WO1999050435A1 - Gpi-122, a novel glycophosphatidylinositol-anchored protein - Google Patents

Gpi-122, a novel glycophosphatidylinositol-anchored protein Download PDF

Info

Publication number
WO1999050435A1
WO1999050435A1 PCT/US1999/006459 US9906459W WO9950435A1 WO 1999050435 A1 WO1999050435 A1 WO 1999050435A1 US 9906459 W US9906459 W US 9906459W WO 9950435 A1 WO9950435 A1 WO 9950435A1
Authority
WO
WIPO (PCT)
Prior art keywords
protein
polynucleotide
seq
sequence
amino acid
Prior art date
Application number
PCT/US1999/006459
Other languages
French (fr)
Inventor
Michael R. Bowman
Patricia D. Lohmar
Nancy L. Wood
Original Assignee
Genetics Institute, Inc.
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 Genetics Institute, Inc. filed Critical Genetics Institute, Inc.
Priority to AU33630/99A priority Critical patent/AU3363099A/en
Priority to CA002326000A priority patent/CA2326000A1/en
Priority to EP99915013A priority patent/EP1066398A4/en
Publication of WO1999050435A1 publication Critical patent/WO1999050435A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention provides novel polynucleotides and proteins encoded by such polynucleotides, along with therapeutic, diagnostic and research utilities for these polynucleotides and proteins.
  • the present invention provides a composition comprising an isolated polynucleotide selected from the group consisting of:
  • a polynucleotide encoding a protein comprising a fragment of the amino acid sequence of SEQ ID NO:2 having biological activity, the fragment comprising eight contiguous amino acids of SEQ ID NO:2;
  • a polynucleotide which is an allelic variant of a polynucleotide of
  • polynucleotide that hybridizes under stringent conditions to any one of the polynucleotides specified in (a)-(i) and that has a length that is at least 25% of the length of SEQ ID NO:l.
  • such polynucleotide comprises the nucleotide sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3730; the nucleotide sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3640; the nucleotide sequence of the full-length protein coding sequence of clone GPI-122 deposited under accession number ATCC 98622; or the nucleotide sequence of a mature protein coding sequence of clone GPI-122 deposited under accession number ATCC 98622.
  • the polynucleotide encodes the full-length or a mature protein encoded by the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622.
  • the present invention provides a polynucleotide encoding a protein comprising the amino acid sequence of SEQ ID NO:2 from amino acid 1 to amino acid 1163.
  • the present invention provides a polynucleotide encoding a protein comprising a fragment of the amino acid sequence of SEQ ID NO:2 having biological activity, the fragment preferably comprising eight (more preferably twenty, most preferably thirty) contiguous amino acids of SEQ ID NO:2, or a polynucleotide encoding a protein comprising a fragment of the amino acid sequence of SEQ ID NO:2 having biological activity, the fragment comprising the amino acid sequence from amino acid 591 to amino acid 600 of SEQ ID NO:2.
  • inventions provide the gene corresponding to the cDNA sequence of SEQ ID NO:l.
  • Further embodiments of the invention provide isolated polynucleotides produced according to a process selected from the group consisting of: (a) a process comprising the steps of:
  • the polynucleotide isolated according to the above process comprises a nucleotide sequence corresponding to the cDNA sequence of SEQ ID NO:l, and extending contiguously from a nucleotide sequence corresponding to the 5' end of SEQ ID NO:l to a nucleotide sequence corresponding to the 3' end of SEQ ID NO:l , but excluding the poly(A) tail at the 3' end of SEQ ID NO:l.
  • the polynucleotide isolated according to the above process comprises a nucleotide sequence corresponding to the cDNA sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3730, and extending contiguously from a nucleotide sequence corresponding to the 5' end of said sequence of
  • SEQ ID NO:l from nucleotide 152 to nucleotide 3730, to a nucleotide sequence corresponding to the 3' end of said sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3730.
  • the polynucleotide isolated according to the above process comprises a nucleotide sequence corresponding to the cDNA sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3640, and extending contiguously from a nucleotide sequence corresponding to the 5' end of said sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3640, to a nucleotide sequence corresponding to the 3' end of said sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3640.
  • the present invention provides a composition comprising a protein, wherein said protein comprises an amino acid sequence selected from the group consisting of:
  • protein comprises the amino acid sequence of SEQ ID NO:2 or the amino acid sequence of SEQ ID NO:2 from amino acid 1 to amino acid 1163.
  • the present invention provides a protein comprising a fragment of the amino acid sequence of SEQ ID NO:2 having biological activity, the fragment preferably comprising eight (more preferably twenty, most preferably thirty) contiguous amino acids of SEQ ID NO:2, or a protein comprising a fragment of the amino acid sequence of SEQ ID NO:2 having biological activity, the fragment comprising the amino acid sequence from amino acid 591 to amino acid 600 of SEQ ID NO:2.
  • the polynucleotide is operably linked to an expression control sequence.
  • the invention also provides a host cell, including bacterial, yeast, insect and mammalian cells, transformed with such polynucleotide compositions. Also provided by the present invention are organisms that have enhanced, reduced, or modified expression of the gene(s) corresponding to the polynucleotide sequences disclosed herein. Processes are also provided for producing a protein, which comprise:
  • the protein produced according to such methods is also provided by the present invention.
  • Protein compositions of the present invention may further comprise a pharmaceutically acceptable carrier.
  • Compositions comprising an antibody which specifically reacts with such protein are also provided by the present invention.
  • such antibody is a monoclonal antibody; also preferably, such antibody is a human antibody.
  • Methods are also provided for preventing, treating or ameliorating a medical condition which comprises administering to a mammalian subject a therapeutically effective amount of a composition comprising a protein of the present invention and a pharmaceutically acceptable carrier.
  • a composition comprising a protein of the present invention and a pharmaceutically acceptable carrier.
  • GPI-122 glycophosphatidylinositol
  • GPI anchors are found to be attached to proteins in mammals, as well as to proteins in lower eukaryotes such as yeast and protozoa. In all cells they are found on the external face of the cell membrane or on the lumenal surface of secretory vesicles.
  • GPI anchors include parasite coat proteins, lymphoid antigens (for example, Thy-1), hydrolytic enzymes, cell adhesion molecules, receptors for small molecules (for example, folate receptor), and the neurturin receptor (Klein et al, 1997, Nature 387: 717-721; Buj-Bello et al, 1997, Nature 387: 721-724).
  • GPI anchors share a core glyco-inositol-1-phospholipid structure, suggesting that a common GPI biosynthetic pathway exists in all eukaryotes.
  • GPI anchors have varying substitutents attached to the carbohydrate portion of the GPI anchor, and the lipid portions of GPI anchors from different species exhibit a striking diversity.
  • GPI anchors can be specifically cleaved through the use of phosphoinositide-specific phospholipase C (PIPLC) (Koke et al., 1991, Protein Expr. Purif. 2(1): 51-58, incorporated by reference herein); treatment of cells with PIPLC releases GPI-anchored proteins from the cell surface.
  • PIPLC phosphoinositide-specific phospholipase C
  • the primary translation products for GPI-anchored proteins have a predominantly hydrophobic sequence at their carboxyl terminus, ranging in size from 17 to about 30 residues, which signals the addition of the GPI anchor and which is cleaved off at the time of anchor addition.
  • Just aminc ⁇ terrninal to the hydrophobic sequence is a stretch of about five to ten hydrophilic amino acids.
  • the residue to which the anchor is added is always one of the small amino acids, such as glycine, aspartic acid, asparagine, alanine, serine, threonine, or cysteine.
  • the two residues carboxy-terminal to the anchor addition site are usually small amino acids also, so that the cleavage/anchor addition site resides in a domain of three small residues.
  • Nucleotide and arrtino acid sequences are reported below for the GPI-122 clone and protein disclosed in the present application.
  • the nucleotide sequence of the GPI-122 clone can readily be determined by sequencing the deposited clone in accordance with known methods. The predicted amino acid sequence (both full-length and mature forms) can then be determined from such nucleotide sequence.
  • amino acid sequence of the protein encoded by a particular clone can also be determined by expression of the clone in a suitable host cell, collecting the protein and determining its sequence.
  • a suitable host cell For the disclosed GPI-122 protein, applicants have identified what they have determined to be the reading frame best identifiable with sequence information available at the time of filing.
  • a "secreted” protein is one which, when expressed in a suitable host cell, is transported across or through a membrane, including transport as a result of signal sequences in its amino acid sequence.
  • "Secreted” proteins include without limitation proteins secreted wholly (e.g., soluble proteins) or partially (e.g., receptors) from the cell in which they are expressed.
  • “Secreted” proteins also include without limitation proteins which are transported across the membrane of the endoplasmic reticulum.
  • GPI-122 is a full-length clone, including the entire coding sequence of a GPI-anchored protein (also referred to herein as "GPI-122 protein")
  • the GPI-122 clone was isolated through the following procedure.
  • the BLAST computer program was used to search the GenBank EST (expressed sequence tag) database for sequences with significant similarity to the nucleotide sequence of Ba3.1 (SEQ ID NO:3), a clone that also encodes a novel GPI-anchored protein.
  • GenBank EST expressed sequence tag
  • Several overlapping human EST sequences were identified with a significant degree of sequence similarity to the Ba3.1 nucleotide sequence.
  • GenBank accession number R99646 yq73e04.rl Homo sapiens cDNA clone 2014385'
  • R99646 clone was not a full-length clone because it did not contain an entire protein-coding sequence.
  • a probe was made using sequence from the 5' end of the R99646 clone; using this probe, two additional clones, each larger than the R99646 clone, were isolated from a phagemid human fetal spleen cDNA library by hybridization under high stringency conditions. The full-length sequences of these clones were determined and it was found that both were still only partial cDNA clones.
  • 5' RACE reactions (Ohara et al, 1989, Proc. Natl. Acad. Sci. USA 86: 5673- 5677, which is incorporated by reference herein) were performed on a human skeletal muscle cDNA library and the RACE reaction products sequenced until a product was identified that contained sequence upstream of the 5' end of an open reading frame.
  • a PCR reaction was performed on a human skeletal muscle library using a first PCR primer derived from the 5' RACE sequence and a second PCR primer derived from a portion of the longest human fetal spleen cDNA clone that was 3' to a SphI restriction site.
  • the resulting "linking" PCR product and the longest human fetal spleen cDNA clone were digested with Notl and SphI, and the appropriate restriction fragments were gel-purified and ligated together.
  • the ligated complete GPI-122 clone was then sequenced in its entirety to confirm that the expected nucleotide sequence was present.
  • amino acids 207 to 219 of SEQ ID NO:2 are a possible leader /signal sequence, with a predicted mature amino acid sequence beginning at amino acid 220; amino acids 310 to 322 of SEQ ID NO:2 are also a possible leader/signal sequence, with a predicted mature amino acid sequence beginning at amino acid 323.
  • Amino acids 1175 to 1193 are a hydrophobic tail region. Arnino-terminal to the hydrophobic tail region is a stretch of several hydrophilic amino acids.
  • Amino acids 1163- 1165 are three small amino acids (threonine, cysteine, and asparagine, respectively) that together with the hydrophobic tail and hydrophilic stretch appear to be a recognition sequence for the addition of a GPI anchor to the threonine 1163 residue, with a concomitant cleavage predicted to remove residues 1164-1193 of the protein.
  • the nucleotide sequence disclosed herein for GPI-122 was searched against the GenBank and GeneSeq nucleotide sequence databases using the BLASTN search protocol.
  • GPI-122 demonstrated at least some similarity with sequences identified as Q60081 (Human brain Expressed Sequence Tag EST02065), as well as several EST sequences including that of GenBank accession number R99646.
  • the predicted amino acid sequence disclosed herein for GPI-122 was searched against the GenPept and GeneSeq amino acid sequence databases using the BLASTP search protocol.
  • the predicted GPI-122 protein demonstrated at least some similarity to sequences identified as Z68314 (K08F8.6 [Caenorhabditis elegans]).
  • GPI-122 proteins and each similar protein or peptide may share at least some activity.
  • the TopPred ⁇ computer program predicts four potential transmembrane domains within the GPI-122 protein sequence, centered around amino acids 500, 900, 980, and 1030 of SEQ ID NO:2, respectively.
  • Fragments of the proteins of the present invention which are capable of exhibiting biological activity are also encompassed by the present invention.
  • Fragments of the protein may be in linear form or they may be cyclized using known methods, for example, as described in H.U. Saragovi, et al, Bio /Technology 10, 773-778 (1992) and in R.S. McDowell, et al, J. Amer. Chem. Soc. 114, 9245-9253 (1992), both of which are incorporated herein by reference.
  • Such fragments may be fused to carrier molecules such as immunoglobulins for many purposes, including increasing the valency of protein binding sites.
  • fragments of the protein may be fused through "linker" sequences to the Fc portion of an immunoglobulin.
  • a bivalent form of the protein such a fusion could be to the Fc portion of an IgG molecule.
  • Other immunoglobulin isotypes may also be used to generate such fusions.
  • a protein - IgM fusion would generate a decavalent form of the protein of the invention.
  • the present invention also provides both full-length and mature forms of the disclosed proteins. The full-length form of the such proteins is identified in the sequence listing by translation of the nucleotide sequence of each disclosed clone.
  • the mature form(s) of such protein may be obtained by expression of the disclosed full-length polynucleotide (preferably those deposited with ATCC) in a suitable mammalian cell or other host cell.
  • the sequence(s) of the mature form(s) of the protein may also be determinable from the amino acid sequence of the full-length form.
  • the present invention also provides genes corresponding to the polynucleotide sequences disclosed herein.
  • “Corresponding genes” are the regions of the genome that are transcribed to produce the mRNAs from which cDNA polynucleotide sequences are derived and may include contiguous regions of the genome necessary for the regulated expression of such genes. Corresponding genes may therefore include but are not limited to coding sequences, 5' and 3' untranslated regions, alternatively spliced exons, introns, promoters, enhancers, and silencer or suppressor elements. The corresponding genes can be isolated in accordance with known methods using the sequence information disclosed herein.
  • Such methods include the preparation of probes or primers from the disclosed sequence information for identification and /or amplification of genes in appropriate genomic libraries or other sources of genomic materials.
  • An "isolated gene” is a gene that has been separated from the adjacent coding sequences, if any, present in the genome of the organism from which the gene was isolated.
  • the chromosomal location corresponding to the polynucleotide sequences disclosed herein may also be determined, for example by hybridizing appropriately labeled polynucleotides of the present invention to chromosomes in situ. It may also be possible to determine the corresponding chromosomal location for a disclosed polynucleotide by identifying significantly similar nucleotide sequences in public databases, such as expressed sequence tags (ESTs), that have already been mapped to particular chromosomal locations. For at least some of the polynucleotide sequences disclosed herein, public database sequences having at least some similarity to the polynucleotide of the present invention have been listed by database accession number.
  • ESTs expressed sequence tags
  • the desired change in gene expression can be achieved through the use of antisense polynucleotides or ribozymes that bind and/or cleave the mRNA transcribed from the gene (Albert and Morris, 1994, Trends Pharmacol. Sci. 15(7): 250-254; Lavarosky et al, 1997,
  • Transgenic animals that have multiple copies of the gene(s) corresponding to the polynucleotide sequences disclosed herein, preferably produced by transformation of cells with genetic constructs that are stably maintained within the transformed cells and their progeny, are provided.
  • organisms are provided in which the gene(s) corresponding to the polynucleotide sequences disclosed herein have been partially or completely inactivated, through insertion of extraneous sequences into the corresponding gene(s) or through deletion of all or part of the corresponding gene(s). Partial or complete gene inactivation can be accomplished through insertion, preferably followed by imprecise excision, of transposable elements (Plasterk, 1992, Bioessays 14(9): 629-633; Zwaal et al, 1993, Proc. Natl. Acad. Sci. USA 90(16): 7431-7435; Clark et al, 1994, Proc. Natl. Acad. Sci.
  • Such organisms are useful for the development of non-human models for the study of disorders involving the corresponding gene(s), and for the development of assay systems for the identification of molecules that interact with the protein product(s) of the corresponding gene(s).
  • the protein of the present invention is membrane-bound (e.g., is a receptor)
  • the present invention also provides for soluble forms of such protein.
  • part or all of the intracellular and transmembrane domains of the protein are deleted such that the protein is fully secreted from the cell in which it is expressed.
  • the intracellular and transmembrane domains of proteins of the invention can be identified in accordance with known techniques for determination of such domains from sequence information.
  • the TopPredLI computer program can be used to predict the location of transmembrane domains in an amino acid sequence, domains which are described by the location of the center of the transmsmbrane domain, with at least ten transmembrane amino acids on each side of the reported central residue(s).
  • Proteins and protein fragments of the present invention include proteins with amino acid sequence lengths that are at least 25%(more preferably at least 50%, and most preferably at least 75%) of the length of a disclosed protein and have at least 60% sequence identity (more preferably, at least 75% identity; most preferably at least 90% or 95% identity) with that disclosed protein, where sequence identity is determined by comparing the amino acid sequences of the proteins when aligned so as to maximize overlap and identity while minimizing sequence gaps.
  • proteins and protein fragments that contain a segment preferably comprising 8 or more (more preferably 20 or more, most preferably 30 or more) contiguous amino acids that shares at least 75% sequence identity (more preferably, at least 85% identity; most preferably at least 95% identity) with any such segment of any of the disclosed proteins.
  • sequence identity may be determined using WU-BLAST
  • WU-BLAST version 2.0 executable programs for several UNIX platforms can be downloaded from ftp://blast.wustl.edu/blast/executables.
  • the complete suite of search programs (BLASTP, BLASTN, BLASTX, TBLASTN, and TBLASTX) is provided at that site, in addition to several support programs.
  • WU-BLAST 2.0 is copyrighted and may not be sold or redistributed in any form or manner without the express written consent of the author; but the posted executables may otherwise be freely used for commercial, nonprofit, or academic purposes.
  • the gapped alignment routines are integral to the database search itself, and thus yield much better sensitivity and selectivity while producing the more easily interpreted output. Gapping can optionally be turned off in all of these programs, if desired.
  • the default amino acid comparison matrix is BLOSUM62, but other amino acid comparison matrices such as PAM can be utilized.
  • Species homologues of the disclosed polynucleotides and proteins are also provided by the present invention.
  • a "species homologue” is a protein or polynucleotide with a different species of origin from that of a given protein or polynucleotide, but with significant sequence similarity to the given protein or polynucleotide.
  • polynucleotide species homologues have at least 60% sequence identity (more preferably, at least 75% identity; most preferably at least 90% identity) with the given polynucleotide, and protein species homologues have at least 30% sequence identity (more preferably, at least 45% identity; most preferably at least 60% identity) with the given protein, where sequence identity is determined by comparing the nucleotide sequences of the polynucleotides or the amino acid sequences of the proteins when aligned so as to maximize overlap and identity while mirvimizing sequence gaps.
  • Species homologues may be isolated and identified by making suitable probes or primers from the sequences provided herein and screening a suitable nucleic acid source from the desired species.
  • species homologues are those isolated from mammalian species. Most preferably, species homologues are those isolated from certain mammalian species such as, for example, Pan troglodytes, Gorilla gorilla, Pongo pygmaeus, Hylobates concolor, Macaca mulatta, Papio papio, Papio hamadryas, Cercopithecus aethiops, Cebus capucinus, Aotus trivirgatus, Sanguinus oedipus, Microcebus murinus, Mus musculus, Rattus norvegicus, Cricetulus griseus, Felis catus, Mustela vison, Canisfamiliaris, Oryctolagus cuniculus, Bos taurus, Ovis aries, Sus scrofa, and Equus caballus, for which genetic maps have been created allowing the identification of syntenic relationships between the genomic organization of genes in one species and the genomic organization of the related genes in another species
  • allelic variants of the disclosed polynucleotides or proteins that is, naturally-occurring alternative forms of the isolated polynucleotides which also encode proteins which are identical or have significantly similar sequences to those encoded by the disclosed polynucleotides.
  • allelic variants have at least 60% sequence identity (more preferably, at least 75% identity; most preferably at least 90% identity) with the given polynucleotide, where sequence identity is determined by comparing the nucleotide sequences of the polynucleotides when aligned so as to maximize overlap and identity while minimizing sequence gaps.
  • Allelic variants may be isolated and identified by making suitable probes or primers from the sequences provided herein and screening a suitable nucleic acid source from individuals of the appropriate species.
  • the invention also includes polynucleotides with sequences complementary to those of the polynucleotides disclosed herein.
  • the present invention also includes polynucleotides that hybridize under reduced stringency conditions, more preferably stringent conditions, and most preferably highly stringent conditions, to polynucleotides described herein. Examples of stringency conditions are shown in the table below: highly stringent conditions are those that are at least as stringent as, for example, conditions A-F; stringent conditions are at least as stringent as, for example, conditions G-L; and reduced stringency conditions are at least as stringent as, for example, conditions M-R.
  • the hybrid length is that anti ⁇ pated for the hybridized reg ⁇ on(s) of the hybridizing polynucleotides.
  • the hybrid length is assumed to be that of the hybridizing polynucleotide.
  • the hybrid length can be determined by aligning the sequences of the polynucleotides and identifying the region or regions of optimal sequence complementarity.
  • SSPE lxSSPE is 0.15M NaCl, lOmM NaH 2 P0 4 , and 1.25mM EDTA, pH 74
  • SSC 0.15M NaCl and 15mM sodium citrate
  • each such hybridizing polynucleotide has a length that is at least 25%(more preferably at least 50%, and most preferably at least 75%) of the length of the polynucleotide of the present invention to which it hybridizes, and has at least 60% sequence identity (more preferably, at least 75% identity; most preferably at least 90% or 95% identity) with the polynucleotide of the present invention to which it hybridizes, where sequence identity is determined by comparing the sequences of the hybridizing polynucleotides when aligned so as to maximize overlap and identity while minimizing sequence gaps.
  • the isolated polynucleotide of the invention may be operably linked to an expression control sequence such as the pMT2 or pED expression vectors disclosed in Kaufman et al, Nucleic Acids Res. 19, 4485-4490 (1991), in order to produce the protein recombinantly.
  • an expression control sequence such as the pMT2 or pED expression vectors disclosed in Kaufman et al, Nucleic Acids Res. 19, 4485-4490 (1991)
  • Many suitable expression control sequences are known in the art. General methods of expressing recombinant proteins are also known and are exemplified in R. Kaufman, Methods in Enzymology 185. 537-566 (1990).
  • operably linked means that the isolated polynucleotide of the invention and an expression control sequence are situated within a vector or cell in such a way that the protein is expressed by a host cell which has been transformed (transfected) with the ligated polynucleotide /expression control sequence.
  • Mammalian host cells include, for example, monkey COS cells, Chinese Hamster
  • Ovary (CHO) cells human kidney 293 cells, human epidermal A431 cells, human Colo205 cells, 3T3 cells, CV-1 cells, other transformed primate cell lines, normal diploid cells, cell strains derived from in vitro culture of primary tissue, primary explants, HeLa cells, mouse L cells, BHK, HL-60, U937, HaK or Jurkat cells.
  • yeast eukaryotes
  • prokaryotes e.g., yeast or any yeast strain capable of expressing heterologous proteins.
  • Potentially suitable bacterial strains include Escherichia coli, Bacillus subtilis, Salmonella typhimurium, or any bacterial
  • the protein 16 strain capable of expressing heterologous proteins. If the protein is made in yeast or bacteria, it may be necessary to modify the protein produced therein, for example by phosphorylation or glycosylation of the appropriate sites, in order to obtain the functional protein. Such covalent attachments may be accomplished using known chemical or enzymatic methods.
  • the protein may also be produced by operably linking the isolated polynucleotide of the invention to suitable control sequences in one or more insect expression vectors, and employing an insect expression system.
  • suitable control sequences in one or more insect expression vectors, and employing an insect expression system.
  • Materials and methods for baculovirus/insect cell expression systems are commercially available in kit form from, e.g., Invitrogen, San Diego, California, U.S.A. (the MaxBac® kit), and such methods are well known in the art, as described in Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987), incorporated herein by reference.
  • an insect cell capable of expressing a polynucleotide of the present invention is "transformed.”
  • the protein of the invention may be prepared by culturing transformed host cells under culture conditions suitable to express the recombinant protein.
  • the resulting expressed protein may then be purified from such culture (i.e., from culture medium or cell extracts) using known purification processes, such as gel filtration and ion exchange chromatography.
  • the purification of the protein may also include an affinity column containing agents which will bind to the protein; one or more column steps over such affinity resins as concanavalin A-agarose, heparin-toyopearl® or Cibacrom blue 3GA Sepharose®; one or more steps involving hydrophobic interaction chromatography using such resins as phenyl ether, butyl ether, or propyl ether; or iirununoaffinity chromatography.
  • the protein of the invention may also be expressed in a form which will facilitate purification.
  • fusion protein such as those of maltose binding protein (MBP), glutathione-S-transferase (GST) or thioredoxin (TRX). Kits for expression and purification of such fusion proteins are commercially available from New England BioLabs (Beverly, MA), Pharmacia (Piscataway, NJ) and Invitrogen Corporation (Carlsbad, CA), respectively.
  • MBP maltose binding protein
  • GST glutathione-S-transferase
  • TRX thioredoxin
  • Kits for expression and purification of such fusion proteins are commercially available from New England BioLabs (Beverly, MA), Pharmacia (Piscataway, NJ) and Invitrogen Corporation (Carlsbad, CA), respectively.
  • the protein can also be tagged with an epitope and subsequently purified by using a specific antibody directed to such epitope.
  • One such epitope (“Flag") is commercially available from the Eastman Kodak Company (New Haven, CT).
  • RP- HPLC reverse-phase high performance liquid chromatography
  • hydrophobic RP-HPLC media e.g., silica gel having pendant methyl or other aliphatic groups
  • Some or all of the foregoing purification steps, in various combinations, can also be employed to provide a substantially homogeneous isolated recombinant protein.
  • the protein thus purified is substantially free of other mammalian proteins and is defined in accordance with the present invention as an "isolated protein.”
  • the protein of the invention may also be expressed as a product of transgenic animals, e.g., as a component of the milk of transgenic cows, goats, pigs, or sheep which are characterized by somatic or germ cells containing a nucleotide sequence encoding the protein.
  • the protein may also be produced by known conventional chemical synthesis. Methods for constructing the proteins of the present invention by synthetic means are known to those skilled in the art.
  • the synthetically-constructed protein sequences by virtue of sharing primary, secondary or tertiary structural and /or conformational characteristics with proteins may possess biological properties in common therewith, including protein activity. Thus, they may be employed as biologically active or immunological substitutes for natural, purified proteins in screening of therapeutic compounds and in immunological processes for the development of antibodies.
  • the proteins provided herein also include proteins characterized by amino acid sequences similar to those of purified proteins but into which modification are naturally provided or deliberately engineered. For example, modifications in the peptide or DNA sequences can be made by those skilled in the art using known techniques.
  • Modifications of interest in the protein sequences may include the alteration, substitution, replacement, insertion or deletion of a selected amino acid residue in the coding sequence.
  • one or more of the cysteine residues may be deleted or replaced with another amino acid to alter the conformation of the molecule.
  • Techniques for such alteration, substitution, replacement, insertion or deletion are well known to those skilled in the art (see, e.g., U.S. Patent No.4,518,584).
  • such alteration, substitution, replacement, insertion or deletion retains the desired activity of the protein.
  • polynucleotides and proteins of the present invention are expected to exhibit one or more of the uses or biological activities (including those associated with assays cited herein) identified below.
  • Uses or activities described for proteins of the present invention may be provided by administration or use of such proteins or by adrriinistration or use of polynucleotides encoding such proteins (such as, for example, in gene therapies or vectors suitable for introduction of DNA).
  • the polynucleotides provided by the present invention can be used by the research community for various purposes.
  • the polynucleotides can be used to express recombinant protein for analysis, characterization or therapeutic use; as markers for tissues in which the corresponding protein is preferentially expressed (either constitutively or at a particular stage of tissue differentiation or development or in disease states); as molecular weight markers on Southern gels; as chromosome markers or tags (when labeled) to identify chromosomes or to map related gene positions; to compare with endogenous DNA sequences in patients to identify potential genetic disorders; as probes to hybridize and thus discover novel, related DNA sequences; as a source of information to derive PCR primers for genetic fingerprinting; as a probe to "subtract-out" known sequences in the process of discovering other novel polynucleotides; for selecting and making oligomers for attachment to a "gene chip” or other support, including for examination of expression patterns; to raise anti-protein antibodies using DNA immunization techniques;
  • the polynucleotide encodes a protein which binds or potentially binds to another protein (such as, for example, in a receptor-ligand interaction)
  • the polynucleotide can also be used in interaction trap assays (such as, for example, those described in Gyuris et al, 1993, Cell 75: 791-803 and in Rossi et al, 1997, Proc. Natl. Acad.
  • the proteins provided by the present invention can similarly be used in assay to determine biological activity, including in a panel of multiple proteins for high- throughput screening; to raise antibodies or to elicit another immune response; as a reagent (including the labeled reagent) in assays designed to quantitatively determine levels of the protein (or its receptor) in biological fluids; as markers for tissues in which the corresponding protein is preferentially expressed (either constitutively or at a particular stage of tissue differentiation or development or in a disease state); and, of course, to isolate correlative receptors or ligands.
  • the protein binds or potentially binds to another protein (such as, for example, in a receptor-ligand interaction)
  • the protein can be used to identify the other protein with which binding occurs or to identify inhibitors of the binding interaction. Proteins involved in these binding interactions can also be used to screen for peptide or small molecule inhibitors or agonists of the binding interaction.
  • Polynucleotides and proteins of the present invention can also be used as nutritional sources or supplements. Such uses include without limitation use as a protein or amino add supplement, use as a carbon source, use as a nitrogen source and use as a source of carbohydrate.
  • the protein or polynucleotide of the invention can be added to the feed of a particular organism or can be administered as a separate solid or liquid preparation, such as in the form of powder, pills, solutions, suspensions or capsules.
  • the protein or polynucleotide of the invention can be added to the medium in or on which the microorganism is cultured.
  • a protein of the present invention may exhibit cytokine, cell proliferation (either inducing or inhibiting) or cell differentiation (either inducing or inhibiting) activity or may
  • cytokine activity is evidenced by any one of a number of routine factor dependent cell proliferation assays for cell lines including, without limitation, 32D, DA2, DA1G, T10, B9, B9/11, BaF3, MC9/G, M+ (preB M+), 2E8, RB5, DAI, 123, T1165, HT2, CTLL2, TF-1, Mo7e and CMK.
  • Assays for T-cell or thymocyte proliferation include without limitation those described in: Current Protocols in Immunology, Ed by J. E. Coligan, A.M. Kruisbeek, D.H. Margulies, E.M. Shevach, W Strober, Pub. Greene Publishing Associates and Wiley- Intersdence (Chapter 3, In Vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); Takai et al., J. Immunol. 137:3494-3500, 1986; BertagnoUi et al., J. Immunol.
  • Assays for cytokine production and /or proliferation of spleen cells, lymph node cells or thymocytes include, without limitation, those described in: Polyclonal T cell stimulation, Kruisbeek, A.M. and Shevach, E.M. In Current Protocols in Immunology. J.E.e.a.
  • Assays for proliferation and differentiation of hematopoietic and lymphopoietic ceUs include, without limitation, those described in: Measurement of Human and Murine
  • Assays for T-cell clone responses to antigens include, without Umitation, those described in: Current Protocols in Immunology, Ed by J. E. Coligan, A.M. Kruisbeek, D.H. Margulies, E.M. Shevach, W Strober, Pub. Greene PubUshing Associates and Wiley-Interscience (Chapter 3, In Vitro assays for Mouse Lymphocyte Function; Chapter 6, Cytokines and their cellular receptors; Chapter 7, Immunologic studies in Humans); Weinberger et al., Proc. Natl. Acad. Sci.
  • a protein of the present invention may also exhibit immune stimulating or immune suppressing activity, induding without Umitation the activities for which assays are described herein.
  • a protein may be useful in the treatment of various immune deficiencies and disorders (including severe combined immunodeficiency (SCID)), e.g., in regulating (up or down) growth and proUferation of T and /or B lymphocytes, as well as effecting the cytolytic activity of NK ceUs and other cell populations.
  • SCID severe combined immunodeficiency
  • These immune deficiendes may be genetic or be caused by viral (e.g., HIV) as well as bacterial or fungal infections, or may result from autoimmune disorders.
  • infectious diseases causes by viral, bacterial, fungal or other infection may be treatable using a protein of the present invention, including infections by HIV, hepatitis viruses, herpesviruses, mycobacteria, Leishmania spp., malaria spp. and various fungal infections such as candidiasis.
  • a protein of the present invention may also be useful where a boost to the immune system generally may be desirable, i.e., in the treatment of cancer.
  • Autoimmune disorders which may be treated using a protein of the present invention include, for example, connective tissue disease, multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, autoimmune pulmonary inflammation, GuUlain-Barre syndrome, autoimmune thyroiditis, insulin dependent diabetes meUitis, myasthenia gravis, graft-versus-host disease and autoimmune inflammatory eye disease.
  • Such a protein of the present invention may also to be useful in the treatment of aUergic reactions and conditions, such as asthma (particularly aUergic asthma) or other respiratory problems.
  • Other conditions, in which immune suppression is desired may also be treatable using a protein of the present invention.
  • T cells may be inhibited by suppressing T cell responses or by inducing specific tolerance in T cells, or both.
  • Immunosuppression of T ceU responses is generaUy an active, non-antigen-specific, process which requires continuous exposure of the T ceUs to the suppressive agent.
  • Tolerance which involves inducing non-responsiveness or anergy in T ceUs, is distinguishable from immunosuppression in that it is generally antigen-specific and persists after exposure to the tolerizing agent has ceased. OperationaUy, tolerance can be demonstrated by the lack of a T cell response upon reexposure to specific antigen in the absence of the tolerizing agent.
  • Down regulating or preventing one or more antigen functions (including without Umitation B lymphocyte antigen functions (such as , for example, B7)), e.g., preventing high level lymphokine synthesis by activated T ceUs, wiU be useful in situations of tissue, skin and organ transplantation and in graft-versus-host disease (GVHD).
  • GVHD graft-versus-host disease
  • blockage of T cell function should result in reduced tissue destruction in tissue transplantation.
  • TypicaUy in tissue transplants, rejection of the transplant is initiated through its recognition as foreign by T cells, f oUowed by an immune reaction that destroys the transplant.
  • a molecule which inhibits or blocks interaction of a B7 lymphocyte antigen with its natural Ugand(s) on immune cells can lead to the binding of the molecule to the natural ligand(s) on the immune ceUs without transmitting the corresponding costimulatory signal.
  • B7 lymphocyte antigen e.g., B7- 1, B7-3 or blocking antibody
  • Blocking B lymphocyte antigen function in this matter prevents cytokine synthesis by immune cells, such as T cells, and thus acts as an immunosuppressant.
  • the lack of costimulation may also be sufficient to anergize the T ceUs, thereby inducing tolerance in a subject. Induction of long-term
  • B lymphocyte antigen-blocking reagents may avoid the necessity of repeated adrninistration of these blocking reagents. To achieve sufficient immunosuppression or tolerance in a subject, it may also be necessary to block the function of a combination of B lymphocyte antigens.
  • the efficacy of particular blocking reagents in preventing organ transplant rejection or GVHD can be assessed using animal models that are predictive of efficacy in humans.
  • Examples of appropriate systems which can be used include allogeneic cardiac grafts in rats and xenogeneic pancreatic islet cell grafts in mice, both of which have been used to examine the immunosuppressive effects of CTLA4Ig fusion proteins in vivo as described in Lenschow et al, Sdence 257:789-792 (1992) and Turka et al, Proc. Natl. Acad. Sci USA, 89:11102-11105 (1992).
  • murine models of GVHD see Paul ed., Fundamental Immunology, Raven Press, New York, 1989, pp. 846-847) can be used to determine the effect of blocking B lymphocyte antigen function in vivo on the development of that disease.
  • Blocking antigen function may also be therapeuticaUy useful for treating autoimmune diseases.
  • Many autoimmune disorders are the result of inappropriate activation of T cells that are reactive against self tissue and which promote the production of cytokines and autoantibodies involved in the pathology of the diseases.
  • Preventing the activation of autoreactive T cells may reduce or eliminate disease symptoms.
  • Administration of reagents which block costimulation of T cells by disrupting receptor:ligand interactions of B lymphocyte antigens can be used to inhibit T cell activation and prevent production of autoantibodies or T cell-derived cytokines which may be involved in the disease process.
  • blocking reagents may induce antigen-specific tolerance of autoreactive T ceUs which could lead to long-term reUef from the disease.
  • the efficacy of blocking reagents in preventing or aUeviating autoimmune disorders can be determined using a number of well-characterized animal models of human autoimmune diseases. Examples include murine experimental autoimmune encephalitis, systemic lupus erythmatosis in MRL/lprfipr mice or NZB hybrid mice, murine autoimmune coUagen arthritis, diabetes meUitus in NOD mice and BB rats, and murine experimental myasthenia gravis (see Paul ed., Fundamental Immunology, Raven
  • Upregulation of an antigen function (preferably a B lymphocyte antigen function), as a means of up regulating immune responses, may also be useful in therapy. Upregulation of immune responses may be in the form of enhancing an existing immune
  • enhancing an immune response through stimulating B lymphocyte antigen function may be useful in cases of viral infection.
  • systemic viral diseases such as influenza, the common cold, and encephaUtis might be aUeviated by the administration of stimulatory forms of B lymphocyte antigens systemically.
  • anti-viral immune responses may be enhanced in an infected patient by removing T cells from the patient, costimulating the T cells in vitro with viral antigen- pulsed APCs either expressing a peptide of the present invention or together with a stimulatory form of a soluble peptide of the present invention and reintroducing the in vitro activated T cells into the patient.
  • Another method of enhancing anti-viral immune responses would be to isolate infected ceUs from a patient, transfect them with a nucleic acid encoding a protein of the present invention as described herein such that the ceUs express aU or a portion of the protein on their surface, and reintroduce the transfected ceUs into the patient.
  • the infected ceUs would now be capable of delivering a costimulatory signal to, and thereby activate, T cells in vivo.
  • up regulation or enhancement of antigen function may be useful in the induction of tumor immunity.
  • Tumor ceUs e.g., sarcoma, melanoma, lymphoma, leukemia, neuroblastoma, carcinoma
  • a nucleic add encoding at least one peptide of the present invention can be administered to a subject to overcome tumor-specific tolerance in the subject. If desired, the tumor ceU can be transfected to express a combination of peptides.
  • tumor cells obtained from a patient can be transfected ex vivo with an expression vector directing the expression of a peptide having B7-2-like activity alone, or in conjunction with a peptide having B7-1-Uke activity and/or B7-3-like activity.
  • the transfected tumor cells are returned to the patient to result in expression of the peptides on the surface of the transfected ceU.
  • gene therapy techniques can be used to target a tumor cell for transfection in vivo.
  • tumor ceUs which lack MHC class I or MHC class II molecules, or which fail to reexpress suffident amounts of MHC class I or MHC class II molecules, can be transfected with nucleic acid encoding all or a portion of (e.g., a cytoplasmic-domain truncated portion) of an MHC class I ⁇ chain protein and ⁇ 2
  • MHC 25 microglobulin protein or an MHC class II a chain protein and an MHC class II ⁇ chain protein to thereby express MHC rouge I or MHC class II proteins on the ceU surface.
  • Expression of the appropriate rouge I or class Et MHC in conjunction with a peptide having the activity of a B lymphocyte antigen (e.g., B7-1, B7-2, B7-3) induces a T cell mediated immune response against the transfected tumor ceU.
  • a B lymphocyte antigen e.g., B7-1, B7-2, B7-3
  • a gene encoding an antisense construct which blocks expression of an MHC class II associated protein, such as the invariant chain can also be cotransfected with a DNA encoding a peptide having the activity of a B lymphocyte antigen to promote presentation of tumor associated antigens and induce tumor specific immunity.
  • a T ceU mediated immune response in a human subject may be sufficient to overcome tumor-specific tolerance in the subject.
  • the activity of a protein of the invention may, among other means, be measured by the following methods:
  • Suitable assays for thymocyte or splenocyte cytotoxicity include, without limitation, those described in: Current Protocols in Immunology, Ed by J. E. CoUgan, A.M. Kruisbeek, D.H. MarguUes, E.M. Shevach, W Strober, Pub. Greene PubUshing Associates and Wney-Interscience (Chapter 3, In Vitro assays for Mouse Lymphocyte Function 3.1- 3.19; Chapter 7, Immunologic studies in Humans); Herrmann et al., Proc. Natl. Acad. Sci. USA 78:2488-2492, 1981; Herrmann et al., J. Immunol. 128:1968-1974, 1982; Handa et al., J.
  • Assays for T-ceU-dependent immunoglobulin responses and isotype switching include, without limitation, those described in: MaUszewski, J. Immunol. 144:3028-3033, 1990; and Assays for B ceU function: In vitro antibody production, Mond, J.J. and Brunswick, M. In Current Protocols in Immunology.
  • MLR Mixed lymphocyte reaction
  • Dendritic cell-dependent assays (which wiU identify, among others, proteins expressed by dendritic cells that activate naive T-cells) include, without limitation, those described in: Guery et al., J. Immunol. 134:536-544, 1995; Inaba et al., Journal of Experimental Medicine 173:549-559, 1991; Macatonia et al., Journal of Immunology 154:5071-5079, 1995; Porgador et al., Journal of Experimental Medicine 182:255-260, 1995; Nair et al., Journal of Virology 67:4062-4069, 1993; Huang et al., Science 264:961-965, 1994; Macatonia et al., Journal of Experimental Medicine 169:1255-1264, 1989; Bhardwaj et al., Journal of Clinical Investigation 94:797-807, 1994; and Inaba et al., Journal of Experimental Medicine 172:631-640, 1990.
  • lymphocyte survival /apoptosis which wiU identify, among others, proteins that prevent apoptosis after superantigen induction and proteins that regulate lymphocyte homeostasis
  • indude without Umitation, those described in: Darzynkiewicz et al., Cytometry 13:795-808, 1992; Gorczyca et al., Leukemia 7:659-670, 1993; Gorczyca et al., Cancer Research 53:1945-1951, 1993; Itoh et al., Cell 66:233-243, 1991; Zacharchuk, Journal of Immunology 145:4037-4045, 1990; Zamai et al., Cytometry 14:891-897, 1993; Gorczyca et al., International Journal of Oncology 1:639-648, 1992.
  • Assays for proteins that influence early steps of T-cell commitment and development include, without Umitation, those described in: Antica et al., Blood 84:111-117, 1994; Fine et al., CeUular Immunology 155:111-122, 1994; Galy et al, Blood 85:2770-2778, 1995; Toki et al., Proc. Nat. Acad Sci. USA 88:7548-7551, 1991.
  • a protein of the present invention may be useful in regulation of hematopoiesis and, consequently, in the treatment of myeloid or lymphoid cell deficiendes. Even marginal biological activity in support of colony forming cells or of factor-dependent ceU lines indicates involvement in regulating hematopoiesis, e.g. in supporting the growth and proliferation of erythroid progenitor cells alone or in combination with other cytokines, thereby indicating utility, for example, in treating various anemias or for use in conjunction with irradiation /chemotherapy to stimulate the production of erythroid precursors and/or erythroid cells; in supporting the growth and proliferation of myeloid
  • 27 ceUs such as granulocytes and monocytes/macrophages (i.e., traditional CSF activity) useful, for example, in conjunction with chemotherapy to prevent or treat consequent myelo-suppression; in supporting the growth and proUferation of megakaryocytes and consequently of platelets thereby aUowing prevention or treatment of various platelet disorders such as thrombocytopenia, and generaUy for use in place of or complimentary to platelet transfusions; and /or in supporting the growth and proUferation of hematopoietic stem ceUs which are capable of maturing to any and aU of the above- mentioned hematopoietic cells and therefore find therapeutic utility in various stem ceU disorders (such as those usuaUy treated with transplantation, including, without limitation, aplastic anemia and paroxysmal nocturnal hemoglobinuria), as well as in repopulating the stem ceU compartment post irradiation/chemotherapy, either in-vivo or ex-vivo (i.
  • Assays for embryonic stem ceU differentiation include, without Umitation, those described in: Johansson et al. CeUular Biology 15:141-151, 1995; Keller et al., Molecular and CeUular Biology 13:473-486, 1993; McClanahan et al, Blood 81:2903-2915, 1993.
  • Assays for stem cell survival and differentiation include, without Umitation, those described in: MethylceUulose colony forming assays, Freshney, M.G. In Culture of Hematopoietic Cells. R.I. Freshney, et al. eds. Vol pp. 265-268, Wiley-Liss, Inc., New York, NY. 1994; Hirayama et al., Proc. Natl. Acad. Sci. USA 89:5907-5911, 1992; Primitive hematopoietic colony forming ceUs with high proliferative potential, McNiece, I.K. and Briddell, R.A. In Culture of Hematopoietic Cells. R.I. Freshney, et al. eds. Vol pp. 23-39,
  • a protein of the present invention also may have utility in compositions used for bone, cartilage, tendon, Ugament and /or nerve tissue growth or regeneration, as well as for wound healing and tissue repair and replacement, and in the treatment of burns, incisions and ulcers.
  • a protein of the present invention which induces cartilage and/or bone growth in circumstances where bone is not normally formed, has appUcation in the healing of bone fractures and cartilage damage or defects in humans and other animals.
  • Such a preparation employing a protein of the invention may have prophylactic use in closed as well as open fracture reduction and also in the improved fixation of artificial joints. De novo bone formation induced by an osteogenic agent contributes to the repair of congenital, trauma induced, or oncologic resection induced craniofacial defects, and also is useful in cosmetic plastic surgery.
  • a protein of this invention may also be used in the treatment of periodontal disease, and in other tooth repair processes. Such agents may provide an environment to attract bone-forming ceUs, stimulate growth of bone-forming ceUs or induce differentiation of progenitors of bone-forming ceUs.
  • a protein of the invention may also be useful in the treatment of osteoporosis or osteoarthritis, such as through stimulation of bone and /or cartilage repair or by blocking inflammation or processes of tissue destruction (coUagenase activity, osteoclast activity, etc.) mediated by inflammatory processes.
  • tissue regeneration activity that may be attributable to the protein of the present invention is tendon/ligament formation.
  • a protein of the present invention which induces tendon/ligament-like tissue or other tissue formation in circumstances where such tissue is not normaUy formed, has appUcation in the healing of tendon or Ugament tears, deformities and other tendon or Ugament defects in humans and other animals.
  • Such a preparation employing a tendon/ligament-like tissue inducing protein may have prophylactic use in preventing damage to tendon or Ugament tissue, as well as use in the improved fixation of tendon or Ugament to bone or other tissues, and
  • compositions of the present invention may provide an environment to attract tendon- or ligament-forming cells, stimulate growth of tendon- or Ugament-forming ceUs, induce differentiation of progenitors of tendon- or Ugament-forming ceUs, or induce growth of tendon/Ugament ceUs or progenitors ex vivo for return in vivo to effect tissue repair.
  • compositions of the invention may also be useful in the treatment of tendinitis, carpal tunnel syndrome and other tendon or Ugament defects.
  • the compositions may also include an appropriate matrix and /or sequestering agent as a carrier as is well known in the art.
  • the protein of the present invention may also be useful for proUferation of neural cells and for regeneration of nerve and brain tissue, i.e. for the treatment of central and peripheral nervous system diseases and neuropathies, as well as mechanical and traumatic disorders, which involve degeneration, death or trauma to neural cells or nerve tissue. More specifically, a protein may be used in the treatment of diseases of the peripheral nervous system, such as peripheral nerve injuries, peripheral neuropathy and localized neuropathies, and central nervous system diseases, such as Alzheimer's, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Shy-Drager syndrome. Further conditions which may be treated in accordance with the present invention indude med anical and traumatic disorders, such as spinal cord disorders, head trauma and cerebrovascular diseases such as stroke. Peripheral neuropathies resulting from chemotherapy or other medical therapies may also be treatable using a protein of the invention.
  • diseases of the peripheral nervous system such as peripheral nerve injuries, peripheral neuropathy and localized neuropathies, and central nervous system diseases, such as Alzheimer's
  • Proteins of the invention may also be useful to promote better or faster closure of non-healing wounds, including without Umitation pressure ulcers, ulcers associated with vascular insufficiency, surgical and traumatic wounds, and the like.
  • a protein of the present invention may also exhibit activity for generation or regeneration of other tissues, such as organs (including, for example, pancreas, Uver, intestine, kidney, skin, endotheUum), musde (smooth, skeletal or cardiac) and vascular (including vascular endotheUum) tissue, or for promoting the growth of cells comprising such tissues.
  • organs including, for example, pancreas, Uver, intestine, kidney, skin, endotheUum
  • musde smooth, skeletal or cardiac
  • vascular including vascular endotheUum
  • a protein of the invention may also exhibit angiogenic activity.
  • a protein of the present invention may also be useful for gut protection or regeneration and treatment of lung or Uver fibrosis, reperfusion injury in various tissues, and conditions resulting from systemic cytokine damage.
  • a protein of the present invention may also be useful for promoting or inhibiting differentiation of tissues described above from precursor tissues or ceUs; or for inhibiting the growth of tissues described above.
  • the activity of a protein of the invention may, among other means, be measured by the following methods:
  • Assays for tissue generation activity include, without Umitation, those described in: International Patent PubUcation No. WO95/ 16035 (bone, cartilage, tendon); International Patent PubUcation No. WO95/ 05846 (nerve, neuronal); International Patent Publication No. WO91/07491 (skin, endotheUum ).
  • Assays for wound healing activity include, without Umitation, those described in:
  • a protein of the present invention may also exhibit activin- or inhibin-related activities, Inhibins are characterized by their ability to inhibit the release of folUcle stimulating hormone (FSH), while activins and are characterized by their ability to stimulate the release of foUide stimulating hormone (FSH).
  • FSH folUcle stimulating hormone
  • a protein of the present invention alone or in heterodimers with a member of the inhibin ⁇ family, may be useful as a contraceptive based on the abiUty of rnhibins to decrease fertiUty in female mammals and decrease spermatogenesis in male mammals. Administration of sufficient amounts of other rnhibins can induce infertiUty in these mammals.
  • the protein of the invention may be useful as a fertiUty inducing therapeutic, based upon the abiUty of activin molecules in stimulating FSH release from cells of the anterior pituitary. See, for example, United States Patent 4,798,885.
  • a protein of the invention may also be useful for advancement of the onset of fertiUty in sexuaUy immature mammals, so as to increase the lifetime reproductive performance of domestic animals such as cows, sheep and pigs.
  • the activity of a protein of the invention may, among other means, be measured by the following methods:
  • a protein of the present invention may have chemotactic or chemokinetic activity (e.g., act as a chemokine) for mammalian ceUs, including, for example, monocytes, fibroblasts, neutrophils, T-ceUs, mast cells, eosinophils, epitheUal and/or endotheUal cells.
  • Chemotactic and chemokinetic proteins can be used to mobilize or attract a desired ceU population to a desired site of action.
  • Chemotactic or chemokinetic proteins provide particular advantages in treatment of wounds and other trauma to tissues, as well as in treatment of localized infections. For example, attraction of lymphocytes, monocytes or neutrophils to tumors or sites of infection may result in improved immune responses against the tumor or infecting agent.
  • a protein or peptide has chemotactic activity for a particular cell population if it can stimulate, directly or indirectly, the directed orientation or movement of such ceU population.
  • the protein or peptide has the abiUty to directly stimulate directed movement of ceUs. Whether a particular protein has chemotactic activity for a population of cells can be readily determined by employing such protein or peptide in any known assay for cell chemotaxis.
  • the activity of a protein of the invention may, among other means, be measured by the following methods:
  • Assays for chemotactic activity consist of assays that measure the abiUty of a protein to induce the migration of ceUs across a membrane as weU as the abiUty of a protein to induce the adhesion of one ceU population to another cell population.
  • a protein of the invention may also exhibit hemostatic or thrombolytic activity.
  • such a protein is expected to be useful in treatment of various coagulation disorders (including hereditary disorders, such as hemophilias) or to enhance coagulation and other hemostatic events in treating wounds resulting from trauma, surgery or other causes.
  • a protein of the invention may also be useful for dissolving or inhibiting formation of thromboses and for treatment and prevention of conditions resulting therefrom (such as, for example, infarction of cardiac and central nervous system vessels (e.g., stroke).
  • the activity of a protein of the invention may, among other means, be measured by the following methods: Assay for hemostatic and thrombolytic activity include, without limitation, those described in: Linet et al., J. Clin. Pharmacol. 26:131-140, 1986; Burdick et al, Thrombosis Res.45:413-419, 1987; Humphrey et al., Fibrinolysis 5:71-79 (1991); Schaub, Prostaglandins 35:467-474, 1988.
  • a protein of the present invention may also demonstrate activity as receptors, receptor Ugands or inhibitors or agonists of receptor/Ugand interactions.
  • receptors and Ugands include, without Umitation, cytokine receptors and their ligands, receptor kinases and their Ugands, receptor phosphatases and their Ugands, receptors involved in ceU-ceU interactions and their Ugands (including without limitation, ceUular adhesion molecules (such as selectins, integrins and their Ugands) and receptor/ligand pairs involved in antigen presentation, antigen recognition and development of ceUular and humoral immune responses).
  • Receptors and Ugands are also useful for screening of potential peptide or small molecule inhibitors of the relevant receptor/Ugand interaction.
  • a protein of the present invention (including, without Umitation, fragments of receptors and Ugands) may themselves be useful as inhibitors of receptor/ligand interactions.
  • the activity of a protein of the invention may, among other means, be measured by the following methods:
  • Suitable assays for receptor-Ugand activity include without Umitation those described in:Current Protocols in Immunology, Ed by J.E. CoUgan, A.M. Kruisbeek, D.H. Margulies, E.M. Shevach, W.Strober, Pub. Greene PubUshing Associates and Wiley-Interscience (Chapter 7.28, Measurement of CeUular Adhesion under static conditions 7.28.1-7.28.22), Takai et al., Proc. Natl. Acad. Sci. USA 84:6864-6868, 1987; Bierer et al., J. Exp. Med. 168:1145-1156, 1988; Rosenstein et al., J. Exp. Med. 169:149-160 1989; Stoltenborg et al., J. Immunol. Methods 175:59-68, 1994; Stitt et al., CeU 80:661-670, 1995.
  • Proteins of the present invention may also exhibit anti-inflammatory activity.
  • the anti-inflarnmatory activity may be achieved by providing a stimulus to ceUs involved in the inflammatory response, by inhibiting or promoting ce -ceU interactions (such as, for example, ceU adhesion), by inhibiting or promoting chemotaxis of ceUs involved in the inflammatory process, inhibiting or promoting cell extravasation, or by stimulating or suppressing production of other factors which more directly inhibit or promote an inflammatory response.
  • Proteins exhibiting such activities can be used to treat inflammatory conditions including chronic or acute conditions), including without limitation inflammation assodated with infection (such as septic shock, sepsis or systemic inflammatory response syndrome (SIRS)), ischemia-reperfusion injury, endotoxin lethaUty, arthritis, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine-induced lung injury, inflammatory bowel disease, Crohn's disease or resulting from over production of cytokines such as TNF or IL-1. Proteins of the invention may also be useful to treat anaphylaxis and hypersensitivity to an antigenic substance or material.
  • Cadherins are calcium-dependent adhesion molecules that appear to play major roles during development, particularly in defining specific cell types. Loss or alteration of normal cadherin expression can lead to changes in cell adhesion properties linked to tumor growth and metastasis. Cadherin malfunction is also implicated in other human diseases, such as pemphigus vulgaris and pemphigus foliaceus (auto-immune blistering skin diseases), Crohn's disease, and some developmental abnormaUties.
  • the cadherin superfarmly includes well over forty members, each with a distinct pattern of expression. All members of the superfamily have in common conserved
  • cadherin domains 34 extraceUular repeats (cadherin domains), but structural differences are found in other parts of the molecule.
  • the cadherin domains bind caldum to form their tertiary structure and thus calcium is required to mediate their adhesion.
  • Only a few amino acids in the first cadherin domain provide the basis for homophiUc adhesion; modification of this recognition site can change the specificity of a cadherin so that instead of recognizing only itself, the mutant molecule can now also bind to a different cadherin.
  • some cadherins engage in heterophiUc adhesion with other cadherins.
  • E-cadherin one member of the cadherin superfamily, is expressed in epitheUal ceU types.
  • PathologicaUy if E-cadherin expression is lost in a tumor, the maUgnant ceUs become invasive and the cancer metastasizes.
  • Transfection of cancer ceU lines with polynucleotides expressing E-cadherin has reversed cancer-associated changes by returning altered ceU shapes to normal, restoring cells' adhesiveness to each other and to their substrate, decreasing the cell growth rate, and drastically reducing anchorage- independent ceU growth.
  • reintiOducing E-cadherin expression reverts carcinomas to a less advanced stage.
  • proteins of the present invention with cadherin activity can be used to treat cancer. Introducing such proteins or polynucleotides into cancer cells can reduce or eliminate the cancerous changes observed in these cells by providing normal cadherin expression.
  • Cancer ceUs have also been shown to express cadherins of a different tissue type than their origin, thus aUowing these cells to invade and metastasize in a different tissue in the body.
  • Proteins of the present invention with cadherin activity, and polynucleotides of the present invention encoding such proteins, can be substituted in these cells for the inappropriately expressed cadherins, restoring normal cell adhesive properties and reducing or eUminating the tendency of the cells to metastasize.
  • proteins of the present invention with cadherin activity can be used to generate antibodies recognizing and binding to cadherins.
  • Such antibodies can be used to block the adhesion of inappropriately expressed tumor-ceU cadherins, preventing the cells from forming a tumor elsewhere.
  • Such an anti-cadherin antibody can also be used as a marker for the grade, pathological type, and prognosis of a cancer, i.e. the more progressed the cancer, the less cadherin expression there wiU be, and this decrease in cadherin expression can be detected by the use of a cadherin-binding antibody.
  • Fragments of proteins of the present invention with cadherin activity preferably a polypeptide comprising a decapeptide of the cadherin recognition site, and polynucleotides of the present invention encoding such protein fragments, can also be used to block cadherin function by binding to cadherins and preventing them from binding in ways that produce undesirable effects.
  • AdditionaUy fragments of proteins of the present invention with cadherin activity, preferably truncated soluble cadherin fragments which have been found to be stable in the circulation of cancer patients, and polynucleotides encoding such protein fragments, can be used to disturb proper ceU-ceU adhesion.
  • Assays for cadherin adhesive and invasive suppressor activity include, without Umitation, those described in: Hortsch et al. J Biol Chem 270 (32): 18809-18817, 1995; Miyaki et al. Oncogene 11: 2547-2552, 1995; Ozawa et al. Cell 63: 1033-1038, 1990.
  • a protein of the invention may exhibit other anti-tumor activities.
  • a protein may inhibit tumor growth directly or indirectly (such as, for example, via antibody-dependent ceU-mediated cytotoxidty (ADCC)).
  • a protein may exhibit its tumor inhibitory activity by acting on tumor tissue or tumor precursor tissue, by inhibiting formation of tissues necessary to support tumor growth (such as, for example, by inhibiting angiogenesis), by causing production of other factors, agents or ceU types which inhibit tumor growth, or by suppressing, eliminating or inhibiting factors, agents or cell types which promote tumor growth.
  • a protein of the invention may also exhibit one or more of the f oUowing additional activities or effects: inhibiting the growth, infection or function of, or killing, infectious agents, including, without Umitation, bacteria, viruses, fungi and other parasites; effecting (suppressing or enhancing) bodily characteristics, including, without limitation, height, weight, hair color, eye color, skin, fat to lean ratio or other tissue pigmentation, or organ or body part size or shape (such as, for example, breast augmentation or diminution, change in bone form or shape); effecting biorhythms or caricadic cycles or rhythms; effecting the fertility of male or female subjects; effecting the metabolism, catabolism, anabolism, processing, utilization, storage or elimination of dietary fat, lipid, protein, carbohydrate, vitamins, minerals, cofactors or other nutritional factors or component(s);
  • a protein of the present invention may be used in a pharmaceutical composition when combined with a pharmaceutically acceptable carrier.
  • a pharmaceutically acceptable carrier may also contain (in addition to protein and a carrier) diluents, fillers, salts, buffers, stabilizers, solubiUzers, and other materials weU known in the art.
  • pharmaceuticalaUy acceptable means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s). The characteristics of the carrier wiU depend on the route of administration.
  • the pharmaceutical composition of the invention may also contain cytokines, lymphokines, or other hematopoietic factors such as M-CSF, GM-CSF, TNF, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IFN, TNFO, TNF1, TNF2, G-CSF, Meg-CSF, thrombopoietin, stem ceU factor, and erythropoietin.
  • the pharmaceutical composition may further contain other agents which either enhance the activity of the protein or compliment its activity or use in treatment.
  • protein of the present invention may be included in formulations of the particular cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent to rninimize side effeds of the cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent.
  • a protein of the present invention may be active in multimers (e.g., heterodimers or homodimers) or complexes with itself or other proteins.
  • multimers e.g., heterodimers or homodimers
  • complexes with itself or other proteins.
  • compositions of the invention may comprise a protein of the invention in such multimeric or complexed form.
  • the pharmaceutical composition of the invention may be in the form of a complex of the protein(s) of present invention along with protein or peptide antigens.
  • the protein and/or peptide antigen wiU deUver a stimulatory signal to both B and T lymphocytes.
  • B lymphocytes wiU respond to antigen through their surface immunoglobulin receptor.
  • T lymphocytes wiU respond to antigen through the T ceU receptor (TCR) foUowing presentation of the antigen by MHC proteins.
  • TCR T ceU receptor
  • MHC and structuraUy related proteins including those encoded by rouge I and class II MHC genes on host ceUs will serve to present the peptide antigen(s) to T lymphocytes.
  • the antigen components could also be suppUed as purified MHC-peptide complexes alone or with co-stimulatory molecules that can directly signal T cells.
  • antibodies able to bind surface immunolgobuUn and other molecules on B ceUs as well as antibodies able to bind the TCR and other molecules on T ceUs can be combined with the pharmaceutical composition of the invention.
  • the pharmaceutical composition of the invention may be in the form of a Uposome in which protein of the present invention is combined, in addition to other pharmaceuticaUy acceptable carriers, with amphipathic agents such as Upids which exist in aggregated form as micelles, insoluble monolayers, Uquid crystals, or lameUar layers in aqueous solution.
  • Suitable Upids for Uposomal formulation indude without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like.
  • Preparation of sud Uposomal formulations is within the level of skill in the art, as disclosed, for example, in U.S. Patent No.4,235,871; U.S. Patent No. 4,501,728; U.S. Patent No.4,837,028; and U.S. Patent No.4,737,323, all of which are incorporated herein by reference.
  • the term "therapeutically effective amount” means the total amount of each active component of the pharmaceutical composition or method that is sufficient to show a meaningful patient benefit, i.e., treatment, healing, prevention or ameUoration of the relevant medical condition, or an increase in rate of treatment, healing, prevention or amelioration of such conditions.
  • a meaningful patient benefit i.e., treatment, healing, prevention or ameUoration of the relevant medical condition, or an increase in rate of treatment, healing, prevention or amelioration of such conditions.
  • the term refers to that ingredient alone.
  • the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, seriaUy or simultaneously.
  • a therapeuticaUy effective amount of protein of the present invention is administered to a mammal having a condition to be treated.
  • Protein of the present invention may be administered in accordance with the method of the invention either alone or in combination with other therapies such as treatments employing cytokines, lymphokines or other hematopoietic factors.
  • protein of the present invention may be administered either simultaneously with the cytokine(s), lymphokine(s), other hematopoietic factor(s), thrombolytic or anti-thrombotic factors, or sequentiaUy. If administered sequentially, the attending physician will decide on the appropriate sequence of administering protein of the present invention in combination with cytokine(s), lymphokine(s), other hematopoietic factor(s), thrombolytic or anti-thrombotic factors.
  • Administration of protein of the present invention used in the pharmaceutical composition or to practice the method of the present invention can be carried out in a variety of conventional ways, such as oral ingestion, inhalation, topical appUcation or cutaneous, subcutaneous, intraperitoneal, parenteral or intravenous injection. Intravenous administration to the patient is preferred.
  • protein of the present invention When a therapeuticaUy effective amount of protein of the present invention is administered oraUy, protein of the present invention will be in the form of a tablet, capsule, powder, solution or elixir.
  • the pharmaceutical composition of the invention may additionaUy contain a solid carrier such as a gelatin or an adjuvant.
  • the tablet, capsule, and powder contain from about 5 to 95% protein of the present invention, and preferably from about 25 to 90% protein of the present invention.
  • a Uquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oU, soybean oil, or sesame oil, or synthetic oUs may be added.
  • the liquid form of the pharmaceutical composition may further contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol.
  • the pharmaceutical composition When administered in liquid form, contains from about 0.5 to 90% by weight of protein of the present invention, and preferably from about 1 to 50% protein of the present invention.
  • protein of the present invention When a therapeuticaUy effective amount of protein of the present invention is administered by intravenous, cutaneous or subcutaneous injection, protein of the present
  • invention wiU be in the form of a pyrogen-free, parenteraUy acceptable aqueous solution.
  • a preferred pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to protein of the present invention, an isotor ⁇ c vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection, or other vehicle as known in the art.
  • the pharmaceutical composition of the present invention may also contain stabuizers, preservatives, buffers, antioxidants, or other additives known to those of skill in the art.
  • the amount of protein of the present invention in the pharmaceutical composition of the present invention wiU depend upon the nature and severity of the condition being treated, and on the nature of prior treatments which the patient has undergone.
  • the attending physician wUl decide the amount of protein of the present invention with which to treat each individual patient. Initially, the attending physician will administer low doses of protein of the present invention and observe the patient's response. Larger doses of protein of the present invention may be administered until the optimal therapeutic effect is obtained for the patient, and at that point the dosage is not increased further.
  • compositions used to practice the method of the present invention should contain about 0.01 ⁇ g to about 100 mg (preferably about O.lng to about 10 mg, more preferably about 0.1 ⁇ g to about 1 mg) of protein of the present invention per kg body weight.
  • the duration of intravenous therapy using the pharmaceutical composition of the present invention wiU vary, depending on the severity of the disease being treated and the condition and potential idiosyncratic response of each individual patient. It is contemplated that the duration of each appUcation of the protein of the present invention will be in the range of 12 to 24 hours of continuous intravenous administration. Ultimately the attending physician wiU dedde on the appropriate duration of intravenous therapy using the pharmaceutical composition of the present invention.
  • Protein of the invention may also be used to immunize animals to obtain polydonal and monoclonal antibodies which specifically react with the protein.
  • antibody includes without limitation a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a single-chain antibody, a CDR-grafted antibody, a humanized antibody, or fragments thereof which bind to the indicated protein.
  • Such term also includes any other species derived from an antibody or antibody sequence which is capable of binding the indicated protein.
  • Antibodies to a particular protein can be produced by methods well known to those skilled in the art.
  • monoclonal antibodies can be produced by generation of antibody-producing hybridomas in accordance with known methods (see for example, Goding, 1983, Monoclonal antibodies: principles and practice, Academic Press Inc., New York; and Yokoyama, 1992, "Production of Monoclonal Antibodies” in Current Protocols in Immunology, Unit 2.5, Greene Publishing Assoc. and John Wiley & Sons).
  • Polyclonal sera and antibodies can be produced by inoculation of a mammalian subject with the relevant protein or fragments thereof in accordance with known methods.
  • Fragments of antibodies, receptors, or other reactive peptides can be produced from the corresponding antibodies by cleavage of and collection of the desired fragments in accordance with known methods (see for example, Goding, supra; and Andrew et al., 1992, "Fragmentation of Immunoglobulins” in Current Protocols in Immunology, Unit 2.8, Greene Publishing Assoc. and John Wiley & Sons).
  • Chimeric antibodies and single chain antibodies can also be produced in accordance with known recombinant methods (see for example, 5,169,939, 5,194,594, and 5,576,184).
  • Humanized antibodies can also be made from corresponding murine antibodies in accordance with well known methods (see for example, U.S. Patent Nos.
  • human antibodies may be produced in non-human animals such as mice that have been genetically altered to express human antibody molecules (see for example Fishwild et al, 1996, Nature Biotechnology 14: 845-851; Mendez etal, 1997, Nature Genetics 15: 146-156 (erratum Nature Genetics 16: 410); and U.S. Patents 5,877,397 and 5,625,126).
  • Such antibodies may be obtained using either the entire protein or fragments thereof as an immunogen.
  • the peptide immunogens additionaUy may contain a cysteine residue at the carboxyl terminus, and are conjugated to a hapten such as keyhole limpet hemocyanin (KLH).
  • KLH keyhole limpet hemocyanin
  • Monoclonal antibodies binding to the protein of the invention may be useful diagnostic agents for the immunodetection of the protein.
  • Neutralizing monoclonal antibodies binding to the protein may also be useful therapeutics for both conditions associated with the protein and also in the treatment of some forms of cancer where
  • the therapeutic method includes administering the composition topically, systematically, or locaUy as an implant or device.
  • the therapeutic composition for use in this invention is, of course, in a pyrogen-free, physiologically acceptable form.
  • the composition may desirably be encapsulated or injected in a viscous form for delivery to the site of bone, cartilage or tissue damage.
  • Topical administration may be suitable for wound healing and tissue repair.
  • TherapeuticaUy useful agents other than a protein of the invention which may also optionaUy be included in the composition as described above, may alternatively or additionaUy, be administered simultaneously or sequentiaUy with the composition in the methods of the invention.
  • the composition would include a matrix capable of delivering the protem-containing composition to the site of bone and/or cartilage damage, providing a structure for the developing bone and cartilage and optimally capable of being resorbed into the body.
  • Such matrices may be formed of materials presently in use for other implanted medical applications.
  • compositions may be biodegradable and chemically defined calcium sulfate, tricalciumphosphate, hydroxyapatite, polylactic acid, polyglycoUc acid and polyanhydrides.
  • potential materials are biodegradable and biologicaUy weU- defined, such as bone or dermal collagen.
  • Further matrices are comprised of pure proteins or extraceUular matrix components.
  • Other potential matrices are nonbiodegradable and chemicaUy defined, such as sintered hydroxapatite, bioglass, aluminates, or other ceramics.
  • Matrices may be comprised of combinations of any of the above mentioned types of material, such as polylactic acid and hydroxyapatite or coUagen and tricaldumphosphate.
  • the bioceramics may be altered in composition, such as in caldum- aluminate-phosphate and processing to alter pore size, particle size, particle shape, and biodegradability.
  • a 50:50 (mole weight) copolymer of lactic acid and glycolic acid in the form of porous particles having diameters ranging from 150 to 800 microns.
  • a sequestering agent such as carboxymethyl cellulose or autologous blood clot, to prevent the protein compositions from disassociating from the matrix.
  • ceUulosic materials such as alkylcelluloses (including hydroxyalkylceUuloses), including methylcellulose, ethylceUulose, hydroxyethylceUulose, hydroxypropylcellulose, hydroxypropyl- methylcellulose, and carboxymethylceUulose, the most preferred being canonic salts of carboxymethylceUulose (CMC).
  • CMC carboxymethylceUulose
  • Other preferred sequestering agents include hyaluronic acid, sodium alginate, poly(ethylene glycol), polyoxyethylene oxide, carboxyvinyl polymer and poly(vinyl alcohol).
  • the amount of sequestering agent useful herein is 0.5-20 wt%, preferably 1-10 wt% based on total formulation weight, which represents the amount necessary to prevent desorbtion of the protein from the polymer matrix and to provide appropriate handling of the composition, yet not so much that the progenitor cells are prevented from infiltrating the matrix, thereby providing the protein the opportunity to assist the osteogenic activity of the progenitor ceUs.
  • proteins of the invention may be combined with other agents benefidal to the treatment of the bone and /or cartilage defed, wound, or tissue in question.
  • agents include various growth factors such as epidermal growth factor
  • EGF platelet derived growth factor
  • TGF- ⁇ transforming growth factors
  • TGF- ⁇ TGF- ⁇
  • IGF insulin-like growth factor
  • the therapeutic compositions are also presently valuable for veterinary applications. Particularly domestic animals and thoroughbred horses, in addition to humans, are desired patients for such treatment with proteins of the present invention.
  • the dosage regimen of a protem-containing pharmaceutical composition to be used in tissue regeneration wiU be determined by the attending physician considering various factors which modify the action of the proteins, e.g., amount of tissue weight desired to be formed, the site of damage, the condition of the damaged tissue, the size of a wound, type of damaged tissue (e.g., bone), the patient's age, sex, and diet, the severity of any infection, time of administration and other clinical factors.
  • the dosage may vary with the type of matrix used in the reconstitution and with inclusion of other proteins in the pharmaceutical composition.
  • the addition of other known growth factors, such as IGF I (insuUn like growth factor I) may also effect
  • Progress can be monitored by periodic assessment of tissue/bone growth and /or repair, for example, X-rays, histomorphometric determinations and tetracycUne labeling.
  • Polynudeotides of the present invention can also be used for gene therapy. Such polynucleotides can be introduced either in vivo or ex vivo into ceUs for expression in a mammaUan subjed. Polynudeotides of the invention may also be administered by other known methods for introduction of nudeic add into a ceU or organism (including, without
  • Cells may also be cultured ex vivo in the presence of proteins of the present invention in order to proliferate or to produce a desired effect on or activity in such cells. Treated ceUs can then be introduced in vivo for therapeutic purposes.

Landscapes

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

Abstract

A novel secreted and glycophosphatidylinositol-anchored protein, GPI-122, is disclosed, as are antibodies that specifically bind to this protein. Polynucleotides encoding GPI-122 are also provided.

Description

GPI-122, A NOVEL GLYCOPHOSPHATIDYLLNOSLTOL-ANCHORED PROTEIN
This application is a continuation-in-part of application Ser. No. 60/XXX,XXX
(converted to a provisional application from non-provisional application Ser. No. 09/049,652), filed March 27, 1998, which is incorporated by reference herein.
HELD OF THE INVENTION
The present invention provides novel polynucleotides and proteins encoded by such polynucleotides, along with therapeutic, diagnostic and research utilities for these polynucleotides and proteins.
BACKGROUND OF THE INVENTION
Technology aimed at the discovery of protein factors (including e.g., cytokines, such as lymphokines, interferons, CSFs and interleukins) has matured rapidly over the past decade. The now routine hybridization cloning and expression cloning techniques clone novel polynucleotides "directly" in the sense that they rely on information directly related to the discovered protein (i.e., partial DNA/amino acid sequence of the protein in the case of hybridization cloning; activity of the protein in the case of expression cloning). More recent "indirect" cloning techniques such as signal sequence cloning, which isolates DNA sequences based on the presence of a now well-recognized secretory leader sequence motif, as well as various PCR-based or low stringency hybridization cloning techniques, have advanced the state of the art by making available large numbers of
DNA/amino acid sequences for proteins that are known to have biological activity by virtue of their secreted nature in the case of leader sequence cloning, or by virtue of the cell or tissue source in the case of PCR-based techniques. It is to these proteins and the polynucleotides encoding them that the present invention is directed. SUMMARY OF THE LNVENΗON In one embodiment, the present invention provides a composition comprising an isolated polynucleotide selected from the group consisting of:
(a) a polynucleotide comprising the nucleotide sequence of SEQ ID NO:l;
(b) a polynucleotide comprising the nucleotide sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3730;
(c) a polynucleotide comprising the nucleotide sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3640; (d) a polynucleotide comprising the nucleotide sequence of the full- length protein coding sequence of clone GPI-122 deposited under accession number ATCC 98622;
(e) a polynucleotide encoding the full-length protein encoded by the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622; (f) a polynucleotide comprising the nucleotide sequence of a mature protein coding sequence of clone GPI-122 deposited under accession number ATCC 98622;
(g) a polynucleotide encoding a mature protein encoded by the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622; (h) a polynucleotide encoding a protein comprising the amino acid sequence of SEQ ID NO:2;
(i) a polynucleotide encoding a protein comprising a fragment of the amino acid sequence of SEQ ID NO:2 having biological activity, the fragment comprising eight contiguous amino acids of SEQ ID NO:2; (j) a polynucleotide which is an allelic variant of a polynucleotide of
(a)-(g) above;
(k) a polynucleotide which encodes a species homologue of the protein of (h) or (i) above ;
(1) a polynucleotide that hybridizes under stringent conditions to any one of the polynucleotides specified in (a)-(i); and
(m) a polynucleotide that hybridizes under stringent conditions to any one of the polynucleotides specified in (a)-(i) and that has a length that is at least 25% of the length of SEQ ID NO:l. Preferably, such polynucleotide comprises the nucleotide sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3730; the nucleotide sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3640; the nucleotide sequence of the full-length protein coding sequence of clone GPI-122 deposited under accession number ATCC 98622; or the nucleotide sequence of a mature protein coding sequence of clone GPI-122 deposited under accession number ATCC 98622. In other preferred embodiments, the polynucleotide encodes the full-length or a mature protein encoded by the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622. In yet other preferred embodiments, the present invention provides a polynucleotide encoding a protein comprising the amino acid sequence of SEQ ID NO:2 from amino acid 1 to amino acid 1163. In further preferred embodiments, the present invention provides a polynucleotide encoding a protein comprising a fragment of the amino acid sequence of SEQ ID NO:2 having biological activity, the fragment preferably comprising eight (more preferably twenty, most preferably thirty) contiguous amino acids of SEQ ID NO:2, or a polynucleotide encoding a protein comprising a fragment of the amino acid sequence of SEQ ID NO:2 having biological activity, the fragment comprising the amino acid sequence from amino acid 591 to amino acid 600 of SEQ ID NO:2.
Other embodiments provide the gene corresponding to the cDNA sequence of SEQ ID NO:l. Further embodiments of the invention provide isolated polynucleotides produced according to a process selected from the group consisting of: (a) a process comprising the steps of:
(i) preparing one or more polynucleotide probes that hybridize in 6X SSC at 65 degrees C to a nucleotide sequence selected from the group consisting of:
(aa) SEQ ID NO:l, but excluding the poly(A) tail at the 3' end of SEQ ID NO:l; and
(ab) the nucleotide sequence of the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622; (ϋ) hybridizing said probe(s) to human genomic DNA in conditions at least as stringent as 4X SSC at 50 degrees C; and
(iii) isolating the DNA polynucleotides detected with the probe(s); and (b) a process comprising the steps of:
(i) preparing one or more polynucleotide primers that hybridize in 6X SSC at 65 degrees C to a nucleotide sequence selected from the group consisting of: (ba) SEQ ID NO:l, but excluding the poly(A) tail at the
3' end of SEQ ID NO:l; and
(bb) the nucleotide sequence of the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622; (ii) hybridizing said primer(s) to human genomic DNA in conditions at least as stringent as 4X SSC at 50 degrees C;
(iii) amplifying human DNA sequences; and (iv) isolating the polynucleotide products of step (b)(iii). Preferably the polynucleotide isolated according to the above process comprises a nucleotide sequence corresponding to the cDNA sequence of SEQ ID NO:l, and extending contiguously from a nucleotide sequence corresponding to the 5' end of SEQ ID NO:l to a nucleotide sequence corresponding to the 3' end of SEQ ID NO:l , but excluding the poly(A) tail at the 3' end of SEQ ID NO:l. Also preferably the polynucleotide isolated according to the above process comprises a nucleotide sequence corresponding to the cDNA sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3730, and extending contiguously from a nucleotide sequence corresponding to the 5' end of said sequence of
SEQ ID NO:l from nucleotide 152 to nucleotide 3730, to a nucleotide sequence corresponding to the 3' end of said sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3730. Also preferably the polynucleotide isolated according to the above process comprises a nucleotide sequence corresponding to the cDNA sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3640, and extending contiguously from a nucleotide sequence corresponding to the 5' end of said sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3640, to a nucleotide sequence corresponding to the 3' end of said sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3640.
In other embodiments, the present invention provides a composition comprising a protein, wherein said protein comprises an amino acid sequence selected from the group consisting of:
(a) the amino acid sequence of SEQ ID NO:2;
(b) the amino acid sequence of SEQ ID NO:2 from amino acid 1 to amino acid 1163; (c) a fragment of the amino acid sequence of SEQ ID NO:2, the fragment comprising eight contiguous amino acids of SEQ ID NO:2; and
(d) the amino acid sequence encoded by the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622; the protein being substantially free from other mammalian proteins. Preferably such protein comprises the amino acid sequence of SEQ ID NO:2 or the amino acid sequence of SEQ ID NO:2 from amino acid 1 to amino acid 1163. In further preferred embodiments, the present invention provides a protein comprising a fragment of the amino acid sequence of SEQ ID NO:2 having biological activity, the fragment preferably comprising eight (more preferably twenty, most preferably thirty) contiguous amino acids of SEQ ID NO:2, or a protein comprising a fragment of the amino acid sequence of SEQ ID NO:2 having biological activity, the fragment comprising the amino acid sequence from amino acid 591 to amino acid 600 of SEQ ID NO:2.
In certain preferred embodiments, the polynucleotide is operably linked to an expression control sequence. The invention also provides a host cell, including bacterial, yeast, insect and mammalian cells, transformed with such polynucleotide compositions. Also provided by the present invention are organisms that have enhanced, reduced, or modified expression of the gene(s) corresponding to the polynucleotide sequences disclosed herein. Processes are also provided for producing a protein, which comprise:
(a) growing a culture of the host cell transformed with such polynucleotide compositions in a suitable culture medium; and
(b) purifying the protein from the culture.
The protein produced according to such methods is also provided by the present invention.
Protein compositions of the present invention may further comprise a pharmaceutically acceptable carrier. Compositions comprising an antibody which specifically reacts with such protein are also provided by the present invention. Preferably, such antibody is a monoclonal antibody; also preferably, such antibody is a human antibody.
Methods are also provided for preventing, treating or ameliorating a medical condition which comprises administering to a mammalian subject a therapeutically effective amount of a composition comprising a protein of the present invention and a pharmaceutically acceptable carrier. DETAILED DESCRIPTION ISOLATED PROTEINS AND POLYNUCLEOTIDES
Applicants have identified a novel human protein, GPI-122, which contains within its amino acid sequence residues that signal the addition of a glycophosphatidylinositol (GPI) anchor.
Certain cell-surface proteins undergo post-translational modification resulting in the covalent addition of a GPI anchor, which serves as their only means of attachment to the cell membrane (Robinson, 1991, Immunology Today 12(1): 35-41, incorporated by reference herein). GPI anchors are found to be attached to proteins in mammals, as well as to proteins in lower eukaryotes such as yeast and protozoa. In all cells they are found on the external face of the cell membrane or on the lumenal surface of secretory vesicles. A wide variety of proteins have GPI anchors, including parasite coat proteins, lymphoid antigens (for example, Thy-1), hydrolytic enzymes, cell adhesion molecules, receptors for small molecules (for example, folate receptor), and the neurturin receptor (Klein et al, 1997, Nature 387: 717-721; Buj-Bello et al, 1997, Nature 387: 721-724). GPI anchors share a core glyco-inositol-1-phospholipid structure, suggesting that a common GPI biosynthetic pathway exists in all eukaryotes. However, GPI anchors have varying substitutents attached to the carbohydrate portion of the GPI anchor, and the lipid portions of GPI anchors from different species exhibit a striking diversity. (Englund, 1993, Ann. Rev. Biochem. 62: 121-138, incorporated by reference herein.) GPI anchors can be specifically cleaved through the use of phosphoinositide-specific phospholipase C (PIPLC) (Koke et al., 1991, Protein Expr. Purif. 2(1): 51-58, incorporated by reference herein); treatment of cells with PIPLC releases GPI-anchored proteins from the cell surface.
The primary translation products for GPI-anchored proteins have a predominantly hydrophobic sequence at their carboxyl terminus, ranging in size from 17 to about 30 residues, which signals the addition of the GPI anchor and which is cleaved off at the time of anchor addition. Just aminc^terrninal to the hydrophobic sequence is a stretch of about five to ten hydrophilic amino acids. The residue to which the anchor is added is always one of the small amino acids, such as glycine, aspartic acid, asparagine, alanine, serine, threonine, or cysteine. In addition, the two residues carboxy-terminal to the anchor addition site are usually small amino acids also, so that the cleavage/anchor addition site resides in a domain of three small residues. (Englund, 1993, Ann. Rev. Biochem. 62: 121- Nucleotide and arrtino acid sequences, as presently determined, are reported below for the GPI-122 clone and protein disclosed in the present application. The nucleotide sequence of the GPI-122 clone can readily be determined by sequencing the deposited clone in accordance with known methods. The predicted amino acid sequence (both full-length and mature forms) can then be determined from such nucleotide sequence. The amino acid sequence of the protein encoded by a particular clone can also be determined by expression of the clone in a suitable host cell, collecting the protein and determining its sequence. For the disclosed GPI-122 protein, applicants have identified what they have determined to be the reading frame best identifiable with sequence information available at the time of filing.
As used herein a "secreted" protein is one which, when expressed in a suitable host cell, is transported across or through a membrane, including transport as a result of signal sequences in its amino acid sequence. "Secreted" proteins include without limitation proteins secreted wholly (e.g., soluble proteins) or partially (e.g., receptors) from the cell in which they are expressed. "Secreted" proteins also include without limitation proteins which are transported across the membrane of the endoplasmic reticulum.
Clone "GPI-122"
A polynucleotide of the present invention has been identified as clone "GPI-122". GPI-122 is a full-length clone, including the entire coding sequence of a GPI-anchored protein (also referred to herein as "GPI-122 protein")
The GPI-122 clone was isolated through the following procedure. The BLAST computer program was used to search the GenBank EST (expressed sequence tag) database for sequences with significant similarity to the nucleotide sequence of Ba3.1 (SEQ ID NO:3), a clone that also encodes a novel GPI-anchored protein. Several overlapping human EST sequences were identified with a significant degree of sequence similarity to the Ba3.1 nucleotide sequence. The longest clone from among these EST sequences, GenBank accession number R99646 (yq73e04.rl Homo sapiens cDNA clone 2014385'), was obtained from the IMAGE consortium and its complete nucleotide sequence was determined. Analysis of the open reading frames encoded by the R99646 clone revealed that the R99646 clone was not a full-length clone because it did not contain an entire protein-coding sequence. A probe was made using sequence from the 5' end of the R99646 clone; using this probe, two additional clones, each larger than the R99646 clone, were isolated from a phagemid human fetal spleen cDNA library by hybridization under high stringency conditions. The full-length sequences of these clones were determined and it was found that both were still only partial cDNA clones. Using 3' PCR primers derived from the 5' end of these human fetal spleen cDNA clones and vector-derived 5' PCR primers, 5' RACE reactions (Ohara et al, 1989, Proc. Natl. Acad. Sci. USA 86: 5673- 5677, which is incorporated by reference herein) were performed on a human skeletal muscle cDNA library and the RACE reaction products sequenced until a product was identified that contained sequence upstream of the 5' end of an open reading frame. To link the sequence from the 5' RACE reaction product to that of the human fetal spleen cDNA clones, a PCR reaction was performed on a human skeletal muscle library using a first PCR primer derived from the 5' RACE sequence and a second PCR primer derived from a portion of the longest human fetal spleen cDNA clone that was 3' to a SphI restriction site. The resulting "linking" PCR product and the longest human fetal spleen cDNA clone were digested with Notl and SphI, and the appropriate restriction fragments were gel-purified and ligated together. The ligated complete GPI-122 clone was then sequenced in its entirety to confirm that the expected nucleotide sequence was present.
The nucleotide sequence of GPI-122 as presently determined is reported in SEQ
ID NO:l. What applicants presently believe to be the proper reading frame and the predicted amino acid sequence of the GPI-122 protein corresponding to the foregoing nucleotide sequence is reported in SEQ ID NO:2. Amino acids 207 to 219 of SEQ ID NO:2 are a possible leader /signal sequence, with a predicted mature amino acid sequence beginning at amino acid 220; amino acids 310 to 322 of SEQ ID NO:2 are also a possible leader/signal sequence, with a predicted mature amino acid sequence beginning at amino acid 323. Amino acids 1175 to 1193 are a hydrophobic tail region. Arnino-terminal to the hydrophobic tail region is a stretch of several hydrophilic amino acids. Amino acids 1163- 1165 are three small amino acids (threonine, cysteine, and asparagine, respectively) that together with the hydrophobic tail and hydrophilic stretch appear to be a recognition sequence for the addition of a GPI anchor to the threonine 1163 residue, with a concomitant cleavage predicted to remove residues 1164-1193 of the protein.
Clone GPI-122 was deposited on December 19, 1997 with the American Type Culture Collection as an original deposit under the Budapest Treaty and was given the accession number ATCC 98622. All restrictions on the availability to the public of the deposited material will be irrevocably removed upon the granting of the patent, except for the requirements specified in 37 C.F.R. § 1.808(b), and the term of the deposit will comply with 37 C.F.R. § 1.806. The clone can be removed from the vector in which it was deposited by performing a Notl/Kpnl digestion (5' site, NotI; 3' site Kpnl). The Notl/Kpnl restriction fragment obtainable from the deposit containing clone GPI-122 should be approximately 5920 bp.
The nucleotide sequence disclosed herein for GPI-122 was searched against the GenBank and GeneSeq nucleotide sequence databases using the BLASTN search protocol. GPI-122 demonstrated at least some similarity with sequences identified as Q60081 (Human brain Expressed Sequence Tag EST02065), as well as several EST sequences including that of GenBank accession number R99646. The predicted amino acid sequence disclosed herein for GPI-122 was searched against the GenPept and GeneSeq amino acid sequence databases using the BLASTP search protocol. The predicted GPI-122 protein demonstrated at least some similarity to sequences identified as Z68314 (K08F8.6 [Caenorhabditis elegans]). Based upon sequence similarity, GPI-122 proteins and each similar protein or peptide may share at least some activity. The TopPredϋ computer program predicts four potential transmembrane domains within the GPI-122 protein sequence, centered around amino acids 500, 900, 980, and 1030 of SEQ ID NO:2, respectively.
Fragments of the proteins of the present invention which are capable of exhibiting biological activity are also encompassed by the present invention. Fragments of the protein may be in linear form or they may be cyclized using known methods, for example, as described in H.U. Saragovi, et al, Bio /Technology 10, 773-778 (1992) and in R.S. McDowell, et al, J. Amer. Chem. Soc. 114, 9245-9253 (1992), both of which are incorporated herein by reference. Such fragments may be fused to carrier molecules such as immunoglobulins for many purposes, including increasing the valency of protein binding sites. For example, fragments of the protein may be fused through "linker" sequences to the Fc portion of an immunoglobulin. For a bivalent form of the protein, such a fusion could be to the Fc portion of an IgG molecule. Other immunoglobulin isotypes may also be used to generate such fusions. For example, a protein - IgM fusion would generate a decavalent form of the protein of the invention. The present invention also provides both full-length and mature forms of the disclosed proteins. The full-length form of the such proteins is identified in the sequence listing by translation of the nucleotide sequence of each disclosed clone. The mature form(s) of such protein may be obtained by expression of the disclosed full-length polynucleotide (preferably those deposited with ATCC) in a suitable mammalian cell or other host cell. The sequence(s) of the mature form(s) of the protein may also be determinable from the amino acid sequence of the full-length form.
The present invention also provides genes corresponding to the polynucleotide sequences disclosed herein. "Corresponding genes" are the regions of the genome that are transcribed to produce the mRNAs from which cDNA polynucleotide sequences are derived and may include contiguous regions of the genome necessary for the regulated expression of such genes. Corresponding genes may therefore include but are not limited to coding sequences, 5' and 3' untranslated regions, alternatively spliced exons, introns, promoters, enhancers, and silencer or suppressor elements. The corresponding genes can be isolated in accordance with known methods using the sequence information disclosed herein. Such methods include the preparation of probes or primers from the disclosed sequence information for identification and /or amplification of genes in appropriate genomic libraries or other sources of genomic materials. An "isolated gene" is a gene that has been separated from the adjacent coding sequences, if any, present in the genome of the organism from which the gene was isolated.
The chromosomal location corresponding to the polynucleotide sequences disclosed herein may also be determined, for example by hybridizing appropriately labeled polynucleotides of the present invention to chromosomes in situ. It may also be possible to determine the corresponding chromosomal location for a disclosed polynucleotide by identifying significantly similar nucleotide sequences in public databases, such as expressed sequence tags (ESTs), that have already been mapped to particular chromosomal locations. For at least some of the polynucleotide sequences disclosed herein, public database sequences having at least some similarity to the polynucleotide of the present invention have been listed by database accession number. Searches using the GenBank accession numbers of these public database sequences can then be performed at an Internet site provided by the National Center for Biotechnology Information having the address http://www.ncbi.nlm.nih.gov/UniGene/, in order to identify "UniGene clusters" of overlapping sequences. Many of the "UniGene clusters" so identified will already have been mapped to particular chromosomal sites. Organisms that have enhanced, reduced, or modified expression of the gene(s) corresponding to the polynucleotide sequences disclosed herein are provided. The desired change in gene expression can be achieved through the use of antisense polynucleotides or ribozymes that bind and/or cleave the mRNA transcribed from the gene (Albert and Morris, 1994, Trends Pharmacol. Sci. 15(7): 250-254; Lavarosky et al, 1997,
10 Biochem. Mol. Med. 62(1): 11-22; and Hampel, 1998, Prog. Nucleic Acid Res. Mol. Biol. 58: 1- 39; all of which are incorporated by reference herein). Transgenic animals that have multiple copies of the gene(s) corresponding to the polynucleotide sequences disclosed herein, preferably produced by transformation of cells with genetic constructs that are stably maintained within the transformed cells and their progeny, are provided. Transgenic animals that have modified genetic control regions that increase or reduce gene expression levels, or that change temporal or spatial patterns of gene expression, are also provided (see European Patent No. 0 649 464 Bl, incorporated by reference herein). In addition, organisms are provided in which the gene(s) corresponding to the polynucleotide sequences disclosed herein have been partially or completely inactivated, through insertion of extraneous sequences into the corresponding gene(s) or through deletion of all or part of the corresponding gene(s). Partial or complete gene inactivation can be accomplished through insertion, preferably followed by imprecise excision, of transposable elements (Plasterk, 1992, Bioessays 14(9): 629-633; Zwaal et al, 1993, Proc. Natl. Acad. Sci. USA 90(16): 7431-7435; Clark et al, 1994, Proc. Natl. Acad. Sci. USA 91(2): 719-722; all of which are incorporated by reference herein), or through homologous recombination, preferably detected by positive/negative genetic selection strategies (Mansour et al, 1988, Nature 336: 348-352; U.S. Patent Nos. 5,464,764; 5,487,992; 5,627,059; 5,631,153; 5,614, 396; 5,616,491; and 5,679,523; all of which are incorporated by reference herein). These organisms with altered gene expression are preferably eukaryotes and more preferably are mammals. Such organisms are useful for the development of non-human models for the study of disorders involving the corresponding gene(s), and for the development of assay systems for the identification of molecules that interact with the protein product(s) of the corresponding gene(s). Where the protein of the present invention is membrane-bound (e.g., is a receptor), the present invention also provides for soluble forms of such protein. In such forms, part or all of the intracellular and transmembrane domains of the protein are deleted such that the protein is fully secreted from the cell in which it is expressed. The intracellular and transmembrane domains of proteins of the invention can be identified in accordance with known techniques for determination of such domains from sequence information. For example, the TopPredLI computer program can be used to predict the location of transmembrane domains in an amino acid sequence, domains which are described by the location of the center of the transmsmbrane domain, with at least ten transmembrane amino acids on each side of the reported central residue(s).
11 Proteins and protein fragments of the present invention include proteins with amino acid sequence lengths that are at least 25%(more preferably at least 50%, and most preferably at least 75%) of the length of a disclosed protein and have at least 60% sequence identity (more preferably, at least 75% identity; most preferably at least 90% or 95% identity) with that disclosed protein, where sequence identity is determined by comparing the amino acid sequences of the proteins when aligned so as to maximize overlap and identity while minimizing sequence gaps. Also included in the present invention are proteins and protein fragments that contain a segment preferably comprising 8 or more (more preferably 20 or more, most preferably 30 or more) contiguous amino acids that shares at least 75% sequence identity (more preferably, at least 85% identity; most preferably at least 95% identity) with any such segment of any of the disclosed proteins.
In particular, sequence identity may be determined using WU-BLAST
(Washington University BLAST) version 2.0 software, which builds upon WU-BLAST version 1.4, which in turn is based on the public domain NCBI-BLAST version 1.4 (Altschul and Gish, 1996, Local alignment statistics, Doolittle ed., Methods in Enzymology 266: 460-480; Altschul et al, 1990, Basic local alignment search tool, Journal of Molecular Biology 215: 403-410; Gish and States, 1993, Identification of protein coding regions by database similarity search, Nature Genetics 3: 266-272; Karlin and Altschul, 1993, Applications and statistics for multiple high-scoring segments in molecular sequences, Proc. Natl. Acad. Sci. USA 90: 5873-5877; all of which are incorporated by reference herein). WU-BLAST version 2.0 executable programs for several UNIX platforms can be downloaded from ftp://blast.wustl.edu/blast/executables. The complete suite of search programs (BLASTP, BLASTN, BLASTX, TBLASTN, and TBLASTX) is provided at that site, in addition to several support programs. WU-BLAST 2.0 is copyrighted and may not be sold or redistributed in any form or manner without the express written consent of the author; but the posted executables may otherwise be freely used for commercial, nonprofit, or academic purposes. In all search programs in the suite - BLASTP, BLASTN, BLASTX, TBLASTN and TBLASTX - the gapped alignment routines are integral to the database search itself, and thus yield much better sensitivity and selectivity while producing the more easily interpreted output. Gapping can optionally be turned off in all of these programs, if desired. The default penalty (Q) for a gap of length one is Q=9 for proteins and BLASTP, and Q=10 for BLASTN, but may be changed to any
12 integer value including zero, one through eight, nine, ten, eleven, twelve through twenty, twenty-one through fifty, fifty-one through one hundred, etc. The default per-residue penalty for extending a gap (R) is R=2 for proteins and BLASTP, and R=10 for BLASTN, but may be changed to any integer value including zero, one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve through twenty, twenty-one through fifty, fifty-one through one hundred, etc. Any combination of values for Q and R can be used in order to align sequences so as to maximize overlap and identity while nrinimizing sequence gaps. The default amino acid comparison matrix is BLOSUM62, but other amino acid comparison matrices such as PAM can be utilized. Species homologues of the disclosed polynucleotides and proteins are also provided by the present invention. As used herein, a "species homologue" is a protein or polynucleotide with a different species of origin from that of a given protein or polynucleotide, but with significant sequence similarity to the given protein or polynucleotide. Preferably, polynucleotide species homologues have at least 60% sequence identity (more preferably, at least 75% identity; most preferably at least 90% identity) with the given polynucleotide, and protein species homologues have at least 30% sequence identity (more preferably, at least 45% identity; most preferably at least 60% identity) with the given protein, where sequence identity is determined by comparing the nucleotide sequences of the polynucleotides or the amino acid sequences of the proteins when aligned so as to maximize overlap and identity while mirvimizing sequence gaps. Species homologues may be isolated and identified by making suitable probes or primers from the sequences provided herein and screening a suitable nucleic acid source from the desired species. Preferably, species homologues are those isolated from mammalian species. Most preferably, species homologues are those isolated from certain mammalian species such as, for example, Pan troglodytes, Gorilla gorilla, Pongo pygmaeus, Hylobates concolor, Macaca mulatta, Papio papio, Papio hamadryas, Cercopithecus aethiops, Cebus capucinus, Aotus trivirgatus, Sanguinus oedipus, Microcebus murinus, Mus musculus, Rattus norvegicus, Cricetulus griseus, Felis catus, Mustela vison, Canisfamiliaris, Oryctolagus cuniculus, Bos taurus, Ovis aries, Sus scrofa, and Equus caballus, for which genetic maps have been created allowing the identification of syntenic relationships between the genomic organization of genes in one species and the genomic organization of the related genes in another species (O'Brien and Seuanez, 1988, Ann. Rev. Genet. 22: 323-351; O'Brien et al, 1993, Nature Genetics 3:103-112; Johansson et al, 1995, Genomics 25: 682-690; Lyons et al, 1997, Nature
13 Genetics 15: 47-56; O'Brien et al., 1997, Trends in Genetics 13(10): 393-399; Carver and Stubbs, 1997, Genome Research 7:1123-1137; all of which are incorporated by reference herein).
The invention also encompasses allelic variants of the disclosed polynucleotides or proteins; that is, naturally-occurring alternative forms of the isolated polynucleotides which also encode proteins which are identical or have significantly similar sequences to those encoded by the disclosed polynucleotides. Preferably, allelic variants have at least 60% sequence identity (more preferably, at least 75% identity; most preferably at least 90% identity) with the given polynucleotide, where sequence identity is determined by comparing the nucleotide sequences of the polynucleotides when aligned so as to maximize overlap and identity while minimizing sequence gaps. Allelic variants may be isolated and identified by making suitable probes or primers from the sequences provided herein and screening a suitable nucleic acid source from individuals of the appropriate species.
The invention also includes polynucleotides with sequences complementary to those of the polynucleotides disclosed herein. The present invention also includes polynucleotides that hybridize under reduced stringency conditions, more preferably stringent conditions, and most preferably highly stringent conditions, to polynucleotides described herein. Examples of stringency conditions are shown in the table below: highly stringent conditions are those that are at least as stringent as, for example, conditions A-F; stringent conditions are at least as stringent as, for example, conditions G-L; and reduced stringency conditions are at least as stringent as, for example, conditions M-R.
14 Stringency Polynucleotide Hybrid Hybridization Temperature and Wash Condition Hybrid Length Buffer* Temperature (bp)t and Buffer*
A DNA:DNA ≥ 50 65°C; lxSSC -or- 65°C; 0.3xSSC
42°C; lxSSC, 50% formamide
B DNA.DNA <50 TB »; lxSSC J.XθJ^-
C DNA.RNA ≥ 50 67°C; lxSSC -or- 67°C; 0.3xSSC
45 °C; lxSSC, 50% formamide
D DNA:R A <50 1 / -X-5θ^-- 1 r-) 1X-5-Λ--
E RNA:RNA ≥ 50 70°C, lxSSC -or- 70°C; 0.3xSSC
50°C, lxSSC, 50% formamide
F RNA:RNA <50 TF »; lxSSC TF »; lxSSC
G DNA:DNA ≥ 50 65°C, 4xSSC -or- 65°C; lxSSC
42 °C, 4xSSC, 50% formamide
H DNA.DNA <50 TH*; 4xSSC i. u / ^eX-JkJ^.
I DNA:RNA ≥ 50 67°C, 4xSSC -or- 67°C; lxSSC
45 °C; 4xSSC, 50% formamide
J DNA:RNA <50 T,*; 4xSSC T,*; 4xSSC
K RNA:RNA ≥ 50 70°C; 4xSSC -or- 67°C; lxSSC
50 °C; 4xSSC, 50% formamide
L RNA.RNA <50 TL*; 2xSSC TL »; 2xSSC
M DNA:DNA ≥ 50 50°C; 4xSSC -or- 50°C; 2xSSC
40°C; 6xSSC, 50% formamide
N DNA:DNA <50 V; 6xSSC TN*; 6xSSC
O DNA:RNA ≥ 50 55°C, 4xSSC -or- 55°C, 2xSSC
42 °C; 6xSSC, 50% formamide
P DNA:RNA <50 TP*; 6xSSC TP*; 6xSSC
Q RNA:RNA ≥ 50 60°C; 4xSSC -or- 60°C; 2xSSC
45 °C; 6xSSC, 50% formamide
R RNA:RNA <50 TR*; 4xSSC TR*; 4xSSC
Figure imgf000017_0001
* The hybrid length is that antiαpated for the hybridized regιon(s) of the hybridizing polynucleotides. When hybridizing a polynucleotide to a target polynucleotide of unknown sequence, the hybrid length is assumed to be that of the hybridizing polynucleotide. When polynucleotides of known sequence are hybridized, the hybrid length can be determined by aligning the sequences of the polynucleotides and identifying the region or regions of optimal sequence complementarity.
*: SSPE (lxSSPE is 0.15M NaCl, lOmM NaH2P04, and 1.25mM EDTA, pH 74) can be substituted for SSC (lxSSC is 0.15M NaCl and 15mM sodium citrate) m the hybridization and wash buffers; washes are performed for 15 minutes after hybridization is complete
TB - TR. The hybridization temperature for hybπds antiαpated to be less than 50 base pairs m length should be 5-10°C less than the melting temperature (Tm) of the hybrid, where Tm is determined according to the following equations. For hybnds less than 18 base pairs in length, Tm(°C) = 2(# of A + T bases) + 4(# of G + C bases). For hybπds between 18 and 49 base pairs in length, Tm(°C) = 81.5 + 16.6(log10[Na+]) + 0.41(%G+C) - (600/N), where N is the number of bases in the hybπd, and [Na+] is the concentration of sodium ions m the hybndization buffer ([Na+] for lxSSC = 0.165 M).
15 Additional examples of stringency conditions for polynucleotide hybridization are provided in Sambrook, J., E.F. Fritsch, and T. Maniatis, 1989, Molecular Cloning: A
Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY, chapters 9 and 11, and Current Protocols in Molecular Biology, 1995, F.M. Ausubel et al, eds., John Wiley & Sons, Inc., sections 2.10 and 6.3-6.4, incorporated herein by reference.
Preferably, each such hybridizing polynucleotide has a length that is at least 25%(more preferably at least 50%, and most preferably at least 75%) of the length of the polynucleotide of the present invention to which it hybridizes, and has at least 60% sequence identity (more preferably, at least 75% identity; most preferably at least 90% or 95% identity) with the polynucleotide of the present invention to which it hybridizes, where sequence identity is determined by comparing the sequences of the hybridizing polynucleotides when aligned so as to maximize overlap and identity while minimizing sequence gaps.
The isolated polynucleotide of the invention may be operably linked to an expression control sequence such as the pMT2 or pED expression vectors disclosed in Kaufman et al, Nucleic Acids Res. 19, 4485-4490 (1991), in order to produce the protein recombinantly. Many suitable expression control sequences are known in the art. General methods of expressing recombinant proteins are also known and are exemplified in R. Kaufman, Methods in Enzymology 185. 537-566 (1990). As defined herein "operably linked" means that the isolated polynucleotide of the invention and an expression control sequence are situated within a vector or cell in such a way that the protein is expressed by a host cell which has been transformed (transfected) with the ligated polynucleotide /expression control sequence.
A number of types of cells may act as suitable host cells for expression of the protein. Mammalian host cells include, for example, monkey COS cells, Chinese Hamster
Ovary (CHO) cells, human kidney 293 cells, human epidermal A431 cells, human Colo205 cells, 3T3 cells, CV-1 cells, other transformed primate cell lines, normal diploid cells, cell strains derived from in vitro culture of primary tissue, primary explants, HeLa cells, mouse L cells, BHK, HL-60, U937, HaK or Jurkat cells. Alternatively, it may be possible to produce the protein in lower eukaryotes such as yeast or in prokaryotes such as bacteria. Potentially suitable yeast strains include Saccharomyces cerevisiae, Schizosaccharomyces pombe, Kluyveromyces strains, Candida, or any yeast strain capable of expressing heterologous proteins. Potentially suitable bacterial strains include Escherichia coli, Bacillus subtilis, Salmonella typhimurium, or any bacterial
16 strain capable of expressing heterologous proteins. If the protein is made in yeast or bacteria, it may be necessary to modify the protein produced therein, for example by phosphorylation or glycosylation of the appropriate sites, in order to obtain the functional protein. Such covalent attachments may be accomplished using known chemical or enzymatic methods.
The protein may also be produced by operably linking the isolated polynucleotide of the invention to suitable control sequences in one or more insect expression vectors, and employing an insect expression system. Materials and methods for baculovirus/insect cell expression systems are commercially available in kit form from, e.g., Invitrogen, San Diego, California, U.S.A. (the MaxBac® kit), and such methods are well known in the art, as described in Summers and Smith, Texas Agricultural Experiment Station Bulletin No. 1555 (1987), incorporated herein by reference. As used herein, an insect cell capable of expressing a polynucleotide of the present invention is "transformed." The protein of the invention may be prepared by culturing transformed host cells under culture conditions suitable to express the recombinant protein. The resulting expressed protein may then be purified from such culture (i.e., from culture medium or cell extracts) using known purification processes, such as gel filtration and ion exchange chromatography. The purification of the protein may also include an affinity column containing agents which will bind to the protein; one or more column steps over such affinity resins as concanavalin A-agarose, heparin-toyopearl® or Cibacrom blue 3GA Sepharose®; one or more steps involving hydrophobic interaction chromatography using such resins as phenyl ether, butyl ether, or propyl ether; or iirununoaffinity chromatography. Alternatively, the protein of the invention may also be expressed in a form which will facilitate purification. For example, it may be expressed as a fusion protein, such as those of maltose binding protein (MBP), glutathione-S-transferase (GST) or thioredoxin (TRX). Kits for expression and purification of such fusion proteins are commercially available from New England BioLabs (Beverly, MA), Pharmacia (Piscataway, NJ) and Invitrogen Corporation (Carlsbad, CA), respectively. The protein can also be tagged with an epitope and subsequently purified by using a specific antibody directed to such epitope. One such epitope ("Flag") is commercially available from the Eastman Kodak Company (New Haven, CT).
17 Finally, one or more reverse-phase high performance liquid chromatography (RP- HPLC) steps employing hydrophobic RP-HPLC media, e.g., silica gel having pendant methyl or other aliphatic groups, can be employed to further purify the protein. Some or all of the foregoing purification steps, in various combinations, can also be employed to provide a substantially homogeneous isolated recombinant protein. The protein thus purified is substantially free of other mammalian proteins and is defined in accordance with the present invention as an "isolated protein."
The protein of the invention may also be expressed as a product of transgenic animals, e.g., as a component of the milk of transgenic cows, goats, pigs, or sheep which are characterized by somatic or germ cells containing a nucleotide sequence encoding the protein.
The protein may also be produced by known conventional chemical synthesis. Methods for constructing the proteins of the present invention by synthetic means are known to those skilled in the art. The synthetically-constructed protein sequences, by virtue of sharing primary, secondary or tertiary structural and /or conformational characteristics with proteins may possess biological properties in common therewith, including protein activity. Thus, they may be employed as biologically active or immunological substitutes for natural, purified proteins in screening of therapeutic compounds and in immunological processes for the development of antibodies. The proteins provided herein also include proteins characterized by amino acid sequences similar to those of purified proteins but into which modification are naturally provided or deliberately engineered. For example, modifications in the peptide or DNA sequences can be made by those skilled in the art using known techniques. Modifications of interest in the protein sequences may include the alteration, substitution, replacement, insertion or deletion of a selected amino acid residue in the coding sequence. For example, one or more of the cysteine residues may be deleted or replaced with another amino acid to alter the conformation of the molecule. Techniques for such alteration, substitution, replacement, insertion or deletion are well known to those skilled in the art (see, e.g., U.S. Patent No.4,518,584). Preferably, such alteration, substitution, replacement, insertion or deletion retains the desired activity of the protein.
Other fragments and derivatives of the sequences of proteins which would be expected to retain protein activity in whole or in part and may thus be useful for screening or other immunological methodologies may also be easily made by those skilled in the art
18 given the disclosures herein. Such modifications are believed to be encompassed by the present invention.
USES AND BIOLOGICAL ACTIVITY The polynucleotides and proteins of the present invention are expected to exhibit one or more of the uses or biological activities (including those associated with assays cited herein) identified below. Uses or activities described for proteins of the present invention may be provided by administration or use of such proteins or by adrriinistration or use of polynucleotides encoding such proteins (such as, for example, in gene therapies or vectors suitable for introduction of DNA).
Research Uses and Utilities
The polynucleotides provided by the present invention can be used by the research community for various purposes. The polynucleotides can be used to express recombinant protein for analysis, characterization or therapeutic use; as markers for tissues in which the corresponding protein is preferentially expressed (either constitutively or at a particular stage of tissue differentiation or development or in disease states); as molecular weight markers on Southern gels; as chromosome markers or tags (when labeled) to identify chromosomes or to map related gene positions; to compare with endogenous DNA sequences in patients to identify potential genetic disorders; as probes to hybridize and thus discover novel, related DNA sequences; as a source of information to derive PCR primers for genetic fingerprinting; as a probe to "subtract-out" known sequences in the process of discovering other novel polynucleotides; for selecting and making oligomers for attachment to a "gene chip" or other support, including for examination of expression patterns; to raise anti-protein antibodies using DNA immunization techniques; and as an antigen to raise anti-DNA antibodies or elicit another immune response. Where the polynucleotide encodes a protein which binds or potentially binds to another protein (such as, for example, in a receptor-ligand interaction), the polynucleotide can also be used in interaction trap assays (such as, for example, those described in Gyuris et al, 1993, Cell 75: 791-803 and in Rossi et al, 1997, Proc. Natl. Acad.
Sci. USA 94: 8405-8410, all of which are incorporated by reference herein) to identify polynucleotides encoding the other protein with which binding occurs or to identify inhibitors of the binding interaction.
19 The proteins provided by the present invention can similarly be used in assay to determine biological activity, including in a panel of multiple proteins for high- throughput screening; to raise antibodies or to elicit another immune response; as a reagent (including the labeled reagent) in assays designed to quantitatively determine levels of the protein (or its receptor) in biological fluids; as markers for tissues in which the corresponding protein is preferentially expressed (either constitutively or at a particular stage of tissue differentiation or development or in a disease state); and, of course, to isolate correlative receptors or ligands. Where the protein binds or potentially binds to another protein (such as, for example, in a receptor-ligand interaction), the protein can be used to identify the other protein with which binding occurs or to identify inhibitors of the binding interaction. Proteins involved in these binding interactions can also be used to screen for peptide or small molecule inhibitors or agonists of the binding interaction.
Any or all of these research utilities are capable of being developed into reagent grade or kit format for commercialization as research products.
Methods for performing the uses listed above are well known to those skilled in the art. References disclosing such methods include without limitation "Molecular Cloning: A Laboratory Manual", 2d ed., Cold Spring Harbor Laboratory Press, Sambrook, J., E.F. Fritsch and T. Maniatis eds., 1989, and "Methods in Enzymology: Guide to Molecular Cloning Techniques", Academic Press, Berger, S.L. and A.R. Kimmel eds., 1987.
Nutritional Uses
Polynucleotides and proteins of the present invention can also be used as nutritional sources or supplements. Such uses include without limitation use as a protein or amino add supplement, use as a carbon source, use as a nitrogen source and use as a source of carbohydrate. In such cases the protein or polynucleotide of the invention can be added to the feed of a particular organism or can be administered as a separate solid or liquid preparation, such as in the form of powder, pills, solutions, suspensions or capsules. In the case of microorganisms, the protein or polynucleotide of the invention can be added to the medium in or on which the microorganism is cultured.
Cytokine and Cell Proliferation /Differentiation Activity
A protein of the present invention may exhibit cytokine, cell proliferation (either inducing or inhibiting) or cell differentiation (either inducing or inhibiting) activity or may
20 induce production of other cytokines in certain cell populations. Many protein factors discovered to date, induding all known cytokines, have exhibited activity in one or more factor-dependent cell proliferation assays, and hence the assays serve as a convenient confirmation of cytokine activity. The activity of a protein of the present invention is evidenced by any one of a number of routine factor dependent cell proliferation assays for cell lines including, without limitation, 32D, DA2, DA1G, T10, B9, B9/11, BaF3, MC9/G, M+ (preB M+), 2E8, RB5, DAI, 123, T1165, HT2, CTLL2, TF-1, Mo7e and CMK. The activity of a protein of the invention may, among other means, be measured by the following methods: Assays for T-cell or thymocyte proliferation include without limitation those described in: Current Protocols in Immunology, Ed by J. E. Coligan, A.M. Kruisbeek, D.H. Margulies, E.M. Shevach, W Strober, Pub. Greene Publishing Associates and Wiley- Intersdence (Chapter 3, In Vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); Takai et al., J. Immunol. 137:3494-3500, 1986; BertagnoUi et al., J. Immunol. 145:1706-1712, 1990; Bertagnolli et al., Cellular Immunology 133:327-341, 1991; BertagnoUi, et al., J. Immunol. 149:3778-3783, 1992; Bowman et al., J. Immunol. 152: 1756-1761, 1994.
Assays for cytokine production and /or proliferation of spleen cells, lymph node cells or thymocytes include, without limitation, those described in: Polyclonal T cell stimulation, Kruisbeek, A.M. and Shevach, E.M. In Current Protocols in Immunology. J.E.e.a.
Coligan eds. Vol 1 pp. 3.12.1-3.12.14, John Wiley and Sons, Toronto. 1994; and
Measurement of mouse and human Interferon γ, Schreiber, R.D. In Current Protocols in
Immunology. J.E.e.a. CoUgan eds. Vol 1 pp. 6.8.1-6.8.8, John WUey and Sons, Toronto. 1994.
Assays for proliferation and differentiation of hematopoietic and lymphopoietic ceUs include, without limitation, those described in: Measurement of Human and Murine
Interleukin 2 and Interleukin 4, Bottomry, K., Davis, L.S. and Lipsky, P.E. In Current Protocols in Immunology. J.E.e.a. Coligan eds. Vol 1 pp. 6.3.1-6.3.12, John Wiley and Sons, Toronto. 1991; deVries et al., J. Exp. Med. 173:1205-1211, 1991; Moreau et al., Nature 336:690-692, 1988; Greenberger et al., Proc. Natl. Acad. Sci. U.S.A. 80:2931-2938, 1983; Measurement of mouse and human interleukin 6 - Nordan, R. In Current Protocols in
Immunology. J.E.e.a. CoUgan eds. Vol 1 pp. 6.6.1-6.6.5, John Wiley and Sons, Toronto. 1991; Smith et al., Proc. Natl. Acad. Sci. U.S.A. 83:1857-1861, 1986; Measurement of human Interleukin 11 - Bennett, F., Giannotti, J., Clark, S.C. and Turner, K. J. In Current Protocols in Immunology. J.E.e.a. CoUgan eds. Vol 1 pp. 6.15.1 John Wiley and Sons, Toronto. 1991;
21 Measurement of mouse and human Interleukin 9 - Ciarletta, A., Giannotti, J., Clark, S.C. and Turner, K.J. In Current Protocols in Immunology. J.E.e.a. Coligan eds. Vol 1 pp. 6.13.1, John Wiley and Sons, Toronto. 1991.
Assays for T-cell clone responses to antigens (which wiU identify, among others, proteins that affect APC-T ceU interactions as weU as direct T-ceU effects by measuring proliferation and cytokine production) include, without Umitation, those described in: Current Protocols in Immunology, Ed by J. E. Coligan, A.M. Kruisbeek, D.H. Margulies, E.M. Shevach, W Strober, Pub. Greene PubUshing Associates and Wiley-Interscience (Chapter 3, In Vitro assays for Mouse Lymphocyte Function; Chapter 6, Cytokines and their cellular receptors; Chapter 7, Immunologic studies in Humans); Weinberger et al., Proc. Natl. Acad. Sci. USA 77:6091-6095, 1980; Weinberger et al., Eur. J. Irnmun. 11:405-411, 1981; Takai et al., J. Immunol. 137:3494-3500, 1986; Takai et al., J. Immunol. 140:508-512, 1988.
Immune Stimulating or Suppressing Activity
A protein of the present invention may also exhibit immune stimulating or immune suppressing activity, induding without Umitation the activities for which assays are described herein. A protein may be useful in the treatment of various immune deficiencies and disorders (including severe combined immunodeficiency (SCID)), e.g., in regulating (up or down) growth and proUferation of T and /or B lymphocytes, as well as effecting the cytolytic activity of NK ceUs and other cell populations. These immune deficiendes may be genetic or be caused by viral (e.g., HIV) as well as bacterial or fungal infections, or may result from autoimmune disorders. More specificaUy, infectious diseases causes by viral, bacterial, fungal or other infection may be treatable using a protein of the present invention, including infections by HIV, hepatitis viruses, herpesviruses, mycobacteria, Leishmania spp., malaria spp. and various fungal infections such as candidiasis. Of course, in this regard, a protein of the present invention may also be useful where a boost to the immune system generally may be desirable, i.e., in the treatment of cancer. Autoimmune disorders which may be treated using a protein of the present invention include, for example, connective tissue disease, multiple sclerosis, systemic lupus erythematosus, rheumatoid arthritis, autoimmune pulmonary inflammation, GuUlain-Barre syndrome, autoimmune thyroiditis, insulin dependent diabetes meUitis, myasthenia gravis, graft-versus-host disease and autoimmune inflammatory eye disease.
22 Such a protein of the present invention may also to be useful in the treatment of aUergic reactions and conditions, such as asthma (particularly aUergic asthma) or other respiratory problems. Other conditions, in which immune suppression is desired (including, for example, organ transplantation), may also be treatable using a protein of the present invention.
Using the proteins of the invention it may also be possible to regulate immune responses in a number of ways. Down regulation may be in the form of inhibiting or blocking an immune response already in progress or may involve preventing the induction of an immune response. The functions of activated T cells may be inhibited by suppressing T cell responses or by inducing specific tolerance in T cells, or both. Immunosuppression of T ceU responses is generaUy an active, non-antigen-specific, process which requires continuous exposure of the T ceUs to the suppressive agent. Tolerance, which involves inducing non-responsiveness or anergy in T ceUs, is distinguishable from immunosuppression in that it is generally antigen-specific and persists after exposure to the tolerizing agent has ceased. OperationaUy, tolerance can be demonstrated by the lack of a T cell response upon reexposure to specific antigen in the absence of the tolerizing agent.
Down regulating or preventing one or more antigen functions (including without Umitation B lymphocyte antigen functions (such as , for example, B7)), e.g., preventing high level lymphokine synthesis by activated T ceUs, wiU be useful in situations of tissue, skin and organ transplantation and in graft-versus-host disease (GVHD). For example, blockage of T cell function should result in reduced tissue destruction in tissue transplantation. TypicaUy, in tissue transplants, rejection of the transplant is initiated through its recognition as foreign by T cells, f oUowed by an immune reaction that destroys the transplant. The administration of a molecule which inhibits or blocks interaction of a B7 lymphocyte antigen with its natural Ugand(s) on immune cells (such as a soluble, monomeric form of a peptide having B7-2 activity alone or in conjunction with a monomeric form of a peptide having an activity of another B lymphocyte antigen (e.g., B7- 1, B7-3) or blocking antibody), prior to transplantation can lead to the binding of the molecule to the natural ligand(s) on the immune ceUs without transmitting the corresponding costimulatory signal. Blocking B lymphocyte antigen function in this matter prevents cytokine synthesis by immune cells, such as T cells, and thus acts as an immunosuppressant. Moreover, the lack of costimulation may also be sufficient to anergize the T ceUs, thereby inducing tolerance in a subject. Induction of long-term
23 tolerance by B lymphocyte antigen-blocking reagents may avoid the necessity of repeated adrninistration of these blocking reagents. To achieve sufficient immunosuppression or tolerance in a subject, it may also be necessary to block the function of a combination of B lymphocyte antigens. The efficacy of particular blocking reagents in preventing organ transplant rejection or GVHD can be assessed using animal models that are predictive of efficacy in humans. Examples of appropriate systems which can be used include allogeneic cardiac grafts in rats and xenogeneic pancreatic islet cell grafts in mice, both of which have been used to examine the immunosuppressive effects of CTLA4Ig fusion proteins in vivo as described in Lenschow et al, Sdence 257:789-792 (1992) and Turka et al, Proc. Natl. Acad. Sci USA, 89:11102-11105 (1992). In addition, murine models of GVHD (see Paul ed., Fundamental Immunology, Raven Press, New York, 1989, pp. 846-847) can be used to determine the effect of blocking B lymphocyte antigen function in vivo on the development of that disease. Blocking antigen function may also be therapeuticaUy useful for treating autoimmune diseases. Many autoimmune disorders are the result of inappropriate activation of T cells that are reactive against self tissue and which promote the production of cytokines and autoantibodies involved in the pathology of the diseases. Preventing the activation of autoreactive T cells may reduce or eliminate disease symptoms. Administration of reagents which block costimulation of T cells by disrupting receptor:ligand interactions of B lymphocyte antigens can be used to inhibit T cell activation and prevent production of autoantibodies or T cell-derived cytokines which may be involved in the disease process. Additionally, blocking reagents may induce antigen-specific tolerance of autoreactive T ceUs which could lead to long-term reUef from the disease. The efficacy of blocking reagents in preventing or aUeviating autoimmune disorders can be determined using a number of well-characterized animal models of human autoimmune diseases. Examples include murine experimental autoimmune encephalitis, systemic lupus erythmatosis in MRL/lprfipr mice or NZB hybrid mice, murine autoimmune coUagen arthritis, diabetes meUitus in NOD mice and BB rats, and murine experimental myasthenia gravis (see Paul ed., Fundamental Immunology, Raven
Press, New York, 1989, pp. 840-856).
Upregulation of an antigen function (preferably a B lymphocyte antigen function), as a means of up regulating immune responses, may also be useful in therapy. Upregulation of immune responses may be in the form of enhancing an existing immune
24 response or eUdting an initial immune response. For example, enhancing an immune response through stimulating B lymphocyte antigen function may be useful in cases of viral infection. In addition, systemic viral diseases such as influenza, the common cold, and encephaUtis might be aUeviated by the administration of stimulatory forms of B lymphocyte antigens systemically.
Alternatively, anti-viral immune responses may be enhanced in an infected patient by removing T cells from the patient, costimulating the T cells in vitro with viral antigen- pulsed APCs either expressing a peptide of the present invention or together with a stimulatory form of a soluble peptide of the present invention and reintroducing the in vitro activated T cells into the patient. Another method of enhancing anti-viral immune responses would be to isolate infected ceUs from a patient, transfect them with a nucleic acid encoding a protein of the present invention as described herein such that the ceUs express aU or a portion of the protein on their surface, and reintroduce the transfected ceUs into the patient. The infected ceUs would now be capable of delivering a costimulatory signal to, and thereby activate, T cells in vivo.
In another application, up regulation or enhancement of antigen function (preferably B lymphocyte antigen function) may be useful in the induction of tumor immunity. Tumor ceUs (e.g., sarcoma, melanoma, lymphoma, leukemia, neuroblastoma, carcinoma) transfected with a nucleic add encoding at least one peptide of the present invention can be administered to a subject to overcome tumor-specific tolerance in the subject. If desired, the tumor ceU can be transfected to express a combination of peptides. For example, tumor cells obtained from a patient can be transfected ex vivo with an expression vector directing the expression of a peptide having B7-2-like activity alone, or in conjunction with a peptide having B7-1-Uke activity and/or B7-3-like activity. The transfected tumor cells are returned to the patient to result in expression of the peptides on the surface of the transfected ceU. Alternatively, gene therapy techniques can be used to target a tumor cell for transfection in vivo.
The presence of the peptide of the present invention having the activity of a B lymphocyte antigen(s) on the surface of the tumor ceU provides the necessary costimulation signal to T cells to induce a T cell mediated immune response against the transfected tumor cells. In addition, tumor ceUs which lack MHC class I or MHC class II molecules, or which fail to reexpress suffident amounts of MHC class I or MHC class II molecules, can be transfected with nucleic acid encoding all or a portion of (e.g., a cytoplasmic-domain truncated portion) of an MHC class I α chain protein and β2
25 microglobulin protein or an MHC class II a chain protein and an MHC class II β chain protein to thereby express MHC dass I or MHC class II proteins on the ceU surface. Expression of the appropriate dass I or class Et MHC in conjunction with a peptide having the activity of a B lymphocyte antigen (e.g., B7-1, B7-2, B7-3) induces a T cell mediated immune response against the transfected tumor ceU. OptionaUy, a gene encoding an antisense construct which blocks expression of an MHC class II associated protein, such as the invariant chain, can also be cotransfected with a DNA encoding a peptide having the activity of a B lymphocyte antigen to promote presentation of tumor associated antigens and induce tumor specific immunity. Thus, the induction of a T ceU mediated immune response in a human subject may be sufficient to overcome tumor-specific tolerance in the subject.
The activity of a protein of the invention may, among other means, be measured by the following methods:
Suitable assays for thymocyte or splenocyte cytotoxicity include, without limitation, those described in: Current Protocols in Immunology, Ed by J. E. CoUgan, A.M. Kruisbeek, D.H. MarguUes, E.M. Shevach, W Strober, Pub. Greene PubUshing Associates and Wney-Interscience (Chapter 3, In Vitro assays for Mouse Lymphocyte Function 3.1- 3.19; Chapter 7, Immunologic studies in Humans); Herrmann et al., Proc. Natl. Acad. Sci. USA 78:2488-2492, 1981; Herrmann et al., J. Immunol. 128:1968-1974, 1982; Handa et al., J. Immunol. 135:1564-1572, 1985; Takai et al, J. Immunol. 137:3494-3500, 1986; Takai et al., J. Immunol. 140:508-512, 1988; Herrmann et al., Proc. Natl. Acad. Sci. USA 78:2488-2492, 1981; Herrmann et al., J. Immunol. 128:1968-1974, 1982; Handa et al., J. Immunol. 135:1564-1572, 1985; Takai et al., J. Immunol. 137:3494-3500, 1986; Bowmanet al., J. Virology 61:1992-1998; Takai et al, J. Immunol. 140:508-512, 1988; BertagnoUi et al., Cellular Immunology 133:327-341, 1991; Brown et al., J. Immunol. 153:3079-3092, 1994.
Assays for T-ceU-dependent immunoglobulin responses and isotype switching (which will identify, among others, proteins that modulate T-ceU dependent antibody responses and that affect Thl/Th2 profiles) include, without limitation, those described in: MaUszewski, J. Immunol. 144:3028-3033, 1990; and Assays for B ceU function: In vitro antibody production, Mond, J.J. and Brunswick, M. In Current Protocols in Immunology.
J.E.e.a. Coligan eds. Vol 1 pp. 3.8.1-3.8.16, John Wiley and Sons, Toronto. 1994.
Mixed lymphocyte reaction (MLR) assays (which wiU identify, among others, proteins that generate predominantly Thl and CTL responses) indude, without Umitation, those described in: Current Protocols in Immunology, Ed by J. E. CoUgan, A.M. Kruisbeek,
26 D.H. MarguUes, E.M. Shevach, W Strober, Pub. Greene PubUshing Associates and Wiley- Intersdence (Chapter 3, In Vitro assays for Mouse Lymphocyte Function 3.1-3.19; Chapter 7, Immunologic studies in Humans); Takai et al., J. Immunol. 137:3494-3500, 1986; Takai et al., J. Immunol. 140:508-512, 1988; BertagnoUi et al., J. Immunol. 149:3778-3783, 1992. Dendritic cell-dependent assays (which wiU identify, among others, proteins expressed by dendritic cells that activate naive T-cells) include, without limitation, those described in: Guery et al., J. Immunol. 134:536-544, 1995; Inaba et al., Journal of Experimental Medicine 173:549-559, 1991; Macatonia et al., Journal of Immunology 154:5071-5079, 1995; Porgador et al., Journal of Experimental Medicine 182:255-260, 1995; Nair et al., Journal of Virology 67:4062-4069, 1993; Huang et al., Science 264:961-965, 1994; Macatonia et al., Journal of Experimental Medicine 169:1255-1264, 1989; Bhardwaj et al., Journal of Clinical Investigation 94:797-807, 1994; and Inaba et al., Journal of Experimental Medicine 172:631-640, 1990.
Assays for lymphocyte survival /apoptosis (which wiU identify, among others, proteins that prevent apoptosis after superantigen induction and proteins that regulate lymphocyte homeostasis) indude, without Umitation, those described in: Darzynkiewicz et al., Cytometry 13:795-808, 1992; Gorczyca et al., Leukemia 7:659-670, 1993; Gorczyca et al., Cancer Research 53:1945-1951, 1993; Itoh et al., Cell 66:233-243, 1991; Zacharchuk, Journal of Immunology 145:4037-4045, 1990; Zamai et al., Cytometry 14:891-897, 1993; Gorczyca et al., International Journal of Oncology 1:639-648, 1992.
Assays for proteins that influence early steps of T-cell commitment and development include, without Umitation, those described in: Antica et al., Blood 84:111-117, 1994; Fine et al., CeUular Immunology 155:111-122, 1994; Galy et al, Blood 85:2770-2778, 1995; Toki et al., Proc. Nat. Acad Sci. USA 88:7548-7551, 1991.
Hematopoiesis Regulating Activity
A protein of the present invention may be useful in regulation of hematopoiesis and, consequently, in the treatment of myeloid or lymphoid cell deficiendes. Even marginal biological activity in support of colony forming cells or of factor-dependent ceU lines indicates involvement in regulating hematopoiesis, e.g. in supporting the growth and proliferation of erythroid progenitor cells alone or in combination with other cytokines, thereby indicating utility, for example, in treating various anemias or for use in conjunction with irradiation /chemotherapy to stimulate the production of erythroid precursors and/or erythroid cells; in supporting the growth and proliferation of myeloid
27 ceUs such as granulocytes and monocytes/macrophages (i.e., traditional CSF activity) useful, for example, in conjunction with chemotherapy to prevent or treat consequent myelo-suppression; in supporting the growth and proUferation of megakaryocytes and consequently of platelets thereby aUowing prevention or treatment of various platelet disorders such as thrombocytopenia, and generaUy for use in place of or complimentary to platelet transfusions; and /or in supporting the growth and proUferation of hematopoietic stem ceUs which are capable of maturing to any and aU of the above- mentioned hematopoietic cells and therefore find therapeutic utility in various stem ceU disorders (such as those usuaUy treated with transplantation, including, without limitation, aplastic anemia and paroxysmal nocturnal hemoglobinuria), as well as in repopulating the stem ceU compartment post irradiation/chemotherapy, either in-vivo or ex-vivo (i.e., in conjunction with bone marrow transplantation or with peripheral progenitor ceU transplantation (homologous or heterologous)) as normal ceUs or geneticaUy manipulated for gene therapy. The activity of a protein of the invention may, among other means, be measured by the following methods:
Suitable assays for proliferation and differentiation of various hematopoietic lines are cited above.
Assays for embryonic stem ceU differentiation (which wiU identify, among others, proteins that influence embryonic differentiation hematopoiesis) include, without Umitation, those described in: Johansson et al. CeUular Biology 15:141-151, 1995; Keller et al., Molecular and CeUular Biology 13:473-486, 1993; McClanahan et al, Blood 81:2903-2915, 1993.
Assays for stem cell survival and differentiation (which will identify, among others, proteins that regulate lympho-hematopoiesis) include, without Umitation, those described in: MethylceUulose colony forming assays, Freshney, M.G. In Culture of Hematopoietic Cells. R.I. Freshney, et al. eds. Vol pp. 265-268, Wiley-Liss, Inc., New York, NY. 1994; Hirayama et al., Proc. Natl. Acad. Sci. USA 89:5907-5911, 1992; Primitive hematopoietic colony forming ceUs with high proliferative potential, McNiece, I.K. and Briddell, R.A. In Culture of Hematopoietic Cells. R.I. Freshney, et al. eds. Vol pp. 23-39,
WUey-Liss, Inc., New York, NY. 1994; Neben et al., Experimental Hematology 22:353-359, 1994; Cobblestone area forming cell assay, Ploemacher, R.E. In Culture of Hematopoietic Cells. R.I. Freshney, et al eds. Vol pp. 1-21, Wiley-Liss, Inc., New York, NY. 1994; Long term bone marrow cultures in the presence of stromal cells, Spooncer, E., Dexter, M. and
28 Allen, T. In Culture of Hematopoietic Cells. R.I. Freshney, et al. eds. Vol pp. 163-179, Wiley-Liss, Inc., New York, NY. 1994; Long term culture initiating cell assay, Sutherland, H.J. In Culture of Hematopoietic Cells. R.I. Freshney, et al. eds. Vol pp. 139-162, Wiley-Liss, Inc., New York, NY. 1994.
Tissue Growth Activity
A protein of the present invention also may have utility in compositions used for bone, cartilage, tendon, Ugament and /or nerve tissue growth or regeneration, as well as for wound healing and tissue repair and replacement, and in the treatment of burns, incisions and ulcers.
A protein of the present invention, which induces cartilage and/or bone growth in circumstances where bone is not normally formed, has appUcation in the healing of bone fractures and cartilage damage or defects in humans and other animals. Such a preparation employing a protein of the invention may have prophylactic use in closed as well as open fracture reduction and also in the improved fixation of artificial joints. De novo bone formation induced by an osteogenic agent contributes to the repair of congenital, trauma induced, or oncologic resection induced craniofacial defects, and also is useful in cosmetic plastic surgery.
A protein of this invention may also be used in the treatment of periodontal disease, and in other tooth repair processes. Such agents may provide an environment to attract bone-forming ceUs, stimulate growth of bone-forming ceUs or induce differentiation of progenitors of bone-forming ceUs. A protein of the invention may also be useful in the treatment of osteoporosis or osteoarthritis, such as through stimulation of bone and /or cartilage repair or by blocking inflammation or processes of tissue destruction (coUagenase activity, osteoclast activity, etc.) mediated by inflammatory processes.
Another category of tissue regeneration activity that may be attributable to the protein of the present invention is tendon/ligament formation. A protein of the present invention, which induces tendon/ligament-like tissue or other tissue formation in circumstances where such tissue is not normaUy formed, has appUcation in the healing of tendon or Ugament tears, deformities and other tendon or Ugament defects in humans and other animals. Such a preparation employing a tendon/ligament-like tissue inducing protein may have prophylactic use in preventing damage to tendon or Ugament tissue, as well as use in the improved fixation of tendon or Ugament to bone or other tissues, and
29 in repairing defects to tendon or Ugament tissue. De novo tendon/Ugament-like tissue formation induced by a composition of the present invention contributes to the repair of congenital, trauma induced, or other tendon or Ugament defects of other origin, and is also useful in cosmetic plastic surgery for attachment or repair of tendons or ligaments. The compositions of the present invention may provide an environment to attract tendon- or ligament-forming cells, stimulate growth of tendon- or Ugament-forming ceUs, induce differentiation of progenitors of tendon- or Ugament-forming ceUs, or induce growth of tendon/Ugament ceUs or progenitors ex vivo for return in vivo to effect tissue repair. The compositions of the invention may also be useful in the treatment of tendinitis, carpal tunnel syndrome and other tendon or Ugament defects. The compositions may also include an appropriate matrix and /or sequestering agent as a carrier as is well known in the art.
The protein of the present invention may also be useful for proUferation of neural cells and for regeneration of nerve and brain tissue, i.e. for the treatment of central and peripheral nervous system diseases and neuropathies, as well as mechanical and traumatic disorders, which involve degeneration, death or trauma to neural cells or nerve tissue. More specifically, a protein may be used in the treatment of diseases of the peripheral nervous system, such as peripheral nerve injuries, peripheral neuropathy and localized neuropathies, and central nervous system diseases, such as Alzheimer's, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Shy-Drager syndrome. Further conditions which may be treated in accordance with the present invention indude med anical and traumatic disorders, such as spinal cord disorders, head trauma and cerebrovascular diseases such as stroke. Peripheral neuropathies resulting from chemotherapy or other medical therapies may also be treatable using a protein of the invention.
Proteins of the invention may also be useful to promote better or faster closure of non-healing wounds, including without Umitation pressure ulcers, ulcers associated with vascular insufficiency, surgical and traumatic wounds, and the like.
It is expected that a protein of the present invention may also exhibit activity for generation or regeneration of other tissues, such as organs (including, for example, pancreas, Uver, intestine, kidney, skin, endotheUum), musde (smooth, skeletal or cardiac) and vascular (including vascular endotheUum) tissue, or for promoting the growth of cells comprising such tissues. Part of the desired effects may be by inhibition or modulation
30 of fibrotic scarring to allow normal tissue to regenerate. A protein of the invention may also exhibit angiogenic activity.
A protein of the present invention may also be useful for gut protection or regeneration and treatment of lung or Uver fibrosis, reperfusion injury in various tissues, and conditions resulting from systemic cytokine damage.
A protein of the present invention may also be useful for promoting or inhibiting differentiation of tissues described above from precursor tissues or ceUs; or for inhibiting the growth of tissues described above.
The activity of a protein of the invention may, among other means, be measured by the following methods:
Assays for tissue generation activity include, without Umitation, those described in: International Patent PubUcation No. WO95/ 16035 (bone, cartilage, tendon); International Patent PubUcation No. WO95/ 05846 (nerve, neuronal); International Patent Publication No. WO91/07491 (skin, endotheUum ). Assays for wound healing activity include, without Umitation, those described in:
Winter, Epidermal Wound Healing, pps. 71-112 (Maibach, HI and Rovee, DT, eds.), Year Book Medical Publishers, Inc., Chicago, as modified by Eaglstein and Mertz, J. Invest. Dermatol 71:382-84 (1978).
Activin /Inhibin Activity
A protein of the present invention may also exhibit activin- or inhibin-related activities, Inhibins are characterized by their ability to inhibit the release of folUcle stimulating hormone (FSH), while activins and are characterized by their ability to stimulate the release of foUide stimulating hormone (FSH). Thus, a protein of the present invention, alone or in heterodimers with a member of the inhibin α family, may be useful as a contraceptive based on the abiUty of rnhibins to decrease fertiUty in female mammals and decrease spermatogenesis in male mammals. Administration of sufficient amounts of other rnhibins can induce infertiUty in these mammals. Alternatively, the protein of the invention, as a homodimer or as a heterodimer with other protein subunits of the inhibin- β group, may be useful as a fertiUty inducing therapeutic, based upon the abiUty of activin molecules in stimulating FSH release from cells of the anterior pituitary. See, for example, United States Patent 4,798,885. A protein of the invention may also be useful for advancement of the onset of fertiUty in sexuaUy immature mammals, so as to increase the lifetime reproductive performance of domestic animals such as cows, sheep and pigs.
31 The activity of a protein of the invention may, among other means, be measured by the following methods:
Assays for activm/inhibin activity indude, without Umitation, those described in: Vale et al., Endocrinology 91:562-572, 1972; Ling et al., Nature 321:779-782, 1986; Vale et al., Nature 321:776-779, 1986; Mason et al., Nature 318:659-663, 1985; Forage et al., Proc. Natl. Acad. Sci. USA 83:3091-3095, 1986.
Chemotactic/Chemokinetic Activity
A protein of the present invention may have chemotactic or chemokinetic activity (e.g., act as a chemokine) for mammalian ceUs, including, for example, monocytes, fibroblasts, neutrophils, T-ceUs, mast cells, eosinophils, epitheUal and/or endotheUal cells. Chemotactic and chemokinetic proteins can be used to mobilize or attract a desired ceU population to a desired site of action. Chemotactic or chemokinetic proteins provide particular advantages in treatment of wounds and other trauma to tissues, as well as in treatment of localized infections. For example, attraction of lymphocytes, monocytes or neutrophils to tumors or sites of infection may result in improved immune responses against the tumor or infecting agent.
A protein or peptide has chemotactic activity for a particular cell population if it can stimulate, directly or indirectly, the directed orientation or movement of such ceU population. Preferably, the protein or peptide has the abiUty to directly stimulate directed movement of ceUs. Whether a particular protein has chemotactic activity for a population of cells can be readily determined by employing such protein or peptide in any known assay for cell chemotaxis.
The activity of a protein of the invention may, among other means, be measured by the following methods:
Assays for chemotactic activity (which wiU identify proteins that induce or prevent chemotaxis) consist of assays that measure the abiUty of a protein to induce the migration of ceUs across a membrane as weU as the abiUty of a protein to induce the adhesion of one ceU population to another cell population. Suitable assays for movement and adhesion indude, without Umitation, those described in: Current Protocols in Immunology, Ed by
J.E. Coligan, A.M. Kruisbeek, D.H. MarguUes, E.M. Shevach, W.Strober, Pub. Greene Publishing Assodates and Wiley-Interscience (Chapter 6.12, Measurement of alpha and beta Chemokines 6.12.1-6.12.28; Taub et al. J. Clin. Invest. 95:1370-1376, 1995; Lind et al.
32 APMIS 103:140-146, 1995; MuUer et al Eur. J. Immunol. 25: 1744-1748; Gruber et al. J. of Immunol. 152:5860-5867, 1994; Johnston et al. J. of Immunol. 153: 1762-1768, 1994.
Hemostatic and Thrombolytic Activity A protein of the invention may also exhibit hemostatic or thrombolytic activity.
As a result, such a protein is expected to be useful in treatment of various coagulation disorders (including hereditary disorders, such as hemophilias) or to enhance coagulation and other hemostatic events in treating wounds resulting from trauma, surgery or other causes. A protein of the invention may also be useful for dissolving or inhibiting formation of thromboses and for treatment and prevention of conditions resulting therefrom (such as, for example, infarction of cardiac and central nervous system vessels (e.g., stroke).
The activity of a protein of the invention may, among other means, be measured by the following methods: Assay for hemostatic and thrombolytic activity include, without limitation, those described in: Linet et al., J. Clin. Pharmacol. 26:131-140, 1986; Burdick et al, Thrombosis Res.45:413-419, 1987; Humphrey et al., Fibrinolysis 5:71-79 (1991); Schaub, Prostaglandins 35:467-474, 1988.
Receptor /Ligand Activity
A protein of the present invention may also demonstrate activity as receptors, receptor Ugands or inhibitors or agonists of receptor/Ugand interactions. Examples of such receptors and Ugands include, without Umitation, cytokine receptors and their ligands, receptor kinases and their Ugands, receptor phosphatases and their Ugands, receptors involved in ceU-ceU interactions and their Ugands (including without limitation, ceUular adhesion molecules (such as selectins, integrins and their Ugands) and receptor/ligand pairs involved in antigen presentation, antigen recognition and development of ceUular and humoral immune responses). Receptors and Ugands are also useful for screening of potential peptide or small molecule inhibitors of the relevant receptor/Ugand interaction. A protein of the present invention (including, without Umitation, fragments of receptors and Ugands) may themselves be useful as inhibitors of receptor/ligand interactions.
The activity of a protein of the invention may, among other means, be measured by the following methods:
33 Suitable assays for receptor-Ugand activity include without Umitation those described in:Current Protocols in Immunology, Ed by J.E. CoUgan, A.M. Kruisbeek, D.H. Margulies, E.M. Shevach, W.Strober, Pub. Greene PubUshing Associates and Wiley-Interscience (Chapter 7.28, Measurement of CeUular Adhesion under static conditions 7.28.1-7.28.22), Takai et al., Proc. Natl. Acad. Sci. USA 84:6864-6868, 1987; Bierer et al., J. Exp. Med. 168:1145-1156, 1988; Rosenstein et al., J. Exp. Med. 169:149-160 1989; Stoltenborg et al., J. Immunol. Methods 175:59-68, 1994; Stitt et al., CeU 80:661-670, 1995.
Anti-Inflammatory Activity
Proteins of the present invention may also exhibit anti-inflammatory activity. The anti-inflarnmatory activity may be achieved by providing a stimulus to ceUs involved in the inflammatory response, by inhibiting or promoting ce -ceU interactions (such as, for example, ceU adhesion), by inhibiting or promoting chemotaxis of ceUs involved in the inflammatory process, inhibiting or promoting cell extravasation, or by stimulating or suppressing production of other factors which more directly inhibit or promote an inflammatory response. Proteins exhibiting such activities can be used to treat inflammatory conditions including chronic or acute conditions), including without limitation inflammation assodated with infection (such as septic shock, sepsis or systemic inflammatory response syndrome (SIRS)), ischemia-reperfusion injury, endotoxin lethaUty, arthritis, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine-induced lung injury, inflammatory bowel disease, Crohn's disease or resulting from over production of cytokines such as TNF or IL-1. Proteins of the invention may also be useful to treat anaphylaxis and hypersensitivity to an antigenic substance or material.
Cadherin /Tumor Invasion Suppressor Activity
Cadherins are calcium-dependent adhesion molecules that appear to play major roles during development, particularly in defining specific cell types. Loss or alteration of normal cadherin expression can lead to changes in cell adhesion properties linked to tumor growth and metastasis. Cadherin malfunction is also implicated in other human diseases, such as pemphigus vulgaris and pemphigus foliaceus (auto-immune blistering skin diseases), Crohn's disease, and some developmental abnormaUties.
The cadherin superfarmly includes well over forty members, each with a distinct pattern of expression. All members of the superfamily have in common conserved
34 extraceUular repeats (cadherin domains), but structural differences are found in other parts of the molecule. The cadherin domains bind caldum to form their tertiary structure and thus calcium is required to mediate their adhesion. Only a few amino acids in the first cadherin domain provide the basis for homophiUc adhesion; modification of this recognition site can change the specificity of a cadherin so that instead of recognizing only itself, the mutant molecule can now also bind to a different cadherin. In addition, some cadherins engage in heterophiUc adhesion with other cadherins.
E-cadherin, one member of the cadherin superfamily, is expressed in epitheUal ceU types. PathologicaUy, if E-cadherin expression is lost in a tumor, the maUgnant ceUs become invasive and the cancer metastasizes. Transfection of cancer ceU lines with polynucleotides expressing E-cadherin has reversed cancer-associated changes by returning altered ceU shapes to normal, restoring cells' adhesiveness to each other and to their substrate, decreasing the cell growth rate, and drastically reducing anchorage- independent ceU growth. Thus, reintiOducing E-cadherin expression reverts carcinomas to a less advanced stage. It is likely that other cadherins have the same invasion suppressor role in carcinomas derived from other tissue types. Therefore, proteins of the present invention with cadherin activity, and polynucleotides of the present invention encoding such proteins, can be used to treat cancer. Introducing such proteins or polynucleotides into cancer cells can reduce or eliminate the cancerous changes observed in these cells by providing normal cadherin expression.
Cancer ceUs have also been shown to express cadherins of a different tissue type than their origin, thus aUowing these cells to invade and metastasize in a different tissue in the body. Proteins of the present invention with cadherin activity, and polynucleotides of the present invention encoding such proteins, can be substituted in these cells for the inappropriately expressed cadherins, restoring normal cell adhesive properties and reducing or eUminating the tendency of the cells to metastasize.
AdditionaUy, proteins of the present invention with cadherin activity, and polynucleotides of the present invention encoding such proteins, can used to generate antibodies recognizing and binding to cadherins. Such antibodies can be used to block the adhesion of inappropriately expressed tumor-ceU cadherins, preventing the cells from forming a tumor elsewhere. Such an anti-cadherin antibody can also be used as a marker for the grade, pathological type, and prognosis of a cancer, i.e. the more progressed the cancer, the less cadherin expression there wiU be, and this decrease in cadherin expression can be detected by the use of a cadherin-binding antibody.
35 Fragments of proteins of the present invention with cadherin activity, preferably a polypeptide comprising a decapeptide of the cadherin recognition site, and polynucleotides of the present invention encoding such protein fragments, can also be used to block cadherin function by binding to cadherins and preventing them from binding in ways that produce undesirable effects. AdditionaUy, fragments of proteins of the present invention with cadherin activity, preferably truncated soluble cadherin fragments which have been found to be stable in the circulation of cancer patients, and polynucleotides encoding such protein fragments, can be used to disturb proper ceU-ceU adhesion.
Assays for cadherin adhesive and invasive suppressor activity include, without Umitation, those described in: Hortsch et al. J Biol Chem 270 (32): 18809-18817, 1995; Miyaki et al. Oncogene 11: 2547-2552, 1995; Ozawa et al. Cell 63: 1033-1038, 1990.
Tumor Inhibition Activity
In addition to the activities described above for immunological treatment or prevention of tumors, a protein of the invention may exhibit other anti-tumor activities. A protein may inhibit tumor growth directly or indirectly (such as, for example, via antibody-dependent ceU-mediated cytotoxidty (ADCC)). A protein may exhibit its tumor inhibitory activity by acting on tumor tissue or tumor precursor tissue, by inhibiting formation of tissues necessary to support tumor growth (such as, for example, by inhibiting angiogenesis), by causing production of other factors, agents or ceU types which inhibit tumor growth, or by suppressing, eliminating or inhibiting factors, agents or cell types which promote tumor growth.
Other Activities A protein of the invention may also exhibit one or more of the f oUowing additional activities or effects: inhibiting the growth, infection or function of, or killing, infectious agents, including, without Umitation, bacteria, viruses, fungi and other parasites; effecting (suppressing or enhancing) bodily characteristics, including, without limitation, height, weight, hair color, eye color, skin, fat to lean ratio or other tissue pigmentation, or organ or body part size or shape (such as, for example, breast augmentation or diminution, change in bone form or shape); effecting biorhythms or caricadic cycles or rhythms; effecting the fertility of male or female subjects; effecting the metabolism, catabolism, anabolism, processing, utilization, storage or elimination of dietary fat, lipid, protein, carbohydrate, vitamins, minerals, cofactors or other nutritional factors or component(s);
36 effecting behavioral characteristics, including, without limitation, appetite, libido, stress, cognition (induding cognitive disorders), depression (induding depressive disorders) and violent behaviors; providing analgesic effects or other pain reducing effects; promoting differentiation and growth of embryonic stem ceUs in lineages other than hematopoietic lineages; hormonal or endocrine activity; in the case of enzymes, correcting deficiencies of the enzyme and treating deficiency-related diseases; treatment of hyperproliferative disorders (such as, for example, psoriasis); immunoglobuUn-Uke activity (such as, for example, the abiUty to bind antigens or complement); and the ability to act as an antigen in a vaccine composition to raise an immune response against such protein or another material or entity which is cross-reactive with such protein.
ADMINISTRATION AND DOSING
A protein of the present invention (from whatever source derived, induding without limitation from recombinant and non-recombinant sources) may be used in a pharmaceutical composition when combined with a pharmaceutically acceptable carrier. Such a composition may also contain (in addition to protein and a carrier) diluents, fillers, salts, buffers, stabilizers, solubiUzers, and other materials weU known in the art. The term "pharmaceuticaUy acceptable" means a non-toxic material that does not interfere with the effectiveness of the biological activity of the active ingredient(s). The characteristics of the carrier wiU depend on the route of administration. The pharmaceutical composition of the invention may also contain cytokines, lymphokines, or other hematopoietic factors such as M-CSF, GM-CSF, TNF, IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IFN, TNFO, TNF1, TNF2, G-CSF, Meg-CSF, thrombopoietin, stem ceU factor, and erythropoietin. The pharmaceutical composition may further contain other agents which either enhance the activity of the protein or compliment its activity or use in treatment. Such additional factors and /or agents may be included in the pharmaceutical composition to produce a synergistic effect with protein of the invention, or to rninimize side effects. Conversely, protein of the present invention may be included in formulations of the particular cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent to rninimize side effeds of the cytokine, lymphokine, other hematopoietic factor, thrombolytic or anti-thrombotic factor, or anti-inflammatory agent.
A protein of the present invention may be active in multimers (e.g., heterodimers or homodimers) or complexes with itself or other proteins. As a result, pharmaceutical
37 compositions of the invention may comprise a protein of the invention in such multimeric or complexed form.
The pharmaceutical composition of the invention may be in the form of a complex of the protein(s) of present invention along with protein or peptide antigens. The protein and/or peptide antigen wiU deUver a stimulatory signal to both B and T lymphocytes. B lymphocytes wiU respond to antigen through their surface immunoglobulin receptor. T lymphocytes wiU respond to antigen through the T ceU receptor (TCR) foUowing presentation of the antigen by MHC proteins. MHC and structuraUy related proteins including those encoded by dass I and class II MHC genes on host ceUs will serve to present the peptide antigen(s) to T lymphocytes. The antigen components could also be suppUed as purified MHC-peptide complexes alone or with co-stimulatory molecules that can directly signal T cells. Alternatively antibodies able to bind surface immunolgobuUn and other molecules on B ceUs as well as antibodies able to bind the TCR and other molecules on T ceUs can be combined with the pharmaceutical composition of the invention.
The pharmaceutical composition of the invention may be in the form of a Uposome in which protein of the present invention is combined, in addition to other pharmaceuticaUy acceptable carriers, with amphipathic agents such as Upids which exist in aggregated form as micelles, insoluble monolayers, Uquid crystals, or lameUar layers in aqueous solution. Suitable Upids for Uposomal formulation indude, without limitation, monoglycerides, diglycerides, sulfatides, lysolecithin, phospholipids, saponin, bile acids, and the like. Preparation of sud Uposomal formulations is within the level of skill in the art, as disclosed, for example, in U.S. Patent No.4,235,871; U.S. Patent No. 4,501,728; U.S. Patent No.4,837,028; and U.S. Patent No.4,737,323, all of which are incorporated herein by reference.
As used herein, the term "therapeutically effective amount" means the total amount of each active component of the pharmaceutical composition or method that is sufficient to show a meaningful patient benefit, i.e., treatment, healing, prevention or ameUoration of the relevant medical condition, or an increase in rate of treatment, healing, prevention or amelioration of such conditions. When appUed to an individual active ingredient, administered alone, the term refers to that ingredient alone. When applied to a combination, the term refers to combined amounts of the active ingredients that result in the therapeutic effect, whether administered in combination, seriaUy or simultaneously.
38 In practicing the method of treatment or use of the present invention, a therapeuticaUy effective amount of protein of the present invention is administered to a mammal having a condition to be treated. Protein of the present invention may be administered in accordance with the method of the invention either alone or in combination with other therapies such as treatments employing cytokines, lymphokines or other hematopoietic factors. When co-administered with one or more cytokines, lymphokines or other hematopoietic factors, protein of the present invention may be administered either simultaneously with the cytokine(s), lymphokine(s), other hematopoietic factor(s), thrombolytic or anti-thrombotic factors, or sequentiaUy. If administered sequentially, the attending physician will decide on the appropriate sequence of administering protein of the present invention in combination with cytokine(s), lymphokine(s), other hematopoietic factor(s), thrombolytic or anti-thrombotic factors.
Administration of protein of the present invention used in the pharmaceutical composition or to practice the method of the present invention can be carried out in a variety of conventional ways, such as oral ingestion, inhalation, topical appUcation or cutaneous, subcutaneous, intraperitoneal, parenteral or intravenous injection. Intravenous administration to the patient is preferred.
When a therapeuticaUy effective amount of protein of the present invention is administered oraUy, protein of the present invention will be in the form of a tablet, capsule, powder, solution or elixir. When administered in tablet form, the pharmaceutical composition of the invention may additionaUy contain a solid carrier such as a gelatin or an adjuvant. The tablet, capsule, and powder contain from about 5 to 95% protein of the present invention, and preferably from about 25 to 90% protein of the present invention. When administered in Uquid form, a Uquid carrier such as water, petroleum, oils of animal or plant origin such as peanut oil, mineral oU, soybean oil, or sesame oil, or synthetic oUs may be added. The liquid form of the pharmaceutical composition may further contain physiological saline solution, dextrose or other saccharide solution, or glycols such as ethylene glycol, propylene glycol or polyethylene glycol. When administered in liquid form, the pharmaceutical composition contains from about 0.5 to 90% by weight of protein of the present invention, and preferably from about 1 to 50% protein of the present invention.
When a therapeuticaUy effective amount of protein of the present invention is administered by intravenous, cutaneous or subcutaneous injection, protein of the present
39 invention wiU be in the form of a pyrogen-free, parenteraUy acceptable aqueous solution. The preparation of such parenteraUy acceptable protein solutions, having due regard to pH, isotonicity, stabihty, and the like, is within the skiU in the art. A preferred pharmaceutical composition for intravenous, cutaneous, or subcutaneous injection should contain, in addition to protein of the present invention, an isotorύc vehicle such as Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, Lactated Ringer's Injection, or other vehicle as known in the art. The pharmaceutical composition of the present invention may also contain stabuizers, preservatives, buffers, antioxidants, or other additives known to those of skill in the art. The amount of protein of the present invention in the pharmaceutical composition of the present invention wiU depend upon the nature and severity of the condition being treated, and on the nature of prior treatments which the patient has undergone. Ultimately, the attending physician wUl decide the amount of protein of the present invention with which to treat each individual patient. Initially, the attending physician will administer low doses of protein of the present invention and observe the patient's response. Larger doses of protein of the present invention may be administered until the optimal therapeutic effect is obtained for the patient, and at that point the dosage is not increased further. It is contemplated that the various pharmaceutical compositions used to practice the method of the present invention should contain about 0.01 μg to about 100 mg (preferably about O.lng to about 10 mg, more preferably about 0.1 μg to about 1 mg) of protein of the present invention per kg body weight.
The duration of intravenous therapy using the pharmaceutical composition of the present invention wiU vary, depending on the severity of the disease being treated and the condition and potential idiosyncratic response of each individual patient. It is contemplated that the duration of each appUcation of the protein of the present invention will be in the range of 12 to 24 hours of continuous intravenous administration. Ultimately the attending physician wiU dedde on the appropriate duration of intravenous therapy using the pharmaceutical composition of the present invention.
Protein of the invention may also be used to immunize animals to obtain polydonal and monoclonal antibodies which specifically react with the protein. As used herein, the term "antibody" includes without limitation a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a single-chain antibody, a CDR-grafted antibody, a humanized antibody, or fragments thereof which bind to the indicated protein.
40 Such term also includes any other species derived from an antibody or antibody sequence which is capable of binding the indicated protein.
Antibodies to a particular protein can be produced by methods well known to those skilled in the art. For example, monoclonal antibodies can be produced by generation of antibody-producing hybridomas in accordance with known methods (see for example, Goding, 1983, Monoclonal antibodies: principles and practice, Academic Press Inc., New York; and Yokoyama, 1992, "Production of Monoclonal Antibodies" in Current Protocols in Immunology, Unit 2.5, Greene Publishing Assoc. and John Wiley & Sons). Polyclonal sera and antibodies can be produced by inoculation of a mammalian subject with the relevant protein or fragments thereof in accordance with known methods. Fragments of antibodies, receptors, or other reactive peptides can be produced from the corresponding antibodies by cleavage of and collection of the desired fragments in accordance with known methods (see for example, Goding, supra; and Andrew et al., 1992, "Fragmentation of Immunoglobulins" in Current Protocols in Immunology, Unit 2.8, Greene Publishing Assoc. and John Wiley & Sons). Chimeric antibodies and single chain antibodies can also be produced in accordance with known recombinant methods (see for example, 5,169,939, 5,194,594, and 5,576,184). Humanized antibodies can also be made from corresponding murine antibodies in accordance with well known methods (see for example, U.S. Patent Nos. 5,530,101, 5,585,089, and 5,693,762). Additionally, human antibodies may be produced in non-human animals such as mice that have been genetically altered to express human antibody molecules (see for example Fishwild et al, 1996, Nature Biotechnology 14: 845-851; Mendez etal, 1997, Nature Genetics 15: 146-156 (erratum Nature Genetics 16: 410); and U.S. Patents 5,877,397 and 5,625,126). Such antibodies may be obtained using either the entire protein or fragments thereof as an immunogen. The peptide immunogens additionaUy may contain a cysteine residue at the carboxyl terminus, and are conjugated to a hapten such as keyhole limpet hemocyanin (KLH). Methods for synthesizing such peptides are known in the art, for example, as in R.P. Merrifield, J. Amer.Chem.Soc. 85, 2149-2154 (1963); J.L. Krstenansky, et al, FEBS Lett. 2U, 10 (1987). Monoclonal antibodies binding to the protein of the invention may be useful diagnostic agents for the immunodetection of the protein. Neutralizing monoclonal antibodies binding to the protein may also be useful therapeutics for both conditions associated with the protein and also in the treatment of some forms of cancer where
41 abnormal expression of the protein is involved. In the case of cancerous ceUs or leukemic cells, neutralizing monoclonal antibodies against the protein may be useful in detecting and preventing the metastatic spread of the cancerous cells, which may be mediated by the protein. For compositions of the present invention which are useful for bone, cartilage, tendon or Ugament regeneration, the therapeutic method includes administering the composition topically, systematically, or locaUy as an implant or device. When administered, the therapeutic composition for use in this invention is, of course, in a pyrogen-free, physiologically acceptable form. Further, the composition may desirably be encapsulated or injected in a viscous form for delivery to the site of bone, cartilage or tissue damage. Topical administration may be suitable for wound healing and tissue repair. TherapeuticaUy useful agents other than a protein of the invention which may also optionaUy be included in the composition as described above, may alternatively or additionaUy, be administered simultaneously or sequentiaUy with the composition in the methods of the invention. Preferably for bone and /or cartilage formation, the composition would include a matrix capable of delivering the protem-containing composition to the site of bone and/or cartilage damage, providing a structure for the developing bone and cartilage and optimally capable of being resorbed into the body. Such matrices may be formed of materials presently in use for other implanted medical applications.
The choice of matrix material is based on biocompatibuity, biodegradabiUty, mechanical properties, cosmetic appearance and interface properties. The particular application of the compositions will define the appropriate formulation. Potential matrices for the compositions may be biodegradable and chemically defined calcium sulfate, tricalciumphosphate, hydroxyapatite, polylactic acid, polyglycoUc acid and polyanhydrides. Other potential materials are biodegradable and biologicaUy weU- defined, such as bone or dermal collagen. Further matrices are comprised of pure proteins or extraceUular matrix components. Other potential matrices are nonbiodegradable and chemicaUy defined, such as sintered hydroxapatite, bioglass, aluminates, or other ceramics. Matrices may be comprised of combinations of any of the above mentioned types of material, such as polylactic acid and hydroxyapatite or coUagen and tricaldumphosphate. The bioceramics may be altered in composition, such as in caldum- aluminate-phosphate and processing to alter pore size, particle size, particle shape, and biodegradability.
42 Presently preferred is a 50:50 (mole weight) copolymer of lactic acid and glycolic acid in the form of porous particles having diameters ranging from 150 to 800 microns.
In some applications, it wiU be useful to utilize a sequestering agent, such as carboxymethyl cellulose or autologous blood clot, to prevent the protein compositions from disassociating from the matrix.
A preferred farrύly of sequestering agents is ceUulosic materials such as alkylcelluloses (including hydroxyalkylceUuloses), including methylcellulose, ethylceUulose, hydroxyethylceUulose, hydroxypropylcellulose, hydroxypropyl- methylcellulose, and carboxymethylceUulose, the most preferred being canonic salts of carboxymethylceUulose (CMC). Other preferred sequestering agents include hyaluronic acid, sodium alginate, poly(ethylene glycol), polyoxyethylene oxide, carboxyvinyl polymer and poly(vinyl alcohol). The amount of sequestering agent useful herein is 0.5-20 wt%, preferably 1-10 wt% based on total formulation weight, which represents the amount necessary to prevent desorbtion of the protein from the polymer matrix and to provide appropriate handling of the composition, yet not so much that the progenitor cells are prevented from infiltrating the matrix, thereby providing the protein the opportunity to assist the osteogenic activity of the progenitor ceUs.
In further compositions, proteins of the invention may be combined with other agents benefidal to the treatment of the bone and /or cartilage defed, wound, or tissue in question. These agents include various growth factors such as epidermal growth factor
(EGF), platelet derived growth factor (PDGF), transforming growth factors (TGF-α and
TGF-β), and insulin-like growth factor (IGF).
The therapeutic compositions are also presently valuable for veterinary applications. Particularly domestic animals and thoroughbred horses, in addition to humans, are desired patients for such treatment with proteins of the present invention.
The dosage regimen of a protem-containing pharmaceutical composition to be used in tissue regeneration wiU be determined by the attending physician considering various factors which modify the action of the proteins, e.g., amount of tissue weight desired to be formed, the site of damage, the condition of the damaged tissue, the size of a wound, type of damaged tissue (e.g., bone), the patient's age, sex, and diet, the severity of any infection, time of administration and other clinical factors. The dosage may vary with the type of matrix used in the reconstitution and with inclusion of other proteins in the pharmaceutical composition. For example, the addition of other known growth factors, such as IGF I (insuUn like growth factor I), to the final composition, may also effect
43 the dosage. Progress can be monitored by periodic assessment of tissue/bone growth and /or repair, for example, X-rays, histomorphometric determinations and tetracycUne labeling.
Polynudeotides of the present invention can also be used for gene therapy. Such polynucleotides can be introduced either in vivo or ex vivo into ceUs for expression in a mammaUan subjed. Polynudeotides of the invention may also be administered by other known methods for introduction of nudeic add into a ceU or organism (including, without
Umitation, in the form of viral vectors or naked DNA).
Cells may also be cultured ex vivo in the presence of proteins of the present invention in order to proliferate or to produce a desired effect on or activity in such cells. Treated ceUs can then be introduced in vivo for therapeutic purposes.
Patent and Uterature references cited herein are incorporated by reference as if fully set forth.
44

Claims

What is claimed is:
1. An isolated polynucleotide selected from the group consisting of:
(a) a polynucleotide comprising the nudeotide sequence of SEQ ID NO:l;
(b) a polynucleotide comprising the nucleotide sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3730;
(c) a polynucleotide comprising the nucleotide sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3640;
(d) a polynucleotide comprising the nucleotide sequence of the fuU- length protein coding sequence of clone GPI-122 deposited under accession number ATCC 98622;
(e) a polynucleotide encoding the full-length protein encoded by the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622;
(f) a polynucleotide encoding a protein comprising the amino acid sequence of SEQ ID NO:2;
(g) a polynucleotide encoding a protein comprising a fragment of the amino add sequence of SEQ ID NO:2, the fragment comprising eight contiguous amino acids of SEQ ID NO:2;
(h) a polynucleotide that hybridizes under conditions at least as stringent as 4X SSC at 65 degrees C, or 4X SSC at 42 degrees C with 50% formamide, to any one of the polynucleotides specified in (a)-(g); and
(i) a polynucleotide that hybridizes under conditions at least as stringent as 4X SSC at 50 degrees C, or 6X SSC at 40 degrees C with 50% formamide, to any one of the polynucleotides specified in (a)-(g), and that has a length that is at least 25% of the length of SEQ ID NO:l.
2. The polynucleotide of claim 1 wherein said polynucleotide is operably linked to at least one expression control sequence.
3. A host cell transformed with the polynucleotide of claim 2.
4. The host cell of claim 3, wherein said ceU is a mammalian ceU.
45
5. A process for producing a protein encoded by the polynudeotide of claim 2, which process comprises:
(a) growing a culture of the host ceU of claim 3 in a suitable culture medium; and
(b) purifying said protein from the culture.
6. A protein produced according to the process of claim 5.
7. An isolated polynucleotide encoding the protein of claim 6.
8. The polynucleotide of claim 7, wherein the polynucleotide comprises the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622.
9. A protein comprising an amino acid sequence selected from the group consisting of:
(a) the amino acid sequence of SEQ ID NO:2;
(b) the amino acid sequence of SEQ ID NO:2 from amino acid 1 to amino acid 1163;
(c) a fragment of the amino acid sequence of SEQ ID NO:2, the fragment comprising eight contiguous amino acids of SEQ ID NO:2; and
(d) the amino acid sequence encoded by the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622; the protein being substantiaUy free from other mammalian proteins.
10. The protein of claim 9, wherein said protein comprises the amino acid sequence of SEQ ID NO:2.
11. The protein of claim 9, wherein said protein comprises the amino acid sequence of SEQ ID NO:2 from amino acid 1 to amino acid 1163.
12. A composition comprising the protein of claim 9 and a pharmaceutically acceptable carrier.
46
13. The polynucleotide of claim 1, wherein the polynucleotide comprises the nucleotide sequence of SEQ ID NO:l.
14. The polynucleotide of claim 1, wherein the polynucleotide comprises the nucleotide sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3730.
15. The polynudeotide of claim 1, wherein the polynucleotide comprises the nucleotide sequence of SEQ ID NO:l from nucleotide 152 to nucleotide 3640.
16. The polynucleotide of claim 1, wherein the polynucleotide comprises the nudeotide sequence of the fuU-length protein coding sequence of clone GPI-122 deposited under accession number ATCC 98622.
17. The polynucleotide of daim 1, wherein the polynucleotide encodes the fuU- length protein encoded by the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622.
18. The polynucleotide of claim 1, wherein the polynucleotide comprises the nucleotide sequence of a mature protein coding sequence of GPI-122 deposited under accession number ATCC 98622.
19. The polynucleotide of claim 1, wherein the polynucleotide encodes a mature protein encoded by the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622.
20. The polynucleotide of claim 1, wherein the polynucleotide encodes a protein comprising the amino acid sequence of SEQ ID NO:2.
21. The polynucleotide of claim 1, wherein the polynucleotide encodes a protein comprising a fragment of the amino add sequence of SEQ ID NO:2, the fragment comprising eight contiguous amino acids of SEQ ID NO:2.
47
22. The protein of claim 9, wherein the protein comprises a fragment of the amino acid sequence of SEQ ID NO:2, the fragment comprising eight contiguous amino acids of SEQ ID NO:2.
23. The protein of claim 9, wherein the protein comprises the amino acid sequence encoded by the cDNA insert of clone GPI-122 deposited under accession number ATCC 98622.
24. An antibody that reacts specifically with the protein of claim 9.
25. The antibody of claim 24, wherein the antibody is a monoclonal antibody.
26. The antibody of claim 24, wherein the antibody is a human antibody.
27. A method for preventing, treating or ameliorating a medical condition which comprises administering to a mammaUan subject a therapeuticaUy effective amount of a composition of claim 12.
28. A method for preventing, treating or ameUorating a medical condition which comprises adrriinistering to a mammaUan subject a therapeuticaUy effective amount of an antibody of claim 24.
48
PCT/US1999/006459 1998-03-27 1999-03-26 Gpi-122, a novel glycophosphatidylinositol-anchored protein WO1999050435A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
AU33630/99A AU3363099A (en) 1998-03-27 1999-03-26 Gpi-122, a novel glycophosphatidylinositol-anchored protein
CA002326000A CA2326000A1 (en) 1998-03-27 1999-03-26 Gpi-122, a novel glycophosphatidylinositol-anchored protein
EP99915013A EP1066398A4 (en) 1998-03-27 1999-03-26 Gpi-122, a novel glycophosphatidylinositol-anchored protein

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US4965298A 1998-03-27 1998-03-27
US09/049,652 1998-03-27

Publications (1)

Publication Number Publication Date
WO1999050435A1 true WO1999050435A1 (en) 1999-10-07

Family

ID=21960956

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1999/006459 WO1999050435A1 (en) 1998-03-27 1999-03-26 Gpi-122, a novel glycophosphatidylinositol-anchored protein

Country Status (4)

Country Link
EP (1) EP1066398A4 (en)
AU (1) AU3363099A (en)
CA (1) CA2326000A1 (en)
WO (1) WO1999050435A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002012318A1 (en) * 2000-06-12 2002-02-14 Biowindow Gene Development Inc. Shanghai A novel peptide--glycophosphatidylinositol f11.22 and the polynucleotide coding this novel peptide

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5374548A (en) * 1986-05-02 1994-12-20 Genentech, Inc. Methods and compositions for the attachment of proteins to liposomes using a glycophospholipid anchor

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5374548A (en) * 1986-05-02 1994-12-20 Genentech, Inc. Methods and compositions for the attachment of proteins to liposomes using a glycophospholipid anchor

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
CONZELMANN et al., "Biosynthesis of Glycophosphoinositol Anchors in Saccharomyces Cerevisiae", CELL BIOLOGY INTERNATIONAL REPORTS, 1991, Vol. 15, No. 9, pages 863-873, XP002919035 *
FIELD M C, MENON A K, CROSS G A M: "DEVELOPMENTAL VARIATION OF GLYCOSYLPHOSPHATIDYLINODITOL MEMBRANE ANCHORS IN TRYPANOSOMA BRUCEI IN VITRO BIOSYNTHESIS OF INTERMEDIATES IN THE CONSTRUCTION OF THE GPI ANCHOR OF THE MAJOR PROCYCLE SURFACE GLYCOPROTEIN", JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY FOR BIOCHEMISTRY AND MOLECULAR BIOLOGY, US, vol. 267, no. 08, 15 March 1992 (1992-03-15), US, pages 5324 - 5327, XP002919034, ISSN: 0021-9258 *
METZ et al., "Characterization of the Plasma Glycosylphosphatidylinositol-Specific Phospholipase D (GPI-PLD)", CELL BIOLOGY INTERNATIONAL REPORTS, 1991, Vol. 15, No. 9, pages 875-882, XP002919033 *
See also references of EP1066398A4 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002012318A1 (en) * 2000-06-12 2002-02-14 Biowindow Gene Development Inc. Shanghai A novel peptide--glycophosphatidylinositol f11.22 and the polynucleotide coding this novel peptide

Also Published As

Publication number Publication date
AU3363099A (en) 1999-10-18
EP1066398A4 (en) 2002-03-20
CA2326000A1 (en) 1999-10-07
EP1066398A1 (en) 2001-01-10

Similar Documents

Publication Publication Date Title
US8048984B2 (en) Human GIL-19/AE289 proteins
EP2357193B1 (en) Human GIL-19/AE289 proteins and polynucleotides encoding same
WO1998040485A1 (en) Human secreted protein, ba3.1, and polynucleotides encoding same
WO1999041284A1 (en) Secreted proteins and polynucleotides encoding them
WO1999046287A1 (en) Secreted proteins and polynucleotides encoding them
WO2000050592A1 (en) Secreted proteins and polynucleotides encoding them
EP1897950B1 (en) Human GIL-19/AE289 proteins and polynucleotides encoding same
WO1999026973A1 (en) Secreted proteins and polynucleotides encoding them
EP1066398A1 (en) Gpi-122, a novel glycophosphatidylinositol-anchored protein
EP1051481A1 (en) Secreted proteins and polynucleotides encoding them
WO1999037674A1 (en) Secreted proteins and polynucleotides encoding them
US20020150563A1 (en) GPI-122, a novel glycophosphatidylinositol-anchored protein
EP1068308A1 (en) Secreted proteins and polynucleotides encoding them
WO2001075133A1 (en) Secreted proteins and polynucleotides encoding them
EP1037970A1 (en) Secreted proteins and polynucleotides encoding them
EP1028974A1 (en) Secreted proteins and polynucleotides encoding them
EP1044220A1 (en) Secreted proteins and polynucleotides encoding them
WO2000063692A1 (en) Secreted proteins and polynucleotides encoding them
AU4676100A (en) Human gil-19/ae289 proteins and polynucleotides encoding same
EP1047772A1 (en) Secreted proteins and polynucleotides encoding them
WO2000066148A1 (en) Novel proteins
WO1999028335A1 (en) Secreted proteins and polynucleotides encoding them

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AL AM AT AU AZ BA BB BG BR BY CA CH CN CU CZ DE DK EE ES FI GB GE GH GM HU ID IL IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MD MG MK MN MW MX NO NZ PL PT RO RU SD SE SG SI SK SL TJ TM TR TT UA UG UZ VN YU ZW

AL Designated countries for regional patents

Kind code of ref document: A1

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

121 Ep: the epo has been informed by wipo that ep was designated in this application
ENP Entry into the national phase

Ref document number: 2326000

Country of ref document: CA

Ref document number: 2326000

Country of ref document: CA

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2000 541323

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: KR

WWE Wipo information: entry into national phase

Ref document number: PA/A/2000/009528

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 1999915013

Country of ref document: EP

WWP Wipo information: published in national office

Ref document number: 1999915013

Country of ref document: EP

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Ref document number: 1999915013

Country of ref document: EP

DPE1 Request for preliminary examination filed after expiration of 19th month from priority date (pct application filed from 20040101)