model of multistage tumorigenesis was
examined using Western blot analysis techniques. Cell extracts were prepared
from normal mammary glands and mammary gland tumors by homogenization in
a buffer containing 50 mM NaCl, 20 mM HEPES pH 7.5, 5 mM KC1, 10%
Glycerol, 1 mM PMSF, and protease inhibitors Leupeptin (1.25 g/ml) and
Pepstatin A (1.0 g/ml). Protein concentrations were estimated using the Bradford
reagent. 15 ug of total protein was denatured, separated on SDS-polyacrylamide and analyzed with rabbit polyclonal antibody raised against E6-AP. Purified
human E6-AP protein was used as a control. As seen in Figure 7, there is
approximately a 4.5 -fold increase in E6-AP expression in mammary tumors
compared to that of normal tissue from mammary glands. Furthermore, increased
expression of E6-AP was associated with tumor progression because E6-AP
overexpression was not observed in the earlier stages of mammary tumorigenesis.
EXAMPLE 12 E6-AP Contains Two Independent Separable Functions, Coactivation and Ubiquitin-Ligase Activity
Since E6-AP is a ubiquitin-protein ligase, it was determined if the
coactivation function of E6-AP is dependent on this enzymatic function. It has
been shown that a conserved cysteine (C) residue in E6-AP at position 833 (C833)
forms a thioester bond with ubiquitin and is necessary for the transfer of ubiquitin
to the protein targeted for ubiquitination. The mutation of C833 to alanine (A) or
serine (S) has been shown to eliminate the ubiquitin-protein ligase activity of E6-
AP. Huibregtse et al. (1995), Proc. Natl. Acad. Sci. USA. 92: 2563-2567. In
cotransfection experiments, an E6-AP bearing a C to S mutation at this critical site
was still able to coactivate PR (Table 1) and ER to nearly the same extent as that
of wild-type E6-AP. Furthermore, the C833S mutant of E6-AP also can reverse
squelch the hormone-dependent transcriptional activity of PR to a similar extent
as that of wild-type E6-AP. The data suggest that the ubiquitin-protein ligase
activity of E6-AP is not required for the coactivation function of E6-AP. To further confirm that the ubiquitin-proteasome pathway is not involved in the
coactivation function of E6-AP, a deletion mutant of E6-AP (aa 1-845) was
analyzed which lacks six amino acids at the carboxy terminus and has been shown
to be defective for ubiquitin-protein ligase activity Huibregtse et al. (1995), Proc.
Natl. Acad. Sci. USA. 92: 2563-2567. Like the C833S mutant, this mutant also
retains the ability to coactivate the hormone-dependent transcriptional activity of
PR (Table 1), further confirming that the ubiquitin-protein ligase activity of E6-
AP is not necessary for E6-AP to function as a coactivator. The data indicate that
E6-AP possesses two independent, separable functions; coactivation and
ubiquitin-protein ligase activity.
EXAMPLE 13
The AS Phenotype Results From Defects in the Ubiquitin-protein Ligase Activity of E6-AP
Recently, it was shown that a subset of AS patients express mutant forms
of the E6-AP, rather than possessing large scale deletions of the 15ql l-ql3 region
which contains E6-AP, that are more common. Kishino et al. (1993), Nature
Genet. 15: 70-73., Matsuura et al. (1997), Nature Genet. 15: 74-77.; Sutcliff et al.
(1997), Genome Res. 1: 368-377. In order to determine if the coactivator function
of E6-AP was necessary for development of the AS phenotype, several mutant E6-
AP proteins corresponding to those found in these AS patients were generated
(shown in Table 1). First, the effect of an E6-AP mutant was tested which had a
gross deletion whereby the C-terminal half of the protein was deleted due to a non-sense mutation at codon 417 (R417X). The ability of this AS mutant protein
to coactivate PR was greatly diminished in comparison to the wild-type E6-AP,
but was still able to interact with PR (Figure 2B) indicating that a loss of
coactivation was due to disruption of the activation domain located at aa 170-680.
Furthermore, the loss of coactivation by the R417X mutant was not due to the loss
of expression of mutant protein, since this mutant was able to interact with PR to
the same extent as that of wild-type E6-AP in mammalian cells which were used
to assess coactivation (Figure 2B). The R417X mutant of E6-AP was also unable
to coactivate ER and AR.
Another mutant form of E6-AP was then tested which contained a small
deletion in the hect domain due to a frameshift mutation which results in the
truncation of the last 17 aa of the protein (1-834) and the replacement of four
different amino acids from the new reading frame. This mutant E6-AP was able
to coactivate PR to the same extent as the wild-type E6-AP (Table 1). Similarly,
an artificial mutant which lacks six aa at the extreme C-terminus of E6-AP (aa 1-
845), was also able to act as a coactivator of PR activity (Table 1). Three other
mutants which were evaluated for their ability to coactivate PR transcription
consisted of a missense mutation I804K in which isoleucine 804 was mutated to
lysine, F782Δ, an internal in-frame deletion of phenylalanine 782, and 1-885
Δstop, a read through mutation which results in a longer mutant form of E6-AP.
All three of these mutant forms of E6-AP were able to coactivate PR activity,
demonstrating that the coactivator function of E6-AP is not involved in the central nervous system (CNS) phenotype of AS (Table 1).
TABLE 1
Amino Acid Position Coactivation Ubiquitin activitv
1-851 ++++ +
450-851 + -
680-851 - -
1-845 ++++ -
1-834* ++++ -
1-714 ++ -
R417X* + -
1-240 not tested -
C833S ++++ -
I804K* ++++ +
F782Δ* ++++ -
1-885* Δ Stop ++++ not tested
AS mutant forms of E6-AP
To correlate the ubiquitin-protein ligase activity of E6-AP with AS, the
ubiquitin-ligase function of wild-type and AS mutant forms of E6-AP was tested.
Some AS mutant forms of E6-AP, such as aa 1-834, R417X, and F782Δ were
unable to ubiquitinate a protein (HHR23A) implemented as a target of E6-AP
ubiquitin-protein ligase activity in an in vitro ubiquitin assay (See also Kumar et
al. (1997), J. Biol. Chem. 272: 13548-13554; Van der Spek et al. (1994), Genomics 23: 651-658). The results show that loss of ubiquitin-protein ligase
activity contributes to the AS phenotype in these patients. However, the AS
missense mutant (I804K), was able to ubiquitinate the target protein HHR23 A, to
an extent comparable to that of wild-type E6-AP (Table 1).
REFERENCES
All patents and publications mentioned in the specification are indicative
of the level of those skilled in the art to which the invention pertains. All patents
and publications are herein incorporated by reference to the same extent as if each
individual publication was specifically and individually indicated to be
incorporated by reference.
Allan et al. (1992), J. Biol. Chem. 267: 19513-19520.
Anzick et al. (1997), Science 277: 965-968.
Baniahmad et al. (1993), Proc. Natl. Acad. Sci. USA. 90: 8832-8836.
Baniahmad et al. (1995), Mol. Cell. Biol. 15, 76-86.
Beuten et al. (1996), Human Genet. 97: 294-298.
Bocquel et al. (1989), Nuc. Acids Res. 17: 2581-2594.
Burris et al. (1995), Proc. Natl. Acad. Sci. USA. 922: 9525-9529.
Chen et al. (1997), Cell 90: 569-580.
Conneely et al. (1989),. In A. K. Roy and J. Clark (ed.), Gene regulation by
steroid hormones IV, p. 220-223, Spring- Verlag Press, New York.
Daniel et al. (1998), J. Gen. Virol. 79: 489-499.
Durfee et al. (1993), Gene Dev. 7: 555-569.
Hatakeyama et al. (1997), J. Biol. Chem. 272: 15085-15092.
Hong et al. (1996), Proc. Natl. Acad. Sci. USA. 93: 4948-4952.
Horwitz et al. (1996), Mol Endocrinol. 10: 1167-1177. Huibregtse et al. (1995), Proc. Natl. Acad. Sci. USA. 92: 2563-2567.
Huibregtse et al. (1991), EMBO J. 10: 4129-4135.
Huibregtse et al. (1993) Mol. Cell. Biol. 13: 775-784.
Huibregtse et al. (1993), Mol. Cell. Bio. 13: 4918-4927.
Kishino et al. (1993), Nature Genet. 15: 70-73.
Kumar et al. (1997), J. Biol. Chem. 272: 13548-13554.
Li et al. (1997), Proc. Natl. Acad. USA. 94: 8479-8484.
Matsuura et al. (1997), Nature Genet. 15: 74-77.
Meyer et al. (1989), Cell 57: 433-442.
Ogryzko et al. (1996), Cell 87: 953-959.
Onate et al. (1995), Science 270: 1354-1357.
Perlmann et al. (1997), Cell 90: 391-397.
Pugh et al. (1992), J. Biol. Chem. 267: 679-682.
Scheffher et al. (1993), Cell 75: 495-505.
Shemshedini et al. (1992), J. Biol. Chem. 261: 1834-1839.
Shibata et al. (1997), Recent. Prog. Hormone Res. 52: 141-165.
Spencer et al. (1997), Nature 389: 194-198.
Sutcliff et al. (1997), Genome Res. 1: 368-377.
Tilley et al. (1989), Proc. Natl. Acad. Sci. USA. 86:327-331.
Tjian et al. (1994), Cell 11: 5-8.
Torchia et al. (1997), Nature 387: 677-684.
Tsai et al. (1994), Ann. Rev. Biochem. 63: 451-486. Tukeshita et al. (1997), J. Biol. Chem. 272: 27629-27634.
Nan der Spek et al. (1994), Genomics 23: 651-658.
Vegeto et al. (1992), Cell 69: 703-713.
Voegel et al. (1996), EMBOJ. 15: 3667-3675.
Xu et al. (1996), Proc. Natl. Acad. Sci. USA. 93: 12195-12199.
Xu et al. (1998), Science 279:1922-1924.
Yeh et al. (1996), Proc. Natl. Acad. Sci. USA. 93: 5517-5521.
One skilled in the art readily appreciates that the present invention is well
adapted to carry out the objectives and obtain the ends and advantages mentioned
as well as those inherent therein. Systems, treatments, methods, procedures and
techniques described herein are presently representative of the preferred
embodiments and are intended to be exemplary and are not intended as limitations
of the scope. Changes therein and other uses will occur to those skilled in the art
which are encompassed within the spirit of the invention or defined by the scope
of the pending claims.

Claims

What is claimed is:
1. A method of treating endocrine tumors comprising interfering with a
hormone coactivator pathway.
2. The method of claim 1 , wherein said interfering comprises reducing the
concentration of a hormone coactivator.
3. The method of claim 2, wherein said concentration is reduced by
administrating oligonucleotides complementary to the coactivator mRNA.
4. The method of claim 2, wherein said concentration is reduced by the
administration of a chemical agent which inhibits transcription of a coactivator
mRNA.
5. The method of claim 2, wherein said concentration is reduced by the
administration of a chemical agent which inhibits translation of a coactivator
mRNA.
6. The method of claim 2, wherein said concentration is reduced by the
administration of a chemical agent which promotes degradation of a coactivator
mRNA, or d) promotes degradation of a coactivator protein.
7. The method of claim 2, wherein said concentration is reduced by the
administration of a chemical agent which promotes degradation of a coactivator
protein.
8. The method of claim 1 , wherein said interfering comprises neutralizing a
coactivator.
9. The method of claim 8, wherein said neutralizing comprises binding a
coactivator to a substrate which lacks the activity of the natural endogenous
receptor.
10. The method of claim 1 , wherein said interfering comprises blocking the
coactivator binding site on the target nuclear receptor with a transcriptionally
incompetent substrate.
11. The method of claim 1 , wherein said interfering comprises any
combination of one or more of the methods selected from the group consisting of
reducing the concentration of a hormone coactivator;
administrating oligonucleotides complementary to the coactivator
mRNA;
administrating a chemical agent which inhibits transcription of a
coactivator mRNA; administrating a chemical agent which inhibits translation of a
coactivator mRNA;
administrating a chemical agent which promotes degradation of a
coactivator mRNA,
administrating a chemical agent which promotes degradation of a
coactivator protein;
neutralizing a coactivator;
binding a coactivator to a substrate which lacks the activity of the
natural endogenous receptor; and
blocking the coactivator binding site on the target nuclear receptor
with a transcriptionally incompetent substrate.
12. The method of claim 1, wherein said endocrine tumors are selected from a
group consisting of mammary tumors, endometrial tumors, and prostate tumors.
13. The method of claim 1, wherein said hormone coactivator is E6-AP.
14. A vector containing an agent which interferes with a hormone coactivator
pathway.
15. The vector of claim 14, wherein said vector comprises a viral vector
coding for said agent.
16. The vector of claim 14, wherein said agent binds to a nuclear hormone
coactivator.
17. The vector of claim 14, wherein said nuclear hormone coactivator is
selected from a group consisting of E6- AP, SRC- 1 , TIF2, p/CIP, and ARA70.
18. The vector of claim 14, wherein said agent binds at or about the site for
coactivator binding on a nuclear hormone receptor.
19. The vector of claim 18, wherein said nuclear hormone receptor is selected
from a group consisting of PR, ER, TR, RARα, AR, and GR.
20. The vector of claim 14, wherein said agent is comprises a nucleotide
sequence complementary to a hormone coactivator mRNA.
21. The vector of claim 14, wherein said hormone coactivator pathway is that
ofE6-AP.
22. A method for screening a tissue sample for endocrine tumors comprising
assaying said tissue for nuclear hormone coactivator mRNA or protein wherein an
increased level indicates tumor presence.
23. The method of claim 22, wherein said endocrine tumors are selected from
a group consisting of mammary tumors, endometrial tumors, and prostate tumors. "
24. The method of claim 22, wherein said nuclear hormone coactivator is from
a group consisting of E6-AP, SRC-1, TIF2, p/CIP, and ARA70.
25. The method of claim 22, wherein said tissue is assayed for E6-AP and p53
concentrations.
26. The method of claim 22, wherein said assay comprises the use of
antibodies directed against specific nuclear coactivators.
27. The method of claim 22, wherein said assay comprises an antibody
directed against E6-AP.
28. The method of claim 22, wherein said assay comprises antibodies directed
against E6-AP and p53.
29. The method of claim 22, wherein said assay comprises the measurement of
specific nuclear hormone coactivator mRNA by RT-PCR.
30. The method of claim 22, wherein said assay comprises the measurement of specific nuclear hormone coactivator mRNA by Northern hybridization.
31. A method for screening a compound for the potential to regulate endocrine
tumor growth comprising contacting a tissue with said compound and
subsequently assaying that tissue for nuclear hormone coactivator mRNA or
protein wherein a lowered nuclear hormone coactivator level indicates the ability
of said compound to inhibit endocrine tumor growth.
32. The method of claim 31 , wherein said tissue consists of cells selected from
a group consisting of MCF-7, T47D, MDA-MB-231, LnCaP, PC3, and RUCA-1
cells.
33. The method of claim 31, wherein said endocrine tumor is selected from a
group consisting of mammary, endometrial, and prostate tumors.
34. The method of claim 31 , wherein said nuclear hormone coactivator is
selected from a group consisting of E6-AP, SRC-1, TIF2, p/CIP, and ARA70.
35. The method of claim 31 , wherein said nuclear hormone coactivator is E6-
AP.
36. The method of claim 31 , wherein p53 and the coactivator E6-AP are assayed.
37. A method for the evaluation of endocrine tumorogenicity of environmental
agents comprising contacting a tissue with said compound and subsequently
assaying that tissue for nuclear hormone coactivator mRNA or protein wherein an
increased level of nuclear hormone coactivator level indicates tumorogenicity of
an agent.
38. The method of claim 37, wherein said tissue comprises cells from a group
consisting of fibroblasts, epithelial cells, COS cells, HeLa cells, blood cells, and
mammary-derived cells which do not express high levels of E6-AP.
39. The method of claim 37, wherein said tissue comprises freshly isolated
cells from a group consisting of mammary, endometrial, and prostate tissues.
40. The method of claim 37, wherein said nuclear hormone coactivator is
selected from a group consisting of E6-AP, SRC-1, TIF2, p/CIP, and ARA70.
41. The method of claim 37, wherein p53 and the coactivator E6-AP are
assayed.
42. A method for the detection of Angelman syndrome comprising the steps of:
a) obtaining a cell extract from a patient tissue sample,
b) incubating said cell extract with a reaction solution comprising ubiquitin
and a ubiquitin-binding target protein,
c) size fractionating the ubiquitin/target protein products, and
d) comparing the ubiquitin/target protein product size to that produced by a
standard,
wherein a decreased ubiquitin/target protein product size compared with that of a
wild type standard indicates Angelman syndrome.
43. The method of claim 42, wherein said tissue sample is from a group
consisting of blood cells, amniocentesis-derived cells, epithelial cells, and
fibroblasts.
44. The method of claim 42, wherein said ubiquitin-ligase activity is E6-AP
dependent.
45. The method of claim 42, wherein said ubiquitin-ligase activity is measured
from recombinant E6-AP protein generated from said tissue sample.
46. The method of claim 42, wherein said target protein is 35S-Met labeled
HHR23A.
47. The method of claim 42, wherein said standard a cell extract from E6-AP
+/+ cells.
48. The method of claim 42, wherein said standard is a recombinant E6-AP
protein.
49. The method of claim 42, wherein said standard is a cell extract from E6-
AP +/- cells.
50. The method of claim 42, wherein said standard is a recombinant AS
mutant E6-AP protein.
51. The method of claim 42, wherein said target protein is made amenable to
size fractionation and subsequent detection via one of several methods including
incorporation of a radioisotope, fluorescent tag, or antibody recognized epitope.
52. A method of detecting a cancerous tumor comprising comparing the
ubiquitin-ligase activity of a tissue or blood sample to a non-tumor control,
wherein a decrease in said ubiquitin-ligase activity indicates that said tumor is
cancerous.
53. The method of Claim 52, wherein said tissue sample is obtained from a mammary gland biopsy.
54. A method of monitoring the progression or regression of a cancerous
tumor comprising comparing the ubiquitin-ligase activity of tissue or blood
samples to a non-tumor control and previously tested samples, wherein a change
in the ubiquitin-ligase activity over time compared to said control and said
previously tested samples is indicative of progression or regression of said tumor.
55. The method of Claim 54, wherein said tissue sample is obtained from a
mammary gland biopsy.
PCT/US2000/000288 1999-01-07 2000-01-06 Novel coactivator overexpressed in endocrine tumors WO2000040756A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU27207/00A AU2720700A (en) 1999-01-07 2000-01-06 Novel coactivator overexpressed in endocrine tumors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US11502599P 1999-01-07 1999-01-07
US60/115,025 1999-01-07

Publications (1)

Publication Number Publication Date
WO2000040756A1 true WO2000040756A1 (en) 2000-07-13

Family

ID=22358886

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/000288 WO2000040756A1 (en) 1999-01-07 2000-01-06 Novel coactivator overexpressed in endocrine tumors

Country Status (2)

Country Link
AU (1) AU2720700A (en)
WO (1) WO2000040756A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5736330A (en) * 1995-10-11 1998-04-07 Luminex Corporation Method and compositions for flow cytometric determination of DNA sequences
US5888740A (en) * 1997-09-19 1999-03-30 Genaco Biomedical Products, Inc. Detection of aneuploidy and gene deletion by PCR-based gene- dose co-amplification of chromosome specific sequences with synthetic sequences with synthetic internal controls
US5914389A (en) * 1993-07-30 1999-06-22 The United States Of America As Represented By The Department Of Health And Human Services E6 associated protein
US5981699A (en) * 1994-01-04 1999-11-09 Mitotix, Inc. Human ubiquitin conjugating enzyme
US6004528A (en) * 1997-09-18 1999-12-21 Bergstein; Ivan Methods of cancer diagnosis and therapy targeted against the cancer stemline

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5914389A (en) * 1993-07-30 1999-06-22 The United States Of America As Represented By The Department Of Health And Human Services E6 associated protein
US5981699A (en) * 1994-01-04 1999-11-09 Mitotix, Inc. Human ubiquitin conjugating enzyme
US5736330A (en) * 1995-10-11 1998-04-07 Luminex Corporation Method and compositions for flow cytometric determination of DNA sequences
US6004528A (en) * 1997-09-18 1999-12-21 Bergstein; Ivan Methods of cancer diagnosis and therapy targeted against the cancer stemline
US5888740A (en) * 1997-09-19 1999-03-30 Genaco Biomedical Products, Inc. Detection of aneuploidy and gene deletion by PCR-based gene- dose co-amplification of chromosome specific sequences with synthetic sequences with synthetic internal controls

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BEER-ROMERO P. ET AL.: "Antisense targeting of E6AP elevates p53 in HPV-infected cells but not in normal cells", ONCOGENE, vol. 14, 1997, pages 595 - 602, XP002925920 *
KUMAR S. ET AL.: "Identification of HHR23A as a Substrate for E6-associated Protein-mediated Ubiquitination", J. BIOL. CHEM., vol. 26, no. 25, 25 June 1999 (1999-06-25), pages 18,785 - 18,792, XP002925919 *

Also Published As

Publication number Publication date
AU2720700A (en) 2000-07-24

Similar Documents

Publication Publication Date Title
Nawaz et al. The Angelman syndrome-associated protein, E6-AP, is a coactivator for the nuclear hormone receptor superfamily
Wen et al. The A and B isoforms of the human progesterone receptor operate through distinct signaling pathways within target cells
Verma et al. The ubiquitin-conjugating enzyme UBCH7 acts as a coactivator for steroid hormone receptors
Thenot et al. Estrogen receptor cofactors expression in breast and endometrial human cancer cells
JP2009242417A (en) Expression analysis of fkbp nucleic acid and polypeptide useful for diagnosis and treatment of prostate cancer
Ray et al. Cyclin G‐associated kinase: A novel androgen receptor‐interacting transcriptional coactivator that is overexpressed in hormone refractory prostate cancer
Wise et al. Identification of domains of c-Jun mediating androgen receptor transactivation
US5789170A (en) Specific co-activator for human androgen receptor
JP2009106282A (en) Expression analysis of kiaa nucleic acid and polypeptide useful for diagnosing and treating prostate cancer
Betney et al. Role of conserved hydrophobic amino acids in androgen receptor AF-1 function
JP2008156360A (en) Novel modulator of non-genomic activity of nuclear receptors (mnar) and uses thereof
US7074566B2 (en) Method for testing hormonal effects of substances
WO2000040756A1 (en) Novel coactivator overexpressed in endocrine tumors
JP3862703B2 (en) Method for determining the hormonal action of a substance
AU2002212142B2 (en) Coaster, a human coactivator of steroid receptors
JP2005000056A (en) Hormone-dependent cancer marker and its utilization
WO2000026232A1 (en) Repressor of estrogen receptor activity
Schreiber The transcriptional coactivator PGC-1 [alpha] as a modulator of ERR [alpha] and GR signaling: function in mitochondrial biogenesis
AU2002212142A1 (en) Coaster, a human coactivator of steroid receptors
Superfamily The Angelman Syndrome-Associated
Lonard et al. Roles of ER, SRC-1, and CBP Phosphorylation in Estrogen Receptor-Regulated Gene Expression
WO2005005473A1 (en) Methods to modulate the activity of the oestrogen receptor
Eng Molecular mechanisms of estrogen receptor signaling
WO2002018420A2 (en) Cofactors of the pregnane x receptor and methods of use
Li et al. The Ubiquitin-Conjugating Enzyme UBCH7

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AU CA JP

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase