WO2003002741A2 - Nucleic acids encoding nuclear receptors - Google Patents

Nucleic acids encoding nuclear receptors Download PDF

Info

Publication number
WO2003002741A2
WO2003002741A2 PCT/IB2002/002356 IB0202356W WO03002741A2 WO 2003002741 A2 WO2003002741 A2 WO 2003002741A2 IB 0202356 W IB0202356 W IB 0202356W WO 03002741 A2 WO03002741 A2 WO 03002741A2
Authority
WO
WIPO (PCT)
Prior art keywords
gene
nucleic acid
polypeptide
agent
expression
Prior art date
Application number
PCT/IB2002/002356
Other languages
French (fr)
Other versions
WO2003002741A8 (en
WO2003002741A3 (en
Inventor
Roger A. Moraga Martinez
Gunnar Thor Sigurdsson
Original Assignee
Decode Genetics Ehf.
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 Decode Genetics Ehf. filed Critical Decode Genetics Ehf.
Priority to AU2002304350A priority Critical patent/AU2002304350A1/en
Publication of WO2003002741A2 publication Critical patent/WO2003002741A2/en
Publication of WO2003002741A3 publication Critical patent/WO2003002741A3/en
Publication of WO2003002741A8 publication Critical patent/WO2003002741A8/en

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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70567Nuclear receptors, e.g. retinoic acid receptor [RAR], RXR, nuclear orphan receptors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • NRs Nuclear receptors
  • NRs are a superfamily of ligand-activated transcription factors that mediate the transmission of extracellular signals into the cell to produce a cellular response. Unlike integral membrane receptors and membrane associated receptors, the nuclear receptors reside in either the cytoplasm or nucleus of eukaryotic cells. NRs recognize similar DNA sequences that contain two or more nucleotide DNA-binding half-sites arranged as direct repeats or inverted repeats. The receptors bind to specific DNA sequences and control gene transcription in the nucleus. Many nuclear receptors undergo a major conformational change upon binding a specific ligand. They regulate the assembly of a transcriptional preinitiation complex at the promoter of target genes and modulate their expression in response to ligand. In particular, NRs repress or stimulate transcription by recruiting corepressor or coactivatior proteins, in addition to directly contacting the basal transcription machinery.
  • NRs are involved in an enormous range of biological processes, and have been found to play roles in eontrolling development, differentiation and physiological function and homeostasis.
  • the NR superfamily includes receptors for steriod hormones, such as estrogen, androgen, progesterone, hepatocyte nuclear factor 4, thyroid hormone, hormonal forms of viamin A and D, retinoids, peroxisomal activators, mineralocoricoids, glucocorticoids, ecdysone and many others including orphan receptors whose ligand have not been identified.
  • the present invention relates to nuclear receptor genes, particularly nucleic acids comprising NR genes, and the amino acids encoded by such nucleic acids.
  • nucleic acids comprising NR genes
  • amino acids encoded by such nucleic acids One such sequence is shown in Table I.
  • Table I the NR entry lists the name (e.g., "MOOSE04608”), the University of California at Santa Cruz contig designation from which the sequence was analyzed (e.g., "ctgl7328”), the exon locations (e.g., "3473973.3474369, 3497170 . . .”), followed by the amino acid sequence and the nucleic acid sequence.
  • Sub-family information on the sequence is shown in Table ⁇ .
  • the sequence the following information is provided: the University of California at Santa Cruz contig designation from which the sequence was analyzed (e.g., "ctgl7328"), the name (e.g., "MOOSE04608”), and the subfamily to which the sequence appears to belong. The assignments were made on the basis of the best E- value with which the sequence aligned.
  • the isolated nucleic acid molecule comprises a nucleotide sequence of SEQ ID NO: 1, as shown in Table I, and the complements thereof.
  • the invention further relates to a nucleic acid molecule which hybridizes under high stringency conditions to a nucleotide sequence of SEQ ID NO: 1, as shown in Table I, and the complements thereof.
  • the invention additionally relates to isolated nucleic acid molecules (e.g., cDNA molecules) encoding an NR polypeptide (e.g., encoding a polypeptide of SEQ ID NO: 2, as shown in Table I).
  • the invention further provides a method for assaying a sample for the presence of a nucleic acid molecule comprising all or a portion of an NR in a sample, comprising contacting said sample with a second nucleic acid molecule comprising a nucleotide sequence encoding an NR polypeptide (e.g., one of SEQ ID NO: 1, as shown in Table I, or the complement of one of SEQ ID NO: 1; a nucleotide sequence encoding SEQ ID NO: 2, as shown in Table I), or a fragment or derivative thereof, under conditions appropriate for selective hybridization.
  • the invention additionally provides a method for assaying a sample for the level of expression of an NR polypeptide, or fragment or derivative thereof, comprising detecting (directly or indirectly) the level of expression of the NR polypeptide, fragment or derivative thereof.
  • the invention also relates to a vector comprising an isolated nucleic acid molecule of the invention operatively linked to a regulatory sequence, as well as to a recombinant host cell comprising the vector.
  • the invention also provides a method for preparing a polypeptide encoded by an isolated nucleic acid molecule described herein (an NR polypeptide), comprising culturing a recombinant host cell of the invention under conditions suitable for expression of said nucleic acid molecule.
  • the invention further provides an isolated polypeptide encoded by isolated nucleic acid molecules of the invention (e.g., NR polypeptide), as well as fragments or derivatives thereof.
  • the polypeptide comprises an amino acid sequence of SEQ ID NO: 2, as shown in Table I.
  • the invention also relates to an isolated polypeptide comprising an amino acid sequence which is greater than about 90 percent identical to an amino acid sequence of SEQ ID NO: 2, preferably about 95, 96, 97, 98 or 99 percent identical.
  • the invention also relates to an antibody, or an antigen-binding fragment thereof, which selectively binds to a polypeptide of the invention, as well as to a method for assaying the presence of a polypeptide encoded by an isolated nucleic acid molecule of the invention in a sample, comprising contacting said sample with an antibody which specifically binds to the encoded polypeptide.
  • the invention further relates to methods of diagnosing a predisposition to a condition mediated by NRs.
  • the methods of diagnosing such a predisposition in an individual include detecting the presence of a mutation in NR, as well as detecting alterations in expression of an NR polypeptide, such as the presence of different splicing variants of NR polypeptides.
  • the alterations in expression can be quantitative, qualitative, or both quantitative and qualitative.
  • the invention additionally relates to an assay for identifying agents which alter (e.g., enhance or inhibit) the activity or expression of one or more NR polypeptides.
  • a cell, cellular fraction, or solution containing an NR polypeptide or a fragment or derivative thereof can be contacted with an agent to be tested, and the level of NR polypeptide expression or activity can be assessed.
  • the activity or expression of more than one NR polypeptides can be assessed concurrently (e.g., the cell, cellular fraction, or solution can contain more than one type of NR polypeptide, such as different splicing variants, and the levels of the different polypeptides or splicing variants can be assessed).
  • the invention relates to assays to identify polypeptides which interact with one or more NR polypeptides.
  • a first vector is used which includes a nucleic acid encoding a DNA binding domain and also an NR polypeptide, splicing variant, or fragment or derivative thereof
  • a second vector is used which includes a nucleic acid encoding a transcription activation domain and also a nucleic acid encoding a polypeptide which potentially may interact with the NR polypeptide, splicing variant, or fragment or derivative thereof (e.g., an NR polypeptide binding agent or receptor).
  • Incubation of yeast containing both the first vector and the second vector under appropriate conditions allows identification of polypeptides which interact with the NR polypeptide or fragment or derivative thereof, and thus can be agents which alter the activity of expression of an NR polypeptide.
  • Agents that enhance or inhibit NR polypeptide expression or activity are also included in the current invention, as are methods of altering (enhancing or inhibiting) NR polypeptide expression or activity by contacting a cell containing NR and/or polypeptide, or by contacting the NR polypeptide, with an agent that enhances or inhibits expression or activity of NR or polypeptide.
  • the invention pertains to pharmaceutical compositions comprising the nucleic acids of the invention, the polypeptides of the invention, and or the agents that alter activity of NR polypeptide.
  • the invention further pertains to methods of treating conditions mediated by NRs, by administering NR therapeutic agents, such as nucleic acids of the invention, polypeptides of the invention, the agents that alter activity of NR polypeptide, or compositions comprising the nucleic acids, polypeptides, and/or the agents that alter activity of NR polypeptide.
  • NR therapeutic agents such as nucleic acids of the invention, polypeptides of the invention, the agents that alter activity of NR polypeptide, or compositions comprising the nucleic acids, polypeptides, and/or the agents that alter activity of NR polypeptide.
  • the present invention relates to nucleic acids comprising nuclear receptors ("NRs"), and the NR amino acids encoded by those nucleic acids.
  • NRs nuclear receptors
  • the NR superfamily includes the receptors for many important signaling pathways, including, but not limited to, hormone receptors, steroid hormone receptors, estrogen receptors, angrogen receptors, progesterone receptors, growth factors receptors, orphan nuclear retinoid receptors, pregnane X receptors, and others.
  • These receptors are involved in the treatment of infections and various diseases and conditions, including, but not limited to, cancers, primary biliary cirrhosis, inflammatory bowel disease, type 1 diabetes, malignant melanoma, rheumatoid arthritis, neuropsychiatric disorders, abnormal bone resorption, cardiovascular disease, central nervous system disorders, glaucoma, ocular hypertension, fungal infections, as well as others.
  • cancers primary biliary cirrhosis, inflammatory bowel disease, type 1 diabetes, malignant melanoma, rheumatoid arthritis, neuropsychiatric disorders, abnormal bone resorption, cardiovascular disease, central nervous system disorders, glaucoma, ocular hypertension, fungal infections, as well as others.
  • ESTs expressed sequence tags
  • HMMs Hidden Markov Models
  • DiAlign 2 works based on segment-to-segment comparisons instead of arbitrary thresholds for gap opening and extension, which makes it ideally suited for building models that represent an entire, full-length sequence, since the alignments built this way have more match states that would be assigned as insertion states when using other alignment algorithms.
  • the models were built using the standard HMMer package. To search for new genes, a genome-wide scan was done on the University of the University of the University of
  • the cross-validation of the results was done in two steps. First, all of the hits with an E- value lower than 10 " that do not overlap with one another were selected, and in the event of overlapping, the one with lowest E- value was selected. After selecting all of those matches, the DNA sequences were compared against the RefSeq database (Pruitt et al, Trends Genet. 16(l):44-47, 2000) using BLAST (Altschul et al. Nucleic Acids Res. 25:3389-3402, 1997).
  • the diseases were linked to the HMM genes in the following manner: (1) the HMM gene models were compared to the consensus of the human genome sequence, located and the results kept in a relational database; (2) all possible markers (Sequence Tagged Sites (STS's)) (public or deCODE genetics) are also located in the same consensus using ePCR or BLAT and results kept in a relational database; and (3) LOD scores for diseases are linked to markers. A span of one LOD drop around the marker was also given. A computer program takes each LOD peak and links it to the consensus through the markerhit in the database. The database is then queried for all HMM genes within the span of one LOD drop or a mimmum of 15 Mb in each direction from the marker. The output is the name of the peak marker and its distance to the HMM gene.
  • STS's Sequence Tagged Sites
  • NR refers to an isolated nucleic acid molecule shown in Table I, and consisting of SEQ ID NO: 1, and also to a portion or fragment of the isolated nucleic acid molecule (e.g., cDNA or the gene) that encodes NR polypeptide (e.g. , a polypeptide selected from the group shown in Table I, and consisting of SEQ ID NO: 2).
  • the isolated nucleic acid molecule comprises a nucleic acid molecule of SEQ ID NO: 1 or the complement of such a nucleic acid molecule.
  • the isolated nucleic acid molecules of the present invention can be RNA, for example, mRNA, or DNA, such as cDNA and genomic DNA.
  • DNA molecules can be double-stranded or single-stranded; single stranded RNA or DNA can be either the coding, or sense, strand or the non-coding, or antisense, strand.
  • the nucleic acid molecule can include all or a portion of the coding sequence of the gene and can further comprise additional non-coding sequences such as introns and non-coding 3' and 5' sequences (including regulatory sequences, for example).
  • the nucleic acid molecule can be fused to a marker sequence, for example, a sequence that encodes a polypeptide to assist in isolation or purification of the polypeptide.
  • Such sequences include, but are not limited to, those which encode a glutathione-S-transferase (GST) fusion protein and those which encode a hemagglutinin A (HA) polypeptide marker from influenza.
  • an "isolated" nucleic acid molecule is one that is separated from nucleic acids which normally flank the gene or nucleotide sequence (as in genomic sequences) and/or has been completely or partially purified from other transcribed sequences (e.g., as in an RNA library).
  • an isolated nucleic acid of the invention may be substantially isolated with respect to the complex cellular milieu in which it naturally occurs, or culture medium when produced by recombinant techniques, or chemical precursors or other chemicals when chemically synthesized, hi some instances, the isolated material will form part of a composition (for example, a crude extract containing other substances), buffer system or reagent mix.
  • an isolated nucleic acid molecule comprises at least about 50, 80 or 90% (on a molar basis) of all macromolecular species present.
  • genomic DNA the term “isolated” also can refer to nucleic acid molecules which are separated from the chromosome with which the genomic DNA is naturally associated.
  • the isolated nucleic acid molecule can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb or 0.1 kb of nucleotides which flank the nucleic acid molecule in the genomic DNA of the cell from which the nucleic acid molecule is derived.
  • the nucleic acid molecule can be fused to other coding or regulatory sequences and still be considered isolated.
  • recombinant DNA contained in a vector is included in the definition of "isolated" as used herein.
  • isolated nucleic acid molecules include recombinant DNA molecules in heterologous host cells, as well as partially or substantially purified DNA molecules in solution.
  • isolated nucleic acid molecules also encompass in vivo and in vitro RNA transcripts of the DNA molecules of the present invention.
  • An isolated nucleic acid molecule or nucleotide sequence can include a nucleic acid molecule or nucleotide sequence which is synthesized chemically or by recombinant means. Therefore, recombinant DNA contained in a vector are included in the definition of "isolated” as used herein.
  • isolated nucleotide sequences include recombinant DNA molecules in heterologous organisms, as well as partially or substantially purified DNA molecules in solution. In vivo and in vitro RNA transcripts of the DNA molecules of the present invention are also encompassed by "isolated" nucleotide sequences.
  • Such isolated nucleotide sequences are useful in the manufacture of the encoded polypeptide, as probes for isolating homologous sequences (e.g., from other mammalian species), for gene mapping (e.g. , by in situ hybridization with chromosomes), or for detecting expression of the gene in tissue (e.g., human tissue), such as by Northern blot analysis.
  • homologous sequences e.g., from other mammalian species
  • gene mapping e.g. , by in situ hybridization with chromosomes
  • tissue e.g., human tissue
  • the present invention also pertains to nucleic acid molecules which are not necessarily found in nature but which encode an NR polypeptide (e.g., a polypeptide having an amino acid sequence comprising an amino acid sequence of SEQ ID NO: 2), or another splicing variant of an NR polypeptide or polymorphic variant thereof.
  • NR polypeptide e.g., a polypeptide having an amino acid sequence comprising an amino acid sequence of SEQ ID NO: 2
  • DNA molecules which comprise a sequence that is different from the naturally-occurring nucleotide sequence but which, due to the degeneracy of the genetic code, encode an NR polypeptide of the present invention are also the subject of this invention.
  • the invention also encompasses nucleotide sequences encoding portions (fragments), or encoding variant polypeptides such as analogues or derivatives of an NR polypeptide.
  • Such variants can be naturally-occurring, such as in the case of allelic variation or single nucleotide polymorphisms, or non- naturally-occurring, such as those induced by various mutagens and mutagenic processes.
  • Intended variations include, but are not limited to, addition, deletion and substitution of one or more nucleotides which can result in conservative or non- conservative amino acid changes, including additions and deletions.
  • the nucleotide (and/or resultant amino acid) changes are silent or conserved; that is, they do not alter the characteristics or activity of an NR polypeptide.
  • nucleotide sequences are fragments that comprise one or more polymorphic microsatellite markers, hi another preferred embodiment, the nucleotide sequences are fragments that comprise one or more single nucleotide polymorphisms in an NR gene.
  • nucleic acid molecules of the invention can include, for example, labeling, methylation, internucleotide modifications such as uncharged linkages (e.g. , methyl phosphonates, phosphotriesters, phosphoamidates, carbamates), charged linkages (e.g., phosphorothioates, phosphorodithioates), pendent moieties (e.g., polypeptides), intercalators (e.g., acridine, psoralen), chelators, alkylators, and modified linkages (e.g., alpha anomeric nucleic acids).
  • uncharged linkages e.g. , methyl phosphonates, phosphotriesters, phosphoamidates, carbamates
  • charged linkages e.g., phosphorothioates, phosphorodithioates
  • pendent moieties e.g., polypeptides
  • intercalators e.g., acridine, p
  • synthetic molecules that mimic nucleic acid molecules in the ability to bind to a designated sequences via hydrogen bonding and other chemical interactions.
  • Such molecules include, for example, those in which peptide linkages substitute for phosphate linkages in the backbone of the molecule.
  • the invention also pertains to nucleic acid molecules which hybridize under high stringency hybridization conditions, such as for selective hybridization, to a nucleotide sequence described herein (e.g., nucleic acid molecules which specifically hybridize to a nucleotide sequence encoding polypeptides described herein, and, optionally, have an activity of the polypeptide).
  • the invention includes variants described herein which hybridize under high stringency hybridization conditions (e.g., for selective hybridization) to a nucleotide sequence comprising a nucleotide sequence of SEQ ID NO: 1.
  • the invention includes variants described herein which hybridize under high stringency hybridization conditions (e.g., for selective hybridization) to a nucleotide sequence encoding an amino acid sequence of SEQ ID NO: 2 or a polymorphic variant thereof, hi a preferred embodiment, the variant which hybridizes under high stringency hybridizations has an activity of an NR.
  • nucleic acid molecules can be detected and/or isolated by specific hybridization (e.g., under high stringency conditions).
  • Specific hybridization refers to the ability of a first nucleic acid to hybridize to a second nucleic acid in a manner such that the first nucleic acid does not hybridize to any nucleic acid other than to the second nucleic acid (e.g.
  • “Stringency conditions” for hybridization is a term of art which refers to the incubation and wash conditions, e.g., conditions of temperature and buffer concentration, which permit hybridization of a particular nucleic acid to a second nucleic acid; the first nucleic acid may be perfectly (i.e., 100%) complementary to the second, or the first and second may share some degree of complementarity which is less than perfect (e.g., 70%, 75%, 85%, 90%, 95%). For example, certain high stringency conditions can be used which distinguish perfectly complementary nucleic acids from those of less complementarity.
  • the exact conditions which determine the stringency of hybridization depend not only on ionic strength (e.g., 0.2X SSC, 0.1X SSC), temperature (e.g., room temperature, 42°C, 68°C) and the concentration of destabilizing agents such as formamide or denaturing agents such as SDS, but also on factors such as the length of the nucleic acid sequence, base composition, percent mismatch between hybridizing sequences and the frequency of occurrence of subsets of that sequence within other non-identical sequences.
  • equivalent conditions can be determined by varying one or more of these parameters while maintaining a similar degree of identity or similarity between the two nucleic acid molecules.
  • conditions are used such that sequences at least about 60%, at least about 70%, at least about 80%o, at least about 90% or at least about 95%> or more identical to each other remain hybridized to one another.
  • hybridization conditions By varying hybridization conditions from a level of stringency at which no hybridization occurs to a level at which hybridization is first observed, conditions which will allow a given sequence to hybridize (e.g., selectively) with the most similar sequences in the sample can be determined. Exemplary conditions are described in Krause, M.H. and S.A. Aaronson,
  • washing is the step in which conditions are usually set so as to determine a minimum level of complementarity of the hybrids. Generally, starting from the lowest temperature at which only homologous hybridization occurs, each °C by which the final wash temperature is reduced (holding SSC concentration constant) allows an increase by 1% in the maximum extent of mismatching among the sequences that hybridize. Generally, doubling the concentration of SSC results in an increase in T m of ⁇ 17°C . Using these guidelines, the washing temperature can be determined empirically for high, moderate or low stringency, depending on the level of mismatch sought.
  • a low stringency wash can comprise washing in a solution containing 0.2X SSC/0.1% SDS for 10 minutes at room temperature;
  • a moderate stringency wash can comprise washing in a prewarmed solution (42°C) solution containing 0.2X SSC/0.1% SDS for 15 minutes at 42°C;
  • a high stringency wash can comprise washing in prewarmed (68°C) solution containing 0. IX SSC/0.1%SDS for 15 minutes at 68°C.
  • washes can be performed repeatedly or sequentially to obtain a desired result as known in the art. Equivalent conditions can be determined by varying one or more of the parameters given as an example, as known in the art, while maintaining a similar degree of identity or similarity between the target nucleic acid molecule and the primer or probe used.
  • the default parameters of the respective programs e.g., NBLAST
  • Another preferred, non-limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller CABIOS 4(1): 11-17, 1998. Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package (Accelrys, Cambridge, UK).
  • a PAM120 weight residue table When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12 , and a gap penalty of 4 can be used. Additional algorithms for sequence analysis are known in the art and include ADVANCE and ADAM as described in Torellis and Robotti, Comput. Appl Biosci. 10:3-5, 1994; and FASTA described in Pearson and Lipman Proc. Natl. Acad. Sci. USA 85:2444-8, 1988. h another embodiment, the percent identity between two amino acid sequences can be accomplished using the GAP program in the GCG software package using either a BLOSUM63 matrix or a PAM250 matrix, and a gap weight of 12, 10, 8, 6, or 4 and a length weight of 2, 3, or 4.
  • the percent identity between two nucleic acid sequences can be accomplished using the GAP program in the GCG software package, using a gap weight of 50 and a length weight of 3.
  • the present invention also provides isolated nucleic acid molecules that contain a fragment or portion that hybridizes under highly stringent conditions to a nucleotide sequence comprising a nucleotide sequence of SEQ ID NO: 1, or the complement of such a sequence, and also provides isolated nucleic acid molecules that contain a fragment or portion that hybridizes under highly stringent conditions to a nucleotide sequence encoding an amino acid sequence selected from SEQ ID NO: 2, or polymorphic variant thereof.
  • the nucleic acid fragments of the invention are at least about 15, preferably at least about 18, 20, 23 or 25 nucleotides, and can be 30, 40, 50, 100, 200 or more nucleotides in length. Longer fragments, for example, 30 or more nucleotides in length, which encode antigenic polypeptides described herein are particularly useful, such as for the generation of antibodies as described below.
  • the nucleic acid fragments of the invention are used as probes or primers in assays such as those described herein.
  • Probes or “primers” are oligonucleotides that hybridize in a base-specific manner to a complementary strand of nucleic acid molecules.
  • probes and primers include polypeptide nucleic acids, as described in Nielsen et al, Science 254:1497-1500, 1991.
  • a probe or primer comprises a region of nucleotide sequence that hybridizes to at least about 15, typically about 20-25, and more typically about 40, 50 or 75, consecutive nucleotides of a nucleic acid molecule comprising a contiguous nucleotide sequence of SEQ JD NO: 1, or the complement of such a sequence, or a sequence encoding an amino acid sequence selected from SEQ ID NO: 2, or polymorphic variant thereof, h preferred embodiments, a probe or primer comprises 100 or fewer nucleotides, preferably from 6 to 50 nucleotides, preferably from 12 to 30 nucleotides.
  • the probe or primer is at least 70% identical to the contiguous nucleotide sequence or to the complement of the contiguous nucleotide sequence, preferably at least 80% identical, more preferably at least 90% identical, even more preferably at least 95% identical, or even capable of selectively hybridizing to the contiguous nucleotide sequence or to the complement of the contiguous nucleotide sequence.
  • the probe or primer further comprises a label, e.g., radioisotope, fluorescent compound, enzyme, or enzyme co-factor.
  • the nucleic acid molecules of the invention such as those described above can be identified and isolated using standard molecular biology techniques and the sequence information provided herein.
  • nucleic acid molecules can be amplified and isolated by the polymerase chain reaction using synthetic oligonucleotide primers designed based on one or more of the sequences of SEQ JJD NO: 1, or the complement of such a sequence, or designed based on nucleotides based on sequences encoding one or more of the amino acid sequences provided herein.
  • synthetic oligonucleotide primers designed based on one or more of the sequences of SEQ JJD NO: 1, or the complement of such a sequence, or designed based on nucleotides based on sequences encoding one or more of the amino acid sequences provided herein.
  • the nucleic acid molecules can be amplified using cDNA, mRNA or genomic DNA as a template, cloned into an appropriate vector and characterized by DNA sequence analysis.
  • LCR ligase chain reaction
  • NASBA nucleic acid based sequence amplification
  • the latter two amplification methods involve isothermal reactions based on isothermal transcription, which produce both single stranded RNA (ssRNA) and double stranded DNA (dsDNA) as the amplification products in a ratio of about 30 or 100 to 1, respectively.
  • the amplified DNA can be radiolabelled and used as a probe for screening a cDNA library derived from human cells, mRNA in zap express, ZIPLOX or other suitable vector.
  • Corresponding clones can be isolated, DNA can obtained following in vivo excision, and the cloned insert can be sequenced in either or both orientations by art recognized methods to identify the correct reading frame encoding a polypeptide of the appropriate molecular weight.
  • the direct analysis of the nucleotide sequence of nucleic acid molecules of the present invention can be accomplished using well-known methods that are commercially available. See, for example, Sambrook et al, Molecular Cloning, A Laboratory Manual (2nd Ed., CSHP, New York 1989); Zyskind et al, Recombinant DNA Laboratory Manual, (Acad. Press, 1988)). Using these or similar methods, the polypeptide and the DNA encoding the polypeptide can be isolated, sequenced and further characterized.
  • Antisense nucleic acid molecules of the invention can be designed using the nucleotide sequences of SEQ JD NO: 1 and/or the complement of SEQ ID NO: 1, and/or a portion of SEQ ID NO: 1, or the complement of SEQ JD NO: 1 and/or a sequence encoding the amino acid sequences of SEQ JD NO: 2, or encoding a portion of SEQ ID NO: 2, and constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art.
  • an antisense nucleic acid molecule e.g., an antisense oligonucleotide
  • an antisense nucleic acid molecule can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used.
  • the antisense nucleic acid molecule can be produced biologically using an expression vector into which a nucleic acid molecule has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid molecule will be of an antisense orientation to a target nucleic acid of interest).
  • the isolated nucleic acid sequences of the invention can be used as molecular weight markers on Southern gels, and as chromosome markers which are labeled to map related gene positions.
  • the nucleic acid sequences can also be used to compare with endogenous DNA sequences in patients to identify one or more of the disorders described above, and as probes, such as to hybridize and discover related DNA sequences or to subtract out known sequences from a sample.
  • the nucleic acid sequences can further be used to derive primers for genetic finge rinting, to raise anti-polypeptide antibodies using DNA immunization techniques, and as an antigen to raise anti-DNA antibodies or elicit immune responses.
  • nucleotide sequences identified herein can be used in numerous ways as polynucleotide reagents. For example, these sequences can be used to: (i) map their respective genes on a chromosome; and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue typing); and (iii) aid in forensic identification of a biological sample. Additionally, the nucleotide sequences of the invention can be used to identify and express recombinant polypeptides for analysis, characterization or therapeutic use, or as markers for tissues in which the corresponding polypeptide is expressed, either constitutively, during tissue differentiation, or in diseased states.
  • nucleic acid sequences can additionally be used as reagents in the screening and/or diagnotic assays described herein, and can also be included as components of kits (e.g., reagent kits) for use in the screening and/or diagnostic assays described herein.
  • kits e.g., reagent kits
  • nucleic acid constructs containing a nucleic acid molecule of SEQ JD NO: 1 and the complements thereof (or a portion thereof).
  • the constructs comprise a vector (e.g., an expression vector) into which a sequence of the invention has been inserted in a sense or antisense orientation.
  • vector refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked.
  • vector refers to a circular double stranded DNA loop into which additional DNA segments can be ligated.
  • viral vector refers to a viral vector, wherein additional DNA segments can be ligated into the viral genome.
  • Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors).
  • Other vectors e.g., non-episomal mammalian vectors
  • certain vectors, expression vectors are capable of directing the expression of genes to which they are operably linked
  • expression vectors of utility in recombinant DNA techniques are often in the form of plasmids.
  • the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adeno viruses and adeno-associated viruses) that serve equivalent functions.
  • Preferred recombinant expression vectors of the invention comprise a nucleic acid molecule of the invention in a form suitable for expression of the nucleic acid molecule in a host cell.
  • the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operably linked to the nucleic acid sequence to be expressed.
  • "operably linked” or "operatively linked” is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell).
  • regulatory sequence is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, “Gene Expression Technology", Methods in Enzymology 185, Academic Press, San Diego, CA (1990). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cell and those which direct expression of the nucleotide sequence only in certain host cells (e.g. , tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed and the level of expression of polypeptide desired.
  • the expression vectors of the invention can be introduced into host cells to thereby produce polypeptides, including fusion polypeptides, encoded by nucleic acid molecules as described herein.
  • the recombinant expression vectors of the invention can be designed for expression of a polypeptide of the invention in prokaryotic or eukaryotic cells, e.g., bacterial cells such as E. coli, insect cells (using baculovirus expression vectors), yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, supra.
  • the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
  • host cell and "recombinant host cell” are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • a host cell can be any prokaryotic or eukaryotic cell.
  • a nucleic acid molecule of the invention can be expressed in bacterial cells (e.g., E. coli), insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells).
  • bacterial cells e.g., E. coli
  • insect cells e.g., yeast or mammalian cells
  • mammalian cells such as Chinese hamster ovary cells (CHO) or COS cells
  • Other suitable host cells are known to those skilled in the art.
  • Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques.
  • transformation and transfection are intended to refer to a variety of art-recognized techniques for introducing a foreign nucleic acid molecule (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, hpofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (supra), and other laboratory manuals.
  • a gene that encodes a selectable marker (e.g., for resistance to antibiotics) is generally introduced into the host cells along with the gene of interest.
  • selectable markers include those that confer resistance to drugs, such as G418, hygromycin and methotrexate.
  • Nucleic acid molecules encoding a selectable marker can be introduced into a host cell on the same vector as the nucleic acid molecule of the invention or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid molecule can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).
  • a host cell of the invention such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) a polypeptide of the invention.
  • the invention further provides methods for producing a polypeptide using the host cells of the invention.
  • the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding a polypeptide of the invention has been introduced) in a suitable medium such that the polypeptide is produced.
  • the method further comprises isolating the polypeptide from the medium or the host cell.
  • a host cell of the invention can also be used to produce nonhuman transgenic animals.
  • a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which a nucleic acid molecule of the invention has been introduced (e.g. , an exogenous NR gene, or an exogenous nucleic acid encoding an NR polypeptide).
  • a nucleic acid molecule of the invention e.g. , an exogenous NR gene, or an exogenous nucleic acid encoding an NR polypeptide.
  • Such host cells can then be used to create non-human transgenic animals in which exogenous nucleotide sequences have been introduced into the genome or homologous recombinant animals in which endogenous nucleotide sequences have been altered.
  • transgenic animal is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene.
  • rodent such as a rat or mouse
  • transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens and amphibians.
  • a transgene is exogenous DNA which is integrated into the genome of a cell from which a transgenic animal develops and which remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal.
  • an "homologous recombinant animal” is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal.
  • Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wihnut et al. (1997) Nature 385:810-813 and PCT Publication Nos. WO 97/07668 and WO 97/07669.
  • the present invention also pertains to isolated polypeptides encoded by NRs ("NR polypeptides”) and fragments and variants thereof, as well as polypeptides encoded by nucleotide sequences described herein (e.g., other splicing variants).
  • polypeptide refers to a polymer of amino acids, and not to a specific length; thus, peptides, oligopeptides and proteins are included within the definition of a polypeptide.
  • a polypeptide is said to be “isolated” or “purified” when it is substantially free of cellular material when it is isolated from recombinant and non-recombinant cells, or free of chemical precursors or other chemicals when it is chemically synthesized.
  • a polypeptide can be joined to another polypeptide with which it is not normally associated in a cell (e.g. , in a "fusion protein") and still be “isolated” or “purified.”
  • polypeptides of the invention can be purified to homogeneity. It is understood, however, that preparations in which the polypeptide is not purified to homogeneity are useful. The critical feature is that the preparation allows for the desired function of the polypeptide, even in the presence of considerable amounts of other components. Thus, the invention encompasses various degrees of purity.
  • the language "substantially free of cellular material” includes preparations of the polypeptide having less than about 30% (by dry weight) other proteins (i.e., contaminating protein), less than about 20% other proteins, less than about 10% other proteins, or less than about 5% other proteins.
  • a polypeptide When a polypeptide is recombinantly produced, it can also be substantially free of culture medium, i.e., culture medium represents less than about 20%, less than about 10%, or less than about 5% of the volume of the polypeptide preparation.
  • the language "substantially free of chemical precursors or other chemicals” includes preparations of the polypeptide in which it is separated from chemical precursors or other chemicals that are involved in its synthesis, hi one embodiment, the language “substantially free of chemical precursors or other chemicals” includes preparations of the polypeptide having less than about 30% (by dry weight) chemical precursors or other chemicals, less than about 20% chemical precursors or other chemicals, less than about 10% chemical precursors or other chemicals, or less than about 5% chemical precursors or other chemicals.
  • a polypeptide of the invention comprises an amino acid sequence encoded by a nucleic acid molecule comprising a nucleotide sequence of SEQ JD NO: 1, or the complement of such a nucleic acid, or portions thereof, e.g., SEQ JD NO: 1, or a portion or polymorphic variant thereof.
  • the polypeptides of the invention also encompass fragment and sequence variants.
  • Variants include a substantially homologous polypeptide encoded by the same genetic locus in an organism, i.e., an allelic variant, as well as other splicing variants.
  • Variants also encompass polypeptides derived from other genetic loci in an organism, but having substantial homology to a polypeptide encoded by a nucleic acid molecule comprising a nucleotide sequence of SEQ JD NO: 1, or a complement of such a sequence, or portions thereof, or having substantial homology to a polypeptide encoded by a nucleic acid molecule comprising a nucleotide sequence of nucleotide sequences encoding SEQ JD NO: 2, or polymorphic variants thereof.
  • Variants also include polypeptides substantially homologous or identical to these polypeptides but derived from another organism, i.e., an ortholog.
  • Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by chemical synthesis. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by recombinant methods. As used herein, two polypeptides (or a region of the polypeptides) are substantially homologous or identical when the amino acid sequences are at least about 45-55%, typically at least about 70-75%, more typically at least about 80-85%, and most typically greater than about 90% or more homologous or identical.
  • a substantially homologous amino acid sequence will be encoded by a nucleic acid molecule hybridizing to one or more of SEQ ID NO: 1, or portion thereof, under stringent conditions as more particularly described above, or will be encoded by a nucleic acid molecule hybridizing to a nucleic acid sequence encoding one of SEQ ID NO: 2, a portion thereof or polymorphic variant thereof, under stringent conditions as more particularly described thereof.
  • the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of one polypeptide or nucleic acid molecule for optimal alignment with the other polypeptide or nucleic acid molecule).
  • amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in one sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the other sequence, then the molecules are homologous at that position.
  • amino acid or nucleic acid "homology” is equivalent to amino acid or nucleic acid "identity”.
  • the percent homology between the two sequences is a function of the number of identical positions shared by the sequences (i.e., percent homology equals the number of identical positions/total number of positions times 100).
  • the invention also encompasses polypeptides having a lower degree of identity but having sufficient similarity so as to perform one or more of the same functions performed by a polypeptide encoded by a nucleic acid molecule of the invention. Similarity is determined by conserved amino acid substitution. Such substitutions are those that substitute a given amino acid in a polypeptide by another amino acid of like characteristics. Conservative substitutions are likely to be phenotypically silent.
  • a variant polypeptide can differ in amino acid sequence by one or more substitutions, deletions, insertions, inversions, fusions, and truncations or a combination of any of these.
  • variant polypeptides can be fully functional or can lack function in one or more activities.
  • Fully functional variants typically contain only conservative variation or variation in non-critical residues or in non-critical regions.
  • Functional variants can also contain substitution of similar amino acids that result in no change or an insignificant change in function. Alternatively, such substitutions may positively or negatively affect function to some degree.
  • Non-functional variants typically contain one or more non-conservative amino acid substitutions, deletions, insertions, inversions, or truncation or a substitution, insertion, inversion, or deletion in a critical residue or critical region.
  • Amino acids that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (Cunningham et al, Science 244:1081-1085 (1989)). The latter procedure introduces single alanine mutations at every residue in the molecule. The resulting mutant molecules are then tested for biological activity in vitro, or in vitro proliferative activity. Sites that are critical for polypeptide activity can also be determined by structural analysis such as crystallization, nuclear magnetic resonance or photoaffmity labeling (Smith et al, J. Mol Biol 224:899-904 (1992); de Vos et al, Science 255:306-312 (1992)).
  • the invention also includes polypeptide fragments of the polypeptides of the invention. Fragments can be derived from a polypeptide encoded by a nucleic acid molecule comprising SEQ JD NO: 1, or a complement of such a nucleic acid (e.g., SEQ JD NO: 1, or other variants). However, the invention also encompasses fragments of the variants of the polypeptides described herein. As used herein, a fragment comprises at least 6 contiguous amino acids. Useful fragments include those that retain one or more of the biological activities of the polypeptide as well as fragments that can be used as an immunogen to generate polypeptide-specific antibodies.
  • Biologically active fragments can comprise a domain, segment, or motif that has been identified by analysis of the polypeptide sequence using well-known methods, e.g., signal peptides, extracellular domains, one or more transmembrane segments or loops, ligand binding regions, zinc finger domains, DNA binding domains, acylation sites, glycosylation sites, or phosphorylation sites. Fragments can be discrete (not fused to other amino acids or polypeptides) or can be within a larger polypeptide. Further, several fragments can be comprised within a single larger polypeptide. In one embodiment a fragment designed for expression in a host can have heterologous pre- and pro-polypeptide regions fused to the amino terminus of the polypeptide fragment and an additional region fused to the carboxyl terminus of the fragment.
  • the invention thus provides chimeric or fusion polypeptides.
  • These comprise a polypeptide of the invention operatively linked to a heterologous protein or polypeptide having an amino acid sequence not substantially homologous to the polypeptide.
  • “Operatively linked” indicates that the polypeptide and the heterologous protein are fused in-frame.
  • the heterologous protein can be fused to the N-terminus or C-terminus of the polypeptide.
  • the fusion polypeptide does not affect function of the polypeptide per se.
  • the fusion polypeptide can be a GST-fusion polypeptide in which the polypeptide sequences are fused to the C-terminus of the GST sequences.
  • fusion polypeptides include, but are not limited to, enzymatic fusion polypeptides, for example ⁇ -galactosidase fusions, yeast two-hybrid GAL fusions, poly-His fusions and Ig fusions.
  • enzymatic fusion polypeptides for example ⁇ -galactosidase fusions, yeast two-hybrid GAL fusions, poly-His fusions and Ig fusions.
  • Such fusion polypeptides, particularly poly-His fusions can facilitate the purification of recombinant polypeptide.
  • hi certain host cells e.g., mammalian host cells
  • expression and/or secretion of a polypeptide can be increased by using a heterologous signal sequence. Therefore, in another embodiment, the fusion polypeptide contains a heterologous signal sequence at its N-terminus.
  • EP-A-O 464 533 discloses fusion proteins comprising various portions of immunoglobulin constant regions.
  • the Fc is useful in therapy and diagnosis and thus results, for example, in improved pharmacokinetic properties (EP-A 0232 262).
  • human proteins have been fused with Fc portions for the purpose of high-throughput screening assays to identify antagonists. Bennett et al, J. Mol Recog. 8:52-58 (1995) and Johanson et al, J. Biol Chem. 270:16:9459-9471 (1995).
  • this invention also encompasses soluble fusion polypeptides containing a polypeptide of the invention and various portions of the constant regions of heavy or light chains of immunoglobulins of various subclass (I g G, IgM, IgA, IgE).
  • a chimeric or fusion polypeptide can be produced by standard recombinant DNA techniques. For example, DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques. hi another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of nucleic acid fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive nucleic acid fragments which can subsequently be annealed and re-amplified to generate a chimeric nucleic acid sequence (see Ausubel et al. , Current Protocols in Molecular Biology, 1992).
  • a nucleic acid molecule encoding a polypeptide of the invention can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the polypeptide.
  • the isolated polypeptide can be purified from cells that naturally express it, purified from cells that have been altered to express it (recombinant), or synthesized using known protein synthesis methods.
  • the polypeptide is produced by recombinant DNA techniques.
  • a nucleic acid molecule encoding the polypeptide is cloned into an expression vector, the expression vector introduced into a host cell and the polypeptide expressed in the host cell.
  • polypeptide can then be isolated from the cells by an appropriate purification scheme using standard protein purification techniques.
  • polypeptides of the present invention can be used as a molecular weight marker on SDS-PAGE gels or on molecular sieve gel filtration columns using art-recognized methods.
  • the polypeptides of the present invention can be used to raise antibodies or to elicit an immune response.
  • the polypeptides can also be used as a reagent, e.g., a labeled reagent, in assays to quantitatively dete ⁇ nine levels of the polypeptide or a molecule to which it binds (e.g., a ligand) in biological fluids.
  • polypeptides can also be used as markers for cells or tissues in which the corresponding polypeptide is preferentially expressed, either constitutively, during tissue differentiation, or in a diseased state.
  • the polypeptides can be used to isolate a corresponding binding agent, e.g., ligand, such as, for example, in an interaction trap assay, and to screen for peptide or small molecule antagonists or agonists of the binding interaction.
  • polypeptides and polypeptide fragments of the invention e.g., having an amino acid sequence of SEQ JD NO: 2 or a portion thereof, or having an amino acid sequence encoded by a nucleic acid molecule comprising all or a portion of SEQ ID NO: 1, or a complement or another variant or portion thereof.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically binds an antigen.
  • a molecule that specifically binds to a polypeptide of the invention is a molecule that binds to that polypeptide or a fragment thereof, but does not substantially bind other molecules in a sample, e.g., a biological sample, which naturally contains the polypeptide.
  • immunologically active portions of immunoglobulin molecules include F(ab) and F(ab') 2 fragments which can be generated by treating the antibody with an enzyme such as pepsin.
  • the invention provides polyclonal and monoclonal antibodies that bind to a polypeptide of the invention.
  • a monoclonal antibody composition thus typically displays a single binding affinity for a particular polypeptide of the invention with which it immunoreacts.
  • Polyclonal antibodies can be prepared as described above by immunizing a suitable subject with a desired immunogen, e.g., polypeptide of the invention or fragment thereof.
  • a desired immunogen e.g., polypeptide of the invention or fragment thereof.
  • the antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized polypeptide.
  • ELISA enzyme linked immunosorbent assay
  • the antibody molecules directed against the polypeptide can be isolated from the mammal (e.g., from the blood) and further purified by well-known techniques, such as protein A chromatography to obtain the IgG fraction.
  • antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497, the human B cell hybridoma technique (Kozbor et al. (1983) Immunol. Today 4:72), the EBV-hybridoma technique (Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, h e, pp. 77-96) or tiioma techniques.
  • standard techniques such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497, the human B cell hybridoma technique (Kozbor et al. (1983) Immunol. Today 4:72), the EBV-hybridoma technique (Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, h e, pp. 77-9
  • hybridomas The technology for producing hybridomas is well known (see generally Current Protocols in Immunology (1994) Coligan et al. (eds.) John Wiley & Sons, Inc., New York, NY). Briefly, an immortal cell line (typically a myeloma) is fused to lymphocytes (typically splenocytes) from a mammal immunized with an immunogen as described above, and the culture supernatants of the resulting hybridoma cells are screened to identify a hybridoma producing a monoclonal antibody that binds a polypeptide of the invention.
  • lymphocytes typically splenocytes
  • a monoclonal antibody to a polypeptide of the invention can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with the polypeptide to thereby isolate immunoglobulin library members that bind the polypeptide.
  • Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01; and the Stratagene SurfZAPTM Phage Display Kit, Catalog No. 240612). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, U.S.
  • recombinant antibodies such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the invention.
  • Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art.
  • antibodies of the invention e.g., a monoclonal antibody
  • a polypeptide-specific antibody can facilitate the purification of natural polypeptide from cells and of recombinantly produced polypeptide expressed in host cells.
  • an antibody specific for a polypeptide of the invention can be used to detect the polypeptide (e.g., in a cellular lysate, cell supernatant, or tissue sample) in order to evaluate the abundance and pattern of expression of the polypeptide.
  • Antibodies can be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, ⁇ -galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin;
  • an example of a luminescent material includes luminol;
  • examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include I, I, S or H.
  • the present invention also pertains to a method of diagnosing or aiding in the diagnosis of a disease or condition associated with an NR gene or gene product in an individual.
  • Diagnostic assays can be designed for assessing NR gene expression, or for assessing activity of NR polypeptides of the invention, hi one embodiment, the assays are used in the context of a biological sample (e.g., blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a disease or condition associated with an NR, or a defect in an NR.
  • a biological sample e.g., blood, serum, cells, tissue
  • the invention also provides for prognostic (or predictive) assays for determining whether an individual is susceptible to a disease of condition associated with a NR, e.g., if an individual is at risk for addiction to an opoid. For example, mutations in the gene can be assayed in a biological sample. Such assays can be used for prognostic or predictive purpose to thereby prophylactically treat an individual prior to the onset of symptoms associated with a susceptibility to a disease or condition associated with an NR.
  • Another aspect of the invention pertains to assays for monitoring the influence of agents (e.g., drugs, compounds or other agents) on the gene expression or activity of polypeptides of the invention, as well as to assays for identifying agents which bind to a polypeptides.
  • agents e.g., drugs, compounds or other agents
  • assays for identifying agents which bind to a polypeptides are described in further detail in the following sections.
  • nucleic acids, probes, primers, polypeptides and antibodies described herein can be used in methods of diagnosis of a susceptibility to a disease or condition associated with an NR, as well as in kits useful for diagnosis of a susceptibility to a disease or condition associated with an NR.
  • diagnosis of a susceptibility to a disease or condition associated with an NR is made by detecting a polymorphism in an NR as described herein.
  • the polymorphism can be a mutation in an NR, such as the insertion or deletion of a single nucleotide, or of more than one nucleotide, resulting in a frame shift mutation; the change of at least one nucleotide, resulting in a change in the encoded amino acid; the change of at least one nucleotide, resulting in the generation of a premature stop codon; the deletion of several nucleotides, resulting in a deletion of one or more amino acids encoded by the nucleotides; the insertion of one or several nucleotides, such as by unequal recombination or gene conversion, resulting in an interruption of the coding sequence of the gene; duplication of all or a part of the gene; transposition of all or apart of the gene; or rearrangement of all or a part of the gene
  • More than one such mutation may be present in a single gene.
  • sequence changes cause a mutation in the polypeptide encoded by an NR gene.
  • the mutation is a frame shift mutation
  • the frame shift can result in a change in the encoded amino acids, and/or can result in the generation of a premature stop codon, causing generation of a truncated polypeptide.
  • a polymorphism associated with a susceptibility to a disease or condition associated with an NR can be a synonymous mutation in one or more nucleotides (i.e., a mutation that does not result in a change in the polypeptide encoded by an NR gene).
  • a polymorphism may alter splicing sites, affect the stability or transport of mRNA, or otherwise affect the transcription or translation of the gene.
  • An NR gene that has any of the mutations described above is referred to herein as a "mutant gene.” h a first method of diagnosing a susceptibility to a susceptibility to a disease or condition associated with an NR, hybridization methods, such as Southern analysis, Northern analysis, or in situ hybridizations, can be used (see Current Protocols in Molecular Biology, Ausubel, F. et al, eds., John Wiley & Sons, including all supplements through 1999).
  • a biological sample from a test subject (a "test sample") of genomic DNA, RNA, or cDNA, is obtained from an individual suspected of having, being susceptible to or predisposed for, or carrying a defect for, a susceptibility to a disease or condition associated with an NR (the "test individual").
  • the individual can be an adult, child, or fetus.
  • the test sample can be from any source which contains genomic DNA, such as a blood sample, sample of amniotic fluid, sample of cerebrospinal fluid, or tissue sample from skin, muscle, buccal or conjunctival mucosa, placenta, gastrointestinal tract or other organs.
  • a test sample of DNA from fetal cells or tissue can be obtained by appropriate methods, such as by amniocentesis or chorionic villus sampling.
  • the DNA, RNA, or cDNA sample is then examined to determine whether a polymorphism in an NR is present, and/or to determine which splicing variant(s) encoded by the NR is present.
  • the presence of the polymorphism or splicing variant(s) can be indicated by hybridization of the gene in the genomic DNA, RNA, or cDNA to a nucleic acid probe.
  • a “nucleic acid probe”, as used herein, can be a DNA probe or an RNA probe; the nucleic acid probe can contain at least one polymorphism in an NR or contains a nucleic acid encoding a particular splicing variant of an NR.
  • the probe can be any of the nucleic acid molecules described above (e.g., the gene, a fragment, a vector comprising the gene, a probe or primer, etc.).
  • a hybridization sample is formed by contacting the test sample containing an NR, with at least one nucleic acid probe.
  • a preferred probe for detecting mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to mRNA or genomic DNA sequences described herein.
  • the nucleic acid probe can be, for example, a full-length nucleic acid molecule, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to appropriate mRNA or genomic DNA.
  • the nucleic acid probe can be all or a portion of one of SEQ ID NO : 1 , or the complement thereof, or a portion thereof; or can be a nucleic acid encoding a portion of one of SEQ ID NO: 2.
  • probes and primers discussed under the heading, "Nucleic Acids of the Invention” are described above (see e.g., probes and primers discussed under the heading, "Nucleic Acids of the Invention").
  • the hybridization sample is maintained under conditions which are sufficient to allow specific hybridization of the nucleic acid probe to anNR.
  • Specific hybridization indicates exact hybridization (e.g., with no mismatches).
  • Specific hybridization can be performed under high stringency conditions or moderate stringency conditions, for example, as described above. In a particularly preferred embodiment, the hybridization conditions for specific hybridization are high stringency.
  • Specific hybridization if present, is then detected using standard methods. If specific hybridization occurs between the nucleic acid probe and the NR in the test sample, then the NR has the polymorphism, or is the splicing variant, that is present in the nucleic acid probe. More than one nucleic acid probe can also be used concurrently in this method. Specific hybridization of any one of the nucleic acid probes is indicative of a polymo ⁇ hism in the NR, or of the presence of a particular splicing variant encoding the NR and is therefore diagnostic for a susceptibility to a susceptibility to a disease or condition associated with an NR. In Northern analysis (see Current Protocols in Molecular Biology, Ausubel,
  • the hybridization methods described above are used to identify the presence of a polymorphism or a particular splicing variant, associated with a susceptibility to a susceptibility to a disease or condition associated with an NR.
  • a test sample of RNA is obtained from the individual by appropriate means.
  • Specific hybridization of a nucleic acid probe, as described above, to RNA from the individual is indicative of a polymorphism in an NR, or of the presence of a particular splicing variant encoded by an NR, and is therefore diagnostic for a susceptibility to a susceptibility to a disease or condition associated with an NR.
  • nucleic acid probes For representative examples of use of nucleic acid probes, see, for example, U.S. Patents No. 5,288,611 and 4,851,330.
  • PNA peptide nucleic acid
  • a nucleic acid probe can be used instead of a nucleic acid probe in the hybridization methods described above.
  • PNA is a DNA mimic having a peptide-like, inorganic backbone, such as N-(2-aminoethyl)glycine units, with an organic base (A, G, C, T or U) attached to the glycine nitrogen via a methylene carbonyl linker (see, for example, Nielsen, P.E. et al, Bioconjugate Chemistry 5, American Chemical Society, p. 1 (1994).
  • the PNA probe can be designed to specifically hybridize to a gene having a polymorphism associated with a susceptibility to a susceptibility to a disease or condition associated with an NR. Hybridization of the PNA probe to an NR is diagnostic for a susceptibility to a susceptibility to a disease or condition associated with an NR.
  • mutation analysis by restriction digestion can be used to detect a mutant gene, or genes containing a polymorphism(s), if the mutation or polymorphism in the gene results in the creation or elimination of a restriction site.
  • a test sample containing genomic DNA is obtained from the individual.
  • PCR Polymerase chain reaction
  • NR and, if necessary, the flanking sequences
  • RFLP analysis is conducted as described (see Current Protocols in Molecular Biology, supra).
  • the digestion pattern of the relevant DNA fragment indicates the presence or absence of the mutation or polymorphism in the NR, and therefore indicates the presence or absence of this susceptibility to a susceptibility to a disease or condition associated with an NR.
  • Sequence analysis can also be used to detect specific polymorphisms in an NR.
  • a test sample of DNA or RNA is obtained from the test individual. PCR or other appropriate methods can be used to amplify the gene, and/or its flanking sequences, if desired.
  • the sequence of anNR, or a fragment of the gene, or cDNA, or fragment of the cDNA, or mRNA, or fragment of the mRNA is determined, using standard methods.
  • the sequence of the gene, gene fragment, cDNA, cDNA fragment, mRNA, or mRNA fragment is compared with the known nucleic acid sequence of the gene, cDNA (e.g., of SEQ JD NO: 1, or a complement thereof, or a nucleic acid sequence encoding SEQ JD NO: 2 or a fragment thereof) or mRNA, as appropriate.
  • the presence of a polymorphism in the NR indicates that the individual has a susceptibility to a susceptibility to a disease or condition associated with an NR.
  • Allele-specific oligonucleotides can also be used to detect the presence of a polymorphism in an NR, through the use of dot-blot hybridization of amplified oligonucleotides with allele-specific oligonucleotide (ASO) probes (see, for example, Saiki, R. et al, Nature 324:163-166 (1986)).
  • ASO allele-specific oligonucleotide
  • an “allele-specific oligonucleotide” (also referred to herein as an “allele-specific oligonucleotide probe”) is an oligonucleotide of approximately 10-50 base pairs, preferably approximately 15-30 base pairs, that specifically hybridizes to an NR, and that contains a polymorphism associated with a susceptibility to a susceptibility to a disease or condition associated with an NR.
  • An allele-specific oligonucleotide probe that is specific for particular polymorphisms in an NR can be prepared, using standard methods (see Current Protocols in Molecular Biology, supra).
  • a test sample of DNA is obtained from the individual.
  • PCR can be used to amplify all or a fragment of an NR, and its flanking sequences.
  • the DNA containing the amplified NR (or fragment of the gene) is dot-blotted, using standard methods (see Current Protocols in Molecular Biology, supra), and the blot is contacted with the oligonucleotide probe. The presence of specific hybridization of the probe to the amplified NR is then detected.
  • Specific hybridization of an allele-specific oligonucleotide probe to DNA from the individual is indicative of a polymorphism in the NR, and is therefore indicative of a susceptibility to a susceptibility to a disease or condition associated with an NR.
  • arrays of oligonucleotide probes that are complementary to target nucleic acid sequence segments from an individual can be used to identify polymorphisms in an NR.
  • an oligonucleotide array can be used.
  • Oligonucleotide arrays typically comprise a plurality of different oligonucleotide probes that are coupled to a surface of a substrate in different known locations. These oligonucleotide arrays, also described as "GenechipsTM,” have been generally described in the art, for example, U.S. Pat. No. 5,143,854 and PCT patent publication Nos. WO 90/15070 and 92/10092.
  • arrays can generally be produced using mechanical synthesis methods or light directed synthesis methods which incorporate a combination of photolithographic methods and solid phase oligonucleotide synthesis methods. See Fodor et al, Science 251:767-777 (1991), Pirrung et al, U.S. Pat. No. 5,143,854 (see also PCT Application No. WO 90/15070) and Fodor et al , PCT Publication No. WO
  • a target nucleic acid sequence which includes one or more previously identified polymorphic markers is amplified by well known amplification techniques, e.g., PCR. Typically, this involves the use of primer sequences that are complementary to the two strands of the target sequence both upstream and downstream from the polymorphism. Asymmetric PCR techniques may also be used. Amplified target, generally incorporating a label, is then hybridized with the array under appropriate conditions. Upon completion of hybridization and washing of the array, the array is scanned to determine the position on the array to which the target sequence hybridizes.
  • amplification techniques e.g., PCR.
  • Amplified target generally incorporating a label
  • the hybridization data obtained from the scan is typically in the form of fluorescence intensities as a function of location on the array.
  • arrays can include multiple detection blocks, and thus be capable of analyzing multiple, specific polymorphisms.
  • detection blocks may be grouped within a single array or in multiple, separate arrays so that varying, optimal conditions may be used during the hybridization of the target to the array. For example, it may often be desirable to provide for the detection of those polymorphisms that fall within G-C rich stretches of a genomic sequence, separately from those falling in A-T rich segments. This allows for the separate optimization of hybridization conditions for each situation.
  • nucleic acid analysis can be used to detect polymorphisms in an NR or variants encoding by an NR.
  • Representative methods include direct manual sequencing (Church and Gilbert, Proc. Natl. Acad. Sci. USA 81:1991-1995 (1988); Sanger, F. et al. Proc. Natl Acad. Sci. USA 74:5463-5467 (1977); Beavis et al, U.S. Pat. No. 5,288,644); automated fluorescent sequencing; single-stranded conformation polymorphism assays (SSCP); clamped denaturing gel electiophoresis (CDGE); denaturing gradient gel electiophoresis (DGGE) (Sheffield, V.C. et al.
  • diagnosis of a susceptibility to a susceptibility to a disease or condition associated with an NR can also be made by examining expression and/or composition of an NR polypeptide, by a variety of methods, including enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations and immunofiuorescence.
  • ELISAs enzyme linked immunosorbent assays
  • Western blots immunoprecipitations and immunofiuorescence.
  • a test sample from an individual is assessed for the presence of an alteration in the expression and/or an alteration in composition of the polypeptide encoded by an NR, or for the presence of a particular variant encoded by an NR.
  • An alteration in expression of a polypeptide encoded by an NR can be, for example, an alteration in the quantitative polypeptide expression (i.e., the amount of polypeptide produced); an alteration in the composition of a polypeptide encoded by an NR is an alteration in the qualitative polypeptide expression (e.g., expression of a mutant NR polypeptide or of a different splicing variant), hi a preferred embodiment, diagnosis of a susceptibility to a susceptibility to a disease or condition associated with an NR is made by detecting a particular splicing variant encoded by that NR, or a particular pattern of splicing variants.
  • An "alteration" in the polypeptide expression or composition refers to an alteration in expression or composition in a test sample, as compared with the expression or composition of polypeptide by an NR in a control sample.
  • a control sample is a sample that corresponds to the test sample (e.g., is from the same type of cells), and is from an individual who is not affected by a susceptibility to a disease or condition associated with an NR.
  • An alteration in the expression or composition of the polypeptide in the test sample, as compared with the control sample is indicative of a susceptibility to a susceptibility to a disease or condition associated with an NR.
  • the presence of one or more different splicing variants in the test sample, or the presence of significantly different amounts of different splicing variants in the test sample, as compared with the control sample, is indicative of a susceptibility to a susceptibility to a disease or condition associated with an NR.
  • Various means of examining expression or composition of the polypeptide encoded by an NR can be used, including spectroscopy, colorimetry, electiophoresis, isoelectric focusing, and immunoassays (e.g., David et al, U.S. Pat. No. 4,376,110) such as immunoblotting (see also Current Protocols in Molecular Biology, particularly Chapter 10).
  • an antibody capable of binding to the polypeptide e.g., as described above, preferably an antibody with a detectable label
  • Antibodies can be polyclonal, or more preferably, monoclonal.
  • An intact antibody, or a fragment thereof e.g. , Fab or F(ab') 2
  • the term "labeled", with regard to the probe or antibody is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently labeled streptavidin.
  • Western blotting analysis using an antibody as described above that specifically binds to a polypeptide encoded by a mutant NR, or an antibody that specifically binds to a polypeptide encoded by a non-mutant gene, or an antibody that specifically binds to a particular splicing variant encoded by an NR, can be used to identify the presence in a test sample of a particular splicing variant or of a polypeptide encoded by a polymorphic or mutant NR, or the absence in a test sample of a particular splicing variant or of a polypeptide encoded by a non-polymorphic or non-mutant gene.
  • the presence of a polypeptide encoded by a polymorphic or mutant gene, or the absence of a polypeptide encoded by a non-polymorphic or non- mutant gene, is diagnostic for a susceptibility to a susceptibility to a disease or condition associated with an NR, as is the presence (or absence) of particular splicing variants encoded by the NR gene.
  • the level or amount of polypeptide encoded by an NR in a test sample is compared with the level or amount of the polypeptide encoded by the NR in a control sample.
  • a level or amount of the polypeptide in the test sample that is higher or lower than the level or amount of the polypeptide in the control sample, such that the difference is statistically significant, is indicative of an alteration in the expression of the polypeptide encoded by the NR, and is diagnostic for a susceptibility to a susceptibility to a disease or condition associated with that NR.
  • the composition of the polypeptide encoded by an NR in a test sample is compared with the composition of the polypeptide encoded by the NR in a control sample (e.g., the presence of different splicing variants).
  • a difference in the composition of the polypeptide in the test sample, as compared with the composition of the polypeptide in the control sample, is diagnostic for a susceptibility to a susceptibility to a disease or condition associated with that NR.
  • both the level or amount and the composition of the polypeptide can be assessed in the test sample and in the control sample.
  • a difference in the amount or level of the polypeptide in the test sample, compared to the control sample; a difference in composition in the test sample, compared to the control sample; or both a difference in the amount or level, and a difference in the composition is indicative of a susceptibility to a susceptibility to a disease or condition associated with that NR.
  • Kits useful in the methods of diagnosis comprise components useful in any of the methods described herein, including for example, hybridization probes or primers as decribed herein (e.g. , labeled probes or primers), reagents for detection of labeled molecules, restriction enzymes (e.g., for RFLP analysis), allele-specific oligonucleotides, antibodies which bind to mutant or to non-mutant (native) NR polypeptide, means for amplification of nucleic acids comprising an NR, or means for analyzing the nucleic acid sequence of an NR or for analyzing the amino acid sequence of an NR polypeptide, etc.
  • hybridization probes or primers as decribed herein e.g. , labeled probes or primers
  • restriction enzymes e.g., for RFLP analysis
  • allele-specific oligonucleotides e.g., antibodies which bind to mutant or to non-mutant (native) NR polypeptide
  • the invention provides methods (also referred to herein as “screening assays”) for identifying the presence of a nucleotide that hybridizes to a nucleic acid of the invention, as well as for identifying the presence of a polypeptide encoded by a nucleic acid of the invention.
  • the presence (or absence) of a nucleic acid molecule of interest in a sample can be assessed by contacting the sample with a nucleic acid comprising a nucleic acid of the invention (e.g., a nucleic acid having the sequence of SEQ JD NO: 1, or the complement thereof, or a nucleic acid encoding an amino acid having the sequence of SEQ ID NO: 2, or a fragment or variant of such nucleic acids), under stringent conditions as described above, and then assessing the sample for the presence (or absence) of hybridization.
  • a nucleic acid comprising a nucleic acid of the invention e.g., a nucleic acid having the sequence of SEQ JD NO: 1, or the complement thereof, or a nucleic acid encoding an amino acid having the sequence of SEQ ID NO: 2, or a fragment or variant of such nucleic acids
  • high stringency conditions are conditions appropriate for selective hybridization
  • a sample containing the nucleic acid molecule of interest is contacted with a nucleic acid containing a contiguous nucleotide sequence (e.g., a primer or a probe as described above) that is at least partially complementary to a part of the nucleic acid molecule of interest (e.g., an NR nucleic acid), and the contacted sample is assessed for the presence or absence of hybridization
  • the nucleic acid containing a contiguous nucleotide sequence is completely complementary to a part of the nucleic acid molecule of interest.
  • all or a portion of the nucleic acid of interest can be subjected to amplification prior to performing the hybridization.
  • the presence (or absence) of a polypeptide of interest, such as a polypeptide of the invention or a fragment or variant thereof, in a sample can be assessed by contacting the sample with an antibody that specifically hybridizes to the polypeptide of interest (e.g., an antibody such as those described above), and then assessing the sample for the presence (or absence) of binding of the antibody to the polypeptide of interest.
  • an antibody that specifically hybridizes to the polypeptide of interest e.g., an antibody such as those described above
  • the invention provides methods for identifying agents (e.g., fusion proteins, polypeptides, peptidomimetics, prodrugs, other receptors associated with NRs, binding agents, antibodies, small molecules or other drugs, or ribozymes which alter (e.g., increase or decrease) the activity of the polypeptides described herein, or which otherwise interact with the polypeptides herein.
  • agents e.g., fusion proteins, polypeptides, peptidomimetics, prodrugs, other receptors associated with NRs, binding agents, antibodies, small molecules or other drugs, or ribozymes which alter (e.g., increase or decrease) the activity of the polypeptides described herein, or which otherwise interact with the polypeptides herein.
  • agents can be agents which bind to polypeptides described herein (e.g., NR binding agents); which have a stimulatory or inhibitory effect on, for example, activity of polypeptides of the invention; or which change (e.g., enhance or inhibit
  • the invention provides assays for screening candidate or test agents that bind to or modulate the activity of polypeptides described herein (or biologically active portion(s) thereof), as well as agents identifiable by the assays.
  • Test agents can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the 'one-bead one-compound' library method; and synthetic library methods using affinity chromatography selection.
  • biological libraries include polypeptide libraries, while the other four approaches are applicable to polypeptide, non-peptide oligomer or small molecule libraries of compounds (Lam, K.S., Anticancer Drug Des. 12:145 (1997)).
  • a cell, cell lysate, or solution containing or expressing an NR polypeptide e.g., one of SEQ ID NO: 2, or another splicing variant encoded by an NR
  • an agent to be tested e.g., one of SEQ ID NO: 2, or another splicing variant encoded by an NR
  • the polypeptide can be contacted directly with the agent to be tested.
  • the level (amount) of NR activity is assessed (e.g., the level (amount) of NR activity is measured, either directly or indirectly), and is compared with the level of activity in a control (i.e., the level of activity of the NR polypeptide or active fragment or derivative thereof in the absence of the agent to be tested). If the level of the activity in the presence of the agent differs, by an amount that is statistically significant, from the level of the activity in the absence of the agent, then the agent is an agent that alters the activity of an NR polypeptide. An increase in the level of NR activity relative to a control, indicates that the agent is an agent that enhances (is an agonist of) NR activity.
  • a decrease in the level of NR activity relative to a control indicates that the agent is an agent that inhibits (is an antagonist of) NR activity.
  • the level of activity of an NR polypeptide or derivative or fragment thereof in the presence of the agent to be tested is compared with a control level that has previously been established. A level of the activity in the presence of the agent that differs from the control level by an amount that is statistically significant indicates that the agent alters NR activity.
  • the present invention also relates to an assay for identifying agents which alter the expression of an NR gene (e.g., antisense nucleic acids, fusion proteins, polypeptides, peptidomimetics, prodrugs, other receptors associated with NRs, G- proteins, binding agents, antibodies, small molecules or other drugs, or ribozymes) which alter (e.g., increase or decrease) expression (e.g., transcription or translation) of the gene or which otherwise interact with the nucleic acids described herein, as well as agents identifiable by the assays.
  • an NR gene e.g., antisense nucleic acids, fusion proteins, polypeptides, peptidomimetics, prodrugs, other receptors associated with NRs, G- proteins, binding agents, antibodies, small molecules or other drugs, or ribozymes
  • alter e.g., increase or decrease expression (e.g., transcription or translation) of the gene or which otherwise interact with the nucleic acids described herein, as well as agents identifiable
  • the solution can comprise, for example, cells containing the nucleic acid or cell lysate containing the nucleic acid; alternatively, the solution can be another solution which comprises elements necessary for transcription/translation of the nucleic acid. Cells not suspended in solution can also be employed, if desired.
  • the level and/or pattern of NR expression e.g., the level and/or pattern of mRNA or of protein expressed, such as the level and/or pattern of different splicing variants
  • a control i.e., the level and/or pattern of the NR expression in the absence of the agent to be tested.
  • the agent is an agent that alters the expression of NR.
  • Enhancement of NR expression indicates that the agent is an agonist of NR activity.
  • inhibition of NR expression indicates that the agent is an antagonist of NR activity.
  • the level and/or pattern of NR polypeptide(s) e.g., different splicing variants
  • the level and/or pattern of NR polypeptide(s) in the presence of the agent to be tested, is compared with a control level and/or pattern that has previously been established.
  • agents which alter the expression of an NR gene or which otherwise interact with the nucleic acids described herein can be identified using a cell, cell lysate, or solution containing a nucleic acid encoding the promoter region of the NR gene operably linked to a reporter gene. After contact with an agent to be tested, the level of expression of the reporter gene (e.g.
  • the level of mRNA or of protein expressed is assessed, and is compared with the level of expression in a control (i.e., the level of the expression of the reporter gene in the absence of the agent to be tested). If the level in the presence of the agent differs, by an amount or in a manner that is statistically significant, from the level in the absence of the agent, then the agent is an agent that alters the expression of the NR, as indicated by its ability to alter expression of a gene that is operably linked to the NR gene promoter. Enhancement of the expression of the reporter indicates that the agent is an agonist of NR activity. Similarly, inhibition of the expression of the reporter indicates that the agent is an antagonist of NR activity.
  • the level of expression of the reporter in the presence of the agent to be tested is compared with a control level that has previously been established.
  • a level in the presence of the agent that differs from the control level by an amount or in a manner that is statistically significant indicates that the agent alters f expression.
  • Agents which alter the amounts of different splicing variants encoded by an NR e.g. , an agent which enhances activity of a first splicing variant, and which inhibits activity of a second splicing variant
  • agents which are agonists of activity of a first splicing variant and antagonists of activity of a second splicing variant can easily be identified using these methods described above.
  • assays can be used to assess the impact of a test agent on the activity of a polypeptide in relation to an NR binding agent.
  • a cell that expresses a compound that interacts with an NR herein referred to as a "NR binding agent", which can be a polypeptide or other molecule that interacts with an NR, such as a G-protein
  • NR binding agent which can be a polypeptide or other molecule that interacts with an NR, such as a G-protein
  • a cell lysate or a solution containing the NR binding agent can be used.
  • test agents can be enzymatically labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product. It is also within the scope of this invention to determine the ability of a test agent to interact with the polypeptide without the labeling of any of the interactants.
  • a microphysiometer can be used to detect the interaction of a test agent with an NR or an NR binding agent without the labeling of either the test agent, NR, or the NR binding agent. McConnell, H.M. et al, Science 257:1906-1912 (1992).
  • a "microphysiometer” e.g., CytosensorTM
  • LAPS light-addressable potentiometric sensor
  • Changes in this acidification rate can be used as an indicator of the interaction between ligand and polypeptide.
  • these receptors can be used to screen for compounds that are agonists for use in treating a susceptibility to a disease or condition associated with an NR or antagonists for studying a susceptibility to a disease or condition associated with an NR.
  • Drugs could be designed to regulate NR activation which in turn can be used to regulate signaling pathways and transcription events of genes downstream.
  • assays can be used to identify polypeptides that interact with one or more NR polypeptides, as described herein.
  • a yeast two-hybrid system such as that described by Fields and Song (Fields, S. and Song, O., Nature 340:245-246 (1989)) can be used to identify polypeptides that interact with one or more NR polypeptides.
  • vectors are constructed based on the flexibility of a transcription factor which has two functional domains (a DNA binding domain and a transcription activation domain).
  • a first vector which includes a nucleic acid encoding a DNA binding domain and also an NR polypeptide, splicing variant, or fragment or derivative thereof
  • a second vector is used which includes a nucleic acid encoding a transcription activation domain and also a nucleic acid encoding a polypeptide which potentially may interact with the NR polypeptide, splicing variant, or fragment or derivative thereof (e.g.
  • an NR polypeptide binding agent or G-protein an NR polypeptide binding agent or G-protein.
  • incubation of yeast containing the first vector and the second vector under appropriate conditions e.g., mating conditions such as used in the MatchmakerTM system from Clontech (Palo Alto, California, USA)
  • appropriate conditions e.g., mating conditions such as used in the MatchmakerTM system from Clontech (Palo Alto, California, USA)
  • Such polypeptides may be useful as agents which alter the activity of expression of an NR polypeptide, as described above.
  • NR NR
  • NR binding agent or other components of the assay on a solid support
  • binding of a test agent to the polypeptide, or interaction of the polypeptide with a binding agent in the presence and absence of a test agent can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtitre plates, test tubes, and micro-centrifuge tubes.
  • a fusion protein e.g., a glutathione-S-transferase fusion protein
  • a fusion protein e.g., a glutathione-S-transferase fusion protein
  • modulators of expression of nucleic acid molecules of the invention are identified in a method wherein a cell, cell lysate, or solution containing a nucleic acid encoding an NR is contacted with a test agent and the expression of appropriate mRNA or polypeptide (e.g., splicing variant(s)) in the cell, cell lysate, or solution, is determined.
  • appropriate mRNA or polypeptide e.g., splicing variant(s)
  • the level of expression of appropriate mRNA or polypeptide(s) in the presence of the test agent is compared to the level of expression of mRNA or polypeptide(s) in the absence of the test agent.
  • the test agent can then be identified as a modulator of expression based on this comparison. For example, when expression of mRNA or polypeptide is greater (statistically significantly greater) in the presence of the test agent than in its absence, the test agent is identified as a stimulator or enhancer of the mRNA or polypeptide expression. Alternatively, when expression of the mRNA or polypeptide is less (statistically significantly less) in the presence of the test agent than in its absence, the test agent is identified as an inhibitor of the mRNA or polypeptide expression.
  • the level of mRNA or polypeptide expression in the cells can be determined by methods described herein for detecting mRNA or polypeptide.
  • This invention further pertains to novel agents identified by the above-described screening assays. Accordingly, it is within the scope of this invention to further use an agent identified as described herein in an appropriate animal model.
  • an agent identified as described herein e.g., a test agent that is a modulating agent, an antisense nucleic acid molecule, a specific antibody, or a polypeptide-binding agent
  • an agent identified as described herein can be used in an animal model to determine the efficacy, toxicity, or side effects of treatment with such an agent.
  • an agent identified as described herein can be used in an animal model to determine the mechanism of action of such an agent.
  • this invention pertains to uses of novel agents identified by the above-described screening assays for treatments as described herein.
  • an agent identified as described herein can be used to alter activity of a polypeptide encoded by an NR, or to alter expression of an NR, by contacting the polypeptide or the gene (or contacting a cell comprising the polypeptide or the gene) with the agent identified as described herein.
  • the present invention also pertains to pharmaceutical compositions comprising nucleic acids described herein, particularly nucleotides encoding the polypeptides described herein; comprising polypeptides described herein (e.g., SEQ ID NO: 2); and/or comprising other splicing variants encoded by an NR; and/or an agent that alters (e.g., enhances or inhibits) NR gene expression or NR polypeptide activity as described herein.
  • nucleic acids described herein particularly nucleotides encoding the polypeptides described herein; comprising polypeptides described herein (e.g., SEQ ID NO: 2); and/or comprising other splicing variants encoded by an NR; and/or an agent that alters (e.g., enhances or inhibits) NR gene expression or NR polypeptide activity as described herein.
  • a polypeptide, protein e.g., a G-protein
  • an agent that alters NR gene expression or an NR binding agent or binding partner, fragment, fusion protein or prodrug thereof, or a nucleotide or nucleic acid construct (vector) comprising a nucleotide of the present invention, or an agent that alters NR polypeptide activity
  • a physiologically acceptable carrier or excipient can be formulated with a physiologically acceptable carrier or excipient to prepare a pharmaceutical composition.
  • the carrier and composition can be sterile. The formulation should suit the mode of administration.
  • Suitable pharmaceutically acceptable carriers include but are not limited to water, salt solutions (e.g., NaCl), saline, buffered saline, alcohols, glycerol, ethanol, gum arabic, vegetable oils, benzyl alcohols, polyethylene glycols, gelatin, carbohydrates such as lactose, amylose or starch, dextrose, magnesium stearate, talc, silicic acid, viscous paraffin, perfume oil, fatty acid esters, hydroxymethylcellulose, polyvinyl pyrolidone, etc., as well as combinations thereof.
  • the pharmaceutical preparations can, if desired, be mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, flavoring and/or aromatic substances and the like which do not deleteriously react with the active agents.
  • auxiliary agents e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, flavoring and/or aromatic substances and the like which do not deleteriously react with the active agents.
  • the composition can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • the composition can be a liquid solution, suspension, emulsion, tablet, pill, capsule, sustained release formulation, or powder.
  • the composition can be fonnulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, polyvinyl pyrolhdone, sodium saccharine, cellulose, magnesium carbonate, etc.
  • compositions of introduction of these compositions include, but are not limited to, intradermal, intramuscular, intraperitoneal, intraocular, intravenous, subcutaneous, topical, oral and intranasal.
  • Other suitable methods of introduction can also include gene therapy (as described below), rechargeable or biodegradable devices, particle acceleration devises ("gene guns") and slow release polymeric devices.
  • the pharmaceutical compositions of this invention can also be administered as part of a combinatorial therapy with other agents.
  • compositions for intravenous administration typically are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampule or sachette indicating the quantity of active agent.
  • the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water, saline or dextrose/water.
  • an ampule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • nonsprayable forms viscous to semi-solid or solid forms comprising a carrier compatible with topical application and having a dynamic viscosity preferably greater than water
  • Suitable formulations include but are not limited to solutions, suspensions, emulsions, creams, ointments, powders, enemas, lotions, sols, liniments, salves, aerosols, etc., which are, if desired, sterilized or mixed with auxiliary agents, e.g., preservatives, stabilizers, wetting agents, buffers or salts for influencing osmotic pressure, etc.
  • auxiliary agents e.g., preservatives, stabilizers, wetting agents, buffers or salts for influencing osmotic pressure, etc.
  • the agent may be incorporated into a cosmetic formulation.
  • sprayable aerosol preparations wherein the active ingredient, preferably in combination with a solid or liquid inert carrier material, is packaged in a squeeze bottle or in admixture with a pressurized volatile, normally gaseous propellant, e.g., pressurized air.
  • a pressurized volatile, normally gaseous propellant e.g., pressurized air.
  • Agents described herein can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2- ethylamino ethanol, histidine, procaine, etc.
  • the agents are administered in a therapeuticaUy effective amount.
  • the amount of agents which will be therapeuticaUy effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques.
  • in vitro or in vivo assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the symptoms of a susceptibility to a disease or condition associated with an NR, and should be decided according to the judgment of a practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use of sale for human administration.
  • the pack or kit can be labeled with information regarding mode of administration, sequence of drug administration (e.g., separately, sequentially or concurrently), or the like.
  • the pack or kit may also include means for reminding the patient to take the therapy.
  • the pack or kit can be a single unit dosage of the combination therapy or it can be a plurality of unit dosages, hi particular, the agents can be separated, mixed together in any combination, present in a single vial or tablet. Agents assembled in a blister pack or other dispensing means is preferred.
  • unit dosage is intended to mean a dosage that is dependent on the individual phannacodynamics of each agent and administered in FDA approved dosages in standard time courses.
  • the present invention also pertains to methods of treatment (prophylactic and/or therapeutic) for a susceptibility to a disease or condition associated with an NR, using an NR therapeutic agent.
  • NR therapeutic agent is an agent that alters (e.g., enhances or inhibits) NR polypeptide activity and/or NR gene expression, as described herein (e.g., an NR agonist or antagonist).
  • NR therapeutic agents can alter NR polypeptide activity or gene expression by a variety of means, such as, for example, by providing additional NR polypeptide or by upregulating the transcription or translation of the NR gene; by altering posttranslational processing of the NR polypeptide; by altering transcription of NR splicing variants; or by interfering with NR polypeptide activity (e.g. , by binding to an NR polypeptide), or by downregulating the transcription or translation of an NR gene.
  • NR therapeutic agents include the following: nucleic acids or fragments or derivatives thereof described herein, particularly nucleotides encoding the polypeptides described herein and vectors comprising such nucleic acids (e.g., a gene, cDNA, and/or mRNA, such as a nucleic acid encoding an NR polypeptide or active fragment or derivative thereof, or an oligonucleotide; for example, one of SEQ ID NO: 1, or a complement thereof, or a nucleic acid encoding SEQ ID NOS:3-4, or fragments or derivatives thereof); polypeptides described herein (e.g., SEQ JD NO: 2, and/or other splicing variants encoded by an NR, or fragments or derivatives thereof); other polypeptides (e.g., G-proteins); NR binding agents; peptidomimetics; fusion proteins or prodrugs thereof; antibodies (e.g., an antibody to a mutant NR polypeptide, or an antibody
  • An NR therapeutic agent that is a nucleic acid is used in the treatment of a susceptibility to a disease or condition associated with an NR.
  • treatment refers not only to ameliorating symptoms associated with the disease, but also preventing or delaying the onset of the disease, and also lessening the severity or frequency of symptoms of the disease.
  • the therapy is designed to alter (e.g., inhibit or enhance), replace or supplement activity of anNR polypeptide in an individual.
  • an NR therapeutic agent can be administered in order to upregulate or increase the expression or availability of the NR gene or of specific splicing variants of NR, or, conversely, to downregulate or decrease the expression or availability of the NR gene or specific splicing variants of the NR.
  • Upregulation or increasing expression or availability of a native NR gene or of a particular splicing variant could interfere with or compensate for the expression or activity of a defective gene or another splicing variant;
  • downregulation or decreasing expression or availability of a native NR gene or of a particular splicing variant could minimize the expression or activity of a defective gene or the particular splicing variant and thereby minimize the impact of the defective gene or the particular splicing variant.
  • the NR therapeutic agent(s) are administered in a therapeuticaUy effective amount (i.e., an amount that is sufficient to treat the disease, such as by ameliorating symptoms associated with the disease, preventing or delaying the onset of the disease, and/or also lessening the severity or frequency of symptoms of the disease).
  • a therapeuticaUy effective amount i.e., an amount that is sufficient to treat the disease, such as by ameliorating symptoms associated with the disease, preventing or delaying the onset of the disease, and/or also lessening the severity or frequency of symptoms of the disease.
  • the amount which will be therapeuticaUy effective in the treatment of a particular individual's disorder or condition will depend on the symptoms and severity of the disease, and can be determined by standard clinical techniques.
  • in vitro or in vivo assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of a practitioner
  • a nucleic acid of the invention e.g., a nucleic acid encoding an NR polypeptide, such as SEQ ID NO: 1, or a complement thereof; or another nucleic acid that encodes an NR polypeptide or a splicing variant, derivative or fragment thereof, such as a nucleic acid encoding SEQ ID NOS: 1-2
  • a nucleic acid of the invention can be used, either alone or in a pharmaceutical composition as described above.
  • an NR or a cDNA encoding an NR polypeptide can be introduced into cells (either in vitro or in vivo) such that the cells produce native NR polypeptide.
  • cells that have been transformed with the gene or cDNA or a vector comprising the gene or cDNA can be introduced (or re-introduced) into an individual affected with the disease.
  • cells which, in nature, lack native NR expression and activity, or have mutant NR expression and activity, or have expression of a disease-associated NR splicing variant can be engineered to express the NR polypeptide or an active fragment of the NR polypeptide (or a different variant of the NR polypeptide).
  • nucleic acid encoding an NR polypeptide, or an active fragment or derivative thereof can be introduced into an expression vector, such as a viral vector, and the vector can be introduced into appropriate cells in an animal.
  • an expression vector such as a viral vector
  • Other gene transfer systems including viral and nonviral transfer systems, can be used.
  • nonviral gene transfer methods such as calcium phosphate coprecipitation, mechanical techniques (e.g., microinjection); membrane fusion- mediated transfer via liposomes; or direct DNA uptake, can also be used.
  • a nucleic acid of the invention in another embodiment, can be used in "antisense" therapy, in which a nucleic acid (e.g., an oligonucleotide) which specifically hybridizes to the mRNA and/or genomic DNA of an NR is administered or generated in situ.
  • a nucleic acid e.g., an oligonucleotide
  • the antisense nucleic acid that specifically hybridizes to the mRNA and/or DNA inhibits expression of the NR polypeptide, e.g., by inhibiting translation and/or transcription.
  • Binding of the antisense nucleic acid can be by conventional base pair complementarity, or, for example, in the case of binding to DNA duplexes, through specific interaction in the major groove of the double helix.
  • An antisense construct of the present invention can be delivered, for example, as an expression plasmid as described above. When the plasmid is transcribed in the cell, it produces RNA which is complementary to a portion of the mRNA and/or DNA which encodes the NR polypeptide.
  • the antisense construct can be an oligonucleotide probe which is generated ex vivo and introduced into cells; it then inhibits expression by hybridizing with the mRNA and/or genomic DNA of the NR.
  • the oligonucleotide probes are modified oligonucleotides which are resistant to endogenous nucleases, e.g. exonucleases and/or endonucleases, thereby rendering them stable in vivo.
  • exemplary nucleic acid molecules for use as antisense oligonucleotides are phosphoramidate, phosphothioate and methylphosphonate analogs of DNA (see also U.S. Pat. NO. 5,176,996; 5,264,564; and 5,256,775).
  • oligodeoxyribonucleotides derived from the translation initiation site are preferred.
  • oligonucleotides mRNA, cDNA or DNA
  • the antisense oligonucleotides bind to NR mRNA transcripts and prevent translation. Absolute complementarity, although preferred, is not required.
  • a sequence "complementary" to a portion of an RNA indicates that a sequence has sufficient complementarity to be able to hybridize with the RNA, forming a stable duplex; in the case of double-stranded antisense nucleic acids, a single strand of the duplex DNA may thus be tested, or triplex formation may be assayed.
  • the ability to hybridize will depend on both the degree of complementarity and the length of the antisense nucleic acid, as described in detail above. Generally, the longer the hybridizing nucleic acid, the more base mismatches with an RNA it may contain and still form a stable duplex (or triplex, as the case may be).
  • One skilled in the art can ascertain a tolerable degree of mismatch by use of standard procedures.
  • the oligonucleotides used in antisense therapy can be DNA, RNA, or chimeric mixtures or derivatives or modified versions thereof, single-stranded or double-stranded.
  • the oligonucleotides can be modified at the base moiety, sugar moiety, or phosphate backbone, for example, to improve stability of the molecule, hybridization, etc.
  • the oligonucleotides can include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger et al, Proc. Natl. Acad. Sci.
  • the oligonucleotide maybe conjugated to another molecule (e.g., a peptide, hybridization triggered cross-linking agent, transport agent, hybridization-triggered cleavage agent).
  • the antisense molecules are delivered to cells which express NR in vivo.
  • a number of methods can be used for delivering antisense DNA or RNA to cells; e.g., antisense molecules can be injected directly into the tissue site, or modified antisense molecules, designed to target the desired cells (e.g., antisense linked to peptides or antibodies that specifically bind receptors or antigens expressed on the target cell surface) can be administered systematically.
  • a recombinant DNA construct is utilized in which the antisense oligonucleotide is placed under the control of a strong promoter (e.g., pol JJI or pol JJ).
  • a vector can be introduced in vivo such that it is taken up by a cell and directs the transcription of an antisense RNA.
  • Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired antisense RNA.
  • Such vectors can be constructed by recombinant DNA technology methods standard in the art and described above.
  • a plasmid, cosmid, YAC or viral vector can be used to prepare the recombinant DNA construct which can be introduced directly into the tissue site.
  • viral vectors can be used which selectively infect the desired tissue, in which case administration may be accomplished by another route (e.g., systemically).
  • Endogenous NR expression can also be reduced by inactivating or "knocking out" NR or its promoter using targeted homologous recombination (e.g., see Smithies et al, Nature 317:230-234 (1985); Thomas & Capecchi, Cell 51:503-512 (1987); Thompson et al, Cell 5:313-321 (1989)).
  • a mutant, non-functional NR (or a completely unrelated DNA sequence) flanked by DNA homologous to the endogenous NR can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells that express the NR in vivo.
  • the recombinant DNA constructs can be directly administered or targeted to the required site in vivo using appropriate vectors, as described above.
  • expression of non-mutant NRs can be increased using a similar method: targeted homologous recombination can be used to insert a DNA construct comprising a non- mutant, functional NR, e.g., a gene having SEQ ID NO: 1, or the complement thereof, or a portion thereof, in place of a mutant NR in the cell, as described above, hi another embodiment, targeted homologous recombination can be used to insert a DNA construct comprising a nucleic acid that encodes an NR polypeptide variant that differs from that present in the cell.
  • endogenous NR expression can be reduced by targeting deoxyribonucleotide sequences complementary to the regulatory region of an NR (i.e., the NR promoter and/or enhancers) to form triple helical structures that prevent transcription of the NR in target cells in the body.
  • NR i.e., the NR promoter and/or enhancers
  • triple helical structures that prevent transcription of the NR in target cells in the body.
  • the antisense constructs described herein by antagonizing the normal biological activity of one of the NR proteins, can be used in the manipulation of tissue, e.g., tissue differentiation, both in vivo and for ex vivo tissue cultures.
  • tissue e.g., tissue differentiation
  • the anti-sense techniques e.g., microinjection of antisense molecules, or transfection with plasmids whose transcripts are anti-sense with regard to an NR mRNA or gene sequence
  • Such techniques can be utilized in cell culture, but can also be used in the creation of transgenic animals.
  • NR therapeutic agents as described herein can also be used in the treatment or prevention of a susceptibility to a disease or condition associated with an NR.
  • the therapeutic agents can be delivered in a composition, as described above, or by themselves. They can be administered systemically, or can be targeted to a particular tissue.
  • the therapeutic agents can be produced by a variety of means, including chemical synthesis; recombinant production; in vivo production (e.g., a transgenic animal, such as U.S. Pat. No. 4,873,316 to Meade et ⁇ l), for example, and can be isolated using standard means such as those described herein.
  • a combination of any of the above methods of treatment e.g., administration of non-mutant NR polypeptide in conjunction with antisense therapy targeting mutant NR mRNA; administration of a first splicing variant encoded by an NR in conjunction with antisense therapy targeting a second splicing encoded by an NR
  • administration of non-mutant NR polypeptide in conjunction with antisense therapy targeting mutant NR mRNA can also be used.

Abstract

Nucleic acids encoding nuclear receptors, and methods of using same are disclosed.

Description

NUCLEIC ACIDS ENCODING NUCLEAR RECEPTORS
RELATED APPLICATIONS
This application is a continuation-in-part application of and claims priority to U.S Provisional Application 60/302,452, filed June 29, 2001 and to U.S. Provisional Application 60/332,976, filed November 6, 2001, the entire teachings of which are incorporated herein by reference.
BACKGROUND OF THE INVENTION
Nuclear receptors ("NRs") are a superfamily of ligand-activated transcription factors that mediate the transmission of extracellular signals into the cell to produce a cellular response. Unlike integral membrane receptors and membrane associated receptors, the nuclear receptors reside in either the cytoplasm or nucleus of eukaryotic cells. NRs recognize similar DNA sequences that contain two or more nucleotide DNA-binding half-sites arranged as direct repeats or inverted repeats. The receptors bind to specific DNA sequences and control gene transcription in the nucleus. Many nuclear receptors undergo a major conformational change upon binding a specific ligand. They regulate the assembly of a transcriptional preinitiation complex at the promoter of target genes and modulate their expression in response to ligand. In particular, NRs repress or stimulate transcription by recruiting corepressor or coactivatior proteins, in addition to directly contacting the basal transcription machinery.
NRs are involved in an enormous range of biological processes, and have been found to play roles in eontrolling development, differentiation and physiological function and homeostasis. The NR superfamily includes receptors for steriod hormones, such as estrogen, androgen, progesterone, hepatocyte nuclear factor 4, thyroid hormone, hormonal forms of viamin A and D, retinoids, peroxisomal activators, mineralocoricoids, glucocorticoids, ecdysone and many others including orphan receptors whose ligand have not been identified. SUMMARY OF THE INVENTION
The present invention relates to nuclear receptor genes, particularly nucleic acids comprising NR genes, and the amino acids encoded by such nucleic acids. One such sequence is shown in Table I. In Table I, the NR entry lists the name (e.g., "MOOSE04608"), the University of California at Santa Cruz contig designation from which the sequence was analyzed (e.g., "ctgl7328"), the exon locations (e.g., "3473973.3474369, 3497170 . . ."), followed by the amino acid sequence and the nucleic acid sequence.
Sub-family information on the sequence is shown in Table π. For the sequence, the following information is provided: the University of California at Santa Cruz contig designation from which the sequence was analyzed (e.g., "ctgl7328"), the name (e.g., "MOOSE04608"), and the subfamily to which the sequence appears to belong. The assignments were made on the basis of the best E- value with which the sequence aligned. hi one embodiment, the isolated nucleic acid molecule comprises a nucleotide sequence of SEQ ID NO: 1, as shown in Table I, and the complements thereof. The invention further relates to a nucleic acid molecule which hybridizes under high stringency conditions to a nucleotide sequence of SEQ ID NO: 1, as shown in Table I, and the complements thereof. The invention additionally relates to isolated nucleic acid molecules (e.g., cDNA molecules) encoding an NR polypeptide (e.g., encoding a polypeptide of SEQ ID NO: 2, as shown in Table I).
The invention further provides a method for assaying a sample for the presence of a nucleic acid molecule comprising all or a portion of an NR in a sample, comprising contacting said sample with a second nucleic acid molecule comprising a nucleotide sequence encoding an NR polypeptide (e.g., one of SEQ ID NO: 1, as shown in Table I, or the complement of one of SEQ ID NO: 1; a nucleotide sequence encoding SEQ ID NO: 2, as shown in Table I), or a fragment or derivative thereof, under conditions appropriate for selective hybridization. The invention additionally provides a method for assaying a sample for the level of expression of an NR polypeptide, or fragment or derivative thereof, comprising detecting (directly or indirectly) the level of expression of the NR polypeptide, fragment or derivative thereof.
The invention also relates to a vector comprising an isolated nucleic acid molecule of the invention operatively linked to a regulatory sequence, as well as to a recombinant host cell comprising the vector. The invention also provides a method for preparing a polypeptide encoded by an isolated nucleic acid molecule described herein (an NR polypeptide), comprising culturing a recombinant host cell of the invention under conditions suitable for expression of said nucleic acid molecule. The invention further provides an isolated polypeptide encoded by isolated nucleic acid molecules of the invention (e.g., NR polypeptide), as well as fragments or derivatives thereof. In a particular embodiment, the polypeptide comprises an amino acid sequence of SEQ ID NO: 2, as shown in Table I. The invention also relates to an isolated polypeptide comprising an amino acid sequence which is greater than about 90 percent identical to an amino acid sequence of SEQ ID NO: 2, preferably about 95, 96, 97, 98 or 99 percent identical.
The invention also relates to an antibody, or an antigen-binding fragment thereof, which selectively binds to a polypeptide of the invention, as well as to a method for assaying the presence of a polypeptide encoded by an isolated nucleic acid molecule of the invention in a sample, comprising contacting said sample with an antibody which specifically binds to the encoded polypeptide.
The invention further relates to methods of diagnosing a predisposition to a condition mediated by NRs. The methods of diagnosing such a predisposition in an individual include detecting the presence of a mutation in NR, as well as detecting alterations in expression of an NR polypeptide, such as the presence of different splicing variants of NR polypeptides. The alterations in expression can be quantitative, qualitative, or both quantitative and qualitative.
The invention additionally relates to an assay for identifying agents which alter (e.g., enhance or inhibit) the activity or expression of one or more NR polypeptides. For example, a cell, cellular fraction, or solution containing an NR polypeptide or a fragment or derivative thereof, can be contacted with an agent to be tested, and the level of NR polypeptide expression or activity can be assessed. The activity or expression of more than one NR polypeptides can be assessed concurrently (e.g., the cell, cellular fraction, or solution can contain more than one type of NR polypeptide, such as different splicing variants, and the levels of the different polypeptides or splicing variants can be assessed). hi another embodiment, the invention relates to assays to identify polypeptides which interact with one or more NR polypeptides. In a yeast two- hybrid system, for example, a first vector is used which includes a nucleic acid encoding a DNA binding domain and also an NR polypeptide, splicing variant, or fragment or derivative thereof, and a second vector is used which includes a nucleic acid encoding a transcription activation domain and also a nucleic acid encoding a polypeptide which potentially may interact with the NR polypeptide, splicing variant, or fragment or derivative thereof (e.g., an NR polypeptide binding agent or receptor). Incubation of yeast containing both the first vector and the second vector under appropriate conditions allows identification of polypeptides which interact with the NR polypeptide or fragment or derivative thereof, and thus can be agents which alter the activity of expression of an NR polypeptide.
Agents that enhance or inhibit NR polypeptide expression or activity are also included in the current invention, as are methods of altering (enhancing or inhibiting) NR polypeptide expression or activity by contacting a cell containing NR and/or polypeptide, or by contacting the NR polypeptide, with an agent that enhances or inhibits expression or activity of NR or polypeptide.
Additionally, the invention pertains to pharmaceutical compositions comprising the nucleic acids of the invention, the polypeptides of the invention, and or the agents that alter activity of NR polypeptide. The invention further pertains to methods of treating conditions mediated by NRs, by administering NR therapeutic agents, such as nucleic acids of the invention, polypeptides of the invention, the agents that alter activity of NR polypeptide, or compositions comprising the nucleic acids, polypeptides, and/or the agents that alter activity of NR polypeptide. DETATLED DESCRIPTION OF THE INVENTION
The present invention relates to nucleic acids comprising nuclear receptors ("NRs"), and the NR amino acids encoded by those nucleic acids.
The NR superfamily includes the receptors for many important signaling pathways, including, but not limited to, hormone receptors, steroid hormone receptors, estrogen receptors, angrogen receptors, progesterone receptors, growth factors receptors, orphan nuclear retinoid receptors, pregnane X receptors, and others.
These receptors are involved in the treatment of infections and various diseases and conditions, including, but not limited to, cancers, primary biliary cirrhosis, inflammatory bowel disease, type 1 diabetes, malignant melanoma, rheumatoid arthritis, neuropsychiatric disorders, abnormal bone resorption, cardiovascular disease, central nervous system disorders, glaucoma, ocular hypertension, fungal infections, as well as others. With the availability of complete genomic sequences for many organisms today, including Homo sapiens, it has become clear that there is a need for data mining techniques to extract the information in them, e.g., gene prediction programs. Of these, the most successful ones are those based on the comparison of known protein or protein-derived information, or those that use expressed sequence tags (ESTs) to predict gene location and structure.
One such algorithm is Gene Wise. It bases its exon prediction on the use of Hidden Markov Models (HMMs) of proteins to be compared against a genomic sequence, so that the translation of the sequence will match the model in a similar way to other HMM profile searches (Eddy, Curr. Opin. Struct. Biol 6(3):361-5, 1996), and allowing the presence of long insertions as long as they include donor and acceptor site sequences at both ends.
To take advantage of the algorithm, the models for different protein families must be built so that they represent the full-length sequences instead of the most common features in them. This is a major difference with existing HMM databases such as Pfam (Sonnhammer et al, Proteins 28(3):405-20, 1997), in which each model is built to represent a family of proteins as broad as possible with minimum overlap between them.
In the present approach, the sequences were subdivided in several families so that the similarity inside of a group of them was over 50%. Given this approach, there are several points of overlap between different families when analyzing a sequence, so the discrimination must be done after the search is completed.
Several resources that include expert-supervised classifications are used to select the best groups of sequences, e.g., the NRdb (Horn et al, Nucleic Acids Res. 26(l):275-9, 1998), PKR (Smith et al, Trends Biochem. Sci. 22(l l):444-6, 1997), NuclearRdb (Horn et al, Nucleic Acids Res. 29:346-349, 2001), IOCH (Le Novere et al, Nucleic Acids Res. 27(l):340-2, 1999), Enzyme (Bairoch, Nucleic Acids Res. 28:304-305, 2000) and Swiss-Prot (Bairoch et al, Nucleic Acids Res. 28:45-48, 2000). When none is available, or the sequences included in some groups are too disrinatly related, the grouping must be done manually, using the ClustalW (Thompson et al, Nucleic Acids Res. 22:4673-4680, 1994) package to measure the distance between different sequences.
The present model was built from multiple sequence alignments of the different protein families obtained with DiAlign 2 (Morgenstern, Bioinformatics 15(3):211-8, 1999). DiAlign works based on segment-to-segment comparisons instead of arbitrary thresholds for gap opening and extension, which makes it ideally suited for building models that represent an entire, full-length sequence, since the alignments built this way have more match states that would be assigned as insertion states when using other alignment algorithms. The models were built using the standard HMMer package. To search for new genes, a genome-wide scan was done on the University of
California at Santa Cruz sequences, using the Gene Wise algorithm. It translates the genomic sequence on the fly to proteins and can therefore maintain a reading frame through insertions and deletions. The algorithm also rewards gaps in the genomic sequence relative to the model if they are encapsulated within introns, like splice structure. For each superfamily of proteins, a classification was obtained in which the sequences are grouped by length and similarity. Each one of these groups was then used to build a HMM profile representing this group of sequences. This approach aims to have models that can represent the full length of the encoded proteins for a whole range of proteins, without being too specific for any one of them or being too general, as would be a HMM built for large groups of sequences. This classification was based either on existing expert-supervised classifications, or by retrieval of sequences and classification based on pairwise alignment distances.
These models were then searched against the October 2000 Fixed Release (with its subsequent corrections) and the April 2001 Fixed Release of the Santa Cruz contigs using the Paracel GeneMatcher+ Hardware Accelerator with the Gene Wise algorithm. The sequences were chopped at 100 Kb with an overlap of 1 Kb. Each one of the superfamilies required between 3 and 6 days to complete and generate results. The results represent the coding regions of the complete final protein as it is found in the organism.
The cross-validation of the results was done in two steps. First, all of the hits with an E- value lower than 10" that do not overlap with one another were selected, and in the event of overlapping, the one with lowest E- value was selected. After selecting all of those matches, the DNA sequences were compared against the RefSeq database (Pruitt et al, Trends Genet. 16(l):44-47, 2000) using BLAST (Altschul et al. Nucleic Acids Res. 25:3389-3402, 1997).
Over 80% of the sequences were 90% or more identical to an existing human RefSeq entry and/or mRNA from GenBank. The differences are usually due to picking the wrong model for a certain sequence that appears as a hit more than once in different famihes, being a different valid splice variant, which can be tested by comparing to the EST database, or by addition of a small last exon to complete the match instead of accept an stop codon in a previous one. hi all of such cases, the results are easily and quickly corrected by eye. Very rarely the algorithm will actually make a wrong prediction, which is consistent with the expected behaviour (Guigo et al, Genome Res. 10(10):1631-42, 2000). Of the remaining sequences, over 50% have a match over 90% identical in the public domain protein databases, and the differences between those sequences in the databases and the potential translations is basically the same as the differences between the DNA sequences and the RefSeq entries. The full sequence of NR genes are shown in Table I as SEQ ID NO: 1. The amino acids encoded by these nucleic acids are shown in Table I as SEQ ID NO: 2.
A number of the genes were found to be linked with markers known to be associated with human diseases genes. These are shown in Table DI. The diseases were linked to the HMM genes in the following manner: (1) the HMM gene models were compared to the consensus of the human genome sequence, located and the results kept in a relational database; (2) all possible markers (Sequence Tagged Sites (STS's)) (public or deCODE genetics) are also located in the same consensus using ePCR or BLAT and results kept in a relational database; and (3) LOD scores for diseases are linked to markers. A span of one LOD drop around the marker was also given. A computer program takes each LOD peak and links it to the consensus through the markerhit in the database. The database is then queried for all HMM genes within the span of one LOD drop or a mimmum of 15 Mb in each direction from the marker. The output is the name of the peak marker and its distance to the HMM gene.
NUCLEIC ACIDS OF THE INVENTION
Accordingly, the invention pertains to isolated nucleic acid molecules comprising human NR genes. The term, "NR", as used herein, refers to an isolated nucleic acid molecule shown in Table I, and consisting of SEQ ID NO: 1, and also to a portion or fragment of the isolated nucleic acid molecule (e.g., cDNA or the gene) that encodes NR polypeptide (e.g. , a polypeptide selected from the group shown in Table I, and consisting of SEQ ID NO: 2). hi a preferred embodiment, the isolated nucleic acid molecule comprises a nucleic acid molecule of SEQ ID NO: 1 or the complement of such a nucleic acid molecule.
The isolated nucleic acid molecules of the present invention can be RNA, for example, mRNA, or DNA, such as cDNA and genomic DNA. DNA molecules can be double-stranded or single-stranded; single stranded RNA or DNA can be either the coding, or sense, strand or the non-coding, or antisense, strand. The nucleic acid molecule can include all or a portion of the coding sequence of the gene and can further comprise additional non-coding sequences such as introns and non-coding 3' and 5' sequences (including regulatory sequences, for example). Additionally, the nucleic acid molecule can be fused to a marker sequence, for example, a sequence that encodes a polypeptide to assist in isolation or purification of the polypeptide. Such sequences include, but are not limited to, those which encode a glutathione-S-transferase (GST) fusion protein and those which encode a hemagglutinin A (HA) polypeptide marker from influenza.
An "isolated" nucleic acid molecule, as used herein, is one that is separated from nucleic acids which normally flank the gene or nucleotide sequence (as in genomic sequences) and/or has been completely or partially purified from other transcribed sequences (e.g., as in an RNA library). For example, an isolated nucleic acid of the invention may be substantially isolated with respect to the complex cellular milieu in which it naturally occurs, or culture medium when produced by recombinant techniques, or chemical precursors or other chemicals when chemically synthesized, hi some instances, the isolated material will form part of a composition (for example, a crude extract containing other substances), buffer system or reagent mix. h other circumstances, the material may be purified to essential homogeneity, for example as determined by PAGE or column chromatography such as HPLC. Preferably, an isolated nucleic acid molecule comprises at least about 50, 80 or 90% (on a molar basis) of all macromolecular species present. With regard to genomic DNA, the term "isolated" also can refer to nucleic acid molecules which are separated from the chromosome with which the genomic DNA is naturally associated. For example, the isolated nucleic acid molecule can contain less than about 5 kb, 4 kb, 3 kb, 2 kb, 1 kb, 0.5 kb or 0.1 kb of nucleotides which flank the nucleic acid molecule in the genomic DNA of the cell from which the nucleic acid molecule is derived. The nucleic acid molecule can be fused to other coding or regulatory sequences and still be considered isolated. Thus, recombinant DNA contained in a vector is included in the definition of "isolated" as used herein. Also, isolated nucleic acid molecules include recombinant DNA molecules in heterologous host cells, as well as partially or substantially purified DNA molecules in solution. "Isolated" nucleic acid molecules also encompass in vivo and in vitro RNA transcripts of the DNA molecules of the present invention. An isolated nucleic acid molecule or nucleotide sequence can include a nucleic acid molecule or nucleotide sequence which is synthesized chemically or by recombinant means. Therefore, recombinant DNA contained in a vector are included in the definition of "isolated" as used herein. Also, isolated nucleotide sequences include recombinant DNA molecules in heterologous organisms, as well as partially or substantially purified DNA molecules in solution. In vivo and in vitro RNA transcripts of the DNA molecules of the present invention are also encompassed by "isolated" nucleotide sequences. Such isolated nucleotide sequences are useful in the manufacture of the encoded polypeptide, as probes for isolating homologous sequences (e.g., from other mammalian species), for gene mapping (e.g. , by in situ hybridization with chromosomes), or for detecting expression of the gene in tissue (e.g., human tissue), such as by Northern blot analysis.
The present invention also pertains to nucleic acid molecules which are not necessarily found in nature but which encode an NR polypeptide (e.g., a polypeptide having an amino acid sequence comprising an amino acid sequence of SEQ ID NO: 2), or another splicing variant of an NR polypeptide or polymorphic variant thereof. Thus, for example, DNA molecules which comprise a sequence that is different from the naturally-occurring nucleotide sequence but which, due to the degeneracy of the genetic code, encode an NR polypeptide of the present invention are also the subject of this invention. The invention also encompasses nucleotide sequences encoding portions (fragments), or encoding variant polypeptides such as analogues or derivatives of an NR polypeptide. Such variants can be naturally-occurring, such as in the case of allelic variation or single nucleotide polymorphisms, or non- naturally-occurring, such as those induced by various mutagens and mutagenic processes. Intended variations include, but are not limited to, addition, deletion and substitution of one or more nucleotides which can result in conservative or non- conservative amino acid changes, including additions and deletions. Preferably the nucleotide (and/or resultant amino acid) changes are silent or conserved; that is, they do not alter the characteristics or activity of an NR polypeptide. In one preferred embodiment, the nucleotide sequences are fragments that comprise one or more polymorphic microsatellite markers, hi another preferred embodiment, the nucleotide sequences are fragments that comprise one or more single nucleotide polymorphisms in an NR gene.
Other alterations of the nucleic acid molecules of the invention can include, for example, labeling, methylation, internucleotide modifications such as uncharged linkages (e.g. , methyl phosphonates, phosphotriesters, phosphoamidates, carbamates), charged linkages (e.g., phosphorothioates, phosphorodithioates), pendent moieties (e.g., polypeptides), intercalators (e.g., acridine, psoralen), chelators, alkylators, and modified linkages (e.g., alpha anomeric nucleic acids). Also included are synthetic molecules that mimic nucleic acid molecules in the ability to bind to a designated sequences via hydrogen bonding and other chemical interactions. Such molecules include, for example, those in which peptide linkages substitute for phosphate linkages in the backbone of the molecule.
The invention also pertains to nucleic acid molecules which hybridize under high stringency hybridization conditions, such as for selective hybridization, to a nucleotide sequence described herein (e.g., nucleic acid molecules which specifically hybridize to a nucleotide sequence encoding polypeptides described herein, and, optionally, have an activity of the polypeptide). In one embodiment, the invention includes variants described herein which hybridize under high stringency hybridization conditions (e.g., for selective hybridization) to a nucleotide sequence comprising a nucleotide sequence of SEQ ID NO: 1. hi another embodiment, the invention includes variants described herein which hybridize under high stringency hybridization conditions (e.g., for selective hybridization) to a nucleotide sequence encoding an amino acid sequence of SEQ ID NO: 2 or a polymorphic variant thereof, hi a preferred embodiment, the variant which hybridizes under high stringency hybridizations has an activity of an NR. Such nucleic acid molecules can be detected and/or isolated by specific hybridization (e.g., under high stringency conditions). "Specific hybridization," as used herein, refers to the ability of a first nucleic acid to hybridize to a second nucleic acid in a manner such that the first nucleic acid does not hybridize to any nucleic acid other than to the second nucleic acid (e.g. , when the first nucleic acid has a higher similarity to the second nucleic acid than to any other nucleic acid in a sample wherein the hybridization is to be performed). "Stringency conditions" for hybridization is a term of art which refers to the incubation and wash conditions, e.g., conditions of temperature and buffer concentration, which permit hybridization of a particular nucleic acid to a second nucleic acid; the first nucleic acid may be perfectly (i.e., 100%) complementary to the second, or the first and second may share some degree of complementarity which is less than perfect (e.g., 70%, 75%, 85%, 90%, 95%). For example, certain high stringency conditions can be used which distinguish perfectly complementary nucleic acids from those of less complementarity. "High stringency conditions", "moderate stringency conditions" and "low stringency conditions" for nucleic acid hybridizations are explained on pages 2.10.1-2.10.16 and pages 6.3.1-6.3.6 in Current Protocols in Molecular Biology (Ausubel, F.M. et al, "Current Protocols in Molecular Biology", John Wiley & Sons, 1998 and 2001), the entire teachings of which are incorporated by reference herein). The exact conditions which determine the stringency of hybridization depend not only on ionic strength (e.g., 0.2X SSC, 0.1X SSC), temperature (e.g., room temperature, 42°C, 68°C) and the concentration of destabilizing agents such as formamide or denaturing agents such as SDS, but also on factors such as the length of the nucleic acid sequence, base composition, percent mismatch between hybridizing sequences and the frequency of occurrence of subsets of that sequence within other non-identical sequences. Thus, equivalent conditions can be determined by varying one or more of these parameters while maintaining a similar degree of identity or similarity between the two nucleic acid molecules. Typically, conditions are used such that sequences at least about 60%, at least about 70%, at least about 80%o, at least about 90% or at least about 95%> or more identical to each other remain hybridized to one another. By varying hybridization conditions from a level of stringency at which no hybridization occurs to a level at which hybridization is first observed, conditions which will allow a given sequence to hybridize (e.g., selectively) with the most similar sequences in the sample can be determined. Exemplary conditions are described in Krause, M.H. and S.A. Aaronson,
Methods in Enzymology 200:546-556, 1991, and in, Ausubel, et al, "Current Protocols in Molecular Biology" , John Wiley & Sons, 1998 and 2002, which describes the determination of washing conditions for moderate or low stringency conditions. Washing is the step in which conditions are usually set so as to determine a minimum level of complementarity of the hybrids. Generally, starting from the lowest temperature at which only homologous hybridization occurs, each °C by which the final wash temperature is reduced (holding SSC concentration constant) allows an increase by 1% in the maximum extent of mismatching among the sequences that hybridize. Generally, doubling the concentration of SSC results in an increase in Tm of ~ 17°C . Using these guidelines, the washing temperature can be determined empirically for high, moderate or low stringency, depending on the level of mismatch sought.
For example, a low stringency wash can comprise washing in a solution containing 0.2X SSC/0.1% SDS for 10 minutes at room temperature; a moderate stringency wash can comprise washing in a prewarmed solution (42°C) solution containing 0.2X SSC/0.1% SDS for 15 minutes at 42°C; and a high stringency wash can comprise washing in prewarmed (68°C) solution containing 0. IX SSC/0.1%SDS for 15 minutes at 68°C. Furthermore, washes can be performed repeatedly or sequentially to obtain a desired result as known in the art. Equivalent conditions can be determined by varying one or more of the parameters given as an example, as known in the art, while maintaining a similar degree of identity or similarity between the target nucleic acid molecule and the primer or probe used.
The percent identity of two nucleotide or amino acid sequences can be determined by aligning the sequences for optimal comparison purposes (e.g., gaps can be introduced in the sequence of a first sequence). The nucleotides or amino acids at corresponding positions are then compared, and the percent identity between the two sequences is a function of the number of identical positions shared by the sequences (i.e., % identity = # of identical positions/total # of positions x 100). h certain embodiments, the length of a sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 60%, and even more preferably at least 70%, 80%, 90% or 95% of the length of the reference sequence. The actual comparison of the two sequences can be accomplished by well-known methods, for example, using a mathematical algorithm. A preferred, non-limiting example of such a mathematical algorithm is described in Karlin et al, Proc. Natl. Acad. Sci. USA 90:5873-5877, 1993. Such an algorithm is incorporated into the NBLAST and XBLAST programs (version 2.0) as described in Altschul et al,
Nucleic Acids Res. 25:389-3402, 1997. When utilizing BLAST and Gapped BLAST programs, the default parameters of the respective programs (e.g., NBLAST) can be used. In one embodiment, parameters for sequence comparison can be set at score=100, wordlength=12, or can be varied (e.g., W=5 or W=20). Another preferred, non-limiting example of a mathematical algorithm utilized for the comparison of sequences is the algorithm of Myers and Miller CABIOS 4(1): 11-17, 1998. Such an algorithm is incorporated into the ALIGN program (version 2.0) which is part of the GCG sequence alignment software package (Accelrys, Cambridge, UK). When utilizing the ALIGN program for comparing amino acid sequences, a PAM120 weight residue table, a gap length penalty of 12 , and a gap penalty of 4 can be used. Additional algorithms for sequence analysis are known in the art and include ADVANCE and ADAM as described in Torellis and Robotti, Comput. Appl Biosci. 10:3-5, 1994; and FASTA described in Pearson and Lipman Proc. Natl. Acad. Sci. USA 85:2444-8, 1988. h another embodiment, the percent identity between two amino acid sequences can be accomplished using the GAP program in the GCG software package using either a BLOSUM63 matrix or a PAM250 matrix, and a gap weight of 12, 10, 8, 6, or 4 and a length weight of 2, 3, or 4. hi yet another embodiment, the percent identity between two nucleic acid sequences can be accomplished using the GAP program in the GCG software package, using a gap weight of 50 and a length weight of 3. The present invention also provides isolated nucleic acid molecules that contain a fragment or portion that hybridizes under highly stringent conditions to a nucleotide sequence comprising a nucleotide sequence of SEQ ID NO: 1, or the complement of such a sequence, and also provides isolated nucleic acid molecules that contain a fragment or portion that hybridizes under highly stringent conditions to a nucleotide sequence encoding an amino acid sequence selected from SEQ ID NO: 2, or polymorphic variant thereof. The nucleic acid fragments of the invention are at least about 15, preferably at least about 18, 20, 23 or 25 nucleotides, and can be 30, 40, 50, 100, 200 or more nucleotides in length. Longer fragments, for example, 30 or more nucleotides in length, which encode antigenic polypeptides described herein are particularly useful, such as for the generation of antibodies as described below.
In a related aspect, the nucleic acid fragments of the invention are used as probes or primers in assays such as those described herein. "Probes" or "primers" are oligonucleotides that hybridize in a base-specific manner to a complementary strand of nucleic acid molecules. Such probes and primers include polypeptide nucleic acids, as described in Nielsen et al, Science 254:1497-1500, 1991.
Typically, a probe or primer comprises a region of nucleotide sequence that hybridizes to at least about 15, typically about 20-25, and more typically about 40, 50 or 75, consecutive nucleotides of a nucleic acid molecule comprising a contiguous nucleotide sequence of SEQ JD NO: 1, or the complement of such a sequence, or a sequence encoding an amino acid sequence selected from SEQ ID NO: 2, or polymorphic variant thereof, h preferred embodiments, a probe or primer comprises 100 or fewer nucleotides, preferably from 6 to 50 nucleotides, preferably from 12 to 30 nucleotides. In other embodiments, the probe or primer is at least 70% identical to the contiguous nucleotide sequence or to the complement of the contiguous nucleotide sequence, preferably at least 80% identical, more preferably at least 90% identical, even more preferably at least 95% identical, or even capable of selectively hybridizing to the contiguous nucleotide sequence or to the complement of the contiguous nucleotide sequence. Often, the probe or primer further comprises a label, e.g., radioisotope, fluorescent compound, enzyme, or enzyme co-factor. The nucleic acid molecules of the invention such as those described above can be identified and isolated using standard molecular biology techniques and the sequence information provided herein. For example, nucleic acid molecules can be amplified and isolated by the polymerase chain reaction using synthetic oligonucleotide primers designed based on one or more of the sequences of SEQ JJD NO: 1, or the complement of such a sequence, or designed based on nucleotides based on sequences encoding one or more of the amino acid sequences provided herein. See generally PCR Technology: Principles and Applications for DNA Amplification (ed. H.A. Erlich, Freeman Press, NY, NY, 1992); PCR Protocols: A Guide to Methods and Applications (Eds. Innis et al, Academic Press, San Diego, CA, 1990); Mattila et al, Nucl. Acids Res. 19:4967, 1991; Eckβrt et al, PCR Methods and Applications 1:17, 1991; PCR (eds. McPherson et al, JRL Press, Oxford); and U.S. Patent 4,683,202. The nucleic acid molecules can be amplified using cDNA, mRNA or genomic DNA as a template, cloned into an appropriate vector and characterized by DNA sequence analysis.
Other suitable amplification methods include the ligase chain reaction (LCR) (see Wu and Wallace, Genomics 4:560, 1989, Landegren et al, Science 241:1077, 1988, transcription amplification (Kwoh et al, Proc. Natl. Acad. Sci. USA 86:1173, 1989), and self-sustained sequence replication (Guatelli et al, Proc. Nat. Acad. Sci. USA 87: 1874, 1990) and nucleic acid based sequence amplification (NASBA). The latter two amplification methods involve isothermal reactions based on isothermal transcription, which produce both single stranded RNA (ssRNA) and double stranded DNA (dsDNA) as the amplification products in a ratio of about 30 or 100 to 1, respectively. The amplified DNA can be radiolabelled and used as a probe for screening a cDNA library derived from human cells, mRNA in zap express, ZIPLOX or other suitable vector. Corresponding clones can be isolated, DNA can obtained following in vivo excision, and the cloned insert can be sequenced in either or both orientations by art recognized methods to identify the correct reading frame encoding a polypeptide of the appropriate molecular weight. For example, the direct analysis of the nucleotide sequence of nucleic acid molecules of the present invention can be accomplished using well-known methods that are commercially available. See, for example, Sambrook et al, Molecular Cloning, A Laboratory Manual (2nd Ed., CSHP, New York 1989); Zyskind et al, Recombinant DNA Laboratory Manual, (Acad. Press, 1988)). Using these or similar methods, the polypeptide and the DNA encoding the polypeptide can be isolated, sequenced and further characterized.
Antisense nucleic acid molecules of the invention can be designed using the nucleotide sequences of SEQ JD NO: 1 and/or the complement of SEQ ID NO: 1, and/or a portion of SEQ ID NO: 1, or the complement of SEQ JD NO: 1 and/or a sequence encoding the amino acid sequences of SEQ JD NO: 2, or encoding a portion of SEQ ID NO: 2, and constructed using chemical synthesis and enzymatic ligation reactions using procedures known in the art. For example, an antisense nucleic acid molecule (e.g., an antisense oligonucleotide) can be chemically synthesized using naturally occurring nucleotides or variously modified nucleotides designed to increase the biological stability of the molecules or to increase the physical stability of the duplex formed between the antisense and sense nucleic acids, e.g., phosphorothioate derivatives and acridine substituted nucleotides can be used. Alternatively, the antisense nucleic acid molecule can be produced biologically using an expression vector into which a nucleic acid molecule has been subcloned in an antisense orientation (i.e., RNA transcribed from the inserted nucleic acid molecule will be of an antisense orientation to a target nucleic acid of interest).
In general, the isolated nucleic acid sequences of the invention can be used as molecular weight markers on Southern gels, and as chromosome markers which are labeled to map related gene positions. The nucleic acid sequences can also be used to compare with endogenous DNA sequences in patients to identify one or more of the disorders described above, and as probes, such as to hybridize and discover related DNA sequences or to subtract out known sequences from a sample. The nucleic acid sequences can further be used to derive primers for genetic finge rinting, to raise anti-polypeptide antibodies using DNA immunization techniques, and as an antigen to raise anti-DNA antibodies or elicit immune responses. Portions or fragments of the nucleotide sequences identified herein (and the corresponding complete gene sequences) can be used in numerous ways as polynucleotide reagents. For example, these sequences can be used to: (i) map their respective genes on a chromosome; and, thus, locate gene regions associated with genetic disease; (ii) identify an individual from a minute biological sample (tissue typing); and (iii) aid in forensic identification of a biological sample. Additionally, the nucleotide sequences of the invention can be used to identify and express recombinant polypeptides for analysis, characterization or therapeutic use, or as markers for tissues in which the corresponding polypeptide is expressed, either constitutively, during tissue differentiation, or in diseased states. The nucleic acid sequences can additionally be used as reagents in the screening and/or diagnotic assays described herein, and can also be included as components of kits (e.g., reagent kits) for use in the screening and/or diagnostic assays described herein.
Another aspect of the invention pertains to nucleic acid constructs containing a nucleic acid molecule of SEQ JD NO: 1 and the complements thereof (or a portion thereof). Yet another aspect of the invention pertains to nucleic acid constructs containing a nucleic acid molecule encoding an amino acid sequence of SEQ JD NO: 2 or polymorphic variant thereof. The constructs comprise a vector (e.g., an expression vector) into which a sequence of the invention has been inserted in a sense or antisense orientation. As used herein, the term "vector" refers to a nucleic acid molecule capable of transporting another nucleic acid to which it has been linked. One type of vector is a "plasmid", which refers to a circular double stranded DNA loop into which additional DNA segments can be ligated. Another type of vector is a viral vector, wherein additional DNA segments can be ligated into the viral genome. Certain vectors are capable of autonomous replication in a host cell into which they are introduced (e.g., bacterial vectors having a bacterial origin of replication and episomal mammalian vectors). Other vectors (e.g., non-episomal mammalian vectors) are integrated into the genome of a host cell upon introduction into the host cell, and thereby are replicated along with the host genome. Moreover, certain vectors, expression vectors, are capable of directing the expression of genes to which they are operably linked, h general, expression vectors of utility in recombinant DNA techniques are often in the form of plasmids. However, the invention is intended to include such other forms of expression vectors, such as viral vectors (e.g., replication defective retroviruses, adeno viruses and adeno-associated viruses) that serve equivalent functions.
Preferred recombinant expression vectors of the invention comprise a nucleic acid molecule of the invention in a form suitable for expression of the nucleic acid molecule in a host cell. This means that the recombinant expression vectors include one or more regulatory sequences, selected on the basis of the host cells to be used for expression, which is operably linked to the nucleic acid sequence to be expressed. Within a recombinant expression vector, "operably linked" or "operatively linked" is intended to mean that the nucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the nucleotide sequence (e.g., in an in vitro transcription/translation system or in a host cell when the vector is introduced into the host cell). The term "regulatory sequence" is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals). Such regulatory sequences are described, for example, in Goeddel, "Gene Expression Technology", Methods in Enzymology 185, Academic Press, San Diego, CA (1990). Regulatory sequences include those which direct constitutive expression of a nucleotide sequence in many types of host cell and those which direct expression of the nucleotide sequence only in certain host cells (e.g. , tissue-specific regulatory sequences). It will be appreciated by those skilled in the art that the design of the expression vector can depend on such factors as the choice of the host cell to be transformed and the level of expression of polypeptide desired. The expression vectors of the invention can be introduced into host cells to thereby produce polypeptides, including fusion polypeptides, encoded by nucleic acid molecules as described herein.
The recombinant expression vectors of the invention can be designed for expression of a polypeptide of the invention in prokaryotic or eukaryotic cells, e.g., bacterial cells such as E. coli, insect cells (using baculovirus expression vectors), yeast cells or mammalian cells. Suitable host cells are discussed further in Goeddel, supra. Alternatively, the recombinant expression vector can be transcribed and translated in vitro, for example using T7 promoter regulatory sequences and T7 polymerase.
Another aspect of the invention pertains to host cells into which a recombinant expression vector of the invention has been introduced. The terms "host cell" and "recombinant host cell" are used interchangeably herein. It is understood that such terms refer not only to the particular subject cell but also to the progeny or potential progeny of such a cell. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
A host cell can be any prokaryotic or eukaryotic cell. For example, a nucleic acid molecule of the invention can be expressed in bacterial cells (e.g., E. coli), insect cells, yeast or mammalian cells (such as Chinese hamster ovary cells (CHO) or COS cells). Other suitable host cells are known to those skilled in the art. Vector DNA can be introduced into prokaryotic or eukaryotic cells via conventional transformation or transfection techniques. As used herein, the terms "transformation" and "transfection" are intended to refer to a variety of art-recognized techniques for introducing a foreign nucleic acid molecule (e.g., DNA) into a host cell, including calcium phosphate or calcium chloride co-precipitation, DEAE-dextran-mediated transfection, hpofection, or electroporation. Suitable methods for transforming or transfecting host cells can be found in Sambrook, et al. (supra), and other laboratory manuals.
For stable transfection of mammalian cells, it is known that, depending upon the expression vector and transfection technique used, only a small fraction of cells may integrate the foreign DNA into their genome, hi order to identify and select these integrants, a gene that encodes a selectable marker (e.g., for resistance to antibiotics) is generally introduced into the host cells along with the gene of interest. Preferred selectable markers include those that confer resistance to drugs, such as G418, hygromycin and methotrexate. Nucleic acid molecules encoding a selectable marker can be introduced into a host cell on the same vector as the nucleic acid molecule of the invention or can be introduced on a separate vector. Cells stably transfected with the introduced nucleic acid molecule can be identified by drug selection (e.g., cells that have incorporated the selectable marker gene will survive, while the other cells die).
A host cell of the invention, such as a prokaryotic or eukaryotic host cell in culture, can be used to produce (i.e., express) a polypeptide of the invention. Accordingly, the invention further provides methods for producing a polypeptide using the host cells of the invention. In one embodiment, the method comprises culturing the host cell of invention (into which a recombinant expression vector encoding a polypeptide of the invention has been introduced) in a suitable medium such that the polypeptide is produced. In another embodiment, the method further comprises isolating the polypeptide from the medium or the host cell.
The host cells of the invention can also be used to produce nonhuman transgenic animals. For example, in one embodiment, a host cell of the invention is a fertilized oocyte or an embryonic stem cell into which a nucleic acid molecule of the invention has been introduced (e.g. , an exogenous NR gene, or an exogenous nucleic acid encoding an NR polypeptide). Such host cells can then be used to create non-human transgenic animals in which exogenous nucleotide sequences have been introduced into the genome or homologous recombinant animals in which endogenous nucleotide sequences have been altered. Such animals are useful for studying the function and/or activity of the nucleotide sequence and polypeptide encoded by the sequence and for identifying and/or evaluating modulators of their activity. As used herein, a "transgenic animal" is a non-human animal, preferably a mammal, more preferably a rodent such as a rat or mouse, in which one or more of the cells of the animal includes a transgene. Other examples of transgenic animals include non-human primates, sheep, dogs, cows, goats, chickens and amphibians. A transgene is exogenous DNA which is integrated into the genome of a cell from which a transgenic animal develops and which remains in the genome of the mature animal, thereby directing the expression of an encoded gene product in one or more cell types or tissues of the transgenic animal. As used herein, an "homologous recombinant animal" is a non-human animal, preferably a mammal, more preferably a mouse, in which an endogenous gene has been altered by homologous recombination between the endogenous gene and an exogenous DNA molecule introduced into a cell of the animal, e.g., an embryonic cell of the animal, prior to development of the animal.
Methods for generating transgenic animals via embryo manipulation and microiηjection, particularly animals such as mice, have become conventional in the art and are described, for example, in U.S. Patent NO. 4,736,866 and 4,870,009, U.S. Pat. No. 4,873,191 and in Hogan, Manipulating the Mouse Embryo (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., 1986). Methods for constructing homologous recombination vectors and homologous recombinant animals are described further in Bradley (1991) Current Opinion in BioTechnology 2:823-829 and in PCT Publication Nos. WO 90/11354, WO 91/01140, WO 92/0968, and WO 93/04169. Clones of the non-human transgenic animals described herein can also be produced according to the methods described in Wihnut et al. (1997) Nature 385:810-813 and PCT Publication Nos. WO 97/07668 and WO 97/07669.
POLYPEPTIDES OF THE INVENTION
The present invention also pertains to isolated polypeptides encoded by NRs ("NR polypeptides") and fragments and variants thereof, as well as polypeptides encoded by nucleotide sequences described herein (e.g., other splicing variants). The term "polypeptide" refers to a polymer of amino acids, and not to a specific length; thus, peptides, oligopeptides and proteins are included within the definition of a polypeptide. As used herein, a polypeptide is said to be "isolated" or "purified" when it is substantially free of cellular material when it is isolated from recombinant and non-recombinant cells, or free of chemical precursors or other chemicals when it is chemically synthesized. A polypeptide, however, can be joined to another polypeptide with which it is not normally associated in a cell (e.g. , in a "fusion protein") and still be "isolated" or "purified."
The polypeptides of the invention can be purified to homogeneity. It is understood, however, that preparations in which the polypeptide is not purified to homogeneity are useful. The critical feature is that the preparation allows for the desired function of the polypeptide, even in the presence of considerable amounts of other components. Thus, the invention encompasses various degrees of purity. In one embodiment, the language "substantially free of cellular material" includes preparations of the polypeptide having less than about 30% (by dry weight) other proteins (i.e., contaminating protein), less than about 20% other proteins, less than about 10% other proteins, or less than about 5% other proteins.
When a polypeptide is recombinantly produced, it can also be substantially free of culture medium, i.e., culture medium represents less than about 20%, less than about 10%, or less than about 5% of the volume of the polypeptide preparation. The language "substantially free of chemical precursors or other chemicals" includes preparations of the polypeptide in which it is separated from chemical precursors or other chemicals that are involved in its synthesis, hi one embodiment, the language "substantially free of chemical precursors or other chemicals" includes preparations of the polypeptide having less than about 30% (by dry weight) chemical precursors or other chemicals, less than about 20% chemical precursors or other chemicals, less than about 10% chemical precursors or other chemicals, or less than about 5% chemical precursors or other chemicals.
In one embodiment, a polypeptide of the invention comprises an amino acid sequence encoded by a nucleic acid molecule comprising a nucleotide sequence of SEQ JD NO: 1, or the complement of such a nucleic acid, or portions thereof, e.g., SEQ JD NO: 1, or a portion or polymorphic variant thereof. However, the polypeptides of the invention also encompass fragment and sequence variants. Variants include a substantially homologous polypeptide encoded by the same genetic locus in an organism, i.e., an allelic variant, as well as other splicing variants. Variants also encompass polypeptides derived from other genetic loci in an organism, but having substantial homology to a polypeptide encoded by a nucleic acid molecule comprising a nucleotide sequence of SEQ JD NO: 1, or a complement of such a sequence, or portions thereof, or having substantial homology to a polypeptide encoded by a nucleic acid molecule comprising a nucleotide sequence of nucleotide sequences encoding SEQ JD NO: 2, or polymorphic variants thereof. Variants also include polypeptides substantially homologous or identical to these polypeptides but derived from another organism, i.e., an ortholog. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by chemical synthesis. Variants also include polypeptides that are substantially homologous or identical to these polypeptides that are produced by recombinant methods. As used herein, two polypeptides (or a region of the polypeptides) are substantially homologous or identical when the amino acid sequences are at least about 45-55%, typically at least about 70-75%, more typically at least about 80-85%, and most typically greater than about 90% or more homologous or identical. A substantially homologous amino acid sequence, according to the present invention, will be encoded by a nucleic acid molecule hybridizing to one or more of SEQ ID NO: 1, or portion thereof, under stringent conditions as more particularly described above, or will be encoded by a nucleic acid molecule hybridizing to a nucleic acid sequence encoding one of SEQ ID NO: 2, a portion thereof or polymorphic variant thereof, under stringent conditions as more particularly described thereof. To determine the percent homology or identity of two amino acid sequences, or of two nucleic acid sequences, the sequences are aligned for optimal comparison purposes (e.g., gaps can be introduced in the sequence of one polypeptide or nucleic acid molecule for optimal alignment with the other polypeptide or nucleic acid molecule). The amino acid residues or nucleotides at corresponding amino acid positions or nucleotide positions are then compared. When a position in one sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the other sequence, then the molecules are homologous at that position. As used herein, amino acid or nucleic acid "homology" is equivalent to amino acid or nucleic acid "identity". The percent homology between the two sequences is a function of the number of identical positions shared by the sequences (i.e., percent homology equals the number of identical positions/total number of positions times 100).
The invention also encompasses polypeptides having a lower degree of identity but having sufficient similarity so as to perform one or more of the same functions performed by a polypeptide encoded by a nucleic acid molecule of the invention. Similarity is determined by conserved amino acid substitution. Such substitutions are those that substitute a given amino acid in a polypeptide by another amino acid of like characteristics. Conservative substitutions are likely to be phenotypically silent. Typically seen as conservative substitutions are the replacements, one for another, among the aliphatic amino acids Ala, Val, Leu and He; interchange of the hydroxyl residues Ser and Thr, exchange of the acidic residues Asp and Glu, substitution between the amide residues Asn and Gin, exchange of the basic residues Lys and Arg and replacements among the aromatic residues Phe and Tyr. Guidance concerning which amino acid changes are likely to be phenotypically silent are found in Bowie et al, Science 247:1306-1310 (1990). A variant polypeptide can differ in amino acid sequence by one or more substitutions, deletions, insertions, inversions, fusions, and truncations or a combination of any of these. Further, variant polypeptides can be fully functional or can lack function in one or more activities. Fully functional variants typically contain only conservative variation or variation in non-critical residues or in non-critical regions. Functional variants can also contain substitution of similar amino acids that result in no change or an insignificant change in function. Alternatively, such substitutions may positively or negatively affect function to some degree. Non-functional variants typically contain one or more non-conservative amino acid substitutions, deletions, insertions, inversions, or truncation or a substitution, insertion, inversion, or deletion in a critical residue or critical region. Amino acids that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (Cunningham et al, Science 244:1081-1085 (1989)). The latter procedure introduces single alanine mutations at every residue in the molecule. The resulting mutant molecules are then tested for biological activity in vitro, or in vitro proliferative activity. Sites that are critical for polypeptide activity can also be determined by structural analysis such as crystallization, nuclear magnetic resonance or photoaffmity labeling (Smith et al, J. Mol Biol 224:899-904 (1992); de Vos et al, Science 255:306-312 (1992)). The invention also includes polypeptide fragments of the polypeptides of the invention. Fragments can be derived from a polypeptide encoded by a nucleic acid molecule comprising SEQ JD NO: 1, or a complement of such a nucleic acid (e.g., SEQ JD NO: 1, or other variants). However, the invention also encompasses fragments of the variants of the polypeptides described herein. As used herein, a fragment comprises at least 6 contiguous amino acids. Useful fragments include those that retain one or more of the biological activities of the polypeptide as well as fragments that can be used as an immunogen to generate polypeptide-specific antibodies.
Biologically active fragments (peptides which are, for example, 6, 9, 12, 15, 16, 20, 30, 35, 36, 37, 38, 39, 40, 50, 100 or more amino acids in length) can comprise a domain, segment, or motif that has been identified by analysis of the polypeptide sequence using well-known methods, e.g., signal peptides, extracellular domains, one or more transmembrane segments or loops, ligand binding regions, zinc finger domains, DNA binding domains, acylation sites, glycosylation sites, or phosphorylation sites. Fragments can be discrete (not fused to other amino acids or polypeptides) or can be within a larger polypeptide. Further, several fragments can be comprised within a single larger polypeptide. In one embodiment a fragment designed for expression in a host can have heterologous pre- and pro-polypeptide regions fused to the amino terminus of the polypeptide fragment and an additional region fused to the carboxyl terminus of the fragment.
The invention thus provides chimeric or fusion polypeptides. These comprise a polypeptide of the invention operatively linked to a heterologous protein or polypeptide having an amino acid sequence not substantially homologous to the polypeptide. "Operatively linked" indicates that the polypeptide and the heterologous protein are fused in-frame. The heterologous protein can be fused to the N-terminus or C-terminus of the polypeptide. h one embodiment the fusion polypeptide does not affect function of the polypeptide per se. For example, the fusion polypeptide can be a GST-fusion polypeptide in which the polypeptide sequences are fused to the C-terminus of the GST sequences. Other types of fusion polypeptides include, but are not limited to, enzymatic fusion polypeptides, for example β-galactosidase fusions, yeast two-hybrid GAL fusions, poly-His fusions and Ig fusions. Such fusion polypeptides, particularly poly-His fusions, can facilitate the purification of recombinant polypeptide. hi certain host cells (e.g., mammalian host cells), expression and/or secretion of a polypeptide can be increased by using a heterologous signal sequence. Therefore, in another embodiment, the fusion polypeptide contains a heterologous signal sequence at its N-terminus.
EP-A-O 464 533 discloses fusion proteins comprising various portions of immunoglobulin constant regions. The Fc is useful in therapy and diagnosis and thus results, for example, in improved pharmacokinetic properties (EP-A 0232 262). In drug discovery, for example, human proteins have been fused with Fc portions for the purpose of high-throughput screening assays to identify antagonists. Bennett et al, J. Mol Recog. 8:52-58 (1995) and Johanson et al, J. Biol Chem. 270:16:9459-9471 (1995). Thus, this invention also encompasses soluble fusion polypeptides containing a polypeptide of the invention and various portions of the constant regions of heavy or light chains of immunoglobulins of various subclass (IgG, IgM, IgA, IgE).
A chimeric or fusion polypeptide can be produced by standard recombinant DNA techniques. For example, DNA fragments coding for the different polypeptide sequences are ligated together in-frame in accordance with conventional techniques. hi another embodiment, the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers. Alternatively, PCR amplification of nucleic acid fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive nucleic acid fragments which can subsequently be annealed and re-amplified to generate a chimeric nucleic acid sequence (see Ausubel et al. , Current Protocols in Molecular Biology, 1992).
Moreover, many expression vectors are commercially available that already encode a fusion moiety (e.g., a GST protein). A nucleic acid molecule encoding a polypeptide of the invention can be cloned into such an expression vector such that the fusion moiety is linked in-frame to the polypeptide. The isolated polypeptide can be purified from cells that naturally express it, purified from cells that have been altered to express it (recombinant), or synthesized using known protein synthesis methods. In one embodiment, the polypeptide is produced by recombinant DNA techniques. For example, a nucleic acid molecule encoding the polypeptide is cloned into an expression vector, the expression vector introduced into a host cell and the polypeptide expressed in the host cell. The polypeptide can then be isolated from the cells by an appropriate purification scheme using standard protein purification techniques. h general, polypeptides of the present invention can be used as a molecular weight marker on SDS-PAGE gels or on molecular sieve gel filtration columns using art-recognized methods. The polypeptides of the present invention can be used to raise antibodies or to elicit an immune response. The polypeptides can also be used as a reagent, e.g., a labeled reagent, in assays to quantitatively deteπnine levels of the polypeptide or a molecule to which it binds (e.g., a ligand) in biological fluids. The polypeptides can also be used as markers for cells or tissues in which the corresponding polypeptide is preferentially expressed, either constitutively, during tissue differentiation, or in a diseased state. The polypeptides can be used to isolate a corresponding binding agent, e.g., ligand, such as, for example, in an interaction trap assay, and to screen for peptide or small molecule antagonists or agonists of the binding interaction.
ANTIBODIES OF THE INVENTION Polyclonal and/or monoclonal antibodies that specifically bind one form of the gene product but not to the other form of the gene product are also provided. Antibodies are also provided that bind a portion of either the variant or the reference gene product that contains the polymorphic site or sites. The invention provides antibodies to the polypeptides and polypeptide fragments of the invention, e.g., having an amino acid sequence of SEQ JD NO: 2 or a portion thereof, or having an amino acid sequence encoded by a nucleic acid molecule comprising all or a portion of SEQ ID NO: 1, or a complement or another variant or portion thereof. The term "antibody" as used herein refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically binds an antigen. A molecule that specifically binds to a polypeptide of the invention is a molecule that binds to that polypeptide or a fragment thereof, but does not substantially bind other molecules in a sample, e.g., a biological sample, which naturally contains the polypeptide. Examples of immunologically active portions of immunoglobulin molecules include F(ab) and F(ab')2 fragments which can be generated by treating the antibody with an enzyme such as pepsin. The invention provides polyclonal and monoclonal antibodies that bind to a polypeptide of the invention. The term "monoclonal antibody" or "monoclonal antibody composition", as used herein, refers to a population of antibody molecules that contain only one species of an antigen binding site capable of immunoreacting with a particular epitope of a polypeptide of the invention. A monoclonal antibody composition thus typically displays a single binding affinity for a particular polypeptide of the invention with which it immunoreacts.
Polyclonal antibodies can be prepared as described above by immunizing a suitable subject with a desired immunogen, e.g., polypeptide of the invention or fragment thereof. The antibody titer in the immunized subject can be monitored over time by standard techniques, such as with an enzyme linked immunosorbent assay (ELISA) using immobilized polypeptide. If desired, the antibody molecules directed against the polypeptide can be isolated from the mammal (e.g., from the blood) and further purified by well-known techniques, such as protein A chromatography to obtain the IgG fraction. At an appropriate time after immunization, e.g., when the antibody titers are highest, antibody-producing cells can be obtained from the subject and used to prepare monoclonal antibodies by standard techniques, such as the hybridoma technique originally described by Kohler and Milstein (1975) Nature 256:495-497, the human B cell hybridoma technique (Kozbor et al. (1983) Immunol. Today 4:72), the EBV-hybridoma technique (Cole et al. (1985), Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, h e, pp. 77-96) or tiioma techniques. The technology for producing hybridomas is well known (see generally Current Protocols in Immunology (1994) Coligan et al. (eds.) John Wiley & Sons, Inc., New York, NY). Briefly, an immortal cell line (typically a myeloma) is fused to lymphocytes (typically splenocytes) from a mammal immunized with an immunogen as described above, and the culture supernatants of the resulting hybridoma cells are screened to identify a hybridoma producing a monoclonal antibody that binds a polypeptide of the invention.
Any of the many well known protocols used for fusing lymphocytes and immortalized cell lines can be applied for the purpose of generating a monoclonal antibody to a polypeptide of the invention (see, e.g., Current Protocols in Immunology, supra; Galfre et al, Nature 266:55052 (1977); R.H. Kenneth, in Monoclonal Antibodies: A New Dimension In Biological Analyses, Plenum Publishing Corp., New York, New York (1980); and Lerner, Yale J. Biol. Med. 54:387-402 (1981). Moreover, the ordinarily skilled worker will appreciate that there are many variations of such methods that also would be useful.
Alternative to preparing monoclonal antibody-secreting hybridomas, a monoclonal antibody to a polypeptide of the invention can be identified and isolated by screening a recombinant combinatorial immunoglobulin library (e.g., an antibody phage display library) with the polypeptide to thereby isolate immunoglobulin library members that bind the polypeptide. Kits for generating and screening phage display libraries are commercially available (e.g., the Pharmacia Recombinant Phage Antibody System, Catalog No. 27-9400-01; and the Stratagene SurfZAP™ Phage Display Kit, Catalog No. 240612). Additionally, examples of methods and reagents particularly amenable for use in generating and screening antibody display library can be found in, for example, U.S. Patent No. 5,223,409; PCT Publication No. WO 92/18619; PCT Publication No. WO 91/17271; PCT Publication No. WO 92/20791; PCT Publication No. WO 92/15679; PCT Publication No. WO 93/01288; PCT Publication No. WO 92/01047; PCT Publication No. WO 92/09690; PCT Publication No. WO 90/02809; Fuchs et al, Bio/Technology 9:1370-1372 (1991); Hay et al, Hum. Antϊbod. Hybridomas 3:81-85 (1992); Huse et al, Science 246:1275-1281 (1989); Griffiths et al., EMBOJ. 12:725-734 (1993).
Additionally, recombinant antibodies, such as chimeric and humanized monoclonal antibodies, comprising both human and non-human portions, which can be made using standard recombinant DNA techniques, are within the scope of the invention. Such chimeric and humanized monoclonal antibodies can be produced by recombinant DNA techniques known in the art. In general, antibodies of the invention (e.g., a monoclonal antibody) can be used to isolate a polypeptide of the invention by standard techniques, such as affinity chromatography or immunoprecipitation. A polypeptide-specific antibody can facilitate the purification of natural polypeptide from cells and of recombinantly produced polypeptide expressed in host cells. Moreover, an antibody specific for a polypeptide of the invention can be used to detect the polypeptide (e.g., in a cellular lysate, cell supernatant, or tissue sample) in order to evaluate the abundance and pattern of expression of the polypeptide. Antibodies can be used diagnostically to monitor protein levels in tissue as part of a clinical testing procedure, e.g., to, for example, determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, and radioactive materials. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, β-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin, and examples of suitable radioactive material include I, I, S or H.
DIAGNOSTIC AND SCREENING ASSAYS OF THE INVENTION
The present invention also pertains to a method of diagnosing or aiding in the diagnosis of a disease or condition associated with an NR gene or gene product in an individual. Diagnostic assays can be designed for assessing NR gene expression, or for assessing activity of NR polypeptides of the invention, hi one embodiment, the assays are used in the context of a biological sample (e.g., blood, serum, cells, tissue) to thereby determine whether an individual is afflicted with a disease or condition associated with an NR, or a defect in an NR. The invention also provides for prognostic (or predictive) assays for determining whether an individual is susceptible to a disease of condition associated with a NR, e.g., if an individual is at risk for addiction to an opoid. For example, mutations in the gene can be assayed in a biological sample. Such assays can be used for prognostic or predictive purpose to thereby prophylactically treat an individual prior to the onset of symptoms associated with a susceptibility to a disease or condition associated with an NR. Another aspect of the invention pertains to assays for monitoring the influence of agents (e.g., drugs, compounds or other agents) on the gene expression or activity of polypeptides of the invention, as well as to assays for identifying agents which bind to a polypeptides. These and other assays and agents are described in further detail in the following sections.
DIAGNOSTIC ASSAYS
The nucleic acids, probes, primers, polypeptides and antibodies described herein can be used in methods of diagnosis of a susceptibility to a disease or condition associated with an NR, as well as in kits useful for diagnosis of a susceptibility to a disease or condition associated with an NR.
In one embodiment of the invention, diagnosis of a susceptibility to a disease or condition associated with an NR is made by detecting a polymorphism in an NR as described herein. The polymorphism can be a mutation in an NR, such as the insertion or deletion of a single nucleotide, or of more than one nucleotide, resulting in a frame shift mutation; the change of at least one nucleotide, resulting in a change in the encoded amino acid; the change of at least one nucleotide, resulting in the generation of a premature stop codon; the deletion of several nucleotides, resulting in a deletion of one or more amino acids encoded by the nucleotides; the insertion of one or several nucleotides, such as by unequal recombination or gene conversion, resulting in an interruption of the coding sequence of the gene; duplication of all or a part of the gene; transposition of all or apart of the gene; or rearrangement of all or a part of the gene. More than one such mutation may be present in a single gene. Such sequence changes cause a mutation in the polypeptide encoded by an NR gene. For example, if the mutation is a frame shift mutation, the frame shift can result in a change in the encoded amino acids, and/or can result in the generation of a premature stop codon, causing generation of a truncated polypeptide. Alternatively, a polymorphism associated with a susceptibility to a disease or condition associated with an NR can be a synonymous mutation in one or more nucleotides (i.e., a mutation that does not result in a change in the polypeptide encoded by an NR gene). Such a polymorphism may alter splicing sites, affect the stability or transport of mRNA, or otherwise affect the transcription or translation of the gene. An NR gene that has any of the mutations described above is referred to herein as a "mutant gene." h a first method of diagnosing a susceptibility to a susceptibility to a disease or condition associated with an NR, hybridization methods, such as Southern analysis, Northern analysis, or in situ hybridizations, can be used (see Current Protocols in Molecular Biology, Ausubel, F. et al, eds., John Wiley & Sons, including all supplements through 1999). For example, a biological sample from a test subject (a "test sample") of genomic DNA, RNA, or cDNA, is obtained from an individual suspected of having, being susceptible to or predisposed for, or carrying a defect for, a susceptibility to a disease or condition associated with an NR (the "test individual"). The individual can be an adult, child, or fetus. The test sample can be from any source which contains genomic DNA, such as a blood sample, sample of amniotic fluid, sample of cerebrospinal fluid, or tissue sample from skin, muscle, buccal or conjunctival mucosa, placenta, gastrointestinal tract or other organs. A test sample of DNA from fetal cells or tissue can be obtained by appropriate methods, such as by amniocentesis or chorionic villus sampling. The DNA, RNA, or cDNA sample is then examined to determine whether a polymorphism in an NR is present, and/or to determine which splicing variant(s) encoded by the NR is present. The presence of the polymorphism or splicing variant(s) can be indicated by hybridization of the gene in the genomic DNA, RNA, or cDNA to a nucleic acid probe. A "nucleic acid probe", as used herein, can be a DNA probe or an RNA probe; the nucleic acid probe can contain at least one polymorphism in an NR or contains a nucleic acid encoding a particular splicing variant of an NR. The probe can be any of the nucleic acid molecules described above (e.g., the gene, a fragment, a vector comprising the gene, a probe or primer, etc.). To diagnose a susceptibility to a susceptibility to a disease or condition associated with an NR, a hybridization sample is formed by contacting the test sample containing an NR, with at least one nucleic acid probe. A preferred probe for detecting mRNA or genomic DNA is a labeled nucleic acid probe capable of hybridizing to mRNA or genomic DNA sequences described herein. The nucleic acid probe can be, for example, a full-length nucleic acid molecule, or a portion thereof, such as an oligonucleotide of at least 15, 30, 50, 100, 250 or 500 nucleotides in length and sufficient to specifically hybridize under stringent conditions to appropriate mRNA or genomic DNA. For example, the nucleic acid probe can be all or a portion of one of SEQ ID NO : 1 , or the complement thereof, or a portion thereof; or can be a nucleic acid encoding a portion of one of SEQ ID NO: 2. Other suitable probes for use in the diagnostic assays of the invention are described above (see e.g., probes and primers discussed under the heading, "Nucleic Acids of the Invention"). The hybridization sample is maintained under conditions which are sufficient to allow specific hybridization of the nucleic acid probe to anNR. "Specific hybridization", as used herein, indicates exact hybridization (e.g., with no mismatches). Specific hybridization can be performed under high stringency conditions or moderate stringency conditions, for example, as described above. In a particularly preferred embodiment, the hybridization conditions for specific hybridization are high stringency.
Specific hybridization, if present, is then detected using standard methods. If specific hybridization occurs between the nucleic acid probe and the NR in the test sample, then the NR has the polymorphism, or is the splicing variant, that is present in the nucleic acid probe. More than one nucleic acid probe can also be used concurrently in this method. Specific hybridization of any one of the nucleic acid probes is indicative of a polymoφhism in the NR, or of the presence of a particular splicing variant encoding the NR and is therefore diagnostic for a susceptibility to a susceptibility to a disease or condition associated with an NR. In Northern analysis (see Current Protocols in Molecular Biology, Ausubel,
F. et al, eds., John Wiley & Sons, supra) the hybridization methods described above are used to identify the presence of a polymorphism or a particular splicing variant, associated with a susceptibility to a susceptibility to a disease or condition associated with an NR. For Northern analysis, a test sample of RNA is obtained from the individual by appropriate means. Specific hybridization of a nucleic acid probe, as described above, to RNA from the individual is indicative of a polymorphism in an NR, or of the presence of a particular splicing variant encoded by an NR, and is therefore diagnostic for a susceptibility to a susceptibility to a disease or condition associated with an NR.
For representative examples of use of nucleic acid probes, see, for example, U.S. Patents No. 5,288,611 and 4,851,330.
Alternatively, a peptide nucleic acid (PNA) probe can be used instead of a nucleic acid probe in the hybridization methods described above. PNA is a DNA mimic having a peptide-like, inorganic backbone, such as N-(2-aminoethyl)glycine units, with an organic base (A, G, C, T or U) attached to the glycine nitrogen via a methylene carbonyl linker (see, for example, Nielsen, P.E. et al, Bioconjugate Chemistry 5, American Chemical Society, p. 1 (1994). The PNA probe can be designed to specifically hybridize to a gene having a polymorphism associated with a susceptibility to a susceptibility to a disease or condition associated with an NR. Hybridization of the PNA probe to an NR is diagnostic for a susceptibility to a susceptibility to a disease or condition associated with an NR. hi another method of the invention, mutation analysis by restriction digestion can be used to detect a mutant gene, or genes containing a polymorphism(s), if the mutation or polymorphism in the gene results in the creation or elimination of a restriction site. A test sample containing genomic DNA is obtained from the individual. Polymerase chain reaction (PCR) can be used to amplify an NR (and, if necessary, the flanking sequences) in the test sample of genomic DNA from the test individual. RFLP analysis is conducted as described (see Current Protocols in Molecular Biology, supra). The digestion pattern of the relevant DNA fragment indicates the presence or absence of the mutation or polymorphism in the NR, and therefore indicates the presence or absence of this susceptibility to a susceptibility to a disease or condition associated with an NR. Sequence analysis can also be used to detect specific polymorphisms in an NR. A test sample of DNA or RNA is obtained from the test individual. PCR or other appropriate methods can be used to amplify the gene, and/or its flanking sequences, if desired. The sequence of anNR, or a fragment of the gene, or cDNA, or fragment of the cDNA, or mRNA, or fragment of the mRNA, is determined, using standard methods. The sequence of the gene, gene fragment, cDNA, cDNA fragment, mRNA, or mRNA fragment is compared with the known nucleic acid sequence of the gene, cDNA (e.g., of SEQ JD NO: 1, or a complement thereof, or a nucleic acid sequence encoding SEQ JD NO: 2 or a fragment thereof) or mRNA, as appropriate. The presence of a polymorphism in the NR indicates that the individual has a susceptibility to a susceptibility to a disease or condition associated with an NR.
Allele-specific oligonucleotides can also be used to detect the presence of a polymorphism in an NR, through the use of dot-blot hybridization of amplified oligonucleotides with allele-specific oligonucleotide (ASO) probes (see, for example, Saiki, R. et al, Nature 324:163-166 (1986)). An "allele-specific oligonucleotide" (also referred to herein as an "allele-specific oligonucleotide probe") is an oligonucleotide of approximately 10-50 base pairs, preferably approximately 15-30 base pairs, that specifically hybridizes to an NR, and that contains a polymorphism associated with a susceptibility to a susceptibility to a disease or condition associated with an NR. An allele-specific oligonucleotide probe that is specific for particular polymorphisms in an NR can be prepared, using standard methods (see Current Protocols in Molecular Biology, supra). To identify polymorphisms in the gene that are associated with a susceptibility to a susceptibility to a disease or condition associated with an NR, a test sample of DNA is obtained from the individual. PCR can be used to amplify all or a fragment of an NR, and its flanking sequences. The DNA containing the amplified NR (or fragment of the gene) is dot-blotted, using standard methods (see Current Protocols in Molecular Biology, supra), and the blot is contacted with the oligonucleotide probe. The presence of specific hybridization of the probe to the amplified NR is then detected. Specific hybridization of an allele-specific oligonucleotide probe to DNA from the individual is indicative of a polymorphism in the NR, and is therefore indicative of a susceptibility to a susceptibility to a disease or condition associated with an NR.
In another embodiment, arrays of oligonucleotide probes that are complementary to target nucleic acid sequence segments from an individual, can be used to identify polymorphisms in an NR. For example, in one embodiment, an oligonucleotide array can be used. Oligonucleotide arrays typically comprise a plurality of different oligonucleotide probes that are coupled to a surface of a substrate in different known locations. These oligonucleotide arrays, also described as "Genechips™," have been generally described in the art, for example, U.S. Pat. No. 5,143,854 and PCT patent publication Nos. WO 90/15070 and 92/10092. These arrays can generally be produced using mechanical synthesis methods or light directed synthesis methods which incorporate a combination of photolithographic methods and solid phase oligonucleotide synthesis methods. See Fodor et al, Science 251:767-777 (1991), Pirrung et al, U.S. Pat. No. 5,143,854 (see also PCT Application No. WO 90/15070) and Fodor et al , PCT Publication No. WO
92/10092 and U.S. Pat. No. 5,424,186, the entire teachings of each of which are incorporated by reference herein. Techniques for the synthesis of these arrays using mechanical synthesis methods are described in, e.g., U.S. Pat. Nos. 5,384,261, the entire teachings of which are incorporated by reference herein. Once an oligonucleotide array is prepared, a nucleic acid of interest is hybridized with the array and scanned for polymorphisms. Hybridization and scanning are generally carried out by methods described herein and also in, e.g., Published PCT Application Nos. WO 92/10092 and WO 95/11995, and U.S. Pat. No. 5,424,186, the entire teachings of which are incorporated by reference herein, hi brief, a target nucleic acid sequence which includes one or more previously identified polymorphic markers is amplified by well known amplification techniques, e.g., PCR. Typically, this involves the use of primer sequences that are complementary to the two strands of the target sequence both upstream and downstream from the polymorphism. Asymmetric PCR techniques may also be used. Amplified target, generally incorporating a label, is then hybridized with the array under appropriate conditions. Upon completion of hybridization and washing of the array, the array is scanned to determine the position on the array to which the target sequence hybridizes. The hybridization data obtained from the scan is typically in the form of fluorescence intensities as a function of location on the array. Although primarily described in terms of a single detection block, e.g., for detection of a single polymorphism, arrays can include multiple detection blocks, and thus be capable of analyzing multiple, specific polymorphisms. In alternate arrangements, it will generally be understood that detection blocks may be grouped within a single array or in multiple, separate arrays so that varying, optimal conditions may be used during the hybridization of the target to the array. For example, it may often be desirable to provide for the detection of those polymorphisms that fall within G-C rich stretches of a genomic sequence, separately from those falling in A-T rich segments. This allows for the separate optimization of hybridization conditions for each situation.
Additional description of use of oligonucleotide arrays for detection of polymorphisms can be found, for example, in U.S. Patents 5,858,659 and 5,837,832, the entire teachings of which are incorporated by reference herein.
Other methods of nucleic acid analysis can be used to detect polymorphisms in an NR or variants encoding by an NR. Representative methods include direct manual sequencing (Church and Gilbert, Proc. Natl. Acad. Sci. USA 81:1991-1995 (1988); Sanger, F. et al. Proc. Natl Acad. Sci. USA 74:5463-5467 (1977); Beavis et al, U.S. Pat. No. 5,288,644); automated fluorescent sequencing; single-stranded conformation polymorphism assays (SSCP); clamped denaturing gel electiophoresis (CDGE); denaturing gradient gel electiophoresis (DGGE) (Sheffield, V.C. et al. Proc. Natl. Acad. Sci. USA 86:232-236 (1989)), mobility shift analysis (Orita, M. et al, Proc. Natl. Acad. Sci. USA 86:2766-2770 (1989)), restriction enzyme analysis (Flavell et al, Cell 15:25 (1978); Geever, et al, Proc. Natl. Acad. Sci. USA 78:5081 (1981)); heteroduplex analysis; chemical mismatch cleavage (CMC) (Cotton et al, Proc. Natl. Acad. Sci. USA 85:4397-4401 (1985)); RNase protection assays (Myers, R.M. et al, Science 230:1242 (1985)); use of polypeptides which recognize nucleotide mismatches, such as E. coli mutS protein; allele-specific PCR, for example. In another embodiment of the invention, diagnosis of a susceptibility to a susceptibility to a disease or condition associated with an NR can also be made by examining expression and/or composition of an NR polypeptide, by a variety of methods, including enzyme linked immunosorbent assays (ELISAs), Western blots, immunoprecipitations and immunofiuorescence. A test sample from an individual is assessed for the presence of an alteration in the expression and/or an alteration in composition of the polypeptide encoded by an NR, or for the presence of a particular variant encoded by an NR. An alteration in expression of a polypeptide encoded by an NR can be, for example, an alteration in the quantitative polypeptide expression (i.e., the amount of polypeptide produced); an alteration in the composition of a polypeptide encoded by an NR is an alteration in the qualitative polypeptide expression (e.g., expression of a mutant NR polypeptide or of a different splicing variant), hi a preferred embodiment, diagnosis of a susceptibility to a susceptibility to a disease or condition associated with an NR is made by detecting a particular splicing variant encoded by that NR, or a particular pattern of splicing variants.
Both such alterations (quantitative and qualitative) can also be present. An "alteration" in the polypeptide expression or composition, as used herein, refers to an alteration in expression or composition in a test sample, as compared with the expression or composition of polypeptide by an NR in a control sample. A control sample is a sample that corresponds to the test sample (e.g., is from the same type of cells), and is from an individual who is not affected by a susceptibility to a disease or condition associated with an NR. An alteration in the expression or composition of the polypeptide in the test sample, as compared with the control sample, is indicative of a susceptibility to a susceptibility to a disease or condition associated with an NR. Similarly, the presence of one or more different splicing variants in the test sample, or the presence of significantly different amounts of different splicing variants in the test sample, as compared with the control sample, is indicative of a susceptibility to a susceptibility to a disease or condition associated with an NR. Various means of examining expression or composition of the polypeptide encoded by an NR can be used, including spectroscopy, colorimetry, electiophoresis, isoelectric focusing, and immunoassays (e.g., David et al, U.S. Pat. No. 4,376,110) such as immunoblotting (see also Current Protocols in Molecular Biology, particularly Chapter 10). For example, in one embodiment, an antibody capable of binding to the polypeptide (e.g., as described above), preferably an antibody with a detectable label, can be used. Antibodies can be polyclonal, or more preferably, monoclonal. An intact antibody, or a fragment thereof (e.g. , Fab or F(ab')2) can be used. The term "labeled", with regard to the probe or antibody, is intended to encompass direct labeling of the probe or antibody by coupling (i.e., physically linking) a detectable substance to the probe or antibody, as well as indirect labeling of the probe or antibody by reactivity with another reagent that is directly labeled. Examples of indirect labeling include detection of a primary antibody using a fluorescently labeled secondary antibody and end-labeling of a DNA probe with biotin such that it can be detected with fluorescently labeled streptavidin.
Western blotting analysis, using an antibody as described above that specifically binds to a polypeptide encoded by a mutant NR, or an antibody that specifically binds to a polypeptide encoded by a non-mutant gene, or an antibody that specifically binds to a particular splicing variant encoded by an NR, can be used to identify the presence in a test sample of a particular splicing variant or of a polypeptide encoded by a polymorphic or mutant NR, or the absence in a test sample of a particular splicing variant or of a polypeptide encoded by a non-polymorphic or non-mutant gene. The presence of a polypeptide encoded by a polymorphic or mutant gene, or the absence of a polypeptide encoded by a non-polymorphic or non- mutant gene, is diagnostic for a susceptibility to a susceptibility to a disease or condition associated with an NR, as is the presence (or absence) of particular splicing variants encoded by the NR gene. In one embodiment of this method, the level or amount of polypeptide encoded by an NR in a test sample is compared with the level or amount of the polypeptide encoded by the NR in a control sample. A level or amount of the polypeptide in the test sample that is higher or lower than the level or amount of the polypeptide in the control sample, such that the difference is statistically significant, is indicative of an alteration in the expression of the polypeptide encoded by the NR, and is diagnostic for a susceptibility to a susceptibility to a disease or condition associated with that NR. Alternatively, the composition of the polypeptide encoded by an NR in a test sample is compared with the composition of the polypeptide encoded by the NR in a control sample (e.g., the presence of different splicing variants). A difference in the composition of the polypeptide in the test sample, as compared with the composition of the polypeptide in the control sample, is diagnostic for a susceptibility to a susceptibility to a disease or condition associated with that NR. hi another embodiment, both the level or amount and the composition of the polypeptide can be assessed in the test sample and in the control sample. A difference in the amount or level of the polypeptide in the test sample, compared to the control sample; a difference in composition in the test sample, compared to the control sample; or both a difference in the amount or level, and a difference in the composition, is indicative of a susceptibility to a susceptibility to a disease or condition associated with that NR.
Kits (e.g., reagent kits) useful in the methods of diagnosis comprise components useful in any of the methods described herein, including for example, hybridization probes or primers as decribed herein (e.g. , labeled probes or primers), reagents for detection of labeled molecules, restriction enzymes (e.g., for RFLP analysis), allele-specific oligonucleotides, antibodies which bind to mutant or to non-mutant (native) NR polypeptide, means for amplification of nucleic acids comprising an NR, or means for analyzing the nucleic acid sequence of an NR or for analyzing the amino acid sequence of an NR polypeptide, etc.
SCREENING ASSAYS AND AGENTS IDENTIFIED THEREBY
The invention provides methods (also referred to herein as "screening assays") for identifying the presence of a nucleotide that hybridizes to a nucleic acid of the invention, as well as for identifying the presence of a polypeptide encoded by a nucleic acid of the invention. In one embodiment, the presence (or absence) of a nucleic acid molecule of interest (e.g., a nucleic acid that has significant homology with a nucleic acid of the invention) in a sample can be assessed by contacting the sample with a nucleic acid comprising a nucleic acid of the invention (e.g., a nucleic acid having the sequence of SEQ JD NO: 1, or the complement thereof, or a nucleic acid encoding an amino acid having the sequence of SEQ ID NO: 2, or a fragment or variant of such nucleic acids), under stringent conditions as described above, and then assessing the sample for the presence (or absence) of hybridization. In a preferred embodiment, high stringency conditions are conditions appropriate for selective hybridization, h another embodiment, a sample containing the nucleic acid molecule of interest is contacted with a nucleic acid containing a contiguous nucleotide sequence (e.g., a primer or a probe as described above) that is at least partially complementary to a part of the nucleic acid molecule of interest (e.g., an NR nucleic acid), and the contacted sample is assessed for the presence or absence of hybridization, hi a preferred embodiment, the nucleic acid containing a contiguous nucleotide sequence is completely complementary to a part of the nucleic acid molecule of interest.
In any of these embodiments, all or a portion of the nucleic acid of interest can be subjected to amplification prior to performing the hybridization. In another embodiment, the presence (or absence) of a polypeptide of interest, such as a polypeptide of the invention or a fragment or variant thereof, in a sample can be assessed by contacting the sample with an antibody that specifically hybridizes to the polypeptide of interest (e.g., an antibody such as those described above), and then assessing the sample for the presence (or absence) of binding of the antibody to the polypeptide of interest.
In another embodiment, the invention provides methods for identifying agents (e.g., fusion proteins, polypeptides, peptidomimetics, prodrugs, other receptors associated with NRs, binding agents, antibodies, small molecules or other drugs, or ribozymes which alter (e.g., increase or decrease) the activity of the polypeptides described herein, or which otherwise interact with the polypeptides herein. For example, such agents can be agents which bind to polypeptides described herein (e.g., NR binding agents); which have a stimulatory or inhibitory effect on, for example, activity of polypeptides of the invention; or which change (e.g., enhance or inhibit) the ability of the polypeptides of the invention to interact with NR binding agents (e.g. , G-proteins, other receptors associated with NRs, or other binding agents); or which alter posttranslational processing of the NR polypeptide (e.g., agents that alter proteolytic processing to direct the polypeptide from where it is normally synthesized to another location in the cell, such as the cell surface; agents that alter proteolytic processing such that more polypeptide is released from the cell, etc. hi one embodiment, the invention provides assays for screening candidate or test agents that bind to or modulate the activity of polypeptides described herein (or biologically active portion(s) thereof), as well as agents identifiable by the assays. Test agents can be obtained using any of the numerous approaches in combinatorial library methods known in the art, including: biological libraries; spatially addressable parallel solid phase or solution phase libraries; synthetic library methods requiring deconvolution; the 'one-bead one-compound' library method; and synthetic library methods using affinity chromatography selection. The biological library approach is limited to polypeptide libraries, while the other four approaches are applicable to polypeptide, non-peptide oligomer or small molecule libraries of compounds (Lam, K.S., Anticancer Drug Des. 12:145 (1997)).
In one embodiment, to identify agents which alter the activity of an NR polypeptide, a cell, cell lysate, or solution containing or expressing an NR polypeptide (e.g., one of SEQ ID NO: 2, or another splicing variant encoded by an NR), or a fragment or derivative thereof (as described above), can be contacted with an agent to be tested; alternatively, the polypeptide can be contacted directly with the agent to be tested. The level (amount) of NR activity is assessed (e.g., the level (amount) of NR activity is measured, either directly or indirectly), and is compared with the level of activity in a control (i.e., the level of activity of the NR polypeptide or active fragment or derivative thereof in the absence of the agent to be tested). If the level of the activity in the presence of the agent differs, by an amount that is statistically significant, from the level of the activity in the absence of the agent, then the agent is an agent that alters the activity of an NR polypeptide. An increase in the level of NR activity relative to a control, indicates that the agent is an agent that enhances (is an agonist of) NR activity. Similarly, a decrease in the level of NR activity relative to a control, indicates that the agent is an agent that inhibits (is an antagonist of) NR activity. In another embodiment, the level of activity of an NR polypeptide or derivative or fragment thereof in the presence of the agent to be tested, is compared with a control level that has previously been established. A level of the activity in the presence of the agent that differs from the control level by an amount that is statistically significant indicates that the agent alters NR activity. The present invention also relates to an assay for identifying agents which alter the expression of an NR gene (e.g., antisense nucleic acids, fusion proteins, polypeptides, peptidomimetics, prodrugs, other receptors associated with NRs, G- proteins, binding agents, antibodies, small molecules or other drugs, or ribozymes) which alter (e.g., increase or decrease) expression (e.g., transcription or translation) of the gene or which otherwise interact with the nucleic acids described herein, as well as agents identifiable by the assays. For example, a solution containing a nucleic acid encoding an NR polypeptide (e.g. , an NR gene) can be contacted with an agent to be tested. The solution can comprise, for example, cells containing the nucleic acid or cell lysate containing the nucleic acid; alternatively, the solution can be another solution which comprises elements necessary for transcription/translation of the nucleic acid. Cells not suspended in solution can also be employed, if desired. The level and/or pattern of NR expression (e.g., the level and/or pattern of mRNA or of protein expressed, such as the level and/or pattern of different splicing variants) is assessed, and is compared with the level and/or pattern of expression in a control (i.e., the level and/or pattern of the NR expression in the absence of the agent to be tested). If the level and/or pattern in the presence of the agent differs, by an amount or in a manner that is statistically significant, from the level and/or pattern in the absence of the agent, then the agent is an agent that alters the expression of NR. Enhancement of NR expression indicates that the agent is an agonist of NR activity. Similarly, inhibition of NR expression indicates that the agent is an antagonist of NR activity. In another embodiment, the level and/or pattern of NR polypeptide(s) (e.g., different splicing variants) in the presence of the agent to be tested, is compared with a control level and/or pattern that has previously been established. A level and/or pattern in the presence of the agent that differs from the control level and/or pattern by an amount or in a manner that is statistically significant indicates that the agent alters NR expression. hi another embodiment of the invention, agents which alter the expression of an NR gene or which otherwise interact with the nucleic acids described herein, can be identified using a cell, cell lysate, or solution containing a nucleic acid encoding the promoter region of the NR gene operably linked to a reporter gene. After contact with an agent to be tested, the level of expression of the reporter gene (e.g. , the level of mRNA or of protein expressed) is assessed, and is compared with the level of expression in a control (i.e., the level of the expression of the reporter gene in the absence of the agent to be tested). If the level in the presence of the agent differs, by an amount or in a manner that is statistically significant, from the level in the absence of the agent, then the agent is an agent that alters the expression of the NR, as indicated by its ability to alter expression of a gene that is operably linked to the NR gene promoter. Enhancement of the expression of the reporter indicates that the agent is an agonist of NR activity. Similarly, inhibition of the expression of the reporter indicates that the agent is an antagonist of NR activity. In another embodiment, the level of expression of the reporter in the presence of the agent to be tested, is compared with a control level that has previously been established. A level in the presence of the agent that differs from the control level by an amount or in a manner that is statistically significant indicates that the agent alters f expression.
Agents which alter the amounts of different splicing variants encoded by an NR (e.g. , an agent which enhances activity of a first splicing variant, and which inhibits activity of a second splicing variant), as well as agents which are agonists of activity of a first splicing variant and antagonists of activity of a second splicing variant, can easily be identified using these methods described above.
In other embodiments of the invention, assays can be used to assess the impact of a test agent on the activity of a polypeptide in relation to an NR binding agent. For example, a cell that expresses a compound that interacts with an NR (herein referred to as a "NR binding agent", which can be a polypeptide or other molecule that interacts with an NR, such as a G-protein) is contacted with an NR in the presence of a test agent, and the ability of the test agent to alter the interaction between the NR and the NR binding agent is determined. Alternatively, a cell lysate or a solution containing the NR binding agent, can be used. An agent which binds to the NR or the NR binding agent can alter the interaction by interfering with, or enhancing the ability of the NR to bind to, associate with, or otherwise interact with the NR binding agent. Determining the ability of the test agent to bind to an NR or an NR binding agent can be accomplished, for example, by coupling the test agent with a radioisotope or enzymatic label such that binding of the test agent to the polypeptide can be determined by detecting the labeled with 2 I, 35S, C or H, either directly or indirectly, and the radioisotope detected by direct counting of radioemmission or by scintillation counting. Alternatively, test agents can be enzymatically labeled with, for example, horseradish peroxidase, alkaline phosphatase, or luciferase, and the enzymatic label detected by determination of conversion of an appropriate substrate to product. It is also within the scope of this invention to determine the ability of a test agent to interact with the polypeptide without the labeling of any of the interactants. For example, a microphysiometer can be used to detect the interaction of a test agent with an NR or an NR binding agent without the labeling of either the test agent, NR, or the NR binding agent. McConnell, H.M. et al, Science 257:1906-1912 (1992). As used herein, a "microphysiometer" (e.g., Cytosensor™) is an analytical instrument that measures the rate at which a cell acidifies its environment using a light-addressable potentiometric sensor (LAPS). Changes in this acidification rate can be used as an indicator of the interaction between ligand and polypeptide. Thus, these receptors can be used to screen for compounds that are agonists for use in treating a susceptibility to a disease or condition associated with an NR or antagonists for studying a susceptibility to a disease or condition associated with an NR. Drugs could be designed to regulate NR activation which in turn can be used to regulate signaling pathways and transcription events of genes downstream.
In another embodiment of the invention, assays can be used to identify polypeptides that interact with one or more NR polypeptides, as described herein. For example, a yeast two-hybrid system such as that described by Fields and Song (Fields, S. and Song, O., Nature 340:245-246 (1989)) can be used to identify polypeptides that interact with one or more NR polypeptides. In such a yeast two- hybrid system, vectors are constructed based on the flexibility of a transcription factor which has two functional domains (a DNA binding domain and a transcription activation domain). If the two domains are separated but fused to two different proteins that interact with one another, transcriptional activation can be achieved, and transcription of specific markers (e.g., nutritional markers such as His and Ade, or color markers such as lacZ) can be used to identify the presence of interaction and transcriptional activation. For example, in the methods of the invention, a first vector is used which includes a nucleic acid encoding a DNA binding domain and also an NR polypeptide, splicing variant, or fragment or derivative thereof, and a second vector is used which includes a nucleic acid encoding a transcription activation domain and also a nucleic acid encoding a polypeptide which potentially may interact with the NR polypeptide, splicing variant, or fragment or derivative thereof (e.g. , an NR polypeptide binding agent or G-protein). Incubation of yeast containing the first vector and the second vector under appropriate conditions (e.g., mating conditions such as used in the Matchmaker™ system from Clontech (Palo Alto, California, USA)) allows identification of colonies which express the markers of interest. These colonies can be examined to identify the polypeptide(s) which interact with the NR polypeptide or fragment or derivative thereof. Such polypeptides may be useful as agents which alter the activity of expression of an NR polypeptide, as described above. hi more than one embodiment of the above assay methods of the present invention, it may be desirable to immobilize either NR, the NR binding agent, or other components of the assay on a solid support, in order to facilitate separation of complexed from uncomplexed forms of one or both of the polypeptides, as well as to accommodate automation of the assay. Binding of a test agent to the polypeptide, or interaction of the polypeptide with a binding agent in the presence and absence of a test agent, can be accomplished in any vessel suitable for containing the reactants. Examples of such vessels include microtitre plates, test tubes, and micro-centrifuge tubes. In one embodiment, a fusion protein (e.g., a glutathione-S-transferase fusion protein) can be provided which adds a domain that allows NR or an NR binding agent to be bound to a matrix or other solid support. In another embodiment, modulators of expression of nucleic acid molecules of the invention are identified in a method wherein a cell, cell lysate, or solution containing a nucleic acid encoding an NR is contacted with a test agent and the expression of appropriate mRNA or polypeptide (e.g., splicing variant(s)) in the cell, cell lysate, or solution, is determined. The level of expression of appropriate mRNA or polypeptide(s) in the presence of the test agent is compared to the level of expression of mRNA or polypeptide(s) in the absence of the test agent. The test agent can then be identified as a modulator of expression based on this comparison. For example, when expression of mRNA or polypeptide is greater (statistically significantly greater) in the presence of the test agent than in its absence, the test agent is identified as a stimulator or enhancer of the mRNA or polypeptide expression. Alternatively, when expression of the mRNA or polypeptide is less (statistically significantly less) in the presence of the test agent than in its absence, the test agent is identified as an inhibitor of the mRNA or polypeptide expression. The level of mRNA or polypeptide expression in the cells can be determined by methods described herein for detecting mRNA or polypeptide.
This invention further pertains to novel agents identified by the above-described screening assays. Accordingly, it is within the scope of this invention to further use an agent identified as described herein in an appropriate animal model. For example, an agent identified as described herein (e.g., a test agent that is a modulating agent, an antisense nucleic acid molecule, a specific antibody, or a polypeptide-binding agent) can be used in an animal model to determine the efficacy, toxicity, or side effects of treatment with such an agent. Alternatively, an agent identified as described herein can be used in an animal model to determine the mechanism of action of such an agent. Furthermore, this invention pertains to uses of novel agents identified by the above-described screening assays for treatments as described herein. In addition, an agent identified as described herein can be used to alter activity of a polypeptide encoded by an NR, or to alter expression of an NR, by contacting the polypeptide or the gene (or contacting a cell comprising the polypeptide or the gene) with the agent identified as described herein. PHARMACEUTICAL COMPOSITIONS
The present invention also pertains to pharmaceutical compositions comprising nucleic acids described herein, particularly nucleotides encoding the polypeptides described herein; comprising polypeptides described herein (e.g., SEQ ID NO: 2); and/or comprising other splicing variants encoded by an NR; and/or an agent that alters (e.g., enhances or inhibits) NR gene expression or NR polypeptide activity as described herein. For instance, a polypeptide, protein (e.g., a G-protein), an agent that alters NR gene expression, or an NR binding agent or binding partner, fragment, fusion protein or prodrug thereof, or a nucleotide or nucleic acid construct (vector) comprising a nucleotide of the present invention, or an agent that alters NR polypeptide activity, can be formulated with a physiologically acceptable carrier or excipient to prepare a pharmaceutical composition. The carrier and composition can be sterile. The formulation should suit the mode of administration.
Suitable pharmaceutically acceptable carriers include but are not limited to water, salt solutions (e.g., NaCl), saline, buffered saline, alcohols, glycerol, ethanol, gum arabic, vegetable oils, benzyl alcohols, polyethylene glycols, gelatin, carbohydrates such as lactose, amylose or starch, dextrose, magnesium stearate, talc, silicic acid, viscous paraffin, perfume oil, fatty acid esters, hydroxymethylcellulose, polyvinyl pyrolidone, etc., as well as combinations thereof. The pharmaceutical preparations can, if desired, be mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring, flavoring and/or aromatic substances and the like which do not deleteriously react with the active agents.
The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. The composition can be a liquid solution, suspension, emulsion, tablet, pill, capsule, sustained release formulation, or powder. The composition can be fonnulated as a suppository, with traditional binders and carriers such as triglycerides. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, polyvinyl pyrolhdone, sodium saccharine, cellulose, magnesium carbonate, etc. Methods of introduction of these compositions include, but are not limited to, intradermal, intramuscular, intraperitoneal, intraocular, intravenous, subcutaneous, topical, oral and intranasal. Other suitable methods of introduction can also include gene therapy (as described below), rechargeable or biodegradable devices, particle acceleration devises ("gene guns") and slow release polymeric devices. The pharmaceutical compositions of this invention can also be administered as part of a combinatorial therapy with other agents.
The composition can be formulated in accordance with the routine procedures as a pharmaceutical composition adapted for administration to human beings. For example, compositions for intravenous administration typically are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water, saline or dextrose/water. Where the composition is administered by injection, an ampule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
For topical application, nonsprayable forms, viscous to semi-solid or solid forms comprising a carrier compatible with topical application and having a dynamic viscosity preferably greater than water, can be employed. Suitable formulations include but are not limited to solutions, suspensions, emulsions, creams, ointments, powders, enemas, lotions, sols, liniments, salves, aerosols, etc., which are, if desired, sterilized or mixed with auxiliary agents, e.g., preservatives, stabilizers, wetting agents, buffers or salts for influencing osmotic pressure, etc. The agent may be incorporated into a cosmetic formulation. For topical application, also suitable are sprayable aerosol preparations wherein the active ingredient, preferably in combination with a solid or liquid inert carrier material, is packaged in a squeeze bottle or in admixture with a pressurized volatile, normally gaseous propellant, e.g., pressurized air.
Agents described herein can be formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with free amino groups such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with free carboxyl groups such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2- ethylamino ethanol, histidine, procaine, etc.
The agents are administered in a therapeuticaUy effective amount. The amount of agents which will be therapeuticaUy effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques. In addition, in vitro or in vivo assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the symptoms of a susceptibility to a disease or condition associated with an NR, and should be decided according to the judgment of a practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems. The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use of sale for human administration. The pack or kit can be labeled with information regarding mode of administration, sequence of drug administration (e.g., separately, sequentially or concurrently), or the like. The pack or kit may also include means for reminding the patient to take the therapy. The pack or kit can be a single unit dosage of the combination therapy or it can be a plurality of unit dosages, hi particular, the agents can be separated, mixed together in any combination, present in a single vial or tablet. Agents assembled in a blister pack or other dispensing means is preferred. For the purpose of this invention, unit dosage is intended to mean a dosage that is dependent on the individual phannacodynamics of each agent and administered in FDA approved dosages in standard time courses.
METHODS OF THERAPY
The present invention also pertains to methods of treatment (prophylactic and/or therapeutic) for a susceptibility to a disease or condition associated with an NR, using an NR therapeutic agent. A "NR therapeutic agent" is an agent that alters (e.g., enhances or inhibits) NR polypeptide activity and/or NR gene expression, as described herein (e.g., an NR agonist or antagonist). NR therapeutic agents can alter NR polypeptide activity or gene expression by a variety of means, such as, for example, by providing additional NR polypeptide or by upregulating the transcription or translation of the NR gene; by altering posttranslational processing of the NR polypeptide; by altering transcription of NR splicing variants; or by interfering with NR polypeptide activity (e.g. , by binding to an NR polypeptide), or by downregulating the transcription or translation of an NR gene. Representative NR therapeutic agents include the following: nucleic acids or fragments or derivatives thereof described herein, particularly nucleotides encoding the polypeptides described herein and vectors comprising such nucleic acids (e.g., a gene, cDNA, and/or mRNA, such as a nucleic acid encoding an NR polypeptide or active fragment or derivative thereof, or an oligonucleotide; for example, one of SEQ ID NO: 1, or a complement thereof, or a nucleic acid encoding SEQ ID NOS:3-4, or fragments or derivatives thereof); polypeptides described herein (e.g., SEQ JD NO: 2, and/or other splicing variants encoded by an NR, or fragments or derivatives thereof); other polypeptides (e.g., G-proteins); NR binding agents; peptidomimetics; fusion proteins or prodrugs thereof; antibodies (e.g., an antibody to a mutant NR polypeptide, or an antibody to a non-mutant NR polypeptide, or an antibody to a particular splicing variant encoded by an NR, as described above); ribozymes; other small molecules; and other agents that alter (e.g., enhance or inhibit) NR gene expression or polypeptide activity, or that regulate transcription of NR splicing variants (e.g., agents that affect which splicing variants are expressed, or that affect the amount of each splicing variant that is expressed. More than one NR therapeutic agent can be used concurrently, if desired.
An NR therapeutic agent that is a nucleic acid is used in the treatment of a susceptibility to a disease or condition associated with an NR. The term, "treatment" as used herein, refers not only to ameliorating symptoms associated with the disease, but also preventing or delaying the onset of the disease, and also lessening the severity or frequency of symptoms of the disease. The therapy is designed to alter (e.g., inhibit or enhance), replace or supplement activity of anNR polypeptide in an individual. For example, an NR therapeutic agent can be administered in order to upregulate or increase the expression or availability of the NR gene or of specific splicing variants of NR, or, conversely, to downregulate or decrease the expression or availability of the NR gene or specific splicing variants of the NR. Upregulation or increasing expression or availability of a native NR gene or of a particular splicing variant could interfere with or compensate for the expression or activity of a defective gene or another splicing variant; downregulation or decreasing expression or availability of a native NR gene or of a particular splicing variant could minimize the expression or activity of a defective gene or the particular splicing variant and thereby minimize the impact of the defective gene or the particular splicing variant.
The NR therapeutic agent(s) are administered in a therapeuticaUy effective amount (i.e., an amount that is sufficient to treat the disease, such as by ameliorating symptoms associated with the disease, preventing or delaying the onset of the disease, and/or also lessening the severity or frequency of symptoms of the disease). The amount which will be therapeuticaUy effective in the treatment of a particular individual's disorder or condition will depend on the symptoms and severity of the disease, and can be determined by standard clinical techniques. In addition, in vitro or in vivo assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of a practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems. In one embodiment, a nucleic acid of the invention (e.g., a nucleic acid encoding an NR polypeptide, such as SEQ ID NO: 1, or a complement thereof; or another nucleic acid that encodes an NR polypeptide or a splicing variant, derivative or fragment thereof, such as a nucleic acid encoding SEQ ID NOS: 1-2) can be used, either alone or in a pharmaceutical composition as described above. For example, an NR or a cDNA encoding an NR polypeptide, either by itself or included within a vector, can be introduced into cells (either in vitro or in vivo) such that the cells produce native NR polypeptide. If necessary, cells that have been transformed with the gene or cDNA or a vector comprising the gene or cDNA can be introduced (or re-introduced) into an individual affected with the disease. Thus, cells which, in nature, lack native NR expression and activity, or have mutant NR expression and activity, or have expression of a disease-associated NR splicing variant, can be engineered to express the NR polypeptide or an active fragment of the NR polypeptide (or a different variant of the NR polypeptide). hi a preferred embodiment, nucleic acid encoding an NR polypeptide, or an active fragment or derivative thereof, can be introduced into an expression vector, such as a viral vector, and the vector can be introduced into appropriate cells in an animal. Other gene transfer systems, including viral and nonviral transfer systems, can be used. Alternatively, nonviral gene transfer methods, such as calcium phosphate coprecipitation, mechanical techniques (e.g., microinjection); membrane fusion- mediated transfer via liposomes; or direct DNA uptake, can also be used.
Alternatively, in another embodiment of the invention, a nucleic acid of the invention; a nucleic acid complementary to a nucleic acid of the invention; or a portion of such a nucleic acid (e.g., an oligonucleotide as described below), can be used in "antisense" therapy, in which a nucleic acid (e.g., an oligonucleotide) which specifically hybridizes to the mRNA and/or genomic DNA of an NR is administered or generated in situ. The antisense nucleic acid that specifically hybridizes to the mRNA and/or DNA inhibits expression of the NR polypeptide, e.g., by inhibiting translation and/or transcription. Binding of the antisense nucleic acid can be by conventional base pair complementarity, or, for example, in the case of binding to DNA duplexes, through specific interaction in the major groove of the double helix. An antisense construct of the present invention can be delivered, for example, as an expression plasmid as described above. When the plasmid is transcribed in the cell, it produces RNA which is complementary to a portion of the mRNA and/or DNA which encodes the NR polypeptide. Alternatively, the antisense construct can be an oligonucleotide probe which is generated ex vivo and introduced into cells; it then inhibits expression by hybridizing with the mRNA and/or genomic DNA of the NR. hi one embodiment, the oligonucleotide probes are modified oligonucleotides which are resistant to endogenous nucleases, e.g. exonucleases and/or endonucleases, thereby rendering them stable in vivo. Exemplary nucleic acid molecules for use as antisense oligonucleotides are phosphoramidate, phosphothioate and methylphosphonate analogs of DNA (see also U.S. Pat. NO. 5,176,996; 5,264,564; and 5,256,775). Additionally, general approaches to constructing oligomers useful in antisense therapy are also described, for example, by Van der Krol et αl, (Biotechniques 6:958-976 (1988)); and Stein et αl. (Cancer Res. 48:2659-2668 (1988)). With respect to antisense DNA, oligodeoxyribonucleotides derived from the translation initiation site are preferred. To perform antisense therapy, oligonucleotides (mRNA, cDNA or DNA) are designed that are complementary to mRNA encoding the NR. The antisense oligonucleotides bind to NR mRNA transcripts and prevent translation. Absolute complementarity, although preferred, is not required. A sequence "complementary" to a portion of an RNA, as referred to herein, indicates that a sequence has sufficient complementarity to be able to hybridize with the RNA, forming a stable duplex; in the case of double-stranded antisense nucleic acids, a single strand of the duplex DNA may thus be tested, or triplex formation may be assayed. The ability to hybridize will depend on both the degree of complementarity and the length of the antisense nucleic acid, as described in detail above. Generally, the longer the hybridizing nucleic acid, the more base mismatches with an RNA it may contain and still form a stable duplex (or triplex, as the case may be). One skilled in the art can ascertain a tolerable degree of mismatch by use of standard procedures.
The oligonucleotides used in antisense therapy can be DNA, RNA, or chimeric mixtures or derivatives or modified versions thereof, single-stranded or double-stranded. The oligonucleotides can be modified at the base moiety, sugar moiety, or phosphate backbone, for example, to improve stability of the molecule, hybridization, etc. The oligonucleotides can include other appended groups such as peptides (e.g., for targeting host cell receptors in vivo), or agents facilitating transport across the cell membrane (see, e.g., Letsinger et al, Proc. Natl. Acad. Sci. USA 86:6553-6556 (1989); Lemaitie et al, Proc. Natl. Acad Sci. USA 84:648-652 (1987); PCT International Publication No. WO 88/09810) or the blood-brain barrier (see, e.g., PCT International Publication No. WO 89/10134), or hybridization-triggered cleavage agents (see, e.g., Krol et al, Bio/Techniques 6:958-976 (1988)) or intercalating agents. (See, e.g., Zon, Pharm. Res. 5:539-549 (1988)). To this end, the oligonucleotide maybe conjugated to another molecule (e.g., a peptide, hybridization triggered cross-linking agent, transport agent, hybridization-triggered cleavage agent).
The antisense molecules are delivered to cells which express NR in vivo. A number of methods can be used for delivering antisense DNA or RNA to cells; e.g., antisense molecules can be injected directly into the tissue site, or modified antisense molecules, designed to target the desired cells (e.g., antisense linked to peptides or antibodies that specifically bind receptors or antigens expressed on the target cell surface) can be administered systematically. Alternatively, in a preferred embodiment, a recombinant DNA construct is utilized in which the antisense oligonucleotide is placed under the control of a strong promoter (e.g., pol JJI or pol JJ). The use of such a construct to transfect target cells in the patient results in the transcription of sufficient amounts of single stranded RNAs that will form complementary base pairs with the endogenous NR transcripts and thereby prevent translation of the NR mRNA. For example, a vector can be introduced in vivo such that it is taken up by a cell and directs the transcription of an antisense RNA. Such a vector can remain episomal or become chromosomally integrated, as long as it can be transcribed to produce the desired antisense RNA. Such vectors can be constructed by recombinant DNA technology methods standard in the art and described above. For example, a plasmid, cosmid, YAC or viral vector can be used to prepare the recombinant DNA construct which can be introduced directly into the tissue site. Alternatively, viral vectors can be used which selectively infect the desired tissue, in which case administration may be accomplished by another route (e.g., systemically).
Endogenous NR expression can also be reduced by inactivating or "knocking out" NR or its promoter using targeted homologous recombination (e.g., see Smithies et al, Nature 317:230-234 (1985); Thomas & Capecchi, Cell 51:503-512 (1987); Thompson et al, Cell 5:313-321 (1989)). For example, a mutant, non-functional NR (or a completely unrelated DNA sequence) flanked by DNA homologous to the endogenous NR (either the coding regions or regulatory regions of NR) can be used, with or without a selectable marker and/or a negative selectable marker, to transfect cells that express the NR in vivo. Insertion of the DNA construct, via targeted homologous recombination, results in inactivation of the NR. The recombinant DNA constructs can be directly administered or targeted to the required site in vivo using appropriate vectors, as described above. Alternatively, expression of non-mutant NRs can be increased using a similar method: targeted homologous recombination can be used to insert a DNA construct comprising a non- mutant, functional NR, e.g., a gene having SEQ ID NO: 1, or the complement thereof, or a portion thereof, in place of a mutant NR in the cell, as described above, hi another embodiment, targeted homologous recombination can be used to insert a DNA construct comprising a nucleic acid that encodes an NR polypeptide variant that differs from that present in the cell.
Alternatively, endogenous NR expression can be reduced by targeting deoxyribonucleotide sequences complementary to the regulatory region of an NR (i.e., the NR promoter and/or enhancers) to form triple helical structures that prevent transcription of the NR in target cells in the body. (See generally, Helene, C, Anticancer Drug Des. 6(6):569-84 (1991); Helene, C, et al, Ann. N. Y. Acad. Sci. 660:27-36 (1992); and Maher, L. J., Bioassays 14(12):807-15 (1992)). Likewise, the antisense constructs described herein, by antagonizing the normal biological activity of one of the NR proteins, can be used in the manipulation of tissue, e.g., tissue differentiation, both in vivo and for ex vivo tissue cultures. Furthermore, the anti-sense techniques (e.g., microinjection of antisense molecules, or transfection with plasmids whose transcripts are anti-sense with regard to an NR mRNA or gene sequence) can be used to investigate the role of one or NR in developmental events, as well as the normal cellular function of the NRs in adult tissue. Such techniques can be utilized in cell culture, but can also be used in the creation of transgenic animals. In yet another embodiment of the invention, other NR therapeutic agents as described herein can also be used in the treatment or prevention of a susceptibility to a disease or condition associated with an NR. The therapeutic agents can be delivered in a composition, as described above, or by themselves. They can be administered systemically, or can be targeted to a particular tissue. The therapeutic agents can be produced by a variety of means, including chemical synthesis; recombinant production; in vivo production (e.g., a transgenic animal, such as U.S. Pat. No. 4,873,316 to Meade et αl), for example, and can be isolated using standard means such as those described herein.
A combination of any of the above methods of treatment (e.g., administration of non-mutant NR polypeptide in conjunction with antisense therapy targeting mutant NR mRNA; administration of a first splicing variant encoded by an NR in conjunction with antisense therapy targeting a second splicing encoded by an NR), can also be used.
The teachings of all publications cited herein are incorporated herein by reference in their entirety.
While this invention has been particularly shown and described with references to preferred embodiments thereof, it will be understood by those skilled in the art that various changes in form and details may be made therein without departing from the scope of the invention encompassed by the appended claims. Table I
MOOSE04608 ctgl7328 3473973..3474369, 3497170..3497264,
3539651..3539696, 3559055..3559165, 3559637.-3559798, 3560481..3560549, 3562954..3563118, 3563704..3563898, 3564786..3564850 5 MSSDDPvHLGSSCGSFIKTEPSSPSSGJDALSHHSPSGSSDASGGFGLALGTFiA NGLDSPPMFAGAGLGGTPCRKSYEDCASGLMEDSAIKCEYMLNAIPKRLCLV CGDJASGYHYGVASCEACKAFFKRTIQGNIEYSCPATNECEITKRRRKSCQA CRFMKCLACRFMKCLKVGMLKEGVRLDRVRGGRQKYKRRLDSESSPYLSL QISPPAKKPLTKJVSYLLVAEPDiaYAMPPPGMPEGDIKALTTLCDLADRELV l o VΠGWAKHJPDFKGLSGAESVCSMFQAVWTSLRCPLIGVVAETRNLFCPKDPI
WT"^-ADIQGELGSLRJPAJVQVSLTLSYPKCRFFTLNKTIFLGQQSSKWEHEEP EALGRPESLDEAVLPAVSPAQGHPCTPRGTGHPQTLPVASSGASWSQSMHH HVRLIFKLFVEMVFCHVA (SEQ ID NO: 2) atgtcctcggacgacaggcacctgggctccagctgcggctccttcatcaagactgagccgtccagcccgtcctcgggcat
15 cgatgccctcagccaccacagccccagtggctcgtccgacgccagcggcggctttggcctggccctgggcacccacgc caacggtctggactcgccacccatgtttgcaggcgccgggctgggaggcaccccatgccgcaagagctacgaggactg tgccagcggcatcatggaggactcggccatcaagtgcgagtacatgctcaacgccatccccaagcgcctgtgcctcgtgt gcggggacattgcctctggctaccactacggcgtggcctcctgcgaggcttgcaaggccttcttcaagaggactatccaa gggaacattgagtacagctgcccggccaccaacgagtgcgagatcaccaaacggaggcgcaagtcctgccaggcctg 0 ccgcttcatgaaatgcctcgcctgccgcttcatgaaatgcctcaaagtggggatgctgaaggaaggtgtgcgccttgatcg agtgcgtggaggccgtcagaaatacaagcgacggctggactcagagagcagcccatacctgagcttacaaatttctcca cctgctaaaaagccattgaccaagattgtctcatacctactggtggctgagccggacaagctctatgccatgcctccccctg gtatgcctgagggggacatcaaggccctgaccactctctgtgacctggcagaccgagagcttgtggtcatcattggctgg gccaagcacatcccagattttaagggcctttctggtgcggaaagtgtgtgttccatgtttcaggctgtgtggacttctcttcgc 5 tgtcccctgataggagttgtggctgagacaaggaatttattttgccccaaggatcccatttggaacaaggctgacatccagg gggagcttggttcactcagaatcccagccattgttcaagtcagtctgacattgtcctaccccaaatgcaggttttttaccttga ataaaacaattttcctggggcagcagagctccaagtgggagcatgaggagccagaagctctgggcaggcccgagagcc ttgatgaggcagtgctgcctgcagtgagcccggcacagggacacccctgcactcccaggggcacggggcacccccag accctcccggtagcctcttcaggggcctcttggtcccagagcatgcaccaccatgtccggctaatttttaaattatttgtaga 0 gatggtgttttgccatgttgcc (SEQ ID NO: 1) Table π
ctgl7328_MOOSE04608.xml 3B Estrogen-related
π m- ΉDNVLSII ps^pj-usgoμsg aε (809W)HSOOPM) sβzzoasoow
9l7:doιpαoi9UojoaD VaHD iri oi
Figure imgf000062_0001
1/snooι
Bra9iqdoziqos
qπ εεx- ΉONVISICI
Figure imgf000062_0002
aε (soθ oasoow) sβzzoasooiΛ.
01 :doj αθl9uojoa£) a]A[O ZS'£-V°^1 lSπa∞ψvm jsnooi;
Figure imgf000062_0003
qπ rarz, :aoNV Siα
Figure imgf000062_0004
aε (809W)asoow) sβzzoasoow 01
8 :doxp αOT Quo jo aONV WD Z/ φo ISZSHFJ∞PBlfl: csnooq
Braiμsy
qπ ε-ει- ΉONVLSKI pa ∞-røsαiisa aε (809W)asooκ) sβz oasoow
ox :dojpαoi9uojoao aiΛ[0 ε' oa wois a∞ψ s ooa c. tsp pypTo ran9TOT S
Figure imgf000062_0005
-19" 9sεzo/zoaι/x3d zoo/εo OΛV

Claims

CLAJJVISWhat is claimed is:
1. An isolated nucleic acid molecule comprising a nuclear receptor (NR) gene, wherein the NR gene has a nucleotide sequence as shown in Table I, or the complements of the nucleic acid sequences as shown in Table I.
2. A nucleic acid encoding a polypeptide, wherein the polypeptide has an amino acid sequence of the group of amino acid sequence as shown in Table I.
3. An isolated nucleic acid molecule which hybridizes under high stringency conditions to a nucleotide sequence as shown in Table I, or the complements of the group of nucleic acid sequences as shown in Table I.
4. An isolated nucleic molecule which hybridizes under high stringency conditions to a nucleotide sequence encoding an amino acid sequence as shown in Table I.
5. A method for assaying for the presence of a first nucleic acid molecule in a sample, comprising contacting said sample with a second nucleic acid molecule, where the second nucleic acid molecule comprises a nucleotide sequence as shown in Table I, and hybridizes to the first nucleic acid under high stringency conditions.
6. A vector comprising an isolated nucleic acid molecule of: (a) the nucleic acid sequence as shown in Table I;
(b) the complement of one of the nucleic acid sequence as shown in Table I; or
(c) a nucleic acid encoding an amino acid molecule as shown in Table I; where the nucleic acid molecule is operably linked to a regulatory sequence.
7. A recombinant host cell comprising the vector of Claim 6.
8. A method for producing a polypeptide encoded by an isolated nucleic acid molecule, comprising culturing the recombinant host cell of Claim 7 under conditions suitable for expression of the nucleic acid molecule.
9. An isolated polypeptide encoded by the nucleotide sequence as shown in Table I, or the complements thereof.
10. The isolated polypeptide of Claim 9, wherein the polypeptide has an amino acid sequence as shown in Table I.
11. An isolated polypeptide comprising an amino acid sequence, wherein the amino acid sequence is greater than about 95% identical to an amino acid sequence as shown in Table I.
12. A fusion protein comprising an isolated polypeptide of Claim 2.
13. A fusion protein comprising an isolated polypeptide of Claim 11.
14. An antibody, or an antigen-binding fragment thereof, wliich selectively binds to a polypeptide of Claim 2, or to a fragment or variant of said amino acid sequence.
15. An antibody, or an antigen-binding fragment thereof, which selectively binds to a polypeptide of Claim 11, or to a fragment or variant of said amino acid sequence.
16. A method of assaying for the presence of a polypeptide encoded by an isolated nucleic acid molecule according to Claim 1 in a sample, the method comprising contacting the sample with an antibody which specifically binds to the encoded polypeptide.
17. A method of identifying an agent which alters the activity of an NR, the method comprising:
(a) contacting a polypeptide of Claim 9, or a derivative or fragment thereof, with an agent to be tested; (b) assessing the level of activity of the polypeptide or derivative or fragment thereof; and (c) comparing the level of activity with a level of activity of the polypeptide or active derivative or fragment thereof in the absence of the agent; wherein if the level of activity of the polypeptide or derivative or fragment thereof in the presence of the agent differs, by an amount that is statistically significant, from the level in the absence of the agent, then the agent is an agent that alters activity of an NR.
18. An agent which alters the activity of an NR, identifiable according to the method of Claim 17.
19. The agent of Claim 18, where the agent is of: an NR gene binding agent; a peptidomimetic; a fusion protein; a prodrug; an antibody; and a ribozyme.
20. A method of altering activity of a polypeptide encoded by an NR gene, comprising contacting the polypeptide with an agent of Claim 19.
21. A method of identifying an agent which alters interaction of the polypeptide of Claim 9 with an NR gene binding agent, comprising: a) contacting the polypeptide or a derivative or fragment thereof, and the binding agent, with an agent to be tested; b) assessing the interaction of the polypeptide or derivative or fragment thereof with the binding agent; and c) comparing the level of interaction with a level of interaction of the polypeptide or derivative or fragment thereof with the binding agent in the absence of the agent, wherein if the level of interaction of the polypeptide or derivative or fragment thereof in the presence of the agent differs by an amount that is statistically significant, from the level of interaction in the absence of the agent, then the agent is an agent that alters interaction of the polypeptide with the binding agent.
22. An agent which alters interaction of an NR gene polypeptide with an NR gene binding agent, identifiable according to the method of Claim 21.
23. An agent which alters interaction of an NR gene polypeptide with an NR gene binding agent, selected from the group consisting of: a second NR gene binding agent; a peptidomimetic; a fusion protein; a prodrug; an antibody; and a ribozyme.
24. A method of altering interaction of an NR gene polypeptide with an NR gene binding agent, comprising contacting the NR gene polypeptide and/or the NR gene binding agent with an agent of Claim 23.
25. A method of identifying an agent which alters expression of an NR gene, comprising the steps of: a) contacting a solution containing a nucleic acid comprising the promoter region of the NR gene operably linked to a reporter gene with an agent to be tested; b) assessing the level of expression of the reporter gene; and c) comparing the level of expression with a level of expression of the reporter gene in the absence of the agent, wherein if the level of expression of the reporter gene in the presence of the agent differs, by an amount that is statistically significant, from the level of expression in the absence of the agent, then the agent is an agent that alters expression of the NR gene.
26. An agent which alters expression of the NR gene, identifiable according to the method of Claim 25.
27. A method of identifying an agent which alters expression of an NR gene, comprising the steps of: a) contacting a solution containing a nucleic acid of Claim 1 or a derivative or fragment thereof with an agent to be tested; b) assessing expression of the nucleic acid, derivative or fragment; and c) comparing expression with expression of the nucleic acid, derivative or fragment in the absence of the agent, wherein if expression of the nucleotide, derivative or fragment in the presence of the agent differs, by an amount that is statistically significant, from the expression in the absence of the agent, then the agent is an agent that alters expression of the NR gene.
28. The method of Claim 27, wherein the expression of the nucleotide, derivative or fragment in the presence of the agent comprises expression of one or more splicing variant(s) that differ in kind or in quantity from the expression of one or more splicing variant(s) the absence of the agent.
29. An agent which alters expression of an NR gene, identifiable according to the method of Claim 27.
30. An agent which alters expression of an NR gene, selected from the group consisting of: antisense nucleic acid to an NR gene; an NR gene polypeptide; an NR gene receptor; an NR gene binding agent; a peptidomimetic; a fusion protein; a prodrug thereof; an antibody; and a ribozyme.
31. A method of altering expression of an NR gene, comprising contacting a cell containing an NR gene with an agent of Claim 30.
32. A method of identifying a polypeptide which interacts with an NR gene polypeptide, comprising employing a yeast two-hybrid system using a first vector which comprises a nucleic acid encoding a DNA binding domain and an NR gene polypeptide, splicing variant, or a fragment or derivative thereof, and a second vector which comprises a nucleic acid encoding a transcription activation domain and a nucleic acid encoding a test polypeptide, wherein if transcriptional activation occurs in the yeast two-hybrid system, the test polypeptide is a polypeptide which interacts with an NR polypeptide.
33. An NR gene therapeutic agent selected from the group consisting of: an NR gene or fragment or derivative thereof; a polypeptide encoded by an NR gene; an NR gene binding agent; a peptidomimetic; a fusion protein; a prodrug; an antibody; an agent that alters NR gene expression; an agent that alters activity of a polypeptide encoded by an NR gene; an agent that alters posttranscriptional processing of a polypeptide encoded by an NR gene; an agent that alters interaction of an NR gene with an NR gene binding agent; an agent that alters transcription of splicing variants encoded by an NR gene; and a ribozyme.
34. A pharmaceutical composition comprising an NR gene therapeutic agent of Claim 33.
35. The pharmaceutical composition of Claim 34, wherein the NR gene therapeutic agent is an isolated nucleic acid molecule comprising an NR gene or fragment or derivative thereof.
36. The pharmaceutical composition of Claim 34, wherein the NR gene therapeutic agent is a polypeptide encoded by the NR gene.
37. A method of treating a disease or condition associated with an NR in an individual, comprising administering an NR gene therapeutic agent to the individual, in a therapeuticaUy effective amount.
38. The method of Claim 37, wherein the NR gene therapeutic agent is an NR gene agonist.
39. The method of Claim 38 wherein the NR gene therapeutic agent is an NR gene antagonist.
40. A transgenic animal comprising a nucleic acid selected from the group consisting of: an exogenous NR gene and a nucleic acid encoding an NR gene polypeptide.
41. A method for assaying a sample for the presence of an NR gene nucleic acid, comprising: a) contacting said sample with a nucleic acid comprising a contiguous nucleotide sequence which is at least partially complementary to a part of the sequence of said NR gene nucleic acid under conditions appropriate for hybridization, and b) assessing whether hybridization has occurred between an NR gene nucleic acid and said nucleic acid comprising a contiguous nucleotide sequence which is at least partially complementary to a part of the sequence of said NR gene nucleic acid; where if hybridization has occurred, an NR gene is present in the nucleic acid.
42. The method of Claim 41, wherein said nucleic acid comprising a contiguous nucleotide sequence is completely complementary to a part of the sequence of said NR gene nucleic acid.
43. The method of Claim 41 , comprising amplification of at least part of said NR gene nucleic acid.
44. The method of Claim 41, wherein said contiguous nucleotide sequence is 100 or fewer nucleotides in length and is either: a) at least 80% identical to a contiguous sequence of nucleotides in one of the nucleic acid sequences as shown in Table I; b) at least 80% identical to the complement of a contiguous sequence of nucleotides in one of the nucleic acid sequences as shown in Table I; or c) capable of selectively hybridizing to said NR gene nucleic acid.
45. A reagent for assaying a sample for the presence of an NR gene nucleic acid, said reagent comprising a nucleic acid comprising a contiguous nucleotide sequence which is at least partially complementary to a part of the nucleotide sequence of said NR gene nucleic acid.
46. The reagent of Claim 45, wherein the nucleic acid comprises a contiguous nucleotide sequence which is completely complementary to a part of the nucleotide sequence of said NR gene nucleic acid.
47. A reagent kit for assaying a sample for the presence of an NR gene nucleic acid, comprising in separate containers: a) one or more labeled nucleic acids comprising a contiguous nucleotide sequence which is at least partially complementary to a part of the nucleotide sequence of said NR gene nucleic acid, and b) reagents for detection of said label.
48. The reagent kit of Claim 47, wherein the labeled nucleic acid comprises a contiguous nucleotide sequences which is completely complementary to a part of the nucleotide sequence of said NR gene nucleic acid.
49. A reagent kit for assaying a sample for the presence of an NR gene nucleic acid, comprising one or more nucleic acids comprising a contiguous nucleotide sequence which is at least partially complementary to a part of the nucleotide sequence of said NR gene nucleic acid, and which is capable of acting as a primer for said NR gene nucleic acid when maintained under conditions for primer extension.
50. The use of a nucleic acid which is 100 or fewer nucleotides in length and which is either: a) at least 80% identical to a contiguous sequence of as shown in Table I; b) at least 80% identical to the complement of a contiguous sequence of nucleotide as shown in Table I; or c) capable of selectively hybridizing to said NR gene nucleic acid, for assaying a sample for the presence of an NR gene nucleic acid.
51. The use of a first nucleic acid which is 100 or fewer nucleotides in length and which is either: a) at least 80% identical to a contiguous sequence of nucleotides as shown in Table I; b) at least 80% identical to the complement of a contiguous sequence of nucleotides in the nucleic acid sequence as shown in Table I; or c) capable of selectively hybridizing to said NR gene nucleic acid; for assaying a sample for the presence of an NR gene nucleic acid that has at least one nucleotide difference from the first nucleic acid.
52. The use of a nucleic acid which is 100 or fewer nucleotides in length and which is either: a) at least 80% identical to a contiguous sequence of nucleotides as shown in Table I; b) at least 80% identical to the complement of a contiguous sequence of nucleotides in the nucleic acid sequence as shown in Table I; or c) capable of selectively hybridizing to said NR gene nucleic acid; for diagnosing a susceptibility to a disease or condition associated with an NR.
PCT/IB2002/002356 2001-06-29 2002-06-24 Nucleic acids encoding nuclear receptors WO2003002741A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002304350A AU2002304350A1 (en) 2001-06-29 2002-06-24 Nucleic acids encoding nuclear receptors

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US30245201P 2001-06-29 2001-06-29
US60/302,452 2001-06-29
US33297601P 2001-11-06 2001-11-06
US60/332,976 2001-11-06

Publications (3)

Publication Number Publication Date
WO2003002741A2 true WO2003002741A2 (en) 2003-01-09
WO2003002741A3 WO2003002741A3 (en) 2003-03-13
WO2003002741A8 WO2003002741A8 (en) 2003-08-07

Family

ID=26972938

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2002/002356 WO2003002741A2 (en) 2001-06-29 2002-06-24 Nucleic acids encoding nuclear receptors

Country Status (2)

Country Link
AU (1) AU2002304350A1 (en)
WO (1) WO2003002741A2 (en)

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5071773A (en) * 1986-10-24 1991-12-10 The Salk Institute For Biological Studies Hormone receptor-related bioassays
EP0866127A2 (en) * 1997-03-17 1998-09-23 Smithkline Beecham Plc HE8AN36, a steroid hormone receptor homolog
EP0939123A2 (en) * 1998-01-15 1999-09-01 Smithkline Beecham Plc Estrogen receptor-related polypetide SBPERR4
WO2000042180A1 (en) * 1999-01-14 2000-07-20 Kyowa Hakko Kogyo Co., Ltd. Novel protein

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5071773A (en) * 1986-10-24 1991-12-10 The Salk Institute For Biological Studies Hormone receptor-related bioassays
EP0866127A2 (en) * 1997-03-17 1998-09-23 Smithkline Beecham Plc HE8AN36, a steroid hormone receptor homolog
EP0939123A2 (en) * 1998-01-15 1999-09-01 Smithkline Beecham Plc Estrogen receptor-related polypetide SBPERR4
WO2000042180A1 (en) * 1999-01-14 2000-07-20 Kyowa Hakko Kogyo Co., Ltd. Novel protein

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
CHEN F ET AL: "Identification of two hERR2-related novel nuclear receptors utilizing bioinformatics and inverse PCR" GENE, ELSEVIER BIOMEDICAL PRESS. AMSTERDAM, NL, vol. 228, no. 1-2, 4 March 1999 (1999-03-04), pages 101-109, XP004159140 ISSN: 0378-1119 *
GIGUÈRE ET AL: "Identification of a new class of steroid hormone receptors" RECHERCHE, LA, SOCIETE D'EDITIONS SCIENTIFIQUES. PARIS, FR, vol. 331, 7 January 1988 (1988-01-07), pages 91-94, XP002136381 ISSN: 0029-5671 *
PETTERSSON K ET AL: "EXPRESSION OF A NOVEL MEMBER OF ESTROGEN RESPONSE ELEMENT-BINDING NUCLEAR RECEPTORS IS RESTRICTED TO THE EARLY STAGES OF CHORION FORMATION DURING MOUSE EMBRYOGENESIS" MECHANISMS OF DEVELOPMENT, ELSEVIER SCIENCE IRELAND LTD, IE, vol. 54, no. 2, February 1996 (1996-02), pages 211-223, XP002914542 ISSN: 0925-4773 *
WHITFIELD G KERR ET AL: "Steroid hormone receptors: Evolution, ligands, and molecular basis of biologic function." JOURNAL OF CELLULAR BIOCHEMISTRY, no. Suppl. 32-33, 1999, pages 110-122, XP002224043 ISSN: 0730-2312 *
YANG N ET AL: "ESTROGEN-RELATED RECEPTOR, HERR1, MODULATES ESTROGEN RECEPTOR-MEDIATED RESPONSE OF HUMAN LACTOFERRIN GENE PROMOTER" JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY OF BIOLOGICAL CHEMISTS, BALTIMORE, MD, US, vol. 271, no. 10, 8 March 1996 (1996-03-08), pages 5795-5804, XP000941916 ISSN: 0021-9258 *

Also Published As

Publication number Publication date
AU2002304350A1 (en) 2003-03-03
WO2003002741A8 (en) 2003-08-07
WO2003002741A3 (en) 2003-03-13

Similar Documents

Publication Publication Date Title
US7244604B2 (en) HDAC9 polypeptides and polynucleotides and uses thereof
US20030054531A1 (en) Human stroke gene
US20020045577A1 (en) Human schizophrenia gene
EP1347992A2 (en) Polymorphic bone morphogenetic protein 2
CA2502359A1 (en) Susceptibility gene for myocardial infarction
WO2003000893A2 (en) Nucleic acids encoding g protein-coupled receptors
US20020094954A1 (en) Human schizophrenia gene
WO2003076658A2 (en) A susceptibility gene for late-onset idiopathic parkinson's disease
AU2003201728B2 (en) Gene for peripheral arterial occlusive disease
WO2003000901A2 (en) Nucleic acids encoding protein kinases
AU2003201728A1 (en) Gene for peripheral arterial occlusive disease
US20060141462A1 (en) Human type II diabetes gene-slit-3 located on chromosome 5q35
WO2003000735A2 (en) Nucleic acids encoding olfactory receptors
WO2003002741A2 (en) Nucleic acids encoding nuclear receptors
WO2003002606A2 (en) Nucleic acids encoding ion channels
US20040142440A1 (en) Seryl transfer RNA synthetase polynucleotides and polypeptides and methods of use thereof
WO2003040393A2 (en) Nucleic acids encoding proteases
US20020165144A1 (en) Human schizophrenia gene
WO2004065938A2 (en) Human osteoporosis gene
WO2003062469A2 (en) Gene matn3 or matrilin-3 linked to osteoarthritis treatment
WO2004041193A2 (en) HUMAN TYPE II DIABETES GENE-Kv CHANNEL-INTERACTING PROTEIN (KChIP1) LOCATED ON CHROMOSOME 5
WO2003040392A2 (en) Nucleic acids encoding very long chain fatty acid biosynthesis enzymes
JP2006515992A (en) Human osteoporosis gene
AU2002322112A1 (en) HDAC9 polypeptides and polynucleotides and uses thereof

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
AK Designated states

Kind code of ref document: A3

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: A3

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

CFP Corrected version of a pamphlet front page

Free format text: UNDER (54) PUBLISHED TITLE REPLACED BY CORRECT TITLE

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