WO2002070699A2 - Nouveaux cofacteurs du recepteur alpha des oestrogenes et procedes d'utilisation - Google Patents

Nouveaux cofacteurs du recepteur alpha des oestrogenes et procedes d'utilisation Download PDF

Info

Publication number
WO2002070699A2
WO2002070699A2 PCT/EP2002/002189 EP0202189W WO02070699A2 WO 2002070699 A2 WO2002070699 A2 WO 2002070699A2 EP 0202189 W EP0202189 W EP 0202189W WO 02070699 A2 WO02070699 A2 WO 02070699A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
cofactor
nucleic acid
polypeptide
protein
Prior art date
Application number
PCT/EP2002/002189
Other languages
English (en)
Other versions
WO2002070699A3 (fr
Inventor
Michael Albers
Silvia Ellwanger
Eva Löser
Manfred KÖGL
Original Assignee
Lion Bioscience Ag
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 Lion Bioscience Ag filed Critical Lion Bioscience Ag
Priority to AU2002249247A priority Critical patent/AU2002249247A1/en
Publication of WO2002070699A2 publication Critical patent/WO2002070699A2/fr
Publication of WO2002070699A3 publication Critical patent/WO2002070699A3/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out

Definitions

  • Multicellular organisms are dependent on advanced mechanisms of information transfer between cells and body compartments.
  • the information that is transmitted can be highly complex and can result in the alteration of genetic programs involved in cellular proliferation, differentiation or reproduction.
  • the signals, such as hormones are often simple molecules, such as peptides, fatty acids, or cholesterol derivatives.
  • NR nuclear receptors
  • steroid hormones for example, estrogens and glucocorticoids and other cholesterol-derivatives
  • vitamin D for example, vitamin D
  • ecdysone for example, ecdysone
  • cis and trans retinoic acid for example, thyroid hormone, bile acids, fatty acids (and other perbxisomal proliferators), as well as so-called orphan receptors, proteins that are structurally similar to other members of this group, but for which no ligands are known (Escriva, H.
  • Orphan receptors may be indicative of unknown signaling pathways in the cell or may be nuclear receptors that function without ligand activation. The activation of transcription by some of these orphan receptors may occur in the absence of an exogenous ligand and/or through signal transduction pathways originating from the cell surface (Mangelsdorf, D. J. et al., The nuclear receptor superfamily: the second decade, Cell 83, 835-839, 1995).
  • a DNA-binding domain hereinafter referred to as "DBD” usually comprises two zinc finger elements and recognizes a specific Hormone Responsive Element hereinafter referred to as "HRE" within the promoters of responsive genes.
  • HRE Hormone Responsive Element
  • Specific amino acid residues in the “DBD” have been shown to confer DNA sequence binding specificity (Schena, M. & Yamamoto, K.R., Mammalian Glucocorticoid Receptor Derivatives Enhance Transcription in Yeast, Science, 241:965-967, 1988).
  • a Ligand-binding-domain hereinafter referred to as "LBD" is at the carboxy-terminal region of known NRs.
  • the LBD appears to interfere with the interaction of the DBD with its HRE. Hormone binding seems to result in a conformational change in the NR and thus opens this interference (Brzozowski et al., Molecular basis of agonism and antagonism in the oestogen receptor, Nature, 389, 753 - 758, 1997; Wagner et al, A structural role for hormone in the thyroid hormone receptor, Nature, 378, 690 - 697. 1995).
  • TAF transcription activation functions
  • Acidic residues in the amino-terminal domains of some nuclear receptors may be important for these transcription factors to interact with RNA polymerase. TAF activity may be dependent on interactions with other protein factors or nuclear components (Diamond et al, Transcription Factor Interactions: Selectors of Positive or Negative Regulation from a Single DNA Element, Science, 249:1266-1272 , 1990).
  • Certain oncoproteins e.g., c-Jun and c-Fos
  • GR glucocorticoid receptors
  • the receptors for estrogen, vitamins A and D, and fatty acids have been shown to interact, either physically or functionally, with the Jun and Fos components of AP-1 in the transactivation of steroid- or AP-1 regulated genes.
  • Coactivators of transcription are proposed to bridge between sequence specific transcription factors, the basal transcription machinery and in addition to influence the chromatin structure of a target cell.
  • proteins like SRC-1, ACTR, and Gripl which are also cofactors of NRs similar to those disclosed in this invention, interact with NRs in a ligand enhanced manner (Heery et al., A signature motif in transcriptional coactivators mediates binding to nuclear receptors, Nature, 387, 733 - 736; Heinzel et al., A complex containing N-CoR, mSin3 and histone deacetylase mediates transcriptional repression, Nature 387, 43 - 47, 1997). Furthermore, the physical interaction with negative receptor-interacting proteins or corepressors has been demonstrated (Xu et al., Coactivator and Corepressor complexes in nuclear receptor function, Curr Opin Genet Dev, 9 (2), 140 - 147, 1999).
  • HRE hormone response element
  • the present invention relates to the identification of novel interacting proteins of the estrogen receptor alpha.
  • the estrogen receptors are nuclear steroid receptors that mediate the effects of estrogen in the body and are therefore involved in the regulation of important developmental and physiological processes such as sexual differentiation and behaviour, fertility, cardiovascular function, brain function, bone generation and resorption as well as cell proliferation and carcinogenesis.
  • Estrogen receptors exist in two isoforms, which are encoded on two separate genes.
  • the two isoforms termed estrogen receptor alpha and estrogen receptor beta (hereinafter referred to as ER alpha and ER beta, respectively) share some degree of structural and functional similarity.
  • ER alpha and ER beta respectively
  • differences with respect to structure and tissue expression patterns have been recognised which suggest that the two estrogen receptors fulfil distinct physiological roles in many tissues.
  • ligands exist or have been chemically designed (both agonists and antagonists), which selectively modulate the action of only one of the two isoforms, thereby opening ways to more specifically treat medical indications influenced by the action of estrogen (reviewed in Katzenellenbogen et al., Recent Prog Horm Res 55, 163-193 (2000) and Barkhem et al., Mol Pharmacol 54, 105-12 (1998)).
  • both the alpha and beta isoforms are expressed in a range of tissues such as the central nervous system, the cardiovascular system, the immune system, the urogenital tract, the gastrointestinal tract, the bone, the lungs, the mammary gland and the uterus
  • expression of one isoform can be predominant in some cell types.
  • expression of ER alpha in the adult uterus and in the mammary glands is more pronounced than ER beta expression, whereas in the urogenital tract, ER beta seems to be the physiological important form (reviewed in Gustafsson, J Endocr 163, 379-383 (1999)).
  • ER alpha seems to be responsible for most of estrogen's effects on reproduction and reproductive organs, which are fully compromised in its absence in adult female mice (Lubahn et al, PNAS 90, 11162-11166 (1993)).
  • Females are infertile with hypoplastic uteri and hypere- mic ovaries and they lack breast tissue development. Males are also infertile.
  • SERMs selective estrogen receptor modulators
  • antioxidant-regulated genes such as the QR gene, which products control the concentrations of free radicals and reactive oxygen - important players in the onset and course of cancer - might be regulated by ER beta.
  • An alternative way, in which cell proliferation could be controlled by ER beta was suggested by Poelzl et al., PNAS 97, 2836-2839 (2000).
  • ER beta but not ER alpha, was demonstrated to interact directly and specifically with a cell- cycle regulatory protein, MAD2 (mitosis arrest-deficient 2) in a ligand independent manner. This could suggest, that the regulatory functions of ER beta in cell proliferation might be mediated through direct protein-protein contacts with a cell cycle spindle assembly protein and thus in a way distinct from the established function of the ERs as transcription factors.
  • the present invention relates to the identification of novel interacting polypeptides of the estrogen receptor alpha.
  • the identification and characterisation of protein factors which modulate ER transactivation activity could be of great benefit for the treatment of numerous diseases such as osteoporosis and other bone diseases, failures in reproductive functions, cancer, cardiovascular diseases such as atherosclerosis, as well as the prevention of hot flushes, mood changes and Alzheimer's disease.
  • the present invention provides novel proteins, nucleic acids, and methods useful for developing and identifying compounds for the treatment of these diseases.
  • the invention also provides for methods to test if a certain compound promotes or disrupts the interaction of these proteins with ER alpha, allowing the screening for compounds with estrogen-regulated cellular effects.
  • These novel proteins interact, presumably also in vivo, with the ER alpha receptor and shall hereinafter collectively be referred to as "cofactors" or "CFs", although some of them in fact do belong to the nuclear receptor family of polypeptides.
  • the importance of this invention is manifested in the effects of the CFs to modulate genes involved in cellular functions like regulation of metabolism and cell homeostasis, cell proliferation and differentiation, pathological cellular aberrations, or cellular defense mechanisms.
  • the CF proteins are useful for screening for ligands of the ER alpha thereby providing for agents which influence the activity of ER alpha and thus the activity of genes controlled by ER alpha.
  • the present invention provides isolated nucleic acid sequences for novel CFs.
  • the present invention provides the cDNA sequences encoding human CFs.
  • nucleic acid sequences have a variety of uses. For example, they are useful for making vectors and for transforming cells, both of which are ultimately useful for production of the CF polypeptides. They are also useful as scientific research tools for developing nucleic acid probes for determining expression levels of the cofactor genes, e.g., to identify diseased or otherwise abnormal states. They are useful for developing analytical tools such as anti sense oligonucleotides for selectively inhibiting expression of the cofactor genes to determine physiological responses.
  • a homogenous composition comprising the cofactor proteins.
  • the protein is useful for screening drugs for agonist and antagonist activity, and, therefore, for screening for drugs useful in regulating physiological responses associated with the cofactors according to the invention.
  • antagonists to the CFs could be used to treat metabolic disorders, immunological indications, hormonal dysfunctions, neurosystemic diseases.
  • the proteins are also useful for developing antibodies for detection of the proteins.
  • vectors such as plasmids, comprising the cofactor nucleic acid sequences that may further comprise additional regulatory elements, e.g., promoters,
  • transformed cells that express the cofactors
  • nucleic acid probes e.g., nucleic acid probes
  • antisense oligonucleotides e.g., agonists, (f) antagonists, and (g) transgenic mammals.
  • transgenic mammals e.g., transgenic mammals.
  • Further aspects of the invention comprise methods for making and using the foregoing compounds and compositions.
  • the present invention comprises, in part, novel cofactors (CF16, CF17, CF18, CF19, CF40, CF41, CF42, and CF43) of the mammalian ER alpha.
  • novel cofactors CF16, CF17, CF18, CF19, CF40, CF41, CF42, and CF43
  • cofactors are those having an amino acid sequence substantially the same as SEQ ID NOs. 3, 6, 9, 12, 15, 18, 21, and/or 24.
  • cofactor As used herein, if reference to the cofactor is made or the cofactor "X", wherein "X” stands for the number designating the cofactor, it is meant as a reference to any protein having an amino acid sequence substantially the same as SEQ ID NO. 3 for CF16, SEQ ID NO. 6 for CF17, SEQ ID NO. 9 for CF18, SEQ ID NO. 12 for CF19, SEQ ID NO. 15 for CF40, SEQ ID NO. 18 for CF41, SEQ ID NO. 21 for CF42, and SEQ ID NO. 24 for CF43.
  • the present invention also comprises the nucleic acid sequences encoding the cofactors 16 to 20, which nucleic acid sequences are substantially the same as SEQ ID NO. 1 for CF16, SEQ ID NO. 4 for CF17, SEQ ID NO. 7 for CF18, SEQ ID NO. 10 for CF19, SEQ ID NO. 13 for CF40, SEQ ID NO. 16 for CF41, SEQ ID NO. 19 for CF42, and SEQ ID NO. 22 for CF43 all encoding human cofactors as preferred embodiments and/or the complements thereof as shown in SEQ ID NO. 2 for CF16, SEQ ID NO. 5 for CF17, SEQ ID NO. 8 for CF18, SEQ ID NO. 11 for CF19, SEQ ID NO. 14 for CF40, SEQ ID NO. 17 for CF41, SEQ ID NO. 20 for CF42, and SEQ ID NO. 23 for CF43.
  • complement refers to the complementary strand of the nucleic acid according to the invention, thus the strand that would hybridize to the nucleic acid according to the invention.
  • complements refers to the complementary strand of the nucleic acid according to the invention, thus the strand that would hybridize to the nucleic acid according to the invention.
  • all DNA sequences herein are however written in 5 '-3' orientation, thus the complements depicted are actually “reverse” complements For simplification purposes they are however some times referred to simply as “complements”.
  • a protein "having an amino acid sequence substantially the same as SEQ ID NO x" means a protein whose amino acid sequence is the same as SEQ ID NO x or differs only in a way such that at least 50% of the residues compared in a sequence alignment with SEQ ID NO. x are identical, preferably 75% of the residues are identical, even more preferably 95% of the residues are identical and most preferably at least 98% of the residues are identical
  • a molecule having a nucleotide sequence substantially the same as SEQ ID NO y means a nucleic acid encoding a protein "having an amino acid sequence substantially the same as SEQ ID NO y+1" (wherein “y+1” is the number of the amino acid sequence for which nucleotide sequence "y” codes) as defined above.
  • This definition is intended to encompass natural allelic variations in the CF sequences.
  • Cloned nucleic acid provided by the present invention may encode CF proteins of any species of origin, including (but not limited to), for example, mouse, rat, rabbit, hamster, cat, dog, pig, primate, and human.
  • nucleic acids provided by the invention encode CFs of mammalian, preferably mouse and most preferably human origin.
  • Nucleic acid hybridization probes provided by the invention are nucleic acids consisting essentially of the nucleotide sequences complementary to any sequence depicted in SEQ ID NO. 1, 4, 7, 10, 13, 16, 19, and 22, and/or the complements thereof as shown in SEQ ID NO. 2, 5, 8, 11, 14, 17, 20, and 23, or parts thereof which are effective in nucleic acid hybridization.
  • Nucleic acid hybridization probes provided by the invention are nucleic acids capable of detecting i.e. hybridizing to the gene encoding the polypeptides according to SEQ ID Nos: 3, 6, 9, 12, 15, 18, 21, and 24.
  • Nucleic acid probes are useful for detecting CF gene expression in cells and tissues using techniques well-known in the art, including, but not limited to, Northern blot hybridization, in situ hybridization, and Southern hybridization to reverse transcriptase - polymerase chain reaction product DNAs.
  • the probes provided by the present invention including oligonucleotide probes derived therefrom, are also useful for Southern hybridization of mammalian, preferably human, genomic DNA for screening for restriction fragment length polymorphism (RFLP) associated with certain genetic disorders.
  • RFLP restriction fragment length polymorphism
  • complementary means a nucleic acid having a sequence that is sufficiently complementary in the Watson- Crick sense to a target nucleic acid to bind to the target under physiological conditions or experimental conditions those skilled in the art routinely use when employing probes.
  • nucleic acid sequence will hybridize with a complementary nucleic acid sequence under highly stringent conditions as defined herein, even though some mismatches may be present.
  • Such closely matched, but not perfectly complementary sequences are also encompassed by the present invention.
  • differences may occur through genetic code degeneracy, or by naturally occurring or man made mutations and such mismatched sequences would still be encompassed by the present claimed invention.
  • the nucleotide sequences of the nuclear cofactors SEQ ID NOs: 1, 4, 7, 10, 13, 16, 19, and 22, and/or their complements SEQ ID NOs 2, 5, 8, 11, 14, 17, 20, and 23 can be used to derive oligonucleotide fragments (probes) of various length. Stretches of 17 to 30 nucleotides are used frequently but depending on the screening parameters longer sequences as 40, 50, 100, 150 up to the full length of the sequence may be used. Those probes can be synthesized chemically and are obtained readily from commercial oligonucleotide providers. Chemical synthesis has improved over the years and chemical synthesis of oligonucleotides as long as 100-200 bases is possible.
  • probes can also be obtained by biochemical de novo synthesis of single stranded DNA.
  • nucleotide sequence of the nuclear receptors or their complements serve as a template and the corresponding complementary strand is synthesized.
  • a variety of standard techniques such as nick translation or primer extension from specific primers or short random oligonucleotides can be used to synthesize the probe (Sambrook, J., Fritsch, E.F. & Maniatis , T. Molecular cloning: a laboratory manual. Cold Spring Harbor Press, Cold Spring Harbor, 1989)).
  • Nucleic acid reproduction technologies exemplified by the polymerase chain reaction (Saiki, R.K.
  • this can be done by cloning the sequence in a vector which carries the respective promoter sequence. It is also possible to introduce the needed sequence by synthesizing a primer with the needed promoter in the form of a 5' "tail". The chemical synthesis of a RNA probe is another option.
  • Appropriate means are available to detect the event of a hybridization.
  • labels and detection systems e.g. radioactive isotopes, fluorescent, or chemiluminescent molecules which can be linked to the probe.
  • methods of introducing haptens which can be detected by antibodies or other ligands such as the avidin/biotin high affinity binding system.
  • Hybridization can take place in solution or on solid phase or in combinations of the two, e.g. hybridization in solution and subsequent capture of the hybridization product onto a solid phase by immobilized antibodies or by ligand coated magnetic beads.
  • Hybridization probes act by forming selectively duplex molecules with complementary stretches of a sequence of a gene or a cDNA.
  • the selectivity of the process can be controlled by varying the conditions of hybridization.
  • stringent conditions for the hybridization e.g. low salt in the range of 0.02 M to 0.15 M salt and/or high temperatures in the range from 50°C degrees centigrade to 70°C degrees centigrade.
  • Stringency can be further improved by the addition of formamide to the hybridisation solution.
  • stringent hybridization conditions are those where between 0.02 M to 0.15 M salt and/or high temperatures in the range from 50°C degrees centigrade to 70°C degrees centigrade are applied.
  • highly stringent hybridization conditions are those where between 0.02 - 0.3 M salt and 65°C degrees centigrade are applied for about 5 to 18 hours of hybridization time and additionally, the sample filters are washed twice for about 15 minutes each at between 60°C - 65°C degrees centigrade, wherein the first washing fluid contains about 0.1 M salt (NaCI and/or Sodium Citrate) and the second contains only about 0.02 M salt (NaCI and/or Sodium Citrate).
  • first washing fluid contains about 0.1 M salt (NaCI and/or Sodium Citrate) and the second contains only about 0.02 M salt (NaCI and/or Sodium Citrate).
  • the following conditions are considered to be highly stringent:
  • Unspecific hybridization products are removed by washing the reaction products repeatedly in 2 x SSC solution and increasing the temperature.
  • the nucleotide sequences of the cofactors CF16 to CF19 and CF40 to CF43 or their complements can be used to design primers for a polymerase chain reaction. Due to the degeneracy of the genetic code the respective amino acid sequence is used to design oligonucleotides in which varying bases coding for the same amino acid are included. Numerous design rules for degenerate primers have been published (Compton et al, 1990). As in hybridization there are a number of factors known to vary the stringency of the PCR. The most important parameter is the annealing temperature. To allow annealing of primers with imperfect matches annealing temperatures are often much lower than the standard annealing temperature of 55°C, e.g.
  • PCR reaction products can be cloned. Either the PCR product is cloned directly, with reagents and protocols from commercial manufacturers (e.g. from Invitrogen, San Diego, USA). Alternatively, restriction sites can be introduced intro the PCR product via a 5'-tail of the PCR primers and used for cloning.
  • GENETIC VARIANTS GENETIC VARIANTS
  • Fragments from the nucleotide sequence of the cofactors or their complements can be used to cover the whole sequence with overlapping sets of PCR primers. These primers are used to produce PCR products using genomic DNA from a human diversity panel of healthy individuals or genomic DNA from individuals which are phenotypically conspicuous.
  • the PCR products can be screened for polymorphisms, for example by denaturing gradient gel electrophoresis, binding to proteins detecting mismatches or cleaving heteroduplices or by denaturing high-performance liquid chromatography. Products which display mutations need to be sequenced to identify the nature of the mutation. Alternatively, PCR products can be se- quenced directly omitting the mutation screening step to identify genetic polymorphisms.
  • genetic variants are identified and are associated with a discrete phenotype, these genetic variations can be included in diagnostic assays.
  • the normal variation of the human population is of interest in designing screening assays as some variants might interact better or worse with a respective lead, i.e. therapeutic or potentially therapeutic substance (a pharmacody- namic application).
  • Polymorphisms or mutations which can be correlated to phenotypic outcome are a tool to extend the knowledge and the commercial applicability of the nucleotide sequences of the cofactors CF16 to CF19 and CF40 to CF43 or their complements or their gene products, as variants might have a slightly different molecular behavior or desired properties.
  • Disease-causing mutations or polymorphisms allow the replacement of this disease inducing gene copy with a wild-type copy by means of gene therapy approaches and/or the modulation of the activity of the gene product by drugs.
  • DNA which encodes cofactor CF16 to CF19 and CF40 to CF43 may be obtained, in view of the instant disclosure, by chemical synthesis, by screening reverse transcripts of mRNA from appropriate cells or cell line cultures, by screening genomic libraries from appropriate cells, or by combinations of these procedures, as illustrated below.
  • oligonucleotide probes generated from the CF nucleotide sequences information provided herein. These oligonucleotides are in addition useful to isolate a full length cDNA from an appropriate cDNA library. Probes may be labeled with a detectable group such as a fluorescent group, a radioactive atom or a chemiluminescent group in accordance with known procedures and used in conventional hybridization assays, as described in greater detail in the examples below.
  • the CF nucleotide sequences may be obtained by use of the polymerase chain reaction (PCR) procedure, with the PCR oligonucleotide primers being produced from the CF nucleotide sequences provided herein, according to SEQ ID NO 1, SEQ ID NO. 4, SEQ ID NO. 7, SEQ ID NO. 10, SEQ ID NO. 13, SEQ ID NO. 16, SEQ ID NO. 19, and SEQ ID NO. 22, and/or the complements thereof as shown in SEQ ID NO. 2, SEQ ID NO. 5, SEQ ID NO. 8, SEQ ID NO. 11, SEQ ID NO. 14, SEQ ID NO. 17, SEQ ID NO. 20, and SEQ ID NO. 23, or parts thereof.
  • PCR polymerase chain reaction
  • nucleic acid according to the invention may be labeled, e.g. for use as a probe.
  • single and differential labeling agents and methods any agents and methods which are known in the art can be used provided that they do not significantly altering the stability or function of said primer in the DNA sequencing method of the present invention.
  • single and differential labels may consist of the group comprising enzymes such as ⁇ - galactosidase, alkaline phosphatase and peroxidase, enzyme substrates, coenzymes, dyes, chromophores, fluorescent, chemiluminescent and bioluminescent labels such as FITC, Cy5, Cy5.5, Cy7, Texas-Red and IRD40(Chen et al. (1993), J. Chromatog. A 652: 355-360 and Kambara et al. (1992), Electrophoresis 13: 542-546), ligands or haptens such as biotin, and radioactive isotopes such as 3 H, 35 S, 32 P 125 I and 14 C.
  • enzymes such as ⁇ - galactosidase, alkaline phosphat
  • the CF nucleic acids or polypeptides may be synthesized in host cells transformed with a recombinant expression construct comprising a nucleic acid encoding any of the cofactors according to the invention, namely CF16 to CF19 and CF40 to CF43.
  • a recombinant expression construct can also be comprised of a vector that is a replicable DNA construct.
  • Amplification vectors do not require expression control domains. All that is needed is the ability to replicate in a host, usually conferred by an origin of replication, and a selection gene to facilitate recognition of transformants. See, Sambrook et al., Molecular Cloning: A Laboratory Manual (2nd Edition, Cold Spring Harbor Press, New York, 1989).
  • An expression vector comprises a polynucleotide operatively linked to a prokaryotic promoter.
  • an expression vector is a polynucleotide operatively linked to an enhancer-promoter that is a eukaryotic promoter, and the expression vector further has a polyade- nylation signal that is positioned 3' of the carboxy-terminal amino acid and within a transcriptional unit of the encoded polypeptide.
  • a promoter is a region of a DNA molecule typically within about 500 nucleotide pairs in front of (upstream of) the point at which transcription begins (i.e., a transcription start site).
  • a vector contains a replicon and control sequences which are derived from species compatible with the host cell. The vector ordinarily carries a replication site, as well as marking sequences which are capable of providing phenotypic selection in transformed cells.
  • An enhancer provides specificity of time, location and expression level for a particular encoding region (e.g., gene).
  • a major function of an enhancer is to increase the level of transcription of a coding sequence in a cell.
  • enhancer-promoter means a composite unit that contains both enhancer and promoter elements.
  • An enhancer-promoter is operatively linked to a coding sequence that encodes at least one gene product.
  • An enhancer-promoter used in a vector construct of the present invention may be any enhancer-promoter that drives expression in a prokaryotic or eukaryotic cell to be trans- formed/transfected.
  • a coding sequence of an expression vector is operatively linked to a transcription terminating region.
  • RNA polymerase transcribes an encoding DNA sequence through a site where poly- adenylation occurs.
  • An expression vector that comprises a polynucleotide that encodes one of the the polypeptides of the cofactors CF16 to CF19 and CF40 to CF43 is meant to include a sequence of nucleotides encoding a CF polypeptide sufficient in length to distinguish said segment from a polynucleotide segment encoding a non- cofactor polypeptide.
  • a polypeptide of the invention may also encode biologically functional polypeptides or peptides which have variant amino acid sequences, such as with changes selected based on considerations such as the relative hydropathic score of the amino acids being exchanged.
  • variant sequences are those isolated from natural sources or induced in the sequences disclosed herein using a mutagenic procedure such as site-directed mutagenesis.
  • an expression vector of the present invention may contain regulatory elements for optimized translation of the polypeptide in prokaryotic or eukaryotic systems. These sequences are operatively located around the transcription start site and are most likely similar to ribosome recognition sites like prokaryotic ribosome binding sites (RBS) or eukaryotic Kozak sequences as known in the art (Kozak M., Initiation of translation in prokaryotes and eukaryotes. Gene 234, 187-208 (1999)).
  • RBS prokaryotic ribosome binding sites
  • Kozak sequences as known in the art
  • An expression vector of the present invention is useful both as a means for preparing quantities of the CFs' polypeptide-encoding DNA itself, and as a means for preparing the encoded CFs' polypeptide and peptides. It is contemplated that where cofactor polypeptides of the invention are made by recombinant means, one may employ either prokaryotic or eukaryotic expression vectors as shuttle systems.
  • a vector such as a plasmid, that incorporates a eukaryotic origin of replication.
  • a vector such as a plasmid, that incorporates a eukaryotic origin of replication.
  • the invention provides homogeneous compositions of mammalian cofactor polypeptides produced by transformed prokaryotic or eukaryotic cells as provided herein. Such homogeneous compositions are intended to be comprised of mammalian cofactor protein that comprises at least 90% of the protein in such homogenous composition.
  • the invention also provides membrane preparation from cells expressing the mammalian cofactors polypeptides as the result of transformation with a recombinant expression construct, as described here.
  • recombinant protein or coding sequence both also include tagged versions of the polypeptides depicted in SEQ ID NO. 3, SEQ ID NO. 6, SEQ ID NO. 9, SEQ ID NO. 12, SEQ ID NO. 15, SEQ ID NO. 18, SEQ ID NO. 21, and/or SEQ ID NO. 24 and fusion proteins of said proteins with any other recombinant protein.
  • Tagged versions here means that small epitopes of 3-20 amino acids are added to the original protein by extending the coding sequence either at the 5 'or the 3 'terminus leading to N- terminal or C-terminal extended proteins respectively, or that such small epitopes are included elsewhere in the protein.
  • Fusion proteins where the added sequences are coding for longer proteins, varying between 2 and 100 kDa.
  • Tags and fusion proteins are usually used to facilitate purification of recombinant proteins by specific antibodies or affinity matrices or to increase solubility of recombinant proteins within the expression host. Fusion proteins are also of major use as essential parts of yeast two hybrid screens for interaction partners of recombinant proteins.
  • EEF alpha Tubulin
  • B-tag QYPALT
  • E tag GAPVPYPDPLEPR
  • c-myc Tag EQKLISEEDL
  • Flag epitope DYKDDDDK
  • HA tag YPYDVPDYA
  • 6 or 10 x His Tag HSV
  • QPELAPEDPED Pk-Tag
  • GKPIPNPLLGLDST protein C
  • EDQVDPRLIDGK T7
  • MASMTGGQQMG MASMTGGQQMG
  • VSV-G YTDIEMNRLGK
  • Fusion proteines may include Thiore- doxin, Glutathiontransferase (GST), Maltose binding Protein (MBP), Cellulose Binding protein, calmodulin binding protein, chitin binding protein, ubiquitin, the Fc part of Immuno- globulins, and the IgG binding domain of Staphylococcus aureus protein A.
  • vector constructs harboring recombinant cofactors as set forth in SEQ ID NO. 1, SEQ ID NO. 4, SEQ ID NO. 7, SEQ ID NO. 10, SEQ ID NO. 13, SEQ ID NO. 16, SEQ ID NO. 19, and/or SEQ ID NO. 22 and/or the complements thereof SEQ ID NO. 2, SEQ ID NO. 5, SEQ ID NO. 8, SEQ ID NO. 11, SEQ ID NO. 14, SEQ ID NO. 17, SEQ ID NO. 20, and/or SEQ ID NO. 23 are transformed or transfected into appropriate host cells.
  • a recombinant host cell of the present invention is transfected with a polynucleotide SEQ ID NO. 1, SEQ ID NO. 4, SEQ ID NO. 7, SEQ ID NO. 10, SEQ ID NO. 13, SEQ ID NO. 16, SEQ ID NO. 19, and/or SEQ ID NO. 22.
  • Means of transforming or transfecting cells with exogenous polynucleotide such as DNA molecules are well known in the art and include techniques such as calcium-phosphate- or DEAE-dextran-mediated transfection, protoplast fusion, electroporation, liposome mediated transfection, direct microinjection and virus infection (Sambrook et al., 1989).
  • transfection mediated by either calcium phosphate or DEAE-dextran The most widely used method for transfection of eukaryotic cells is transfection mediated by either calcium phosphate or DEAE-dextran. Although the mechanism remains obscure, it is believed that the transfected DNA enters the cytoplasm of the cell by endocytosis and is transported to the nucleus. Depending on the cell type, up to 90% of a population of cultured cells may be transfected at any one time. Because of its high efficiency, transfection mediated by calcium phosphate or DEAE-dextran is the method of choice for studies requiring transient expression of the foreign nucleic acid in large numbers of cells. Calcium phosphate-mediated transfection is also used to establish cell lines that integrate copies of the foreign DNA, which are usually arranged in head-to-tail tandem arrays into the host cell genome.
  • protoplasts derived from bacteria carrying high numbers of copies of a plasmid of interest are mixed directly with cultured mammalian cells. After fusion of the cell membranes (usually with polyethylene glycol), the contents of the bacterium are delivered into the cytoplasm of the mammalian cells and the plasmid DNA is transported to the nucleus.
  • Protoplast fusion is not as efficient as transfection for many of the cell lines that are commonly used for transient expression assays, but it is useful for cell lines in which endocytosis of DNA occurs inefficiently. Protoplast fusion frequently yields multiple copies of the plasmid DNA tandemly integrated into the host chromosome.
  • Electroporation may be extremely efficient and may be used both for transient expression of cloned genes and for establishment of cell lines that carry integrated copies of the gene of interest. Electroporation, in contrast to calcium phosphate-mediated transfection and protoplast fusion, frequently gives rise to cell lines that carry one, or at most a few, integrated copies of the foreign DNA.
  • Liposome transfection involves encapsulation of DNA and RNA within liposomes, followed by fusion of the liposomes with the cell membrane. The mechanism of how DNA is delivered into the cell is unclear but transfection efficiencies may be as high as 90%.
  • Direct microinjection of a DNA molecule into nuclei has the advantage of not exposing DNA to cellular compartments such as low-pH endosomes. Microinjection is therefore used primarily as a method to establish lines of cells that carry integrated copies of the DNA of interest.
  • adenovirus vector-mediated cell transfection has been reported for various cells (Stratford-Perricaudet et al, 1992).
  • the commonly used viral vectors for the transfer of genes according to the current state of the art are mainly retroviral, lentiviral, adenoviral and adeno-associated viral vectors. These are circular nucleotide sequences derived from natural viruses in which at least the viral structural protein encoding genes are replaced by the construct to be transferred.
  • Retroviral vector systems provide the prerequisite for a long-lasting expression of the transgene by the stable, but non-directed integration into the genome of the host.
  • Vectors of the younger generation possess no irrelevant and potentially immunogenic proteins, furthermore, there is no pre-existing immunity of the recipient in view of the vector.
  • Retroviruses contain an RNA-genome, that is packed into a lipid coating, which consists out of parts of the host cell membrane and viral proteins.
  • the RNA-genome is reversely transcribed and integrated into the target-cell DNA using the enzyme integrase. This can subsequently be transcribed and translated by the infected cell, thereby viral compounds are produced which then form retroviras particles. RNA will then be exclusively included in the newly produced viruses.
  • gag which codes for viral structural proteins, so-called group-specific antigenes, pol for Enzyme like reverse transcriptase and integrase and env for the noirenvelope" protein, which is responsible for the binding of the host specific receptor.
  • group-specific antigenes pol for Enzyme like reverse transcriptase and integrase and env for the noirenvelope protein, which is responsible for the binding of the host specific receptor.
  • the production of the replication incompetent viruses occurs after transfection in so-called packaging-celllines which are in addition provided with gag/pol-encoding genes and express those decisin trans” and thereby complement the formation of replicationincompeten (i.e. gag/pol-deletet) transgene virus particles.
  • An alternative is cotransfection of the essential virus genes, wherein only the transgen containing vector carries the packaging signal.
  • Novel, non-viral vectors consist out of autonomically and self-integrating DNA sequences, the transposons, which are introduced into the host cell by, e.g. liposomal transfection and were for the first time successfully used for the expression of human transgenes in mammalian cells (Yant et al, 2000).
  • a transfected cell may be prokaryotic or eukaryotic, transfection may be transient or stable. Where it is of interest to produce a full length human CF16 to CF19 and CF40 to CF 43 protein, cultured mammalian or human cells are of particular interest.
  • the recombinant host cells of the present invention are prokaryotic host cells.
  • eukaryotic microbes such as yeast may also be used illustrative examples for suitable cells and organisms for expression of recombinant proteins are belonging to but not limited to the following examples: Insect cells, such as Drosophila Sf21, SF9 cells or others, Expression strains of Escherichia coli, such as XL1 blue, BRL21, Ml 5, Saccharomyces cerevisiae, Schizosaccharomyces pombe, Hansenula polymorpha and Pichia pastoris strains, immortalized mammalian cell lines such as AtT-20, VERO and HeLa cells, Chinese hamster ovary (CHO) cell lines, and W138, BHK, COSM6, COS-7, 293 and MDCK cells, BHK-21 cells, Att 20HeLa cells, HeK 294, T47 D cells and others.
  • Expression of recombinant proteins within the scope of this invention can also be performed in vitro. This may occur by a two step procedure, thereby producing first mRNA by in vitro transcription of an apt polynucleotide construct followed by in vitro translation with convenient cellular extracts. These cellular extracts may be reticulocyte lysates but are not limited to this type.
  • In vitro transcription may be performed by T7 or SP6 DNA polymerase or any other RNA polymerase which can recognize per se or with the help of accessory factors the promoter sequence contained in the recombinant DNA construct of choice.
  • one of the recently made available one step coupled transcription/translation systems may be used for in vitro translation of DNA coding for the proteins of this invention.
  • TNT® T7 Quick System by Promega.
  • transfected cells are maintained for a period of time sufficient for expression of the recombinant cofactor proteins according to the invention.
  • a suitable maintenance time depends strongly on the cell type and organism used and is easily ascertainable by one skilled in the art. Typically, maintenance time is from about 2 hours to about 14 days. For the same reasons and for sake of protein stability and solubility incubation temperatures during maintenance time may vary from 20°C to 42 °C.
  • Recombinant proteins are recovered or collected either from the transfected cells or the medium in which those cells are cultured. Recovery comprises cell disruption, isolation and purification of the recombinant protein. Isolation and purification techniques for polypeptides are well-known in the art and include such procedures as precipitation, filtration, chromatography, electrophoresis and the like.
  • purification includes but is not limited to affinity purification of tagged or nontagged recombinant proteins.
  • affinity purification of tagged proteines small molecules such as gluthathione, maltose or chitin, specific proteins such as the IgG binding domain of Staphylococcus aureus protein A, antibodies or specific chelates which bind with high affinity to the tag of the recombinant protein are employed.
  • affinity purification of non-tagged proteins specific monoclonal or polyclonal antibodies, which were raised against said protein, can be used.
  • immobilized specific interactors of said protein may be employed for affinity purification. Interactors include native or recombinant proteins as well as native or artificial specific low molecular weight ligands.
  • the protein itself may be produced using chemical methods to synthesize any of the amino acid sequences according to the invention (SEQ ID NO. 3, SEQ ID NO. 6, SEQ ID NO. 9, SEQ ID NO. 12, SEQ ID NO. 15, SEQ ID NO. 18, SEQ ID NO. 21, and/or SEQ ID NO. 24) or that is encoded by the nucleotide sequences according to the invention and/or the complements thereof or a portion thereof.
  • peptide synthesis can be performed using conventional Merrifield solid phase f-Moc or t-Boc chemistry or various solid-phase techniques (Roberge, J. Y. et al.
  • the newly synthesized peptide(s) may be substantially purified by preparative high performance liquid chromatography (e.g., Creighton, T. (1983) Proteins, Structures and Molecular Principles, WH Freeman and Co., New York, N. Y.).
  • the composition of the synthetic peptides may be confirmed by amino acid analysis or sequencing (e.g., the Edman degradation procedure; Creighton, supra). Additionally, the amino acid sequences according to the invention, i.e. SEQ ID NO. 3, SEQ ID NO. 6, SEQ ID NO. 9, SEQ ID NO. 12, SEQ ID NO.
  • SEQ ID NO. 18, SEQ ID NO. 21, and SEQ ID NO. 24 or the sequence that is encoded by SEQ ID NO. 1, SEQ ID NO. 4, SEQ ID NO. 7, SEQ ID NO. 10, SEQ ID NO. 13, SEQ ID NO. 16, SEQ ID NO. 19, and SEQ ID NO. 22 or any part thereof, may be altered during direct synthesis and/or combined using chemical methods with sequences from other proteins, or any part thereof, to produce a variant polypeptide.
  • CF16 to CF19 and CF40 to CF43 all bind ER alpha, presumably also in vivo.
  • one or more of the CFs are complexed with the ER alpha polypeptide or portions thereof, preferentially in vitro.
  • the ER alpha polypep- tide receptor is encoded by a genomic nucleic acid sequence according to SEQ ID NO. 25 or 38.
  • the receptor has an amino acid sequence according to SEQ ID NO. 27 or 30.
  • Such complexes are particularly suited for all forms of binding or screening assays (see also below).
  • such assays are performed with complexes of the receptor(s) associated with one or more of the CF proteins.
  • a trimeric complex consisting of ER alpha homodimers bound to one of the CFs.
  • ER alpha may bind in monomeric form to one of the CFs.
  • Such complexes may be used in binding and screening assays as outlined below.
  • the entire CF polypeptide is part of the complex or alternatively only a portion, e.g. a truncated fragment of the other polypeptide (ER alpha) is part of the complex.
  • the present invention concerns a method for identifying new inhibitory or stimulatory substances of the cofactors according to the invention, these substances may be termed as “candidate substances”. It is contemplated that this screening technique proves useful in the general identification of compounds that serve the purpose of inhibiting or stimulating cofactor activity.
  • DHEA beta-Hydroxy-5alpha-androstan-17-one
  • the pharmaceutical agents to be screened can also be derived from chemical compositions or man-made compounds.
  • the candidate substances could also include monoclonal or polyclonal antibodies, peptides or proteins, such as those derived from recombinant DNA technology or by other means, including chemical peptide synthesis.
  • the active compounds may include fragments or parts or derivatives of naturally-occurring compounds or may be only found as active combinations of known compounds which are other- wise inactive. We anticipate that such screens will in some cases lead to the isolation of agonists of nuclear receptors or cofactors, in other cases to the isolation of antagonists. In other instances, substances will be identified that have mixed agonistic and antagonistic effects, or affect nuclear receptors or cofactors in any other way.
  • the invention concerns the isolation of substance inhibiting the interaction of the cofactor protein and ER alpha.
  • substances are useful for the development of drugs against diseases as listed above.
  • Substances disrupting the interactions may be isolated by a variety of screening methods including the two hybrid system or the reverse two hybrid system (Lenna CA. and Hannink, M. 1996, Nucl. Acids Res. 24: 3341-3347), or any variation of cellular or cell free assays as described in this invention, as is obvious to anyone skilled in the art.
  • the binding of the cofactor protein and ER alpha can be used to monitor the binding of a substance to one of the binding partners.
  • the substance which can be a small molecule such as a ligand to a nuclear receptor, will lead to a change in the allosteric conformation of the binding protein which in consequence leads to a loss of the interaction of the two proteins.
  • ligand-dependent protein- protein interactions one can design assays where the protein-protein interaction serves as a surrogate read-out for the binding of one of the proteins to small molecule ligand. Any assay method which is useful for the measurement of protein-protein interactions can be used for such an indirect assay.
  • a recombinant cell line To identify a candidate substance capable of influencing the cofactor protein activity, one first obtains a recombinant cell line.
  • a reporter such as luciferase, fluorescent proteins such as green or red fluorescent protein
  • beta- galactosidase alpha-galactosidase
  • beta-lactamase alpha-galactosidase
  • chloramphenicol-acetyl-transferase beta-
  • the amount of reporter protein present reflects the activity of the cofactor.
  • This recombinant cell line is then screened for the effect of substances on the expression of the reporters, thus measuring the effect of these substances on the activity of the cofactor.
  • These substances can be derived from natural sources, such as fungal extracts, plant extracts, bacterial extracts, higher eukaryotic cell extracts, or even extracts from animal sources, or marine, forest or soil samples, may be assayed for the presence of potentially useful pharmaceutical agents. It will be understood that that the pharmaceutical agents to be screened may be derived from chemical compositions or man-made compounds.
  • the candidate substances can also include monoclonal or polyclonal antibodies, peptides or proteins, such as those derived from recombinant DNA technology or by other means, including chemical peptide synthesis.
  • the active compounds may include fragments or parts or derivatives of naturally-occurring compounds or may be only found as active combinations of known compounds which are otherwise inactive.
  • the assay can be performed by firstly bringing a suitable cell containing a reporter gene which transcription is influenced by the cofactors activity in contact with a compound and secondly monitoring the expression of the reporter gene to evaluate the effect of the compound on the activity of the cofactor.
  • assays are included where measuring the activity of di- or multimeric complexes of the cofactor and other proteins such as ER alpha. Further included are assays aiming at the identification of compounds which specifically influence only the monomeric, homodimeric or homomultimeric form of the cofactor, or influencing only multimeric forms of the cofactor. Such assays include measuring the effect of a compound on the cofactor in the absence of a binding partner, and measuring the effect of a compound on the cofactor in the presence of a binding partner, such as ER alpha.
  • a binding partner such as ER alpha
  • a cell line where the activity of ER alpha or any other nuclear receptor determines the expression of a reporter can be obtained by generating an artificial promoter upstream of the reporter gene, which contains preferably multiple copies of HREs to which ER alpha or any other nuclear receptor binds.
  • transgenic animals described in the invention can be used to derive cell lines useful for cellular screening assays.
  • Cell lines useful for such an assay include many different kinds of cells, including prokaryotic, animal, fungal, plant and human cells.
  • Yeast cells can be used in this assay, including Saccharomyces cerevisiae and Schizosaccharomyces pombe cells.
  • the two hybrid assay relies on reconstituting in vivo a functional transcriptional activator protein from two separate fusion proteins.
  • the method makes use of chimeric genes which express hybrid proteins.
  • a first hybrid gene comprises the coding sequence for a DNA-binding domain of a transcriptional activator fused in frame to the coding sequence for a cofactor.
  • the second hybrid protein encodes a transcriptional activation domain fused in frame to another gene, for example ER alpha.
  • cofactor and ER alpha proteins are able to interact, they bring into close proximity the two domains of the transcriptional activator. This proximity is sufficient to cause transcription of a reporter gene which is operably linked to a transcriptional regulatory site responsive to the transcriptional activator, and expression of the reporter gene can be detected and used to score for the interaction of the cofactor and ER alpha proteins.
  • Suitable host cells for such assays include yeast cells, but also mammalian cells or bacterial cells.
  • Two hybrid systems using hybrid protein fusions with other proteins than transcription factors, including enzymes such as beta-galactosidase or dihydrofolate reductase may also be applied. These assays are useful both to monitor the effect of a compound, including peptides, proteins or nucleic acids on an interaction of a cofactor with a given binding partner, as well as to identify novel proteins or nucleic acids interacting with the cofactor.
  • Recombinant forms of the polypeptides according to SEQ ID NO. 3, SEQ ID NO. 6, SEQ ID NO. 9, SEQ ID NO. 12, SEQ ID NO. 15, SEQ ID NO. 18, SEQ ID NO. 21, or SEQ ID NO. 24 can be used in cell-free screening assays aiming at the isolation of compounds affecting the activity of cofactors.
  • the cofactor polypeptides are brought into contact with a substance to test if the substance has an effect on the activity of the cofactor.
  • the detection of an interaction between an agent and a cofactor may be accomplished through techniques well-known in the art. These techniques include but are not limited to centrifugation, chromatography, electrophoresis and spectroscopy. The use of isotopically labeled reagents in conjunction with these techniques or alone is also contemplated. Commonly used radioactive isotopes include 3 H, 14 C, 22 Na, 32 P, 33 P, 35 S, 45 Ca, 60 Co, 125 I, and 131 I. Commonly used stable isotopes include 2 H, C, 15 N, 18 O.
  • an agent binds to any of the cofactors of the present invention
  • the binding may be detected by using radiolabeled agent or radiolabeled cofactor.
  • radiolabeled agent or radiolabeled cofactor is utilized, the agent-cofactor complex may be detected by liquid scintillation or by exposure to x-ray film or phosho-imaging devices.
  • One way to screen for substances affecting cofactor activity is to measure the effect of the substance on the binding affinity of the cofactor to other proteins or molecules, such as activators or repressors, DNA, RNA, other proteins, antibodies peptides or other substances, including chemical compounds known to affect receptor activity or to a nuclear receptor itself.
  • Assays measuring the binding of a protein to a ligand are well known in the art, such as ELISA assays, FRET assays, bandshift assays, plasmon-resonance based assays, scintilUation proximity assays, fluorescence polarization assays, alpha screen assays.
  • a mixture containing a cofactor polypeptide, effector and candidate substance is allowed to incubate.
  • the unbound effector is separable from any effector/cofactor complex so formed.
  • One then simply measures the amount of each e.g., versus a control to which no candidate substance has been added). This measurement may be made at various time points where velocity data is desired. From this, one determines the ability of the candidate substance to alter or modify the function of the cofactor.
  • TLC thin layer chromatographic methods
  • HPLC high-density lipoprotein
  • spectrophotometric gas chroma- tographic/mass spectrophotometric or NMR analyses.
  • Another method of separation is to immobilize one of the binding partners on a solid support, and to wash away any unbound material. It is contemplated that any such technique may be employed so long as it is capable of differentiating between the effector and complex, and may be used to determine enzymatic function such as by identifying or quantifying the substrate and product.
  • a screening assay in which candidate agent binding of cofactors is analysed can include a number of conditions. These conditions include but are not limited to pH, temperature, tonic- ity, the presence of relevant other proteins, and relevant modifications to the polypeptide such as glycosylation or lipidation. It is contemplated that the cofactors can be expressed and utilized in a prokaryotic or eukaryotic cell.
  • the host cell expressing the cofactors can be used whole or the cofactor can be isolated from the host cell.
  • the cofactor can be membrane bound in the membrane of the host cell or it can be free in the cytosol of the host cell.
  • the host cell can also be fractionated into sub-cellular fractions where the cofactor can be found. For example, cells expressing the cofactor can be fractionated into the nuclei, the endoplasmic reticulum, vesicles, or the membrane surfaces of the cell.
  • pH is preferably from about a value of 6.0 to a value of about 8.0, more preferably from about a value of about 6.8 to a value of about 7.8, and most preferably, about 7.4.
  • temperature is from about 20°C degrees to about 50°C degrees more preferably, from about 30°C degrees to about 40°C degrees and even more preferably about 37°C de- grees.
  • Osmolality is preferably from about 5 milliosmols per liter (mosm/L) to about 400 mosn ⁇ /1, and more preferably, from about 200 milliosmols per liter to about 400 mosm/1 and, even more preferably from about 290 mosm/L to about 310 mosm/L.
  • cofactors include sodium, potassium, calcium, magnesium, and chloride.
  • prosthetic groups small, non-peptide molecules, known as prosthetic groups may also be required.
  • Other biological conditions needed for cofactor function are well- known in the art.
  • proteins can be reconstituted in artificial membranes, vesicles or liposomes. (Danboldt et al.,1990).
  • the present invention contemplates that the cofactor can be incorporated into artificial membranes, vesicles or liposomes.
  • the reconstituted cofactor can be utilized in screening assays.
  • a cofactor of the present invention can be coupled to a solid support, e.g., to agarose beads, polyacrylamide beads, polyacrylic, sepharose beads or other solid matrices capable of being coupled to polypeptides.
  • a solid support e.g., to agarose beads, polyacrylamide beads, polyacrylic, sepharose beads or other solid matrices capable of being coupled to polypeptides.
  • Well-known coupling agents include cyanogen bromide (CNBr), carbonyldiimidazole, tosyl chloride, diaminopimelimidate, and glutar aldehyde.
  • a typical screening assay for identifying candidate substances one employs the same recombinant expression host as the starting source for obtaining the cofactor polypeptide, generally prepared in the form of a crude homogenate. Recombinant cells expressing the cofactor are washed and homogenized to prepare a crude polypeptide homogenate in a desirable buffer such as disclosed herein. In a typical assay, an amount of polypeptide from the cell homogenate, is placed into a small volume of an appropriate assay buffer at an appropriate pH.
  • Candidate substances such as agonists and antagonists, are added to the admixture in convenient concentrations and the interaction between the candidate substance and the cofactor polypeptide is monitored.
  • screening assays for the testing of candidate substances are designed to allow the determination of structure-activity relationships of agonists or antagonists with the cofactors, e.g., comparisons of binding between naturally-occurring hormones or other substances capable of interacting with or otherwise modulating the cofactor; or comparison of the activity caused by the binding of such molecules to the cofactor.
  • the polypeptides of the invention are crystallized in order to carry out x-ray crystallographic studies as a means of evaluating interactions with candidate substances or other molecules with the cofactor polypeptide.
  • the purified recombinant polypeptides of the invention i.e. of the cofactors according to the invention, when crystallized in a suitable form, are amenable to detection of intra-molecular interactions by x-ray crystallography.
  • the structure of the polypeptides can be determined using nuclear magnetic resonance.
  • This invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising an effective amount of an agonist or antagonist drug identified by the method described herein and a pharmaceutically acceptable carrier.
  • Such drugs and carrier can be administered by various routes, for example oral, subcutaneous, intramuscular, intravenous or intracerebral.
  • the preferred route of administration would be oral at daily doses of about 0.01 -100 mg/kg.
  • This invention provides a method of treating diseases such as cancer, cardiovascular diseases, bone diseases, hormonal dysfunctions and others by altering the activity of the cofactor thereby influencing the binding affinity of the cofactor to ER alpha.
  • the recombinant expression constructs of the present invention are useful in molecular biology to transform cells which do not ordinarily express the CF16 to CF19 and CF40 to CF43 polypeptides to express these cofactors upon transformation.
  • Such cells are useful as intermediates for making cellular preparations useful for cofactor binding assays, which are in turn useful for drug screening.
  • the recombinant expression constructs of the present invention are also useful in gene therapy.
  • Cloned genes of the present invention, or fragments thereof, may also be used in gene therapy carried out by homologous recombination or site-directed mutagenesis. See generally Thomas & Capecchi, Cell 51, 503-512 (1987); Bertling, Bioscience Reports 7, 107-112 (1987); Smithies et al., Nature 317, 230-234 (1985).
  • Oligonucleotides of the present invention are useful as diagnostic tools for probing cofactor gene expression in tissues.
  • tissues are probed in situ with oligonucleotide probes carrying detectable groups by conventional autoradiographic techniques, as explained in greater detail in the examples below, to investigate native expression of this cofactor or pathological conditions relating thereto.
  • chromosomes can be probed to investigate the presence or absence of the CF genes, and potential pathological conditions related thereto, as also illustrated by the Examples below.
  • Probes according to the invention should generally be at least about 15 nucleotides in length to prevent binding to random sequences, but, under the appropriate circumstances may be smaller.
  • Another aspect of the invention includes antibodies specifically reactive with the proteins or any parts of the proteins according to the invention and or polypeptides encoded by the nucleotide sequences of the cofactors.
  • the term tauantibody refers to intact molecules as well as fragments thereof, such as Fa, F(ab).sub.2, and Fv, which are capable of binding the epitopic determinant.
  • immunogens derived from the polypeptide according to the invention and/or encoded by the nucleic acids according to the invention anti-protein/anti-peptide an- tiserum or monoclonal antibodies can be made by standard protocols (E. Howell & D. Lane. Antibodies: A Laboratory Manual. Cold Spring Harbor Laboratory (1988)).
  • a polyclonal antibody is prepared by immunizing a mammal, such as a mouse, a hamster or rabbit with an immunogenic form of the cofactors according to the invention depending on which of these are desired) of the present invention, and collecting antisera from that immunized animal. Because of the relatively large blood volume of rabbits, a rabbit is a preferred choice for production of polyclonal antibodies.
  • fusion proteins As an immunizing antigen, fusion proteins, intact polypeptides or fragments containing small peptides of interest can be used. They can be derived by expression from a cDNA transfected in a host cell with subsequent recovering of the protein peptide or peptides can be synthesized chemically (e.g. oligopeptides with 10-15 residues in length).
  • Important tools for monitoring the function of the cofactor gene according to the present invention i.e. encoded by a sequence according to SEQ ID NO. 1, SEQ ID NO. 4, SEQ ID NO. 7, SEQ ID NO. 10, SEQ ID NO. 13, SEQ ID NO. 16, SEQ ID NO. 19, or SEQ ID NO. 22 are antibodies against various domains of the proteins according to the invention.
  • a given polypeptide or polynucleotide may vary in its immunogenicity. It is often necessary to couple the immunogen (e.g. the polypeptide) with a carrier. Commonly used carriers that are chemically coupled to peptides include bovine serum albumin (BSA) and keyhole limpet hemocyanin (KLH). The coupled peptide is then used to immunize the animal in the presence of an adjuvant, a non-specific stimulator of the immune response in order to enhance immunogenicity. The production of polyclonal antibodies is monitored by detection of antibody titers in plasma or serum at various time points following immunization. Standard ELISA or other immunoassays can be used with the immunogen as antigen to assess the levels of antibodies. When a desired level of immunogenicity is obtained, the immunized animal may be bled and the serum isolated, stored and purified.
  • BSA bovine serum albumin
  • KLH keyhole limpet hemocyanin
  • antibody-producing cells e.g. spleen cells
  • immunized animal preferably mouse or rat
  • immortalizing cells such as myeloma cells
  • myeloma cells e.g. spleen cells
  • myeloma cell e.g. the murine NS-1 myeloma cell.
  • Such techniques are well known in the art, and include, for example, the hybridoma technique (originally developed by Kohler & Milstein. Nature 256: 495-497 (1975)), the human B cell hybridoma technique (Kozbar et al.
  • the fused spleen/myeloma cells are cultured in a selective medium to select fused spleen/myeloma cells from the parental cells.
  • Fused cells are separated from the mixture of non-fused parental cells, for example, by the addition of agents that block the de novo synthesis of nucleotides in the tissue culture media. This culturing provides a population of hy- bridomas from which specific hybridomas are selected.
  • selection of hybridomas is performed by culturing the cells by single-clone dilution in microtiter plates, followed by testing the individual clonal supernatants for reactivity with an antigen-polypeptide. The selected clones may then be propagated indefinitely to provide the monoclonal antibody in convenient quantity.
  • antibodies which specifically bind the polypeptides according to the invention and/or encoded by the nucleotide sequences of the cofactors or their complements provides an important utility in immunolocalization studies, and may play an important role in the diagnosis and treatment of such diseases and disorders as metabolic disorders, immunological indications, hormonal dysfunctions and/or neurosystemic diseases.
  • the antibodies may be employed to identify tissues, organs, and cells which express the cofactors.
  • Antibodies can be used diagnostically in immuno-precipitation and immuno-blotting to detect and evaluate co- factor protein levels in tissue or from cells in bodily fluid as part of a clinical testing procedure.
  • Monoclonal antibodies provided by the present invention are also produced by recombinant genetic methods well known to those of skill in the art, and the present invention encompasses antibodies made by such methods that are immunologically reactive with an epitope of a mammalian cofactor protein or peptide according to the invention.
  • the present invention encompasses fragments of the antibody that are immunologically reactive with an epitope of a cofactor protein or peptide. Such fragments are produced by any number of methods, including but not limited to proteolytic cleavage, chemical synthesis or preparation of such fragments by means of genetic engineering technology.
  • the present in- vention also encompasses single-chain antibodies that are immunologically reactive with an epitope of a cofactor protein or peptide made by methods known to those of skill in the art.
  • the invention also includes chimeric antibodies, comprised of light chain and heavy chain peptides immunologically reactive to an epitope that is a cofactor protein or peptide according to the invention.
  • the chimeric antibodies embodied in the present invention include those that are derived from naturally occurring antibodies as well as chimeric antibodies made by means of genetic engineering technology well known to those of skill in the art as, for example, humanized antibodies.
  • the antibodies can also be chemically and/or enzymatically modified, for example carry a glycosylation and/or a label, like a fluorescent or radioactive label.
  • SEQ ID NO. 3 SEQ ID NO. 6, SEQ ID NO. 9, SEQ ID NO. 12, SEQ ID NO. 15, SEQ ID NO. 18, SEQ ID NO. 21, and/or SEQ ID NO. 24 which rely on the use of phage display systems and related systems, such as described in Hoogenboom HR, de Bruine AP, Hufton SE, Hoet RM, Arends JW, Roovers RC, Immunotechnology 1998 Jun;4(l):l-20, and references therein.
  • the present invention also encompasses one or more epitopes of a cofactor protein or peptide that is comprised of sequences and/or a conformation of sequences present in the cofactor proteins or peptide molecule.
  • epitopes may be naturally occurring, or may be the result of proteolytic cleavage of the cofactor proteins or peptides and isolation of an epitope- containing peptide or may be obtained by synthesis of an epitope-containing peptide using a method of genetic engineering technology and synthesized by genetically engineered prokaryotic or eukaryotic cells.
  • ANTISENSE OLIGONUCLEOTIDES AGAINST CF16, CF17, CF18, CF19, CF40, CF41, CF42 and CF43 GENE TRANSCRIPTS
  • Antisense oligonucleotides are short single stranded DNA or RNA molecules which may be used to block the availability of the cofactor messenger(s). Synthetic derivatives of ribonu- cleotides or desoxyribonucleotides and/or PNAs (see above) are equally possible. These are potential candidate agents which may interact with the cofactor according to the invention.
  • sequence of an antisense oligonucleotide is at least partially complementary to the sequence of the cofactor of interest.
  • the complementarity of the sequence is in any case high enough to enable the antisense oligonucleotide to bind to the nucleic acid according to the invention or parts thereof (SEQ ID NO1, SEQ ID NO. 4, SEQ ID NO. 7, SEQ ID NO. 10, SEQ ID NO. 13, SEQ ID NO. 16, SEQ ID NO.
  • Antisense oligonucleotides can be conjugated to different other molecules in order to deliver them to the cell or tissue expressing any of the cofactor genes.
  • the antisense oligonucleotide can be conjugated to a carrier protein (e.g. ferritin) in order to direct the oligonucleotide towards the desired target tissue, i.e. in case of ferritin predominantly to the liver.
  • a carrier protein e.g. ferritin
  • Antisense expression constructs are expression vector systems that allow the expression - either inducible or uninducible - of a complementary sequence to the CF16 to CF19 and CF40 to CF43 cofactor sequences according to the invention.
  • the potential possibility of such an approach has been demonstrated in many different model systems (von Ruden T, Gilboa E, Inhibition of human T-cell leukemia virus type I replication in primary human T cells that express antisense RNA, J Virol 1989 Feb;63(2):677-82; Nemir M, Bhattacharyya D, Li X, Singh K, Mukherjee AB, Mukherjee BB, Targeted inhibition of osteopontin expression in the mammary gland causes abnormal morphogenesis and lactation deficiency, J Biol Chem 2000 Jan 14;275(2):969-76; Ma L, Gauville C, Berthois Y, Millot G, Johnson GR, Calvo F Antisense expression for amphiregulin suppresses tumorigenicity of
  • an antisense expression construct can be constructed with virtually any expression vector capable of fulfilling at least the basic requirements known to those skilled in the art.
  • retroviral expression systems or tissue specific gene expression systems are preferred.
  • Current standard technologies for delivering antisense constructs are performed through a conjugation of constructs with liposomes and related, complex-forming compounds, which are transferred via electroporation techniques or via particle-mediated "gene gun” technologies into the cell. Other techniques may be envisioned by one skilled in the art.
  • Microinjection still plays a major role in most gene transfer techniques for the generation of germ-line mutants expressing foreign DNA (including antisense RNA constructs) and is preferred embodiment of the present invention.
  • Ribozymes are either RNA molecules (Gibson SA, Pellenz C, Hutchison RE, Davey FR, Shillitoe EJ, Induction of apoptosis in oral cancer cells by an anti-bcl-2 ribozyme delivered by an adenovirus vector, Clin Cancer Res 2000 Jan;6(l):213-22; Folini M, Colella G, Villa R, Lualdi S, Daidone MG, Zaffaroni N, Inhibition of Telomerase Activity by a Hammerhead Ribozyme Targeting the RNA Component of Telomerase in Human Melanoma Cells, J Invest Dermatol 2000 Feb;114(2):259-267; Halatsch ME, Schmidt U, Botefur IC, Holland JF, Oh- numa T, Marked inhibition of glioblastoma target cell tumorigenicity in vitro by retrovirus- mediated transfer of a hairpin ribozyme against deletion-mutant epidermal growth factor receptor messenger RNA,
  • the catalytic activity located in one part of the RNA (or DNA) molecule can be "targeted" to a specific sequence of interest by fusing the enzymatically active RNA molecule sequence with a short stretch of RNA (or DNA) sequence that is complementary to the cofactor gene transcript of interest.
  • a construct will, when introduced into a cell either physically or via gene transfer of a ribozyme expression construct find the corresponding cofactor sequence (our sequence of interest or also targeted in RNA) and bind via its sequence-specific part to said sequence.
  • the catalytic activity attached to the construct usually associated with a special nucleic acid structure (people distinguish so called “hammerhead” structures and "hairpin” structures), will then cleave the targeted RNA.
  • the targeted mRNA will be destroyed and cannot be translated efficiently, thus the protein encoded by the mRNA derived from cofactor will not be expressed or at least will be expressed at significantly reduced amounts.
  • the invention covers inducible ribozyme constructs (Koizumi M, Soukup GA, Kerr JN, Breaker RR, Allosteric selection of ribozymes that respond to the second messengers cGMP and cAMP, Nat Struct Biol 1999 Nov;6(l 1):1062-1071).
  • the invention concerns the use of "bivalent" ribozymes (multimers of catalytically active nucleic acids) as described in (Hamada M, Kuwabara T, Warashina M, Nakayama A, Taira K, Specificity of novel allosterically trans- and cis- activated connected maxizymes that are designed to suppress BCR-ABL expression FEBS Lett 1999 Nov 12;461(l-2):77-85).
  • non-human transgenic animals grown from germ cells transformed with a CF16, CF17, CF18, CF19, CF40, CF41, CF42 OR CF43 nucleic acid sequence according to the invention and that express the cofactor according to the invention and offspring and descendants thereof.
  • transgenic non-human mammals comprising a homologous recombination knockout of the native cofactors, as well as transgenic non-human mammals grown from germ cells transformed with nucleic acid antisense to the nucleic acids of the invention and offspring and descendants thereof.
  • non-human transgenic animals in which the native cofactor has been replaced with the human ortholog are also encompassed by the invention.
  • Transgenic animals according to the invention can be made using well known techniques with the nucleic acids disclosed herein.
  • Such transgenic animals are useful for screening for and determining the physiological effects of the cofactor agonists and antagonist.
  • transgenic animals are useful for developing drugs to regulate physiological activities in which the cofactors participate.
  • the amino acid sequences of the present invention can be used for structural drug design.
  • Aim is to produce structural analogs of biologically active polypeptides of interest or of small molecules with which they interact (e.g. agonists, antagonists or inhibitors) in order to design drugs which are, for example, more active or stable forms of the polypeptide, or which, for example, enhance or interfere with the function of a polypeptide in vivo.
  • one first determines the three-dimensional structure of a protein of interest, i.e. the cofactor, by computer-modeling, x-ray crystallography or a combination of both approaches.
  • Figure 1 shows yeast-two-hybrid interactions of CF16 tested against a set of nuclear receptors and cofactors (see Example 4).
  • a Gal4-DNA binding domain-CF16 fusion protein was tested for interactions against a panel of Gal4 activation domain fusion proteins as described in the text. Values for interactions are shown as fold activation over the CF16 interaction with the Gal4 activation domain only (protein 1-1 upper panel and protein 13-1 lower panel).
  • Gal4 activation domain fusion proteins tested are:
  • SEQ ID No 1 to 3 show CF16 (tremblnewIAK023173) sequences, in particular the cDNA Sequence (Seq ID No 1) the Reverse Complement of the cDNA Sequence (Seq ID No 2) and the amino acid sequence (Seq ID No 3),
  • SEQ ID No 4 to 6 show CF17 (nageneseqIZ41321) sequences, in particular the cDNA Sequence (Seq ID No 4) the Reverse Complement of the cDNA Sequence (Seq ID No 5) and the amino acid sequence (Seq ID No 6),
  • SEQ ID No 7 to 9 show CF18 (Nkx2.2(aageneseqiY25173)) sequences, in particular the cDNA Sequence (Seq ID No 7) the Reverse Complement of the cDNA Sequence (Seq ID No 8) and the amino acid sequence (Seq ID No 9),
  • SEQ ID No 10 to 12 show CF19 (CAM2(trembllAF 112472)) sequences, in particular the cDNA Sequence (Seq ID No 10) the Reverse Complement of the cDNA Sequence (Seq ID No 11) and the amino acid sequence (Seq ID No 12),
  • SEQ ID No 13 to 15 show CF40 sequences (NR26BP5, NM_014958), in particular the cDNA Sequence (Seq ID No 13) the Reverse Complement of the cDNA Sequence (Seq ID No 14) and the amino acid sequence (Seq ID No 15),
  • SEQ ID No 16 to 18 show CF41 sequences (NR26BP7; aageneseq
  • SEQ ID No 19 to 21 show CF42 sequences (NR51BP1, nageneseqlC76971lC76971 Human ORFX ORF2526 polynucleotide), in particular the cDNA Sequence (Seq ID No 19) the Reverse Complement of the cDNA Sequence (Seq ID No 20) and the amino acid sequence (Seq ID No 21),
  • SEQ ID No 22 to 24 show CF43 sequences (NR51BP2, embl
  • SEQ ID No 28 to 30 show ER alpha bait sequences, in particular the cDNA Sequence (Seq ID No 28) the Reverse Complement of the cDNA Sequence (Seq ID No 29) and the amino acid sequence of the ER alpha ligand binding domain bait (Seq ID No 30),
  • Site-specific DNA cleavage is performed by treatment with the suitable restriction enzyme (or enzymes) under conditions that are generally understood in the art, and the particulars of which are specified by the manufacturer of these commercially available restriction enzymes. See, e.g., New England Biolabs, Product Catalog. In general, about 1 ⁇ g of plasmid and/or DNA sequence is cleaved by one unit of enzyme in about 20 ⁇ l of buffer solution. Often excess of restriction enzyme is used to ensure complete digestion of the DNA substrate. Incubation times of about one hour to two hours at about 37°C are workable, although variations are tolerable.
  • Transformed host cells are cells which have been transformed or transfected with recombinant expression constructs made using recombinant DNA techniques and comprising cofactor encoding sequences.
  • Preferred host cells for transient transfection are COS-7 cells.
  • Transformed host cells may ordinarily express one of the CF16 to CF19 and CF40 to CF43 cofactors but host cells transformed for purposes of cloning or amplifying nucleic acid hybridisation probe DNA need not express the cofactors. When expressed, the cofactor proteins will typically be located in the host cell membrane.
  • Cultures of cells derived from multicellular organisms are desirable hosts for recombinant nuclear receptor protein synthesis.
  • any higher eukaryotic cell culture is workable, whether from vertebrate or invertebrate culture.
  • mammalian cells are preferred. Propagation of such cells in cell culture has become a routine procedure. See Tissue Culture (Academic Press, Kruse & Patterson, Eds., 1973).
  • useful host cell lines are bacteria cells, insect cells, yeast cells, human 293 cells, VERO and HeLa cells, LMTK- cells, and WI138, BHK, COS-7, CV, and MDCK cell lines. Human 293 cells are preferred.
  • a multiple tissue northern blot (Clontech, Palo Alto) is hybridized to a labeled probe.
  • the blot contains about 0.3 to 3 ⁇ g of poly A RNA derived from various tissues.
  • Hybridization may be carried out in a hybridization solution such as one containing SSC (see Maniatis et al, ibid) at an optimized temperature between 50°c and 70°C, preferably 65°C.
  • the filter may be washed and a film exposed for signal detection (see also: Maniatis et al., Molecular Cloning: A laboratory Manual, Cold Spring Harbor Laboratory Press, N.Y.(1989)).
  • RNA is obtained from various tissues and used to prepare cDNA expression libraries by using for example an Invittogen kit. (Invittogen Corporation, San Diego).
  • the chosen library may be screened under stringent condition (see definitions above) by using CF16, CF17, CF18, CF19, CF40, CF41, CF42 or CF43 specific probes.
  • the cDNA insert of positive clones is subsequently sequenced and cloned in a suitable expression vector.
  • full length cofactor clones from various species are obtained by using RACE PCR technology.
  • suitable cDNA libraries are constructed or purchased.
  • the first strand cDNA is used directly in RACE PCR reactions using a RACE cDNA amplification kit according to the manufactures protocol (Clontech, Palo Alto). Amplified fragments are purified, cloned and subsequently used for sequence analysis.
  • genomic libraries (Clontech, Palo Alto) are screened with a receptor specific probe under stringent conditions. Positive clones are isolated and the complete DNA sequence of the putative receptor is determined by sequence analysis (Maniatis et al., Molecular Cloning: A laboratory Manual, Cold Spring Harbor Laboratory Press, N.Y.(1989)).
  • EXAMPLE 4 ISOLATION OF THE COFACTOR PROTEINS BY USE OF THE YEAST TWO-HYBRID SYSTEM
  • a yeast two-hybrid assay was performed using methods such as described by Fields and Song Nature 340, ⁇ p245 (1989), Bartel et al, Biotechniques 14, pp920 (1993) and Lee et al. Nature 374 pp91-4 (1995).
  • a sequence encoding amino acids (aa) 249-595 of ER alpha (containing the ligand binding domain; LBD) or alternatively a sequence encoding the full length ER alpha protein (amino acids 1-595) was cloned into the vector pGBT9 (Clontech) in such way that, after transformation of the haploid yeast strain CGI 945 (Clontech), a hybrid protein is expressed consisting of the DNA-binding domain (BD) of the Gal4 transcription factor fused N-terminally to amino acids 249-595 of ER alpha (SEQ ID NO. 33) or to amino acids 1-595 of ER alpha (SEQ ID NO. 30), respectively.
  • BD DNA-binding domain
  • CGI 945 cells expressing the Gal4BD::ER al- pha(aa 249-595) or the Gal4BD::ER alpha (aal-595) fusion protein were mated to cells of strain Y187 (Clontech) containing a library of Gal4 transcription activation domain (AD) fusion plasmids with human cDNA generated from a range of tissues inserted into the vector pACT2 (Clontech). All libraries were purchased from Clontech Laboratories (MATCHMAKER human cDNA libraries) and included Cat.
  • HL4040AH (aorta), HL4041AH (chondrocytes), HY4004AH (brain), HY4035AH (testis), HY4024AH (liver), HY4042AH (heart), HY4053AH (bone marrow), HY4028AH (fetal brain), HY4043AH (kidney), HY4051AH (ovary), HY4047AH (skeletal muscle) and HY4000AA (hela).
  • the two- hybrid screens were essentially performed following the Clontech "Pretransformed Matchmaker Libraries User Manual” (PT3183-1): Transformed CGI 945 and Y187 cells were mated in order to coexpress the Gal4BD::ER alpha (aa249-595) fusion protein or the Gal4BD::ER alpha(aal-595) fusion protein, respectively, and the Gal4AD fusion proteins encoded on the library plasmids within one cell.
  • CF16, CF17, CF18, CF19, CF40, and CF41 Six novel cofactors (CF16, CF17, CF18, CF19, CF40, and CF41) interacting with the Gal4BD::ER alpha(aa249-595) fusion protein were isolated using this approach: CF16 was isolated from the aorta, bone marrow, testis, skeletal muscle and brain cDNA libraries, CF17 from the brain and testis libraries, CF18 and CF19 from the brain cDNA library, CF40 (NR26BP5) from the liver and kidney cDNA libraries and CF41 (NR26BP7) from the kidney cDNA library.
  • CF16 was isolated from the aorta, bone marrow, testis, skeletal muscle and brain cDNA libraries
  • CF17 from the brain and testis libraries
  • CF18 and CF19 from the brain cDNA library
  • CF40 NR26BP5
  • CF41 NR26BP7
  • CF42 (NR51BP1) was isolated from the heart and aorta libraries and CF43 (NR51BP2) was isolated from the brain and aorta libraries.
  • yeast two hybrid approach could be set up in a more directed manner, in such way that a DNA fragment encoding the full length CF16 protein is cloned into the vector pGBT9 so that a fusion protein is expressed in which CF16 is fused with its N-terminus to the Gal4 DNA binding domain.
  • the vector is then transformed into yeast strain CGI 945 (Clontech). The cells are grown at 30 °C to an optical density (OD 600 nm) of 1.0.
  • a number of protein encoding fragments or full length open reading frames are cloned into the vector pGAD424 (Clontech) in such way that proteins are expressed which are fused at their N-tennini to the Gal4 transcription activation domain.
  • the resulting plasmids are transformed into yeast strain Y187 (Clontech).
  • the Y187- transformants are grown as described above. After that 25 ⁇ l aliquots of the cultures become mixed in the wells of a 96 well microtiterplate with 25 ⁇ l aliquots of the CGI 945 transformants containing the CF16 DNA binding domain fusion protein.
  • the selective medium contains in addition 17 ⁇ -estradiol in 250 nM concentration.
  • the cells are incubated for exactly 72 hours at 30 °C in order to select for histidine prototroph clones i.e. expression of Gal4 activation domain fusion proteins interacting with CF16.
  • MELl reporter gene activation is tested via fluorimetric detection of 4-Methylumbelliferone (4-Mu) in the medium.
  • 4-Mu is one of the products resulting from cleavage of 4-Mu-X by alpha-galactosidase, the MELl gene product. 4-Mu emits fluorescent light of 465 nm wavelength, if excited with light of 360 nm wavelength. Thus the fluorescence units measured in each well give an indication on the relative strength of the interaction of a given Gal4 activation domain fusion protein with CF16. It turns out that amongst the proteins tested, the ligand binding domain of ERalpha interacted in a strictly estradiol dependent fashion with CF16 (Fig.1 ; compare interactions 2-6 in A and 2-6 in B).
  • estradiol a set of other chemical molecules known to influence the function of specific nuclear receptors was added to the medium: these included rifampicin (in 1 ⁇ M concentration), vitamin D3 (1 ⁇ M), all-trans retinoic acid (1 ⁇ M), 9-cis retinoic acid (1 ⁇ M), dexamethasone (0.5 ⁇ M), androstane (0.5 ⁇ M), linoleic acid (10 ⁇ M), aldosterone (0.5 ⁇ M), triiodothyronine (1 ⁇ M). No additional interactions could be de- tected as compared to the experiment done without the addition of ligand. This indicates the specificity of the ligand dependent interaction of CF16 with the estrogen receptor alpha.
  • EXAMPLE 5 DETECTION OF MUTANT ALLELES OF THE GENES ACCORDING TO THE INVENTION AND THEIR UTILISATION FOR DIAGNOSTIC PURPOSES.
  • alteration of the wild-type cofactor gene is detected.
  • the method can be performed by detecting the wild-type cofactor gene and confirming the lack of cause of the disease as a result of the locus.
  • “Alteration of the wild-type gene” encompasses all forms of mutations including deletions, insertions and point mutations in the coding and non-coding regions. Deletions may be of the entire gene or of only a portion. Point mutations may result in stop codons, frameshift mutations or amino acid substitutions. Somatic mutations are those which occur only in certain tissues and are not inherited in the germline. Germline mutations can be found in any of a body's tissue and are mostly inherited. Point mutational events may occur in regulatory regions, such as the promoter of the gene, leading to loss or dimunition of expression of the mRNA. Point mutations may also abolish proper RNA processing, leading to loss of expression of the cofactor gene product or to a decrease in mRNA stability or translation efficiency.
  • Applicable diagnostic techniques include, but are not limited to fluorescent in situ hybridization (FISH), direct DNA sequencing, PFGE analysis, Southern blot analysis, single stranded conformation analysis (SSCA), RNAse protection assay, allele-specific oligonucleotide (ASO), dot blot analysis, hybridization using nucleic acid modified with gold nanoparticles and PCR-SSCP, as discussed in detail further below.
  • FISH fluorescent in situ hybridization
  • direct DNA sequencing PFGE analysis
  • Southern blot analysis single stranded conformation analysis
  • SSCA single stranded conformation analysis
  • ASO allele-specific oligonucleotide
  • dot blot analysis hybridization using nucleic acid modified with gold nanoparticles and PCR-SSCP, as discussed in detail further below.
  • DNA microchip technology can be applied.
  • the presence of a disease due to a germline mutation of a cofactor can be ascertained by testing any tissue of the diseased human for mutations of the cofactor gene. For instance, a person who has inherited a germline mutation in the cofactor gene, especially one that will alter the interaction of the cofactor with the ER alpha protein, will be prone to develop a disease, such as cancer, bone diseases or defects in reproductive organs.
  • the presence of such a mutation can be determined by extracting DNA from any tissue of the body. For example, blood can be drawn and DNA extracted from blood cells and analyzed.
  • prenatal diagnosis of the disease will be possible by testing fetal cells, placental cells or amniotic cells for mutations in the cofactor gene.
  • Direct genomic DNA Sequencing can detect sequence variations of cofactor genes (Nucleic Acids Res 1997 May 15;25(10):2032-2034 Direct DNA sequence determination from total genomic DNA. Kilger C, Paabo S, Biol. Chem. 1997 Feb; 378(2):99-105, Direct exponential amplification and sequencing (DEXAS) of genomic DNA. Kilger C, Paabo S, DE 19653439.9 and DE 19653494.1). Another way is to make use of the single-stranded conformation polymorphism assay (SSCP; Orita et al., PNAS 86, 2166 (1989)). Variations in the DNA sequence of the cofactor gene from the wild-type sequence will be detected due to a shifted mobility of the corresponding DNA-fragments in SSCP gels.
  • SSCP single-stranded conformation polymorphism assay
  • SNPs single nucleotide polymorphisms
  • SNPs in coding or regulatory regions of genes which are thought to contribute to a disease physiology can have a direct impact on the phenotype, e.g. change a quantitative readout of disease physiology, for example the age of onset of heart attack.
  • Association and linkage studies with related individuals therefore provide an excellent means to test or verify a hypothesis on the functional impact of the gene of interest on disease physiology in vivo, in humans.
  • the ER alpha protein is known to control the expression of numerous estrogen responsive genes, which are implicated in the regulation of physiological and developmental processes such as sexual differentiation and behavior, fertility, cardiovascular function, brain function, bone generation and resorption as well as cell proliferation and carcinogenesis. Proteins interacting with ER alpha and/or the cofactors according to the invention are involved in the function of ER alpha. Therefore, alterations in the cofactors are useful for determining the genetic state of a person with respect to its capability to respond to estrogens.
  • DNA samples can be prepared from normal individuals and from persons being affected by the disease and these samples can be cut by one or more restriction enzymes and applied to Southern analysis. Southern blots displaying hybridizing fragments differing in length from the control DNA when probed with sequences near or including the cofactor locus could indicate a possible mutation. If large DNA fragments are used it is appropriate to separate these fragments by pulsed field gel electrophoresis (PFGE).
  • PFGE pulsed field gel electrophoresis
  • Detection of point mutations may be accomplished by amplification, for instance by PCR, from genomic or cDNA and sequencing of the amplified nucleic or by molecular cloning of the cofactor allele and sequencing the allele using techniques well known in the art.
  • restriction fragment length polymorphism (RFLP) probes for the gene or surrounding marker genes can be used to score for alteration of an allele or an insertion in a polymorphic fragment.
  • RFLP restriction fragment length polymorphism
  • the SSCP detects a band which migrates differently because the variation causes a difference in single strand, intra molecular base pairing.
  • the R ⁇ Ase protection assay involves cleavage of the mutant fragment into two or more smaller fragments. By using DGGE variations in the D ⁇ A can be detected by differences in the migration rates of mutant compared to normal al- leles in a denaturing gradient gel. In the mutS assay, the protein binds only to sequences that contain a nucleotide mismatch in a hetero duplex between mutant and wild-type sequences.
  • Mismatches are hybridised nucleic acid duplexes in which the two strands are not 100% complementary. Lack of total homology may be due to deletions, insertions, inversions or substitutions. Mismatch detection can be used to detect point mutations in the gene or the corresponding mR ⁇ A product. While these techniques are less sensitive than sequencing, they can preferably be used when a large number of samples shall be tested.
  • An example of a mismatch cleavage method is the R ⁇ Ase protection assay.
  • the method involves the use of a labeled ribonucleotide probe which is complementary to the wild-type sequence of the cofactor gene coding se- quence.
  • the riboprobe and either mR ⁇ A or D ⁇ A isolated from the person are hybridised together and subsequently digested with the enzyme R ⁇ ase A which is able to detect some mismatches in a duplex R ⁇ A structure. If a mismatch is detected by the enzyme, it cleaves at the site of the mismatch.
  • R ⁇ A product when the annealed R ⁇ A preparation is separated on an electrophoretic gel matrix, if a mismatch has been detected and cleaved by R ⁇ Ase A, an R ⁇ A product will be seen which is smaller than the full length duplex R ⁇ A for the riboprobe and the mR ⁇ A or D ⁇ A. If the riboprobe comprises only a fragment of the mR ⁇ A or the gene, it is advantageous to use a number of probes to screen the whole mRNA sequence for mismatches.
  • DNA probes can be used to detect mismatch mutations through enzymatic or chemical cleavage (Cotton et al, PNAS 85, 4397 (1988); Shenk et al, PNAS 72, 989 (1975); Novack et al., PNAS 83, 586 (1986)).
  • mismatches can be detected by shifts in the electrophoretic mobility of mismatched duplexes relative to match duplexes (Cariello, Human Genetics 42, 726 (1988)).
  • the cellular mRNA or DNA which might contain a mutation can be amplified using PCR (see below) before hybridisation.
  • Variations in DNA of the cofactor) gene can also be detected using Southern hybridisation, especially if the changes are major rearrangements, such as deletions or insertions.
  • DNA sequences of the cofactor gene which have been amplified by PCR may also be screened using allele specific probes. These probes are nucleic acid oligomers, each of which contains a region of the gene sequence harboring a known mutation. For instance, one oli- gomer could be about 25 nucleotides in length corresponding to a portion of the gene sequence. By using a number of such allele-specific probes, PCR amplification products can be screened to identify the presence of a previously discovered mutation in the gene.
  • Hybridisation of allele-specific probes with amplified cofactor sequences can be performed, for example, on a nylon filter. Under high stringency hybridisation conditions, the hybridisation of a particular probe should indicate the presence of the same mutation in the tissue as in the allele-specific probe.
  • the newly developed technique of nucleic acid analysis via microchip technology is also applicable to the present invention.
  • this technique thousands of distinct nucleotide probes are built up in an array on a silicon chip.
  • Nucleic acid to be analysed is fluorescently labeled and hybridised to the probes on the chip. It is also possible to study nucleic acid-protein interactions using these nucleic acid microchips.
  • This technique one can determine the presence of mutations or even sequence the nucleic acid being analysed or one can measure expression of a gene of interest.
  • This method is one of parallel processing of thousands of probes at once and can tremendously accelerate the analysis.
  • the most definite test for mutations in a candidate locus is to directly compare genomic co- factor sequences from patients with those from normal individuals. Alternatively one could sequence mRNA after amplification (for example by PCR) thereby eliminating the necessity of determining the exon structure of the respective gene.
  • Mutations from patients falling outside the coding region of the cofactor gene can be detected by examining the noncoding regions, such as introns and regulatory sequences within or near the genes. Early indications of mutations in noncoding regions could be for example the abundance or abnormal size of mRNA products in patients as compared to control individuals as detected by northern blot analysis.
  • Alteration of cofactor expression can be detected by any technique known in the art. These include northern blot analysis, PCR amplification and RNAse protection. Diminished mRNA expression indicates an alteration in the wild-type gene sequence. Alterations of wild-type genes can also be detected by screening for alteration of cofactor protein. For example, monoclonal antibodies against cofactor protein can be used to screen a tissue. Lack of cognate antigen would indicate a mutation. Antibodies specific for products of mutant alleles could also be used to detect mutant gene product. These kind of immunological assays could be done in any convenient format known in the art. These include western blots, immunohistochemical assays and ELISA assays.
  • Any means for detecting an altered cofactor protein can be used to detect alteration of the wild-type cofactor gene.
  • Functional assays such as protein binding determinations can be used.
  • assays can be used which detect the cofactors' biochemical function. Finding a mutant cofactor gene product indicates an alteration of the co- factor wild-type gene.
  • One such binding assay tests the binding of cofactor protein with wild- type ER alpha protein.
  • wild-type ER alpha protein or the domain interacting with the cofactor protein can be used in a protein binding assay or biochemical function assay to detect normal or mutant proteins.
  • a mutant cofactor gene or gene product or a mutant ER alpha protein can also be detected in other human body samples, such as serum, stool, urine and sputum.
  • Other human body samples such as serum, stool, urine and sputum.
  • the same techniques discussed above for detection of mutant genes or gene products in tissues can be applied to other body samples. By screening such body samples, a simple early diagnosis can be achieved for the disease) resulting from a mutation in the cofactor gene.
  • EXAMPLE 6 A CELL BASED ASSAY FOR MEASURING THE BINDING OF THE COFACTOR CF16, CF17, CF18, CF19, CF40, CF41, CF42 OR CF43, RESPECTIVELY TO ER ALPHA.
  • the DNA sequence encoding the open reading frame of the respective cofactor is transferred into the vector pVP16 (Clontech) to allow the expression of a fusion protein of the cofactor with the strong transactivation domain of the VP16 protein (of herpes simplex virus) in mammalian cells under the control of the strong CMV promoter.
  • the reporter the luciferase gene is cloned under the control of a minimal promoter containing a ER alpha-responsive DNA element.
  • This vector also expresses a second enzyme, e.g. beta- galactosidase, under the control of a constitutive promoter, to allow normalisation for transfection efficiency between experiments.
  • a third vector contains the ER alpha gene under the control of the strong CMV promoter.
  • CV-1 cells are then transiently transfected with different combinations of the three plasmids.
  • Transfection is done by standard methods, e.g. by use of the CalPhos Maximizer (Clontech, #8021-1,-2).
  • CalPhos Maximizer Clontech, #8021-1,-2
  • Interaction of the cofactor protein with ER alpha will lead to a strong transactivation due to the attached VP16 domain of the cofactor fusion protein.
  • interaction of the cofactor with ER alpha will result in increased luciferase activity.
  • Inclusion of the cofactor VP16 will result in increased luciferase activity as compared to transfection of the ER alpha and the reporter alone.
  • extracts are prepared of the transfected cells 48 to 72 hours after transfection, and luciferase activity is determined. To normalise for transfection efficiency, beta-galactosidase activity is also determined.
  • ER alpha Addition of substances known or suspected to influence the binding of ER alpha to the co- factor are added to the medium of the transfected cells. These substances are added at different timepoints prior to cell lysis, typically ranging between 18 hours to a five minutes before cell lysis. Luciferase activity is taken as a measure of the effect of these substances on the binding of the cofactor to ER alpha. To avoid activation of ER alpha by substances contained in the serum of the medium, charcoal stripped serum has to be used for these experiments. In an alternative setting of the experiment, the DNA-binding domain of ER alpha is replaced with the DNA-binding domain of the yeast GAL4 transcription factor.
  • the luciferase is expressed under the control of GAL4-responsive upstream activating sequences.
  • Expression of luciferase again is an indication for binding of the cofactor CF16- VP16, CF17-VP16, CF18-VP16, CF19-VP16, CF40-VP16, CF41-VP16, CF42-VP16 or CF43-VP16, fusion respectively to the GAL4-ER alpha fusion.
  • This setting is also referred to as the mammalian two hybrid system. A description of the experiment is found in the manual to the mammalian MATCHMAKER Two-Hybrid Assay Kit from Clontech, # PT3002-1, catalogue #K1602-1)
  • Substances activating nuclear receptors cause an exchange of the proteins bound to the receptors, thus effecting the dissociation of some proteins and promoting the binding of other proteins.
  • ER alpha-activating compounds and ER alpha-inactivating compounds by monitoring the binding of the respective CF cofactor to ER alpha.
  • stably transfected cell lines which contain copies of the two different expression constructs for ER alpha and the respective CF cofactor as well as the reporter construct stably integrated into the chromosomes of the cells.
  • DNA sequences encoding the open reading frame of the cofactor and the ER alpha gene are each transferred separately into the vector pENTRY (Life Technologies) to allow efficient construction of a diverse set of expression constructs.
  • the open reading frame is then recom- bined into the vector pDEST17 for expression in E. coli strain BL21 as a fusion protein to a six-histidine tag induced by IPTG, as well as into the pDEST15 for expression as a fusion protein with glutathione S-transferase (GST).
  • the plasmids pDEST15, pDEST17and pENTRY are purchased from LIFE TECHNOLOGIES.
  • the open reading frame is introduced into the vector pLV-CBDgw for expression as a fusion protein with the calmodu- lin binding protein using recombinant baculoviruses as specified by the manufacturer (Becton Dickinson).
  • pLV-CBDgw is a derivative of the vector pLV1392 (Becton Dickinson) wliich is modified by the insertion of a calmodulin binding protein fragment, followed by the sequence required for recombinational cloning via the Gateway system (Life Technologies). Protein expression is induced and recombinant protein is purified by passage over a Ni-NTA-column, or a glutathione column or a calmodulin column, respectively.
  • a biotinylated (Biotintag Micro biotinylation Kit, Sigma) His-tagged ER alpha protein and the GST fusion of the cofactor are mixed at 0.2- 5 ⁇ M.
  • Antibody to the GST protein is added which is labelled by the europium chelate at a concentration of 1-3 (typical 2.5) nM.
  • Streptavidin which is fluorescently labeled by covalent attachment of allophycocyanin is added at a concentration of 5-30 ⁇ g/ml (typical lO ⁇ g/ml).
  • the europium chelate is stimulated by a flash of light (320nm) and, the emitted light is measured in a delayed (50-200 ⁇ s) time window for 300 to 450 ⁇ s after the flash at 615 nm (fluorescence of europium chelate) and 655nm (fluorescence of APC). Since APC is only excited by the light emitted by the europium chelate, a close proximity of the two different fluoropho- res is required for excitation. The strength of the APC signal, as well as the ratio of the signals from the two fluorophores (i.e. the ratio of the intensities of light emitted at 655 and 615nm) serves as a measure for the interaction of the two proteins. Reaction buffers contain 20mM TrisHCl pH 7.9, 60mM KC1, 4mM MgCl 2 . Reaction volume is 25 ⁇ l. The Wallac VictorV fluorimeter is used for the fluorimetric measurements.
  • the cofactor is used as a biotinylated His-tagged protein, and the ER alpha protein is used as fusion to GST.
  • the His-tagged proteins are replaced by the same proteins fused to the calmodulin binding protein.
  • the detection of the interaction is via biotinylated calmodulin, which is in turn binding to APC- coupled streptavidin.
  • Calcium has to be included in the buffer in the form of 4mM CaCl , to allow complex formation between calmodulin and the calmodulin binding protein.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Zoology (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Toxicology (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Abstract

L'invention concerne de nouveaux cofacteurs du récepteur alpha des oestrogènes désignés par CF16, CF17, CF18, CF19, CF40, CF41, CF42 et CF43 et en particulier les séquences d'acide nucléique isolées codant ces cofacteurs et les polypeptides isolés de ceux-ci. L'invention concerne également des procédés d'isolation et/ou de production d'acides nucléiques ou de protéines ainsi que des procédés d'utilisation de ces cofacteurs, comme l'inhibition ou l'activation de la liaison de ces cofacteurs du récepteur alpha des oestrogènes.
PCT/EP2002/002189 2001-03-01 2002-02-28 Nouveaux cofacteurs du recepteur alpha des oestrogenes et procedes d'utilisation WO2002070699A2 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002249247A AU2002249247A1 (en) 2001-03-01 2002-02-28 Cofactors of the estrogen receptor alpha and methods of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP01105062 2001-03-01
EP01105062.2 2001-03-01

Publications (2)

Publication Number Publication Date
WO2002070699A2 true WO2002070699A2 (fr) 2002-09-12
WO2002070699A3 WO2002070699A3 (fr) 2003-11-13

Family

ID=8176641

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2002/002189 WO2002070699A2 (fr) 2001-03-01 2002-02-28 Nouveaux cofacteurs du recepteur alpha des oestrogenes et procedes d'utilisation

Country Status (2)

Country Link
AU (1) AU2002249247A1 (fr)
WO (1) WO2002070699A2 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1383873A2 (fr) * 2001-03-30 2004-01-28 PE Corporation (NY) Proteines kinase humaines isolees, molecules d'acide nucleique codant les proteines kinase humaines et utilisation correspondante
CN110672763A (zh) * 2019-09-10 2020-01-10 山东省分析测试中心 一种多孔亚胺链共价有机骨架材料在雌激素检测中的应用

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999050664A1 (fr) * 1998-04-01 1999-10-07 Glaxo Group Limited Compositions et methodes de detection des interactions dependant de ligands entre un recepteur nucleaire et un co-activateur

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999050664A1 (fr) * 1998-04-01 1999-10-07 Glaxo Group Limited Compositions et methodes de detection des interactions dependant de ligands entre un recepteur nucleaire et un co-activateur

Non-Patent Citations (9)

* Cited by examiner, † Cited by third party
Title
BARKHEM T ET AL: "DIFFERENTIAL RESPONSE OF ESTROGEN RECEPTOR ALPHA AND ESTROGEN RECEPTOR BETA TO PARTIAL ESTROGEN AGONISTS/ANTAGONISTS" MOLECULAR PHARMACOLOGY, BALTIMORE, MD, US, vol. 54, no. 1, July 1998 (1998-07), pages 105-112, XP000978331 ISSN: 0026-895X cited in the application *
DATABASE EMBL [Online] Database accession no. AK023173 XP002234411 *
DUTERTRE MARTIN ET AL: "Molecular mechanisms of selective estrogen receptor modulator (SERM) action." JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS, vol. 295, no. 2, November 2000 (2000-11), pages 431-437, XP002234407 ISSN: 0022-3565 *
KATZENELLENBOGEN B S ET AL: "ESTROGEN RECEPTOR TRANSCRIPTION AND TRANSACTIVATION ESTROGEN RECEPTOR ALPHA AND ESTROGEN RECEPTOR BETA: REGULATION BY SELECTIVE ESTROGEN RECEPTOR MODULATORS AND IMPORTANCE IN BREAST CANCER" BREAST CANCER RESEARCH, CURRENT SCIENCE, LONDON, GB, vol. 2, no. 5, 2000, pages 335-344, XP001148108 ISSN: 1465-5411 *
KATZENELLENBOGEN BENITA S ET AL: "Molecular mechanisms of estrogen action: Selective ligands and receptor pharmacology." JOURNAL OF STEROID BIOCHEMISTRY AND MOLECULAR BIOLOGY, vol. 74, no. 5, 2000, pages 279-285, XP002234410 ISSN: 0960-0760 *
LANZ R B ET AL: "A STEROID RECEPTOR COACTIVATOR, SRA, FUNCTIONS AS AN RNA AND IS PRESENT IN AN SRC-1 COMPLEX" CELL, CELL PRESS, CAMBRIDGE, NA, US, vol. 97, 2 April 1999 (1999-04-02), pages 16-27, XP002926500 ISSN: 0092-8674 *
MCKENNA NEIL J ET AL: "Nuclear receptor coregulators: Cellular and molecular biology." ENDOCRINE REVIEWS, vol. 20, no. 3, June 1999 (1999-06), pages 321-344, XP002234409 ISSN: 0163-769X *
PAVAO M ET AL: "Estrogen receptor antibodies: specificity and utility in detection, localization and analyses of estrogen receptor alpha and beta" STEROIDS, BUTTERWORTH-HEINEMANN, STONEHAM, MA, US, vol. 66, no. 1, 1 January 2001 (2001-01-01), pages 1-16, XP004220761 ISSN: 0039-128X *
SHANG YONGFENG ET AL: "Cofactor dynamics and sufficiency in estrogen receptor-regulated transcription." CELL, vol. 103, no. 6, 8 December 2000 (2000-12-08), pages 843-852, XP002234408 ISSN: 0092-8674 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP1383873A2 (fr) * 2001-03-30 2004-01-28 PE Corporation (NY) Proteines kinase humaines isolees, molecules d'acide nucleique codant les proteines kinase humaines et utilisation correspondante
EP1383873A4 (fr) * 2001-03-30 2005-05-18 Applera Corp Proteines kinase humaines isolees, molecules d'acide nucleique codant les proteines kinase humaines et utilisation correspondante
CN110672763A (zh) * 2019-09-10 2020-01-10 山东省分析测试中心 一种多孔亚胺链共价有机骨架材料在雌激素检测中的应用

Also Published As

Publication number Publication date
WO2002070699A3 (fr) 2003-11-13
AU2002249247A1 (en) 2002-09-19

Similar Documents

Publication Publication Date Title
US5639616A (en) Isolated nucleic acid encoding a ubiquitous nuclear receptor
US20090023645A1 (en) AIB1, a novel steroid receptor co-activator
JP2012065671A (ja) Macacafascicularis由来のP−糖タンパク質およびその使用
US20020123095A1 (en) Estrogen receptor alpha variants and methods of detection thereof
WO2002077229A2 (fr) Nouveaux cofacteurs du recepteur alpha x du foie et techniques d'utilisation
US6353091B1 (en) Human N-type calcium channel isoform
WO2002070699A2 (fr) Nouveaux cofacteurs du recepteur alpha des oestrogenes et procedes d'utilisation
WO2002018420A2 (fr) Nouveaux cofacteurs du recepteur du pregnane x et leurs methodes d'utilisation
WO2002064783A2 (fr) Nouveaux cofacteurs du recepteur beta d'oestrogenes et procedes d'utilisation
WO2002053585A2 (fr) Cofacteur cf11 de recepteur nucleaire mammifere et ses methodes d'utilisation
WO2002053591A1 (fr) Cofacteur de recepteur nucleaire mammalien cf12 et ses procedes d'utilisation
WO2002042322A2 (fr) Cofacteurs de recepteurs nucleaires mammaliens cf7 et cf8 et procedes d'utilisation
JP2004512816A (ja) エストロゲン受容体β変異体及びその検出方法
WO2002044365A1 (fr) Cofacteur cf9 d'un recepteur nucleaire de mammifere et methodes d'utilisation correspondantes
WO2001062969A2 (fr) Variants de recepteurs oestrogeniques alpha et procede de detection desdits variants
WO2002024728A2 (fr) Nouveau cofacteur mammifere cf6 du recepteur nucleaire et ses methodes d'utilisation
EP1371662A1 (fr) Variant nouveau du recepteur LXR-alpha cellulaire
US6746867B1 (en) Mammalian mesoderm induction early response (MIER) gene family
EP1371661A1 (fr) Variants du récepteur CAR au niveau de son domaine de liaison du ligand
WO2002070697A1 (fr) Recepteur nucleaire l67 de mammifere et ses procedes d'utilisation
EP1328661A2 (fr) Variants alpha du recepteur d'oestrogene et techniques de detection de ces variants
WO2003087140A1 (fr) Fxrb de non-primate comme recepteur hormonal nucleaire sensible au lanosterol et utilisations associees
WO1999019476A2 (fr) Famille de genes non mammiferes a reaction precoce immediate a l'induction mesodermique (nm-mier)

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TN TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP