WO1997047642A1 - Cathepsin k gene - Google Patents

Cathepsin k gene Download PDF

Info

Publication number
WO1997047642A1
WO1997047642A1 PCT/US1996/010346 US9610346W WO9747642A1 WO 1997047642 A1 WO1997047642 A1 WO 1997047642A1 US 9610346 W US9610346 W US 9610346W WO 9747642 A1 WO9747642 A1 WO 9747642A1
Authority
WO
WIPO (PCT)
Prior art keywords
sequence
cathepsin
polynucleotide
polypeptide
seq
Prior art date
Application number
PCT/US1996/010346
Other languages
French (fr)
Inventor
Christine Marie Debouck
Fred Drake
Maxine Gowen
Julie Rood
Gregg A. Hastings
Mark D. Adams
Claire M. Fraser
Norman H. Lee
Ewen F. Kirkness
Judith A. Blake
Lisa M. Fitzgerald
Original Assignee
Smithkline Beecham Corporation
Human Genome Sciences, Inc.
The Institute For Genomic Research
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 Smithkline Beecham Corporation, Human Genome Sciences, Inc., The Institute For Genomic Research filed Critical Smithkline Beecham Corporation
Priority to AU63332/96A priority Critical patent/AU6333296A/en
Priority to PCT/US1996/010346 priority patent/WO1997047642A1/en
Priority to PCT/US1996/014026 priority patent/WO1997047643A1/en
Priority to AU69092/96A priority patent/AU6909296A/en
Publication of WO1997047642A1 publication Critical patent/WO1997047642A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/48Hydrolases (3) acting on peptide bonds (3.4)
    • C12N9/50Proteinases, e.g. Endopeptidases (3.4.21-3.4.25)
    • C12N9/64Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue
    • C12N9/6421Proteinases, e.g. Endopeptidases (3.4.21-3.4.25) derived from animal tissue from mammals
    • C12N9/6472Cysteine endopeptidases (3.4.22)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y304/00Hydrolases acting on peptide bonds, i.e. peptidases (3.4)
    • C12Y304/22Cysteine endopeptidases (3.4.22)
    • C12Y304/22038Cathepsin K (3.4.22.38)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • This invention relates, in part, to newly identified polynucleotides and polypeptides; variants and derivatives of the polynucleotides and polypeptides; processes for making the polynucleotides and the polypeptides, and their variants and derivatives; agonists and antagonists of the polypeptides; and uses of the polynucleotides, polypeptides, variants, derivatives, agonists and antagonists.
  • the invention relates to polynucleotides and polypeptides of human cathepsin K, especially genomic sequences of cathepsin K, and most especially promoter and intronic sequences.
  • Bone resorption involves the simultaneous removal of both the mineral and the organic constituents of the extracellular matrix. This occurs mainly in an acidic phagolysosome-like extracellular compartment covered by the ruffled border of osteoclasts. Barron, et al., J. Cell Biol., 101:2210-22, (1985). Osteoclasts are multinucleate giant cells that play key roles in bone resorption. Attached to the bone surface, osteoclasts produce an acidic microenvironment between osteoclasts and bone matrix. In this acidic microenvironment, bone minerals and organic components are solubilized. Organic components, mainly type-I collagen, are thought to be solubilized by protease digestion.
  • cysteine proteinases may play an important role in the degradation of organic components of bone.
  • cathepsins B, L, H, and S can degrade type-I collagen in the acidic condition.
  • Cathepsin L is the most active of the lysosomal cysteine proteases with regard to its ability to hydrolyze azocasein, elastin, and collagen.
  • Cathepsins are proteases that function in the normal physiological as well as pathological degradation of connective tissue. Cathepsins play a major role in intracellular protein degradation and turnover, bone remodeling, and prohormone activation. Marx, J.L., Science. 235:285-286 (1987). Cathepsin B, H, L and S are ubiquitously expressed lysosomal cysteine proteinases that belong to the papain superfamily. They are found at constitutive levels in many tissues in the human including kidney, liver, lung and spleen. Some pathological roles of cathepsins include an involvement in glomerulonephritis, arthritis, and cancer metastasis.
  • cathepsins L and B may be involved in the remodelling of this tissue.
  • These lysosomal cysteine proteases digest extracellular matrix proteins such as elastin, laminin, and type I collagen under acidic conditions. Osteoclast cells require this activity to degrade the organic matrix prior to bone regeneration accomplished by osteoblasts.
  • Several natural and synthetic inhibitors of cysteine proteinases have been effective in inhibiting the degradation of this matrix.
  • the isolation of cathepsins and their role in bone resorption has been the subject of an intensive study.
  • OC-2 has recently been isolated from pure osteoclasts from rabbit bones.
  • the OC-2 was found to encode a possible cysteine proteinase structurally related to cathepsins L and S. Tezuka, K., et al., J. Biol. Chem., 269: 1106- 1109, (1994).
  • An inhibitor of cysteine proteinases and collagenase, Z-Phe-Ala-CHN 2 has been studied for its effect on the resorptive activity of isolated osteoclasts and has been found to inhibit resorption pits in dentine. Delaisse, J.M. et al., Bone, 8:305-313 (1987).
  • the polynucleotide comprises the region encoding human cathepsin K in the sequence set out in Figure 1 [SEQ ID NO: 1 ] or in the genomic DNA (herein "gDNA") in ATCC deposit No. 98035 (referred to herein as the deposited clone).
  • isolated nucleic acid molecules encoding human cathepsin K including mRNAs, cDNAs, genomic DNAs and, in further embodiments of this aspect of the invention, biologically, diagnostically, clinically or therapeutically useful variants, analogs or derivatives thereof, or fragments thereof, including fragments of the variants, analogs and derivatives.
  • cathepsin K polypeptides particularly human cathepsin K polypeptides, that cause or are associated with disease, for example, osteoporosis, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hyperparathyroidism, bone degradation, metastatic tumors, rhemuatoid arthritis, osteoarthritis, peridontal disease and degradation of bone implants and bone protheses, particularly dental implants.
  • disease for example, osteoporosis, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hyperparathyroidism, bone degradation, metastatic tumors, rhemuatoid arthritis, osteoarthritis, peridontal disease and degradation of bone implants and bone protheses, particularly dental implants.
  • cathepsin K novel polypeptides of human origin referred to herein as cathepsin K as well as biologically, diagnostically or therapeutically useful fragments, variants and derivatives thereof, variants and derivatives of the fragments, and analogs of the foregoing.
  • cathepsin K polypeptides comprising culturing host cells having expressibly inco ⁇ orated therein an exogenously-derived human cathepsin K-encoding polynucleotide under conditions for expression of human cathepsin K in the host and then recovering the expressed polypeptide.
  • products, compositions and methods for, among other things: assessing cathepsin K expression in cells by determining cathepsin K polypeptides of cathepsin K-encoding mRNA or hnRNA in vitro, ex vivo or in vivo by exposing cells to cathepsin K polypeptides, polynucleotides or antibodies as disclosed herein; assaying genetic variation and aberrations, such as defects, in cathepsin K polynucleotides, genes and gene control sequences; and administering a cathepsin K polypeptide or polynucleotide to an organism to augment cathepsin K function or remediate cathepsin K dysfunction.
  • probes that hybridize specifically to human cathepsin K sequences.
  • antibodies against cathepsin K polypeptides are provided.
  • the antibodies are highly selective for human cathepsin K.
  • cathepsin K agonists are provided.
  • preferred agonists are molecules that mimic cathepsin K, that bind to cathepsin K-binding molecules or receptor molecules, and that elicit or augment cathepsin K-induced responses.
  • cathepsin K antagonists are those which mimic cathepsin K so as to bind to cathepsin K receptor or binding molecules but not elicit a cathepsin K-induced response or more than one cathepsin K-induced response.
  • preferred antagonists are molecules that bind to or interact with cathepsin K so as to inhibit an effect of cathepsin K or more than one effect of cathepsin K.
  • the agonists and antagonists may be used to mimic, augment or inhibit the action of cathepsin K polypeptides. They may be used, for instance, to treat osteoporosis, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hyperparathyroidism, bone degradation, metastatic tumors, and degradation of bone implants and bone protheses, particularly dental implants.
  • Such antagonists may be particularly useful to treat osteoporosis, Paget's disease, Gaucher's disease, Alzheimer's disease, hyperparathyroidism, bone degradation, metastatic tumors, CNS inflammation, rhemuatoid arthritis, osteoarthritis, periodontal disease and degradation of bone implants and bone protheses, particularly dental implants.
  • compositions comprising a cathepsin K polynucleotide or a cathepsin K polypeptide for administration to cells in vitro, to cells ex vivo and to cells in vivo, or to a multicellular organism.
  • the compositions comprise a cathepsin K polynucleotide for expression of a cathepsin K polypeptide in a host organism for treatment of disease.
  • Particularly preferred in this regard is expression in a human patient for treatment of a dysfunction associated with aberrant endogenous activity of cathepsin K or to provide therapeutic.
  • Figure 1 shows the genomic nudeotide sequence of human cathepsin K [SEQ ID NO: 1].
  • Figure 2 shows the nudeotide, exon-intron boundaries and deduced amino acid sequence of human cathepsin K.
  • Figure 3 shows structural features of cathepsin K [SEQ ID NO: 2-19].
  • Figure 4 shows the intron-exon junctions.
  • Figure 5 shows the regions of similarity between amino acid sequences of cathepsin K, human cathepsins S, L, H, B, D, E, G and rabbit OC2 polypeptides.
  • Figure 6 shows the deduced amino acid sequence of human cathepsin K.
  • DIGESTION of DNA refers to catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA.
  • the various restriction enzymes referred to herein are commercially available and their reaction conditions, cofactors and other requirements for use are known and routine to the skilled artisan.
  • 1 ⁇ g of plasmid or DNA fragment is digested with about 2 units of enzyme in about 20 ml of reaction buffer.
  • isolating DNA fragments for plasmid construction typically 5 to 50 ⁇ g of DNA are digested with 20 to 250 units of enzyme in proportionately larger volumes.
  • GENETIC ELEMENT generally means a polynucleotide comprising a region that encodes a polypeptide or a region that regulates transcription or translation or other processes important to expression of the polypeptide in a host cell, or a polynucleotide comprising both a region that encodes a polypeptide and a region operably linked thereto that regulates expression.
  • Genetic elements may be comprised within a vector that replicates as an episomal element; that is, as a molecule physically independent of the host cell genome. They may be comprised within mini-chromosomes, such as those that arise during amplification of transfected DNA by methotrexate selection in eukaryotic cells. Genetic elements also may be comprised within a host cell genome; not in their natural state but, rather, following manipulation such as isolation, cloning and introduction into a host cell in the form of purified DNA or in a vector, among others.
  • IDENTITY means the degree of sequence relatedness between two polypeptide or two polynucleotides sequences as determined by the identity of the match between two strings of such sequences. Identity can be readily calculated.
  • identity is well known to skilled artisans (Computational Molecular Biology, Lesk, A.M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D.W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A.M., and Griffin, H.G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M.
  • Preferred computer program methods to determine identity between two sequences include, but are not limited to, GCG program package (Devereux, J., et al., Nucleic Acids Research 12(1): 387 (1984)), BLASTP, BLASTN, FASTA (Atschul, S.F. et al., J. Molec. Biol. 215: 403 (1990)).
  • GCG program package Digit, J., et al., Nucleic Acids Research 12(1): 387 (1984)
  • BLASTP BLASTN
  • FASTA Altschul, S.F. et al., J. Molec. Biol. 215: 403 (1990)
  • ISOLATED means altered "by the hand of man” from its natural state; i.e., that, if it occurs in nature, it has been changed or removed from its original environment, or both.
  • a naturally occurring polynucleotide or a polypeptide naturally present in a living animal in its natural state is not “isolated,” but the same polynucleotide or polypeptide separated from some or all of the coexisting materials of its natural is “isolated", as the term is employed herein.
  • polynucleotides can be joined to other polynucleotides, such as DNAs, for mutagenesis, to form fusion proteins, and for propagation or expression in a host, for instance.
  • the isolated polynucleotides, alone or joined to other polynucleotides such as vectors, can be introduced into host cells, in culture or in whole organisms. Introduced into host cells in culture or in whole organisms, such DNAs still would be isolated, as the term is used herein, because they would not be in their naturally occurring form or environment.
  • polynucleotides and polypeptides may occur in a composition, such as a media formulations, solutions for introduction of polynucleotides or polypeptides, for example, into cells, compositions or solutions for chemical or enzymatic reactions, for instance, which are not naturally occurring compositions, and, therein remain isolated polynucleotides or polypeptides within the meaning of that term as it is employed herein.
  • a composition such as a media formulations, solutions for introduction of polynucleotides or polypeptides, for example, into cells, compositions or solutions for chemical or enzymatic reactions, for instance, which are not naturally occurring compositions, and, therein remain isolated polynucleotides or polypeptides within the meaning of that term as it is employed herein.
  • LIGATION refers to the process of forming phosphodiester bonds between two or more polynucleotides, which most often are double stranded DNAs.
  • Techniques for ligation are well known to the art and protocols for ligation are described in standard laboratory manuals and references, such as, for instance, Sambrook et al., MOLECULAR CLONING, A LABORATORY MANUAL, 2nd Ed.; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1989) and Maniatis et al., pg. 146, as cited below.
  • OLIGONUCLEOTIDE(S) refers to relatively short polynucleotides. Often the term refers to single-stranded deoxyribonucleotides, but it can refer as well to single-, double-, or triple-stranded ribonucleotides, antisense polynucleotides, RNA:DNA hybrids and double-stranded DNAs, among others. Oligonudeotides, such as single-stranded DNA probe oligonudeotides, often are synthesized by chemical methods, such as those implemented on automated oligonudeotide synthesizers.
  • oligonudeotides can be made by a variety of other methods, including in vitro recombinant DNA-mediated techniques and by expression of DNAs in cells and organisms. Initially, chemically synthesized DNAs typically are obtained without a 5' phosphate. The 5' ends of such oligonudeotides are not substrates for phosphodiester bond formation by ligation reactions that employ DNA ligases typically used to form recombinant DNA molecules. Where ligation of such oligonudeotides is desired, a phosphate can be added by standard techniques, such as those that employ a kinase and ATP.
  • the 3' end of a chemically synthesized oligonudeotide generally has a free hydroxyl group and, in the presence of a ligase, such as T4 DNA ligase, readily will form a phosphodiester bond with a 5' phosphate of another polynucleotide, such as another oligonudeotide. As is well known, this reaction can be prevented selectively, where desired, by removing the 5' phosphates of the other polynucleotide(s) prior to ligation.
  • PLASMIDS generally are designated herein by a lower case letter/? preceded and/or followed by capital letters and/or numbers, in accordance with standard naming conventions that are familiar to those of skill in the art.
  • plasmids disclosed herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids by routine application of well known, published procedures.
  • Many plasmids and other cloning and expression vectors that can be used in accordance with the present invention are well known and readily available to those of skill in the art.
  • those of skill readily may construct any number of other plasmids suitable for use in the invention. The properties, construction and use of such plasmids, as well as other vectors, in the present invention will be readily apparent to those of skill from the present disclosure.
  • POLYNUCLEOTIDE(S) generally refers to any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA.
  • polynucleotides as used herein refers to, among others, single-and double-stranded DNA, DNA that is a mixture of single-and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions.
  • polynucleotide as used herein refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA.
  • the strands in such regions may be from the same molecule or from different molecules.
  • the regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules.
  • One of the molecules of a triple-helical region often is an oligonudeotide.
  • polynucleotide includes DNAs or RNAs as described above that contain one or more modified bases.
  • DNAs or RNAs with backbones modified for stability or for other reasons are “polynucleotides” as that term is intended herein.
  • DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples are polynucleotides as the term is used herein.
  • polynucleotide as it is employed herein embraces such chemically, enzymatically or metabolically modified forms of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including simple and complex cells, inter alia.
  • POLYPEPTIDES includes all polypeptides as described below.
  • the basic structure of polypeptides is well known and has been described in innumerable textbooks and other publications in the art.
  • the term is used herein to refer to any peptide or protein comprising two or more amino acids joined to each other in a linear chain by peptide bonds.
  • the term refers to both short chains, which also commonly are referred to in the art as peptides, oligopeptides and oligomers, for example, and to longer chains, which generally are referred to in the art as proteins, of which there are many types.
  • polypeptides often contain amino acids other than the 20 amino acids commonly referred to as the 20 naturally occurring amino acids, and that many amino acids, including the terminal amino acids, may be modified in a given polypeptide, either by natural processes, such as processing and other post-translational modifications, but also by chemical modification techniques which are well known to the art. Even the common modifications that occur naturally in polypeptides are too numerous to list exhaustively here, but they are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature, and they are well known to those of skill in the art.
  • polypeptides of the present are, to name an illustrative few, acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nudeotide or nudeotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cystine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as
  • polypeptides are not always entirely linear.
  • polypeptides may be branched as a result of ubiquitination, and they may be circular, with or without branching, generally as a result of posttranslation events, including natural processing event and events brought about by human manipulation which do not occur naturally.
  • Circular, branched and branched circular polypeptides may be synthesized by non-translation natural process and by entirely synthetic methods, as well.
  • Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini.
  • blockage of the amino or carboxyl group in a polypeptide, or both, by a covalent modification is common in naturally occurring and synthetic polypeptides and such modifications may be present in polypeptides of the present invention, as well.
  • the amino terminal residue of polypeptides made in E. coli, prior to proteolytic processing almost invariably will be N-formylmethionine.
  • polypeptides made by expressing a cloned gene in a host for instance, the nature and extent of the modifications in large part will be determined by the host cell posttranslational modification capacity and the modification signals present in the polypeptide amino acid sequence.
  • glycosylation often does not occur in bacterial hosts such as E. coli. Accordingly, when glycosylation is desired, a polypeptide should be expressed in a glycosylating host, generally a eukaryotic cell.
  • Insect cells often carry out the same posttranslational glycosylations as mammalian cells and, for this reason, insect cell expression systems have been developed to express efficiently mammalian proteins having native patterns of glycosylation, inter alia. Similar considerations apply to other modifications.
  • polypeptide encompasses all such modifications, particularly those that are present in polypeptides synthesized by expressing a polynucleotide in a host cell.
  • VARIANT(S) of polynucleotides or polypeptides are polynucleotides or polypeptides that differ from a reference polynucleotide or polypeptide, respectively. Variants in this sense are described below and elsewhere in the present disclosure in greater detail.
  • changes in the nudeotide sequence of the variant may be silent. That is, they may not alter the amino acids encoded by the polynucleotide. Where alterations are limited to silent changes of this type a variant will encode a polypeptide with the same amino acid sequence as the reference.
  • changes in the nudeotide sequence of the variant may alter the amino acid sequence of a polypeptide encoded by the reference polynucleotide. Such nudeotide changes may result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence, as discussed below.
  • a polypeptide that differs in amino acid sequence from another, reference polypeptide is Generally, differences are limited so that the sequences of the reference and the variant are closely similar overall and, in many region, identical.
  • a variant and reference polypeptide may differ in amino acid sequence by one or more substitutions, additions, deletions, fusions and truncations, which may be present in any combination.
  • RECEPTOR MOLECULE refers to molecules which bind or interact specifically with cathepsin K polypeptides of the present invention, including not only classic receptors and enzymatic substrates, both of which are preferred, but also other molecules that specifically bind to or interact with polypeptides of the invention (which also may be referred to as “binding molecules” and “interaction molecules,” respectively and as “cathepsin K binding molecules” and “cathepsin K interaction molecules.” These cathepsin K binding molecules also include, for example, cathepsin K substrate analogs.
  • Binding between polypeptides of the invention and such molecules, including receptor or binding or interaction molecules may be exclusive to polypeptides of the invention, which is very highly preferred, or it may be highly specific for polypeptides of the invention, which is highly preferred, or it may be highly specific to a group of proteins that includes polypeptides of the invention, which is preferred, or it may be specific to several groups of proteins at least one of which includes polypeptides of the invention.
  • Receptors also may be non-naturally occurring, such as antibodies and antibody-derived reagents that bind specifically to polypeptides of the invention.
  • the present invention relates to novel cathepsin K polypeptides and polynucleotides, among other things, as described in greater detail below.
  • the invention relates to polypeptides and polynucleotides of a novel human cathepsin K, which is related by amino acid sequence homology to rabbit OC-2 and human cathepsin O cDNA. Tezuka, K., et al., J. Biol. Chem., 269: 1106- 1109, (1994).
  • the invention relates especially to cathepsin K having the nudeotide sequences set out in Figure 1 [SEQ ID NO: 1], and to the cathepsin K nudeotide sequences of the gDNA in ATCC Deposit No. 98035, which is herein referred to as "the deposited clone” or as the "gDNA of the deposited clone.”
  • the nudeotide sequences set out in Figure 1 [SEQ ID NO: 1] were obtained by sequencing the gDNA of the deposited clone, as more specifically set forth elsewhere herein.
  • the sequence of the deposited clone is controlling as to any discrepancies between the two.
  • isolated polynucleotides which encode the cathepsin K polypeptide having the deduced amino acid sequence of Figure 2 [SEQ ID NO:20] (see also Figure 6 for the deduced amino acid sequence) or the cathepsin K polypeptide encoded by the gDNA in the deposited clone.
  • a polynucleotide of the present invention encoding human cathepsin K polypeptide may be obtained using standard cloning and screening procedures, such as those for cloning gDNAs using DNA from cells of a human as starting material.
  • standard cloning and screening procedures such as those for cloning gDNAs using DNA from cells of a human as starting material.
  • the polynucleotide set out in Figure 1 [SEQ ID NO: 1] was discovered in a human gDNA library as described in Example 1.
  • Human cathepsin K of the invention is structurally related to other proteins of the cathepsin family, as shown by the results of sequencing the gDNA encoding human cathepsin K in the deposited clone.
  • the gDNA sequence thus obtained is set out in Figure 1 [SEQ ID NO: 1]. It contains a non-contiguous open reading frame encoding, after intron removal, but including all exons, a protein of about 329 amino acid residues.
  • Polynucleotides of the present invention may be in the form of RNA, such as mRNA or hnRNA, or in the form of DNA, including, for instance, cDNA and gDNA obtained by cloning or produced by chemical synthetic techniques or by a combination thereof.
  • the DNA may be triple-stranded, double-stranded or single-stranded.
  • Single-stranded DNA may be the coding strand, also known as the sense strand, or it may be the non-coding strand, also referred to as the anti-sense strand.
  • the coding sequence which encodes the polypeptide may be identical to the exon sequence of the polynucleotide shown in Figure 1 [SEQ ID NO: 1] or that of the deposited clone.
  • It also may be a polynucleotide with a different sequence, which, as a result of the redundancy (degeneracy) of the genetic code, encodes the polypeptide of the DNA of Figure 2 [SEQ ID NO:20] or of the deposited gDNA, including, but not limited to, splice variants transcribed from such gDNA.
  • Polynucleotides of the present invention which encode the polypeptide of Figure 1 [SEQ ID NO: 1] or the polypeptide encoded by the deposited gDNA may include, but are not limited to the coding sequence for the mature polypeptide, by itself; the coding sequence for the mature polypeptide and additional coding sequences, such as those encoding a leader or secretory sequence, such as a pre-, or pro- or prepro- protein sequence; the coding sequence of the mature polypeptide, with or without the aforementioned additional coding sequences, together with additional, non-coding sequences, including for example, but not limited to introns and non-coding 5' and 3' sequences, such as the transcribed, non-translated sequences that play a role in transcription, mRNA processing - including splicing and polyadenylation signals, for example - ribosome binding and stability of mRNA; additional coding sequence which codes for additional amino acids, such as those which provide additional functionalities.
  • the polypeptide may be fused to a marker sequence, such as a peptide, which facilitates purification of the fused polypeptide.
  • the marker sequence is a hexa-histidine peptide, such as the tag provided in the vector pQE-9, among others, many of which are commercially available.
  • hexa-histidine provides for convenient purification of the fusion protein.
  • the HA tag corresponds to an epitope derived of influenza hemagglutinin protein, which has been described by Wilson et al., Cell 37: 767 ( 1984), for instance.
  • polynucleotide encoding a polypeptide encompasses polynucleotides which include a sequence encoding a polypeptide of the present invention, particularly the human cathepsin K having the amino acid sequence set out in Figure 2 [SEQ ID NO:20] or the amino acid sequence of the human cathepsin K encoded by the gDNA of the deposited clone.
  • the term encompasses polynucleotides that include a single continuous region or discontinuous regions encoding the polypeptide (for example, interrupted by introns) together with additional regions, that also may contain coding and/or non-coding sequences.
  • the present invention further relates to variants of the herein above described polynucleotides which encode for fragments, analogs and derivatives of the polypeptide having the deduced amino acid sequence of Figure 2 [SEQ ID NO:20] or the polypeptide encoded by the exons of the gDNA of the deposited clone, including, but not limited to, splice variants transcribed from such gDNA.
  • a variant of the polynucleotide may be a naturally occurring variant such as a naturally occurring allelic variant or splice variant, or it may be a variant that is not known to occur naturally.
  • Such non-naturally occurring variants of the polynucleotide may be made by mutagenesis techniques, including those applied to polynucleotides, cells or organisms.
  • Such non-naturally occurring variants of the polynucleotide may be made by modifying splice acceptor, donor and/or branch sites, or by expressing the gDNA in cells where it is not naturally expressed, or cell extracts made from such cells.
  • variants in this regard are variants that differ from the aforementioned polynucleotides by nudeotide substitutions, deletions or additions.
  • the substitutions, deletions or additions may involve one or more nucleotides.
  • the variants may be altered in coding or non-coding regions or both. Alterations in the coding regions may produce conservative or non-conservative amino acid substitutions, deletions or additions.
  • polynucleotides encoding cathepsin K variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments which have the amino acid sequence of the cathepsin K polypeptide of Figure 2 [SEQ ID NO:20] or of the deposit in which several, a few, 5 to 10, 1 to 5, 1 to 3, 2, 1 or no amino acid residues are substituted, deleted or added, in any combination.
  • silent substitutions, additions and deletions which do not alter the properties and activities of the cathepsin K.
  • conservative substitutions are particularly preferred in this regard.
  • polynucleotides that are at least 70% identical to a polynucleotide encoding the cathepsin K polypeptide having the amino acid sequence set out in Figure 2 [SEQ ID NO:20], or variants, close homologs, derivatives and analogs thereof, as described above, and polynucleotides which are complementary to such polynucleotides.
  • polynucleotides that comprise a region that is at least 80% identical to a polynucleotide encoding the cathepsin K polypeptide of the gDNA of the deposited clone and polynucleotides complementary thereto.
  • polynucleotides at least 90% identical to the same are particularly preferred, and among these particularly preferred polynucleotides, those with at least 95% are especially preferred. Furthermore, those with at least 97% are highly preferred among those with at least 95%, and among these those with at least 98% and at least 99% are particularly highly preferred, with at least 99% being the more preferred.
  • Still further preferred embodiments of the invention are polynucleotides comprising cathepsin K intron polynucleotide sequences, particularly polynucleotides comprising intron 1 [SEQ ID NO: 4], 2 [SEQ ID NO: 6], 3 [SEQ ID NO: 8], 4 [SEQ ID NO: 10], 5 [SEQ ID NO: 12], 6 [SEQ ID NO: 14] or 7[SEQ ID NO: 16], having the intron polynucleotide sequence set out in Figures 1 [SEQ ID NO: 1] and 3 [SEQ ID NO: 2-19], or variants, close homologs, derivatives and analogs thereof, as described above, and polynucleotides which are complementary to such polynucleotides.
  • Still other preferred embodiments of the invention are polynucleotides comprising cathepsin K exon polynucleotide sequences, particularly polynucleotides comprising exon 1 [SEQ ID NO: 3], 2 [SEQ ID NO: 5], 3 [SEQ ID NO: 7], 4 [SEQ ID NO: 9], 5 [SEQ ID NO: 1 1], 6 [SEQ ID NO: 13], 7 [SEQ ID NO: 15] or 8 [SEQ ID NO: 17], having the exon polynucleotide sequence set out in Figures 1 [SEQ ID NO: 1] and 3 [SEQ ID NO: 2-19], or variants, close homologs, derivatives and analogs thereof, as described above, and polynucleotides which are complementary to such polynucleotides.
  • polynucleotides comprising cathepsin K exon 1 [SEQ ID NO: 3], 2 [SEQ ID NO: 5], 3 [SEQ ID NO: 7], 4 [SEQ ID NO: 9], 5 [SEQ ID NO: 1 1], 6 [SEQ ID NO: 13], 7 [SEQ ID NO: 15] or 8 [SEQ ID NO: 17], operatively linked to the intron of a gene other than cathepsin K.
  • More preferred embodiments of the invention are differentially spliced polynucleotides, particularly those comprising any one or more of the following exon-exon pairs: 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 2-4, 2-5, 2-6, 2-7, 2-8, 3-4, 3-5, 3-6, 3-7, 3-8, 4-5, 4-6, 4-7, 4-8, 5-7, 5-8, or 6-8.
  • Particularly preferred embodiments of the invention are differentially spliced polynucleotides which encode polypeptides which function in cells, especially those which have a biological activity of cathepsin K, most especially those expressed in human cells.
  • Polynucleotides comprising exon-exon pairs may be a naturally occurring variant such as a naturally occurring splice variant, or it may be a variant that is not known to occur naturally.
  • Such non-naturally occurring variants of the polynucleotide may be made by mutagenesis techniques, including those applied to polynucleotides, cells or organisms.
  • Such non-naturally occurring variants of the polynucleotide may be made by modifying splice acceptor, donor and/or branch sites, or by expressing the gDNA in cells where it is not naturally expressed, or cell extracts made from such cells.
  • Exon-exon pairs can be full, fused exons or can be fused fragments of exons with a splice junction present.
  • Preferred exon-exon pairs comprising exon fragments may be made from at least two exons, one of which comprises an operable splice donor site and the other of which comprises an operable splice acceptor site and which both are operatively linked by an intron.
  • Particularly preferred embodiments in this respect are polynucleotides which encode polypeptides which retain substantially the same biological function or activity as the mature polypeptide encoded by the cDNA of Figure 2 [SEQ ID NO:20] or the gDNA of the deposited clone.
  • the present invention further relates to polynucleotides that hybridize to the herein above-described sequences.
  • the present invention especially relates to polynucleotides which hybridize under stringent conditions to the herein above-described polynucleotides.
  • stringent conditions means hybridization will occur only if there is at least 95% and preferably at least 97% identity between the sequences.
  • polynucleotides of the invention may be used as a hybridization probe for cDNA and genomic DNA to isolate full-length cDNAs and genomic clones encoding cathepsin K and to isolate cDNA and genomic clones of other genes that have a high sequence similarity to the human cathepsin K gene.
  • Such probes generally will comprise at least 15 bases.
  • such probes will have at least 30 bases and may have at least 50 bases.
  • Particularly preferred probes will have at least 30 bases and will have 50 bases or less.
  • the coding region of the cathepsin K gene may be isolated by screening using the known DNA sequence to synthesize an oligonudeotide probe.
  • a labeled oligonudeotide having a sequence complementary to that of a gene of the present invention is then used to screen a library of human cDNA, genomic DNA or mRNA to determine which members of the library the probe hybridizes to.
  • the polynucleotides and polypeptides of the present invention may be employed as research reagents and materials for discovery of treatments and diagnostics to human disease, as further discussed herein relating to polynucleotide assays, inter alia.
  • the polynucleotides may encode a polypeptide which is the mature protein plus additional amino or carboxyl-terminal amino acids, or amino acids interior to the mature polypeptide (when the mature form has more than one polypeptide chain, for instance).
  • Such sequences may play a role in processing of a protein from precursor to a mature form, may facilitate protein trafficking, may prolong or shorten protein half-life or may facilitate manipulation of a protein for assay or production, among other things.
  • the additional amino acids may be processed away from the mature protein by cellular enzymes.
  • a precursor protein, having the mature form of the polypeptide fused to one or more prosequences may be an inactive form of the polypeptide.
  • inactive precursors When prosequences are removed such inactive precursors generally are activated. Some or all of the prosequences may be removed before activation. Generally, such precursors are called proproteins.
  • a polynucleotide of the present invention may encode a mature protein, a mature protein plus a leader sequence (which may be referred to as a preprotein), a precursor of a mature protein having one or more prosequences which are not the leader sequences of a preprotein, or a preproprotein, which is a precursor to a proprotein, having a leader sequence and one or more prosequences, which generally are removed during processing steps that produce active and mature forms of the polypeptide.
  • a leader sequence which may be referred to as a preprotein
  • a precursor of a mature protein having one or more prosequences which are not the leader sequences of a preprotein or a preproprotein, which is a precursor to a proprotein, having a leader sequence and one or more prosequences, which generally are removed during processing steps that produce active and mature forms of the polypeptide.
  • Deposited materials A deposit containing a human cathepsin K gDNA has been deposited with the American Type Culture Collection, as noted above. Also as noted above, the gDNA deposit is referred to herein as "the deposited clone" or as "the gDNA of the deposited clone.” The deposited clone was deposited with the American Type Culture
  • the deposited material is a PI cosmid that contains the full length cathepsin K gDNA, referred to as "PlSacB2CatK/P129" upon deposit.
  • the deposit has been made under the terms of the Budapest Treaty on the international recognition of the deposit of micro-organisms for purposes of patent procedure. The strain will be irrevocably and without restriction or condition released to the public upon the issuance of a patent.
  • the deposit is provided merely as convenience to those of skill in the art and is not an admission that a deposit is required for enablement, such as that required under 35 U.S.C. section 112.
  • sequence of the polynucleotides contained in the deposited material, as well as the amino acid sequence of the polypeptide encoded thereby, are controlling in the event of any conflict with any description of sequences herein.
  • a license may be required to make, use or sell the deposited materials, and no such license is hereby granted.
  • the present invention further relates to a human cathepsin K polypeptide which has the deduced amino acid sequence of Figure 2 [SEQ ID NO:20], which is encoded by an unspliced or differentially spliced hnRNA or mRNA transcribed from the sequence of Figure 1 [SEQ ID NO: 1], or which has the amino acid sequence encoded by the deposited clone.
  • polypetides encoded by the cathepsin K gDNA comprising missense or nonsense mutations, or those polypeptides encoded by unspliced or partially spliced hnRNAs which still comprise at least one intron, particularly those polypeptides which are naturally found in cells, especially human cells.
  • Frameshift mutations have been shown to be associated with disease (Hoi, FA, et al. Journal of Medical Genetics, 1995, 32 (1), 52-56).
  • Preferred polypeptides provided by the invention are encoded by differentially spliced polynucleotides, particularly those polypeptides encoded by polynucleotides comprising any one or more of the following exon-exon pairs: 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 2-4, 2-5, 2-6, 2-7, 2-8, 3-4, 3-5, 3-6, 3-7, 3-8, 4-5, 4-6, 4-7, 4-8, 5-7, 5-8, or 6-8.
  • Particularly preferred embodiments of the invention are polypeptides encoded by differentially spliced polynucleotides, which polypeptides function in cells, especially those which have a biological activity of cathepsin K, most especially those expressed in human cells.
  • Still further preferred embodiments of the invention are polypeptides encoded by polynucleotides comprising exon polynucleotide sequences, particularly polynucleotides comprising cathepsin K exon 1 [SEQ ID NO: 3], 2 [SEQ ID NO: 5], 3 [SEQ ID NO: 7], 4 [SEQ ID NO: 9], 5 [SEQ ID NO: 11], 6 [SEQ ID NO: 13], 7 [SEQ ID NO: 15] or 8 [SEQ ID NO: 17], having the exon polynucleotide sequence set out in Figures 1 [SEQ ID NO: 1] and 3 [SEQ ID NO: 2-19], or variants, close homologs, derivatives and analogs thereof, as described above, and polypeptides encoded by polynucleotides which are complementary to such polynucleotides.
  • polypeptides encoded by polynucleotides comprising comprising cathepsin K exon exon 1 [SEQ ID NO: 3], 2 [SEQ ID NO: 5], 3 [SEQ ID NO: 7], 4 [SEQ ID NO: 9], 5 [SEQ ID NO: 1 1], 6 [SEQ ID NO: 13], 7 [SEQ ID NO: 15] or 8 [SEQ ID NO: 17], operatively liked to the intron of a gene other then cathepsin K, or joined to an exon of another gene.
  • the invention also relates to fragments, analogs and derivatives of these polypeptides.
  • fragment when referring to the polypeptide of Figure 2 [SEQ ID NO:20], a polypeptide encoded by an unspliced or differentially spliced hnRNA or mRNA transcribed from the sequence of Figure 1 [SEQ ID NO: 1], or that encoded by the deposited gDNA, means a polypeptide which retains essentially the same biological function or activity as such polypeptide.
  • an analog includes a proprotein which can be activated by cleavage of the proprotein portion to produce an active mature polypeptide.
  • the polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide. In certain preferred embodiments it is a recombinant polypeptide.
  • the fragment, derivative or analog of the polypeptide of Figure 2 [SEQ ID NO:20], or that encoded by an unspliced or differentially spliced hnRNA or mRNA transcribed from the sequence of Figure 1 [SEQ ID NO: 1], or that encoded by the gDNA in the deposited clone may be (i) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the mature polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the mature polypeptide, such as a leader or secretory sequence or a sequence which
  • particularly preferred embodiments of the invention in this regard are polypeptides having the amino acid sequence of cathepsin K set out in Figure 2 [SEQ ID NO:20], variants, analogs, derivatives and fragments thereof, and variants, analogs and derivatives of the fragments.
  • particularly preferred embodiments of the invention in this regard are polypeptides having the amino acid sequence of the cathepsin K of the gDNA in the deposited clone, variants, analogs, derivatives and fragments thereof, and variants, analogs and derivatives of the fragments.
  • substitutions are those that vary from a reference by conservative amino acid substitutions. Such substitutions are those that substitute a given amino acid in a polypeptide by another amino acid of like characteristics. 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, Tyr.
  • variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments having the amino acid sequence of the cathepsin K polypeptide of Figure 2 [SEQ ID NO: 20] or of the gDNA in the deposited clone, in which several, a few, 5 to 10, 1 to 5, 1 to 3, 2, 1 or no amino acid residues are substituted, deleted or added, in any combination.
  • silent substitutions, additions and deletions which do not alter the properties and activities of the cathepsin K.
  • conservative substitutions are also especially preferred in this regard.
  • polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.
  • polypeptides of the present invention include the polypeptide encoded by at least one of the exons of SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 1 1, SEQ ID NO: 13, SEQ ID NO: 15 or SEQ ID NO: 17, (in particular the mature polypeptide) as well as polypeptides which have at least 70% similarity (preferably at least 70% identity) to the polypeptide encoded by at least one of the exons of SEQ ID NO: SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 1 1, SEQ ID NO: 13, SEQ ID NO: 15 or SEQ ID NO: 17 and more preferably at least 90% similarity (more preferably at least 90% identity) to the polypeptide encoded by at least one of the exons of SEQ ID NO: SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 1 1 , SEQ ID NO
  • similarity between two polypeptides is determined by comparing the amino acid sequence and its conserved amino acid substitutes of one polypeptide to the sequence of a second polypeptide.
  • Fragments or portions of the polypeptides of the present invention may be employed for producing the corresponding full-length polypeptide by peptide synthesis; therefore, the fragments may be employed as intermediates for producing the full-length polypeptides. Fragments or portions of the polynucleotides of the present invention may be used to synthesize full-length polynucleotides of the present invention.
  • polypeptides comprising fragments of cathepsin K, most particularly fragments of the cathepsin K having the amino acids encoded by the exons set out in Figure 1 [SEQ ID NO: 1], or having the amino acid encoded by the exon sequence of the cathepsin K of the deposited clone, and exon fragments or variants and derivatives of the cathepsin K of Figure 1 [SEQ ID NO: 1] or of the deposited clone.
  • a fragment is a polypeptide having an amino acid sequence that entirely is the same as part but not all of the amino acid sequence of the aforementioned cathepsin K polypeptides and variants or derivatives thereof.
  • Such fragments may be "free-standing,” i.e., not part of or fused to other amino acids or polypeptides, such as, for example, an exon, or they may be comprised within a larger polypeptide of which they form a part or region.
  • the presently discussed fragments most preferably form a single continuous region.
  • several fragments may be comprised within a single larger polypeptide.
  • fragments in one aspect of the meaning intended herein refers to the portion or portions of a fusion polypeptide or fusion protein derived from cathepsin K.
  • polypeptide fragments of the invention there may be mentioned those which are encoded by the polynucleotide sequence comprising cathepsin K exon 1, 2, 3, 4, 5, 6, 7 or 8, having the exon or intron 1,2,3,4,5,6 or 7 polynucleotide sequences respectively as set out in Figures 1 [SEQ ID NO: 1] and 3 [SEQ ID NO: 2-19], or variants, close homologs, derivatives and analogs thereof, as described above, and polypeptides encoded by polynucleotides which are complementary to such polynucleotides.
  • about 65-90 amino acids in this context means a polypeptide fragment of 65 plus or minus several, a few, 5, 4, 3, 2 or 1 amino acids to 90 plus or minus several a few, 5, 4, 3, 2 or 1 amino acid residues, i.e., ranges as broad as 65 minus several amino acids to 90 plus several amino acids to as narrow as 65 plus several amino acids to 90 minus several amino acids.
  • Truncation mutants include cathepsin K polypeptides having the amino acid sequence encoded by the exons of Figure 1 [SEQ ID NO: 1], or of the deposited clone, or of variants or derivatives thereof, except for deletion of a continuous series of residues (that is, a continuous region, part or portion) that includes the amino terminus, or a continuous series of residues that includes the carboxyl terminus or, as in double truncation mutants, deletion of two continuous series of residues, one including the amino terminus and one including the carboxyl terminus. Fragments having the size ranges set out about also are preferred embodiments of truncation fragments, which are especially preferred among fragments generally.
  • fragments characterized by structural or functional attributes of cathepsin K are also preferred in this aspect of the invention.
  • Preferred embodiments of the invention in this regard include fragments that comprise alpha-helix and alpha-helix forming regions ("alpha-regions"), beta-sheet and beta-sheet-forming regions ("beta-regions"), turn and turn-forming regions ("turn-regions”), coil and coil-forming regions ("coil-regions”), hydrophilic regions, hydrophobic regions, alpha amphipathic regions, beta amphipathic regions, flexible regions, surface-forming regions and high antigenic index regions of cathepsin K.
  • Certain preferred regions include Garnier-Robson alpha-regions, beta-regions, turn-regions and coil-regions, Chou-Fasman alpha-regions, beta-regions and turn-regions, Kyte-Doolittle hydrophilic regions and hydrophilic regions, Eisenberg alpha and beta amphipathic regions, Karplus-Schulz flexible regions, Emini surface-forming regions and Jameson-Wolf high antigenic index regions.
  • fragments in this regard are those that comprise regions of cathepsin K that combine several structural features, such as several of the features set out above.
  • the exon sequences of Figure 1 [SEQ ID NO: 1] which all are characterized by encoding amino acid compositions highly characteristic of turn-regions, hydrophilic regions, flexible-regions, surface-forming regions, and high antigenic index-regions, are especially highly preferred regions.
  • Such regions may be comprised within a larger polypeptide or may be by themselves a preferred fragment of the present invention, as discussed above. It will be appreciated that the term "about” as used in this paragraph has the meaning set out above regarding fragments in general.
  • Further preferred regions are those that mediate activities of cathepsin K.
  • Most highly preferred in this regard are fragments that have a chemical, biological, antigenic or other activity of cathepsin K, including those with a similar activity or an improved activity, or with a decreased undesirable activity.
  • the invention also relates to, among others, polynucleotides encoding the aforementioned fragments, polynucleotides that hybridize to polynucleotides encoding the fragments, particularly those that hybridize under stringent conditions, and polynucleotides, such as PCR primers, for amplifying polynucleotides that encode the fragments.
  • preferred polynucleotides are those that correspondent to the preferred fragments, as discussed above.
  • polynucleotides are genetic elements of cathepsin K, including, but not being limited to, a polyadenylation region, enhancers, a promoter, a cap site introns, exons, and splice sites (references describing these elements include, Darnel, J. et al. Molecular Cell Biology, second edition, W.H. Freeman, New York (1990); Watson, J.D., et al. Molecular Biology of the Gene, Benjamin/Cummings Pub., Menlo Park, CA, (1987)).
  • Untranslated regions contain many elements important in regulating gene expression. Mutations and markers in these regions have also been associated with disease (Ozawa T, et al., European Journal of Immunogenetics, APR 1995, 22 (2), 163-169).
  • a preferred embodiment of the invention is the 5'UTR, particularly the sequence set forth in Figure 3(A) [SEQUENCE ID NO: 2]. Mutations and markers in the 5' UTR have been associated with disease (Carlock L, et al., Human Genetics, APR 1994, 93 (4), 457-459).
  • a particularly preferred polynucleotide is an enhancer and promoter in the 5' UTR region of the cathepsin K gDNA.
  • Enhancers are often found in the 5' UTR and upregulate gene expression (see Miller et al., Biotechniques 7: 980-990 (1989) for a general reference on promoters).
  • the enhancer of the present invention can be operatively fused to heterologous genes to upregulate gene expression. It is believed that the enhancer promoter will regulate tissue-specific gene expression, being particularly useful to express genes is osteoclast and leukocytes, particularly macrophages cells.
  • a particularly preferred polynucleotide is the enhancer promoter having the sequence set forth in Figure 3(A) [SEQUENCE ID NO: 2].
  • Transcription factors are often associated with the enhancer and promoter and act to modulate the function of these regions and binding sites for these factors have been described (Faisst, Steffen and Meyer, Silke, Nucleic Acids Research, Vol. 20, No. 1, pp. 3-26, 1991 ; Smale, Stephen T., Transcription: Mechanisms and Regulation, Raven Press, Ltd. pp. 63-81 (1994)). These sites bind such factors as, for example, Spl, Apl, and Ap3 which are involved in transcription initiation
  • FIG. 3(S) Preferred canonical binding sites for transcription factors are underlined in Figure 3(S) [SEQUENCE ID NO: 2].
  • the Pu Box in Figure 3(S) [SEQUENCE ID NO: 2] has been described to be present in a macrophage gene, a cell in which cathepsin K is also found (Zhang, Dong-Er, Mol. and Cell. Biol., Vol. 14, No. 1, pp. 373-381 (1994)).
  • the present invention provides a promoter region that is useful, among other things, for the mediation of tissue-specific expression in osteoclasts and leukocytes, particularly macrophages.
  • a Pu box (AGGAA), present in the enhancer and promoter region has also been observed in a macrophage cell line (THP1).
  • Pu boxes in the sequences in the invention are provided. These Pu boxes are believed to be active in the cathepsin K gene in macrophages.
  • the promoter is particularly useful for the study of the control of cathepsin K gene expression, particularly as a region to be probed to diagnose disease. Vitamin D response elements have been found in the 5'UTR of known genes (Kahlen, Jean-Pierre and Carlberg, Carsten, Biochemical & Biophysical Research Communications, Vol. 202, No. 3, pp.
  • a further preferred polynucleotide is a cap site located 49 base pairs upstream of the ATG start codon of the sequence set forth in ( Figure 2).
  • a further preferred embodiment of the invention is the promoter region of cathepsin K ( Figure 3(A) and (S) [SEQUENCE ID NO: 2]).
  • Functional promoter region sequences have been described (Corden, J., et al., Science, 209, pp. 1406- 1414 (1990)).
  • a non-canonical promoter region in the sequence of cathepsin K set forth in Figures 3(A) [SEQUENCE ID NO: 2] and (S) [SEQUENCE ID NO: 2] comprises an A-T rich stretch at 19-27 base pairs upstream of the start codon ATG. Mutations in the TATA box region of promoters have been shown to be associated with disease (Peltoketo H, et al., Genomics, 1994, 23 (1 ), 250-252).
  • the 3' untranslated region of cathepsin K is a preferred polynucleotide of the invention, especially that polynucleotide set forth in Figure 3(Q) [SEQUENCE ID NO: 18], especially that region set forth in Figure 3(R) [SEQUENCE ID NO: 19]. Mutations in the 3' UTR have been associated with disease (Saito A, et al., Journal of the American Society of Nephrology, 1994, 4 (9), 1649-1653; Payne SJ, et al., Human Molecular Genetics, 1994, 3 (2), 390).
  • the polyadenylation region set forth in Figure 3(Q) [SEQUENCE ID NO: 18] is also a preferred polynucleotide of the 3' UTR.
  • the polyadenylation region comprises two copies of the canonical polyadenylation hexanucleotide, AAT AAA.
  • the polyadenylation region can be used, for example, in expression vectors to mediate mRNA 3' end formation (see, for example Gil, A. et al, Nature 312:413-414 (London) (1984)).
  • polynucleotides of the invention are the splice sites, including, but not limited to the splice donors, splice acceptors and the splice branchpoint. Splice junctions formation is essential for the proper creation of an open reading frame (Mount, Stephen, M., Department of Molecular Biophysics and Biochemistry, Yale University, Sterling Hall of Medicine, New Haven, CT, USA, IRL Press Limited, London, pp. 459-472 (1981)). Diseases associated with the improper formation of the splice junction are known. Particularly preferred splice junction polynucleotides are set forth in Figure 4. Introns comprise elements important in gene expression and in the formation of mature mRNA.
  • Preferred embodiments of the invention are the cathepsin K introns, particularly those introns having the sequences set forth in Figure 3 (C, E, G, I, K, M, and O) [SEQUENCE ID NO: 4, 6, 8, 10, 14, and 16].
  • Polymo ⁇ hisms in the introns can serve as markers for disease following linkage analysis.
  • genetic analyses described herein can be used to locate mutations in the introns associated with and/or causing disease.
  • Another preferred embodiment is a cathepsin K intronic enhancer.
  • Intron 3 does not follow consensus splice junction GT/AG rule. This intron/exon boundaries was verified by sequencing of the PI clone and the genomic DNA. GC/AG splice junctions though not common, have been described(Mount, Stephen, M., Department of Molecular Biophysics and Biochemistry, Yale University, Sterling Hall of Medicine, New Haven, CT, USA, IRL Press Limited, London, pp. 459-472 ( 1981 )).
  • cathepsin K exons particularly those exons having the sequences set forth in Figure 3 (B, D, F, H, J, L, N, and P) [SEQUENCE ID NO: 3, 5, 7, 9, 1 1, 13, 15 and 17 respectively].
  • Polymo ⁇ hisms in the exons can serve as markers for disease following linkage analysis.
  • genetic analyses described herein can be used to locate mutations in the exons associated with and/or causing disease.
  • Polynucleotide fragments of the invention can be used to create ribozymes that inhibit the expression of the cathepsin K gene.
  • General methods for the construction of ribozyme constructs are known in the art (Stram Y, and Molad T, Virus Genes, 1995, 9 (2), 155-159). Skilled artisans can readily adapt these methods using the novel fragments of the invention to create novel ribozyme constructs.
  • Preferred ribozyme constructs comprise sequences which are complementary to the transcribed control elements of the cathepsin K gene, particularly polynucleotides that are complementary to the 5' untranslated region, splice junctions, and 3'untranslated region, especially the polyadenylation region.
  • the fragments of the invention are useful as diagnostic probes for disease, particularly bone disease, such as osteoporosis, and including, for example, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hype ⁇ arathyroidism, bone degradation, metastatic tumors, rhemuatoid arthritis, osteoarthritis, periodontal disease and degradation of bone implants and bone protheses, particularly dental implants.
  • markers for disease can be located in regions of the cathepsin gene, particularly untranslated regions, which are useful with the diagnostic methods of the invention.
  • the present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recombinant techniques.
  • Host cells can be genetically engineered to inco ⁇ orate polynucleotides and express polypeptides of the present invention.
  • polynucleotides may be introduced into host cells using well known techniques of infection, transduction, transfection, transvection and transformation.
  • the polynucleotides may be introduced alone or with other polynucleotides.
  • Such other polynucleotides may be introduced independently, co-introduced or introduced joined to the polynucleotides of the invention.
  • polynucleotides of the invention may be transfected into host cells with another, separate, polynucleotide encoding a selectable marker, using standard techniques for co-transfection and selection in, for instance, mammalian cells.
  • the polynucleotides generally will be stably inco ⁇ orated into the host cell genome.
  • the polynucleotides may be joined to a vector containing a selectable marker for propagation in a host.
  • the vector construct may be introduced into host cells by the aforementioned techniques.
  • a plasmid vector is introduced as DNA in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid.
  • Electroporation also may be used to introduce polynucleotides into a host. If the vector is a virus, it may be packaged in vitro or introduced into a packaging cell and the packaged virus may be transduced into cells.
  • the vector may be, for example, a plasmid vector, a single or double-stranded phage vector, a single or double-stranded RNA or DNA viral vector.
  • Such vectors may be introduced into cells as polynucleotides, preferably DNA, by well known techniques for introducing DNA and RNA into cells.
  • the vectors in the case of phage and viral vectors also may be and preferably are introduced into cells as packaged or encapsidated virus by well known techniques for infection and transduction.
  • Viral vectors may be replication competent or replication defective. In the latter case viral propagation generally will occur only in complementing host cells.
  • vectors are those for expression of polynucleotides and polypeptides of the present invention.
  • such vectors comprise cis-acting control regions effective for expression in a host operatively linked to the polynucleotide to be expressed.
  • Appropriate trans-acting factors either are supplied by the host, supplied by a complementing vector or supplied by the vector itself upon introduction into the host.
  • the vectors provide for specific expression.
  • Such specific expression may be inducible expression or expression only in certain types of cells or both inducible and cell-specific.
  • Particularly preferred among inducible vectors are vectors that can be induced for expression by environmental factors that are easy to manipulate, such as temperature and nutrient additives.
  • a variety of vectors suitable to this aspect of the invention, including constitutive and inducible expression vectors for use in prokaryotic and eukaryotic hosts, are well known and employed routinely by those of skill in the art.
  • the engineered host cells can be cultured in conventional nutrient media, which may be modified as appropriate for, inter alia, activating promoters, selecting transformants or amplifying genes.
  • Culture conditions such as temperature, pH and the like, previously used with the host cell selected for expression generally will be suitable for expression of polypeptides of the present invention as will be apparent to those of skill in the art.
  • vectors can be used to express a polypeptide of the invention.
  • Such vectors include chromosomal, episomal and virus-derived vectors e.g., vectors derived from bacterial plasmids, from bacteriophage, from yeast episomes, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, and vectors derived from combinations thereof, such as those derived from plasmid and bacteriophage genetic elements, such as cosmids and phagemids, all may be used for expression in accordance with this aspect of the present invention.
  • any vector suitable to maintain, propagate or express polynucleotides to express a polypeptide in a host may be used for expression in this regard.
  • the appropriate DNA sequence may be inserted into the vector by any of a variety of well-known and routine techniques.
  • a DNA sequence for expression is joined to an expression vector by cleaving the DNA sequence and the expression vector with one or more restriction endonucleases and then joining the restriction fragments together using T4 DNA ligase.
  • Procedures for restriction and ligation that can be used to this end are well known and routine to those of skill. Suitable procedures in this regard, and for constructing expression vectors using alternative techniques, which also are well known and routine to those skill, are set forth in great detail in Sambrook et al. cited elsewhere herein.
  • the DNA sequence in the expression vector is operatively linked to appropriate expression control sequence(s), including, for instance, a promoter to direct mRNA transcription.
  • appropriate expression control sequence(s) including, for instance, a promoter to direct mRNA transcription.
  • promoters include the phage lambda PL promoter, the E. coli lac, t ⁇ and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name just a few of the well-known promoters. It will be understood that numerous promoters not mentioned are suitable for use in this aspect of the invention are well known and readily may be employed by those of skill in the manner illustrated by the discussion and the examples herein.
  • expression constructs will contain sites for transcription initiation and termination, and, in the transcribed region, a ribosome binding site for translation.
  • the coding portion of the mature transcripts expressed by the constructs will include a translation initiating AUG at the beginning and a termination codon appropriately positioned at the end of the polypeptide to be translated.
  • constructs may contain control regions that regulate as well as engender expression. Generally, in accordance with many commonly practiced procedures, such regions will operate by controlling transcription, such as repressor binding sites and enhancers, among others.
  • Vectors for propagation and expression generally will include selectable markers. Such markers also may be suitable for amplification or the vectors may contain additional markers for this pu ⁇ ose.
  • the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells. Preferred markers include dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, tetracycline, kanamycin, and ampicillin resistance genes for culturing E. coli and other bacteria.
  • the vector containing the appropriate DNA sequence as described elsewhere herein, as well as an appropriate promoter, and other appropriate control sequences, may be introduced into an appropriate host using a variety of well known techniques suitable to expression therein of a desired polypeptide.
  • appropriate hosts include bacterial cells, such as E. coli, streptomyces and
  • Salmonella typhimurium cells Salmonella typhimurium cells; fungal cells, such as yeast cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS and Bowes melanoma cells; and plant cells.
  • Hosts for a great variety of expression constructs are well known, and those of skill will be enabled by the present disclosure readily to select a host for expressing a polypeptides in accordance with this aspect of the present invention.
  • the present invention also includes recombinant constructs, such as expression constructs, comprising one or more of the sequences described above.
  • the constructs comprise a vector, such as a plasmid or viral vector, into which such a sequence of the invention has been inserted.
  • the sequence may be inserted in a forward or reverse orientation.
  • the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence.
  • suitable vectors and promoters are known to those of skill in the art, and there are many commercially available vectors suitable for use in the present invention.
  • vectors which are commercially available, are provided by way of example.
  • vectors preferred for use in bacteria are pQE70, pQE60 and pQE-9, available from Qiagen; pBS vectors, Phagescript vectors, Bluescript vectors, pNH8A, pNH16a, pNH18A, pNH46A, available from Stratagene; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia.
  • eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTl and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia. These vectors are listed solely by way of illustration of the many commercially available and well known vectors that are available to those of skill in the art for use in accordance with this aspect of the present invention. It will be appreciated that any other plasmid or vector suitable for, for example, introduction, maintenance, propagation or expression of a polynucleotide or polypeptide of the invention in a host may be used in this aspect of the invention.
  • Promoter regions can be selected from any desired gene using vectors that contain a reporter transcription unit lacking a promoter region, such as a chloramphenicol acetyl transferase ("CAT") transcription unit, downstream of restriction site or sites for introducing a candidate promoter fragment; i.e., a fragment that may contain a promoter.
  • CAT chloramphenicol acetyl transferase
  • introduction into the vector of a promoter-containing fragment at the restriction site upstream of the cat gene engenders production of CAT activity, which can be detected by standard CAT assays.
  • Vectors suitable to this end are well known and readily available. Two such vectors are pKK232-8 and pCM7.
  • promoters for expression of polynucleotides of the present invention include not only well known and readily available promoters, but also promoters that readily may be obtained by the foregoing technique, using a reporter gene.
  • a preferred embodiment of the invention are expression vectors comprising cathepsin K promoter sequences that function as a promoter. Such vector constructs may be used for targeted gene expression in cells which utilize the cathepsin K promoter, for example, osteoclasts and macrophages. Any gene of interest can be expressibly linked to the cathepsin K promoter and expressed in such cells which utilize the cathepsin K promoter.
  • genes which immortalize primary eukaryotic cells may be expressibly linked cathepsin K promoter to immortalize cells, such as, for example, bone cells, including osteoclasts, and macrophages.
  • Certain preferred vectors comprise cathepsin K promoter expressibly linked to a toxin gene, such as for example, ricin, and are useful in methods for the targeted killing of cell populations that utilize the cathepsin K promoter for gene expression.
  • Certain other preferred vectors comprise cathepsin K promoter expressibly linked to a anti-cathepsin K ribozyme or antisense polynucleotide, which are useful in methods for such targeted killing.
  • bacterial promoters suitable for expression of polynucleotides and polypeptides in accordance with the present invention are the E. coli lad and lacZ promoters, the T3 and T7 promoters, the gpt promoter, the lambda PR, PL promoters and the t ⁇ promoter.
  • known eukaryotic promoters suitable in this regard are the CMV immediate early promoter, the HSV thymidine kinase promoter, the early and late SV40 promoters, the promoters of retroviral LTRs, such as those of the Rous sarcoma virus ("RSV"), and metallothionein promoters, such as the mouse metallothionein-I promoter.
  • RSV Rous sarcoma virus
  • the present invention also relates to host cells containing the above-described constructs discussed above.
  • the host cell can be a higher eukaryotic cell, such as a mammalian cell or insect cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al. BASIC METHODS IN MOLECULAR BIOLOGY, (1986).
  • Constructs in host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence.
  • the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
  • Mature proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook et al., MOLECULAR CLONING: A LABORATORY MANUAL, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989).
  • recombinant expression vectors for yeast will include origins of replication, a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence, and a selectable marker to permit isolation of vector containing cells after exposure to the vector.
  • suitable promoters are those derived from the genes that encode glycolytic enzymes such as 3-phosphoglycerate kinase ("PGK”), a-factor, acid phosphatase, and heat shock proteins, among others.
  • PGK 3-phosphoglycerate kinase
  • Selectable markers include the ampicillin resistance gene of E. coli and the t ⁇ l gene of S. cerevisiae.
  • Enhancers are cis-ac ⁇ ng elements of DNA, usually about from 10 to 300 bp that act to increase transcriptional activity of a promoter in a given host cell-type.
  • enhancers include the SV40 enhancer, which is located on the late side of the replication origin at bp 100 to 270, the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
  • Polynucleotides of the invention encoding the heterologous structural sequence of a polypeptide of the invention generally will be inserted into the vector using standard techniques so that it is operably linked to the promoter for expression.
  • the polynucleotide will be positioned so that the transcription start site is located appropriately 5' to a ribosome binding site.
  • the ribosome binding site will be 5' to the AUG that initiates translation of the polypeptide to be expressed.
  • a translation stop codon at the end of the polypeptide and there will be a polyadenylation signal and a transcription termination signal appropriately disposed at the 3' end of the transcribed region.
  • appropriate secretion signals may be inco ⁇ orated into the expressed polypeptide.
  • the signals may be endogenous to the polypeptide or they may be heterologous signals.
  • the polypeptide may be expressed in a modified form, such as a fusion protein, and may include not only secretion signals but also additional heterologous functional regions.
  • a region of additional amino acids, particularly charged amino acids may be added to the N-terminus of the polypeptide to improve stability and persistence in the host cell, during purification or during subsequent handling and storage.
  • a region may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide.
  • the addition of peptide moieties to polypeptides to engender secretion or excretion, to improve stability and to facilitate purification, among others, are familiar and routine techniques in the art.
  • Suitable prokaryotic hosts for propagation, maintenance or expression of polynucleotides and polypeptides in accordance with the invention include
  • useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017).
  • plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017).
  • pBR322 ATCC 37017
  • Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEMl (Promega Biotec, Madison, WI, USA).
  • Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents, such methods are well know to those skilled in the art.
  • mammalian cell culture systems can be employed for expression, as well.
  • mammalian expression systems include the COS-7 lines of monkey kidney fibroblast, described in Gluzman et al., Cell 23: 175 (1981).
  • Other cell lines capable of expressing a compatible vector include for example, the C 127, 3T3, CHO, HeLa, human kidney 293 and BHK cell lines.
  • Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation sites, splice donor and acceptor sites, transcriptional termination sequences, and 5' flanking non-transcribed sequences that are necessary for expression.
  • DNA sequences derived from the SV40 splice sites, and the SV40 polyadenylation sites are used for required non-transcribed genetic elements of these types.
  • the cathepsin K polypeptide can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography ("HPLC") is employed for purification. Well known techniques for refolding protein may be employed to regenerate active conformation when the polypeptide is denatured during isolation and or purification.
  • HPLC high performance liquid chromatography
  • Polypeptides of the present invention include naturally purified products, products of chemical synthetic procedures, and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. In addition, polypeptides of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes. Further illustrative aspects and preferred embodiments of the invention
  • Cathepsin K polynucleotides and polypeptides may be used in accordance with the present invention for a variety of applications, particularly those that make use of the chemical and biological properties cathepsin K.
  • applications in the detection and treatment of disease particularly bone disease, such as osteoporosis, and including, for example, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hype ⁇ arathyroidism, bone degradation, metastatic tumors, rhemuatoid arthritis, osteoarthritis, periodontal disease and degradation of bone implants and bone protheses, particularly dental implants.
  • Additional applications relate to diagnosis and to treatment of disorders of cells, tissues and organisms. These aspects of the invention are illustrated further by the following discussion.
  • This invention is also related to the use of the cathepsin K exons, introns, promoters and polynucleotides to detect complementary polynucleotides such as, for example, as a diagnostic reagent. Detection of a mutated form of cathepsin K associated with a dysfunction will provide a diagnostic tool that can add or define a diagnosis of a disease or susceptibility to a disease which results from under-expression, over-expression or altered expression of cathepsin K, such as, for example, osteoporosis, periodontal disease, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hype ⁇ arathyroidism, and bone degradation, metastatic tumors, and degradation of bone implants and bone protheses, particularly dental implants.
  • Nucleic acids for diagnosis may be obtained from a patient's cells, such as from blood, urine, saliva, tissue biopsy and autopsy material.
  • the genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR prior to analysis (Saiki et al., Nature, 324: 163-166 (1986)).
  • Ligation-mediated amplification may also be used for amplification (Vollach, V., et al., Nucl Acids Res. 22: 2507 (1994).
  • RNA or cDNA may also be used in the same ways.
  • PCR primers complementary to the nucleic acid encoding cathepsin K can be used to identify and analyze cathepsin K expression and mutations. For example, deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to radiolabeled cathepsin K RNA or alternatively, radiolabeled cathepsin K antisense DNA sequences. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melting temperatures.
  • Sequence differences between a reference gene and genes having mutations also may be revealed by direct DNA sequencing.
  • cloned DNA segments may be employed as probes to detect specific DNA segments.
  • the sensitivity of such methods can be greatly enhanced by appropriate use of PCR or another amplification method.
  • a sequencing primer is used with double-stranded PCR product or a single-stranded template molecule generated by a modified PCR.
  • the sequence determination is performed by conventional procedures with radiolabeled nudeotide or by automatic sequencing procedures with fluorescent-tags.
  • DNA sequence differences may be achieved by detection of alteration in electrophoretic mobility of DNA fragments in gels, with or without denaturing agents. Small sequence deletions and insertions can be visualized by high resolution gel electrophoresis. DNA fragments of different sequences may be distinguished on denaturing formamide gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures (see, e.g., Myers et al., Science, 230: 1242 ( 1985)). Sequence changes at specific locations also may be revealed by nuclease protection assays, such as RNase and S 1 protection or the chemical cleavage method (e.g., Cotton et al., Proc. Natl. Acad. Sci., USA, 85: 4397-4401 (1985)).
  • the detection of a specific DNA sequence may be achieved by methods such as hybridization, RNase protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes, (e.g., restriction fragment length polymo ⁇ hisms ("RFLP"), SSCP and Southern blotting of genomic DNA.
  • restriction enzymes e.g., restriction fragment length polymo ⁇ hisms ("RFLP"), SSCP and Southern blotting of genomic DNA.
  • mutations also can be detected by in situ analysis.
  • the sequences of the present invention are also valuable for chromosome identification.
  • the sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome.
  • Few chromosome marking reagents based on actual sequence data (repeat polymo ⁇ hisms) are presently available for marking chromosomal location.
  • the mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease.
  • the gDNA herein disclosed is used to clone genomic DNA of a cathepsin K gene.
  • genomic DNA then is used for in situ chromosome mapping using well known techniques for this pu ⁇ ose.
  • some trial and error may be necessary to identify a genomic probe that gives a good in situ hybridization signal.
  • sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the gDNA. Computer analysis of the 3' untranslated region of the gene is used to rapidly select primers that do not span more than one exon in the genomic DNA complicate the amplification process. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the primer will yield an amplified fragment.
  • PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA to a particular chromosome.
  • sublocalization can be achieved with panels of fragments from specific chromosomes (e.g., radiation hybrid panels) or pools of large genomic clones in an analogous manner.
  • Other mapping strategies that can similarly be used to map to its chromosome include in situ hybridization, prescreening with labeled flow-sorted chromosomes and preselection by hybridization to construct chromosome specific -cDNA libraries.
  • Fluorescence in situ hybridization of a cDNA clone to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step.
  • This technique can be used with gDNA as short as 50 to as long as 600.
  • Verma et al. HUMAN CHROMOSOMES: A MANUAL OF BASIC TECHNIQUES, Pergamon Press, New York ( 1988).
  • a gDNA precisely localized to a chromosomal region associated with the disease could be one of between 50 and 500 potential causative genes. (This assumes 1 megabase mapping resolution and one gene per 20 kb).
  • the present invention also relates to a diagnostic assays such as quantitative and diagnostic assays for detecting levels of cathepsin K protein in cells, tissues and bodily fluids, including determination of normal and abnormal levels of polypeptide.
  • Bodily fluids useful in the diagnostic methods of the invention include, for example, synovial fluid, cerebrospinal fluid, urine, serum, gingival fluid and lymph.
  • a diagnostic assay in accordance with the invention for detecting over-expression of cathepsin K protein compared to normal control tissue samples may be used to detect the presence of disease, for example, osteoporosis, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hype ⁇ arathyroidism, bone degradation, metastatic tumors, rhemuatoid arthritis, osteoarthritis, periodontal disease and degradation of bone implants and bone protheses, particularly dental implants.
  • Assay techniques that can be used to determine levels of a protein, such as an immunoassay for cathepsin K protein of the present invention, in a sample derived from a host are well-known to those of skill in the art.
  • An ELISA assay initially comprises preparing an antibody specific to cathepsin K, preferably a monoclonal antibody.
  • a reporter antibody generally is prepared which binds to the monoclonal antibody.
  • the reporter antibody is attached a detectable reagent such as radioactive, fluorescent or enzymatic reagent, in this example horseradish peroxidase enzyme.
  • a sample is removed from a host and incubated on a solid support, e.g. a polystyrene dish, that binds the proteins in the sample. Any free protein binding sites on the dish are then covered by incubating with a non-specific protein such as bovine serum albumin.
  • a non-specific protein such as bovine serum albumin.
  • the monoclonal antibody is incubated in the dish during which time the monoclonal antibodies attach to any cathepsin K proteins attached to the polystyrene dish. Unbound monoclonal antibody is washed out with buffer.
  • the reporter antibody linked to horseradish peroxidase is placed in the dish resulting in binding of the reporter antibody to any monoclonal antibody bound to cathepsin K.
  • Unattached reporter antibody is then washed out.
  • Reagents for peroxidase activity including a colorimetric substrate are then added to the dish.
  • the amount of color developed in a given time period indicates the amount of cathepsin K protein present in the sample.
  • Quantitative results typically are obtained by reference to a standard curve.
  • a competition assay may be employed wherein antibodies specific to cathepsin K attached to a solid support and labeled cathepsin K and a sample derived from the host are passed over the solid support and the amount of label detected attached to the solid support can be correlated to a quantity of cathepsin K in the sample.
  • the polypeptides, their fragments or other derivatives, or analogs thereof, or cells expressing them can be used as an immunogen to produce antibodies thereto.
  • These antibodies can be, for example, polyclonal or monoclonal antibodies.
  • the present invention also includes chimeric, single chain, and humanized antibodies, as well as Fab fragments, or the product of a Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments.
  • Antibodies generated against the polypeptides corresponding to a sequence of the present invention can be obtained by direct injection of the polypeptides into an animal or by administering the polypeptides to an animal, preferably a nonhuman. The antibody so obtained will then bind the polypeptides itself. In this manner, even a sequence encoding only a fragment of the polypeptides can be used to generate antibodies binding the whole native polypeptides. Such antibodies can then be used to isolate the polypeptide from tissue expressing that polypeptide.
  • any technique which provides antibodies produced by continuous clonal cell line cultures can be used. Examples include the hybridoma technique (Kohler, G. and Milstein, C, Nature 256: 495-497 (1975), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., Immunology Today 4: 72 (1983) and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al., pg. 77-96 in MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc. (1985).
  • such antibodies can be used to detect and treat diseases caused by or associated with mutant cathepsin K or abnormal cathepsin K levels, such as, osteoporosis, periodontal disease, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hype ⁇ arathyroidism, bone degradation, metastatic tumors, and degradation of bone implants and bone protheses, particularly dental implants.
  • Immunization using polynucleotides of the inventions can be carried out using known methods to produce a cathepsin K-specific immune response.
  • This invention provides methods to determine drug responsiveness of individuals having or suspected of having a cathepsin K gene mutation or cathepsin K gene expression abnormality, and also provides reagents to carry out such methods.
  • Individuals may be grouped by their responsiveness to a given compound, particularly drugs, used to treat diseases caused by or associated with a mutation of cathepsin K gene or cathepsin K gene expression.
  • Such individuals may be further grouped by detecting different gene mutations or gene expression level variants. In this manner specific gene mutations and gene expression variants can be readily associated with a certain degree of responsiveness to a compound by an individual).
  • Methods and reagents provided herein can be used to group compound responsiveness by detecting cathepsin K gene mutations and cathepsin K gene expression variants.
  • Other methods for grouping individuals by compound responsivess are known to skilled artisans and can be adapted to use the polypetides and polynucleotides of the invention.
  • the invention also provides algorithms useful in conjunction with a device or embodied in a composition matter which are useful for the diagnosis of diseases caused by or associated with cathepsin K or mutants or variants thereof. Preferred algorithms are provided for disease stratification and staging.
  • This invention also provides a method for identification of molecules, such as receptor molecules, that bind cathepsin K or fragments of cathepsin K of the invention.
  • Genes encoding proteins that bind cathepsin K, such as receptor proteins can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting. Such methods are described in many laboratory manuals such as, for instance, Coligan et al., Current Protocols in Immunology 1(2): Chapter 5 (1991).
  • expression cloning may be employed for this pu ⁇ ose.
  • polyadenylated RNA is prepared from a cell responsive to cathepsin K
  • a cDNA library is created from this RNA, the library is divided into pools and the pools are transfected individually into cells that are not responsive to cathepsin K. The transfected cells then are exposed to labeled cathepsin K.
  • Cathepsin K can be labeled by a variety of well-known techniques including standard methods of radio-iodination or inclusion of a recognition site for a site-specific protein kinase.) Following exposure, the cells are fixed and binding of cathepsin K, or a molecule which binds to cathepsin K, is determined. These procedures conveniently are carried out on glass slides.
  • Pools are identified of cDNA that produced cathepsin K-binding cells. Sub-pools are prepared from these positives, transfected into host cells and screened as described above. Using an iterative sub-pooling and re-screening process, one or more single clones that encode the putative binding molecule or substrate, such as cell matrix, bone matrix or a receptor molecule, and the any of the same can be isolated. Alternatively a labeled ligand can be photoaffinity linked to a cell extract, such as a membrane or a membrane extract, prepared from cells that express a molecule that it binds, such as a receptor molecule. Cross-linked material is resolved by polyacrylamide gel electrophoresis ("PAGE") and exposed to X-ray film.
  • PAGE polyacrylamide gel electrophoresis
  • the labeled complex containing the ligand-receptor can be excised, resolved into peptide fragments, and subjected to protein microsequencing.
  • the amino acid sequence obtained from microsequencing can be used to design unique or degenerate oligonudeotide probes to screen cDNA libraries to identify genes encoding the putative receptor molecule.
  • Polypeptides of the invention also can be used to assess cathepsin K binding capacity of cathepsin K binding molecules, such as receptor molecules, in cells or in cell-free preparations.
  • Agonists and antagonists - assays and molecules The invention also provides a method of screening compounds to identify those which enhance or block the action of cathepsin K in or on cells, such as its interaction with cathepsin K-binding molecules such as receptor and enzymatic substrate molecules.
  • An agonist is a compound which increases the natural biological functions of cathepsin K, while antagonists decrease or eliminate such functions.
  • a cellular compartment such as a membrane, vacuole, inclusion or a preparation of any thereof, such as a membrane-preparation, may be prepared from a cell that expresses a molecule that binds cathepsin K, such as a molecule of a signaling or regulatory pathway modulated by cathepsin K.
  • the preparation is incubated with labeled cathepsin K in the absence or the presence of a candidate molecule which may be a cathepsin K agonist or antagonist.
  • the ability of the candidate molecule to bind the binding molecule, such as a substrate, is reflected in decreased binding of the labeled ligand.
  • Molecules which bind gratuitously, i.e., without inducing the effects of cathepsin K on binding the cathepsin K binding molecule, are most likely to be good antagonists. Molecules that bind well and elicit effects that are the same as or closely related to cathepsin K are agonists.
  • Cathepsin K-like effects of potential agonists and antagonists may by measured, for instance, by determining activity of a second messenger system following interaction of the candidate molecule with a cell or appropriate cell preparation, and comparing the effect with that of cathepsin K or molecules that elicit the same effects as cathepsin K.
  • Second messenger systems that may be useful in this regard include but are not limited to AMP guanylate cyclase, ion channel, phosphoinositide hydrolysis second messenger systems, or compounds which signal the binding of a potential agonists and antagonists to cathepsin K or its substrate.
  • an assay for cathepsin K antagonists is a competitive assay that combines cathepsin K and a potential antagonist with enzymatic substrate or substrate analogs under appropriate conditions for a competitive inhibition assay.
  • Cathepsin K can be labeled, such as by radioactivity, such that the number of cathepsin K molecules bound to a receptor molecule can be determined accurately to assess the effectiveness of the potential antagonist.
  • Potential antagonists include small organic molecules, peptides, polypeptides and antibodies that bind to a polypeptide of the invention and thereby inhibit or extinguish its activity. Potential antagonists also may be small organic molecules, a peptide, a polypeptide such as a closely related protein or antibody that binds the same sites on a binding molecule, such as a receptor molecule, without inducing cathepsin K-induced activities, thereby preventing the action of cathepsin K by excluding cathepsin K from binding.
  • Potential antagonists include a small molecule which binds to and occupies the binding site of the polypeptide thereby preventing binding to cellular binding molecules, such as receptor molecules, such that normal biological activity is prevented.
  • small molecules include but are not limited to small organic molecules, peptides or peptide-like molecules.
  • Antisense molecules can be used to control gene expression through antisense DNA or RNA or through triple-helix formation. Antisense techniques are discussed, for example, in - Okano, J. Neurochem. 56: 560 (1991); OLIGODEOXYNUCLEOTIDES AS ANTISENSE INHIBITORS OF GENE EXPRESSION, CRC Press, Boca Raton, FL (1988). Triple helix formation is discussed in, for instance Lee et al., Nucleic Acids Research 6: 3073 (1979); Cooney et al., Science 241: 456 (1988); and Dervan et al., Science 251 : 1360 (1991).
  • the methods are based on binding of a polynucleotide to a complementary DNA or RNA.
  • the 5' coding portion of a polynucleotide that encodes the mature polypeptide of the present invention may be used to design an antisense RNA oligonudeotide of from about 10 to 40 base pairs in length.
  • a DNA oligonudeotide is designed to be complementary to a region of the gene involved in transcription thereby preventing transcription and the production of cathepsin K.
  • the antisense RNA oligonudeotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into cathepsin K polypeptide.
  • the oligonudeotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of cathepsin K.
  • the antagonists may be employed in a composition with a pharmaceutically acceptable carrier, e.g., as hereinafter described.
  • the antagonists may be employed for instance to treat diseases caused by or associated with mutant cathepsin K or abnormal cathepsin K levels, such as, osteoporosis, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hype ⁇ arathyroidism, bone degradation, metastatic tumors, rhemuatoid arthritis, osteoarthritis, periodontal disease and degradation of bone implants and bone protheses, particularly dental implants.
  • diseases caused by or associated with mutant cathepsin K or abnormal cathepsin K levels such as, osteoporosis, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hype ⁇ arathyroidism, bone degradation, metastatic tumors, rhemuatoid arthritis, osteoarthritis, periodontal disease and degradation of bone implants and bone protheses, particularly dental implants.
  • compositions comprising the polynucleotides or the polypeptides discussed above or the agonists or antagonists.
  • the polypeptides of the present invention may be employed in combination with a non-sterile or sterile carrier or carriers for use with cells, tissues or organisms, such as a pharmaceutical carrier suitable for administration to a subject.
  • a pharmaceutical carrier suitable for administration to a subject such as a pharmaceutical carrier suitable for administration to a subject.
  • Such compositions comprise, for instance, a media additive or a therapeutically effective amount of a polypeptide of the invention and a pharmaceutically acceptable carrier or excipient.
  • Such carriers may include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol and combinations thereof. The formulation should suit the mode of administration.
  • the invention further relates to pharmaceutical packs and kits comprising one or more containers filled with one or more of the ingredients of the aforementioned compositions of the invention.
  • 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, reflecting approval by the agency of the manufacture, use or sale of the product for human administration.
  • Polypeptides and other compounds of the present invention may be employed alone or in conjunction with other compounds, such as therapeutic compounds.
  • compositions may be administered in any effective, convenient manner including, for instance, administration by topical, oral, anal, vaginal, intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal, intraarticular, or intradermal routes among others.
  • compositions generally are administered in an amount effective for treatment or prophylaxis of a specific indication or indications.
  • the compositions are administered in an amount of at least about 10 mg/kg body weight.
  • dose is from about 10 mg/kg to about 1 mg/kg body weight, daily. It will be appreciated that optimum dosage will be determined by standard methods for each treatment modality and indication, taking into account the indication, its severity, route of administration, complicating conditions and the like.
  • cathepsin K polynucleotides, polypeptides, agonists and antagonists that are polypeptides may be employed in accordance with the present invention by expression of such polypeptides in vivo, in treatment modalities often referred to as "gene therapy.”
  • cells from a patient may be engineered with a polynucleotide, such as a DNA or RNA, encoding a polypeptide ex vivo, and the engineered cells then can be provided to a patient to be treated with the polypeptide.
  • a polynucleotide such as a DNA or RNA
  • cells may be engineered ex vivo by the use of a retroviral plasmid vector containing RNA encoding a polypeptide of the present invention.
  • Cells from a patient may also be engineered with a polynucleotide, such as a ribozyme that has been constructed, using well known methods, to inhibit the gene expression of Cathepsin K.
  • a polynucleotide such as a ribozyme that has been constructed
  • Other constructs may also be engineered into a patient's cells to contains antisense stretches of cathepsin K sequence, using well known methods. Such antisense constructs will inhibit Cathepsin K expression in the patient.
  • cells may be engineered in vivo for expression of a polypeptide in vivo by procedures known in the art.
  • a polynucleotide of the invention may be engineered for expression in a replication defective retroviral vector, as discussed above.
  • the retroviral expression construct then may be isolated and introduced into a packaging cell that is transduced with a retroviral plasmid vector containing RNA encoding a polypeptide of the present invention such that the packaging cell now produces infectious viral particles containing the gene of interest.
  • These producer cells may be administered to a patient for engineering cells in vivo and expression of the polypeptide in vivo.
  • Retroviruses from which the retroviral plasmid vectors herein above mentioned may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, adenovirus, Myeloproliferative Sarcoma Virus, and mammary tumor virus.
  • the retroviral plasmid vector is derived from Moloney Murine Leukemia Virus.
  • Such vectors well include one or more promoters for expressing the polypeptide.
  • Suitable promoters which may be employed include, but are not limited to, cathepsin K promoter, a retroviral LTR, an SV40 promoter, and the human cytomegalovirus (CMV) promoter described in Miller et al., Biotechniques 7: 980-990 (1989), or any other promoter (e.g., cellular promoters such as eukaryotic cellular promoters including, but not limited to, the histone, RNA polymerase III, and alpha-actin promoters).
  • CMV human cytomegalovirus
  • viral promoters which may be employed include, but are not limited to, adenovirus promoters, thymidine kinase (TK) promoters, and B19 parvovirus promoters. The selection of a suitable promoter will be apparent to those skilled in the art from the teachings contained herein.
  • Suitable promoters which may be employed include, but are not limited to, adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the rous sarcoma virus (RSV) promoter; inducible promoters, such as the MMT promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAI promoter; human globin promoters; viral thymidine kinase promoters, such as the He ⁇ es Simplex thymidine kinase promoter; retroviral LTRs (including the modified retroviral LTRs herein above described); the alpha-actin promoter; and human growth hormone promoters.
  • the promoter also may be the native promoter which controls the gene encoding the poly
  • the retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines.
  • packaging cells which may be transfected include, but are not limited to, the PE501, PA317, Y-2, Y-AM, PA12, T19-14X, VT-19-17-H2, YCRE, YCRIP, GP+E-86, GP+envAml2, and DAN cell lines as described in Miller, A., Human Gene Therapy 1: 5-14 (1990).
  • the vector may be transduced into the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaPO4 precipitation.
  • the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host.
  • the producer cell line will generate infectious retroviral vector particles, which include the nucleic acid sequence(s) encoding the polypeptides.
  • retroviral vector particles then may be employed to transduce eukaryotic cells, either in vitro or in vivo. The transduced eukaryotic cells will express the nucleic acid sequence(s) encoding the polypeptide.
  • Eukaryotic cells which may be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic stem cells, hepatocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells, and bronchial epithelial cells.
  • N used herein in a nudeotide sequence refers to an unknown nudeotide or nucleotides. All examples were or may be carried out using standard techniques, which are well known and routine to those of skill in the art, except where otherwise described in detail. Routine molecular biology techniques of the following examples can be carried out as described in standard laboratory manuals, such as Sambrook et al., MOLECULAR CLONING: A LABORATORY MANUAL, 2nd Ed.; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989), herein referred to as "Sambrook.” All parts or amounts set out in the following examples are by weight, unless otherwise specified.
  • ligations were accomplished using standard buffers, incubation temperatures and times, approximately equimolar amounts of the DNA fragments to be ligated and approximately 10 units of T4 DNA ligase ("ligase”) per 0.5 ⁇ g of DNA.
  • ligase T4 DNA ligase
  • cDNA as disclosed in U.S. Patent Number 5,501,969 was used to isolate the gDNA clone from a gDNA library (Clontech) according to the following method. Primers to adjacent exons (6 of the 7 exons) were prepared. The sequence of these primers is underlined in Figure 2. PCR was performed using standard methods well known in the art. Amplified fragments were cloned into a TA vector (Clontech) and the clones were sequenced by an automated sequencer (Applied BioSystems Model 373) by established methods well known in the art using forward and reverse sequencing primers. The sequence of all internal introns were obtained. 5' and 3' terminal intron sequences were obtained as follows.
  • 5' end primers were designed to obtain sequence for the first intron (see underlined primer in Figure 2), using these primers 2 PI clones were obtained (Genome Systems Inc.). Both clones were full length. PCR was used to confirm the sequence of internal intron-exon boundary junctions (see Example 2). Primers derived from sequence at the 5' end of the PI clones was used to "walk” and sequence along the clone, in a stepwise fashion, using new primers at each sequence step, by routine methods known in the art. Purification of PI clones was carried out as set forth in Example 1(d). "Walking” and sequencing was performed in both directions to confirm cathepsin K gDNA sequence. PCR was again performed using proofreading Taq polymerase (PCR Ultima, Perkin Elmer).
  • a transcription start site was obtained using a 5' RACE kit (Gibco BRL) and the protocol supplied therewith. This site was also confirmed using an RNASe protection assay kit (Hybspeed, RPA Ambion).
  • Example 1 (a)-(d) provide further specifics concerning cloning and sequencing of cathepsin K
  • Intron 1 was identified by utilization of 5' RACE (Gibco BRL) technique to determine 5' UTR sequence from which primer could be designed to PCR from exon 1 to exon 2. (intron 1 starts prior to ATG so PCR may not be readily employed based on cDNA sequence available.) Intron 1 was amplified by PCR on human genomic DNA (Clontech) and cloned into PCRII vector and sequenced as described in Example 1.
  • 5' RACE Gibco BRL
  • Intron 2 was identified by PCR on human genomic DNA from primers designed in exon 2 to exon 3. PCR product was cloned and sequenced using standard methods.
  • Intron 3 Intron 3 was identified by PCR on human genomic DNA from primers designed in exon 3 to exon 4. PCR product was cloned and sequenced using standard methods. Intron 4
  • Intron 4 was identified by PCR on human genomic DNA from primers designed in exon 4 to exon 5. PCR product was cloned and sequenced using standard methods.
  • Introns 5 and 6 were identified by PCR on human genomic DNA from primers designed in exon 5 to exon 7. PCR product was cloned and sequenced using standard methods confirming presence of both introns.
  • Intron 7 was identified by PCR on human genomic DNA from primers designed in exon 7 to exon 8. PCR product was cloned and sequenced using standard methods.
  • PI clone A 5' and 3' untranslated regions were isolated from a single PI clone (Genome Systems Inc.). This PI clone has been identified herein as "PI clone A.” Sequence was obtained by direct sequence walking up and down the PI clone with gene specific primers derived from confirmed cDNA sequence using standard methods. These regions were then cloned via PCR and confirmed by sequence analysis using standard methods. The 5' UTR was additionally amplified by PCR using proofreading Taq Polymerase Ultima in accordance with manufacturer instructions and cloned to eliminate sequence ambiguities. 5' and 3' UTR were further confirmed by PCR on human genomic DNA using standard methods. (c) DNA sequencing of mRNA Cap Site & size of exon 1
  • mRNA Cap Site was determined to be about 48 bp upstream of the start codon based on 5' RACE sequencing. Ribonuclease Protection Assay confirmed a protected fragment of about 48 bp in size indicating that the start site from transcription resides about 48 bp upstream of ATG (start codon). Putative transcription factors have been identified by analysis of sequence with database transcription factor sequence information and these are set forth in Figure 3(S) [SEQUENCE ID NO: 2]. The 1.1 kb 5' UTR fragment was cloned into pCAT expression vectors to further analyze the promoter sequence region.
  • PI clone A colonies were streaked out on Kanamycin LB plates. A single colony was picked and grown O/N in 20 mis with 25 ⁇ g/ml of kanamycin. 500 mis of media (25 ⁇ g/ml kanamycin) was inoculated with 16 mis of the O/N culture and grown for 10 hours. Cells were pelleted by centrifugation and resuspended in 10 mis of Qiagen PI Solution. 10 mis of Qiagen P2 Solution was added and incubated at room temp, for 5 min. 10 mis of Qiagen P3 Solution was added and the mixture left on ice for 15 min. The sample was spun at 10,000g for 15 min. The supernatant was removed and extracted with phenol.
  • the supernatant was then re-extracted with chloroform.
  • the DNA was precipitated following addition of NaOAc pH 5.2 and 1.1 volumes of isopropanol.
  • the DNA was pelleted by centrifugation for 15 min. at 10,000g and washed with 70% ethanol.
  • 250 ⁇ l of DNA (about 50 ⁇ g) was added to 65 ⁇ l 30% PEG in 1.5M NaCI. 8.5 ⁇ l of 3M NaCI was added and the mixture incubated on ice for 30 min. The sample was spun at 12,000g for 10 min. The supernatant was discarded and the pellet dissolved in 200 ⁇ l distilled water.
  • the DNA was then extracted with chloroform, vortexed and spun at 10,000g for 1 min.
  • the aqueous layer was removed and the DNA precipitated with 40 ⁇ l of NaOAc pH 5.2 and 1 ml of ethanol.
  • the sample was spun at 12,000g for 30 min.
  • the DNA was washed with 1 ml of 70% ethanol and resuspended on distilled water.
  • the DNA was denatured with 0.1 volumes of 2M NaOH and 2mM EDTA and incubated at 37°C for 30 min.
  • the mixture was neutralized with 0.1 volume of 3M NaOAc pH 5.2 and precipitated with 2.5 volumes of ethanol.
  • the denatured DNA was resuspended in distilled water at a concentration of 1 ⁇ g/ ⁇ l 6 ⁇ g/ ⁇ l were used in each sequencing reaction (ABI) using TaqFS.
  • PI DNA was used for FISH analysis (Genome Systems, Inc.) to map to specific chromosome. Prior results done showed by use of 2 PCR somatic cell hybrid panels that the gene mapped to Chromosome 1. FISH analysis confirmed mapping to chromosome 1 and also further mapped the gene to lq21. This is the same locus as is known for cathepsin-S.
  • the DNA sequence encoding human cathepsin K in the deposited polynucleotide is amplified using PCR oligonudeotide primers specific to the amino acid carboxyl terminal sequence of the human cathepsin K protein and to vector sequences 3' to the gene. Additional nucleotides containing restriction sites to facilitate cloning are added to the 5' and 3' sequences respectively.
  • the 5' and 3' oligonudeotide primers are designed with sequences capable of mediating amplification by PCR.
  • the 3' primer has sequences complementary to a portion of the nucleotides of the cathepsin K coding sequence set out in Figure 2, including the stop codon.
  • the restriction sites are compatible with restriction enzyme sites in the bacterial expression vector pQE-70, for example, which is used for bacterial expression in certain of the Examples. (Qiagen, Inc. 9259 Eton Avenue, Chatsworth, CA, 9131 1).
  • pQE-70 encodes ampicillin antibiotic resistance (“Amp r ”) and contains a bacterial origin of replication (“ori”), an IPTG inducible promoter, a ribosome binding site (“RBS”), a 6-His tag and restriction enzyme sites.
  • the amplified human cathepsin K DNA and the vector pQE-70 both are digested with appropriate restriction enzymes to allow ligation with the restriction digested vector, and the digested DNAs then are ligated together. Insertion of the cathepsin K DNA into the restricted vector places the cathepsin K coding region downstream of and operably linked to the vector's IPTG-inducible promoter and in-frame with an initiating AUG appropriately positioned for translation of cathepsin K.
  • the ligation mixture is transformed into competent E. coli cells using standard procedures.
  • E. coli strain M15/rep4 containing multiple copies of the plasmid pREP4, which expresses lac repressor and confers kanamycin resistance ("Kan r "), is used in carrying out the illustrative example described here.
  • This strain which is only one of many that are suitable for expressing cathepsin K, is available commercially from Qiagen.
  • Transformants are identified by their ability to grow on LB plates in the presence of ampicillin (demonstrating Amp 1" ). Plasmid DNA is isolated from resistant colonies and the identity of the cloned DNA are confirmed by restriction analysis.
  • Clones containing the desired constructs are grown overnight ("O/N") in liquid culture in LB media supplemented with both ampicillin ( 100 ug/ml) and kanamycin (25 ug/ml).
  • the O/N culture is used to inoculate a large culture, at a dilution of approximately 1:100 to 1 :250.
  • the cells are grown to an optical density at 600nm ("OD600”) of between 0.4 and 0.6.
  • Isopropyl-B-D-thiogalactopyranoside (“IPTG”) is then added to a final concentration of 1 mM to induce transcription from lac repressor sensitive promoters, by inactivating the lad repressor. Cells subsequently are incubated further for 3 to 4 hours.
  • Inclusion bodies are purified from the disrupted cells using routine collection techniques, and protein is solubilized from the inclusion bodies into 8M urea.
  • the 8M urea solution containing the solubilized protein is passed over a PD-10 column in 2X phosphate buffered saline ("PBS"), thereby removing the urea, exchanging the buffer and refolding the protein.
  • PBS 2X phosphate buffered saline
  • the protein is purified by a further step of chromatography to remove endotoxin. Then, it is sterile filtered.
  • the sterile filtered protein preparation is stored in 2X PBS at a concentration of 95 micrograms per ml.
  • the gDNA sequence encoding the full length human cathepsin K protein is amplified using PCR oligonudeotide primers corresponding to the 5' and 3' sequences of the gene:
  • the 5' and 3' primers are provided with sequences capable of mediating PCR amplification followed by a stretch of about 20 bases of the sequence of cathepsin K of Figure 2. Inserted into an expression vector, as described below, the 5' end of the amplified fragment encoding human cathepsin K provides an efficient signal peptide. An efficient signal for initiation of translation in eukaryotic cells, as described by Kozak, M., J. Mol. Biol. 196: 947-950 (1987) is appropriately located in the vector portion of the construct.
  • the amplified fragment is isolated from a 1 % agarose gel using a commercially available kit ("Geneclean," BIO 101 Inc., La Jolla, Ca.). The fragment then is digested with BamHl and Asp718 and again is purified on a 1% agarose gel. This fragment is designated herein F2.
  • any of the many expression vectors known in the art for baculovirus expression can be used to express the cathepsin K protein in the baculovirus expression system, using standard methods, such as those described in Summers et al, A MANUAL OF METHODS FOR BACULOVIRUS VECTORS AND INSECT CELL CULTURE PROCEDURES, Texas Agricultural Experimental Station Bulletin No. 1555 (1987).
  • a preferred expression vector contains the strong polyhedrin promoter of the Autographa californica nuclear polyhedrosis virus (AcMNPV) followed by convenient restriction sites. The signal peptide of
  • AcMNPV gp67 including the N-terminal methionine, is located just upstream of a BamHl site.
  • the polyadenylation site of the simian virus 40 ("SV40") is used for efficient polyadenylation.
  • SV40 simian virus 40
  • the beta-galactosidase gene from E.coli is inserted in the same orientation as the polyhedrin promoter and is followed by the polyadenylation signal of the polyhedrin gene.
  • the polyhedrin sequences are flanked at both sides by viral sequences for cell-mediated homologous recombination with wild-type viral DNA to generate viable virus that express the cloned polynucleotide.
  • baculovirus vectors could be used in place of pA2-GP, such as pAc373, pVL941 and pAcIMl provided, as those of skill readily will appreciate, that construction provides appropriately located signals for transcription, translation, trafficking and the like, such as an in-frame AUG and a signal peptide, as required.
  • Such vectors are described in Luckow et al., Virology 170: 31-39, among others.
  • the plasmid is digested with the appropriate restriction enzymes, as can readily be determined by the skilled artisan, to remove the insert as a single fragment and the insert fragment is then dephosphorylated using calf intestinal phosphatase, using routine procedures known in the art.
  • V2 The DNA is then isolated from a 1% agarose gel using a commercially available kit ("Geneclean” BIO 101 Inc., La Jolla, Ca.). This vector DNA is designated herein "V2". Fragment F2 and the dephosphorylated plasmid V2 are ligated together with
  • E.coli HB 101 cells are transformed with ligation mix and spread on culture plates. Bacteria are identified that contain the plasmid with the human cathepsin K gene by digesting DNA from individual colonies using with the appropriate restriction nucleases, as can readily be determined by the skilled artisan, and then analyzing the digestion product by gel electrophoresis. The sequence of the cloned fragment is confirmed by DNA sequencing. This plasmid is designated herein pBaccathepsin K.
  • 5 mg of the plasmid pBaccathepsin K is co-transfected with 1.0 mg of a commercially available linearized baculovirus DNA ("BaculoGoldTM baculovirus DNA", Pharmingen, San Diego, CA.), using the lipofection method described by Feigner et al., Proc. Natl. Acad. Sci. USA 84: 7413-7417 (1987).
  • BaculoGoldTM virus DNA and 5 mg of the plasmid pBaccathepsin K are mixed in a sterile well of a microtiter plate containing 50 ml of serum free Grace's medium (Life Technologies Inc., Gaithersburg, MD).
  • plaque assay After four days the supernatant is collected and a plaque assay is performed, as described by Summers and Smith, cited above.
  • An agarose gel with "Blue Gal” (Life Technologies Inc., Gaithersburg) is used to allow easy identification and isolation of ⁇ gal-expressing clones, which produce blue-stained plaques.
  • a detailed description of a "plaque assay” of this type can also be found in the user's guide for insect cell culture and baculovirology distributed by Life Technologies Inc., Gaithersburg, page 9-10).
  • the virus is added to the cells. After appropriate incubation, blue stained plaques are picked with the tip of an Eppendorf pipette.
  • the agar containing the recombinant viruses is then resuspended in an Eppendorf tube containing 200 ml of Grace's medium.
  • the agar is removed by a brief centrifugation and the supernatant containing the recombinant baculovirus is used to infect Sf9 cells seeded in 35 mm dishes. Four days later the supernatants of these culture dishes are harvested and then they are stored at 4°C.
  • a clone containing properly inserted cathepsin K is identified by DNA analysis including restriction mapping and sequencing. This is designated herein as V-cathepsin K.
  • Sf9 cells are grown in Grace's medium supplemented with 10% heat-inactivated FBS.
  • the cells are infected with the recombinant baculovirus V-cathepsin K at a multiplicity of infection ("MOI") of about 2 (about 1 to about 3).
  • MOI multiplicity of infection
  • the medium is removed and is replaced with SF900 II medium minus methionine and cysteine (available from Life Technologies Inc., Gaithersburg). 42 hours later, 5 mCi of 35 S-methionine and 5 mCi 35 S cysteine (available from Amersham) are added.
  • the cells are further incubated for 16 hours and then they are harvested by centrifugation, lysed and the labeled proteins are visualized by SDS-PAGE and autoradiography.
  • Example 5 Expression of cathepsin K in COS cells
  • pCAT-CatK which contains the 1100 bp putative CatK promoter, upstream of the CAT reporter gene was transfected into COS cells by the DEAE-dextran procedure. Transfections were done on COS cells in 100mm dishes and 5 ⁇ g of DNA was used. As controls, pCAT basic, which contains no promoter or enhancer, and pCAT control, which contains the SV40 promoter and enhancer, were also transferred separately. 72 hours after transfection, extracts were made by freeze-thaw and equal amounts of extract protein were used in both 1-hour and overnight CAT assays. No activity was detected in untransfected COS cells.
  • pCAT-CatK showed a 1.4-1.6 fold increase of CAT expression relative to pCAT basic after subtraction of background from untransfected cells. Since it is possible that higher levels of activation may be obtained in the presence of various inducers, activation of the CatK promoter by adding exogenous 1 ,25 di-hydroxy vitamin D3 is believed to occur. Vitamin D has been shown by others to activate transcription of osteocalcin, osteopontin, calcitonin and P450 promoters through interaction with the vitamin D receptor and the vitamin D response element(s) found in these various promoters. The ability of vitamin D to transactivate these promoters is believed thought to play a role in the control of bone formation and reso ⁇ tion. Similar experiments can be performed to assess estrogen responsiveness which is also believed thought to play a role in the control of bone formation and reso ⁇ tion. (b) Expression Vector
  • the expression plasmid, cathepsin K HA is made by cloning a gDNA encoding cathepsin K into the expression vector pcDNAI/Amp (which can be obtained from Invitrogen, Inc.).
  • the expression vector pcDNAI/amp contains: (1) an E.coli origin of replication effective for propagation in E.
  • coli and other prokaryotic cell (2) an ampicillin resistance gene for selection of plasmid-containing prokaryotic cells; (3) an SV40 origin of replication for propagation in eukaryotic cells; (4) a CMV promoter, a polylinker, an SV40 intron, and a polyadenylation signal arranged so that a gDNA conveniently can be placed under expression control of the CMV promoter and operably linked to the SV40 intron and the polyadenylation signal by means of restriction sites in the polylinker.
  • a DNA fragment encoding the entire cathepsin K precursor and a HA tag fused in frame to its 3' end is cloned into the polylinker region of the vector so that recombinant protein expression is directed by the CMV promoter.
  • the HA tag corresponds to an epitope derived from the influenza hemagglutinin protein described by Wilson et al., Cell 37: 767 (1984).
  • the fusion of the HA tag to the target protein allows easy detection of the recombinant protein with an antibody that recognizes the HA epitope.
  • the plasmid construction strategy is as follows.
  • the cathepsin K gDNA of the deposit clone is amplified using primers that contained convenient restriction sites, much as described above regarding the construction of expression vectors for expression of cathepsin K in E. coli and S. furgiperda.
  • primers that contained convenient restriction sites, much as described above regarding the construction of expression vectors for expression of cathepsin K in E. coli and S. furgiperda.
  • one of the primers contains a heamaglutinin tag ("HA tag") as described above.
  • Suitable primers can readily be made by skilled artisans using known methods.
  • the PCR amplified DNA fragment and the vector, pcDNAI/Amp are digested with restriction endonuclease and then ligated.
  • the ligation mixture is transformed into E. coli strain SURE (available from Stratagene Cloning Systems, 11099 North Torrey Pines Road, La Jolla, CA 92037) the transformed culture is plated on ampicillin media plates which then are incubated to allow growth of ampicillin resistant colonies. Plasmid DNA is isolated from resistant colonies and examined by restriction analysis and gel sizing for the presence of the cathepsin K-encoding fragment.
  • COS cells are transfected with an expression vector, as described above, using DEAE-DEXTRAN, as described, for instance, in Sambrook et al., MOLECULAR CLONING: A LABORATORY MANUAL, Cold Spring Laboratory Press, Cold Spring Harbor, New York (1989). Cells are incubated under conditions for expression of cathepsin K by the vector.
  • cathepsin K HA fusion protein is detected by radiolabelling and immunoprecipitation, using methods described in, for example Harlow et al., ANTIBODIES: A LABORATORY MANUAL, 2nd Ed.; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1988). To this end, two days after transfection, the cells are labeled by incubation in media containing 35 S-cysteine for 8 hours. The cells and the media are collected, and the cells are washed and then lysed with detergent-containing RIPA buffer: 150 mM NaCI, 1% NP-40, 0.1 % SDS, 0.5% DOC, 50 mM TRIS, pH 7.5, as described by Wilson et al. cited above.
  • Proteins are precipitated from the cell lysate and from the culture media using an HA-specific monoclonal antibody. The precipitated proteins then are analyzed by SDS-PAGE gels and autoradiography. An expression product of the expected size is seen in the cell lysate, which is not seen in negative controls.
  • Northern blot analysis is carried out to examine the levels of expression of cathepsin K in human tissues, using methods described by, among others, Sambrook et al, cited above.
  • RNAzolTM B system Biotecx Laboratories, Inc. 6023 South Loop East, Houston, TX 77033.
  • RNA is isolated from tissue samples. The RNA is size resolved by electrophoresis through a 1% agarose gel under strongly denaturing conditions. RNA is blotted from the gel onto a nylon filter, and the filter then is prepared for hybridization to a detectably labeled polynucleotide probe.
  • the antisense strand of the coding region of the gDNA insert in the deposited clone is labeled to a high specific activity.
  • the gDNA is labeled by primer extension, using the Prime-It kit, available from Stratagene. The reaction is carried out using 50 ng of the gDNA, following the standard reaction protocol as recommended by the supplier.
  • the labeled polynucleotide is purified away from other labeled reaction components by column chromatography using a Select-G-50 column, obtained from 5-Prime - 3-Prime, Inc. of 5603 Arapahoe Road, Boulder, CO 80303.
  • the labeled probe is hybridized to the filter, at a concentration of 1 ,000,000 cpm/ml, in a small volume of 7% SDS, 0.5 M NaPO ⁇ pH 7.4 at 65°C, overnight.
  • Fibroblasts are obtained from a subject by skin biopsy.
  • the resulting tissue is placed in tissue-culture medium and separated into small pieces. Small chunks of the tissue are placed on a wet surface of a tissue culture flask, approximately ten pieces are placed in each flask.
  • the flask is turned upside down, closed tight and left at room temperature overnight. After 24 hours at room temperature, the flask is inverted - the chunks of tissue remain fixed to the bottom of the flask - and fresh media is added (e.g., Ham's F12 media, with 10% FBS, penicillin and streptomycin).
  • the tissue is then incubated at 37°C for approximately one week. At this time, fresh media is added and subsequently changed every several days. After an additional two weeks in culture, a monolayer of fibroblasts emerges. The monolayer is trypsinized and scaled into larger flasks.
  • a vector for gene therapy is digested with restriction enzymes for cloning a fragment to be expressed.
  • the digested vector is treated with calf intestinal phosphatase to prevent self-ligation.
  • the dephosphorylated, linear vector is fractionated on an agarose gel and purified.
  • Cathepsin K gDNA capable of expressing active cathepsin K is isolated.
  • Preferred constructs use the cathepsin K promoter for cell type-specific gene expression.
  • the ends of the fragment are modified, if necessary, for cloning into the vector. For instance, 5' overhanging may be treated with DNA polymerase to create blunt ends. 3' overhanging ends may be removed using S I nuclease. Linkers may be ligated to blunt ends with T4 DNA ligase.
  • Equal quantities of the Moloney murine leukemia virus linear backbone and the cathepsin K fragment are mixed together and joined using T4 DNA ligase.
  • the ligation mixture is used to transform E. Coli and the bacteria are then plated on agar-containing kanamycin. Kanamycin phenotype and restriction analysis confirm that the vector has the properly inserted gene.
  • Packaging cells are grown in tissue culture to confluent density in Dulbecco's
  • DMEM Modified Eagles Medium
  • CS calf serum
  • penicillin penicillin
  • streptomycin penicillin and streptomycin.
  • the vector containing the cathepsin K gene is introduced into the packaging cells by standard techniques. Infectious viral particles containing the cathepsin K gene are collected from the packaging cells, which now are called producer cells.
  • Fresh media is added to the producer cells, and after an appropriate incubation period media is harvested from the plates of confluent producer cells.
  • the media containing the infectious viral particles, is filtered through a Millipore filter to remove detached producer cells.
  • the filtered media then is used to infect fibroblast cells.
  • Media is removed from a sub-confluent plate of fibroblasts and quickly replaced with the filtered media.
  • Polybrene (Aldrich) may be included in the media to facilitate transduction. After appropriate incubation, the media is removed and replaced with fresh media. If the titer of virus is high, then virtually all fibroblasts will be infected and no selection is required. If the titer is low, then it is necessary to use a retroviral vector that has a selectable marker, such as neo or his, to select out transduced cells for expansion.
  • Engineered fibroblasts then may be injected into humans or animals, including for example, rats and mice, either alone or after having been grown to confluence on microcarrier beads, such as cytodex 3 beads.
  • the injected fibroblasts produce cathepsin K product, and the biological actions of the protein are conveyed to the host.
  • a fragment encoding pro-cathepsin K (no secretion signal) was inserted in the pET22b vector commercially available through Novagen, wherein the inserted gene is under the transcriptional control of the T7 promoter.
  • IL of shake flask grown E. coli expressing pCatK was pelleted (about 2.5g wet weight). The pellet was washed twice with 50 mL TBS+EDTA (50 mM Tris, 150 mM NaCI, 1 mM EDTA, pH 8.0). The washed pellet was solubilized into 25 mL of wash buffer by dispersion with a Tekmar tissuemizer and lysed by sonication on ice. Following centrifugation (13,000xg for 30 min at 4"C), the lysate pellet was again washed with 25 mL of lysis buffer and pelleted.
  • TBS+EDTA 50 mM Tris, 150 mM NaCI, 1 mM EDTA, pH 8.0
  • the washed pellet was solubilized into 25 mL of wash buffer by dispersion with a Tekmar tissuemizer and lysed by sonication on ice. Following centrifugation (13,000xg for 30 min at 4"C
  • the washed lysate pellet was solubilized using the tissuemizer into 25 mL of 50 mM Tris, 150 mM NaCI, 5 mM EDTA, 10 mM DTT, 8 M urea, pH 8.0. After stirring for 15 minutes the sample was centrifuged and the supernatant 0.45 ⁇ m filtered prior to protein assay at 6.75 mg/mL. 6.5 mL of this material (43.88 mg) was refolded by quick dilution into IL of stirring 50 mM Tris, 5 mM EDTA, 0.7 M L-argimne, 10 mM reduced and 1 mM oxidized glutathione, pH 8.0.
  • an approximately 25 mg of refolded, activated mature cathepsin K may be isolated from IL of shaker flask grown E. coli assuming minimal or no losses due to scale-up.
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • CTCAAAGCCC TTAACACTCT AGTTTTTAAA TGTCCTACTG GACATTTTGC CAGTTTGCAA 1620
  • AATTACATGT AAATGGACTA TAAGCAATTG TGTAAGCCAT ATGTCATGCT GCAGGCTGCA 1680
  • AATTGTTCTT AAAATGGAGG ATTTGTAATT AAGAAAGCCA ATGCAAGAAA TGAGTGAAGC 1740
  • TTCCATCCAT AACCTTGAGG CTTCTCTTGG TGTCCATACA TATGAACTGG CTATGAACCA 3300
  • CTGTCAAAAA TCAGGTACTC TCCTTTCTTC TGGGTGTGCA TATGTAATCT GGCATGACCT 3600
  • ATTAAGGTAC ATCTCCCCAT TCCCATTCTG TTATTTAACC TCATTTGTTT CTGTACATTT 3720
  • CACGCGCCAC TACGCCTGGC TAATTTTTCT ATTTTTATAG AGATGCGTTT TCACCATGTT 3960
  • GATTGCTCCA CACAATTATA CAGCTCTGTT GGCTCCTCCT CCCCAGCATG ATGTTTTGTA 5460
  • TCCTCTAGTC ATAGGAAGGA AATCATATCT GAAAGGGAGG CAACCTGAGG GGTTTTTTAT 5940
  • CTAGCTACTC CGGAAGCTGG TTGGGAGGAC AACTTGAGCC TGGGAGTTCA AGGCTGCTGT 6960 GAACTGTGAT CATGTCACTG CTCTCTAACC TGGGTGACAG AGTGAGACCC TGTCCCCAAA 7020
  • GTGAGCCACC ATGCCCCACC CCTTACTCAT TTTTAATTGG ATTGTTTTTT CTCTTTCTTA 7500
  • TTTTCCTCCC ACATTTATGT CTTAAAACAC CTGTAGGGGA TTTTTTTTTT TTTTTTTTTT 9780
  • AATAGTGCTC AATTATGTGC TGGGCAGATG CTCCCTGTGT GCAAGGTGCT TAGTGACATA 11460
  • TCTCCTTCCC CACATCTGTT TATGGAAGAA AATGAAGTTT GGGGTGTGGT TTGAGGAATC 240
  • ATCCACTGCC TCCTTCCCTC CTCCCTACCC TTCCTTCTCT CAGCATTTCT ATCCCCGCCT 1020
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • TGCTTTGTTC TTGAGGTGAA AGGGCACCAG GAAAAGAGGG CAAGGAATTA AGGTACATCT 120
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • AAACCCACAA ACCCACCATG CTATTTTCAC AAATTGTGTG GCTTTATTTT ATATGATCTC 360
  • AATTGTTTGC CAAAATATTT GGAAAAATTT ATACCTGCAA TGTGTAAGAA ATCCCCTTCA 1020
  • CTGTGAACTG TGATCATGTC ACTGCTCTCT AACCTGGGTG ACAGAGTGAG ACCCTGTCCC 1620
  • CAAAAAACAA CAACCGTTTT TTTTTTTGGTAG AGACATTGTC TCGCTATGTT GCCAAGGCTA 1680 GTCTCAAACT CCTGGGCTCA AGCAATCCTC CCACCTCCCC AAAGTGCTGG GATTTATAGA 1740
  • TGTAAGCCAC CATGCCTGGC CTACCCTTTT TTTTTTTTTT TGAAATGGAG TTTTGCTTTT 1800
  • TGCAGTGAGC CAAGATCATG CCATTGCACT CCAGCCTGGG TAACAGAGAC TCTGTTTCAA 3420
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • GAGCATCTTC CCAAGTCTCC CACAGCTTCT GTCCTGTGTT GTCCCTTCTC TTGACTCCCA 1320
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • CTGGGGAGAA AACTGGGGAA ACAAAGGATA TATCCTCATG GCTCGAAATA AGAACAACGC 60 CTGTGGCATT GCCAACCTGG CCAGCTTCCC CAAGATG 97
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • GATTGTCTGT TCAGTTTCCC CATTTGTTTG TGCTTCAAAT GATCCTTCCT ACTTTGCTTC 180
  • MOLECULE TYPE cDNA
  • HYPOTHETICAL NO
  • ANTISENSE NO
  • FRAGMENT TYPE (vi) ORIGINAL SOURCE:
  • GGAGACTAAA TTAAGTGACA TGGAAACTTG GGTCTTGAAA AACATTTTAA GGTTATTTTT 300
  • Trp Glu Lys Asn Leu Lys Tyr lie Ser lie His Asn Leu Glu Ala

Abstract

The invention relates to cathepsin K polypeptides, polynucleotides encoding the polypeptides, methods for producing the polypeptides, in particular by expressing the polynucleotides, and agonists and antagonists of the polypeptides. The invention further relates to methods for utilizing such polynucleotides, polypeptides, agonists and antagonists for applications, which relate, in part, to research, diagnostic and clinical arts.

Description

CATHEPSIN K GENE
This invention relates, in part, to newly identified polynucleotides and polypeptides; variants and derivatives of the polynucleotides and polypeptides; processes for making the polynucleotides and the polypeptides, and their variants and derivatives; agonists and antagonists of the polypeptides; and uses of the polynucleotides, polypeptides, variants, derivatives, agonists and antagonists. In particular, in these and in other regards, the invention relates to polynucleotides and polypeptides of human cathepsin K, especially genomic sequences of cathepsin K, and most especially promoter and intronic sequences.
BACKGROUND OF THE INVENTION
Bone resorption involves the simultaneous removal of both the mineral and the organic constituents of the extracellular matrix. This occurs mainly in an acidic phagolysosome-like extracellular compartment covered by the ruffled border of osteoclasts. Barron, et al., J. Cell Biol., 101:2210-22, (1985). Osteoclasts are multinucleate giant cells that play key roles in bone resorption. Attached to the bone surface, osteoclasts produce an acidic microenvironment between osteoclasts and bone matrix. In this acidic microenvironment, bone minerals and organic components are solubilized. Organic components, mainly type-I collagen, are thought to be solubilized by protease digestion. There is evidence that cysteine proteinases may play an important role in the degradation of organic components of bone. Among cysteine proteinases, cathepsins B, L, H, and S can degrade type-I collagen in the acidic condition. Etherington, D.J. Biochem. J., 127, 685-692 (1972). Cathepsin L is the most active of the lysosomal cysteine proteases with regard to its ability to hydrolyze azocasein, elastin, and collagen.
Cathepsins are proteases that function in the normal physiological as well as pathological degradation of connective tissue. Cathepsins play a major role in intracellular protein degradation and turnover, bone remodeling, and prohormone activation. Marx, J.L., Science. 235:285-286 (1987). Cathepsin B, H, L and S are ubiquitously expressed lysosomal cysteine proteinases that belong to the papain superfamily. They are found at constitutive levels in many tissues in the human including kidney, liver, lung and spleen. Some pathological roles of cathepsins include an involvement in glomerulonephritis, arthritis, and cancer metastasis. Sloan, B.F., and Honn, K.V., Cancer Metastasis Rev., 3:249-263 (1984). Greatly elevated levels of cathepsin L and B mRNA and protein are seen in tumor cells. Cathepsin L mRNA is also induced in fibroblasts treated with tumor promoting agents and growth factors. Kane, S.E. and Gottesman, M.M. Cancer Biology, 1:127-136 (1990). The gene expression and cellular content of a non-cysteine protease, cathepsin D, in Alzheimer's disease brain showed evidence for early up-regulation of the endosomal-lysosomal system. Cataldo AM, et al., Neuron, 1995, 14 (3), 671- 680).
In vitro studies on bone resorption have shown that cathepsins L and B may be involved in the remodelling of this tissue. These lysosomal cysteine proteases digest extracellular matrix proteins such as elastin, laminin, and type I collagen under acidic conditions. Osteoclast cells require this activity to degrade the organic matrix prior to bone regeneration accomplished by osteoblasts. Several natural and synthetic inhibitors of cysteine proteinases have been effective in inhibiting the degradation of this matrix. The isolation of cathepsins and their role in bone resorption has been the subject of an intensive study. OC-2 has recently been isolated from pure osteoclasts from rabbit bones. The OC-2 was found to encode a possible cysteine proteinase structurally related to cathepsins L and S. Tezuka, K., et al., J. Biol. Chem., 269: 1106- 1109, (1994). An inhibitor of cysteine proteinases and collagenase, Z-Phe-Ala-CHN2 has been studied for its effect on the resorptive activity of isolated osteoclasts and has been found to inhibit resorption pits in dentine. Delaisse, J.M. et al., Bone, 8:305-313 (1987). Also, the effect of human recombinant cystatin C, a cysteine proteinase inhibitor, on bone resorption in vitro has been evaluated, and has been shown to significantly inhibit bone resorption which has been stimulated by parathyroid hormone. Lerner, U.H. and Grubb Anders, Journal of Bone and Mineral Research, 7:433-439, (1989). Further, a cDNA clone encoding the human cysteine protease cathepsin L has been recombinantly manufactured and expressed at high levels in E. coli in a T7 expression system. Recombinant human procathepsin L was successfully expressed at high levels and purified as both procathepsin L and active processed cathepsin L forms. Information about the possible function of the propeptide in cathepsin L folding and/or processing and about the necessity for the light chain of the enzyme for protease activity was obtained by expressing and purifying mutant enzymes carrying structural alterations in these regions. Smith, S.M. and Gottesman, M.M., J. Bio Chem., 264:20487-20495, (1989). There has also been reported the expression of a functional human cathepsin S in Saccharomyces cerevisiae and the characterization of the recombinant enzyme. Bromme, D. et al., J. Biol. Chem., 268:4832-4838 (1993).
SUMMARY OF THE INVENTION Toward these ends, and others, it is an object of the present invention to provide polypeptides, inter alia, that have been identified as novel cathepsin K by homology between the amino acid sequence set out in Figure 5 and known amino acid sequences of other proteins such as rabbit OC-2 and human cathepsin O cDNA. Tezuka, K., et al., J. Biol. Chem., 269:1106-1109, (1994). It is a further object of the invention, moreover, to provide polynucleotides that encode cathepsin K, particularly polynucleotides that encode the polypeptide herein designated cathepsin K.
In a particularly preferred embodiment of this aspect of the invention the polynucleotide comprises the region encoding human cathepsin K in the sequence set out in Figure 1 [SEQ ID NO: 1 ] or in the genomic DNA (herein "gDNA") in ATCC deposit No. 98035 (referred to herein as the deposited clone).
In accordance with this aspect of the invention there are provided isolated nucleic acid molecules encoding human cathepsin K, including mRNAs, cDNAs, genomic DNAs and, in further embodiments of this aspect of the invention, biologically, diagnostically, clinically or therapeutically useful variants, analogs or derivatives thereof, or fragments thereof, including fragments of the variants, analogs and derivatives.
Among the particularly preferred embodiments of this aspect of the invention are naturally occurring allelic variants of human cathepsin K. It also is an object of the invention to provide cathepsin K polypeptides, particularly human cathepsin K polypeptides, that cause or are associated with disease, for example, osteoporosis, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hyperparathyroidism, bone degradation, metastatic tumors, rhemuatoid arthritis, osteoarthritis, peridontal disease and degradation of bone implants and bone protheses, particularly dental implants. In accordance with this aspect of the invention there are provided novel polypeptides of human origin referred to herein as cathepsin K as well as biologically, diagnostically or therapeutically useful fragments, variants and derivatives thereof, variants and derivatives of the fragments, and analogs of the foregoing.
Among the particularly preferred embodiments of this aspect of the invention are variants of human cathepsin K encoded by naturally occurring alleles of the human cathepsin K gene.
It is another object of the invention to provide a process for producing the aforementioned polypeptides, polypeptide fragments, variants and derivatives, fragments of the variants and derivatives, and analogs of the foregoing.
In a preferred embodiment of this aspect of the invention there are provided methods for producing the aforementioned cathepsin K polypeptides comprising culturing host cells having expressibly incoφorated therein an exogenously-derived human cathepsin K-encoding polynucleotide under conditions for expression of human cathepsin K in the host and then recovering the expressed polypeptide.
In accordance with yet another object of the invention there are methods to determine drug responsiveness of individuals having or suspected of having a defect in the cathepsin K gene. In accordance with yet another object the invention there are provided products, compositions, processes and methods that utilize the aforementioned polypeptides and polynucleotides for research, biological, clinical and therapeutic purposes, inter alia.
In accordance with certain preferred embodiments of this aspect of the invention, there are provided products, compositions and methods, inter alia, for, among other things: assessing cathepsin K expression in cells by determining cathepsin K polypeptides of cathepsin K-encoding mRNA or hnRNA in vitro, ex vivo or in vivo by exposing cells to cathepsin K polypeptides, polynucleotides or antibodies as disclosed herein; assaying genetic variation and aberrations, such as defects, in cathepsin K polynucleotides, genes and gene control sequences; and administering a cathepsin K polypeptide or polynucleotide to an organism to augment cathepsin K function or remediate cathepsin K dysfunction.
In accordance with certain preferred embodiments of this and other aspects of the invention there are provided probes that hybridize specifically to human cathepsin K sequences. In certain additional preferred embodiments of this aspect of the invention there are provided antibodies against cathepsin K polypeptides. In certain particularly preferred embodiments in this regard, the antibodies are highly selective for human cathepsin K.
In accordance with another aspect of the present invention, there are provided cathepsin K agonists. Among preferred agonists are molecules that mimic cathepsin K, that bind to cathepsin K-binding molecules or receptor molecules, and that elicit or augment cathepsin K-induced responses. Also among preferred agonists are molecules that interact with cathepsin K or cathepsin K polypeptides, or with other modulators of cathepsin K activities, and thereby potentiate or augment an effect of cathepsin K or more than one effect of cathepsin K.
In accordance with yet another aspect of the present invention, there are provided cathepsin K antagonists. Among preferred antagonists are those which mimic cathepsin K so as to bind to cathepsin K receptor or binding molecules but not elicit a cathepsin K-induced response or more than one cathepsin K-induced response. Also among preferred antagonists are molecules that bind to or interact with cathepsin K so as to inhibit an effect of cathepsin K or more than one effect of cathepsin K.
The agonists and antagonists may be used to mimic, augment or inhibit the action of cathepsin K polypeptides. They may be used, for instance, to treat osteoporosis, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hyperparathyroidism, bone degradation, metastatic tumors, and degradation of bone implants and bone protheses, particularly dental implants. Such antagonists may be particularly useful to treat osteoporosis, Paget's disease, Gaucher's disease, Alzheimer's disease, hyperparathyroidism, bone degradation, metastatic tumors, CNS inflammation, rhemuatoid arthritis, osteoarthritis, periodontal disease and degradation of bone implants and bone protheses, particularly dental implants.
In a further aspect of the invention there are provided compositions comprising a cathepsin K polynucleotide or a cathepsin K polypeptide for administration to cells in vitro, to cells ex vivo and to cells in vivo, or to a multicellular organism. In certain particularly preferred embodiments of this aspect of the invention, the compositions comprise a cathepsin K polynucleotide for expression of a cathepsin K polypeptide in a host organism for treatment of disease. Particularly preferred in this regard is expression in a human patient for treatment of a dysfunction associated with aberrant endogenous activity of cathepsin K or to provide therapeutic.
Other objects, features, advantages and aspects of the present invention will become apparent to those of skill from the following description. It should be understood, however, that the following description and the specific examples, while indicating preferred embodiments of the invention, are given by way of illustration only. Various changes and modifications within the spirit and scope of the disclosed invention will become readily apparent to those skilled in the art from reading the following description and from reading the other parts of the present disclosure.
BRIEF DESCRIPTION OF THE DRAWINGS The following drawings depict certain embodiments of the invention. They are illustrative only and do not limit the invention otherwise disclosed herein.
Figure 1 shows the genomic nudeotide sequence of human cathepsin K [SEQ ID NO: 1].
Figure 2 shows the nudeotide, exon-intron boundaries and deduced amino acid sequence of human cathepsin K.
Figure 3 (A - S) shows structural features of cathepsin K [SEQ ID NO: 2-19].
Figure 4 shows the intron-exon junctions.
Figure 5 shows the regions of similarity between amino acid sequences of cathepsin K, human cathepsins S, L, H, B, D, E, G and rabbit OC2 polypeptides.
Figure 6 shows the deduced amino acid sequence of human cathepsin K.
GLOSSARY
The following illustrative explanations are provided to facilitate understanding of certain terms used frequently herein, particularly in the examples. The explanations are provided as a convenience and are not limitative of the invention. DIGESTION of DNA refers to catalytic cleavage of the DNA with a restriction enzyme that acts only at certain sequences in the DNA. The various restriction enzymes referred to herein are commercially available and their reaction conditions, cofactors and other requirements for use are known and routine to the skilled artisan. For analytical purposes, typically, 1 μg of plasmid or DNA fragment is digested with about 2 units of enzyme in about 20 ml of reaction buffer. For the purpose of isolating DNA fragments for plasmid construction, typically 5 to 50 μg of DNA are digested with 20 to 250 units of enzyme in proportionately larger volumes.
Appropriate buffers and substrate amounts for particular restriction enzymes are described in standard laboratory manuals, such as those referenced below, and they are specified by commercial suppliers.
Incubation times of about 1 hour at 37°C are ordinarily used, but conditions may vary in accordance with standard procedures, the supplier's instructions and the particulars of the reaction. After digestion, reactions may be analyzed, and fragments may be purified by electrophoresis through an agarose or polyacrylamide gel, using well known methods that are routine for those skilled in the art.
GENETIC ELEMENT generally means a polynucleotide comprising a region that encodes a polypeptide or a region that regulates transcription or translation or other processes important to expression of the polypeptide in a host cell, or a polynucleotide comprising both a region that encodes a polypeptide and a region operably linked thereto that regulates expression.
Genetic elements may be comprised within a vector that replicates as an episomal element; that is, as a molecule physically independent of the host cell genome. They may be comprised within mini-chromosomes, such as those that arise during amplification of transfected DNA by methotrexate selection in eukaryotic cells. Genetic elements also may be comprised within a host cell genome; not in their natural state but, rather, following manipulation such as isolation, cloning and introduction into a host cell in the form of purified DNA or in a vector, among others. IDENTITY means the degree of sequence relatedness between two polypeptide or two polynucleotides sequences as determined by the identity of the match between two strings of such sequences. Identity can be readily calculated. While there exist a number of methods to measure identity between two polynucleotide or polypeptide sequences, the term "identity" is well known to skilled artisans (Computational Molecular Biology, Lesk, A.M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D.W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A.M., and Griffin, H.G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991). Methods commonly employed to determine identity between two sequences include, but are not limited to disclosed in Guide to Huge Computers, Martin J. Bishop, ed., Academic Press, San Diego, 1994, and Carillo, H., and Lipman, D., SIAM J. Applied Math., 48: 1073 (1988). Preferred methods to determine identity are designed to give the largest match between the two sequences tested. Such methods are codified in computer programs. Preferred computer program methods to determine identity between two sequences include, but are not limited to, GCG program package (Devereux, J., et al., Nucleic Acids Research 12(1): 387 (1984)), BLASTP, BLASTN, FASTA (Atschul, S.F. et al., J. Molec. Biol. 215: 403 (1990)). ISOLATED means altered "by the hand of man" from its natural state; i.e., that, if it occurs in nature, it has been changed or removed from its original environment, or both.
For example, a naturally occurring polynucleotide or a polypeptide naturally present in a living animal in its natural state is not "isolated," but the same polynucleotide or polypeptide separated from some or all of the coexisting materials of its natural is "isolated", as the term is employed herein.
As part of or following isolation, such polynucleotides can be joined to other polynucleotides, such as DNAs, for mutagenesis, to form fusion proteins, and for propagation or expression in a host, for instance. The isolated polynucleotides, alone or joined to other polynucleotides such as vectors, can be introduced into host cells, in culture or in whole organisms. Introduced into host cells in culture or in whole organisms, such DNAs still would be isolated, as the term is used herein, because they would not be in their naturally occurring form or environment. Similarly, the polynucleotides and polypeptides may occur in a composition, such as a media formulations, solutions for introduction of polynucleotides or polypeptides, for example, into cells, compositions or solutions for chemical or enzymatic reactions, for instance, which are not naturally occurring compositions, and, therein remain isolated polynucleotides or polypeptides within the meaning of that term as it is employed herein.
LIGATION refers to the process of forming phosphodiester bonds between two or more polynucleotides, which most often are double stranded DNAs. Techniques for ligation are well known to the art and protocols for ligation are described in standard laboratory manuals and references, such as, for instance, Sambrook et al., MOLECULAR CLONING, A LABORATORY MANUAL, 2nd Ed.; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1989) and Maniatis et al., pg. 146, as cited below.
OLIGONUCLEOTIDE(S) refers to relatively short polynucleotides. Often the term refers to single-stranded deoxyribonucleotides, but it can refer as well to single-, double-, or triple-stranded ribonucleotides, antisense polynucleotides, RNA:DNA hybrids and double-stranded DNAs, among others. Oligonudeotides, such as single-stranded DNA probe oligonudeotides, often are synthesized by chemical methods, such as those implemented on automated oligonudeotide synthesizers. However, oligonudeotides can be made by a variety of other methods, including in vitro recombinant DNA-mediated techniques and by expression of DNAs in cells and organisms. Initially, chemically synthesized DNAs typically are obtained without a 5' phosphate. The 5' ends of such oligonudeotides are not substrates for phosphodiester bond formation by ligation reactions that employ DNA ligases typically used to form recombinant DNA molecules. Where ligation of such oligonudeotides is desired, a phosphate can be added by standard techniques, such as those that employ a kinase and ATP.
The 3' end of a chemically synthesized oligonudeotide generally has a free hydroxyl group and, in the presence of a ligase, such as T4 DNA ligase, readily will form a phosphodiester bond with a 5' phosphate of another polynucleotide, such as another oligonudeotide. As is well known, this reaction can be prevented selectively, where desired, by removing the 5' phosphates of the other polynucleotide(s) prior to ligation. PLASMIDS generally are designated herein by a lower case letter/? preceded and/or followed by capital letters and/or numbers, in accordance with standard naming conventions that are familiar to those of skill in the art. Starting plasmids disclosed herein are either commercially available, publicly available on an unrestricted basis, or can be constructed from available plasmids by routine application of well known, published procedures. Many plasmids and other cloning and expression vectors that can be used in accordance with the present invention are well known and readily available to those of skill in the art. Moreover, those of skill readily may construct any number of other plasmids suitable for use in the invention. The properties, construction and use of such plasmids, as well as other vectors, in the present invention will be readily apparent to those of skill from the present disclosure.
POLYNUCLEOTIDE(S) generally refers to any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA. Thus, for instance, polynucleotides as used herein refers to, among others, single-and double-stranded DNA, DNA that is a mixture of single-and double-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded or a mixture of single- and double-stranded regions.
In addition, polynucleotide as used herein refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA. The strands in such regions may be from the same molecule or from different molecules. The regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules. One of the molecules of a triple-helical region often is an oligonudeotide.
As used herein, the term polynucleotide includes DNAs or RNAs as described above that contain one or more modified bases. Thus, DNAs or RNAs with backbones modified for stability or for other reasons are "polynucleotides" as that term is intended herein. Moreover, DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples, are polynucleotides as the term is used herein.
It will be appreciated that a great variety of modifications have been made to DNA and RNA that serve many useful purposes known to those of skill in the art. The term polynucleotide as it is employed herein embraces such chemically, enzymatically or metabolically modified forms of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including simple and complex cells, inter alia.
POLYPEPTIDES, as used herein, includes all polypeptides as described below. The basic structure of polypeptides is well known and has been described in innumerable textbooks and other publications in the art. In this context, the term is used herein to refer to any peptide or protein comprising two or more amino acids joined to each other in a linear chain by peptide bonds. As used herein, the term refers to both short chains, which also commonly are referred to in the art as peptides, oligopeptides and oligomers, for example, and to longer chains, which generally are referred to in the art as proteins, of which there are many types.
It will be appreciated that polypeptides often contain amino acids other than the 20 amino acids commonly referred to as the 20 naturally occurring amino acids, and that many amino acids, including the terminal amino acids, may be modified in a given polypeptide, either by natural processes, such as processing and other post-translational modifications, but also by chemical modification techniques which are well known to the art. Even the common modifications that occur naturally in polypeptides are too numerous to list exhaustively here, but they are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature, and they are well known to those of skill in the art. Among the known modifications which may be present in polypeptides of the present are, to name an illustrative few, acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nudeotide or nudeotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cystine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
Such modifications are well known to those of skill and have been described in great detail in the scientific literature. Several particularly common modifications, glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation, for instance, are described in most basic texts, such as, for instance PROTEINS - STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993). Many detailed reviews are available on this subject, such as, for example, those provided by Wold, F., Posttranslational Protein Modifications: Perspectives and Prospects, pgs. 1-12 in POSTTRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York (1983); Seifter et al., Analysis for protein modifications and nonprotein cofactors, Meth. Enzymol. 182: 626-646 (1990) and Rattan et al., Protein Synthesis: Posttranslational Modifications and Aging, Ann. N.Y. Acad. Sci. 663: 48-62 (1992).
It will be appreciated, as is well known and as noted above, that polypeptides are not always entirely linear. For instance, polypeptides may be branched as a result of ubiquitination, and they may be circular, with or without branching, generally as a result of posttranslation events, including natural processing event and events brought about by human manipulation which do not occur naturally. Circular, branched and branched circular polypeptides may be synthesized by non-translation natural process and by entirely synthetic methods, as well.
Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. In fact, blockage of the amino or carboxyl group in a polypeptide, or both, by a covalent modification, is common in naturally occurring and synthetic polypeptides and such modifications may be present in polypeptides of the present invention, as well. For instance, the amino terminal residue of polypeptides made in E. coli, prior to proteolytic processing, almost invariably will be N-formylmethionine.
The modifications that occur in a polypeptide often will be a function of how it is made. For polypeptides made by expressing a cloned gene in a host, for instance, the nature and extent of the modifications in large part will be determined by the host cell posttranslational modification capacity and the modification signals present in the polypeptide amino acid sequence. For instance, as is well known, glycosylation often does not occur in bacterial hosts such as E. coli. Accordingly, when glycosylation is desired, a polypeptide should be expressed in a glycosylating host, generally a eukaryotic cell. Insect cells often carry out the same posttranslational glycosylations as mammalian cells and, for this reason, insect cell expression systems have been developed to express efficiently mammalian proteins having native patterns of glycosylation, inter alia. Similar considerations apply to other modifications.
It will be appreciated that the same type of modification may be present in the same or varying degree at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications.
In general, as used herein, the term polypeptide encompasses all such modifications, particularly those that are present in polypeptides synthesized by expressing a polynucleotide in a host cell.
VARIANT(S) of polynucleotides or polypeptides, as the term is used herein, are polynucleotides or polypeptides that differ from a reference polynucleotide or polypeptide, respectively. Variants in this sense are described below and elsewhere in the present disclosure in greater detail.
(1) A polynucleotide that differs in nudeotide sequence from another, reference polynucleotide. Generally, differences are limited so that the nudeotide sequences of the reference and the variant are closely similar overall and, in many regions, identical. As noted below, changes in the nudeotide sequence of the variant may be silent. That is, they may not alter the amino acids encoded by the polynucleotide. Where alterations are limited to silent changes of this type a variant will encode a polypeptide with the same amino acid sequence as the reference. Also as noted below, changes in the nudeotide sequence of the variant may alter the amino acid sequence of a polypeptide encoded by the reference polynucleotide. Such nudeotide changes may result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence, as discussed below.
(2) A polypeptide that differs in amino acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference and the variant are closely similar overall and, in many region, identical. A variant and reference polypeptide may differ in amino acid sequence by one or more substitutions, additions, deletions, fusions and truncations, which may be present in any combination.
RECEPTOR MOLECULE, as used herein, refers to molecules which bind or interact specifically with cathepsin K polypeptides of the present invention, including not only classic receptors and enzymatic substrates, both of which are preferred, but also other molecules that specifically bind to or interact with polypeptides of the invention (which also may be referred to as "binding molecules" and "interaction molecules," respectively and as "cathepsin K binding molecules" and "cathepsin K interaction molecules." These cathepsin K binding molecules also include, for example, cathepsin K substrate analogs. Binding between polypeptides of the invention and such molecules, including receptor or binding or interaction molecules may be exclusive to polypeptides of the invention, which is very highly preferred, or it may be highly specific for polypeptides of the invention, which is highly preferred, or it may be highly specific to a group of proteins that includes polypeptides of the invention, which is preferred, or it may be specific to several groups of proteins at least one of which includes polypeptides of the invention. Receptors also may be non-naturally occurring, such as antibodies and antibody-derived reagents that bind specifically to polypeptides of the invention.
DESCRIPTION OF THE INVENTION The present invention relates to novel cathepsin K polypeptides and polynucleotides, among other things, as described in greater detail below. In particular, the invention relates to polypeptides and polynucleotides of a novel human cathepsin K, which is related by amino acid sequence homology to rabbit OC-2 and human cathepsin O cDNA. Tezuka, K., et al., J. Biol. Chem., 269: 1106- 1109, (1994). The invention relates especially to cathepsin K having the nudeotide sequences set out in Figure 1 [SEQ ID NO: 1], and to the cathepsin K nudeotide sequences of the gDNA in ATCC Deposit No. 98035, which is herein referred to as "the deposited clone" or as the "gDNA of the deposited clone." It will be appreciated that the nudeotide sequences set out in Figure 1 [SEQ ID NO: 1] were obtained by sequencing the gDNA of the deposited clone, as more specifically set forth elsewhere herein. Hence, the sequence of the deposited clone is controlling as to any discrepancies between the two.
Polynucleotides
In accordance with one aspect of the present invention, there are provided isolated polynucleotides which encode the cathepsin K polypeptide having the deduced amino acid sequence of Figure 2 [SEQ ID NO:20] (see also Figure 6 for the deduced amino acid sequence) or the cathepsin K polypeptide encoded by the gDNA in the deposited clone.
Using the information provided herein, such as the polynucleotide sequence set out in Figure 1 [SEQ ID NO: 1], a polynucleotide of the present invention encoding human cathepsin K polypeptide may be obtained using standard cloning and screening procedures, such as those for cloning gDNAs using DNA from cells of a human as starting material. Illustrative of the invention, the polynucleotide set out in Figure 1 [SEQ ID NO: 1] was discovered in a human gDNA library as described in Example 1.
Human cathepsin K of the invention is structurally related to other proteins of the cathepsin family, as shown by the results of sequencing the gDNA encoding human cathepsin K in the deposited clone. The gDNA sequence thus obtained is set out in Figure 1 [SEQ ID NO: 1]. It contains a non-contiguous open reading frame encoding, after intron removal, but including all exons, a protein of about 329 amino acid residues.
Polynucleotides of the present invention may be in the form of RNA, such as mRNA or hnRNA, or in the form of DNA, including, for instance, cDNA and gDNA obtained by cloning or produced by chemical synthetic techniques or by a combination thereof. The DNA may be triple-stranded, double-stranded or single-stranded. Single-stranded DNA may be the coding strand, also known as the sense strand, or it may be the non-coding strand, also referred to as the anti-sense strand. The coding sequence which encodes the polypeptide may be identical to the exon sequence of the polynucleotide shown in Figure 1 [SEQ ID NO: 1] or that of the deposited clone. It also may be a polynucleotide with a different sequence, which, as a result of the redundancy (degeneracy) of the genetic code, encodes the polypeptide of the DNA of Figure 2 [SEQ ID NO:20] or of the deposited gDNA, including, but not limited to, splice variants transcribed from such gDNA.
Polynucleotides of the present invention which encode the polypeptide of Figure 1 [SEQ ID NO: 1] or the polypeptide encoded by the deposited gDNA may include, but are not limited to the coding sequence for the mature polypeptide, by itself; the coding sequence for the mature polypeptide and additional coding sequences, such as those encoding a leader or secretory sequence, such as a pre-, or pro- or prepro- protein sequence; the coding sequence of the mature polypeptide, with or without the aforementioned additional coding sequences, together with additional, non-coding sequences, including for example, but not limited to introns and non-coding 5' and 3' sequences, such as the transcribed, non-translated sequences that play a role in transcription, mRNA processing - including splicing and polyadenylation signals, for example - ribosome binding and stability of mRNA; additional coding sequence which codes for additional amino acids, such as those which provide additional functionalities. Thus, for instance, the polypeptide may be fused to a marker sequence, such as a peptide, which facilitates purification of the fused polypeptide. In certain preferred embodiments of this aspect of the invention, the marker sequence is a hexa-histidine peptide, such as the tag provided in the vector pQE-9, among others, many of which are commercially available. As described in Gentz et al., Proc. Natl. Acad. Sci., USA 86: 821-824 (1989), for instance, hexa-histidine provides for convenient purification of the fusion protein. The HA tag corresponds to an epitope derived of influenza hemagglutinin protein, which has been described by Wilson et al., Cell 37: 767 ( 1984), for instance. In accordance with the foregoing, the term "polynucleotide encoding a polypeptide" as used herein encompasses polynucleotides which include a sequence encoding a polypeptide of the present invention, particularly the human cathepsin K having the amino acid sequence set out in Figure 2 [SEQ ID NO:20] or the amino acid sequence of the human cathepsin K encoded by the gDNA of the deposited clone. The term encompasses polynucleotides that include a single continuous region or discontinuous regions encoding the polypeptide (for example, interrupted by introns) together with additional regions, that also may contain coding and/or non-coding sequences. The present invention further relates to variants of the herein above described polynucleotides which encode for fragments, analogs and derivatives of the polypeptide having the deduced amino acid sequence of Figure 2 [SEQ ID NO:20] or the polypeptide encoded by the exons of the gDNA of the deposited clone, including, but not limited to, splice variants transcribed from such gDNA. A variant of the polynucleotide may be a naturally occurring variant such as a naturally occurring allelic variant or splice variant, or it may be a variant that is not known to occur naturally. Such non-naturally occurring variants of the polynucleotide may be made by mutagenesis techniques, including those applied to polynucleotides, cells or organisms. Such non-naturally occurring variants of the polynucleotide may be made by modifying splice acceptor, donor and/or branch sites, or by expressing the gDNA in cells where it is not naturally expressed, or cell extracts made from such cells.
Among variants in this regard are variants that differ from the aforementioned polynucleotides by nudeotide substitutions, deletions or additions. The substitutions, deletions or additions may involve one or more nucleotides. The variants may be altered in coding or non-coding regions or both. Alterations in the coding regions may produce conservative or non-conservative amino acid substitutions, deletions or additions.
Among the particularly preferred embodiments of the invention in this regard are polynucleotide sequence of cathepsin K set out in Figure 1 [SEQ ID NO: 1] or the polynucleotide sequence of cathepsin K of the gDNA of the deposited clone; variants, analogs, derivatives and fragments thereof, and fragments of the variants, analogs and derivatives.
Further particularly preferred in this regard are polynucleotides encoding cathepsin K variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments, which have the amino acid sequence of the cathepsin K polypeptide of Figure 2 [SEQ ID NO:20] or of the deposit in which several, a few, 5 to 10, 1 to 5, 1 to 3, 2, 1 or no amino acid residues are substituted, deleted or added, in any combination. Especially preferred among these are silent substitutions, additions and deletions, which do not alter the properties and activities of the cathepsin K. Also especially preferred in this regard are conservative substitutions. Most highly preferred are polypeptides having the amino acid sequence of Figure 2 [SEQ ID NO:20] or of the deposit, without substitutions.
Further preferred embodiments of the invention are polynucleotides that are at least 70% identical to a polynucleotide encoding the cathepsin K polypeptide having the amino acid sequence set out in Figure 2 [SEQ ID NO:20], or variants, close homologs, derivatives and analogs thereof, as described above, and polynucleotides which are complementary to such polynucleotides. Alternatively, most highly preferred are polynucleotides that comprise a region that is at least 80% identical to a polynucleotide encoding the cathepsin K polypeptide of the gDNA of the deposited clone and polynucleotides complementary thereto. In this regard, polynucleotides at least 90% identical to the same are particularly preferred, and among these particularly preferred polynucleotides, those with at least 95% are especially preferred. Furthermore, those with at least 97% are highly preferred among those with at least 95%, and among these those with at least 98% and at least 99% are particularly highly preferred, with at least 99% being the more preferred. Still further preferred embodiments of the invention are polynucleotides comprising cathepsin K intron polynucleotide sequences, particularly polynucleotides comprising intron 1 [SEQ ID NO: 4], 2 [SEQ ID NO: 6], 3 [SEQ ID NO: 8], 4 [SEQ ID NO: 10], 5 [SEQ ID NO: 12], 6 [SEQ ID NO: 14] or 7[SEQ ID NO: 16], having the intron polynucleotide sequence set out in Figures 1 [SEQ ID NO: 1] and 3 [SEQ ID NO: 2-19], or variants, close homologs, derivatives and analogs thereof, as described above, and polynucleotides which are complementary to such polynucleotides. Other preferred embodiments of the invention are polynucleotides comprising cathepsin K intron 1 [SEQ ID NO: 4], 2 [SEQ ID NO: 6], 3 [SEQ ID NO: 8], 4 [SEQ ID NO: 10], 5 [SEQ ID NO: 12], 6 [SEQ ID NO: 14] or 7[SEQ ID NO: 16], operatively linked to the exon of a gene other than cathepsin K, or joining a cathepsin K exon and an exon of another gene.
Still other preferred embodiments of the invention are polynucleotides comprising cathepsin K exon polynucleotide sequences, particularly polynucleotides comprising exon 1 [SEQ ID NO: 3], 2 [SEQ ID NO: 5], 3 [SEQ ID NO: 7], 4 [SEQ ID NO: 9], 5 [SEQ ID NO: 1 1], 6 [SEQ ID NO: 13], 7 [SEQ ID NO: 15] or 8 [SEQ ID NO: 17], having the exon polynucleotide sequence set out in Figures 1 [SEQ ID NO: 1] and 3 [SEQ ID NO: 2-19], or variants, close homologs, derivatives and analogs thereof, as described above, and polynucleotides which are complementary to such polynucleotides. Other preferred embodiments of the invention are polynucleotides comprising cathepsin K exon 1 [SEQ ID NO: 3], 2 [SEQ ID NO: 5], 3 [SEQ ID NO: 7], 4 [SEQ ID NO: 9], 5 [SEQ ID NO: 1 1], 6 [SEQ ID NO: 13], 7 [SEQ ID NO: 15] or 8 [SEQ ID NO: 17], operatively linked to the intron of a gene other than cathepsin K. More preferred embodiments of the invention are differentially spliced polynucleotides, particularly those comprising any one or more of the following exon-exon pairs: 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 2-4, 2-5, 2-6, 2-7, 2-8, 3-4, 3-5, 3-6, 3-7, 3-8, 4-5, 4-6, 4-7, 4-8, 5-7, 5-8, or 6-8. Particularly preferred embodiments of the invention are differentially spliced polynucleotides which encode polypeptides which function in cells, especially those which have a biological activity of cathepsin K, most especially those expressed in human cells. Polynucleotides comprising exon-exon pairs may be a naturally occurring variant such as a naturally occurring splice variant, or it may be a variant that is not known to occur naturally. Such non-naturally occurring variants of the polynucleotide may be made by mutagenesis techniques, including those applied to polynucleotides, cells or organisms. Such non-naturally occurring variants of the polynucleotide may be made by modifying splice acceptor, donor and/or branch sites, or by expressing the gDNA in cells where it is not naturally expressed, or cell extracts made from such cells. Exon-exon pairs can be full, fused exons or can be fused fragments of exons with a splice junction present. Preferred exon-exon pairs comprising exon fragments may be made from at least two exons, one of which comprises an operable splice donor site and the other of which comprises an operable splice acceptor site and which both are operatively linked by an intron.
Particularly preferred embodiments in this respect, moreover, are polynucleotides which encode polypeptides which retain substantially the same biological function or activity as the mature polypeptide encoded by the cDNA of Figure 2 [SEQ ID NO:20] or the gDNA of the deposited clone.
The present invention further relates to polynucleotides that hybridize to the herein above-described sequences. In this regard, the present invention especially relates to polynucleotides which hybridize under stringent conditions to the herein above-described polynucleotides. As herein used, the term "stringent conditions" means hybridization will occur only if there is at least 95% and preferably at least 97% identity between the sequences.
As discussed additionally herein regarding polynucleotide assays of the invention, for instance, polynucleotides of the invention as discussed above, may be used as a hybridization probe for cDNA and genomic DNA to isolate full-length cDNAs and genomic clones encoding cathepsin K and to isolate cDNA and genomic clones of other genes that have a high sequence similarity to the human cathepsin K gene. Such probes generally will comprise at least 15 bases. Preferably, such probes will have at least 30 bases and may have at least 50 bases. Particularly preferred probes will have at least 30 bases and will have 50 bases or less. For example, the coding region of the cathepsin K gene may be isolated by screening using the known DNA sequence to synthesize an oligonudeotide probe. A labeled oligonudeotide having a sequence complementary to that of a gene of the present invention is then used to screen a library of human cDNA, genomic DNA or mRNA to determine which members of the library the probe hybridizes to.
The polynucleotides and polypeptides of the present invention may be employed as research reagents and materials for discovery of treatments and diagnostics to human disease, as further discussed herein relating to polynucleotide assays, inter alia. The polynucleotides may encode a polypeptide which is the mature protein plus additional amino or carboxyl-terminal amino acids, or amino acids interior to the mature polypeptide (when the mature form has more than one polypeptide chain, for instance). Such sequences may play a role in processing of a protein from precursor to a mature form, may facilitate protein trafficking, may prolong or shorten protein half-life or may facilitate manipulation of a protein for assay or production, among other things. As generally is the case in situ, the additional amino acids may be processed away from the mature protein by cellular enzymes.
A precursor protein, having the mature form of the polypeptide fused to one or more prosequences may be an inactive form of the polypeptide. When prosequences are removed such inactive precursors generally are activated. Some or all of the prosequences may be removed before activation. Generally, such precursors are called proproteins.
In sum, a polynucleotide of the present invention may encode a mature protein, a mature protein plus a leader sequence (which may be referred to as a preprotein), a precursor of a mature protein having one or more prosequences which are not the leader sequences of a preprotein, or a preproprotein, which is a precursor to a proprotein, having a leader sequence and one or more prosequences, which generally are removed during processing steps that produce active and mature forms of the polypeptide.
Deposited materials A deposit containing a human cathepsin K gDNA has been deposited with the American Type Culture Collection, as noted above. Also as noted above, the gDNA deposit is referred to herein as "the deposited clone" or as "the gDNA of the deposited clone." The deposited clone was deposited with the American Type Culture
Collection, 12301 Park Lawn Drive, Rockville, Maryland 20852, USA, on April 26, 1996, and assigned ATCC Deposit No. 98035.
The deposited material is a PI cosmid that contains the full length cathepsin K gDNA, referred to as "PlSacB2CatK/P129" upon deposit. The deposit has been made under the terms of the Budapest Treaty on the international recognition of the deposit of micro-organisms for purposes of patent procedure. The strain will be irrevocably and without restriction or condition released to the public upon the issuance of a patent. The deposit is provided merely as convenience to those of skill in the art and is not an admission that a deposit is required for enablement, such as that required under 35 U.S.C. section 112.
The sequence of the polynucleotides contained in the deposited material, as well as the amino acid sequence of the polypeptide encoded thereby, are controlling in the event of any conflict with any description of sequences herein.
A license may be required to make, use or sell the deposited materials, and no such license is hereby granted.
Polypeptides
The present invention further relates to a human cathepsin K polypeptide which has the deduced amino acid sequence of Figure 2 [SEQ ID NO:20], which is encoded by an unspliced or differentially spliced hnRNA or mRNA transcribed from the sequence of Figure 1 [SEQ ID NO: 1], or which has the amino acid sequence encoded by the deposited clone. Also provided are polypetides encoded by the cathepsin K gDNA comprising missense or nonsense mutations, or those polypeptides encoded by unspliced or partially spliced hnRNAs which still comprise at least one intron, particularly those polypeptides which are naturally found in cells, especially human cells. Frameshift mutations have been shown to be associated with disease (Hoi, FA, et al. Journal of Medical Genetics, 1995, 32 (1), 52-56).
Preferred polypeptides provided by the invention are encoded by differentially spliced polynucleotides, particularly those polypeptides encoded by polynucleotides comprising any one or more of the following exon-exon pairs: 1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 2-4, 2-5, 2-6, 2-7, 2-8, 3-4, 3-5, 3-6, 3-7, 3-8, 4-5, 4-6, 4-7, 4-8, 5-7, 5-8, or 6-8. Particularly preferred embodiments of the invention are polypeptides encoded by differentially spliced polynucleotides, which polypeptides function in cells, especially those which have a biological activity of cathepsin K, most especially those expressed in human cells.
Still further preferred embodiments of the invention are polypeptides encoded by polynucleotides comprising exon polynucleotide sequences, particularly polynucleotides comprising cathepsin K exon 1 [SEQ ID NO: 3], 2 [SEQ ID NO: 5], 3 [SEQ ID NO: 7], 4 [SEQ ID NO: 9], 5 [SEQ ID NO: 11], 6 [SEQ ID NO: 13], 7 [SEQ ID NO: 15] or 8 [SEQ ID NO: 17], having the exon polynucleotide sequence set out in Figures 1 [SEQ ID NO: 1] and 3 [SEQ ID NO: 2-19], or variants, close homologs, derivatives and analogs thereof, as described above, and polypeptides encoded by polynucleotides which are complementary to such polynucleotides. Other preferred embodiments of the invention are polypeptides encoded by polynucleotides comprising comprising cathepsin K exon exon 1 [SEQ ID NO: 3], 2 [SEQ ID NO: 5], 3 [SEQ ID NO: 7], 4 [SEQ ID NO: 9], 5 [SEQ ID NO: 1 1], 6 [SEQ ID NO: 13], 7 [SEQ ID NO: 15] or 8 [SEQ ID NO: 17], operatively liked to the intron of a gene other then cathepsin K, or joined to an exon of another gene.
The invention also relates to fragments, analogs and derivatives of these polypeptides. The terms "fragment," "derivative" and "analog" when referring to the polypeptide of Figure 2 [SEQ ID NO:20], a polypeptide encoded by an unspliced or differentially spliced hnRNA or mRNA transcribed from the sequence of Figure 1 [SEQ ID NO: 1], or that encoded by the deposited gDNA, means a polypeptide which retains essentially the same biological function or activity as such polypeptide. Thus, an analog includes a proprotein which can be activated by cleavage of the proprotein portion to produce an active mature polypeptide. The polypeptide of the present invention may be a recombinant polypeptide, a natural polypeptide or a synthetic polypeptide. In certain preferred embodiments it is a recombinant polypeptide.
The fragment, derivative or analog of the polypeptide of Figure 2 [SEQ ID NO:20], or that encoded by an unspliced or differentially spliced hnRNA or mRNA transcribed from the sequence of Figure 1 [SEQ ID NO: 1], or that encoded by the gDNA in the deposited clone may be (i) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the mature polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the mature polypeptide, such as a leader or secretory sequence or a sequence which is employed for purification of the mature polypeptide or a proprotein sequence. Such fragments, derivatives and analogs are deemed to be within the scope of those skilled in the art from the teachings herein.
Among the particularly preferred embodiments of the invention in this regard are polypeptides having the amino acid sequence of cathepsin K set out in Figure 2 [SEQ ID NO:20], variants, analogs, derivatives and fragments thereof, and variants, analogs and derivatives of the fragments. Alternatively, particularly preferred embodiments of the invention in this regard are polypeptides having the amino acid sequence of the cathepsin K of the gDNA in the deposited clone, variants, analogs, derivatives and fragments thereof, and variants, analogs and derivatives of the fragments.
Among preferred variants are those that vary from a reference by conservative amino acid substitutions. Such substitutions are those that substitute a given amino acid in a polypeptide by another amino acid of like characteristics. 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, Tyr.
Further particularly preferred in this regard are variants, analogs, derivatives and fragments, and variants, analogs and derivatives of the fragments, having the amino acid sequence of the cathepsin K polypeptide of Figure 2 [SEQ ID NO: 20] or of the gDNA in the deposited clone, in which several, a few, 5 to 10, 1 to 5, 1 to 3, 2, 1 or no amino acid residues are substituted, deleted or added, in any combination. Especially preferred among these are silent substitutions, additions and deletions, which do not alter the properties and activities of the cathepsin K. Also especially preferred in this regard are conservative substitutions. Most highly preferred are polypeptides having the amino acid sequence of Figure 2 [SEQ ID NO:20] or the deposited clone without substitutions.
The polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to homogeneity.
The polypeptides of the present invention include the polypeptide encoded by at least one of the exons of SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 1 1, SEQ ID NO: 13, SEQ ID NO: 15 or SEQ ID NO: 17, (in particular the mature polypeptide) as well as polypeptides which have at least 70% similarity (preferably at least 70% identity) to the polypeptide encoded by at least one of the exons of SEQ ID NO: SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 1 1, SEQ ID NO: 13, SEQ ID NO: 15 or SEQ ID NO: 17 and more preferably at least 90% similarity (more preferably at least 90% identity) to the polypeptide encoded by at least one of the exons of SEQ ID NO: SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 1 1 , SEQ ID NO: 13, SEQ ID NO: 15 or SEQ ID NO: 17 and still more preferably at least 95% similarity (still more preferably at least 95% identity) to the polypeptide encoded by at least one of the exons of SEQ ID NO: SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 1 1, SEQ ID NO: 13, SEQ ID NO: 15 or SEQ ID NO: 17 and also include portions of such polypeptides with such portion of the polypeptide generally containing at least 30 amino acids and more preferably at least 50 amino acids.
As known in the art "similarity" between two polypeptides is determined by comparing the amino acid sequence and its conserved amino acid substitutes of one polypeptide to the sequence of a second polypeptide.
Fragments or portions of the polypeptides of the present invention may be employed for producing the corresponding full-length polypeptide by peptide synthesis; therefore, the fragments may be employed as intermediates for producing the full-length polypeptides. Fragments or portions of the polynucleotides of the present invention may be used to synthesize full-length polynucleotides of the present invention.
Fragments Also among preferred embodiments of this aspect of the present invention are polypeptides comprising fragments of cathepsin K, most particularly fragments of the cathepsin K having the amino acids encoded by the exons set out in Figure 1 [SEQ ID NO: 1], or having the amino acid encoded by the exon sequence of the cathepsin K of the deposited clone, and exon fragments or variants and derivatives of the cathepsin K of Figure 1 [SEQ ID NO: 1] or of the deposited clone.
In this regard a fragment is a polypeptide having an amino acid sequence that entirely is the same as part but not all of the amino acid sequence of the aforementioned cathepsin K polypeptides and variants or derivatives thereof.
Such fragments may be "free-standing," i.e., not part of or fused to other amino acids or polypeptides, such as, for example, an exon, or they may be comprised within a larger polypeptide of which they form a part or region. When comprised within a larger polypeptide, the presently discussed fragments most preferably form a single continuous region. However, several fragments may be comprised within a single larger polypeptide. For instance, certain preferred embodiments relate to a fragment of a cathepsin K polypeptide of the present comprised within a precursor polypeptide designed for expression in a host and having heterologous pre and pro-polypeptide regions fused to the amino terminus of the cathepsin K fragment and an additional region fused to the carboxyl terminus of the fragment. Therefore, fragments in one aspect of the meaning intended herein, refers to the portion or portions of a fusion polypeptide or fusion protein derived from cathepsin K.
As representative examples of polypeptide fragments of the invention, there may be mentioned those which are encoded by the polynucleotide sequence comprising cathepsin K exon 1, 2, 3, 4, 5, 6, 7 or 8, having the exon or intron 1,2,3,4,5,6 or 7 polynucleotide sequences respectively as set out in Figures 1 [SEQ ID NO: 1] and 3 [SEQ ID NO: 2-19], or variants, close homologs, derivatives and analogs thereof, as described above, and polypeptides encoded by polynucleotides which are complementary to such polynucleotides.
In this context about includes the particularly recited range and ranges larger or smaller by several, a few, 5, 4, 3, 2 or 1 amino acid at either extreme or at both extremes. For instance, about 65-90 amino acids in this context means a polypeptide fragment of 65 plus or minus several, a few, 5, 4, 3, 2 or 1 amino acids to 90 plus or minus several a few, 5, 4, 3, 2 or 1 amino acid residues, i.e., ranges as broad as 65 minus several amino acids to 90 plus several amino acids to as narrow as 65 plus several amino acids to 90 minus several amino acids.
Highly preferred in this regard are the recited ranges plus or minus as many as 5 amino acids at either or at both extremes. Particularly highly preferred are the recited ranges plus or minus as many as 3 amino acids at either or at both the recited extremes. Especially particularly highly preferred are ranges plus or minus 1 amino acid at either or at both extremes or the recited ranges with no additions or deletions. Most highly preferred of all in this regard are fragments encoded by each of the exons of cathepsin K. Among especially preferred fragments of the invention are truncation mutants of cathepsin K. Truncation mutants include cathepsin K polypeptides having the amino acid sequence encoded by the exons of Figure 1 [SEQ ID NO: 1], or of the deposited clone, or of variants or derivatives thereof, except for deletion of a continuous series of residues (that is, a continuous region, part or portion) that includes the amino terminus, or a continuous series of residues that includes the carboxyl terminus or, as in double truncation mutants, deletion of two continuous series of residues, one including the amino terminus and one including the carboxyl terminus. Fragments having the size ranges set out about also are preferred embodiments of truncation fragments, which are especially preferred among fragments generally. Also preferred in this aspect of the invention are fragments characterized by structural or functional attributes of cathepsin K. Preferred embodiments of the invention in this regard include fragments that comprise alpha-helix and alpha-helix forming regions ("alpha-regions"), beta-sheet and beta-sheet-forming regions ("beta-regions"), turn and turn-forming regions ("turn-regions"), coil and coil-forming regions ("coil-regions"), hydrophilic regions, hydrophobic regions, alpha amphipathic regions, beta amphipathic regions, flexible regions, surface-forming regions and high antigenic index regions of cathepsin K. Certain preferred regions include Garnier-Robson alpha-regions, beta-regions, turn-regions and coil-regions, Chou-Fasman alpha-regions, beta-regions and turn-regions, Kyte-Doolittle hydrophilic regions and hydrophilic regions, Eisenberg alpha and beta amphipathic regions, Karplus-Schulz flexible regions, Emini surface-forming regions and Jameson-Wolf high antigenic index regions.
Among highly preferred fragments in this regard are those that comprise regions of cathepsin K that combine several structural features, such as several of the features set out above. In this regard, the exon sequences of Figure 1 [SEQ ID NO: 1], which all are characterized by encoding amino acid compositions highly characteristic of turn-regions, hydrophilic regions, flexible-regions, surface-forming regions, and high antigenic index-regions, are especially highly preferred regions. Such regions may be comprised within a larger polypeptide or may be by themselves a preferred fragment of the present invention, as discussed above. It will be appreciated that the term "about" as used in this paragraph has the meaning set out above regarding fragments in general.
Further preferred regions are those that mediate activities of cathepsin K. Most highly preferred in this regard are fragments that have a chemical, biological, antigenic or other activity of cathepsin K, including those with a similar activity or an improved activity, or with a decreased undesirable activity.
It will be appreciated that the invention also relates to, among others, polynucleotides encoding the aforementioned fragments, polynucleotides that hybridize to polynucleotides encoding the fragments, particularly those that hybridize under stringent conditions, and polynucleotides, such as PCR primers, for amplifying polynucleotides that encode the fragments. In these regards, preferred polynucleotides are those that correspondent to the preferred fragments, as discussed above. Other preferred polynucleotides are genetic elements of cathepsin K, including, but not being limited to, a polyadenylation region, enhancers, a promoter, a cap site introns, exons, and splice sites (references describing these elements include, Darnel, J. et al. Molecular Cell Biology, second edition, W.H. Freeman, New York (1990); Watson, J.D., et al. Molecular Biology of the Gene, Benjamin/Cummings Pub., Menlo Park, CA, (1987)).
Untranslated regions contain many elements important in regulating gene expression. Mutations and markers in these regions have also been associated with disease (Ozawa T, et al., European Journal of Immunogenetics, APR 1995, 22 (2), 163-169). A preferred embodiment of the invention is the 5'UTR, particularly the sequence set forth in Figure 3(A) [SEQUENCE ID NO: 2]. Mutations and markers in the 5' UTR have been associated with disease (Carlock L, et al., Human Genetics, APR 1994, 93 (4), 457-459). A particularly preferred polynucleotide is an enhancer and promoter in the 5' UTR region of the cathepsin K gDNA. Enhancers are often found in the 5' UTR and upregulate gene expression (see Miller et al., Biotechniques 7: 980-990 (1989) for a general reference on promoters). The enhancer of the present invention can be operatively fused to heterologous genes to upregulate gene expression. It is believed that the enhancer promoter will regulate tissue-specific gene expression, being particularly useful to express genes is osteoclast and leukocytes, particularly macrophages cells. A particularly preferred polynucleotide is the enhancer promoter having the sequence set forth in Figure 3(A) [SEQUENCE ID NO: 2]. Transcription factors are often associated with the enhancer and promoter and act to modulate the function of these regions and binding sites for these factors have been described (Faisst, Steffen and Meyer, Silke, Nucleic Acids Research, Vol. 20, No. 1, pp. 3-26, 1991 ; Smale, Stephen T., Transcription: Mechanisms and Regulation, Raven Press, Ltd. pp. 63-81 (1994)). These sites bind such factors as, for example, Spl, Apl, and Ap3 which are involved in transcription initiation
(Faisst, Steffen and Meyer, Silke, Nucleic Acids Research, Vol. 20, No. 1, pp. 3-26, 1991). Preferred canonical binding sites for transcription factors are underlined in Figure 3(S) [SEQUENCE ID NO: 2]. The Pu Box in Figure 3(S) [SEQUENCE ID NO: 2] has been described to be present in a macrophage gene, a cell in which cathepsin K is also found (Zhang, Dong-Er, Mol. and Cell. Biol., Vol. 14, No. 1, pp. 373-381 (1994)). The present invention provides a promoter region that is useful, among other things, for the mediation of tissue-specific expression in osteoclasts and leukocytes, particularly macrophages. A Pu box (AGGAA), present in the enhancer and promoter region has also been observed in a macrophage cell line (THP1). Pu boxes in the sequences in the invention are provided. These Pu boxes are believed to be active in the cathepsin K gene in macrophages. RT-PCR performed in THP1 cells, using cathepsin K sequence as a probe, showed expression. The promoter is particularly useful for the study of the control of cathepsin K gene expression, particularly as a region to be probed to diagnose disease. Vitamin D response elements have been found in the 5'UTR of known genes (Kahlen, Jean-Pierre and Carlberg, Carsten, Biochemical & Biophysical Research Communications, Vol. 202, No. 3, pp. 1366-1372 (1994); Darwish, Hisham and DeLuca, Hector, Critical Reviews in Eukaryotic Gene Expression, 3(2):89-l 16 (1993); Carlberg, Carsten, Eur. J. Biochem. 231, pp. 517-527 (1995); Ohyama, Yoshihiko, J. Biol. Chem., Vol. 269, No. 14, pp. 10545-10550 (1994)). Portions of vitamin D ("vD half sites") responsive elements and calcium ion responsive elements ("Ca half pairs") are present in the 3' UTR sequence as set forth in Figure 3(S) [SEQUENCE ID NO: 2]. Such sites have been described (Katz, Ronald, W., Subauste, Jose, S., and Koenig, Ronald J., J. Biol. Chem., Vol. 270, No. 10, pp. 5238-5242 (1995)). Other half sites present in the sequence of the 5'UTR set forth in Figure 3(A and S) [SEQUENCE ID NO: 2] include osteopontin/parathyroid hormone responsive element, calcitrol response element and osteocalcin half site (see, for example, Juge-Aubry, Cristiana, et al., J. Biol. Chem., Vol. 270, No. 30, pp. 181 17-18122 (1995)). Promoter factor binding sites found in the promoter and enhancer region and provided in the invention are also found in cathepsin K introns. Estrogen response elements are also expected to be present in cathepsin K 5' UTR. Skilled artisans can readily find such elements using the methods provided herein.
A further preferred polynucleotide is a cap site located 49 base pairs upstream of the ATG start codon of the sequence set forth in (Figure 2).
A further preferred embodiment of the invention is the promoter region of cathepsin K (Figure 3(A) and (S) [SEQUENCE ID NO: 2]). Functional promoter region sequences have been described (Corden, J., et al., Science, 209, pp. 1406- 1414 (1990)). A non-canonical promoter region in the sequence of cathepsin K set forth in Figures 3(A) [SEQUENCE ID NO: 2] and (S) [SEQUENCE ID NO: 2] comprises an A-T rich stretch at 19-27 base pairs upstream of the start codon ATG. Mutations in the TATA box region of promoters have been shown to be associated with disease (Peltoketo H, et al., Genomics, 1994, 23 (1 ), 250-252).
The 3' untranslated region of cathepsin K is a preferred polynucleotide of the invention, especially that polynucleotide set forth in Figure 3(Q) [SEQUENCE ID NO: 18], especially that region set forth in Figure 3(R) [SEQUENCE ID NO: 19]. Mutations in the 3' UTR have been associated with disease (Saito A, et al., Journal of the American Society of Nephrology, 1994, 4 (9), 1649-1653; Payne SJ, et al., Human Molecular Genetics, 1994, 3 (2), 390). The polyadenylation region set forth in Figure 3(Q) [SEQUENCE ID NO: 18] is also a preferred polynucleotide of the 3' UTR. The polyadenylation region comprises two copies of the canonical polyadenylation hexanucleotide, AAT AAA. The polyadenylation region can be used, for example, in expression vectors to mediate mRNA 3' end formation (see, for example Gil, A. et al, Nature 312:413-414 (London) (1984)).
Other particularly preferred polynucleotides of the invention are the splice sites, including, but not limited to the splice donors, splice acceptors and the splice branchpoint. Splice junctions formation is essential for the proper creation of an open reading frame (Mount, Stephen, M., Department of Molecular Biophysics and Biochemistry, Yale University, Sterling Hall of Medicine, New Haven, CT, USA, IRL Press Limited, London, pp. 459-472 (1981)). Diseases associated with the improper formation of the splice junction are known. Particularly preferred splice junction polynucleotides are set forth in Figure 4. Introns comprise elements important in gene expression and in the formation of mature mRNA. Mutations and markers in introns have been shown to be associated with diseases (Peral, G. et al., Human Molecular Genetics, APR 1995, 4 (4), 569-574; Chrysogelos, S.A., Nucleic Acids Research, 1993, 21 (24), 5736-5741; Ameis, D., Journal of Lipid Research, 1995, 36 (2), 241-250). The splice junctions have also been shown to be associated with disease (Ameis D, et al., Journal of Lipid Research, FEB 1995, 36 (2), 241-250; Petrini JHJ, et al., Journal of Immunology, 1994, 152 (1), 176-183; Kleiman FE, et al., Human Genetics, 1994, 94 (3), 279-282). Alternative splicing and cryptic splice sites selection also have been shown to be associated with disease (Arakawa H, et al., Human Molecular Genetics, 1994, 3 (4), 565-568; Tieu PT, et al., Human Mutation, 1994, 3 (3), 333-336; Reale MA, et al., Cancer Research, 1994, 54 (16), 4493-4501). Introns may also comprise enhancer elements as part of their sequence.
Preferred embodiments of the invention are the cathepsin K introns, particularly those introns having the sequences set forth in Figure 3 (C, E, G, I, K, M, and O) [SEQUENCE ID NO: 4, 6, 8, 10, 14, and 16]. Polymoφhisms in the introns can serve as markers for disease following linkage analysis. Moreover, genetic analyses described herein can be used to locate mutations in the introns associated with and/or causing disease.
Another preferred embodiment is a cathepsin K intronic enhancer. Intron 3 does not follow consensus splice junction GT/AG rule. This intron/exon boundaries was verified by sequencing of the PI clone and the genomic DNA. GC/AG splice junctions though not common, have been described(Mount, Stephen, M., Department of Molecular Biophysics and Biochemistry, Yale University, Sterling Hall of Medicine, New Haven, CT, USA, IRL Press Limited, London, pp. 459-472 ( 1981 )). Further preferred embodiments of the invention are the cathepsin K exons, particularly those exons having the sequences set forth in Figure 3 (B, D, F, H, J, L, N, and P) [SEQUENCE ID NO: 3, 5, 7, 9, 1 1, 13, 15 and 17 respectively]. Polymoφhisms in the exons can serve as markers for disease following linkage analysis. Moreover, genetic analyses described herein can be used to locate mutations in the exons associated with and/or causing disease.
Polynucleotide fragments of the invention can be used to create ribozymes that inhibit the expression of the cathepsin K gene. General methods for the construction of ribozyme constructs are known in the art (Stram Y, and Molad T, Virus Genes, 1995, 9 (2), 155-159). Skilled artisans can readily adapt these methods using the novel fragments of the invention to create novel ribozyme constructs. Preferred ribozyme constructs comprise sequences which are complementary to the transcribed control elements of the cathepsin K gene, particularly polynucleotides that are complementary to the 5' untranslated region, splice junctions, and 3'untranslated region, especially the polyadenylation region.
The fragments of the invention, particularly regions in the untranslated region, the promoter and introns are useful as diagnostic probes for disease, particularly bone disease, such as osteoporosis, and including, for example, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hypeφarathyroidism, bone degradation, metastatic tumors, rhemuatoid arthritis, osteoarthritis, periodontal disease and degradation of bone implants and bone protheses, particularly dental implants. Moreover, markers for disease can be located in regions of the cathepsin gene, particularly untranslated regions, which are useful with the diagnostic methods of the invention.
Vectors, host cells, expression
The present invention also relates to vectors which include polynucleotides of the present invention, host cells which are genetically engineered with vectors of the invention and the production of polypeptides of the invention by recombinant techniques. Host cells can be genetically engineered to incoφorate polynucleotides and express polypeptides of the present invention. For instance, polynucleotides may be introduced into host cells using well known techniques of infection, transduction, transfection, transvection and transformation. The polynucleotides may be introduced alone or with other polynucleotides. Such other polynucleotides may be introduced independently, co-introduced or introduced joined to the polynucleotides of the invention.
Thus, for instance, polynucleotides of the invention may be transfected into host cells with another, separate, polynucleotide encoding a selectable marker, using standard techniques for co-transfection and selection in, for instance, mammalian cells. In this case the polynucleotides generally will be stably incoφorated into the host cell genome.
Alternatively, the polynucleotides may be joined to a vector containing a selectable marker for propagation in a host. The vector construct may be introduced into host cells by the aforementioned techniques. Generally, a plasmid vector is introduced as DNA in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. Electroporation also may be used to introduce polynucleotides into a host. If the vector is a virus, it may be packaged in vitro or introduced into a packaging cell and the packaged virus may be transduced into cells. A wide variety of techniques suitable for making polynucleotides and for introducing polynucleotides into cells in accordance with this aspect of the invention are well known and routine to those of skill in the art. Such techniques are reviewed at length in Sambrook et al. cited above, which is illustrative of the many laboratory manuals that detail these techniques. In accordance with this aspect of the invention the vector may be, for example, a plasmid vector, a single or double-stranded phage vector, a single or double-stranded RNA or DNA viral vector. Such vectors may be introduced into cells as polynucleotides, preferably DNA, by well known techniques for introducing DNA and RNA into cells. The vectors, in the case of phage and viral vectors also may be and preferably are introduced into cells as packaged or encapsidated virus by well known techniques for infection and transduction. Viral vectors may be replication competent or replication defective. In the latter case viral propagation generally will occur only in complementing host cells.
Preferred among vectors, in certain respects, are those for expression of polynucleotides and polypeptides of the present invention. Generally, such vectors comprise cis-acting control regions effective for expression in a host operatively linked to the polynucleotide to be expressed. Appropriate trans-acting factors either are supplied by the host, supplied by a complementing vector or supplied by the vector itself upon introduction into the host.
In certain preferred embodiments in this regard, the vectors provide for specific expression. Such specific expression may be inducible expression or expression only in certain types of cells or both inducible and cell-specific. Particularly preferred among inducible vectors are vectors that can be induced for expression by environmental factors that are easy to manipulate, such as temperature and nutrient additives. A variety of vectors suitable to this aspect of the invention, including constitutive and inducible expression vectors for use in prokaryotic and eukaryotic hosts, are well known and employed routinely by those of skill in the art.
The engineered host cells can be cultured in conventional nutrient media, which may be modified as appropriate for, inter alia, activating promoters, selecting transformants or amplifying genes. Culture conditions, such as temperature, pH and the like, previously used with the host cell selected for expression generally will be suitable for expression of polypeptides of the present invention as will be apparent to those of skill in the art.
A great variety of expression vectors can be used to express a polypeptide of the invention. Such vectors include chromosomal, episomal and virus-derived vectors e.g., vectors derived from bacterial plasmids, from bacteriophage, from yeast episomes, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, and vectors derived from combinations thereof, such as those derived from plasmid and bacteriophage genetic elements, such as cosmids and phagemids, all may be used for expression in accordance with this aspect of the present invention. Generally, any vector suitable to maintain, propagate or express polynucleotides to express a polypeptide in a host may be used for expression in this regard.
The appropriate DNA sequence may be inserted into the vector by any of a variety of well-known and routine techniques. In general, a DNA sequence for expression is joined to an expression vector by cleaving the DNA sequence and the expression vector with one or more restriction endonucleases and then joining the restriction fragments together using T4 DNA ligase. Procedures for restriction and ligation that can be used to this end are well known and routine to those of skill. Suitable procedures in this regard, and for constructing expression vectors using alternative techniques, which also are well known and routine to those skill, are set forth in great detail in Sambrook et al. cited elsewhere herein.
The DNA sequence in the expression vector is operatively linked to appropriate expression control sequence(s), including, for instance, a promoter to direct mRNA transcription. Representatives of such promoters include the phage lambda PL promoter, the E. coli lac, tφ and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name just a few of the well-known promoters. It will be understood that numerous promoters not mentioned are suitable for use in this aspect of the invention are well known and readily may be employed by those of skill in the manner illustrated by the discussion and the examples herein.
In general, expression constructs will contain sites for transcription initiation and termination, and, in the transcribed region, a ribosome binding site for translation. The coding portion of the mature transcripts expressed by the constructs will include a translation initiating AUG at the beginning and a termination codon appropriately positioned at the end of the polypeptide to be translated.
In addition, the constructs may contain control regions that regulate as well as engender expression. Generally, in accordance with many commonly practiced procedures, such regions will operate by controlling transcription, such as repressor binding sites and enhancers, among others. Vectors for propagation and expression generally will include selectable markers. Such markers also may be suitable for amplification or the vectors may contain additional markers for this puφose. In this regard, the expression vectors preferably contain one or more selectable marker genes to provide a phenotypic trait for selection of transformed host cells. Preferred markers include dihydrofolate reductase or neomycin resistance for eukaryotic cell culture, tetracycline, kanamycin, and ampicillin resistance genes for culturing E. coli and other bacteria. The vector containing the appropriate DNA sequence as described elsewhere herein, as well as an appropriate promoter, and other appropriate control sequences, may be introduced into an appropriate host using a variety of well known techniques suitable to expression therein of a desired polypeptide. Representative examples of appropriate hosts include bacterial cells, such as E. coli, streptomyces and
Salmonella typhimurium cells; fungal cells, such as yeast cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS and Bowes melanoma cells; and plant cells. Hosts for a great variety of expression constructs are well known, and those of skill will be enabled by the present disclosure readily to select a host for expressing a polypeptides in accordance with this aspect of the present invention.
More particularly, the present invention also includes recombinant constructs, such as expression constructs, comprising one or more of the sequences described above. The constructs comprise a vector, such as a plasmid or viral vector, into which such a sequence of the invention has been inserted. The sequence may be inserted in a forward or reverse orientation. In certain preferred embodiments in this regard, the construct further comprises regulatory sequences, including, for example, a promoter, operably linked to the sequence. Large numbers of suitable vectors and promoters are known to those of skill in the art, and there are many commercially available vectors suitable for use in the present invention.
The following vectors, which are commercially available, are provided by way of example. Among vectors preferred for use in bacteria are pQE70, pQE60 and pQE-9, available from Qiagen; pBS vectors, Phagescript vectors, Bluescript vectors, pNH8A, pNH16a, pNH18A, pNH46A, available from Stratagene; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia. Among preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTl and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia. These vectors are listed solely by way of illustration of the many commercially available and well known vectors that are available to those of skill in the art for use in accordance with this aspect of the present invention. It will be appreciated that any other plasmid or vector suitable for, for example, introduction, maintenance, propagation or expression of a polynucleotide or polypeptide of the invention in a host may be used in this aspect of the invention.
Promoter regions can be selected from any desired gene using vectors that contain a reporter transcription unit lacking a promoter region, such as a chloramphenicol acetyl transferase ("CAT") transcription unit, downstream of restriction site or sites for introducing a candidate promoter fragment; i.e., a fragment that may contain a promoter. As is well known, introduction into the vector of a promoter-containing fragment at the restriction site upstream of the cat gene engenders production of CAT activity, which can be detected by standard CAT assays. Vectors suitable to this end are well known and readily available. Two such vectors are pKK232-8 and pCM7. Thus, promoters for expression of polynucleotides of the present invention include not only well known and readily available promoters, but also promoters that readily may be obtained by the foregoing technique, using a reporter gene. A preferred embodiment of the invention are expression vectors comprising cathepsin K promoter sequences that function as a promoter. Such vector constructs may be used for targeted gene expression in cells which utilize the cathepsin K promoter, for example, osteoclasts and macrophages. Any gene of interest can be expressibly linked to the cathepsin K promoter and expressed in such cells which utilize the cathepsin K promoter. In this manner genes which immortalize primary eukaryotic cells, such as, for example, SV40 T- Antigen, may be expressibly linked cathepsin K promoter to immortalize cells, such as, for example, bone cells, including osteoclasts, and macrophages. Certain preferred vectors comprise cathepsin K promoter expressibly linked to a toxin gene, such as for example, ricin, and are useful in methods for the targeted killing of cell populations that utilize the cathepsin K promoter for gene expression. Certain other preferred vectors comprise cathepsin K promoter expressibly linked to a anti-cathepsin K ribozyme or antisense polynucleotide, which are useful in methods for such targeted killing.
Among known bacterial promoters suitable for expression of polynucleotides and polypeptides in accordance with the present invention are the E. coli lad and lacZ promoters, the T3 and T7 promoters, the gpt promoter, the lambda PR, PL promoters and the tφ promoter. Among known eukaryotic promoters suitable in this regard are the CMV immediate early promoter, the HSV thymidine kinase promoter, the early and late SV40 promoters, the promoters of retroviral LTRs, such as those of the Rous sarcoma virus ("RSV"), and metallothionein promoters, such as the mouse metallothionein-I promoter.
Selection of appropriate vectors and promoters for expression in a host cell is a well known procedure and the requisite techniques for expression vector construction, introduction of the vector into the host and expression in the host are routine skills in the art. The present invention also relates to host cells containing the above-described constructs discussed above. The host cell can be a higher eukaryotic cell, such as a mammalian cell or insect cell, or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al. BASIC METHODS IN MOLECULAR BIOLOGY, (1986).
Constructs in host cells can be used in a conventional manner to produce the gene product encoded by the recombinant sequence. Alternatively, the polypeptides of the invention can be synthetically produced by conventional peptide synthesizers.
Mature proteins can be expressed in mammalian cells, yeast, bacteria, or other cells under the control of appropriate promoters. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention. Appropriate cloning and expression vectors for use with prokaryotic and eukaryotic hosts are described by Sambrook et al., MOLECULAR CLONING: A LABORATORY MANUAL, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989).
Generally, recombinant expression vectors for yeast will include origins of replication, a promoter derived from a highly-expressed gene to direct transcription of a downstream structural sequence, and a selectable marker to permit isolation of vector containing cells after exposure to the vector. Among suitable promoters are those derived from the genes that encode glycolytic enzymes such as 3-phosphoglycerate kinase ("PGK"), a-factor, acid phosphatase, and heat shock proteins, among others. Selectable markers include the ampicillin resistance gene of E. coli and the tφl gene of S. cerevisiae.
Transcription of the DNA encoding the polypeptides of the present invention by higher eukaryotes may be increased by inserting an enhancer sequence into the vector. Enhancers are cis-acύng elements of DNA, usually about from 10 to 300 bp that act to increase transcriptional activity of a promoter in a given host cell-type. Examples of enhancers include the SV40 enhancer, which is located on the late side of the replication origin at bp 100 to 270, the cytomegalovirus early promoter enhancer, the polyoma enhancer on the late side of the replication origin, and adenovirus enhancers.
Polynucleotides of the invention, encoding the heterologous structural sequence of a polypeptide of the invention generally will be inserted into the vector using standard techniques so that it is operably linked to the promoter for expression. The polynucleotide will be positioned so that the transcription start site is located appropriately 5' to a ribosome binding site. The ribosome binding site will be 5' to the AUG that initiates translation of the polypeptide to be expressed. Generally, there will be no other open reading frames that begin with an initiation codon, usually AUG, and lie between the ribosome binding site and the initiating AUG. Also, generally, there will be a translation stop codon at the end of the polypeptide and there will be a polyadenylation signal and a transcription termination signal appropriately disposed at the 3' end of the transcribed region. For secretion of the translated protein into the lumen of the endoplasmic reticulum, into the periplasmic space or into the extracellular environment, appropriate secretion signals may be incoφorated into the expressed polypeptide. The signals may be endogenous to the polypeptide or they may be heterologous signals.
The polypeptide may be expressed in a modified form, such as a fusion protein, and may include not only secretion signals but also additional heterologous functional regions. Thus, for instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence in the host cell, during purification or during subsequent handling and storage. Also, a region may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide. The addition of peptide moieties to polypeptides to engender secretion or excretion, to improve stability and to facilitate purification, among others, are familiar and routine techniques in the art.
Suitable prokaryotic hosts for propagation, maintenance or expression of polynucleotides and polypeptides in accordance with the invention include
Escherichia coli, Bacillus subtilis and Salmonella typhimurium. Various species of Pseudomonas, Streptomyces, and Staphylococcus are suitable hosts in this regard. Moreover, many other hosts also known to those of skill may be employed in this regard. As a representative but non-limiting example, useful expression vectors for bacterial use can comprise a selectable marker and bacterial origin of replication derived from commercially available plasmids comprising genetic elements of the well known cloning vector pBR322 (ATCC 37017). Such commercial vectors include, for example, pKK223-3 (Pharmacia Fine Chemicals, Uppsala, Sweden) and GEMl (Promega Biotec, Madison, WI, USA). These pBR322 "backbone" sections are combined with an appropriate promoter and the structural sequence to be expressed.
Following transformation of a suitable host strain and growth of the host strain to an appropriate cell density, where the selected promoter is inducible it is induced by appropriate means (e.g., temperature shift or exposure to chemical inducer) and cells are cultured for an additional period. Cells typically then are harvested by centrifugation, disrupted by physical or chemical means, and the resulting crude extract retained for further purification.
Microbial cells employed in expression of proteins can be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or use of cell lysing agents, such methods are well know to those skilled in the art.
Various mammalian cell culture systems can be employed for expression, as well. Examples of mammalian expression systems include the COS-7 lines of monkey kidney fibroblast, described in Gluzman et al., Cell 23: 175 (1981). Other cell lines capable of expressing a compatible vector include for example, the C 127, 3T3, CHO, HeLa, human kidney 293 and BHK cell lines.
Mammalian expression vectors will comprise an origin of replication, a suitable promoter and enhancer, and also any necessary ribosome binding sites, polyadenylation sites, splice donor and acceptor sites, transcriptional termination sequences, and 5' flanking non-transcribed sequences that are necessary for expression. In certain preferred embodiments in this regard DNA sequences derived from the SV40 splice sites, and the SV40 polyadenylation sites are used for required non-transcribed genetic elements of these types.
The cathepsin K polypeptide can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography ("HPLC") is employed for purification. Well known techniques for refolding protein may be employed to regenerate active conformation when the polypeptide is denatured during isolation and or purification.
Polypeptides of the present invention include naturally purified products, products of chemical synthetic procedures, and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. In addition, polypeptides of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes. Further illustrative aspects and preferred embodiments of the invention
Cathepsin K polynucleotides and polypeptides may be used in accordance with the present invention for a variety of applications, particularly those that make use of the chemical and biological properties cathepsin K. Among these are applications in the detection and treatment of disease, particularly bone disease, such as osteoporosis, and including, for example, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hypeφarathyroidism, bone degradation, metastatic tumors, rhemuatoid arthritis, osteoarthritis, periodontal disease and degradation of bone implants and bone protheses, particularly dental implants. Additional applications relate to diagnosis and to treatment of disorders of cells, tissues and organisms. These aspects of the invention are illustrated further by the following discussion.
Polynucleotide assays
This invention is also related to the use of the cathepsin K exons, introns, promoters and polynucleotides to detect complementary polynucleotides such as, for example, as a diagnostic reagent. Detection of a mutated form of cathepsin K associated with a dysfunction will provide a diagnostic tool that can add or define a diagnosis of a disease or susceptibility to a disease which results from under-expression, over-expression or altered expression of cathepsin K, such as, for example, osteoporosis, periodontal disease, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hypeφarathyroidism, and bone degradation, metastatic tumors, and degradation of bone implants and bone protheses, particularly dental implants.
Individuals carrying mutations in the human cathepsin K gene may be detected at the DNA level by a variety of techniques. Nucleic acids for diagnosis may be obtained from a patient's cells, such as from blood, urine, saliva, tissue biopsy and autopsy material. The genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR prior to analysis (Saiki et al., Nature, 324: 163-166 (1986)). Ligation-mediated amplification may also be used for amplification (Vollach, V., et al., Nucl Acids Res. 22: 2507 (1994). RNA or cDNA may also be used in the same ways. As an example, PCR primers complementary to the nucleic acid encoding cathepsin K can be used to identify and analyze cathepsin K expression and mutations. For example, deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype. Point mutations can be identified by hybridizing amplified DNA to radiolabeled cathepsin K RNA or alternatively, radiolabeled cathepsin K antisense DNA sequences. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melting temperatures.
Sequence differences between a reference gene and genes having mutations also may be revealed by direct DNA sequencing. In addition, cloned DNA segments may be employed as probes to detect specific DNA segments. The sensitivity of such methods can be greatly enhanced by appropriate use of PCR or another amplification method. For example, a sequencing primer is used with double-stranded PCR product or a single-stranded template molecule generated by a modified PCR. The sequence determination is performed by conventional procedures with radiolabeled nudeotide or by automatic sequencing procedures with fluorescent-tags.
Genetic testing based on DNA sequence differences may be achieved by detection of alteration in electrophoretic mobility of DNA fragments in gels, with or without denaturing agents. Small sequence deletions and insertions can be visualized by high resolution gel electrophoresis. DNA fragments of different sequences may be distinguished on denaturing formamide gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures (see, e.g., Myers et al., Science, 230: 1242 ( 1985)). Sequence changes at specific locations also may be revealed by nuclease protection assays, such as RNase and S 1 protection or the chemical cleavage method (e.g., Cotton et al., Proc. Natl. Acad. Sci., USA, 85: 4397-4401 (1985)).
Thus, the detection of a specific DNA sequence may be achieved by methods such as hybridization, RNase protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes, (e.g., restriction fragment length polymoφhisms ("RFLP"), SSCP and Southern blotting of genomic DNA.
In addition to more conventional gel-electrophoresis and DNA sequencing, mutations also can be detected by in situ analysis.
Chromosome assays
The sequences of the present invention are also valuable for chromosome identification. The sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome. Moreover, there is a current need for identifying particular sites on the chromosome. Few chromosome marking reagents based on actual sequence data (repeat polymoφhisms) are presently available for marking chromosomal location. The mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease. In certain preferred embodiments in this regard, the gDNA herein disclosed is used to clone genomic DNA of a cathepsin K gene. This can be accomplished using a variety of well known techniques and libraries, which generally are available commercially. The genomic DNA then is used for in situ chromosome mapping using well known techniques for this puφose. Typically, in accordance with routine procedures for chromosome mapping, some trial and error may be necessary to identify a genomic probe that gives a good in situ hybridization signal.
In some cases, in addition, sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the gDNA. Computer analysis of the 3' untranslated region of the gene is used to rapidly select primers that do not span more than one exon in the genomic DNA complicate the amplification process. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Only those hybrids containing the human gene corresponding to the primer will yield an amplified fragment.
PCR mapping of somatic cell hybrids is a rapid procedure for assigning a particular DNA to a particular chromosome. Using the present invention with the same oligonudeotide primers, sublocalization can be achieved with panels of fragments from specific chromosomes (e.g., radiation hybrid panels) or pools of large genomic clones in an analogous manner. Other mapping strategies that can similarly be used to map to its chromosome include in situ hybridization, prescreening with labeled flow-sorted chromosomes and preselection by hybridization to construct chromosome specific -cDNA libraries.
Fluorescence in situ hybridization ("FISH") of a cDNA clone to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step. This technique can be used with gDNA as short as 50 to as long as 600. For a review of this technique, see Verma et al., HUMAN CHROMOSOMES: A MANUAL OF BASIC TECHNIQUES, Pergamon Press, New York ( 1988).
Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be correlated with genetic map data. Such data are found, for example, in V. McKusick, MENDELIAN INHERITANCE IN MAN, available on line through Johns Hopkins University, Welch Medical Library. The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinheritance of physically adjacent genes).
Next, it is necessary to determine the differences in the cDNA or genomic sequence between affected and unaffected individuals. If a mutation is observed in some or all of the affected individuals but not in any normal individuals, then the mutation is likely to be the causative agent of the disease.
With current resolution of physical mapping and genetic mapping techniques, a gDNA precisely localized to a chromosomal region associated with the disease could be one of between 50 and 500 potential causative genes. (This assumes 1 megabase mapping resolution and one gene per 20 kb). Polypeptide assays
The present invention also relates to a diagnostic assays such as quantitative and diagnostic assays for detecting levels of cathepsin K protein in cells, tissues and bodily fluids, including determination of normal and abnormal levels of polypeptide. Bodily fluids useful in the diagnostic methods of the invention include, for example, synovial fluid, cerebrospinal fluid, urine, serum, gingival fluid and lymph. Thus, for instance, a diagnostic assay in accordance with the invention for detecting over-expression of cathepsin K protein compared to normal control tissue samples may be used to detect the presence of disease, for example, osteoporosis, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hypeφarathyroidism, bone degradation, metastatic tumors, rhemuatoid arthritis, osteoarthritis, periodontal disease and degradation of bone implants and bone protheses, particularly dental implants. Assay techniques that can be used to determine levels of a protein, such as an immunoassay for cathepsin K protein of the present invention, in a sample derived from a host are well-known to those of skill in the art. Such assay methods include radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA assays. Among these ELISAs frequently are preferred. An ELISA assay initially comprises preparing an antibody specific to cathepsin K, preferably a monoclonal antibody. In addition a reporter antibody generally is prepared which binds to the monoclonal antibody. The reporter antibody is attached a detectable reagent such as radioactive, fluorescent or enzymatic reagent, in this example horseradish peroxidase enzyme.
To carry out an ELISA a sample is removed from a host and incubated on a solid support, e.g. a polystyrene dish, that binds the proteins in the sample. Any free protein binding sites on the dish are then covered by incubating with a non-specific protein such as bovine serum albumin. Next, the monoclonal antibody is incubated in the dish during which time the monoclonal antibodies attach to any cathepsin K proteins attached to the polystyrene dish. Unbound monoclonal antibody is washed out with buffer. The reporter antibody linked to horseradish peroxidase is placed in the dish resulting in binding of the reporter antibody to any monoclonal antibody bound to cathepsin K. Unattached reporter antibody is then washed out. Reagents for peroxidase activity, including a colorimetric substrate are then added to the dish. Immobilized peroxidase, linked to cathepsin K through the primary and secondary antibodies, produces a colored reaction product. The amount of color developed in a given time period indicates the amount of cathepsin K protein present in the sample. Quantitative results typically are obtained by reference to a standard curve. A competition assay may be employed wherein antibodies specific to cathepsin K attached to a solid support and labeled cathepsin K and a sample derived from the host are passed over the solid support and the amount of label detected attached to the solid support can be correlated to a quantity of cathepsin K in the sample.
Antibodies
The polypeptides, their fragments or other derivatives, or analogs thereof, or cells expressing them can be used as an immunogen to produce antibodies thereto. These antibodies can be, for example, polyclonal or monoclonal antibodies. The present invention also includes chimeric, single chain, and humanized antibodies, as well as Fab fragments, or the product of a Fab expression library. Various procedures known in the art may be used for the production of such antibodies and fragments. Antibodies generated against the polypeptides corresponding to a sequence of the present invention can be obtained by direct injection of the polypeptides into an animal or by administering the polypeptides to an animal, preferably a nonhuman. The antibody so obtained will then bind the polypeptides itself. In this manner, even a sequence encoding only a fragment of the polypeptides can be used to generate antibodies binding the whole native polypeptides. Such antibodies can then be used to isolate the polypeptide from tissue expressing that polypeptide.
For preparation of monoclonal antibodies, any technique which provides antibodies produced by continuous clonal cell line cultures can be used. Examples include the hybridoma technique (Kohler, G. and Milstein, C, Nature 256: 495-497 (1975), the trioma technique, the human B-cell hybridoma technique (Kozbor et al., Immunology Today 4: 72 (1983) and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al., pg. 77-96 in MONOCLONAL ANTIBODIES AND CANCER THERAPY, Alan R. Liss, Inc. (1985).
Techniques described for the production of single chain antibodies (U.S. Patent No. 4,946,778) can be adapted to produce single chain antibodies to immunogenic polypeptide products of this invention. Also, transgenic mice, or other organisms such as other mammals, may be used to express antibodies, including for example, humanized antibodies to immunogenic polypeptide products of this invention.
Thus, among others, such antibodies can be used to detect and treat diseases caused by or associated with mutant cathepsin K or abnormal cathepsin K levels, such as, osteoporosis, periodontal disease, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hypeφarathyroidism, bone degradation, metastatic tumors, and degradation of bone implants and bone protheses, particularly dental implants. Immunization using polynucleotides of the inventions can be carried out using known methods to produce a cathepsin K-specific immune response.
Clinical Genomics
This invention provides methods to determine drug responsiveness of individuals having or suspected of having a cathepsin K gene mutation or cathepsin K gene expression abnormality, and also provides reagents to carry out such methods. Individuals may be grouped by their responsiveness to a given compound, particularly drugs, used to treat diseases caused by or associated with a mutation of cathepsin K gene or cathepsin K gene expression. Such individuals may be further grouped by detecting different gene mutations or gene expression level variants. In this manner specific gene mutations and gene expression variants can be readily associated with a certain degree of responsiveness to a compound by an individual). Methods and reagents provided herein can be used to group compound responsiveness by detecting cathepsin K gene mutations and cathepsin K gene expression variants. Other methods for grouping individuals by compound responsivess are known to skilled artisans and can be adapted to use the polypetides and polynucleotides of the invention.
The invention also provides algorithms useful in conjunction with a device or embodied in a composition matter which are useful for the diagnosis of diseases caused by or associated with cathepsin K or mutants or variants thereof. Preferred algorithms are provided for disease stratification and staging.
Cathepsin K binding molecules and assays
This invention also provides a method for identification of molecules, such as receptor molecules, that bind cathepsin K or fragments of cathepsin K of the invention. Genes encoding proteins that bind cathepsin K, such as receptor proteins, can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting. Such methods are described in many laboratory manuals such as, for instance, Coligan et al., Current Protocols in Immunology 1(2): Chapter 5 (1991).
For instance, expression cloning may be employed for this puφose. To this end polyadenylated RNA is prepared from a cell responsive to cathepsin K, a cDNA library is created from this RNA, the library is divided into pools and the pools are transfected individually into cells that are not responsive to cathepsin K. The transfected cells then are exposed to labeled cathepsin K. (Cathepsin K can be labeled by a variety of well-known techniques including standard methods of radio-iodination or inclusion of a recognition site for a site-specific protein kinase.) Following exposure, the cells are fixed and binding of cathepsin K, or a molecule which binds to cathepsin K, is determined. These procedures conveniently are carried out on glass slides.
Pools are identified of cDNA that produced cathepsin K-binding cells. Sub-pools are prepared from these positives, transfected into host cells and screened as described above. Using an iterative sub-pooling and re-screening process, one or more single clones that encode the putative binding molecule or substrate, such as cell matrix, bone matrix or a receptor molecule, and the any of the same can be isolated. Alternatively a labeled ligand can be photoaffinity linked to a cell extract, such as a membrane or a membrane extract, prepared from cells that express a molecule that it binds, such as a receptor molecule. Cross-linked material is resolved by polyacrylamide gel electrophoresis ("PAGE") and exposed to X-ray film. The labeled complex containing the ligand-receptor can be excised, resolved into peptide fragments, and subjected to protein microsequencing. The amino acid sequence obtained from microsequencing can be used to design unique or degenerate oligonudeotide probes to screen cDNA libraries to identify genes encoding the putative receptor molecule. Polypeptides of the invention also can be used to assess cathepsin K binding capacity of cathepsin K binding molecules, such as receptor molecules, in cells or in cell-free preparations.
Agonists and antagonists - assays and molecules The invention also provides a method of screening compounds to identify those which enhance or block the action of cathepsin K in or on cells, such as its interaction with cathepsin K-binding molecules such as receptor and enzymatic substrate molecules. An agonist is a compound which increases the natural biological functions of cathepsin K, while antagonists decrease or eliminate such functions.
For example, a cellular compartment, such as a membrane, vacuole, inclusion or a preparation of any thereof, such as a membrane-preparation, may be prepared from a cell that expresses a molecule that binds cathepsin K, such as a molecule of a signaling or regulatory pathway modulated by cathepsin K. The preparation is incubated with labeled cathepsin K in the absence or the presence of a candidate molecule which may be a cathepsin K agonist or antagonist. The ability of the candidate molecule to bind the binding molecule, such as a substrate, is reflected in decreased binding of the labeled ligand. Molecules which bind gratuitously, i.e., without inducing the effects of cathepsin K on binding the cathepsin K binding molecule, are most likely to be good antagonists. Molecules that bind well and elicit effects that are the same as or closely related to cathepsin K are agonists.
Cathepsin K-like effects of potential agonists and antagonists may by measured, for instance, by determining activity of a second messenger system following interaction of the candidate molecule with a cell or appropriate cell preparation, and comparing the effect with that of cathepsin K or molecules that elicit the same effects as cathepsin K. Second messenger systems that may be useful in this regard include but are not limited to AMP guanylate cyclase, ion channel, phosphoinositide hydrolysis second messenger systems, or compounds which signal the binding of a potential agonists and antagonists to cathepsin K or its substrate. Another example of an assay for cathepsin K antagonists is a competitive assay that combines cathepsin K and a potential antagonist with enzymatic substrate or substrate analogs under appropriate conditions for a competitive inhibition assay. Cathepsin K can be labeled, such as by radioactivity, such that the number of cathepsin K molecules bound to a receptor molecule can be determined accurately to assess the effectiveness of the potential antagonist.
Potential antagonists include small organic molecules, peptides, polypeptides and antibodies that bind to a polypeptide of the invention and thereby inhibit or extinguish its activity. Potential antagonists also may be small organic molecules, a peptide, a polypeptide such as a closely related protein or antibody that binds the same sites on a binding molecule, such as a receptor molecule, without inducing cathepsin K-induced activities, thereby preventing the action of cathepsin K by excluding cathepsin K from binding.
Potential antagonists include a small molecule which binds to and occupies the binding site of the polypeptide thereby preventing binding to cellular binding molecules, such as receptor molecules, such that normal biological activity is prevented. Examples of small molecules include but are not limited to small organic molecules, peptides or peptide-like molecules.
Other potential antagonists include antisense molecules. Antisense technology can be used to control gene expression through antisense DNA or RNA or through triple-helix formation. Antisense techniques are discussed, for example, in - Okano, J. Neurochem. 56: 560 (1991); OLIGODEOXYNUCLEOTIDES AS ANTISENSE INHIBITORS OF GENE EXPRESSION, CRC Press, Boca Raton, FL (1988). Triple helix formation is discussed in, for instance Lee et al., Nucleic Acids Research 6: 3073 (1979); Cooney et al., Science 241: 456 (1988); and Dervan et al., Science 251 : 1360 (1991). The methods are based on binding of a polynucleotide to a complementary DNA or RNA. For example, the 5' coding portion of a polynucleotide that encodes the mature polypeptide of the present invention may be used to design an antisense RNA oligonudeotide of from about 10 to 40 base pairs in length. A DNA oligonudeotide is designed to be complementary to a region of the gene involved in transcription thereby preventing transcription and the production of cathepsin K. The antisense RNA oligonudeotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into cathepsin K polypeptide. The oligonudeotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of cathepsin K.
The antagonists may be employed in a composition with a pharmaceutically acceptable carrier, e.g., as hereinafter described.
The antagonists may be employed for instance to treat diseases caused by or associated with mutant cathepsin K or abnormal cathepsin K levels, such as, osteoporosis, Paget's disease, Gaucher's disease, CNS inflammation, Alzheimer's disease, hypeφarathyroidism, bone degradation, metastatic tumors, rhemuatoid arthritis, osteoarthritis, periodontal disease and degradation of bone implants and bone protheses, particularly dental implants.
Compositions
The invention also relates to compositions comprising the polynucleotides or the polypeptides discussed above or the agonists or antagonists. Thus, the polypeptides of the present invention may be employed in combination with a non-sterile or sterile carrier or carriers for use with cells, tissues or organisms, such as a pharmaceutical carrier suitable for administration to a subject. Such compositions comprise, for instance, a media additive or a therapeutically effective amount of a polypeptide of the invention and a pharmaceutically acceptable carrier or excipient. Such carriers may include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol and combinations thereof. The formulation should suit the mode of administration.
Kits
The invention further relates to pharmaceutical packs and kits comprising one or more containers filled with one or more of the ingredients of the aforementioned compositions of the invention. 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, reflecting approval by the agency of the manufacture, use or sale of the product for human administration.
Administration
Polypeptides and other compounds of the present invention may be employed alone or in conjunction with other compounds, such as therapeutic compounds.
The pharmaceutical compositions may be administered in any effective, convenient manner including, for instance, administration by topical, oral, anal, vaginal, intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal, intraarticular, or intradermal routes among others.
The pharmaceutical compositions generally are administered in an amount effective for treatment or prophylaxis of a specific indication or indications. In general, the compositions are administered in an amount of at least about 10 mg/kg body weight. Preferably, in most cases, dose is from about 10 mg/kg to about 1 mg/kg body weight, daily. It will be appreciated that optimum dosage will be determined by standard methods for each treatment modality and indication, taking into account the indication, its severity, route of administration, complicating conditions and the like. Gene therapy
The cathepsin K polynucleotides, polypeptides, agonists and antagonists that are polypeptides may be employed in accordance with the present invention by expression of such polypeptides in vivo, in treatment modalities often referred to as "gene therapy."
Thus, for example, cells from a patient may be engineered with a polynucleotide, such as a DNA or RNA, encoding a polypeptide ex vivo, and the engineered cells then can be provided to a patient to be treated with the polypeptide. For example, cells may be engineered ex vivo by the use of a retroviral plasmid vector containing RNA encoding a polypeptide of the present invention. Such methods are well-known in the art and their use in the present invention will be apparent from the teachings herein.
Cells from a patient may also be engineered with a polynucleotide, such as a ribozyme that has been constructed, using well known methods, to inhibit the gene expression of Cathepsin K. Other constructs may also be engineered into a patient's cells to contains antisense stretches of cathepsin K sequence, using well known methods. Such antisense constructs will inhibit Cathepsin K expression in the patient.
Similarly, cells may be engineered in vivo for expression of a polypeptide in vivo by procedures known in the art. For example, a polynucleotide of the invention may be engineered for expression in a replication defective retroviral vector, as discussed above. The retroviral expression construct then may be isolated and introduced into a packaging cell that is transduced with a retroviral plasmid vector containing RNA encoding a polypeptide of the present invention such that the packaging cell now produces infectious viral particles containing the gene of interest. These producer cells may be administered to a patient for engineering cells in vivo and expression of the polypeptide in vivo. These and other methods for administering a polypeptide of the present invention by such method should be apparent to those skilled in the art from the teachings of the present invention. Retroviruses from which the retroviral plasmid vectors herein above mentioned may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, retroviruses such as Rous Sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, adenovirus, Myeloproliferative Sarcoma Virus, and mammary tumor virus. In one embodiment, the retroviral plasmid vector is derived from Moloney Murine Leukemia Virus.
Such vectors well include one or more promoters for expressing the polypeptide. Suitable promoters which may be employed include, but are not limited to, cathepsin K promoter, a retroviral LTR, an SV40 promoter, and the human cytomegalovirus (CMV) promoter described in Miller et al., Biotechniques 7: 980-990 (1989), or any other promoter (e.g., cellular promoters such as eukaryotic cellular promoters including, but not limited to, the histone, RNA polymerase III, and alpha-actin promoters). Other viral promoters which may be employed include, but are not limited to, adenovirus promoters, thymidine kinase (TK) promoters, and B19 parvovirus promoters. The selection of a suitable promoter will be apparent to those skilled in the art from the teachings contained herein.
The nucleic acid sequence encoding the polypeptide of the present invention will be placed under the control of a suitable promoter. Suitable promoters which may be employed include, but are not limited to, adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the rous sarcoma virus (RSV) promoter; inducible promoters, such as the MMT promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAI promoter; human globin promoters; viral thymidine kinase promoters, such as the Heφes Simplex thymidine kinase promoter; retroviral LTRs (including the modified retroviral LTRs herein above described); the alpha-actin promoter; and human growth hormone promoters. The promoter also may be the native promoter which controls the gene encoding the polypeptide.
The retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines. Examples of packaging cells which may be transfected include, but are not limited to, the PE501, PA317, Y-2, Y-AM, PA12, T19-14X, VT-19-17-H2, YCRE, YCRIP, GP+E-86, GP+envAml2, and DAN cell lines as described in Miller, A., Human Gene Therapy 1: 5-14 (1990). The vector may be transduced into the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaPO4 precipitation. In one alternative, the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host. The producer cell line will generate infectious retroviral vector particles, which include the nucleic acid sequence(s) encoding the polypeptides. Such retroviral vector particles then may be employed to transduce eukaryotic cells, either in vitro or in vivo. The transduced eukaryotic cells will express the nucleic acid sequence(s) encoding the polypeptide. Eukaryotic cells which may be transduced include, but are not limited to, embryonic stem cells, embryonic carcinoma cells, as well as hematopoietic stem cells, hepatocytes, fibroblasts, myoblasts, keratinocytes, endothelial cells, and bronchial epithelial cells.
EXAMPLES
The present invention is further described by the following examples. The examples are provided solely to illustrate the invention by reference to specific embodiments. These exemplifications, while illustrating certain specific aspects of the invention, do not portray the limitations or circumscribe the scope of the disclosed invention.
Certain terms used herein are explained in the foregoing glossary. An N used herein in a nudeotide sequence refers to an unknown nudeotide or nucleotides. All examples were or may be carried out using standard techniques, which are well known and routine to those of skill in the art, except where otherwise described in detail. Routine molecular biology techniques of the following examples can be carried out as described in standard laboratory manuals, such as Sambrook et al., MOLECULAR CLONING: A LABORATORY MANUAL, 2nd Ed.; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989), herein referred to as "Sambrook." All parts or amounts set out in the following examples are by weight, unless otherwise specified.
Unless otherwise stated size separation of fragments in the examples below was carried out using standard techniques of agarose and polyacrylamide gel electrophoresis ("PAGE") in Sambrook and numerous other references such as, for instance, by Goeddel et al., Nucleic Acids Res. 8: 4057 (1980).
Unless described otherwise, ligations were accomplished using standard buffers, incubation temperatures and times, approximately equimolar amounts of the DNA fragments to be ligated and approximately 10 units of T4 DNA ligase ("ligase") per 0.5 μg of DNA.
Example 1 Isolation and sequencing of human cathepsin K genomic clone
cDNA as disclosed in U.S. Patent Number 5,501,969, was used to isolate the gDNA clone from a gDNA library (Clontech) according to the following method. Primers to adjacent exons (6 of the 7 exons) were prepared. The sequence of these primers is underlined in Figure 2. PCR was performed using standard methods well known in the art. Amplified fragments were cloned into a TA vector (Clontech) and the clones were sequenced by an automated sequencer (Applied BioSystems Model 373) by established methods well known in the art using forward and reverse sequencing primers. The sequence of all internal introns were obtained. 5' and 3' terminal intron sequences were obtained as follows. 5' end primers were designed to obtain sequence for the first intron (see underlined primer in Figure 2), using these primers 2 PI clones were obtained (Genome Systems Inc.). Both clones were full length. PCR was used to confirm the sequence of internal intron-exon boundary junctions (see Example 2). Primers derived from sequence at the 5' end of the PI clones was used to "walk" and sequence along the clone, in a stepwise fashion, using new primers at each sequence step, by routine methods known in the art. Purification of PI clones was carried out as set forth in Example 1(d). "Walking" and sequencing was performed in both directions to confirm cathepsin K gDNA sequence. PCR was again performed using proofreading Taq polymerase (PCR Ultima, Perkin Elmer).
A transcription start site was obtained using a 5' RACE kit (Gibco BRL) and the protocol supplied therewith. This site was also confirmed using an RNASe protection assay kit (Hybspeed, RPA Ambion). Example 1 (a)-(d) provide further specifics concerning cloning and sequencing of cathepsin K
(a) DNA sequencing of intron-exon boundaries Intron-Exon Boundaries
Intron 1
Intron 1 was identified by utilization of 5' RACE (Gibco BRL) technique to determine 5' UTR sequence from which primer could be designed to PCR from exon 1 to exon 2. (intron 1 starts prior to ATG so PCR may not be readily employed based on cDNA sequence available.) Intron 1 was amplified by PCR on human genomic DNA (Clontech) and cloned into PCRII vector and sequenced as described in Example 1.
Intron 2 Intron 2 was identified by PCR on human genomic DNA from primers designed in exon 2 to exon 3. PCR product was cloned and sequenced using standard methods.
Intron 3 Intron 3 was identified by PCR on human genomic DNA from primers designed in exon 3 to exon 4. PCR product was cloned and sequenced using standard methods. Intron 4
Intron 4 was identified by PCR on human genomic DNA from primers designed in exon 4 to exon 5. PCR product was cloned and sequenced using standard methods.
Introns 5 & 6
Introns 5 and 6 were identified by PCR on human genomic DNA from primers designed in exon 5 to exon 7. PCR product was cloned and sequenced using standard methods confirming presence of both introns.
Intron 7
Intron 7 was identified by PCR on human genomic DNA from primers designed in exon 7 to exon 8. PCR product was cloned and sequenced using standard methods.
All introns that were identified by PCR on human genomic DNA were confirmed by PCR of the same regions on PI clone A (see (b) below) clone (Genome Systems, Inc.)
(b) DNA sequencing of 5' and 3' untranslated region (UTR)
5' and 3' untranslated regions were isolated from a single PI clone (Genome Systems Inc.). This PI clone has been identified herein as "PI clone A." Sequence was obtained by direct sequence walking up and down the PI clone with gene specific primers derived from confirmed cDNA sequence using standard methods. These regions were then cloned via PCR and confirmed by sequence analysis using standard methods. The 5' UTR was additionally amplified by PCR using proofreading Taq Polymerase Ultima in accordance with manufacturer instructions and cloned to eliminate sequence ambiguities. 5' and 3' UTR were further confirmed by PCR on human genomic DNA using standard methods. (c) DNA sequencing of mRNA Cap Site & size of exon 1
An mRNA Cap Site was determined to be about 48 bp upstream of the start codon based on 5' RACE sequencing. Ribonuclease Protection Assay confirmed a protected fragment of about 48 bp in size indicating that the start site from transcription resides about 48 bp upstream of ATG (start codon). Putative transcription factors have been identified by analysis of sequence with database transcription factor sequence information and these are set forth in Figure 3(S) [SEQUENCE ID NO: 2]. The 1.1 kb 5' UTR fragment was cloned into pCAT expression vectors to further analyze the promoter sequence region.
(d) PI DNA preparation
PI clone A colonies were streaked out on Kanamycin LB plates. A single colony was picked and grown O/N in 20 mis with 25 μg/ml of kanamycin. 500 mis of media (25 μg/ml kanamycin) was inoculated with 16 mis of the O/N culture and grown for 10 hours. Cells were pelleted by centrifugation and resuspended in 10 mis of Qiagen PI Solution. 10 mis of Qiagen P2 Solution was added and incubated at room temp, for 5 min. 10 mis of Qiagen P3 Solution was added and the mixture left on ice for 15 min. The sample was spun at 10,000g for 15 min. The supernatant was removed and extracted with phenol. The supernatant was then re-extracted with chloroform. The DNA was precipitated following addition of NaOAc pH 5.2 and 1.1 volumes of isopropanol. The DNA was pelleted by centrifugation for 15 min. at 10,000g and washed with 70% ethanol. To clean up the DNA for sequencing, 250 μl of DNA (about 50 μg) was added to 65 μl 30% PEG in 1.5M NaCI. 8.5 μl of 3M NaCI was added and the mixture incubated on ice for 30 min. The sample was spun at 12,000g for 10 min. The supernatant was discarded and the pellet dissolved in 200 μl distilled water. The DNA was then extracted with chloroform, vortexed and spun at 10,000g for 1 min. The aqueous layer was removed and the DNA precipitated with 40 μl of NaOAc pH 5.2 and 1 ml of ethanol. The sample was spun at 12,000g for 30 min. The DNA was washed with 1 ml of 70% ethanol and resuspended on distilled water. Prior to sequencing, the DNA was denatured with 0.1 volumes of 2M NaOH and 2mM EDTA and incubated at 37°C for 30 min. The mixture was neutralized with 0.1 volume of 3M NaOAc pH 5.2 and precipitated with 2.5 volumes of ethanol. The denatured DNA was resuspended in distilled water at a concentration of 1 μg/μl 6 μg/μl were used in each sequencing reaction (ABI) using TaqFS.
Example 2 Chromosomal mapping of cathepsin K
Purified PI DNA was used for FISH analysis (Genome Systems, Inc.) to map to specific chromosome. Prior results done showed by use of 2 PCR somatic cell hybrid panels that the gene mapped to Chromosome 1. FISH analysis confirmed mapping to chromosome 1 and also further mapped the gene to lq21. This is the same locus as is known for cathepsin-S.
Example 3 Alternative sequencing method
The DNA sequence encoding human cathepsin K in the deposited polynucleotide is amplified using PCR oligonudeotide primers specific to the amino acid carboxyl terminal sequence of the human cathepsin K protein and to vector sequences 3' to the gene. Additional nucleotides containing restriction sites to facilitate cloning are added to the 5' and 3' sequences respectively.
The 5' and 3' oligonudeotide primers are designed with sequences capable of mediating amplification by PCR. The 3' primer has sequences complementary to a portion of the nucleotides of the cathepsin K coding sequence set out in Figure 2, including the stop codon. The restriction sites are compatible with restriction enzyme sites in the bacterial expression vector pQE-70, for example, which is used for bacterial expression in certain of the Examples. (Qiagen, Inc. 9259 Eton Avenue, Chatsworth, CA, 9131 1). pQE-70 encodes ampicillin antibiotic resistance ("Ampr") and contains a bacterial origin of replication ("ori"), an IPTG inducible promoter, a ribosome binding site ("RBS"), a 6-His tag and restriction enzyme sites.
The amplified human cathepsin K DNA and the vector pQE-70 both are digested with appropriate restriction enzymes to allow ligation with the restriction digested vector, and the digested DNAs then are ligated together. Insertion of the cathepsin K DNA into the restricted vector places the cathepsin K coding region downstream of and operably linked to the vector's IPTG-inducible promoter and in-frame with an initiating AUG appropriately positioned for translation of cathepsin K. The ligation mixture is transformed into competent E. coli cells using standard procedures. Such procedures are described in Sambrook et al., MOLECULAR CLONING: A LABORATORY MANUAL, 2nd Ed.; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y. (1989). E. coli strain M15/rep4, containing multiple copies of the plasmid pREP4, which expresses lac repressor and confers kanamycin resistance ("Kanr"), is used in carrying out the illustrative example described here. This strain, which is only one of many that are suitable for expressing cathepsin K, is available commercially from Qiagen.
Transformants are identified by their ability to grow on LB plates in the presence of ampicillin (demonstrating Amp1"). Plasmid DNA is isolated from resistant colonies and the identity of the cloned DNA are confirmed by restriction analysis.
Clones containing the desired constructs are grown overnight ("O/N") in liquid culture in LB media supplemented with both ampicillin ( 100 ug/ml) and kanamycin (25 ug/ml). The O/N culture is used to inoculate a large culture, at a dilution of approximately 1:100 to 1 :250. The cells are grown to an optical density at 600nm ("OD600") of between 0.4 and 0.6. Isopropyl-B-D-thiogalactopyranoside ("IPTG") is then added to a final concentration of 1 mM to induce transcription from lac repressor sensitive promoters, by inactivating the lad repressor. Cells subsequently are incubated further for 3 to 4 hours. Cells then are harvested by centrifugation and disrupted, by standard methods. Inclusion bodies are purified from the disrupted cells using routine collection techniques, and protein is solubilized from the inclusion bodies into 8M urea. The 8M urea solution containing the solubilized protein is passed over a PD-10 column in 2X phosphate buffered saline ("PBS"), thereby removing the urea, exchanging the buffer and refolding the protein. The protein is purified by a further step of chromatography to remove endotoxin. Then, it is sterile filtered. The sterile filtered protein preparation is stored in 2X PBS at a concentration of 95 micrograms per ml.
Analysis of the preparation by standard methods of polyacrylamide gel electrophoresis is performed to determine the percent monomeric cathepsin K in the sample.
Example 4 Cloning and expression of human cathepsin K in a baculovirus expression system
The gDNA sequence encoding the full length human cathepsin K protein is amplified using PCR oligonudeotide primers corresponding to the 5' and 3' sequences of the gene:
The 5' and 3' primers are provided with sequences capable of mediating PCR amplification followed by a stretch of about 20 bases of the sequence of cathepsin K of Figure 2. Inserted into an expression vector, as described below, the 5' end of the amplified fragment encoding human cathepsin K provides an efficient signal peptide. An efficient signal for initiation of translation in eukaryotic cells, as described by Kozak, M., J. Mol. Biol. 196: 947-950 (1987) is appropriately located in the vector portion of the construct. The amplified fragment is isolated from a 1 % agarose gel using a commercially available kit ("Geneclean," BIO 101 Inc., La Jolla, Ca.). The fragment then is digested with BamHl and Asp718 and again is purified on a 1% agarose gel. This fragment is designated herein F2.
Any of the many expression vectors known in the art for baculovirus expression can be used to express the cathepsin K protein in the baculovirus expression system, using standard methods, such as those described in Summers et al, A MANUAL OF METHODS FOR BACULOVIRUS VECTORS AND INSECT CELL CULTURE PROCEDURES, Texas Agricultural Experimental Station Bulletin No. 1555 (1987). A preferred expression vector contains the strong polyhedrin promoter of the Autographa californica nuclear polyhedrosis virus (AcMNPV) followed by convenient restriction sites. The signal peptide of
AcMNPV gp67, including the N-terminal methionine, is located just upstream of a BamHl site. The polyadenylation site of the simian virus 40 ("SV40") is used for efficient polyadenylation. For an easy selection of recombinant virus the beta-galactosidase gene from E.coli is inserted in the same orientation as the polyhedrin promoter and is followed by the polyadenylation signal of the polyhedrin gene. The polyhedrin sequences are flanked at both sides by viral sequences for cell-mediated homologous recombination with wild-type viral DNA to generate viable virus that express the cloned polynucleotide.
Many other baculovirus vectors could be used in place of pA2-GP, such as pAc373, pVL941 and pAcIMl provided, as those of skill readily will appreciate, that construction provides appropriately located signals for transcription, translation, trafficking and the like, such as an in-frame AUG and a signal peptide, as required. Such vectors are described in Luckow et al., Virology 170: 31-39, among others. The plasmid is digested with the appropriate restriction enzymes, as can readily be determined by the skilled artisan, to remove the insert as a single fragment and the insert fragment is then dephosphorylated using calf intestinal phosphatase, using routine procedures known in the art. The DNA is then isolated from a 1% agarose gel using a commercially available kit ("Geneclean" BIO 101 Inc., La Jolla, Ca.). This vector DNA is designated herein "V2". Fragment F2 and the dephosphorylated plasmid V2 are ligated together with
T4 DNA ligase. E.coli HB 101 cells are transformed with ligation mix and spread on culture plates. Bacteria are identified that contain the plasmid with the human cathepsin K gene by digesting DNA from individual colonies using with the appropriate restriction nucleases, as can readily be determined by the skilled artisan, and then analyzing the digestion product by gel electrophoresis. The sequence of the cloned fragment is confirmed by DNA sequencing. This plasmid is designated herein pBaccathepsin K.
5 mg of the plasmid pBaccathepsin K is co-transfected with 1.0 mg of a commercially available linearized baculovirus DNA ("BaculoGold™ baculovirus DNA", Pharmingen, San Diego, CA.), using the lipofection method described by Feigner et al., Proc. Natl. Acad. Sci. USA 84: 7413-7417 (1987). lmg of BaculoGold™ virus DNA and 5 mg of the plasmid pBaccathepsin K are mixed in a sterile well of a microtiter plate containing 50 ml of serum free Grace's medium (Life Technologies Inc., Gaithersburg, MD). Afterwards 10 ml Lipofectin plus 90 ml Grace's medium are added, mixed and incubated for 15 minutes at room temperature. Then the transfection mixture is added drop-wise to Sf9 insect cells (ATCC CRL 1711) seeded in a 35 mm tissue culture plate with 1 ml Grace's medium without serum. The plate is rocked back and forth to mix the newly added solution. The plate is then incubated for 5 hours at 27°C. After 5 hours the transfection solution is removed from the plate and 1 ml of Grace's insect medium supplemented with 10% fetal calf serum is added. The plate is put back into an incubator and cultivation is continued at 27°C for four days.
After four days the supernatant is collected and a plaque assay is performed, as described by Summers and Smith, cited above. An agarose gel with "Blue Gal" (Life Technologies Inc., Gaithersburg) is used to allow easy identification and isolation of β gal-expressing clones, which produce blue-stained plaques. (A detailed description of a "plaque assay" of this type can also be found in the user's guide for insect cell culture and baculovirology distributed by Life Technologies Inc., Gaithersburg, page 9-10). Four days after serial dilution, the virus is added to the cells. After appropriate incubation, blue stained plaques are picked with the tip of an Eppendorf pipette. The agar containing the recombinant viruses is then resuspended in an Eppendorf tube containing 200 ml of Grace's medium. The agar is removed by a brief centrifugation and the supernatant containing the recombinant baculovirus is used to infect Sf9 cells seeded in 35 mm dishes. Four days later the supernatants of these culture dishes are harvested and then they are stored at 4°C. A clone containing properly inserted cathepsin K is identified by DNA analysis including restriction mapping and sequencing. This is designated herein as V-cathepsin K.
Sf9 cells are grown in Grace's medium supplemented with 10% heat-inactivated FBS. The cells are infected with the recombinant baculovirus V-cathepsin K at a multiplicity of infection ("MOI") of about 2 (about 1 to about 3). Six hours later the medium is removed and is replaced with SF900 II medium minus methionine and cysteine (available from Life Technologies Inc., Gaithersburg). 42 hours later, 5 mCi of 35S-methionine and 5 mCi 35S cysteine (available from Amersham) are added. The cells are further incubated for 16 hours and then they are harvested by centrifugation, lysed and the labeled proteins are visualized by SDS-PAGE and autoradiography.
Example 5 Expression of cathepsin K in COS cells (a) CAT Assays pCAT-CatK, which contains the 1100 bp putative CatK promoter, upstream of the CAT reporter gene was transfected into COS cells by the DEAE-dextran procedure. Transfections were done on COS cells in 100mm dishes and 5μg of DNA was used. As controls, pCAT basic, which contains no promoter or enhancer, and pCAT control, which contains the SV40 promoter and enhancer, were also transferred separately. 72 hours after transfection, extracts were made by freeze-thaw and equal amounts of extract protein were used in both 1-hour and overnight CAT assays. No activity was detected in untransfected COS cells. pCAT-CatK showed a 1.4-1.6 fold increase of CAT expression relative to pCAT basic after subtraction of background from untransfected cells. Since it is possible that higher levels of activation may be obtained in the presence of various inducers, activation of the CatK promoter by adding exogenous 1 ,25 di-hydroxy vitamin D3 is believed to occur. Vitamin D has been shown by others to activate transcription of osteocalcin, osteopontin, calcitonin and P450 promoters through interaction with the vitamin D receptor and the vitamin D response element(s) found in these various promoters. The ability of vitamin D to transactivate these promoters is believed thought to play a role in the control of bone formation and resoφtion. Similar experiments can be performed to assess estrogen responsiveness which is also believed thought to play a role in the control of bone formation and resoφtion. (b) Expression Vector
The expression plasmid, cathepsin K HA, is made by cloning a gDNA encoding cathepsin K into the expression vector pcDNAI/Amp (which can be obtained from Invitrogen, Inc.). The expression vector pcDNAI/amp contains: (1) an E.coli origin of replication effective for propagation in E. coli and other prokaryotic cell; (2) an ampicillin resistance gene for selection of plasmid-containing prokaryotic cells; (3) an SV40 origin of replication for propagation in eukaryotic cells; (4) a CMV promoter, a polylinker, an SV40 intron, and a polyadenylation signal arranged so that a gDNA conveniently can be placed under expression control of the CMV promoter and operably linked to the SV40 intron and the polyadenylation signal by means of restriction sites in the polylinker.
A DNA fragment encoding the entire cathepsin K precursor and a HA tag fused in frame to its 3' end is cloned into the polylinker region of the vector so that recombinant protein expression is directed by the CMV promoter. The HA tag corresponds to an epitope derived from the influenza hemagglutinin protein described by Wilson et al., Cell 37: 767 (1984). The fusion of the HA tag to the target protein allows easy detection of the recombinant protein with an antibody that recognizes the HA epitope. The plasmid construction strategy is as follows.
The cathepsin K gDNA of the deposit clone is amplified using primers that contained convenient restriction sites, much as described above regarding the construction of expression vectors for expression of cathepsin K in E. coli and S. furgiperda. To facilitate detection, purification and characterization of the expressed cathepsin K, one of the primers contains a heamaglutinin tag ("HA tag") as described above.
Suitable primers can readily be made by skilled artisans using known methods. The PCR amplified DNA fragment and the vector, pcDNAI/Amp, are digested with restriction endonuclease and then ligated. The ligation mixture is transformed into E. coli strain SURE (available from Stratagene Cloning Systems, 11099 North Torrey Pines Road, La Jolla, CA 92037) the transformed culture is plated on ampicillin media plates which then are incubated to allow growth of ampicillin resistant colonies. Plasmid DNA is isolated from resistant colonies and examined by restriction analysis and gel sizing for the presence of the cathepsin K-encoding fragment.
For expression of recombinant cathepsin K, COS cells are transfected with an expression vector, as described above, using DEAE-DEXTRAN, as described, for instance, in Sambrook et al., MOLECULAR CLONING: A LABORATORY MANUAL, Cold Spring Laboratory Press, Cold Spring Harbor, New York (1989). Cells are incubated under conditions for expression of cathepsin K by the vector.
Expression of the cathepsin K HA fusion protein is detected by radiolabelling and immunoprecipitation, using methods described in, for example Harlow et al., ANTIBODIES: A LABORATORY MANUAL, 2nd Ed.; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1988). To this end, two days after transfection, the cells are labeled by incubation in media containing 35S-cysteine for 8 hours. The cells and the media are collected, and the cells are washed and then lysed with detergent-containing RIPA buffer: 150 mM NaCI, 1% NP-40, 0.1 % SDS, 0.5% DOC, 50 mM TRIS, pH 7.5, as described by Wilson et al. cited above. Proteins are precipitated from the cell lysate and from the culture media using an HA-specific monoclonal antibody. The precipitated proteins then are analyzed by SDS-PAGE gels and autoradiography. An expression product of the expected size is seen in the cell lysate, which is not seen in negative controls.
Example 6 Tissue distribution of cathepsin K expression
Northern blot analysis is carried out to examine the levels of expression of cathepsin K in human tissues, using methods described by, among others, Sambrook et al, cited above. For Northern blot analysis, total cellular RNA samples are isolated with RNAzol™ B system (Biotecx Laboratories, Inc. 6023 South Loop East, Houston, TX 77033).
About lOmg of Total RNA is isolated from tissue samples. The RNA is size resolved by electrophoresis through a 1% agarose gel under strongly denaturing conditions. RNA is blotted from the gel onto a nylon filter, and the filter then is prepared for hybridization to a detectably labeled polynucleotide probe.
As a probe to detect mRNA that encodes cathepsin K, the antisense strand of the coding region of the gDNA insert in the deposited clone (or cathepsin K cDNA) is labeled to a high specific activity. The gDNA is labeled by primer extension, using the Prime-It kit, available from Stratagene. The reaction is carried out using 50 ng of the gDNA, following the standard reaction protocol as recommended by the supplier. The labeled polynucleotide is purified away from other labeled reaction components by column chromatography using a Select-G-50 column, obtained from 5-Prime - 3-Prime, Inc. of 5603 Arapahoe Road, Boulder, CO 80303. The labeled probe is hybridized to the filter, at a concentration of 1 ,000,000 cpm/ml, in a small volume of 7% SDS, 0.5 M NaPOφ pH 7.4 at 65°C, overnight.
Thereafter the probe solution is drained and the filter is washed twice at room temperature and twice at 60°C with 0.5 x SSC, 0.1% SDS. The filter then is dried and exposed to film at -70°C overnight with an intensifying screen. Autoradiography shows that mRNA for cathepsin K is abundant in human osteoclasts.
In situ hybridization, using known methods, was also used to show cathespin K expression in human cells. Cathepsin K was shown to be highly abundant in human osteoclast cells.
Example 7 Gene therapeutic expression of human cathepsin K
Fibroblasts are obtained from a subject by skin biopsy. The resulting tissue is placed in tissue-culture medium and separated into small pieces. Small chunks of the tissue are placed on a wet surface of a tissue culture flask, approximately ten pieces are placed in each flask. The flask is turned upside down, closed tight and left at room temperature overnight. After 24 hours at room temperature, the flask is inverted - the chunks of tissue remain fixed to the bottom of the flask - and fresh media is added (e.g., Ham's F12 media, with 10% FBS, penicillin and streptomycin).
The tissue is then incubated at 37°C for approximately one week. At this time, fresh media is added and subsequently changed every several days. After an additional two weeks in culture, a monolayer of fibroblasts emerges. The monolayer is trypsinized and scaled into larger flasks.
A vector for gene therapy is digested with restriction enzymes for cloning a fragment to be expressed. The digested vector is treated with calf intestinal phosphatase to prevent self-ligation. The dephosphorylated, linear vector is fractionated on an agarose gel and purified.
Cathepsin K gDNA capable of expressing active cathepsin K, is isolated.
Preferred constructs use the cathepsin K promoter for cell type-specific gene expression. The ends of the fragment are modified, if necessary, for cloning into the vector. For instance, 5' overhanging may be treated with DNA polymerase to create blunt ends. 3' overhanging ends may be removed using S I nuclease. Linkers may be ligated to blunt ends with T4 DNA ligase.
Equal quantities of the Moloney murine leukemia virus linear backbone and the cathepsin K fragment are mixed together and joined using T4 DNA ligase. The ligation mixture is used to transform E. Coli and the bacteria are then plated on agar-containing kanamycin. Kanamycin phenotype and restriction analysis confirm that the vector has the properly inserted gene.
Packaging cells are grown in tissue culture to confluent density in Dulbecco's
Modified Eagles Medium (DMEM) with 10% calf serum (CS), penicillin and streptomycin. The vector containing the cathepsin K gene is introduced into the packaging cells by standard techniques. Infectious viral particles containing the cathepsin K gene are collected from the packaging cells, which now are called producer cells.
Fresh media is added to the producer cells, and after an appropriate incubation period media is harvested from the plates of confluent producer cells.
The media, containing the infectious viral particles, is filtered through a Millipore filter to remove detached producer cells. The filtered media then is used to infect fibroblast cells. Media is removed from a sub-confluent plate of fibroblasts and quickly replaced with the filtered media. Polybrene (Aldrich) may be included in the media to facilitate transduction. After appropriate incubation, the media is removed and replaced with fresh media. If the titer of virus is high, then virtually all fibroblasts will be infected and no selection is required. If the titer is low, then it is necessary to use a retroviral vector that has a selectable marker, such as neo or his, to select out transduced cells for expansion.
Engineered fibroblasts then may be injected into humans or animals, including for example, rats and mice, either alone or after having been grown to confluence on microcarrier beads, such as cytodex 3 beads. The injected fibroblasts produce cathepsin K product, and the biological actions of the protein are conveyed to the host.
Example 8 Refolding of pCatK Expressed in E. coli
Bacterial expression
A fragment encoding pro-cathepsin K (no secretion signal) was inserted in the pET22b vector commercially available through Novagen, wherein the inserted gene is under the transcriptional control of the T7 promoter. The resulting vector, pET-pCatK, was introduced into BL21(DE3) cells by standard transformation methods. Cells were grown to OD650 = 0.6 and treated with lmM IPTG to induce the T7 promoter. Cells were harvested after 4 hours of aeration at 37°C after addition of IPTG.
Refolding procedure
IL of shake flask grown E. coli expressing pCatK was pelleted (about 2.5g wet weight). The pellet was washed twice with 50 mL TBS+EDTA (50 mM Tris, 150 mM NaCI, 1 mM EDTA, pH 8.0). The washed pellet was solubilized into 25 mL of wash buffer by dispersion with a Tekmar tissuemizer and lysed by sonication on ice. Following centrifugation (13,000xg for 30 min at 4"C), the lysate pellet was again washed with 25 mL of lysis buffer and pelleted. The washed lysate pellet was solubilized using the tissuemizer into 25 mL of 50 mM Tris, 150 mM NaCI, 5 mM EDTA, 10 mM DTT, 8 M urea, pH 8.0. After stirring for 15 minutes the sample was centrifuged and the supernatant 0.45 μm filtered prior to protein assay at 6.75 mg/mL. 6.5 mL of this material (43.88 mg) was refolded by quick dilution into IL of stirring 50 mM Tris, 5 mM EDTA, 0.7 M L-argimne, 10 mM reduced and 1 mM oxidized glutathione, pH 8.0. The solution was layered with N„ covered, and stirred overnight at 4"C. Following concentration to 13.75 mL using an Amicon stirred cell equipped with a YM-10 membrane, the protein concentration was assayed at 2.29 mg/mL yielding 31.49 mg of protein or a 72% recovery through refolding. Upon dialysis into PBS, 0.76 mg/mL of protein was recovered (33%). Dialysis into PBS + 0.5 M NaCI yielded 1.66 mg/mL, a 72% recovery through dialysis. 0.2 mL of this material was activated with the addition of 1/10 volume 0.5 M NaOAC, 0.2 M L-cysteine at pH 4.0, and the pH of the solution (now at pH 7.66) adjusted down to 4.0 with HOAC. 1 % "seed" mature cathepsin K purified from Baculovirus was added after pH adjustment. A significant precipitation also occurred with this material at pH 4.0 however, at 4.25 hr the protein concentration of the supernatant was 0.5 mg/mL resulting in a specific activity of 1.16 μmol/min/mg using Z-F-R-AMC as the substrate. Recovery through activation was 30% of total protein. Again, all detectable activity was inhibited with the addition of about 0.5 mM E64 (cysteine protease inhibitor) in 10% DMSO.
Using this refolding procedure an approximately 25 mg of refolded, activated mature cathepsin K may be isolated from IL of shaker flask grown E. coli assuming minimal or no losses due to scale-up.
It will be clear that the invention may be practiced otherwise than as particularly described in the foregoing description and examples. Numerous modifications and variations of the present invention are possible in light of the above teachings and, therefore, are within the scope of the appended claims. SEQUENCE LISTING
(1) GENERAL INFORMATION
(i) APPLICANTS: SmithKline Beecham Corporation, Human Genome
Sciences, Inc. and Institute for Genomic Research
(ii) TITLE OF THE INVENTION: CATHEPSIN K GENE
(iii) NUMBER OF SEQUENCES: 20
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: SmithKline Beecham Corporation
(B) STREET: 709 Swedeland Road
(C) CITY: King of Prussia
(D) STATE: PA
(E) COUNTRY: USA
(F) ZIP: 19406-2799
(v) COMPUTER READABLE FORM:
(A) MEDIUM TYPE: Diskette
(B) COMPUTER: IBM Compatible
(C) OPERATING SYSTEM: DOS
(D) SOFTWARE: FastSEQ Version 1.5
(vi) CURRENT APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE:
(C) CLASSIFICATION:
(vii) PRIOR APPLICATION DATA:
(A) APPLICATION NUMBER:
(B) FILING DATE:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: Gimmi, Edward R
(B) REGISTRATION NUMBER: 38,891
(C) REFERENCE/DOCKET NUMBER: ATG50006
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: 610-270-4478
(B) TELEFAX: 610-270-5090
(C) TELEX: (2) INFORMATION FOR SEQ ID NO:l:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 14237 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 1 :
GURCATHSNK GNMCDNASUN CSDNGCTTTG GCTCCCAAAG GCCTGGGATT ACAGGCGTGA 60
ACCACTGCGC CTAGCCTGTT AGCAGCTCTT AAAATCCAGA GGCATAAGCC TGTATTTTTG 120
AGGGTTTATG CATGGAATCC AGCTAGAAAC TGAGTCTATT ACAGATCCCA TTTATTATCC 180
TTTCTATTCC AAGAAGCCTT TTTTTCTCCT TCCCCACATC TGTTTATGGA AGAAAATGAA 240
GTTTGGGGTG TGGTTTGAGG AATCAGCTAG ATTCTTATGA TCTGTCACAT GCTTGGATGT 300
TGGGGAAGCA TTTGGAGAAG CTCATGTGAC TTGTCCTAGA TTGGGGATTT TAATTGAGAC 360
AGATGATGTT TATCGGGCAT CCCACCACCT GAGAGTTTTA GCAACAGAGT CACATGTGAG 420
TCCATCAGAA CTTACGGCAT TGATTCAAGT GCTGTCATAA ATAACCAGGA CTGCTGTTTT 480
TGGTTACTTT TAAAGACAGT TTCATCTGGA CTTTCTGGGC ATATCCTCCT TCAGCAAAAC 540
CACATTAGGC TGGGAAAACT ATTCTGCCTG GAAGTAATGA CAACTTGCAA CCAACAAGCT 600
TATAAAAATA CAAAGAATTC TGGAGCCTAT GGCTTCCATT ACATTATTCT TTTATAGCCT 660
TTTATGTTCA TTACCGCATC CCAGAGGTGA GAGTCAGACA CAAATATGAA AATAGGTTTC 720
AATGTTGGAG AGGTAAATCC TAACAGGAAA GGGGTAGGAA AAGATATAAT CCCCCAATAT 780
TAAAATAAAG ATATTGAAGA AGAAGGATGG GAGAGACTAG GGCTGTGTCC TTCCTTTTAC 840
TCACCAAAAG AGAAAGTAAG CTCCTATTTG AGTCAATAGA TATTGAGGTC TTGTTATTTG 900
CCACCAAAGA CAGTCTTGTG AGACTAAATA GCTAGTAATT CCCTACCCTG GCACACATGC 960
TGCATACACA CAGAAACACT GCAAATCCAC TGCCTCCTTC CCTCCTCCCT ACCCTTCCTT 1020
CTCTCAGCAT TTCTATCCCC GCCTCCTCCT CTTACCCAAA TTTTCCAGCC GATCACTGGA 1080
GCTGACTTCC GCAATCCCGA TGGAATAAAT CTAGCACCCC TGATGGTGTG CCCACACTTT 1140
GCTGCCGAAA CGAAGCCAGA CAACAGATTT CCATCAGCAG GTAACGTTTG CAACTTCCTA 1200
GATCTTTTAG CTTTTCATTC CTGTCAATTC TCTGAGTATT AGGGATGTAG TGACTTGAGG 1260
ATCACAATAA ACTTTTAGCC TCTGCAGATG AAAACAGAGA TGCACTTCTT AGGTCATTCC 1320
CTGGCTAAAT AAAATCTGCC TGGAAATCTG TAGAATTCCT TGTATGATTT ATATATATAC 1380
ATACATGATT GTTAGTAAAA GCAAAGTATA TAGGGAATCA TTTCCCCATC CTTCAAGAGT 1440
GGCCTTTCTG CAGTGTTTTC TACTTTGGCC AACAAGGATC AAAACGGTTA ACTCCTTAGT 1500
GAGGAGGAGG AGAGTGGTAT GGGGAGGTAG TAGCTCAGTG CTTCCTGTTC ACTGAGACAT 1560
CTCAAAGCCC TTAACACTCT AGTTTTTAAA TGTCCTACTG GACATTTTGC CAGTTTGCAA 1620
AATTACATGT AAATGGACTA TAAGCAATTG TGTAAGCCAT ATGTCATGCT GCAGGCTGCA 1680 AATTGTTCTT AAAATGGAGG ATTTGTAATT AAGAAAGCCA ATGCAAGAAA TGAGTGAAGC 1740
TAACTAGAGT AAACTTATGA AAAGCTGTGA ATTTCATCAT CATAGAACAT TGCTTTTCAG 1800
TCTGAACATT CTTCTAACAA ACCTTGGATC TGAGGCTTCT TGTCCTTTGC GGCAGCCACA 1860
GTGGGTTTTT GTTGTTAGGG GAAAATAAAA AACCTTGCCC GCAGCATCTG GTTAAGATTA 1920
GGGCAGTTTC CTGCCTAAGG AGGGAAGGGA GAGAAAAAGG AAGAAGAAAT GCATAAGGAG 1980
AATGAGGAGA TATACAATGT CTCAGAAAAC AGGAAACATT GTCCTATTTT CCCTTGTCCT 2040
CTTCTGACAA GATCTGGGAA AGTACCAGAA TTTAGGCACG AAAGAGAAGA ACGCCTCGAA 2100
GAAATGATCA GGAAGCAAAA CTTAGACGGA AATCTCTCCT TTGTGTATTC TGAACCCCAC 2160
TACCACCTTG CTATTTGTCT GTCTCCAAGC CTGCTAGGGA CCCTGGAGGA AACGCACTGA 2220
GCCCATTCTG ATTGTCCAGT TTCTATCCCC CATTTCTGGT TGTGTACGTG TGTGTGTGTG 2280
TGTGTGTGTG TGTGTGTGTG TGTGTGTGTG TGAGAGAGAG AGAGACAGAG AGAGAAACAG 2340
AGAGAGTGTG TGTTGCCTAA ATCTCCCGAG AGAGAGAGAG AGAGAGAGAG AGAGAGAGAG 2400
AGAGAGAAAA GAGAGAAATG GCTAAATCCC CCTAGATCAA AGTCCTTGGA ACCAGATGTA 2460
CCAGCATCCT ATCTAAACAC AGGCCCCTCC TGACTATCAT TGTTTTATCA CCCTTTTTCC 2520
GTCTACCTTT CTCTTCCTCA TAAAGCCTAG TTTTCCTCTG TTTCCCTGCC AAATGGAAGA 2580
GTTTTCCCTA ACTACATTCT TCTGCAGGAT GTGGGGGCTC AAGGTTCTGC TGCTACCTGT 2640
GGTGAGCTTT GCTCTGTACC CTGAGGAGAT ACTGGACACC CACTGGGAGC TATGGAAGAA 2700
GACCCACAGG AAGCAATATA ACAACAAGGT GCCTGGGGTC CTGGAGGGGG CATGGCAGGA 2760
AGGCTGAGAC CTGAGCTCTC TCATCTTAGC TTCCAGACTC CCTTCTTCAA TCCAAATGCT 2820
TTATTCCAAG CAAATCAGTC CCTCTTCCCT AACTCATGTT AACATACGGT TTTCATTCCT 2880
ATGCTTCAAT CATCCTCTTG TCAAACTTGT ATTCCTTCCC TTTGGTTTTA TAAGTGTGTA 2940
ACATTCCTCT TTTGGGAAGA GTCCCAAGAT TAATGCTGTT AATCCATAAG CAATTTTTCT 3000
GTCTCTCCAG AGCTTGTGTG GTTGTTTACA TATTATCTCT CTTCTTGCAG GCTCTTAATT 3060
CCATGGTTAG TTCCCCAACT AAACTGTAAA CTTTTATGAT TGTGAGTTTC CTTTATTCTC 3120
CTAAAACCCT TCACAATATT ACATATGAAC TGTAGACAGT CTATACAAGT ACTGACTATG 3180
CTTTGTTTAG GTGGATGAAA TCTCTCGGCG TTTAATTTGG GAAAAAAACC TGAAGTATAT 3240
TTCCATCCAT AACCTTGAGG CTTCTCTTGG TGTCCATACA TATGAACTGG CTATGAACCA 3300
CCTGGGGGAC ATGGCAAGTA TAGCTTCAGC TCCTGTCCCA CCTGCACCAT TTGCTTTAGT 3360
TCCCTGCTGA TGCCTGGCCT CTTTCTTCTT TGTCTTAGAC CAGTGAAGAG GTGGTTCAGA 3420
AGATGACTGG ACTCAAAGTA CCCCTGTCTC ATTCCCGCAG TAATGACACC CTTTATATCC 3480
CAGAATGGGA AGGTAGAGCC CCAGACTCTG TCGACTATCG AAAGAAAGGA TATGTTACTC 3540
CTGTCAAAAA TCAGGTACTC TCCTTTCTTC TGGGTGTGCA TATGTAATCT GGCATGACCT 3600
TTTCCTTTTT CTGCTGCTTT GTTCTTGAGG TGAAAGGGCA CCAGGAAAAG AGGGCAAGGA 3660
ATTAAGGTAC ATCTCCCCAT TCCCATTCTG TTATTTAACC TCATTTGTTT CTGTACATTT 3720
GGGTTGTTTC TGGTTTTTCT TTTTCTTTTC CCTTTTTTTT TTTTTTTTTT TTTTGAGATA 3780
GAGTCTCACT CTGTCGCCCA GGATGGAGTG CAGTGGTGCA ATCTTGGCTC ACTGCAACCT 3840
ACACCTCCCG GGTTCAAGCG ATTCTCCTGC CTCAGCCTCC TGAGTAGCTG AGATTACAGG 3900
CACGCGCCAC TACGCCTGGC TAATTTTTCT ATTTTTATAG AGATGCGTTT TCACCATGTT 3960
GGCCAGGCTG GTCTTGAACT GACCTCAGGT GATCCACCTG CCTCAGCCTC CCAAAGTGCT 4020
GGGATTAGAG TCATGAGCCA TCGCGGCCTG GTTTTTCTTT ATTACAAATA GTGTTGCAAT 4080
AAGCACCCTT GTGCATATGT TTTTGTGCAC ATGTACAAAT ATTTATGCAA AATAAGTCCT 4140
AAAATTGGAA TTGTTAGGTC ACAAATAATC CTTTCCCCCC CCCCAAATTT TTTTTTTTTT 4200
TTTGAGACAG CGTCTCTGTC ACCCAGGCTG GAGTCCAGTG GCGCAATCAT GGCTCACTGC 4260
AGCCTCAACG TCTCAGGCTC AAGTGATTCT CCAACCTCAG CCTCCCTAGT AGCTGGGAAT 4320 TAGAAGCACA TGCCACCACA CCCAGCTAAT TTTAAAAAAT TTTTTGTTAG AGACAGGGTT 4380
TTGCCATGCT ACCCAAGCTG GTCTCAAATT CCTGGGCTCA AGCAATCTGC CCGCTTCGGC 4440
CTCCCAAAGT GCTAGGATTA CAGACATGAG CCACCATGCC CAGCCCAAAA AAGTTTTTGC 4500
AATCTTACAT TCTTACTAGC ATGAGAATGT CAGTTTTTTC ACAACCCAAA CAACACAGGA 4560
TTGTATCAGC AAGATAAACA ATTGATTTAA CGTTCATTTA ACAAACACTT TTTGACCCCC 4620
AGAACCTACC AGATGCAGTG TTAGGCAGCA GAGACTCAAG ATGACTAAGA CACAACCTGT 4680
GTCCTCAGGA AATCTCAATC TAAAAAAATA GAACAGGAAA GAAAGAAAAA TCTACAATCT 4740
AGCTGCACAA ACAATAATAG CTAATACTTT TTGAGATTTT ATTGTTTGTC AGGAACTTCT 4800
TAACTCTTTA CATGAGTTTA AATATTTAAT CCCTTATAAC AATATTTTAT GCATAGAGAA 4860
ACTGAGACAC AGGCAAATTT AGTAACTTAC CCGGGGTCAC ATAGCTACTG GGTGGCAAAG 4920
TCAGGGTTAG CTCCCAGGAC AAATGCCTCC ACAGCTGGTA CTGTGCTCTG CTTTACTGTA 4980
GCTAATAGTA AAAATGGTAG CAAAAATCAA TAGCAGTAGA ACAGTGCAAC AGATATTAAG 5040
CGGAAGAGGA AGACTCACAA CAATGACAAC ATTTGTGCTG AAATTTTTAA GAACACATGG 5100
AATTTCCTTC AGCCGGGTAG AGAGAAGATA TAGAAATGTA AACACCAAAG ATTCATAGTT 5160
TCTCTGTATC CCTTTCAGGG TCAGTGTGGT TCCTGTTGGG CTTTTAGCTC TGTGGGTGCC 5220
CTGGAGGGCC AACTCAAGAA GAAAACTGGC AAACTCTTAA ATCTGAGTCC CCAGAACCTA 5280
GTGGATTGTG TGTCTGAGAA TGATGGCTGT GGAGGGGCTA CATGACCAAT GCCTTCCAAT 5340
ATGTGCAGAA GAACCGGGGT ATTGACTCTG AAGATGCCTA CCCATATGTG GGACAGGTGA 5400
GATTGCTCCA CACAATTATA CAGCTCTGTT GGCTCCTCCT CCCCAGCATG ATGTTTTGTA 5460
CTGGAAACAA TTCCAGAAAT ACTGTTTTCT GTTATCCTAT CCTGCTTTCT TGATGGAATA 5520
ATTTCCCACA GAAGGCCAAG AAGATTTCCA CAATCTGGGG GAATTTAGGG AGCTTAAGCT 5580
ACTATAGCTC CTATTTGCAT CTCTGCCATG GAGAGAAAAC AGAGGCTAGG CTACCTACCC 5640
CATAGACTTC CGAGCTGGGT TCTATAACCC TCTGCTCAAT TCCTCACTCC CACAACAAAC 5700
CCACAAACCC ACCATGCTAT TTTCACAAAT TGTGTGGCTT TATTTTATAT GATCTCAGTG 5760
TGAGTTTTCA GAACATTTCA GCAAATTATG TAAGTTTACA TGCTAACATC TATAAAATGA 5820
GAGAAAAAAC AAGTTGCTTC ATATAAGAGA TAAGGGATTA ACTCAGTTCC TCCTGCATGA 5880
TCCTCTAGTC ATAGGAAGGA AATCATATCT GAAAGGGAGG CAACCTGAGG GGTTTTTTAT 5940
ACACATAGGG CTGGGTCTGA TAGACAATAT AATGTAGGGC CTTCACAACA GAAACCTCTG 6000
AAACAGGGAC AGCAAGTTTG AGAATAAAAA TGATGGCTAC TGTGTTCTAA GCCGTGTCCT 6060
TAGTGCATTT TTTCTTTTTC TTTTTTTCAT TTAATCTCAT AACAACTCTG TTAGGTAGAC 6120
TTATCTTGAA TGTATAGGTG AGGAAATGGA CACTTAAGGA GATAAGACAG TATAATTCAT 6180
ACCACTAGTA TGTAACAATG TAAGATGTAT CTACCAGGGA TGTTTATCTT CTGCAAACAT 6240
TCCTAGGTAT ATCTCCCATG CACATGTGCA AGAATTTCTT ACTAGGATAT AATGCCTTGG 6300
AACTGAATTG TCTGGGTCTT AGGGTATGTC TGTCTTCACT TTACTACACA ATGTCAAATT 6360
GTTTGCCAAA ATATTTGGAA AAATTTATAC CTGCAATGTG TAAGAAATCC CCTTCAATCA 6420
CCTTTTTATC AGTATGTTTA TCTGGCCATT TGCATTTCTT CTTCAGTGAA TTAACTGTTT 6480
TTATCTCTTG CTCATTTGTT TTTCTTTTTA TTTTTTTGAA ATAGGGTCTT ACTCTGTTGC 6540
CCAAGCTGGA GTGTGGTGAA CAGTCATAGC TCACTGCAGC CTCCACTTCC GGGCTCAAGC 6600
AATCCTCTCG CCTCAGCCTC CCAAATAGCT AGGATATAGG TGCATGCCAT CATGCCCACC 6660
AATTTCAAAA AACCTTTGAA ATTTTTTTTT GTAGAGGCTA GGCATGGTGG CTCATGCCTG 6720
TAATCCCAGC ACTTTGGGAA GCTGAGGTGG GAGGATCGCT TGAGCCCAGC ACTTTGGGAA 6780
GCTGAGGTGG GAGGATCGCT TGAGCCCAGG AATTGGAGGT CGGCCTGATA CAACATAGCA 6840
AGACCTCATC TCTACAGAAA AAATTTTTAA AAGTAGCCAG GTATGATGGC GTGCATAGTT 6900
CTAGCTACTC CGGAAGCTGG TTGGGAGGAC AACTTGAGCC TGGGAGTTCA AGGCTGCTGT 6960 GAACTGTGAT CATGTCACTG CTCTCTAACC TGGGTGACAG AGTGAGACCC TGTCCCCAAA 7020
AAACAACAAC CGTTTTTTTT TGGTAGAGAC ATTGTCTCGC TATGTTGCCA AGGCTAGTCT 7080
CAAACTCCTG GGCTCAAGCA ATCCTCCCAC CTCCCCAAAG TGCTGGGATT TATAGATGTA 7140
AGCCACCATG CCTGGCCTAC CCTTTTTTTT TTTTTTTGAA ATGGAGTTTT GCTTTTGTCA 7200
CCTAGGCTTG AGTGCAGTGG CGCGATCTTG GCTCACTGCA ACCTCCACCT CCTGGATTCA 7260
AGCAATTCTC CTGCCTCAGC CTCCTGAGTA GCTGGGATTA TAGGCACCCG CAACCACGCC 7320
CGGCTAGTTT TTGTATTTTT AGTACAGACA GGGTTTCACC ATGTTGGCCA GGCTGGTCTT 7380
GAACCCCTGA CCTCAGGTGG TCCGCCCGCC TCGGCCTCCC AAAGTGCTGG GATTACAGGT 7440
GTGAGCCACC ATGCCCCACC CCTTACTCAT TTTTAATTGG ATTGTTTTTT CTCTTTCTTA 7500
GCGATTCTTA AAAGTTTAAA GAGAATATTT GGATACAATA CTATGTATTT AAAAGTTGAG 7560
GTCTGTCTTT CCATTCTTTT TATGATGTCT TTCAATCTAC AAAAGTTAAT TTTAATAGCC 7620
TGGCGCCGGT GGATCTCGCT TATTATCCCC TCACTTTGGG AAGCTGAGAT GGGTGGATCA 7680
CAATGTCACG AGATCTTGAC CATCCTTCCT GGCGCGGTGG CTGCTAATGG AAGCGGAACA 7740
CGTATAAAGC CAGTCCGCAC AAACGGTGCT GACCCCGGAT GAATGTCTGC TACTGGGCTA 7800
TCTGGACAAG GGAAAACTCA AGCGCAAAGA TAAAGCAGGT AGCTTGCAGT GGGCTTACAT 7860
GGCGATAGCT AGACTGGGCG GTTTTATGGA CAGCATGCCA ACCGGAATTG CCATCTGGGG 7920
CGCCCTCTGG TAAGGTTGGG AAACCCTGCA AAGTAAACTG GATGGCTTTC TTGCCGCCAA 7980
GGATCTGATG GCGCAGGGGA TCAAGATCTG ATCAAGAGAC AGGATGAGGA TCGTTTCGCA 8040
TGATTGAACA AGATGGATTG CACGCAGGTT CTCCGGCCGC TTGGGTGGAG AGGCTATTCG 8100
GCTATGACTG GGCACAACAG ACAATCGGCT GCTCTGATGC CGCCGTGTTC CGGCTGTCAG 8160
CGCAGGGGCG CCCGGTTCTT TTTGTCAAGA CCGACCTGTC CGGTGCCCTG AATGAACTGC 8220
AGGACGAGGC AGCGCGGCTA TCGTGGCTGG CCACGACGGG CGTTCCTTGC GCAGCTGTGC 8280
TCGACGTTGT CACTGAAGCG GGAAGGGACT GGCTGCTATT GGGCGAAGTG CCGGGGCAGG 8340
ATCTCCTGTC ATCCCACCTT GCTCCTGCCG AGAAAGTATC CATCATGGCT GATGCNACTG 8400
CGTTTCAAAA AAAAAAAAAG TTAATTTTAA TATAGTAAAA TTAGTAAAAG GATTAATTTT 8460
CCCTTTGCAA TTTTTGTAAT GTGTTTTATT CGTTTATGAA TGGAGAAAGG TAAGAAAAAA 8520
TAAAATTTAA AAAAGAAGAG ATGTGGCCAG GTACGGTGGC TCACACCTAT AATCCCAGTA 8580
GTTTGGGAGG CTGAGGCAGG CAGATCACTT GAGGTCAGGA GTTTGAGACC AGCTGGGATA 8640
ACATGGTGAA ACCCCATCTC TACTAAAAAT ACAAAAATTA GCCAGGTGTG ATTGCGCACG 8700
CTTGTAATCC CAGCAGGCTG AGGCAGGAGA ATTGCTCGAA CTCAGGAGGC AGAGGTTGCA 8760
GTGAGCCAAG ATCATGCCAT TGCACTCCAG CCTGGGTAAC AGAGACTCTG TTTCAAAAAA 8820
TAAAAAGATA AAAAGGGAAG AGATCTGATA GGGCGCCCAG AAAAACATTT TAAAGGGGAT 8880
GGTATTATAA GTTTGTTCCC AGCATAATGC CAGGTTATTT CTGACTTTAA AGTATCATCA 8940
CATAATATCT TTTTGAGTCA ATTTCCAAGA TATTCTGTTT CACTTGTAAT TCTGTGTAAT 9000
TTTTGGCACC AGGAGGCATC AGGGATTTGG AGCACATGGC AGAAACAAAG GCATCTTGAA 9060
AAATATCAAG GCAGTAGACC ACTGTAATCT TAAAATGGCA TATCAAATGC TGCTATTGCT 9120
GTTAATATTT AGATAATGTT AGATAATGTA TTTTTTTAGA GGGTATCTCA CTATCTTGCA 9180
CAGGCTGGAG TAGAGTGGCT ATTCACAGCA TGATCACAGT ACACTAAAGG CTCAAACTCC 9240
TGGGCACAAA CAATCCTCCT GCCTCAGCCT GCTGAGTAGT AGATAATAAG TTCTTGTGGA 9300
TGCAACCTTA GGGTTCTGAA GGGGTAGTCT GTAGGAAAAT GAATTGCTGA AAAGAATACA 9360
CCACCTTAAC ATGGGCTATT ATTCGATTCC ATAATTGTGG CTTGCCAATG AAACATTGCT 9420
AACTACCTGT AAAATATAGT GTTGGAAGTC ATAGGCTAAA TTGCTAAGTT CTTTAATCTA 9480
TTTTAGTGTC TTGTTATGTA CTTTTATATT TTGTCTTTGA TGAGAGCACA AGGATCACAC 9540
CAGTTCCCCT GATATAGGTG CAGAGGGCCC AGGTCTTCCC TCTAGCTAAG CCTTGGCCTT 9600 GGCCTCCTAC CCACACAGCA GCTGGTGCCT TCCTGCCCCC TGAGGCTAAT ACATACTATG 9660
TGGCCAGAAG ATGGTTTATG CTTTTTAAAA AAATCTTATT TCAGAAATCT TTCCCTACTG 9720
TTTTCCTCCC ACATTTATGT CTTAAAACAC CTGTAGGGGA TTTTTTTTTT TTTTTTTTTT 9780
TTGAGATGGA GTCTCGCTCT CGCCCAGGCT GGAGTGCAAT GGCGCGATCT TGGCTCACTG 9840
CAAGGTCTGC CTCCCAGGTT CACGCCATTC TCCTGCCTCA GCCTCCCCAG TAGCTGGGAC 9900
TACAGGCGCC CGCTACCACG CCTGGCTAAT TTTTTTGCAT TTTTAGTAGA GACAGGGTTT 9960
CACTGTGTTA GCCAGGATGG TATAGATCTC TGACCTCGTG ATCCACCTTT CTTCAGCCTT 10020
CCAAAGTGCT GGGATTAACA GGCATGGAGC CCCACCGCAC TGGCCTGTAG TTGGTTTTTA 10080
TGTGTGGTGG AAGGCGGGAA TCCTCTTTTC ATATTCGTTT TTGTGAGGAA GAACAGACCC 10140
TCTTTAGAAG CCCTAGACTG CTGCCTCTGT TAGTTCACTG GCATCACTCA AAATATTGGT 10200
TGAGTTTCTT ACTCACTGAC TCATTGCCTA TTGCTTTGTC CTAGTCCTAT TACAATCTTG 10260
TTTCTTCCAG CCAGGAAGAG AGTTGTATGT ACAACCCAAC AGGCAAGGCA GCTAAATGCA 10320
GAGGGTACAG AGAGATCCCC GAGGGGAATG AGAAAGCCCT GAAGAGGGCA GTGGCCCGAG 10380
TGGGACCTGT CTCTGTGGCC ATTGATGCAA GCCTGACCTC CTTCCAGTTT TACAGCAAAG 10440
GTAAGAAGCT GCTGATCCTA TACAGCACTG TCTTTTATGA TACAAACTTG ATGGTTTCTC 10500
GAAGGACCTT GGGTATTTTC AGTACTTAGT TTTTGTATTC ACATGGAGGT GGCCAGAGAG 10560
AAATTAACAA CTGCTGCAGT ATGGAGCAGC ATCTCTGTGG TAAACCCTCC TGACACGGAT 10620
GGAATTCTTC AAACAGTCTC CTAGACTGGG AGATCCCACA GGGTGACCCT TGGATTGCAT 10680
AGAGCCTCAC GCTGGTAGTT TGTATTCTAG GTGTGTATTA TGATGAAAGC TGCAATAGCG 10740
ATAATCTGAA CCATGCGGTT TTGGCAGTGG GATATGGAAT CCAGAAGGGA AACAAGCACT 10800
GGATAATTAA AAACAGGTAA TGATGGGAAC ACTACTTTTG TTATTCAGTC ACCCTTTTAA 10860
CACTCAACCT CACCTCCAGC TTCCCGATAT TCCTTTCTCT GTCCCAAATC AAGAAAAAAT 10920
TATCTCAGAG TTCTCACTTC TATCTTCTCA GTCAGAGGCT CTTAATTCTC AGTCTGACAC 10980
TTAATGGCCA GTGTGTTAGT CCATTTTGCA TTGCCACAAA AGAATACCCG AGACTGGGTA 11040
GTTTATAAAG AAACGAGGTT TGTTTGGCTA TACAAAGCGT GGCACTAGTA TCTGCTCAGC 11100
CTCTGATGAG GCCTCAGAGC TTTTACTCAT GGCAGAAGGC AAAAGAGGGA GCAGGCATGT 11160
CACATAGTGA GAGAGGGAGC AAGAGAGAGA GGGAGGTGCC GACTCTTTAA AGAACCAGCT 11220
CTTGCATGAA CTAATAGAGT GAGAACTCAC TCATCACCAA GGCGATGGCA CCAAGCCATT 11280
CCATGAGGAA TCCACTCTCA TAACCCAAAC ACCTCCCACT ATGCCCCACC TCCCACATTG 11340
GGGATCACAT TTCAGCATGA GACTGGGAGG GGACACACAT CCAAACCATA TCCGCCAGAC 11400
AATAGTGCTC AATTATGTGC TGGGCAGATG CTCCCTGTGT GCAAGGTGCT TAGTGACATA 11460
CATAAACCAA CGAGCAGATG ACACCTTCAG TGAGCTCAGA GCCCAATAAG ACAGACCTAA 11520
CTAACCATGA GATAAAGCAG TACAAAGAAC CAGCAGGAGC TTTGGAATTA CGTATTTTTA 11580
CTTTCTTTTG TCTCTAATGT GATCAGTTTC TTAGATGGTT TCCATTAGCA ATCTGTCTTT 11640
AACAGTAGGG GAGCAGCGTT AAAGGTTTAA TATTCCTTTT GAACAGTTTT TTTCCTTCAA 11700
AATACACTTA AGATACACGT ATATAAGAAC TTGCCAAAGA TTGTGAAGAG AAACATTTTT 11760
TAGAAATAAG ATATAAACAA AAAAAGTTAG TGTTACTTTC CTATGTTGGG GAACAAAGAA 11820
AACTCCAGGG TACCTTGCTT CCCATTTCTC TTTAGCACCT TGTGACTTTT GGGGAGGGGC 11880
AGATTGATAA CAATTATAGT TTTCCTTTCC TGGCTGATCA CCATTAACCT GGCAGCAGCA 11940
CTGGCTAAAT CTCCTGTCCT TAGTGCCCTC CAAGGAGCAG GAGCCCTAGA CTCTGGGTCG 12000
CTGACAGACT CACGCAGTGG TGTTGTTCAA ACCTGAAGCA ACTTTTTATA TCACAGTTCC 12060
AACTCAAGGT GAACCTGAGC ATCTTCCCAA GTCTCCCACA GCTTCTGTCC TGTGTTGTCC 12120
CTTCTCTTGA CTCCCAGGTC CAAGCACTTA CCCTGTTCTT TCATGATCAG GTACCATGTG 12180
TGGAGATAGC TTCCAAGAGA GCTGGGAGGA AGAAAGGACA CACCCGGGCA GGATCAGGAA 12240 CACTGGGGGC CCCTGGAGAA GGGGAGAGTG GGGGAGGGTA CAGGTTTTAA ATAAAATGTG 12300
TTGGTAATTA GAGAATTGCT GGTTGGGGAA AGAGGTCTGA AAACAATTCA GGAAGATAAA 12360
CAAGACAATC TCTCCTCTCT CCTCTTTCTC ACGTCGTCTC TCTTGTCTTC TAGTCTCGCT 12420
ACTCATTTCC TTAGTAATCT CATCCACTCT CATAGTTTCA TCCATCTCTC CTATGGGGTT 12480
TACCCCCAAA TCAAGATCAC CAGCTTCAGC CTCCTTCTTA TGCTCTAAAC TCACATTTTC 12540
AAGATTAATA TTCCCCAAAT ACAGCTCTGA TCATATCACT CTCCCACTCA AAATCCCTCA 12600
CTGGCTCCTC ACGATGATGG GTCACAGAGT AAAGGTGAAG CTTTTTAACC TTGCAGTAAA 12660
GGTAATTCAA CCTGATCTCA ATCTGCCTTT CCAGACATCT CTCCCACTAC ACCCTGTTAG 12720
GCACACTGCT TTTCAGCTAC ATGATCCTAA CAGTGCCCCA CACTTTCCTG CCTCTGTTGT 12780
TCATTTCACA CCCTTCCACT GGCATCCCCT TCCCACAGGT CGAAATTCTA CTTAGCCTTT 12840
TGGCTCAGCT CAAATGCCAC CTCTTACATC AAGCCTCTAA GATTCTCTTG ATCAGAAGGA 12900
ATCTTTCCCT CCTTTGATAC CTACAGTATT ATGCCTTCTC CCTATTTCTT GACTTTAAAC 12960
TCTTTAAAGT TAAAAAACAT CATATTCATT TTTGTGTACC ATCAGTACCT CGCACAATAC 13020
TCAGTAAATA TTTTAATGAA TAAATAAACT GAGAGTACTA AGTATTTTTC TTGATTGGTC 13080
TTACAGCTGG GGAGAAAACT GGGGAAACAA AGGATATATC CTCATGGCTC GAAATAAGAA 13140
CAACGCCTGT GGCATTGCCA ACCTGGCCAG CTTCCCCAAG ATGTGACTCC AGCCAGCCCA 13200
AATCCATCCT GCTCTTCCAT TTCCTTCCAC GATGGTGCAG TGTAACGATG CACTTTGGAA 13260
GGGTGAAGGT GTGCTATTTT TGAAGCAGAT GTGGTGATAC TGAGATTGTC TGTTCAGTTT 13320
CCCCATTTGT TTGTGCTTCA AATGATCCTT CCTACTTTGC TTCTCTCCAC CCATGACCTT 13380
TTTCCACTGT GGCCATCAGG ACTTTCCCTG ACAGCTGTGT ACTCTTAGGC TAAGAGATGT 13440
GACTACAGCC TGCCCCTGAC TGTGTTGTCC CAGGGCTGAT GCTGACAGGT ACAGGCTGGA 13500
GATTTTCACT AGGTTAGATT CTCATTCACG GGACTAGTTA GCTTTAAGCA CCCTAGAGGA 13560
CTAGGGTAAT CTGACTTCTC ACTTCCTAAG TTCCCTTCTA TATCCTCAAG GTAGAAATGT 13620
CTATGTTTTC TACTCCAATT CATAAATCTA TTCATAAGTC TTTGGTACAA GTTTACATGA 13680
TAAAAAGAAA TGTGATTTGT CTTCCCTTCT TTGCACTTTT GAAATAAAGT ATTTATCTCC 13740
TGTCTACAGT TTAATAAATA GCATCTAGTA CACATTCATT TTGTGTTGGA TACTGTGTTA 13800
GGTGCTGGAG GAAAAAAGAT GAATAGAACA TCTTCTATGT ACTTGATGCG CTCACAGTCT 13860
GGTTGTAGAG ACTGTCACAT AAACATTTCA TCCCAATTCA TTTATTTGTT CATTCCTTCA 13920
GCCAATATAT ATTGAGTTCT TACTCTGTGC CAAGAACTGT ACTACATTTC TGGGATTAAG 13980
TGGATATAAG GAGATCTCAG TGTTTAATCT GCCTGAGGGG AGACTAAATT AAGTGACATG 14040
GAAACTTGGG TCTTGAAAAA CATTTTAAGG TTATTTTTTC TTTTCTCTCT CTCTCGCTCT 14100
GTCTTTCTCT CTCTTTCGTC AGGGTCTCCC TCTGTTGCCC AGGCTGGAGT CAGTGGCACT 14160
CATAGCTCAC TGCAGCCTTG ATCTCCTGGG CTCAAGAGTT CTTCCCACCT CAGTCTCCTA 14220
AGTAGCTTGG ACTACGG 14237
(2) INFORMATION FOR SEQ ID NO:2 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1108 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear (ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2 :
GCTTTGGCTC CCAAAGGCCT GGGATTACAG GCGTGAACCA CTGCGCCTAG CCTGTTAGCA 60
GCTCTTAAAA TCCAGAGGCA TAAGCCTGTA TTTTTGAGGG TTTATGCATG GAATCCAGCT 120
AGAAACTGAG TCTATTACAG ATCCCATTTA TTATCCTTTC TATTCCAAGA AGCCTTTTTT 180
TCTCCTTCCC CACATCTGTT TATGGAAGAA AATGAAGTTT GGGGTGTGGT TTGAGGAATC 240
AGCTAGATTC TTATGATCTG TCACATGCTT GGATGTTGGG GAAGCATTTG GAGAAGCTCA 300
TGTGACTTGT CCTAGATTGG GGATTTTAAT TGAGACAGAT GATGTTTATC GGGCATCCCA 360
CCACCTGAGA GTTTTAGCAA CAGAGTCACA TGTGAGTCCA TCAGAACTTA CGGCATTGAT 420
TCAAGTGCTG TCATAAATAA CCAGGACTGC TGTTTTTGGT TACTTTTAAA GACAGTTTCA 480
TCTGGACTTT CTGGGCATAT CCTCCTTCAG CAAAACCACA TTAGGCTGGG AAAACTATTC 540
TGCCTGGAAG TAATGACAAC TTGCAACCAA CAAGCTTATA AAAATACAAA GAATTCTGGA 600
GCCTATGGCT TCCATTACAT TATTCTTTTA TAGCCTTTTA TGTTCATTAC CGCATCCCAG 660
AGGTGAGAGT CAGACACAAA TATGAAAATA GGTTTCAATG TTGGAGAGGT AAATCCTAAC 720
AGGAAAGGGG TAGGAAAAGA TATAATCCCC CAATATTAAA ATAAAGATAT TGAAGAAGAA 780
GGATGGGAGA GACTAGGGCT GTGTCCTTCC TTTTACTCAC CAAAAGAGAA AGTAAGCTCC 840
TATTTGAGTC AATAGATATT GAGGTCTTGT TATTTGCCAC CAAAGACAGT CTTGTGAGAC 900
TAAATAGCTA GTAATTCCCT ACCCTGGCAC ACATGCTGCA TACACACAGA AACACTGCAA 960
ATCCACTGCC TCCTTCCCTC CTCCCTACCC TTCCTTCTCT CAGCATTTCT ATCCCCGCCT 1020
CCTCCTCTTA CCCAAATTTT CCAGCCGATC ACTGGAGCTG ACTTCCGCAA TCCCGATGGA 1080
ATAAATCTAG CACCCCTGAT GGTGTGCC 1108
(2) INFORMATION FOR SEQ ID NO:3 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 48 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE: (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3:
CACACTTTGC TGCCGAAACG AAGCCAGACA ACAGATTTCC ATCAGCAG 48
(2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1427 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4 :
GTAACGTTTG CAACTTCCTA GATCTTTTAG CTTTTCATTC CTGTCAATTC TCTGAGTATT 60
AGGGATGTAG TGACTTGAGG ATCACAATAA ACTTTTAGCC TCTGCAGATG AAAACAGAGA 120
TGCACTTCTT AGGTCATTCC CTGGCTAAAT AAAATCTGCC TGGAAATCTG TAGAATTCCT 180
TGTATGATTT ATATATATAC ATACATGATT GTTAGTAAAA GCAAAGTATA TAGGGAATCA 240
TTTCCCCATC CTTCAAGAGT GGCCTTTCTG CAGTGTTTTC TACTTTGGCC AACAAGGATC 300
AAAACGGTTA ACTCCTTAGT GAGGAGGAGG AGAGTGGTAT GGGGAGGTAG TAGCTCAGTG 360
CTTCCTGTTC ACTGAGACAT CTCAAAGCCC TTAACACTCT AGTTTTTAAA TGTCCTACTG 420
GACATTTTGC CAGTTTGCAA AATTACATGT AAATGGACTA TAAGCAATTG TGTAAGCCAT 480
ATGTCATGCT GCAGGCTGCA AATTGTTCTT AAAATGGAGG ATTTGTAATT AAGAAAGCCA 540
ATGCAAGAAA TGAGTGAAGC TAACTAGAGT AAACTTATGA AAAGCTGTGA ATTTCATCAT 600
CATAGAACAT TGCTTTTCAG TCTGAACATT CTTCTAACAA ACCTTGGATC TGAGGCTTCT 660
TGTCCTTTGC GGCAGCCACA GTGGGTTTTT GTTGTTAGGG GAAAATAAAA AACCTTGCCC 720
GCAGCATCTG GTTAAGATTA GGGCAGTTTC CTGCCTAAGG AGGGAAGGGA GAGAAAAAGG 780
AAGAAGAAAT GCATAAGGAG AATGAGGAGA TATACAATGT CTCAGAAAAC AGGAAACATT 840
GTCCTATTTT CCCTTGTCCT CTTCTGACAA GATCTGGGAA AGTACCAGAA TTTAGGCACG 900
AAAGAGAAGA ACGCCTCGAA GAAATGATCA GGAAGCAAAA CTTAGACGGA AATCTCTCCT 960
TTGTGTATTC TGAACCCCAC TACCACCTTG CTATTTGTCT GTCTCCAAGC CTGCTAGGGA 1020
CCCTGGAGGA AACGCACTGA GCCCATTCTG ATTGTCCAGT TTCTATCCCC CATTTCTGGT 1080
TGTGTACGTG TGTGTGTGTG TGTGTGTGTG TGTGTGTGTG TGTGTGTGTG TGAGAGAGAG 1140
AGAGACAGAG AGAGAAACAG AGAGAGTGTG TGTTGCCTAA ATCTCCCGAG AGAGAGAGAG 1200
AGAGAGAGAG AGAGAGAGAG AGAGAGAAAA GAGAGAAATG GCTAAATCCC CCTAGATCAA 1260
AGTCCTTGGA ACCAGATGTA CCAGCATCCT ATCTAAACAC AGGCCCCTCC TGACTATCAT 1320
TGTTTTATCA CCCTTTTTCC GTCTACCTTT CTCTTCCTCA TAAAGCCTAG TTTTCCTCTG 1380
TTTCCCTGCC AAATGGAAGA GTTTTCCCTA ACTACATTCT TCTGCAG 1427 (2) INFORMATION FOR SEQ ID NO: 5 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 121 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (V) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 5 :
GATGTGGGGG CTCAAGGTTC TGCTGCTACC TGTGGTGAGC TTTGCTCTGT ACCCTGAGGA 60 GATACTGGAC ACCCACTGGG AGCTATGGAA GAAGACCCAC AGGAAGCAAT ATAACAACAA 120 G 121
(2) INFORMATION FOR SEQ ID NO:6 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 462 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (V) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 6 :
GTGCCTGGGG TCCTGGAGGG GGCATGGCAG GAAGGCTGAG ACCTGAGCTC TCTCATCTTA 60
GCTTCCAGAC TCCCTTCTTC AATCCAAATG CTTTATTCCA AGCAAATCAG TCCCTCTTCC 120
CTAACTCATG TTAACATACG GTTTTCATTC CTATGCTTCA ATCATCCTCT TGTCAAACTT 180
GTATTCCTTC CCTTTGGTTT TATAAGTGTG TAACATTCCT CTTTTGGGAA GAGTCCCAAG 240
ATTAATGCTG TTAATCCATA AGCAATTTTT CTGTCTCTCC AGAGCTTGTG TGGTTGTTTA 300
CATATTATCT CTCTTCTTGC AGGCTCTTAA TTCCATGGTT AGTTCCCCAA CTAAACTGTA 360
AACTTTTATG ATTGTGAGTT TCCTTTATTC TCCTAAAACC CTTCACAATA TTACATATGA 420 ACTGTAGACA GTCTATACAA GTACTGACTA TGCTTTGTTT AG 462
(2) INFORMATION FOR SEQ ID NO:7 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 124 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:7 :
GTGGATGAAA TCTCTCGGCG TTTAATTTGG GAAAAAAACC TGAAGTATAT TTCCATCCAT 60 AACCTTGAGG CTTCTCTTGG TGTCCATACA TATGAACTGG CTATGAACCA CCTGGGGGGA 120 CATG 124
(2) INFORMATION FOR SEQ ID NO: 8 :
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 85 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 8:
GCAAGTATAG CTTCAGCTCC TGTCCCACCT GCACCATTTG CTTTAGTTCC CTGCTGATGC 60 CTGGCCTCTT TCTTCTTTGT CTTAG 85 (2) INFORMATION FOR SEQ ID NO:9:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 156 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9 :
ACCAGTGAAG AGGTGGTTCA GAAGATGACT GGACTCAAAG TACCCCTGTC TCATTCCCGC 60 AGTAATGACA CCCTTTATAT CCCAGAATGG GAAGGTAGAG CCCCAGACTC TGTCGACTAT 120 CGAAAGAAAG GATATGTTAC TCCTGTCAAA AATCAG 156
(2) INFORMATION FOR SEQ ID NO: 10:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1624 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 10:
GTACTCTCCT TTCTTCTGGG TGTGCATATG TAATCTGGCA TGACCTTTTC CTTTTTCTGC 60
TGCTTTGTTC TTGAGGTGAA AGGGCACCAG GAAAAGAGGG CAAGGAATTA AGGTACATCT 120
CCCCATTCCC ATTCTGTTAT TTAACCTCAT TTGTTTCTGT ACATTTGGGT TGTTTCTGGT 180
TTTTCTTTTT CTTTTCCCTT TTTTTTTTTT TTTTTTTTTT GAGATAGAGT CTCACTCTGT 240
CGCCCAGGAT GGAGTGCAGT GGTGCAATCT TGGCTCACTG CAACCTACAC CTCCCGGGTT 300
CAAGCGATTC TCCTGCCTCA GCCTCCTGAG TAGCTGAGAT TACAGGCACG CGCCACTACG 360
CCTGGCTAAT TTTTCTATTT TTATAGAGAT GCGTTTTCAC CATGTTGGCC AGGCTGGTCT 420
TGAACTGACC TCAGGTGATC CACCTGCCTC AGCCTCCCAA AGTGCTGGGA TTAGAGTCAT 480 GAGCCATCGC GGCCTGGTTT TTCTTTATTA CAAATAGTGT TGCAATAAGC ACCCTTGTGC 540
ATATGTTTTT GTGCACATGT ACAAATATTT ATGCAAAATA AGTCCTAAAA TTGGAATTGT 600
TAGGTCACAA ATAATCCTTT CCCCCCCCCC AAATTTTTTT TTTTTTTTTG AGACAGCGTC 660
TCTGTCACCC AGGCTGGAGT CCAGTGGCGC AATCATGGCT CACTGCAGCC TCAACGTCTC 720
AGGCTCAAGT GATTCTCCAA CCTCAGCCTC CCTAGTAGCT GGGAATTAGA AGCACATGCC 780
ACCACACCCA GCTAATTTTA AAAAATTTTT TGTTAGAGAC AGGGTTTTGC CATGCTACCC 840
AAGCTGGTCT CAAATTCCTG GGCTCAAGCA ATCTGCCCGC TTCGGCCTCC CAAAGTGCTA 900
GGATTACAGA CATGAGCCAC CATGCCCAGC CCAAAAAAGT TTTTGCAATC TTACATTCTT 960
ACTAGCATGA GAATGTCAGT TTTTTCACAA CCCAAACAAC ACAGGATTGT ATCAGCAAGA 1020
TAAACAATTG ATTTAACGTT CATTTAACAA ACACTTTTTG ACCCCCAGAA CCTACCAGAT 1080
GCAGTGTTAG GCAGCAGAGA CTCAAGATGA CTAAGACACA ACCTGTGTCC TCAGGAAATC 1140
TCAATCTAAA AAAATAGAAC AGGAAAGAAA GAAAAATCTA CAATCTAGCT GCACAAACAA 1200
TAATAGCTAA TACTTTTTGA GATTTTATTG TTTGTCAGGA ACTTCTTAAC TCTTTACATG 1260
AGTTTAAATA TTTAATCCCT TATAACAATA TTTTATGCAT AGAGAAACTG AGACACAGGC 1320
AAATTTAGTA ACTTACCCGG GGTCACATAG CTACTGGGTG GCAAAGTCAG GGTTAGCTCC 1380
CAGGACAAAT GCCTCCACAG CTGGTACTGT GCTCTGCTTT ACTGTAGCTA ATAGTAAAAA 1440
TGGTAGCAAA AATCAATAGC AGTAGAACAG TGCAACAGAT ATTAAGCGGA AGAGGAAGAC 1500
TCACAACAAT GACAACATTT GTGCTGAAAT TTTTAAGAAC ACATGGAATT TCCTTCAGCC 1560
GGGTAGAGAG AAGATATAGA AATGTAAACA CCAAAGATTC ATAGTTTCTC TGTATCCCTT 1620
TCAG 1624
(2) INFORMATION FOR SEQ ID NO: 11:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 218 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 11:
GGTCAGTGTG GTTCCTGTTG GGCTTTTAGC TCTGTGGGTG CCCTGGAGGG CCAACTCAAG 60
AAGAAAACTG GCAAACTCTT AAATCTGAGT CCCCAGAACC TAGTGGATTG TGTGTCTGAG 120
AATGATGGCT GTGGAGGGGC TACATGACCA ATGCCTTCCA ATATGTGCAG AAGAACCGGG 180
GTATTGACTC TGAAGATGCC TACCCATATG TGGGACAG 218 (2) INFORMATION FOR SEQ ID NO: 12:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 4878 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 12:
GTGAGATTGC TCCACACAAT TATACAGCTC TGTTGGCTCC TCCTCCCCAG CATGATGTTT 60
TGTACTGGAA ACAATTCCAG AAATACTGTT TTCTGTTATC CTATCCTGCT TTCTTGATGG 120
AATAATTTCC CACAGAAGGC CAAGAAGATT TCCACAATCT GGGGGAATTT AGGGAGCTTA 180
AGCTACTATA GCTCCTATTT GCATCTCTGC CATGGAGAGA AAACAGAGGC TAGGCTACCT 240
ACCCCATAGA CTTCCGAGCT GGGTTCTATA ACCCTCTGCT CAATTCCTCA CTCCCACAAC 300
AAACCCACAA ACCCACCATG CTATTTTCAC AAATTGTGTG GCTTTATTTT ATATGATCTC 360
AGTGTGAGTT TTCAGAACAT TTCAGCAAAT TATGTAAGTT TACATGCTAA CATCTATAAA 420
ATGAGAGAAA AAACAAGTTG CTTCATATAA GAGATAAGGG ATTAACTCAG TTCCTCCTGC 480
ATGATCCTCT AGTCATAGGA AGGAAATCAT ATCTGAAAGG GAGGCAACCT GAGGGGTTTT 540
TTATACACAT AGGGCTGGGT CTGATAGACA ATATAATGTA GGGCCTTCAC AACAGAAACC 600
TCTGAAACAG GGACAGCAAG TTTGAGAATA AAAATGATGG CTACTGTGTT CTAAGCCGTG 660
TCCTTAGTGC ATTTTTTCTT TTTCTTTTTT TCATTTAATC TCATAACAAC TCTGTTAGGT 720
AGACTTATCT TGAATGTATA GGTGAGGAAA TGGACACTTA AGGAGATAAG ACAGTATAAT 780
TCATACCACT AGTATGTAAC AATGTAAGAT GTATCTACCA GGGATGTTTA TCTTCTGCAA 840
ACATTCCTAG GTATATCTCC CATGCACATG TGCAAGAATT TCTTACTAGG ATATAATGCC 900
TTGGAACTGA ATTGTCTGGG TCTTAGGGTA TGTCTGTCTT CACTTTACTA CACAATGTCA 960
AATTGTTTGC CAAAATATTT GGAAAAATTT ATACCTGCAA TGTGTAAGAA ATCCCCTTCA 1020
ATCACCTTTT TATCAGTATG TTTATCTGGC CATTTGCATT TCTTCTTCAG TGAATTAACT 1080
GTTTTTATCT CTTGCTCATT TGTTTTTCTT TTTATTTTTT TGAAATAGGG TCTTACTCTG 1140
TTGCCCAAGC TGGAGTGTGG TGAACAGTCA TAGCTCACTG CAGCCTCCAC TTCCGGGCTC 1200
AAGCAATCCT CTCGCCTCAG CCTCCCAAAT AGCTAGGATA TAGGTGCATG CCATCATGCC 1260
CACCAATTTC AAAAAACCTT TGAAATTTTT TTTTGTAGAG GCTAGGCATG GTGGCTCATG 1320
CCTGTAATCC CAGCACTTTG GGAAGCTGAG GTGGGAGGAT CGCTTGAGCC CAGCACTTTG 1380
GGAAGCTGAG GTGGGAGGAT CGCTTGAGCC CAGGAATTGG AGGTCGGCCT GATACAACAT 1440
AGCAAGACCT CATCTCTACA GAAAAAATTT TTAAAAGTAG CCAGGTATGA TGGCGTGCAT 1500
AGTTCTAGCT ACTCCGGAAG CTGGTTGGGA GGACAACTTG AGCCTGGGAG TTCAAGGCTG 1560
CTGTGAACTG TGATCATGTC ACTGCTCTCT AACCTGGGTG ACAGAGTGAG ACCCTGTCCC 1620
CAAAAAACAA CAACCGTTTT TTTTTGGTAG AGACATTGTC TCGCTATGTT GCCAAGGCTA 1680 GTCTCAAACT CCTGGGCTCA AGCAATCCTC CCACCTCCCC AAAGTGCTGG GATTTATAGA 1740
TGTAAGCCAC CATGCCTGGC CTACCCTTTT TTTTTTTTTT TGAAATGGAG TTTTGCTTTT 1800
GTCACCTAGG CTTGAGTGCA GTGGCGCGAT CTTGGCTCAC TGCAACCTCC ACCTCCTGGA 1860
TTCAAGCAAT TCTCCTGCCT CAGCCTCCTG AGTAGCTGGG ATTATAGGCA CCCGCAACCA 1920
CGCCCGGCTA GTTTTTGTAT TTTTAGTACA GACAGGGTTT CACCATGTTG GCCAGGCTGG 1980
TCTTGAACCC CTGACCTCAG GTGGTCCGCC CGCCTCGGCC TCCCAAAGTG CTGGGATTAC 2040
AGGTGTGAGC CACCATGCCC CACCCCTTAC TCATTTTTAA TTGGATTGTT TTTTCTCTTT 2100
CTTAGCGATT CTTAAAAGTT TAAAGAGAAT ATTTGGATAC AATACTATGT ATTTAAAAGT 2160
TGAGGTCTGT CTTTCCATTC TTTTTATGAT GTCTTTCAAT CTACAAAAGT TAATTTTAAT 2220
AGCCTGGCGC CGGTGGATCT CGCTTATTAT CCCCTCACTT TGGGAAGCTG AGATGGGTGG 2280
ATCACAATGT CACGAGATCT TGACCATCCT TCCTGGCGCG GTGGCTGCTA ATGGAAGCGG 2340
AACACGTATA AAGCCAGTCC GCACAAACGG TGCTGACCCC GGATGAATGT CTGCTACTGG 2400
GCTATCTGGA CAAGGGAAAA CTCAAGCGCA AAGATAAAGC AGGTAGCTTG CAGTGGGCTT 2460
ACATGGCGAT AGCTAGACTG GGCGGTTTTA TGGACAGCAT GCCAACCGGA ATTGCCATCT 2520
GGGGCGCCCT CTGGTAAGGT TGGGAAACCC TGCAAAGTAA ACTGGATGGC TTTCTTGCCG 2580
CCAAGGATCT GATGGCGCAG GGGATCAAGA TCTGATCAAG AGACAGGATG AGGATCGTTT 2640
CGCATGATTG AACAAGATGG ATTGCACGCA GGTTCTCCGG CCGCTTGGGT GGAGAGGCTA 2700
TTCGGCTATG ACTGGGCACA ACAGACAATC GGCTGCTCTG ATGCCGCCGT GTTCCGGCTG 2760
TCAGCGCAGG GGCGCCCGGT TCTTTTTGTC AAGACCGACC TGTCCGGTGC CCTGAATGAA 2820
CTGCAGGACG AGGCAGCGCG GCTATCGTGG CTGGCCACGA CGGGCGTTCC TTGCGCAGCT 2880
GTGCTCGACG TTGTCACTGA AGCGGGAAGG GACTGGCTGC TATTGGGCGA AGTGCCGGGG 2940
CAGGATCTCC TGTCATCCCA CCTTGCTCCT GCCGAGAAAG TATCCATCAT GGCTGATGCN 3000
ACTGCGTTTC AAAAAAAAAA AAAGTTAATT TTAATATAGT AAAATTAGTA AAAGGATTAA 3060
TTTTCCCTTT GCAATTTTTG TAATGTGTTT TATTCGTTTA TGAATGGAGA AAGGTAAGAA 3120
AAAATAAAAT TTAAAAAAGA AGAGATGTGG CCAGGTACGG TGGCTCACAC CTATAATCCC 3180
AGTAGTTTGG GAGGCTGAGG CAGGCAGATC ACTTGAGGTC AGGAGTTTGA GACCAGCTGG 3240
GATAACATGG TGAAACCCCA TCTCTACTAA AAATACAAAA ATTAGCCAGG TGTGATTGCG 3300
CACGCTTGTA ATCCCAGCAG GCTGAGGCAG GAGAATTGCT CGAACTCAGG AGGCAGAGGT 3360
TGCAGTGAGC CAAGATCATG CCATTGCACT CCAGCCTGGG TAACAGAGAC TCTGTTTCAA 3420
AAAATAAAAA GATAAAAAGG GAAGAGATCT GATAGGGCGC CCAGAAAAAC ATTTTAAAGG 3480
GGATGGTATT ATAAGTTTGT TCCCAGCATA ATGCCAGGTT ATTTCTGACT TTAAAGTATC 3540
ATCACATAAT ATCTTTTTGA GTCAATTTCC AAGATATTCT GTTTCACTTG TAATTCTGTG 3600
TAATTTTTGG CACCAGGAGG CATCAGGGAT TTGGAGCACA TGGCAGAAAC AAAGGCATCT 3660
TGAAAAATAT CAAGGCAGTA GACCACTGTA ATCTTAAAAT GGCATATCAA ATGCTGCTAT 3720
TGCTGTTAAT ATTTAGATAA TGTTAGATAA TGTATTTTTT TAGAGGGTAT CTCACTATCT 3780
TGCACAGGCT GGAGTAGAGT GGCTATTCAC AGCATGATCA CAGTACACTA AAGGCTCAAA 3840
CTCCTGGGCA CAAACAATCC TCCTGCCTCA GCCTGCTGAG TAGTAGATAA TAAGTTCTTG 3900
TGGATGCAAC CTTAGGGTTC TGAAGGGGTA GTCTGTAGGA AAATGAATTG CTGAAAAGAA 3960
TACACCACCT TAACATGGGC TATTATTCGA TTCCATAATT GTGGCTTGCC AATGAAACAT 4020
TGCTAACTAC CTGTAAAATA TAGTGTTGGA AGTCATAGGC TAAATTGCTA AGTTCTTTAA 4080
TCTATTTTAG TGTCTTGTTA TGTACTTTTA TATTTTGTCT TTGATGAGAG CACAAGGATC 4140
ACACCAGTTC CCCTGATATA GGTGCAGAGG GCCCAGGTCT TCCCTCTAGC TAAGCCTTGG 4200
CCTTGGCCTC CTACCCACAC AGCAGCTGGT GCCTTCCTGC CCCCTGAGGC TAATACATAC 4260
TATGTGGCCA GAAGATGGTT TATGCTTTTT AAAAAAATCT TATTTCAGAA ATCTTTCCCT 4320 ACTGTTTTCC TCCCACATTT ATGTCTTAAA ACACCTGTAG GGGATTTTTT TTTTTTTTTT 4380
TTTTTTGAGA TGGAGTCTCG CTCTCGCCCA GGCTGGAGTG CAATGGCGCG ATCTTGGCTC 4440
ACTGCAAGGT CTGCCTCCCA GGTTCACGCC ATTCTCCTGC CTCAGCCTCC CCAGTAGCTG 4500
GGACTACAGG CGCCCGCTAC CACGCCTGGC TAATTTTTTT GCATTTTTAG TAGAGACAGG 4560
GTTTCACTGT GTTAGCCAGG ATGGTATAGA TCTCTGACCT CGTGATCCAC CTTTCTTCAG 4620
CCTTCCAAAG TGCTGGGATT AACAGGCATG GAGCCCCACC GCACTGGCCT GTAGTTGGTT 4680
TTTATGTGTG GTGGAAGGCG GGAATCCTCT TTTCATATTC GTTTTTGTGA GGAAGAACAG 4740
ACCCTCTTTA GAAGCCCTAG ACTGCTGCCT CTGTTAGTTC ACTGGCATCA CTCAAAATAT 4800
TGGTTGAGTT TCTTACTCAC TGACTCATTG CCTATTGCTT TGTCCTAGTC CTATTACAAT 4860
CTTGTTTCTT CCAGCCAG 4878
(2) INFORMATION FOR SEQ ID NO: 13:
(l) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 166 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:13:
GAAGAGAGTT GTATGTACAA CCCAACAGGC AAGGCAGCTA AATGCAGAGG GTACAGAGAG 60 ATCCCCGAGG GGAATGAGAA AGCCCTGAAG AGGGCAGTGG CCCGAGTGGG ACCTGTCTCT 120 GTGGCCATTG ATGCAAGCCT GACCTCCTTC CAGTTTTACA GCAAAG 166
(2) INFORMATION FOR SEQ ID NO: 14:
(I) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 270 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(II) MOLECULE TYPE: cDNA (m) HYPOTHETICAL: NO (iv) ANTISENSE: NO
(v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE: (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 14:
GTAAGAAGCT GCTGATCCTA TACAGCACTG TCTTTTATGA TACAAACTTG ATGGTTTCTC 60
GAAGGACCTT GGGTATTTTC AGTACTTAGT TTTTGTATTC ACATGGAGGT GGCCAGAGAG 120
AAATTAACAA CTGCTGCAGT ATGGAGCAGC ATCTCTGTGG TAAACCCTCC TGACACGGAT 180
GGAATTCTTC AAACAGTCTC CTAGACTGGG AGATCCCACA GGGTGACCCT TGGATTGCAT 240
AGAGCCTCAC GCTGGTAGTT TGTATTCTAG 270
(2) INFORMATION FOR SEQ ID NO: 15:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 106 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 15:
GTGTGTATTA TGATGAAAGC TGCAATAGCG ATAATCTGAA CCATGCGGTT TTGGCAGTGG 60 GATATGGAAT CCAGAAGGGA AACAAGCACT GGATAATTAA AAACAG 106
(2) INFORMATION FOR SEQ ID NO: 16:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 2270 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE: (xi) SEQUENCE DESCRIPTION: SEQ ID NO:16:
GTAATGATGG GAACACTACT TTTGTTATTC AGTCACCCTT TTAACACTCA ACCTCACCTC 60
CAGCTTCCCG ATATTCCTTT CTCTGTCCCA AATCAAGAAA AAATTATCTC AGAGTTCTCA 120
CTTCTATCTT CTCAGTCAGA GGCTCTTAAT TCTCAGTCTG ACACTTAATG GCCAGTGTGT 180
TAGTCCATTT TGCATTGCCA CAAAAGAATA CCCGAGACTG GGTAGTTTAT AAAGAAACGA 240
GGTTTGTTTG GCTATACAAA GCGTGGCACT AGTATCTGCT CAGCCTCTGA TGAGGCCTCA 300
GAGCTTTTAC TCATGGCAGA AGGCAAAAGA GGGAGCAGGC ATGTCACATA GTGAGAGAGG 360
GAGCAAGAGA GAGAGGGAGG TGCCGACTCT TTAAAGAACC AGCTCTTGCA TGAACTAATA 420
GAGTGAGAAC TCACTCATCA CCAAGGCGAT GGCACCAAGC CATTCCATGA GGAATCCACT 480
CTCATAACCC AAACACCTCC CACTATGCCC CACCTCCCAC ATTGGGGATC ACATTTCAGC 540
ATGAGACTGG GAGGGGACAC ACATCCAAAC CATATCCGCC AGACAATAGT GCTCAATTAT 600
GTGCTGGGCA GATGCTCCCT GTGTGCAAGG TGCTTAGTGA CATACATAAA CCAACGAGCA 660
GATGACACCT TCAGTGAGCT CAGAGCCCAA TAAGACAGAC CTAACTAACC ATGAGATAAA 720
GCAGTACAAA GAACCAGCAG GAGCTTTGGA ATTACGTATT TTTACTTTCT TTTGTCTCTA 780
ATGTGATCAG TTTCTTAGAT GGTTTCCATT AGCAATCTGT CTTTAACAGT AGGGGAGCAG 840
CGTTAAAGGT TTAATATTCC TTTTGAACAG TTTTTTTCCT TCAAAATACA CTTAAGATAC 900
ACGTATATAA GAACTTGCCA AAGATTGTGA AGAGAAACAT TTTTTAGAAA TAAGATATAA 960
ACAAAAAAAG TTAGTGTTAC TTTCCTATGT TGGGGAACAA AGAAAACTCC AGGGTACCTT 1020
GCTTCCCATT TCTCTTTAGC ACCTTGTGAC TTTTGGGGAG GGGCAGATTG ATAACAATTA 1080
TAGTTTTCCT TTCCTGGCTG ATCACCATTA ACCTGGCAGC AGCACTGGCT AAATCTCCTG 1140
TCCTTAGTGC CCTCCAAGGA GCAGGAGCCC TAGACTCTGG GTCGCTGACA GACTCACGCA 1200
GTGGTGTTGT TCAAACCTGA AGCAACTTTT TATATCACAG TTCCAACTCA AGGTGAACCT 1260
GAGCATCTTC CCAAGTCTCC CACAGCTTCT GTCCTGTGTT GTCCCTTCTC TTGACTCCCA 1320
GGTCCAAGCA CTTACCCTGT TCTTTCATGA TCAGGTACCA TGTGTGGAGA TAGCTTCCAA 1380
GAGAGCTGGG AGGAAGAAAG GACACACCCG GGCAGGATCA GGAACACTGG GGGCCCCTGG 1440
AGAAGGGGAG AGTGGGGGAG GGTACAGGTT TTAAATAAAA TGTGTTGGTA ATTAGAGAAT 1500
TGCTGGTTGG GGAAAGAGGT CTGAAAACAA TTCAGGAAGA TAAACAAGAC AATCTCTCCT 1560
CTCTCCTCTT TCTCACGTCG TCTCTCTTGT CTTCTAGTCT CGCTACTCAT TTCCTTAGTA 1620
ATCTCATCCA CTCTCATAGT TTCATCCATC TCTCCTATGG GGTTTACCCC CAAATCAAGA 1680
TCACCAGCTT CAGCCTCCTT CTTATGCTCT AAACTCACAT TTTCAAGATT AATATTCCCC 1740
AAATACAGCT CTGATCATAT CACTCTCCCA CTCAAAATCC CTCACTGGCT CCTCACGATG 1800
ATGGGTCACA GAGTAAAGGT GAAGCTTTTT AACCTTGCAG TAAAGGTAAT TCAACCTGAT 1860
CTCAATCTGC CTTTCCAGAC ATCTCTCCCA CTACACCCTG TTAGGCACAC TGCTTTTCAG 1920
CTACATGATC CTAACAGTGC CCCACACTTT CCTGCCTCTG TTGTTCATTT CACACCCTTC 1980
CACTGGCATC CCCTTCCCAC AGGTCGAAAT TCTACTTAGC CTTTTGGCTC AGCTCAAATG 2040
CCACCTCTTA CATCAAGCCT CTAAGATTCT CTTGATCAGA AGGAATCTTT CCCTCCTTTG 2100
ATACCTACAG TATTATGCCT TCTCCCTATT TCTTGACTTT AAACTCTTTA AAGTTAAAAA 2160
ACATCATATT CATTTTTGTG TACCATCAGT ACCTCGCACA ATACTCAGTA AATATTTTAA 2220
TGAATAAATA AACTGAGAGT ACTAAGTATT TTTCTTGATT GGTCTTACAG 2270 (2) INFORMATION FOR SEQ ID NO: 17:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 97 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:17:
CTGGGGAGAA AACTGGGGAA ACAAAGGATA TATCCTCATG GCTCGAAATA AGAACAACGC 60 CTGTGGCATT GCCAACCTGG CCAGCTTCCC CAAGATG 97
(2) INFORMATION FOR SEQ ID NO:18:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 595 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:18:
TGACTCCAGC CAGCCCAAAT CCATCCTGCT CTTCCATTTC CTTCCACGAT GGTGCAGTGT 60
AACGATGCAC TTTGGAAGGG TGAAGGTGTG CTATTTTTGA AGCAGATGTG GTGATACTGA 120
GATTGTCTGT TCAGTTTCCC CATTTGTTTG TGCTTCAAAT GATCCTTCCT ACTTTGCTTC 180
TCTCCACCCA TGACCTTTTT CCACTGTGGC CATCAGGACT TTCCCTGACA GCTGTGTACT 240
CTTAGGCTAA GAGATGTGAC TACAGCCTGC CCCTGACTGT GTTGTCCCAG GGCTGATGCT 300
GACAGGTACA GGCTGGAGAT TTTCACTAGG TTAGATTCTC ATTCACGGGA CTAGTTAGCT 360
TTAAGCACCC TAGAGGACTA GGGTAATCTG ACTTCTCACT TCCTAAGTTC CCTTCTATAT 420
CCTCAAGGTA GAAATGTCTA TGTTTTCTAC TCCAATTCAT AAATCTATTC ATAAGTCTTT 480
GGTACAAGTT TACATGATAA AAAGAAATGT GATTTGTCTT CCCTTCTTTG CACTTTTGAA 540 ATAAAGTATT TATCTCCTGT CTACAGTTTA ATAAATAGCA TCTAGTACAC ATTCA 595
(2) INFORMATION FOR SEQ ID NO: 19:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 459 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: cDNA (iii) HYPOTHETICAL: NO (iv) ANTISENSE: NO (v) FRAGMENT TYPE: (vi) ORIGINAL SOURCE:
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 19:
TTTTGTGTTG GATACTGTGT TAGGTGCTGG AGGAAAAAAG ATGAATAGAA CATCTTCTAT 60
GTACTTGATG CGCTCACAGT CTGGTTGTAG AGACTGTCAC ATAAACATTT CATCCCAATT 120
CATTTATTTG TTCATTCCTT CAGCCAATAT ATATTGAGTT CTTACTCTGT GCCAAGAACT 180
GTACTACATT TCTGGGATTA AGTGGATATA AGGAGATCTC AGTGTTTAAT CTGCCTGAGG 240
GGAGACTAAA TTAAGTGACA TGGAAACTTG GGTCTTGAAA AACATTTTAA GGTTATTTTT 300
TCTTTTCTCT CTCTCTCGCT CTGTCTTTCT CTCTCTTTCG TCAGGGTCTC CCTCTGTTGC 360
CCAGGCTGGA GTCAGTGGCA CTCATAGCTC ACTGCAGCCT TGATCTCCTG GGCTCAAGAG 420
TTCTTCCCAC CTCAGTCTCC TAAGTAGCTT GGACTACGG 459
(2) INFORMATION FOR SEQ ID NO:20:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 329 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: peptide
(iii) HYPOTHETICAL: NO
(iv) ANTISENSE: NO
(v) FRAGMENT TYPE: N-terminal
(vi) ORIGINAL SOURCE: (xi) SEQUENCE DESCRIPTION: SEQ ID NO:20:
Met Trp Gly Leu Lys Val Leu Leu Leu Pro Val Val Ser Phe Ala Leu
1 5 10 15
Tyr Pro Glu Glu lie Leu Asp Thr His Trp Glu Leu Trp Lys Lys Thr
20 25 30
His Arg Lys Gin Tyr Asn Asn Lys Val Asp Glu lie Ser Arg Arg Leu
35 40 45 lie Trp Glu Lys Asn Leu Lys Tyr lie Ser lie His Asn Leu Glu Ala
50 55 60
Ser Leu Gly Val His Thr Tyr Glu Leu Ala Met Asn His Leu Gly Asp 65 70 75 80
Met Thr Ser Glu Glu Val Val Gin Lys Met Thr Gly Leu Lys Val Pro
85 90 95
Leu Ser His Ser Arg Ser Asn Asp Thr Leu Tyr lie Pro Glu Trp Glu
100 105 110
Gly Arg Ala Pro Asp Ser Val Asp Tyr Arg Lys Lys Gly Tyr Val Thr
115 120 125
Pro Val Lys Asn Gin Gly Gin Cys Gly Ser Cys Trp Ala Phe Ser Ser
130 135 140
Val Gly Ala Leu Glu Gly Gin Leu Lys Lys Lys Thr Gly Lys Leu Leu 145 150 155 160
Asn Leu Ser Pro Gin Asn Leu Val Asp Cys Val Ser Glu Asn Asp Gly
165 170 175
Cys Gly Gly Gly Tyr Met Thr Asn Ala Phe Gin Tyr Val Gin Lys Asn
180 185 190
Arg Gly lie Asp Ser Glu Asp Ala Tyr Pro Tyr Val Gly Gin Glu Glu
195 200 205
Ser Cys Met Tyr Asn Pro Thr Gly Lys Ala Ala Lys Cys Arg Gly Tyr
210 215 220
Arg Glu lie Pro Glu Gly Asn Glu Lys Ala Leu Lys Arg Ala Val Ala 225 230 235 240
Arg Val Gly Pro Val Ser Val Ala lie Asp Ala Ser Leu Thr Ser Phe
245 250 255
Gin Phe Tyr Ser Lys Gly Val Tyr Tyr Asp Glu Ser Cys Asn Ser Asp
260 265 270
Asn Leu Asn His Ala Val Leu Ala Val Gly Tyr Gly lie Gin Lys Gly
275 280 285
Asn Lys His Trp lie lie Lys Asn Ser Trp Gly Glu Asn Trp Gly Asn
290 295 300
Lys Gly Tyr lie Leu Met Ala Arg Asn Lys Aεn Asn Ala Cys Gly lie 305 310 315 320
Ala Asn Leu Ala Ser Phe Pro Lys Met 325

Claims

What is claimed is:
1. An isolated polynucleotide comprising a region selected from the group consisting of: a sequence at least 80% identical to the sequence in SEQ ID NO a sequence at least 85% identical to the sequence in SEQ ID NO a sequence at least 90% identical to the sequence in SEQ ID NO a sequence at least 95% identical to the sequence in SEQ ID NO and a sequence at least 97% identical to the sequence in SEQ ID NO
2. An isolated polynucleotide according to claim 1, wherein said region is a genomic DNA or a cDNA.
3. An isolated polynucleotide comprising cathepsin K enhancer or promoter.
4. The isolated polynucleotide of claim 3 having the sequence in SEQ ID NO: 1.
5. An isolated polynucleotide comprising cathepsin K polyadenylation region.
6. The isolated polynucleotide of claim 5 having the sequence in SEQ ID NO: 1.
7. An isolated polynucleotide comprising a cathepsin K intron.
8. The isolated polynucleotide of claim 7 having the sequence in SEQ ID NO: 1.
9. An isolated polynucleotide comprising a sequence selected from the group consisting of: intron 1, 2, 3, 4, 5, 6 and 7.
10. An isolated polypeptide encoded by a polynucleotide comprising a sequence selected from the group consisting of:
intron 1, 2, 3, 4, 5, 6 and 7.
1 1. An isolated polynucleotide comprising a cathepsin K exon.
12. The isolated polynucleotide of claim 1 1 having the sequence in SEQ ID NO: 1.
13. An isolated polynucleotide comprising a sequence selected from the group consisting of:
exon 1, 2, 3, 4, 5, 6, 7 and 8.
14. An isolated polypeptide encoded by a polynucleotide comprising a sequence selected from the group consisting of:
exon 1, 2, 3, 4, 5, 6, 7, and 8.
15. An isolated polynucleotide comprising an exon-exon pairs selected from the group consisting of:
1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 2-4, 2-5, 2-6, 2-7, 2-8, 3-4, 3-5, 3-6, 3-7, 3-8, 4-5, 4-6, 4-7, 4-8, 5-7, 5-8 and 6-8.
16. An isolated polypeptide encoded by a polynucleotide comprising an exon-exon pairs selected from the group consisting of:
1-3, 1-4, 1-5, 1-6, 1-7, 1-8, 2-4, 2-5, 2-6, 2-7, 2-8, 3-4, 3-5, 3-6, 3-7, 3-8, 4-5, 4-6, 4-7, 4-8, 5-7, 5-8 and 6-8.
17. An isolated polynucleotide comprising a region selected from the group consisting of: a sequence at least 80% identical to the human cDNA in ATCC Deposit No.: 98035, a sequence at least 85% identical to the human cDNA in ATCC Deposit No.: 98035, a sequence at least 90% identical to the human cDNA in ATCC Deposit No.: 98035, a sequence at least 95% identical to the human cDNA in ATCC Deposit No.:
98035, and a sequence at least 97% identical to the human cDNA in ATCC Deposit No.: 98035.
18. An isolated polynucleotide comprising a member selected from the group consisting of:
(a) a polynucleotide having at least a 70% identity to a polynucleotide encoding a polypeptide comprising an amino acid sequence set forth in SEQ ID NO:
2; (b) a polynucleotide having at least a 70% identity to a polynucleotide encoding a polypeptide comprising amino acid 1 to amino acid 214 forth in SEQ ID
NO: 2
(c) a polynucleotide which hybridizes to and is at least 70% complementary to the polynucleotide of (a) or (b); and (d) a polynucleotide fragment of the polynucleotide of (a), (b) or (c) wherein said fragment comprises at least 30 consecutive bases of SEQ ID NO: 1.
19. An isolated polynucleotide according to claim 1, wherein said region is the region encoding cathepsin K in the human cDNA insert in ATCC Deposit No.: 98035.
20. An expression vector comprising cis-acting control elements effective for expression in a host cell of an operatively linked polynucleotide according to claim 1.
21. An expression vector according to claim 20, wherein said control elements are effective for inducible expression of said polynucleotide in said host cell.
22. An expression vector comprising cis-acting control elements effective for expression in a host cell of an operatively linked polynucleotide according to claim 18.
23. A host cell having expressibly incorporated therein an expression vector according to claim 22.
24. A host cell having expressibly incoφorated therein an expression vector according to claim 18.
25. A process for making a polypeptide, comprising the step of expressing in a host cell a polynucleotide according to claim 1.
26. A process for making a polypeptide, comprising the step of expressing in a host cell a polynucleotide according to claim 18.
27. A polypeptide of 15 or more amino acids identical in sequence to a continuous region of the amino acid sequence of SEQ ID NO: 20 .
28. A polypeptide of 15 or more amino acids identical in sequence to a continuous region of the amino acid sequence of the polypeptide encoded by the human cDNA in ATCC Deposit No. 98035.
29. A polypeptide of 50 or more amino acids identical in sequence to a continuous region of the amino acid sequence of SEQ ID NO: 20.
30. A polynucleotide of 25 or more nucleotides identical in sequence to a continuous region of a polynucleotide at least 90% identical in sequence to the polynucleotide of SEQ ID NO: 1.
31. A polynucleotide according to claim 23 of 50 or more nucleotides.
32. A polynucleotide according to claim 31 of 75 or more nucleotides.
33. A host cell genetically engineered with the vector of Claim 20.
34. A process for producing a polypeptide comprising: expressing from the host cell of Claim 33 the polypeptide encoded by said DNA.
35. A process for producing cells capable of expressing a polypeptide comprising genetically engineering cells with the vector of Claim 20.
36. A method for determining a cathepsin K-encoding polynucleotide in a sample, comprising the steps of: hybridizing to a sample a probe specific for said polynucleotide under conditions effective for said probe to hybridize specifically to said polynucleotide and determining the hybridization of said probe to polynucleotides in said sample, wherein said probe comprises its sequence a region of 20 or more base pairs at least 90% identical to the polynucleotide sequence of SEQ ID NO: 1.
37. A method for determining a cathepsin K-encoding polynucleotide in a sample, comprising the steps of: hybridizing to a sample a probe specific for said polynucleotide under conditions effective for said probe to hybridize specifically to said polynucleotide and determining the hybridization of said probe to polynucleotides in said sample, wherein said probe comprises its sequence a region of 20 or more base pairs at least 90% identical to the polynucleotide sequence of the human cDNA insert of ATCC deposit No. 98035.
38. A method for detecting in a sample a polypeptide comprising a region at least 90% identical in sequence to the amino acid sequence of residues 1 through
135 of SEQ ID NO: 1, said method comprising: incubating with a sample a reagent that binds specifically to said polypeptide under conditions effective for specific binding and determining the binding of said reagent to said polypeptide in said sample.
39. A method for diagnosing a disease characterized by aberrant expression of a cathepsin K polynucleotide, comprising: hybridizing a probe specific for a polynucleotide comprising a region at least 90% identical in sequence to an RNA or DNA that encodes amino acids 1-135 in SEQ ID NO: 1 under condition effective for specific hybridization and determining hybridization of said probe to said polynucleotide in said sample.
40. A method for diagnosing a disease characterized by aberrant expression of a cathepsin K polypeptide, comprising: incubating with a sample a reagent that binds specifically to a polypeptide comprising a region at least 90% identical in sequence to the amino acid sequence of residues 1 through 135 of SEQ ID NO: 1 under conditions effective for specific binding, and determining the binding of said reagent to said polypeptide in said sample.
41. A compound which inhibits activation of the polypeptide of claim 13.
42. A method for the treatment of a patient having need of cathepsin K comprising: administering to the patient a therapeutically effective amount of the polypeptide of claim 13.
43. The method of Claim 42 wherein said therapeutically effective amount of the polypeptide is administered by providing to the patient DNA encoding said polypeptide and expressing said polypeptide in vivo.
44. A method for the treatment of a patient having need to inhibit a cathepsin K polypeptide comprising: administering to the patient a therapeutically effective amount of the compound of Claim 41.
45. A process for diagnosing a disease or a susceptibility to a disease related to an under-expression of the polypeptide of claim 13 comprising: determining a mutation in a nucleic acid sequence encoding said polypeptide.
46. A diagnostic process comprising: analyzing for the presence of the polypeptide of claim 13 in a sample derived from a host.
47. A method for identifying compounds which bind to and inhibit activation of the polypeptide of claim 13 comprising: contacting a cell expressing on the surface thereof a receptor for the polypeptide, said receptor being associated with a second component capable of providing a detectable signal in response to the binding of a compound to said receptor, with an analytically detectable cathepsin K polypeptide and a compound under conditions to permit binding to the receptor; and determining whether the compound binds to and inhibits the receptor by detecting the absence of a signal generated from the interaction of the cathepsin K with the receptor.
PCT/US1996/010346 1996-06-14 1996-06-14 Cathepsin k gene WO1997047642A1 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
AU63332/96A AU6333296A (en) 1996-06-14 1996-06-14 Cathepsin k gene
PCT/US1996/010346 WO1997047642A1 (en) 1996-06-14 1996-06-14 Cathepsin k gene
PCT/US1996/014026 WO1997047643A1 (en) 1996-06-14 1996-08-26 Cathepsin k gene
AU69092/96A AU6909296A (en) 1996-06-14 1996-08-26 Cathepsin k gene

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/US1996/010346 WO1997047642A1 (en) 1996-06-14 1996-06-14 Cathepsin k gene

Publications (1)

Publication Number Publication Date
WO1997047642A1 true WO1997047642A1 (en) 1997-12-18

Family

ID=22255330

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US1996/010346 WO1997047642A1 (en) 1996-06-14 1996-06-14 Cathepsin k gene
PCT/US1996/014026 WO1997047643A1 (en) 1996-06-14 1996-08-26 Cathepsin k gene

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/US1996/014026 WO1997047643A1 (en) 1996-06-14 1996-08-26 Cathepsin k gene

Country Status (2)

Country Link
AU (2) AU6333296A (en)
WO (2) WO1997047642A1 (en)

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5998470A (en) * 1995-10-30 1999-12-07 Smithkline Beecham Corporation Protease inhibitors
WO2000068414A2 (en) * 1999-05-05 2000-11-16 Merck Frosst Canada & Co. Whole cell assay for cathepsin k
WO2001000798A2 (en) * 1999-06-25 2001-01-04 Hybritech Incorporated Cloning and expressing of human cathepsin k in mammalian cells
US6387682B1 (en) 1994-03-08 2002-05-14 Human Genome Sciences, Inc. Human osteoclast-derived cathepsin
US6534498B1 (en) 1999-11-10 2003-03-18 Smithkline Beecham Corporation Protease inhibitors
US6544767B1 (en) 1994-10-27 2003-04-08 Axys Pharmaceuticals, Inc. Cathespin O2 protease
US6562842B2 (en) 1995-10-30 2003-05-13 Smithkline Beecham Corporation Protease inhibitors
US6583137B1 (en) 1999-11-10 2003-06-24 Smithkline Beecham Corporation Protease inhibitors
US6596715B1 (en) 1999-11-10 2003-07-22 Smithkline Beecham Corporation Protease inhibitors
US7071184B2 (en) 2000-03-21 2006-07-04 Smithkline Beecham Corporation Protease inhibitors
US7282512B2 (en) 2002-01-17 2007-10-16 Smithkline Beecham Corporation Cycloalkyl ketoamides derivatives useful as cathepsin K inhibitors
US7405209B2 (en) 1998-12-23 2008-07-29 Smithkline Beecham Corporation Protease inhibitors

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5501969A (en) * 1994-03-08 1996-03-26 Human Genome Sciences, Inc. Human osteoclast-derived cathepsin

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5501969A (en) * 1994-03-08 1996-03-26 Human Genome Sciences, Inc. Human osteoclast-derived cathepsin

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BIOCHEM. BIOPHYS. RES. COMM., 05 January 1995, Vol. 206, No. 1, INAOKA et al., "Molecular Cloning of Human cDNA for Cathepsin K: Novel Cysteine Proteinase Predominantly Expressed in Bone", pages 89-96. *
BIOL. CHEM. HOPPE-SEYLER, 1995, Vol. 376, BROMME et al., "Human Cathepsin 02, a Novel Cysteine Protease Highly Expressed in Osteoclastomas and Ovary. Molecular Cloning, Sequencing and Tissue Distribution", pages 379-384. *
FEBS LETTERS, 1995, Vol. 357, SHI et al., "Molecular Cloning of Human Cathepsin 0, a Novel Endoproteinase and Homologue of Rabbit 0C2", pages 129-134. *
J. BIOL. CHEM., 14 January 1994, Vol. 269, No. 2, TEZUKA et al., "Molecular Cloning of Possible Cysteine Proteinase Predominantly Expressed in Osteoclasts", pages 1106-1109. *
J. BIOL. CHEM., 24 May 1996, Vol. 271, No. 21, DRAKE et al., "Cathepsin K, but not Cathepsins B, L, or S, is Abundantly Expressed in Human Osteoclasts", pages 12511-12516. *

Cited By (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6387682B1 (en) 1994-03-08 2002-05-14 Human Genome Sciences, Inc. Human osteoclast-derived cathepsin
US6994978B2 (en) 1994-03-08 2006-02-07 Human Genome Sciences, Inc. Human osteoclast derived cathepsin
US7235371B2 (en) 1994-03-08 2007-06-26 Human Genome Sciences, Inc. Human osteoclast derived cathepsin
US6680375B2 (en) 1994-03-08 2004-01-20 Human Genome Sciences, Inc. Human osteoclast-derived cathepsin
US6475487B2 (en) 1994-03-08 2002-11-05 Human Genome Sciences, Inc. Treatment of a patient by administering an antibody that inhibits cathepsin O
US6423507B1 (en) 1994-03-08 2002-07-23 Human Genome Sciences, Inc. Human osteoclast-derived cathepsin
US6544767B1 (en) 1994-10-27 2003-04-08 Axys Pharmaceuticals, Inc. Cathespin O2 protease
US6232342B1 (en) 1995-10-30 2001-05-15 Smithkline Beecham Corporation Protease inhibitors
US6331542B1 (en) 1995-10-30 2001-12-18 Smithkline Beecham Corporation Protease inhibitors
US6284777B1 (en) 1995-10-30 2001-09-04 Smithkline Beecham Corporation Carbohydrazide protease inhibitors
US5998470A (en) * 1995-10-30 1999-12-07 Smithkline Beecham Corporation Protease inhibitors
US6562842B2 (en) 1995-10-30 2003-05-13 Smithkline Beecham Corporation Protease inhibitors
US6057362A (en) * 1995-10-30 2000-05-02 Smithkline Beecham Corporation Protease inhibitors
US6586466B2 (en) 1995-10-30 2003-07-01 Smithkline Beecham Corporation Carbohydrazide-protease inhibitors
US7405209B2 (en) 1998-12-23 2008-07-29 Smithkline Beecham Corporation Protease inhibitors
US6346373B1 (en) 1999-05-05 2002-02-12 Merck Frosst Canada & Co., Whole cell assay for cathepsin K activity
WO2000068414A3 (en) * 1999-05-05 2001-05-17 Merck Frosst Canada Inc Whole cell assay for cathepsin k
WO2000068414A2 (en) * 1999-05-05 2000-11-16 Merck Frosst Canada & Co. Whole cell assay for cathepsin k
WO2001000798A3 (en) * 1999-06-25 2001-08-09 Hybritech Inc Cloning and expressing of human cathepsin k in mammalian cells
WO2001000798A2 (en) * 1999-06-25 2001-01-04 Hybritech Incorporated Cloning and expressing of human cathepsin k in mammalian cells
US6596715B1 (en) 1999-11-10 2003-07-22 Smithkline Beecham Corporation Protease inhibitors
US6583137B1 (en) 1999-11-10 2003-06-24 Smithkline Beecham Corporation Protease inhibitors
US6534498B1 (en) 1999-11-10 2003-03-18 Smithkline Beecham Corporation Protease inhibitors
US7071184B2 (en) 2000-03-21 2006-07-04 Smithkline Beecham Corporation Protease inhibitors
US7563784B2 (en) 2000-03-21 2009-07-21 Smithkline Beecham Corporation Protease inhibitors
US7282512B2 (en) 2002-01-17 2007-10-16 Smithkline Beecham Corporation Cycloalkyl ketoamides derivatives useful as cathepsin K inhibitors

Also Published As

Publication number Publication date
AU6333296A (en) 1998-01-07
AU6909296A (en) 1998-01-07
WO1997047643A1 (en) 1997-12-18

Similar Documents

Publication Publication Date Title
EP0792370B1 (en) Interleukin-1 beta converting enzyme like apoptosis protease-3 and 4
US6391853B1 (en) Human tissue inhibitor of metalloproteinase-4
US6316199B1 (en) Arginase II
US6750034B1 (en) DNA encoding human serine protease D-G
WO1997047642A1 (en) Cathepsin k gene
US6709653B1 (en) Antibodies specific for human inositol monophosphatase H1
US5861298A (en) Cathepsin K gene
US5922546A (en) Human disintegrin metalloprotease KUZ gene
EP0861898A1 (en) Rat Cathepsin K polynucleotide & polypeptide sequence
US6107075A (en) Human elastase IV
CA2360464A1 (en) Human peptidases
US5955339A (en) Human inositol monophosphatase H1
EP0812916A2 (en) Cathepsin k gene
EP0754234A1 (en) DNA ENCODING PRECURSOR OF INTERLEUKIN-1BETA CONVERTING ENZYME-RELATED CYSTEINE PROTEINASE II (ICE rel-II)
US6818429B2 (en) Isolated human protease proteins, nucleic acid molecules encoding human protease proteins, and uses thereof
US20020132331A1 (en) Human DNA Ligase IV
US20040132158A1 (en) Human peptidases
US6835555B1 (en) Interleukin-1 β converting enzyme like apoptosis protease-3 and 4
WO1999002664A1 (en) Human n-arginine dibasic convertase
WO1997033986A1 (en) Arginase ii
US20030211524A1 (en) Isolated human protease proteins, nucleic acid molecules encoding human protease proteins, and uses thereof
MXPA97000151A (en) Elastase iv hum

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AU BB BG BR CA CN CZ EE GE HU IL IS JP KG KP KR LK LR LT LV MD MG MK MN MX NO NZ PL RO SG SI SK TR TT UA US UZ VN AM AZ BY KG KZ MD RU TJ TM

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): KE LS MW SD SZ UG AT BE CH DE DK ES FI FR GB GR IE IT LU MC NL PT SE BF BJ CF CG CI CM GA GN ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: CA

NENP Non-entry into the national phase

Ref country code: JP

Ref document number: 98501547

Format of ref document f/p: F

122 Ep: pct application non-entry in european phase