WO2000043512A1 - Polypeptide humain avec multiples domaines du type facteur de croissance epidermique (egf), denomme zntr2 - Google Patents

Polypeptide humain avec multiples domaines du type facteur de croissance epidermique (egf), denomme zntr2 Download PDF

Info

Publication number
WO2000043512A1
WO2000043512A1 PCT/US2000/001419 US0001419W WO0043512A1 WO 2000043512 A1 WO2000043512 A1 WO 2000043512A1 US 0001419 W US0001419 W US 0001419W WO 0043512 A1 WO0043512 A1 WO 0043512A1
Authority
WO
WIPO (PCT)
Prior art keywords
zntr2
amino acid
seq
nucleic acid
sequence
Prior art date
Application number
PCT/US2000/001419
Other languages
English (en)
Inventor
James L. Holloway
Catherine E. Lofton-Day
Teresa Gilbert
Original Assignee
Zymogenetics, 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 Zymogenetics, Inc. filed Critical Zymogenetics, Inc.
Priority to CA002358873A priority Critical patent/CA2358873A1/fr
Priority to AU28546/00A priority patent/AU2854600A/en
Publication of WO2000043512A1 publication Critical patent/WO2000043512A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates generally to a new polypeptide having diagnostic and therapeutic uses.
  • the present invention relates to a novel polypeptide, designated "Zntr2,” and to nucleic acid molecules encoding Zntr2.
  • hormones and polypeptide growth factors are controlled by hormones and polypeptide growth factors. These diffusable molecules allow cells to communicate with each other and act in concert to form tissues and organs, and to repair and regenerate damaged tissue.
  • hormones and growth factors include the steroid hormones, parathyroid hormone, follicle stimulating hormone, the interferons, the interleukins, platelet derived growth factor, epidermal growth factor, and granulocyte-macrophage colony stimulating factor.
  • Certain receptors are integral membrane proteins that bind with the hormone or growth factor outside the cell, and that are linked to signaling pathways within the cell, such as second messenger systems.
  • Other classes of receptors are soluble intracellular molecules.
  • Epidermal growth factor one of the first growth factors discovered, exerts a potent mitogenic effect on epithelial cells, endothelial cells, and fibroblasts.
  • epidermal growth factor has been used as an adjunct for wound healing, and to treat patients affected by conditions such as necrotizing enterocolitis, Zollinger-Ellison syndrome, gastrointestinal ulceration, and congenital microvillus atrophy (Guglietta and Sullivan, Eur. J. Gastroenterol. Hepatol. 7:945 (1995); Rosenthal et al, In Vivo 11:201 (1997); G ⁇ n ⁇ l et al., J Pharm. Pharmacol. 50:641 (1998)).
  • a peptide domain that characterizes epidermal growth factor consists of about 30 to 50 amino residues and includes three disulfide bonds (see, for example, Nakayama et al, Genomics 51:27 (1998)). This region of epidermal growth factor is believed to play a role in critical extracellular functions, including cell adhesion and receptor-ligand interactions.
  • Epidermal growth factor-like domains are found in a variety of proteins that are either secreted proteins or membrane-bound proteins that contain the domains within the extracellular region. Proteins that contain epidermal growth factor-like domains include differentiation inhibitors, differentiation stimulators, coagulation factors, and growth factor activators, among others.
  • the present invention provides a novel polypeptide, designated "Zntr2.”
  • the present invention also provides variant Zntr2 polypeptides, Zntr2 fusion proteins, nucleic acid molecules encoding such polypeptides and proteins, and methods for using these nucleotide and amino acid sequences.
  • the present invention provides nucleic acid molecules that encode a new human polypeptide, designated as "Zntr2," that is characterized by multiple epidermal growth factor-like domains.
  • Zntr2 a new human polypeptide
  • An illustrative nucleic acid molecule containing a sequence that encodes the Zntr2 polypeptide has the nucleotide sequence of SEQ ID NO:l.
  • the encoded polypeptide has the following amino acid sequence: MNGGAERAMR SLPSLGGLAL LCCAAAAAAA AVASAASAGN
  • the Zntr2 signal sequence includes amino acid residues 1 to 30 of SEQ ID NO:2, although, amino acid residues 9 to 30 can also provide a useful signal sequence.
  • the Zntr2 extracellular domain includes amino acid residues 31 to 507 of SEQ ID NO:2, the transmembrane domain includes amino acid residues 508 to 533 of SEQ ID NO:2, and the intracellular domain includes amino acid residues 534 to 602 of SEQ ID NO:2.
  • the Zntr2 amino acid sequence also contains multiple repeats of two epidermal growth factor related motifs.
  • An epidermal growth factor-like signature has the following structure: x(4)-C-x(0,48)-C-x(3,12)-C-x(l,70)-C-x(l,6)-C-x(2)-G-a- x(0,21)-G-x(2)-C-x, where "x" is any amino acid, "a” is often an aromatic amino acid, and a range within parenthesis indicates the occurrence of "x" (PROSITE sequence No. PDOC00021 of Release 15.0; Bairoch et al, Nucleic Acids Res. 24:2 7 (1997)).
  • This motif is present in SEQ ID NO:2 at amino acid residues 200 to 236, and 250 to 284.
  • the amino acid represented by "a" in the signature is valine, rather than an aromatic amino acid.
  • the region between the fourth and sixth cysteines, which includes two conserved glycine residues, can exhibit the following pattern: C-x-C-x(5)- G-x(2)-C. This pattern can be found at amino acid residues 224 to 235 and 272 to 283 of SEQ ID NO:2.
  • a laminin-type epidermal growth factor-like domain signature which follows the pattern: C-x(l,2)-C-x(5)-G-x(2)-C-x(2)-C-x(3,4)-[FYW]-x(3,15)-C, where "x" is any amino acid, the values in parentheses indicate either the number or range of occurrence of "x,” and acceptable amino acids are indicated in square brackets (PROSITE sequence No. PS01248 of Release 15.0).
  • amino acid residues 224 to 256 (alternatively: amino acid residues 224 to 254, or amino acid residues 224 to 251)
  • amino acid residues 272 to 303 (alternatively: amino acid residues 272 to 301, or amino acid residues 272 to 298)
  • amino acid residues 317 to 351 (alternatively: amino acid residues 317 to 349, or amino acid residues 317 to 346)
  • amino acid residues 371 to 402 alternatively: amino acid residues 371 to 400, or amino acid residues 371 to 397
  • amino acid residues 422 to 449 amino acid residues 224 to 256
  • amino acid residues 272 to 303 (alternatively: amino acid residues 272 to 301, or amino acid residues 272 to 298)
  • amino acid residues 317 to 351 (alternatively: amino acid residues 317 to 349, or amino acid residues 317 to 346)
  • amino acid residues 371 to 402 (alternatively: amino acid
  • Zntr2 chromosome 9 at 9q33. This region is associated with a lethal congenital syndrome, as described below.
  • Northern analyses evidenced two transcript sizes of Zntr2, approximately 3.4 kilobases and 6.5 kilobases in length. Strong expression of the 3.4 kilobase transcript was observed in peripheral blood leukocytes, while weaker expression was seen in spinal cord, bone marrow, spleen, testis, ovary and colon. Every tissue examined showed expression of the larger transcript except thymus. The strongest expression of the larger transcript was observed in brain, spleen, peripheral blood leukocytes and spinal cord. A high level of expression was also found in fetal brain tissue.
  • the present invention provides isolated polypeptides comprising an extracellular domain, wherein the extracellular domain comprises amino acid residues 31 to 507 of the amino acid sequence of SEQ ID NO:2.
  • Such polypeptides may further comprise a transmembrane domain that resides in a carboxyl- terminal position relative to the extracellular domain, wherein the transmembrane domain comprises amino acid residues 508 to 533 of SEQ ID NO:2.
  • polypeptides may also comprise an intracellular domain that resides in a carboxyl- terminal position relative to the transmembrane domain, wherein the intracellular domain comprises amino acid residues 534 to 602 of SEQ ID NO:2, and optionally, a signal secretory sequence that resides in an amino-terminal position relative to the extracellular domain, wherein the signal secretory sequence comprises either amino acid residues 1 to 30 or 9 to 30 of the amino acid sequence of SEQ ID NO:2.
  • the present invention also contemplates isolated polypeptides having an amino acid sequence that is at least 70%, at least 80%, or at least 90% identical to the amino acid sequence of SEQ ID NO:2, the amino acid sequence of amino acid residues 31 to 227 of SEQ ID NO:2, or the amino acid sequence of amino acid residues 31 to 507 of SEQ ID NO:2, wherein such isolated polypeptides can specifically bind with an antibody that specifically binds with a polypeptide consisting of the amino acid sequence of SEQ ID NO:2.
  • An illustrative polypeptide is a polypeptide that comprises amino acid residues 31 to 602 of the amino acid sequence of SEQ ID NO:2.
  • polypeptides include polypeptides that comprise at least one epidermal growth factor-like signature having one of the following motifs: x(4)-C-x-C-x(10)-C- x(6)-C-x-C-x(2)-G-x(2)-G-x(2)-C-x, or x(4)-C-x-C-x(9)-C-x(5)-C-x-C-x(2)-G-x(2)-G- x(2)-C-x, wherein "x" is any amino acid, and wherein the values in parenthesis indicate the number of occurrences of "x.”
  • such polypeptides can comprise at least one amino acid sequence of amino acid residues 200 to 236 of SEQ ID NO:2 or an amino acid sequence of amino acid residues 250 to 284 of SEQ ID NO:2.
  • polypeptides can comprise both amino acid sequences.
  • Other illustrative polypeptides include polypeptides that further comprise at least one laminin-type epidermal growth factor-like domain signature having one of the following motifs: C-x- C-x(5)-G-x(2)-C-x(2)-C-x(3)-F-x(8)-C, C-x-C-x(5)-G-x(2)-C-x(2)-C-x(3)-Y-x(7)-C, C- x(2)-C-x(5)-G-x(2)-C-x(2)-C-x(3)-F-x(9)-C, C-x(2)-C-x(5)-G-x(2)-C-x(2)-C-x(3)-Y- x(6)-C, and C-x(2)-C-x(5)-G-x(2)-C-x(2)-C-x(3)-Y-x(7)-C, wherein "x" is any amino acid, and wherein the values in parenthesis indicate the number of occurrences of "x.”
  • the present invention also contemplates polypeptides that comprise at least 15 contiguous amino acid residues of any of the following amino acid sequences of SEQ ID NO:2: (a) amino acid residues 1 to 220, (b) amino acid residues 31 to 220, (c) amino acid residues 1 to 241, (d) amino acid residues 31 to 241, (e) amino acid residues 1 to 507, (f) amino acid residues 9 to 507, (g) amino acid residues 508 to 533, and (h) amino acid residues 534 to 602, or at least 30 contiguous amino acid residues of any of the following amino acid sequences of SEQ ID NO:2: (i) amino acid residues 1 to 235, (j) amino acid residues 31 to 235, (k) amino acid residues 1 to 256, (1) amino acid residues 31 to 256, (m) amino acid residues 1 to 507, (n) amino acid residues 9 to 507, (o) amino acid residues 508 to 533, and (p) amino acid residue
  • the present invention also includes polypeptides comprising, or consisting of, amino acid residues (a) to (p). Additional illustrative polypeptides described herein include polypeptides comprising, or consisting of, the following amino acid sequences of SEQ ID NO:2: amino acid residues 1 to 206, 31 to 206, 1 to 227, and 31 to 207.
  • the present invention further provides antibodies and antibody fragments that specifically bind with such polypeptides.
  • Exemplary antibodies include polyclonal antibodies, murine monoclonal antibodies, humanized antibodies derived from murine monoclonal antibodies, and human monoclonal antibodies.
  • Illustrative antibody fragments include F(ab') 2 , F(ab) 2 , Fab', Fab, Fv, scFv, and minimal recognition units.
  • the present invention further contemplates anti-idiotype antibodies that bind with such antibodies or antibody fragments.
  • the present invention also includes compositions comprising a carrier and a peptide, polypeptide, antibody, or anti-idiotype antibody described herein.
  • the present invention also provides isolated nucleic acid molecules that encode a Zntr2 polypeptide, wherein the nucleic acid molecule is selected from the group consisting of (a) a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:3, (b) a nucleic acid molecule encoding the amino acid sequence of SEQ ID NO:2, and (c) a nucleic acid molecule that remains hybridized following stringent wash conditions to a nucleic acid molecule having the nucleotide sequence of nucleotides 193-1908 of SEQ ID NO:l, or the complement of nucleotides 193-1908 of SEQ ID NO:l .
  • Illustrative nucleic acid molecules include those in which any difference between the amino acid sequence encoded by the nucleic acid molecule and the corresponding amino acid sequence of SEQ ID NO:2 is due to a conservative amino acid substitution.
  • the present invention further contemplates isolated nucleic acid molecules that comprise a nucleotide sequence of nucleotides 193-1908 of SEQ ID NO:l.
  • an isolated nucleic acid molecule can comprise the nucleotide sequence of nucleotides 103-1908 of SEQ ID NO:l.
  • the present invention also includes vectors and expression vectors comprising such nucleic acid molecules.
  • Such expression vectors may comprise a transcription promoter, and a transcription terminator, wherein the promoter is operably linked with the nucleic acid molecule, and wherein the nucleic acid molecule is operably linked with the transcription terminator.
  • the present invention further includes recombinant host cells comprising these vectors and expression vectors.
  • Illustrative host cells include bacterial, yeast, fungal, insect, mammalian, and plant cells.
  • Recombinant host cells comprising such expression vectors can be used to produce Zntr2 polypeptides by culturing such recombinant host cells that comprise the expression vector and that produce the Zntr2 protein, and, optionally, isolating the Zntr2 protein from the cultured recombinant host cells.
  • the present invention also contemplates methods for detecting the presence of Zntr2 RNA in a biological sample, comprising the steps of (a) contacting a Zntr2 nucleic acid probe under hybridizing conditions with either (i) test RNA molecules isolated from the biological sample, or (ii) nucleic acid molecules synthesized from the isolated RNA molecules, wherein the probe has a nucleotide sequence comprising a portion of the nucleotide sequence of nucleotides 193-1908 of SEQ ID NO:l, or its complement, and (b) detecting the formation of hybrids of the nucleic acid probe and either the test RNA molecules or the synthesized nucleic acid molecules, wherein the presence of the hybrids indicates the presence ofZntr2 RNA in the biological sample.
  • the biological sample can be a human biological sample.
  • the present invention further provides methods for detecting the presence of Zntr2 polypeptide in a biological sample, comprising the steps of: (a) contacting the biological sample with an antibody or an antibody fragment that specifically binds with a polypeptide consisting of the amino acid sequence of SEQ ID NO:2, wherein the contacting is performed under conditions that allow the binding of the antibody or antibody fragment to the biological sample, and (b) detecting any of the bound antibody or bound antibody fragment.
  • Such an antibody or antibody fragment may further comprise a detectable label selected from the group consisting of radioisotope, fluorescent label, chemiluminescent label, enzyme label, bioluminescent label, and colloidal gold.
  • An exemplary biological sample is a human biological sample.
  • kits for performing these detection methods may comprise a container that comprises a nucleic acid molecule, wherein the nucleic acid molecule is selected from the group consisting of (a) a nucleic acid molecule comprising the nucleotide sequence of nucleotides 193-1908 of SEQ ID NO:l, (b) a nucleic acid molecule comprising the complement of nucleotides 193-1908 of the nucleotide sequence of SEQ ID NO:l, (c) a nucleic acid molecule that is a fragment of (a) consisting of at least eight nucleotides, and (d) a nucleic acid molecule that is a fragment of (b) consisting of at least eight nucleotides.
  • kit may also comprise a second container that comprises one or more reagents capable of indicating the presence of the nucleic acid molecule.
  • a kit for detection of Zntr2 protein may comprise a container that comprises an antibody, or an antibody fragment, that specifically binds with a polypeptide consisting of the amino acid sequence of SEQ ID NO:2.
  • the present invention also contemplates isolated nucleic acid molecules comprising a nucleotide sequence that encodes a Zntr2 secretion signal sequence and a nucleotide sequence that encodes a biologically active polypeptide, wherein the Zntr2 secretion signal sequence comprises an amino acid sequence of either residues 1 to 30 of SEQ ID NO:2 or residues 9 to 30 of SEQ ID NO:2.
  • Exemplary biologically active polypeptide include Factor Vila, proinsulin, insulin, follicle stimulating hormone, tissue type plasminogen activator, tumor necrosis factor, interleukin, colony stimulating factor, interferon, erythropoietin, and thrombopoietin.
  • the present invention also provides fusion proteins comprising a Zntr2 secretion signal sequence and a polypeptide, wherein the Zntr2 secretion signal sequence comprises an amino acid sequence of either residues 1 to 30 of SEQ ID NO:2 or residues 9 to 30 of SEQ ID NO:2.
  • the present invention further includes isolated nucleic acid molecules that encode an extracellular Zntr2 domain, wherein the extracellular domain has the amino acid sequence of amino acid residues 31 to 507 of SEQ ID NO:2, and isolated polypeptides consisting of the amino acid sequence of amino acid residues 31 to 507 of SEQ ID NO:2.
  • the present invention also contemplates antibodies that specifically bind with such isolated polypeptides, and anti-idiotype antibodies that specifically bind with such antibodies. Such anti-idiotype antibodies can be used, for example, to mimic the Zntr2 extracellular domain.
  • the present invention provides fusion proteins that comprise a Zntr2 polypeptide moiety, and an immunoglobulin moiety, such as an immunoglobulin heavy chain constant region.
  • nucleic acid or “nucleic acid molecule” refers to polynucleotides, such as deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), oligonucleotides, fragments generated by the polymerase chain reaction (PCR), and fragments generated by any of ligation, scission, endonuclease action, and exonuclease action.
  • Nucleic acid molecules can be composed of monomers that are naturally- occurring nucleotides (such as DNA and RNA), or analogs of naturally-occurring nucleotides (e.g., ⁇ -enantiomeric forms of naturally-occurring nucleotides), or a combination of both.
  • Modified nucleotides can have alterations in sugar moieties and/or in pyrimidine or purine base moieties.
  • Sugar modifications include, for example, replacement of one or more hydroxyl groups with halogens, alkyl groups, amines, and azido groups, or sugars can be functionalized as ethers or esters.
  • the entire sugar moiety can be replaced with sterically and electronically similar structures, such as aza-sugars and carbocyclic sugar analogs.
  • modifications in a base moiety include alkylated purines and pyrimidines, acylated purines or pyrimidines, or other well-known heterocyclic substitutes.
  • Nucleic acid monomers can be linked by phosphodiester bonds or analogs of such linkages.
  • nucleic acid molecule also includes so- called “peptide nucleic acids,” which comprise naturally-occurring or modified nucleic acid bases attached to a polyamide backbone. Nucleic acids can be either single stranded or double stranded.
  • complement of a nucleic acid molecule refers to a nucleic acid molecule having a complementary nucleotide sequence and reverse orientation as compared to a reference nucleotide sequence. For example, the sequence 5' ATGCACGGG 3' is complementary to 5' CCCGTGCAT 3'.
  • contig denotes a nucleic acid molecule that has a contiguous stretch of identical or complementary sequence to another nucleic acid molecule. Contiguous sequences are said to "overlap" a given stretch of a nucleic acid molecule either in their entirety or along a partial stretch of the nucleic acid molecule.
  • degenerate nucleotide sequence denotes a sequence of nucleotides that includes one or more degenerate codons as compared to a reference nucleic acid molecule that encodes a polypeptide.
  • Degenerate codons contain different triplets of nucleotides, but encode the same amino acid residue (i.e., GAU and GAC triplets each encode Asp).
  • structural gene refers to a nucleic acid molecule that is transcribed into messenger RNA (mRNA), which is then translated into a sequence of amino acids characteristic of a specific polypeptide.
  • an "isolated nucleic acid molecule” is a nucleic acid molecule that is not integrated in the genomic DNA of an organism.
  • a DNA molecule that encodes a growth factor that has been separated from the genomic DNA of a cell is an isolated DNA molecule.
  • Another example of an isolated nucleic acid molecule is a chemically-synthesized nucleic acid molecule that is not integrated in the genome of an organism.
  • a nucleic acid molecule that has been isolated from a particular species is smaller than the complete DNA molecule of a chromosome from that species.
  • nucleic acid molecule construct is a nucleic acid molecule, either single- or double-stranded, that has been modified through human intervention to contain segments of nucleic acid combined and juxtaposed in an arrangement not existing in nature.
  • Linear DNA denotes non-circular DNA molecules having free 5' and 3' ends.
  • Linear DNA can be prepared from closed circular DNA molecules, such as plasmids, by enzymatic digestion or physical disruption.
  • cDNA complementary DNA
  • cDNA is a single-stranded DNA molecule that is formed from an mRNA template by the enzyme reverse transcriptase. Typically, a primer complementary to portions of mRNA is employed for the initiation of reverse transcription.
  • cDNA to refer to a double- stranded DNA molecule consisting of such a single-stranded DNA molecule and its complementary DNA strand.
  • cDNA also refers to a clone of a cDNA molecule synthesized from an RNA template.
  • a "promoter” is a nucleotide sequence that directs the transcription of a structural gene. Typically, a promoter is located in the 5' non-coding region of a gene, proximal to the transcriptional start site of a structural gene. Sequence elements within promoters that function in the initiation of transcription are often characterized by consensus nucleotide sequences. These promoter elements include RNA polymerase binding sites, TATA sequences, CAAT sequences, differentiation-specific elements (DSEs; McGehee et al, Mol.
  • CREs cyclic AMP response elements
  • SREs serum response elements
  • GREs glucocorticoid response elements
  • binding sites for other transcription factors such as CRE/ATF (O'Reilly et al, J. Biol. Chem. 267: 9938 (1992)), AP2 (Ye et al., J. Biol. Chem. 269:25728 (1994)), SP1, cAMP response element binding protein (CREB; Loeken, Gene Expr.
  • CRE/ATF O'Reilly et al, J. Biol. Chem. 267: 9938 (1992)
  • AP2 Ye et al., J. Biol. Chem. 269:25728 (1994)
  • SP1, cAMP response element binding protein CREB; Loeken, Gene Expr.
  • a promoter is an inducible promoter, then the rate of transcription increases in response to an inducing agent. In contrast, the rate of transcription is not regulated by an inducing agent if the promoter is a constitutive promoter. Repressible promoters are also known.
  • a “core promoter” contains essential nucleotide sequences for promoter function, including the TATA box and start of transcription. By this definition, a core promoter may or may not have detectable activity in the absence of specific sequences that may enhance the activity or confer tissue specific activity.
  • a “regulatory element” is a nucleotide sequence that modulates the activity of a core promoter.
  • a regulatory element may contain a nucleotide sequence that binds with cellular factors enabling transcription exclusively or preferentially in particular cells, tissues, or organelles. These types of regulatory elements are normally associated with genes that are expressed in a "cell-specific,” “tissue-specific,” or “organelle-specific” manner.
  • An “enhancer” is a type of regulatory element that can increase the efficiency of transcription, regardless of the distance or orientation of the enhancer relative to the start site of transcription.
  • Heterologous DNA refers to a DNA molecule, or a population of DNA molecules, that does not exist naturally within a given host cell.
  • DNA molecules heterologous to a particular host cell may contain DNA derived from the host cell species (i.e., endogenous DNA) so long as that host DNA is combined with non-host DNA (i.e., exogenous DNA).
  • endogenous DNA DNA derived from the host cell species
  • non-host DNA i.e., exogenous DNA
  • a DNA molecule containing a non-host DNA segment encoding a polypeptide operably linked to a host DNA segment comprising a transcription promoter is considered to be a heterologous DNA molecule.
  • a heterologous DNA molecule can comprise an endogenous gene operably linked with an exogenous promoter.
  • a DNA molecule comprising a gene derived from a wild-type cell is considered to be heterologous DNA if that DNA molecule is introduced into a mutant cell that lacks the wild-type gene.
  • a "polypeptide” is a polymer of amino acid residues joined by peptide bonds, whether produced naturally or synthetically. Polypeptides of less than about 10 amino acid residues are commonly referred to as "peptides.”
  • a “protein” is a macromolecule comprising one or more polypeptide chains.
  • a protein may also comprise non-peptidic components, such as carbohydrate groups. Carbohydrates and other non-peptidic substituents may be added to a protein by the cell in which the protein is produced, and will vary with the type of cell. Proteins are defined herein in terms of their amino acid backbone structures; substituents such as carbohydrate groups are generally not specified, but may be present nonetheless.
  • a peptide or polypeptide encoded by a non-host DNA molecule is a
  • heterologous peptide or polypeptide.
  • integrated genetic element is a segment of DNA that has been incorporated into a chromosome of a host cell after that element is introduced into the cell through human manipulation.
  • integrated genetic elements are most commonly derived from linearized plasmids that are introduced into the cells by electroporation or other techniques. Integrated genetic elements are passed from the original host cell to its progeny.
  • a "cloning vector” is a nucleic acid molecule, such as a plasmid, cosmid, or bacteriophage, that has the capability of replicating autonomously in a host cell.
  • Cloning vectors typically contain one or a small number of restriction endonuclease recognition sites that allow insertion of a nucleic acid molecule in a determinable fashion without loss of an essential biological function of the vector, as well as nucleotide sequences encoding a marker gene that is suitable for use in the identification and selection of cells transformed with the cloning vector.
  • Marker genes typically include genes that provide tetracycline resistance or ampicillin resistance.
  • an “expression vector” is a nucleic acid molecule encoding a gene that is expressed in a host cell.
  • an expression vector comprises a transcription promoter, a gene, and a transcription terminator. Gene expression is usually placed under the control of a promoter, and such a gene is said to be “operably linked to” the promoter.
  • a regulatory element and a core promoter are operably linked if the regulatory element modulates the activity of the core promoter.
  • a “recombinant host” is a cell that contains a heterologous nucleic acid molecule, such as a cloning vector or expression vector. In the present context, an example of a recombinant host is a cell that produces Zntr2 from an expression vector.
  • Zntr2 can be produced by a cell that is a "natural source” of Zntr2, and that lacks an expression vector.
  • “Integrative transformants” are recombinant host cells, in which heterologous DNA has become integrated into the genomic DNA of the cells.
  • a “fusion protein” is a hybrid protein expressed by a nucleic acid molecule comprising nucleotide sequences of at least two genes.
  • a fusion protein can comprise at least part of a Zntr2 polypeptide fused with a polypeptide that binds an affinity matrix.
  • Such a fusion protein provides a means to isolate large quantities of Zntr2 using affinity chromatography.
  • Receptor denotes a cell-associated protein that binds to a bioactive molecule termed a "ligand.” This interaction mediates the effect of the ligand on the cell.
  • Receptors can be membrane bound, cytosolic or nuclear; monomeric (e.g., thyroid stimulating hormone receptor, beta-adrenergic receptor) or multimeric (e.g., PDGF receptor, growth hormone receptor, IL-3 receptor, GM-CSF receptor, G-CSF receptor, erythropoietin receptor and IL-6 receptor).
  • Membrane-bound receptors are characterized by a multi-domain structure comprising an extracellular ligand-binding domain and an intracellular effector domain that is typically involved in signal transduction. In certain membrane-bound receptors, the extracellular ligand-binding domain and the intracellular effector domain are located in separate polypeptides that comprise the complete functional receptor.
  • the binding of ligand to receptor results in a conformational change in the receptor that causes an interaction between the effector domain and other molecule(s) in the cell, which in turn leads to an alteration in the metabolism of the cell.
  • Metabolic events that are often linked to receptor-ligand interactions include gene transcription, phosphorylation, dephosphorylation, increases in cyclic AMP production, mobilization of cellular calcium, mobilization of membrane lipids, cell adhesion, hydrolysis of inositol lipids and hydrolysis of phospholipids.
  • secretory signal sequence denotes a nucleotide sequence that encodes a peptide (a "secretory peptide") that, as a component of a larger polypeptide, directs the larger polypeptide through a secretory pathway of a cell in which it is synthesized.
  • secretory peptide a nucleotide sequence that encodes a peptide that, as a component of a larger polypeptide, directs the larger polypeptide through a secretory pathway of a cell in which it is synthesized.
  • secretory signal sequence denotes a nucleotide sequence that encodes a peptide (a "secretory peptide”) that, as a component of a larger polypeptide, directs the larger polypeptide through a secretory pathway of a cell in which it is synthesized.
  • the larger polypeptide is commonly cleaved to remove the secretory peptide during transit through the secretory pathway.
  • isolated polypeptide is a polypeptide that is essentially free from contaminating cellular components, such as carbohydrate, lipid, or other proteinaceous impurities associated with the polypeptide in nature.
  • a preparation of isolated polypeptide contains the polypeptide in a highly purified form, i.e., at least about 80% pure, at least about 90% pure, at least about 95%) pure, greater than 95% pure, or greater than 99% pure.
  • SDS sodium dodecyl sulfate
  • isolated does not exclude the presence of the same polypeptide in alternative physical forms, such as dimers or alternatively glycosylated, derivatized, or phosphorylated forms.
  • amino-terminal and “carboxyl-terminal” are used herein to denote positions within polypeptides. Where the context allows, these terms are used with reference to a particular sequence or portion of a polypeptide to denote proximity or relative position. For example, a certain sequence positioned carboxyl-terminal to a reference sequence within a polypeptide is located proximal to the carboxyl terminus of the reference sequence, but is not necessarily at the carboxyl terminus of the complete polypeptide.
  • expression refers to the biosynthesis of a gene product.
  • expression involves transcription of the structural gene into mRNA and the translation of mRNA into one or more polypeptides.
  • splice variant is used herein to denote alternative forms of RNA transcribed from a gene. Splice variation arises naturally through use of alternative splicing sites within a transcribed RNA molecule, or less commonly between separately transcribed RNA molecules, and may result in several mRNAs transcribed from the same gene. Splice variants may encode polypeptides having altered amino acid sequence. The term splice variant is also used herein to denote a polypeptide encoded by a splice variant of an mRNA transcribed from a gene.
  • immunomodulator includes cytokines, stem cell growth factors, lymphotoxins, co-stimulatory molecules, hematopoietic factors, and synthetic analogs of these molecules.
  • complement/anti-complement pair denotes non-identical moieties that form a non-covalently associated, stable pair under appropriate conditions.
  • biotin and avidin are prototypical members of a complement/anti-complement pair.
  • Other exemplary complement/anti-complement pairs include receptor/ligand pairs, antibody/antigen (or hapten or epitope) pairs, sense/antisense polynucleotide pairs, and the like.
  • the complement/anti-complement pair preferably has a binding affinity of less than 10 9 M " '.
  • An "anti-idiotype antibody” is an antibody that binds with the variable region domain of an immunoglobulin. In the present context, an anti-idiotype antibody binds with the variable region of an anti-Zntr2 antibody, and thus, an anti-idiotype antibody mimics an epitope of Zntr2.
  • an “antibody fragment” is a portion of an antibody such as F(ab') 2 , F(ab) 2 , Fab', Fab, and the like. Regardless of structure, an antibody fragment binds with the same antigen that is recognized by the intact antibody. For example, an anti-Zntr2 monoclonal antibody fragment binds with an epitope of Zntr2.
  • antibody fragment also includes a synthetic or a genetically engineered polypeptide that binds to a specific antigen, such as polypeptides consisting of the light chain variable region, "Fv” fragments consisting of the variable regions of the heavy and light chains, recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker ("scFv proteins”), and minimal recognition units consisting of the amino acid residues that mimic the hypervariable region.
  • scFv proteins recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker
  • a “chimeric antibody” is a recombinant protein that contains the variable domains and complementary determining regions derived from a rodent antibody, while the remainder of the antibody molecule is derived from a human antibody.
  • Humanized antibodies are recombinant proteins in which murine complementarity determining regions of a monoclonal antibody have been transferred from heavy and light variable chains of the murine immunoglobulin into a human variable domain.
  • a “therapeutic agent” is a molecule or atom which is conjugated to an antibody moiety to produce a conjugate which is useful for therapy.
  • therapeutic agents include drugs, toxins, immunomodulators, chelators, boron compounds, photoactive agents or dyes, and radioisotopes.
  • a “detectable label” is a molecule or atom which can be conjugated to an antibody moiety to produce a molecule useful for diagnosis.
  • detectable labels include chelators, photoactive agents, radioisotopes, fluorescent agents, paramagnetic ions, or other marker moieties.
  • the term "affinity tag” is used herein to denote a polypeptide segment that can be attached to a second polypeptide to provide for purification or detection of the second polypeptide or provide sites for attachment of the second polypeptide to a substrate.
  • any peptide or protein for which an antibody or other specific binding agent is available can be used as an affinity tag.
  • Affinity tags include a poly- histidine tract, protein A (Nilsson et al, EMBO J.
  • naked antibody is an entire antibody, as opposed to an antibody fragment, which is not conjugated with a therapeutic agent. Naked antibodies include both polyclonal and monoclonal antibodies, as well as certain recombinant antibodies, such as chimeric and humanized antibodies. As used herein, the term "antibody component" includes both an entire antibody and an antibody fragment.
  • an “immunoconjugate” is a conjugate of an antibody component with a therapeutic agent or a detectable label.
  • antibody fusion protein refers to a recombinant molecule that comprises an antibody component and a therapeutic agent.
  • therapeutic agents suitable for such fusion proteins include immunomodulators ("antibody-immunomodulator fusion protein”) and toxins (“antibody-toxin fusion protein”).
  • tumor associated antigen is a protein normally not expressed, or expressed at lower levels, by a normal counterpart cell.
  • tumor associated antigens include alpha-fetoprotein, carcinoembryonic antigen, and Her-2/neu. Many other illustrations of tumor associated antigens are known to those of skill in the art. See, for example, Urban et al, Ann. Rev. Immunol. 10:617 (1992).
  • a “target polypeptide” or a “target peptide” is an amino acid sequence that comprises at least one epitope, and that is expressed on a target cell, such as a tumor cell, or a cell that carries an infectious agent antigen.
  • T cells recognize peptide epitopes presented by a major histocompatibility complex molecule to a target polypeptide or target peptide and typically lyse the target cell or recruit other immune cells to the site of the target cell, thereby killing the target cell.
  • An “antigenic peptide” is a peptide which will bind a major histocompatibility complex molecule to form an MHC-peptide complex which is recognized by a T cell, thereby inducing a cytotoxic lymphocyte response upon presentation to the T cell.
  • antigenic peptides are capable of binding to an appropriate major histocompatibility complex molecule and inducing a cytotoxic T cells response, such as cell lysis or specific cytokine release against the target cell which binds or expresses the antigen.
  • the antigenic peptide can be bound in the context of a class I or class II major histocompatibility complex molecule, on an antigen presenting cell or on a target cell.
  • RNA polymerase II catalyzes the transcription of a structural gene to produce mRNA.
  • a nucleic acid molecule can be designed to contain an RNA polymerase II template in which the RNA transcript has a sequence that is complementary to that of a specific mRNA.
  • the RNA transcript is termed an "anti- sense RNA” and a nucleic acid molecule that encodes the anti-sense RNA is termed an "anti-sense gene.”
  • Anti-sense RNA molecules are capable of binding to mRNA molecules, resulting in an inhibition of mRNA translation.
  • an "anti-sense oligonucleotide specific for Zntr2" or an “Zntr2 anti- sense oligonucleotide” is an oligonucleotide having a sequence (a) capable of forming a stable triplex with a portion of the Zntr2 gene, or (b) capable of forming a stable duplex with a portion of an mRNA transcript of the Zntr2 gene.
  • a "ribozyme” is a nucleic acid molecule that contains a catalytic center.
  • the term includes RNA enzymes, self-splicing RNAs, self-cleaving RNAs, and nucleic acid molecules that perform these catalytic functions.
  • a nucleic acid molecule that encodes a ribozyme is termed a "ribozyme gene.”
  • an “external guide sequence” is a nucleic acid molecule that directs the endogenous ribozyme, RNase P, to a particular species of intracellular mRNA, resulting in the cleavage of the mRNA by RNase P.
  • a nucleic acid molecule that encodes an external guide sequence is termed an "external guide sequence gene.”
  • variant Zntr2 gene refers to nucleic acid molecules that encode a polypeptide having an amino acid sequence that is a modification of SEQ ID NO:2. Such variants include naturally-occurring polymorphisms of Zntr2 genes, as well as synthetic genes that contain conservative amino acid substitutions of the amino acid sequence of SEQ ID NO:2. Additional variant forms of Zntr2 genes are nucleic acid molecules that contain insertions or deletions of the nucleotide sequences described herein. A variant Zntr2 gene can be identified by determining whether the gene hybridizes with a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l, or its complement, under stringent conditions.
  • variant Zntr2 genes can be identified by sequence comparison. Two amino acid sequences have "100%) amino acid sequence identity” if the amino acid residues of the two amino acid sequences are the same when aligned for maximal correspondence. Similarly, two nucleotide sequences have "100% nucleotide sequence identity” if the nucleotide residues of the two nucleotide sequences are the same when aligned for maximal correspondence. Sequence comparisons can be performed using standard software programs such as those included in the LASERGENE bioinformatics computing suite, which is produced by DNASTAR (Madison, Wisconsin).
  • a variant gene or polypeptide encoded by a variant gene may be characterized by its ability to bind specifically to an anti-Zntr2 antibody.
  • allelic variant is used herein to denote any of two or more alternative forms of a gene occupying the same chromosomal locus. Allelic variation arises naturally through mutation, and may result in phenotypic polymorphism within populations. Gene mutations can be silent (no change in the encoded polypeptide) or may encode polypeptides having altered amino acid sequence.
  • allelic variant is also used herein to denote a protein encoded by an allelic variant of a gene.
  • ortholog denotes a polypeptide or protein obtained from one species that is the functional counterpart of a polypeptide or protein from a different species. Sequence differences among orthologs are the result of speciation.
  • Parenters are distinct but structurally related proteins made by an organism. Paralogs are believed to arise through gene duplication. For example, ⁇ - globin, ⁇ -globin, and myoglobin are paralogs of each other.
  • the present invention includes functional fragments of Zntr2 genes.
  • a "functional fragment" of a Zntr2 gene refers to a nucleic acid molecule that encodes a portion of a Zntr2 polypeptide specifically binds with an anti-Zntr2 antibody.
  • Nucleic acid molecules encoding a human Zntr2 gene can be obtained by screening a human cDNA or genomic library using polynucleotide probes based upon SEQ ID NO: 1. These techniques are standard and well-established. As an illustration, a nucleic acid molecule that encodes a human Zntr2 gene can be isolated from a human cDNA library. In this case, the first step would be to prepare the cDNA library by isolating RNA from peripheral blood leukocytes, brain, spleen, or spinal cord tissue, using methods well-known to those of skill in the art.
  • RNA isolation techniques must provide a method for breaking cells, a means of inhibiting RNase-directed degradation of RNA, and a method of separating RNA from DNA, protein, and polysaccharide contaminants.
  • total RNA can be isolated by freezing tissue in liquid nitrogen, grinding the frozen tissue with a mortar and pestle to lyse the cells, extracting the ground tissue with a solution of phenol/chloroform to remove proteins, and separating RNA from the remaining impurities by selective precipitation with lithium chloride (see, for example, Ausubel et ⁇ l (eds.), Short Protocols in Molecular Biology, 3 r " Edition, pages 4-1 to 4-6 (John Wiley & Sons 1995) ["Ausubel (1995)”]; Wu et al, Methods in Gene Biotechnology, pages 33-41 (CRC Press, Inc. 1997) ["Wu (1997)”]).
  • total RNA can be isolated from tissue by extracting ground tissue with guanidinium isothiocyanate, extracting with organic solvents, and separating RNA from contaminants using differential centrifugation (see, for example, Chirgwin et al, Biochemistry 18:52 (1979); Ausubel (1995) at pages 4-1 to 4-6; Wu (1997) at pages 33-41).
  • poly(A) + RNA In order to construct a cDNA library, poly(A) + RNA must be isolated from a total RNA preparation. Poly(A) + RNA can be isolated from total RNA using the standard technique of oligo(dT)-cellulose chromatography (see, for example, Aviv and Leder, Proc. Nat'l Acad. Sci. USA 69: 40 (1972); Ausubel (1995) at pages 4-11 to 4- 12).
  • Double-stranded cDNA molecules are synthesized from poly(A) + RNA using techniques well-known to those in the art. (see, for example, Wu (1997) at pages 41-46). Moreover, commercially available kits can be used to synthesize double- stranded cDNA molecules. For example, such kits are available from Life Technologies, Inc. (Gaithersburg, MD), CLONTECH Laboratories, Inc. (Palo Alto, CA), Promega Corporation (Madison, WI) and STRATAGENE (La Jolla, CA). Various cloning vectors are appropriate for the construction of a cDNA library.
  • a cDNA library can be prepared in a vector derived from bacteriophage, such as a ⁇ gtlO vector. See, for example, Huynh et al, "Constructing and Screening cDNA Libraries in ⁇ gtlO and ⁇ gtl l," in DNA Cloning: A Practical
  • double-stranded cDNA molecules can be inserted into a plasmid vector, such as a PBLUESCRIPT vector (STRATAGENE; La Jolla, CA), a LAMDAGEM-4 (Promega Corp.) or other commercially available vectors.
  • a plasmid vector such as a PBLUESCRIPT vector (STRATAGENE; La Jolla, CA), a LAMDAGEM-4 (Promega Corp.) or other commercially available vectors.
  • Suitable cloning vectors also can be obtained from the American Type Culture Collection
  • the cDNA library is inserted into a prokaryotic host, using standard techniques.
  • a cDNA library can be introduced into competent E. coli DH5 cells, which can be obtained, for example, from
  • a human genomic library can be prepared by means well-known in the art
  • Genomic DNA can be isolated by lysing tissue with the detergent Sarkosyl, digesting the lysate with proteinase K, clearing insoluble debris from the lysate by centrifugation, precipitating nucleic acid from the lysate using isopropanol, and purifying resuspended
  • DNA fragments that are suitable for the production of a genomic library can be obtained by the random shearing of genomic DNA or by the partial digestion of genomic DNA with restriction endonucleases.
  • Genomic DNA fragments can be inserted into a vector, such as a bacteriophage or cosmid vector, in accordance with conventional techniques, such as the use of restriction enzyme digestion to provide appropriate termini, the use of alkaline phosphatase treatment to avoid undesirable joining of DNA molecules, and ligation with appropriate ligases. Techniques for such manipulation are well-known in the art (see, for example, Ausubel (1995) at pages 5-1 to 5-6; Wu (1997) at pages 307-
  • Nucleic acid molecules that encode a human Zntr2 gene can also be obtained using the polymerase chain reaction (PCR) with oligonucleotide primers having nucleotide sequences that are based upon the nucleotide sequences of the human Zntr2 gene, as described herein.
  • PCR polymerase chain reaction
  • General methods for screening libraries with PCR are provided by, for example, Yu et al , "Use of the Polymerase Chain Reaction to Screen
  • human genomic libraries can be obtained from commercial sources such as Research Genetics (Huntsville, AL) and the American Type Culture Collection (Manassas, VA).
  • a library containing cDNA or genomic clones can be screened with one or more polynucleotide probes based upon SEQ ID NO:l, using standard methods (see, for example, Ausubel (1995) at pages 6-1 to 6-11).
  • Anti-Zntr2 antibodies produced as described below, can also be used to isolate DNA sequences that encode human Zntr2 genes from cDNA libraries.
  • the antibodies can be used to screen ⁇ gtl l expression libraries, or the antibodies can be used for immunoscreening following hybrid selection and translation (see, for example, Ausubel (1995) at pages 6-12 to 6-16; Margolis et al, "Screening ⁇ expression libraries with antibody and protein probes," in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), pages 1-14 (Oxford University Press 1995)).
  • a Zntr2 gene can be obtained by synthesizing nucleic acid molecules using mutually priming long oligonucleotides and the nucleotide sequences described herein (see, for example, Ausubel (1995) at pages 8-8 to 8-9).
  • the nucleic acid molecules of the present invention can also be synthesized with "gene machines” using protocols such as the phosphoramidite method. If chemically-synthesized double stranded DNA is required for an application such as the synthesis of a gene or a gene fragment, then each complementary strand is made separately.
  • the production of short genes 60 to 80 base pairs is technically straightforward and can be accomplished by synthesizing the complementary strands and then annealing them. For the production of longer genes (>300 base pairs), however, special strategies may be required, because the coupling efficiency of each cycle during chemical DNA synthesis is seldom 100%).
  • Zntr2 cDNA or Zntr2 genomic fragment can be determined using standard methods.
  • Zntr2 polynucleotide sequences disclosed herein can also be used as probes or primers to clone 5' non-coding regions of a Zntr2 gene.
  • Promoter elements from a Zntr2 gene can be used to direct the expression of heterologous genes in, for example, peripheral blood leukocytes of transgenic animals or patients undergoing gene therapy.
  • the identification of genomic fragments containing a Zntr2 promoter or regulatory element can be achieved using well- established techniques, such as deletion analysis (see, generally, Ausubel (1995)).
  • Cloning of 5' flanking sequences also facilitates production of Zntr2 proteins by "gene activation," according to the general methods disclosed in U.S. Patent No. 5,641,670. Briefly, expression of an endogenous Zntr2 gene in a cell is altered by introducing into the Zntr2 locus a DNA construct comprising at least a targeting sequence, a regulatory sequence, an exon, and an unpaired splice donor site.
  • the targeting sequence is a Zntr2 5' non-coding sequence that permits homologous recombination of the construct with the endogenous Zntr2 locus, whereby the sequences within the construct become operably linked with the endogenous Zntr2 coding sequence.
  • an endogenous Zntr2 promoter can be replaced or supplemented with other regulatory sequences to provide enhanced, tissue-specific, or otherwise regulated expression.
  • SEQ ID NO:3 is a degenerate nucleotide sequence that encompasses all nucleic acid molecules that encode the Zntr2 polypeptides of SEQ ID NO:2. Those skilled in the art will recognize that the degenerate sequence of SEQ ID NO:3.
  • NO:3 also provides all RNA sequences encoding SEQ ID NO:2, by substituting U for
  • the present invention contemplates Zntr2 polypeptide-encoding nucleic acid molecules comprising nucleotide 103 to nucleotide 1908 of SEQ ID NO:l, and their
  • RNA equivalents Table 1 sets forth the one-letter codes used within SEQ ID NO: 3 to denote degenerate nucleotide positions. "Resolutions” are the nucleotides denoted by a code letter. "Complement” indicates the code for the complementary nucleotide(s). For example, the code Y denotes either C or T, and its complement R denotes A or G, A being complementary to T, and G being complementary to C.
  • degenerate codons used in SEQ ID NO:3, encompassing all possible codons for a given amino acid, are set forth in Table 2.
  • degenerate codon representative of all possible codons encoding an amino acid.
  • WSN can, in some circumstances, encode arginine
  • MGN can, in some circumstances, encode serine
  • some polynucleotides encompassed by the degenerate sequence may encode variant amino acid sequences, but one of ordinary skill in the art can easily identify such variant sequences by reference to the amino acid sequence of SEQ ID NO:2. Variant sequences can be readily tested for functionality as described herein. Different species can exhibit "preferential codon usage.” In general, see, see,
  • preferential codon usage or “preferential codons” is a term of art referring to protein translation codons that are most frequently used in cells of a certain species, thus favoring one or a few representatives of the possible codons encoding each amino acid (See Table 2).
  • the amino acid threonine (Thr) may be encoded by ACA, ACC, ACG, or ACT, but in mammalian cells ACC is the most commonly used codon; in other species, for example, insect cells, yeast, viruses or bacteria, different Thr codons may be preferential.
  • Preferential codons for a particular species can be introduced into the polynucleotides of the present invention by a variety of methods known in the art.
  • preferential codon sequences into recombinant DNA can, for example, enhance production of the protein by making protein translation more efficient within a particular cell type or species. Therefore, the degenerate codon sequence disclosed in SEQ ID NO: 3 serves as a template for optimizing expression of polynucleotides in various cell types and species commonly used in the art and disclosed herein. Sequences containing preferential codons can be tested and optimized for expression in various species, and tested for functionality as disclosed herein.
  • the present invention further provides variant polypeptides and nucleic acid molecules that represent counterparts from other species (orthologs). These species include, but are not limited to mammalian, avian, amphibian, reptile, fish, insect and other vertebrate and invertebrate species. Of particular interest are Zntr2 polypeptides from other mammalian species, including murine, porcine, ovine, bovine, canine, feline, equine, and other primate polypeptides. Orthologs of human Zntr2 can be cloned using information and compositions provided by the present invention in combination with conventional cloning techniques.
  • a cDNA can be cloned using mRNA obtained from a tissue or cell type that expresses Zntr2 as disclosed herein. Suitable sources of mRNA can be identified by probing northern blots with probes designed from the sequences disclosed herein. A library is then prepared from mRNA of a positive tissue or cell line. A Zntr2-encoding cDNA molecule can then be isolated by a variety of methods, such as by probing with a complete or partial human cDNA or with one or more sets of degenerate probes based on the disclosed sequences. A cDNA can also be cloned using the polymerase chain reaction with primers designed from the representative human Zntr2 sequences disclosed herein. Within an additional method, the cDNA library can be used to transform or transfect host cells, and expression of the cDNA of interest can be detected with an antibody to Zntr2 polypeptide. Similar techniques can also be applied to the isolation of genomic clones.
  • SEQ ID NO:l represents a single allele of human Zntr2, and that allelic variation and alternative splicing are expected to occur. Allelic variants of this sequence can be cloned by probing cDNA or genomic libraries from different individuals according to standard procedures. Allelic variants of the nucleotide sequence shown in SEQ ID NO:l, including those containing silent mutations and those in which mutations result in amino acid sequence changes, are within the scope of the present invention, as are proteins which are allelic variants of SEQ ID NO:2.
  • cDNA molecules generated from alternatively spliced mRNAs, which retain the properties of the Zntr2 polypeptide are included within the scope of the present invention, as are polypeptides encoded by such cDNAs and mRNAs.
  • Allelic variants and splice variants of these sequences can be cloned by probing cDNA or genomic libraries from different individuals or tissues according to standard procedures known in the art.
  • the isolated nucleic acid molecules can hybridize under stringent conditions to nucleic acid molecules comprising nucleotide sequences disclosed herein.
  • such nucleic acid molecules can hybridize under stringent conditions to nucleic acid molecules comprising the nucleotide sequence of SEQ ID NO:l, to nucleic acid molecules having the nucleotide sequence of nucleotides 103 to 1908 of SEQ ID NO:l, or to nucleic acid molecules having a nucleotide sequence complementary to SEQ ID NO:l or to nucleotides 103 to 1908 of SEQ ID NO:l .
  • stringent conditions are selected to be about 5°C lower than the thermal melting point (T m ) for the specific sequence at a defined ionic strength and pH.
  • T m is the temperature (under defined ionic strength and pH) at which 50%> of the target sequence hybridizes to a perfectly matched probe.
  • a pair of nucleic acid molecules can hybridize if the nucleotide sequences have some degree of complementarity.
  • Hybrids can tolerate mismatched base pairs in the double helix, but the stability of the hybrid is influenced by the degree of mismatch.
  • the T m of the mismatched hybrid decreases by 1°C for every 1-1.5% base pair mismatch. Varying the stringency of the hybridization conditions allows control over the degree of mismatch that will be present in the hybrid.
  • the degree of stringency increases as the hybridization temperature increases and the ionic strength of the hybridization buffer decreases.
  • Stringent hybridization conditions encompass temperatures of about 5-25°C below the T m of the hybrid and a hybridization buffer having up to 1 M Na + .
  • the above conditions are meant to serve as a guide and it is well within the abilities of one skilled in the art to adapt these conditions for use with a particular polypeptide hybrid.
  • the T m for a specific target sequence is the temperature (under defined conditions) at which 50% of the target sequence will hybridize to a perfectly matched probe sequence.
  • Those conditions which influence the T m include, the size and base pair content of the polynucleotide probe, the ionic strength of the hybridization solution, and the presence of destabilizing agents in the hybridization solution.
  • Sequence analysis software such as OLIGO 6.0 (LSR; Long Lake, MN) and Primer Premier 4.0 (Premier Biosoft International; Palo Alto, CA), as well as sites on the Internet, are available tools for analyzing a given sequence and calculating T m based on user defined criteria. Such programs can also analyze a given sequence under defined conditions and identify suitable probe sequences. Typically, hybridization of longer polynucleotide sequences, >50 base pairs, is performed at temperatures of about 20-25°C below the calculated T m . For smaller probes, ⁇ 50 base pairs, hybridization is typically carried out at the T m or 5-10°C below. This allows for the maximum rate of hybridization for DNA- DNA and DNA-RNA hybrids.
  • the length of the polynucleotide sequence influences the rate and stability of hybrid formation. Smaller probe sequences, ⁇ 50 base pairs, reach equilibrium with complementary sequences rapidly, but may form less stable hybrids. Incubation times of anywhere from minutes to hours can be used to achieve hybrid formation. Longer probe sequences come to equilibrium more slowly, but form more stable complexes even at lower temperatures. Incubations are allowed to proceed overnight or longer. Generally, incubations are carried out for a period equal to three times the calculated Cot time. Cot time, the time it takes for the polynucleotide sequences to reassociate, can be calculated for a particular sequence by methods known in the art.
  • the base pair composition of polynucleotide sequence will effect the thermal stability of the hybrid complex, thereby influencing the choice of hybridization temperature and the ionic strength of the hybridization buffer.
  • A-T pairs are less stable than G-C pairs in aqueous solutions containing sodium chloride. Therefore, the higher the G-C content, the more stable the hybrid. Even distribution of G and C residues within the sequence also contribute positively to hybrid stability.
  • the base pair composition can be manipulated to alter the T m of a given sequence.
  • 5-methyldeoxycytidine can be substituted for deoxycytidine and 5-bromodeoxuridine can be substituted for thymidine to increase the T m whereas 7-deazz-2'- deoxyguanosine can be substituted for guanosine to reduce dependence on T m .
  • the ionic concentration of the hybridization buffer also affects the stability of the hybrid.
  • Hybridization buffers generally contain blocking agents such as Denhardt's solution (Sigma Chemical Co., St.
  • hybridization buffers contain from between 10 mM - 1 M Na + .
  • destabilizing or denaturing agents such as formamide, tetralkylammonium salts, guanidinium cations or thiocyanate cations to the hybridization solution will alter the T m of a hybrid.
  • formamide is used at a concentration of up to 50% to allow incubations to be carried out at more convenient and lower temperatures.
  • Formamide also acts to reduce non-specific background when using RNA probes.
  • a nucleic acid molecule encoding a variant Zntr2 polypeptide can be hybridized with a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l (or its complement) at 42°C overnight in a solution comprising 50% formamide, 5xSSC (lxSSC: 0.15 M sodium chloride and 15 mM sodium citrate), 50 mM sodium phosphate (pH 7.6), 5x Denhardt's solution (lOOx Denhardt's solution: 2%> (w/v) Ficoll 400, 2% (w/v) polyvinylpyrrolidone, and 2% (w/v) bovine serum albumin), 10% dextran sulfate, and 20 ⁇ g/ml denatured, sheared salmon sperm DNA.
  • 5xSSC lxSSC: 0.15 M sodium chloride and 15 mM sodium citrate
  • 50 mM sodium phosphate pH 7.6
  • 5x Denhardt's solution l
  • hybridization mixture can be incubated at a higher temperature, such as about 65°C, in a solution that does not contain formamide.
  • a higher temperature such as about 65°C
  • premixed hybridization solutions are available (e.g., EXPRESSHYB Hybridization Solution from CLONTECH Laboratories, Inc.), and hybridization can be performed according to the manufacturer's instructions.
  • nucleic acid molecules can be washed to remove non-hybridized nucleic acid molecules under stringent conditions, or under highly stringent conditions.
  • Typical stringent washing conditions include washing in a solution of 0.5x - 2x SSC with 0.1% sodium dodecyl sulfate (SDS) at 55 - 65°C.
  • nucleic acid molecules encoding a variant Zntr2 polypeptide remain hybridized, following a wash under stringent washing conditions, with a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l (or its complement), in which the wash stringency is equivalent to 0.5x - 2x SSC with 0.1% SDS at 55 - 65°C, including 0.5x SSC with 0.1% SDS at 55°C, or 2xSSC with 0.1% SDS at 65°C.
  • wash stringency is equivalent to 0.5x - 2x SSC with 0.1% SDS at 55 - 65°C, including 0.5x SSC with 0.1% SDS at 55°C, or 2xSSC with 0.1% SDS at 65°C.
  • SSPE for SSC in the wash solution.
  • Typical highly stringent washing conditions include washing in a solution of O.lx - 0.2x SSC with 0.1% sodium dodecyl sulfate (SDS) at 50 - 65°C.
  • SDS sodium dodecyl sulfate
  • nucleic acid molecules encoding a variant Zntr2 polypeptide remain hybridized, following a wash under highly stringent washing conditions, with a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l (or its complement), in which the wash stringency is equivalent to O.lx - 0.2x SSC with 0.1% SDS at 50 - 65°C, including O.lx SSC with 0.1% SDS at 50°C, or 0.2xSSC with 0.1% SDS at 65°C.
  • the present invention also provides isolated Zntr2 polypeptides that have a substantially similar sequence identity to the polypeptides of SEQ ID NO:2, or their orthologs.
  • substantially similar sequence identity is used herein to denote polypeptides having 70%, 80%>, 90%>, 95% or greater than 95% sequence identity to the sequences shown in SEQ ID NO:2, or their orthologs.
  • the present invention also contemplates Zntr2 variant nucleic acid molecules that can be identified using two criteria: a determination of the similarity between the encoded polypeptide with the amino acid sequence of SEQ ID NO:2, and a hybridization assay, as described above.
  • Such Zntr2 variants include nucleic acid molecules (1) that remain hybridized, following a wash under stringent washing conditions, with a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l (or its complement), in which the wash stringency is equivalent to 0.5x - 2x SSC with 0.1% SDS at 55 - 65°C, and (2) that encode a polypeptide having 70%, 80%, 90%, 95% or greater than 95% sequence identity to the amino acid sequence of SEQ ID NO:2.
  • Zntr2 variants can be characterized as nucleic acid molecules (1) that remain hybridized, following a wash under highly stringent washing conditions, with a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l (or its complement), in which the wash stringency is equivalent to O.lx - 0.2x SSC with 0.1% SDS at 50 - 65°C, and (2) that encode a polypeptide having 70%, 80%, 90%, 95% or greater than 95% sequence identity to the amino acid sequence of SEQ ID NO:2.
  • Percent sequence identity is determined by conventional methods. See, for example, Altschul et al, Bull. Math. Bio. 48:603 (1986), and Henikoff and Henikoff, Proc. Natl Acad. Sci. USA #9:10915 (1992). Briefly, two amino acid sequences are aligned to optimize the alignment scores using a gap opening penalty of 10, a gap extension penalty of 1 , and the "BLOSUM62" scoring matrix of Henikoff and Henikoff (ibid.) as shown in Table 3 (amino acids are indicated by the standard one- letter codes). The percent identity is then calculated as: ([Total number of identical matches]/ [length of the longer sequence plus the number of gaps introduced into the longer sequence in order to align the two sequences])(100).
  • the "FASTA" similarity search algorithm of Pearson and Lipman is a suitable protein alignment method for examining the level of identity shared by an amino acid sequence disclosed herein and the amino acid sequence of a putative Zntr2 variant.
  • the FASTA algorithm is described by Pearson and Lipman, Proc. Nat'l Acad. Sci. USA 55:2444 (1988), and by Pearson, Meth. Enzymol 183:63 (1990).
  • the ten regions with the highest density of identities are then rescored by comparing the similarity of all paired amino acids using an amino acid substitution matrix, and the ends of the regions are "trimmed" to include only those residues that contribute to the highest score.
  • the trimmed initial regions are examined to determine whether the regions can be joined to form an approximate alignment with gaps.
  • the highest scoring regions of the two amino acid sequences are aligned using a modification of the Needleman- Wunsch- Sellers algorithm (Needleman and Wunsch, J Mol. Biol 48:444 (1970); Sellers, SIAM J. Appl Math. 26:787 (1974)), which allows for amino acid insertions and deletions.
  • FASTA can also be used to determine the sequence identity of nucleic acid molecules using a ratio as disclosed above.
  • the ktup value can range between one to six, preferably from three to six, most preferably three, with other parameters set as described above.
  • the present invention includes nucleic acid molecules that encode a polypeptide having a conservative amino acid change, compared with the amino acid sequence of SEQ ID NO:2. That is, variants can be obtained that contain one or more amino acid substitutions of SEQ ID NO:2, in which an alkyl amino acid is substituted for an alkyl amino acid in a Zntr2 amino acid sequence, an aromatic amino acid is substituted for an aromatic amino acid in a Zntr2 amino acid sequence, a sulfur- containing amino acid is substituted for a sulfur-containing amino acid in a Zntr2 amino acid sequence, a hydroxy-containing amino acid is substituted for a hydroxy- containing amino acid in a Zntr2 amino acid sequence, an acidic amino acid is substituted for an acidic amino acid in a Zntr2 amino acid sequence, a basic amino acid is substituted for a basic amino acid in a Zntr2 amino acid sequence, or a dibasic monocarboxylic amino acid is substituted for a dibasic
  • a “conservative amino acid substitution” is illustrated by a substitution among amino acids within each of the following groups: (1) glycine, alanine, valine, leucine, and isoleucine, (2) phenylalanine, tyrosine, and tryptophan, (3) serine and threonine, (4) aspartate and glutamate, (5) glutamine and asparagine, and (6) lysine, arginine and histidine.
  • the BLOSUM62 table is an amino acid substitution matrix derived from about 2,000 local multiple alignments of protein sequence segments, representing highly conserved regions of more than 500 groups of related proteins (Henikoff and Henikoff, Proc. Nat'l Acad. Sci. USA 59:10915 (1992)). Accordingly, the BLOSUM62 substitution frequencies can be used to define conservative amino acid substitutions that may be introduced into the amino acid sequences of the present invention. Although it is possible to design amino acid substitutions based solely upon chemical properties (as discussed above), the language "conservative amino acid substitution” can refer to a substitution represented by a BLOSUM62 value of greater than -1. For example, an amino acid substitution is conservative if the substitution is characterized by a BLOSUM62 value of 0, 1, 2, or 3.
  • certain conservative amino acid substitutions are characterized by a BLOSUM62 value of at least 1 (e.g., 1, 2 or 3), while other conservative amino acid substitutions are characterized by a BLOSUM62 value of at least 2 (e.g., 2 or 3).
  • Conservative amino acid changes in a Zntr2 gene can be introduced by substituting nucleotides for the nucleotides recited in SEQ ID NO:l .
  • Such "conservative amino acid” variants can be obtained, for example, by oligonucleotide- directed mutagenesis, linker-scanning mutagenesis, mutagenesis using the polymerase chain reaction, and the like (see Ausubel (1995) at pages 8-10 to 8-22; and McPherson (ed.), Directed Mutagenesis: A Practical Approach (IRL Press 1991)).
  • a variant Zntr2 polypeptide can be identified by the ability to specifically bind anti- Zntr2 antibodies. Particular variants of Zntr2 are characterized by having greater than
  • amino acid sequence e.g., SEQ ID NO:2
  • variation in amino acid sequence is due to one or more conservative amino acid substitutions.
  • the proteins of the present invention can also comprise non-naturally occurring amino acid residues.
  • Non-naturally occurring amino acids include, without limitation, tr ⁇ ra-3-methylproline, 2,4-methanoproline, cw-4-hydroxyproline, trans-4- hydroxyproline, N-methylglycine, ⁇ //o-threonine, methylthreonine, hydroxyethylcysteine, hydroxyethylhomocysteine, nitroglutamine, homoglutamine, pipecolic acid, thiazolidine carboxylic acid, dehydroproline, 3- and 4-methylproline, 3,3-dimethylproline, tert-leucine, norvaline, 2-azaphenylalanine, 3-azaphenylalanine, 4-azaphenylalanine, and 4-fluorophenylalanine.
  • E. coli cells are cultured in the absence of a natural amino acid that is to be replaced (e.g., phenylalanine) and in the presence of the desired non-naturally occurring amino acid(s) (e.g., 2-azaphenylalanine, 3-azaphenylalanine, 4-azaphenylalanine, or 4- fluorophenylalanine).
  • a natural amino acid that is to be replaced e.g., phenylalanine
  • desired non-naturally occurring amino acid(s) e.g., 2-azaphenylalanine, 3-azaphenylalanine, 4-azaphenylalanine, or 4- fluorophenylalanine.
  • non-naturally occurring amino acid is incorporated into the protein in place of its natural counterpart. See, Koide et al, Biochem. 33:7470 (1994). Naturally occurring amino acid residues can be converted to non-naturally occurring species by in vitro chemical modification. Chemical modification can be combined with site-directed mutagenesis to further expand the range of substitutions (Wynn and Richards, Protein Sci. 2:395 (1993)).
  • Non-conservative amino acids, amino acids that are not encoded by the genetic code, non-naturally occurring amino acids, and unnatural amino acids may be substituted for Zntr2 amino acid residues.
  • Essential amino acids in the polypeptides of the present invention can be identified according to procedures known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (Cunningham and Wells, Science 244: 081 (1989), Bass et al, Proc. Nat'lAcad. Sci. USA 55:4498 (1991), Coombs and Corey, "Site-Directed Mutagenesis and Protein Engineering," in Proteins: Analysis and Design, Angeletti (ed.), pages 259-311 (Academic Press, Inc. 1998)).
  • Zntr2 ligand binding domains can also be determined by physical analysis of structure, as determined by such techniques as nuclear magnetic resonance, crystallography, electron diffraction or photoaffinity labeling, in conjunction with mutation of putative contact site amino acids. See, for example, de Vos et al, Science 255:306 (1992), Smith et al, J Mol Biol 224:899 (1992), and Wlodaver et al, FEBS Lett. 309:59 (1992). Moreover, Zntr2 labeled with biotin or FITC can be used for expression cloning of Zntr2 ligands.
  • variants of the disclosed Zntr2 nucleotide and polypeptide sequences can also be generated through DNA shuffling as disclosed by Stemmer, Nature 370:389 (1994), Stemmer, Proc. Nat 'I Acad. Sci. USA 97:10747 (1994), and international publication No. WO 97/20078. Briefly, variant DNAs are generated by in vitro homologous recombination by random fragmentation of a parent DNA followed by reassembly using PCR, resulting in randomly introduced point mutations. This technique can be modified by using a family of parent DNAs, such as allelic variants or DNAs from different species, to introduce additional variability into the process.
  • Mutagenesis methods as disclosed herein can be combined with high- throughput, automated screening methods to detect activity of cloned, mutagenized polypeptides in host cells.
  • Mutagenized DNA molecules that encode biologically active polypeptides, or polypeptides that bind with anti-Zntr2 antibodies can be recovered from the host cells and rapidly sequenced using modern equipment. These methods allow the rapid determination of the importance of individual amino acid residues in a polypeptide of interest, and can be applied to polypeptides of unknown structure.
  • the present invention also includes "functional fragments" of Zntr2 polypeptides and nucleic acid molecules encoding such functional fragments.
  • Routine deletion analyses of nucleic acid molecules can be performed to obtain functional fragments of a nucleic acid molecule that encodes a Zntr2 polypeptide.
  • DNA molecules having the nucleotide sequence of SEQ ID NO:l can be digested with Bal 1 nuclease to obtain a series of nested deletions. The fragments are then inserted into expression vectors in proper reading frame, and the expressed polypeptides are isolated and tested for the ability to bind anti-Zntr2 antibodies.
  • exonuclease digestion is to use oligonucleotide-directed mutagenesis to introduce deletions or stop codons to specify production of a desired fragment.
  • particular fragments of a Zntr2 gene can be synthesized using the polymerase chain reaction.
  • Methods for identifying functional domains are well-known to those of skill in the art. For example, studies on the truncation at either or both termini of interferons have been summarized by Horisberger and Di Marco, Pharmac. Ther. 66:507 (1995).
  • standard techniques for functional analysis of proteins are described by, for example, Treuter et al, Molec. Gen. Genet.
  • Illustrative functional fragments of Zntr2 include a signal sequence (amino acid residues 1 to 30 or amino acid residues 9 to 30 of SEQ ID NO:2), an extracellular domain (31 to 507 of SEQ ID NO:2), a transmembrane domain (amino acid residues 508 to 533 of SEQ ID NO:2), and an intracellular domain (amino acid residues 534 to 602 of SEQ ID NO:2).
  • a polypeptide that comprises the Zntr2 extracellular domain, but lacks a transmembrane domain, is considered to be a "soluble Zntr2 receptor."
  • Examples of soluble Zntr2 receptors include an extracellular domain (e.g.
  • amino acid residues 31 to 507 of SEQ ID NO:2) amino acid residues 31 to 507 of SEQ ID NO:2
  • a signal sequence with an extracellular domain e.g., amino acid residues 1 to 507, or 9 to 507, of SEQ ID NO:2
  • an extracellular domain with an intracellular domain e.g., amino acid residues 31-507 and amino acid residues 534 to 602 of SEQ ID NO:2
  • a signal sequence with an extracellular domain and an intracellular domain e.g., amino acid residues 1 to 507 (or, 9 to 507) and amino acid residues 534 to 602 of SEQ ID NO:2), and the like.
  • the transmembrane domain can be used to construct membrane bound fusion proteins.
  • Such fusion proteins can comprise a soluble polypeptide moiety and a Zntr2 transmembrane domain.
  • the present invention also contemplates functional fragments of a Zntr2 gene that has amino acid changes, compared with the amino acid sequence of SEQ ID NO:2.
  • a variant Zntr2 gene can be identified on the basis of structure by determining the level of identity with nucleotide and amino acid sequences of SEQ ID NOs:l and 2, as discussed above.
  • An alternative approach to identifying a variant gene on the basis of structure is to determine whether a nucleic acid molecule encoding a potential variant Zntr2 gene can hybridize to a nucleic acid molecule having the nucleotide sequence of SEQ ID NO:l, as discussed above.
  • the present invention also provides polypeptide fragments or peptides comprising an epitope-bearing portion of a Zntr2 polypeptide described herein.
  • Such fragments or peptides may comprise an "immunogenic epitope," which is a part of a protein that elicits an antibody response when the entire protein is used as an immunogen.
  • Immunogenic epitope-bearing peptides can be identified using standard methods (see, for example, Geysen et al, Proc. Nat'l Acad. Sci. USA 81:3998 (1983)).
  • polypeptide fragments or peptides may comprise an
  • antigenic epitope which is a region of a protein molecule to which an antibody can specifically bind. Certain epitopes consist of a linear or contiguous stretch of amino acids, and the antigenicity of such an epitope is not disrupted by denaturing agents. It is known in the art that relatively short synthetic peptides that can mimic epitopes of a protein can be used to stimulate the production of antibodies against the protein (see, for example, Sutcliffe et al, Science 219:660 (1983)). Accordingly, antigenic epitope- bearing peptides and polypeptides of the present invention are useful to raise antibodies that bind with the polypeptides described herein.
  • Antigenic epitope-bearing peptides and polypeptides can contain at least four to ten amino acids, at least ten to fifteen amino acids, or about 15 to about 30 amino acids of SEQ ID NO:2.
  • Such epitope-bearing peptides and polypeptides can be produced by fragmenting a Zntr2 polypeptide, or by chemical peptide synthesis, as described herein.
  • epitopes can be selected by phage display of random peptide libraries (see, for example, Lane and Stephen, Curr. Opin. Immunol. 5:268 (1993), and Cortese et al, Curr. Opin. Biotechnol. 7:616 (1996)).
  • the gene encodes a polypeptide that may be characterized by its ability to bind specifically to an anti-Zntr2 antibody.
  • the present invention includes a computer-readable medium encoded with a data structure that provides at least one of the following sequences: SEQ ID NO:l, SEQ ID NO:2, and SEQ ID NO:3. Suitable forms of computer-readable media include magnetic media and optically-readable media.
  • magnétique media examples include a hard or fixed drive, a random access memory (RAM) chip, a floppy disk, digital linear tape (DLT), a disk cache, and a ZIP disk.
  • Optically readable media are exemplified by compact discs (e.g., CD-read only memory (ROM), CD-rewritable (RW), and CD-recordable), and digital versatile/video discs (DVD) (e.g., DVD-ROM, DVD-RAM, and DVD+RW).
  • compact discs e.g., CD-read only memory (ROM), CD-rewritable (RW), and CD-recordable
  • DVD digital versatile/video discs
  • Fusion proteins of Zntr2 can be used to express Zntr2 in a recombinant host, and to isolate expressed Zntr2. As described below, particular Zntr2 fusion proteins also have uses in diagnosis and therapy.
  • fusion protein comprises a peptide that guides a Zntr2 polypeptide from a recombinant host cell.
  • a secretory signal sequence also known as a signal peptide, a leader sequence, prepro sequence or pre sequence
  • the secretory signal sequence may be derived from Zntr2
  • a suitable signal sequence may also be derived from another secreted protein or synthesized de novo.
  • the secretory signal sequence is operably linked to a Zntr2- encoding sequence such that the two sequences are joined in the correct reading frame and positioned to direct the newly synthesized polypeptide into the secretory pathway of the host cell.
  • Secretory signal sequences are commonly positioned 5' to the nucleotide sequence encoding the polypeptide of interest, although certain secretory signal sequences may be positioned elsewhere in the nucleotide sequence of interest (see, e.g., Welch et al, U.S. Patent No. 5,037,743; Holland et al, U.S. Patent No. 5,143,830).
  • yeast signal sequence is preferred for expression in yeast cells.
  • suitable yeast signal sequences are those derived from yeast mating phermone ⁇ -factor (encoded by the MFal gene), invertase (encoded by the SUC2 gene), or acid phosphatase (encoded by the PH05 gene). See, for example, Romanos et al. , "Expression of Cloned Genes in Yeast," in DNA Cloning 2: A Practical Approach, 2 nd Edition, Glover and Hames (eds.), pages 123-167 (Oxford University Press 1995).
  • Zntr2 can be expressed as a fusion protein comprising a glutathione S-transferase polypeptide.
  • Glutathione S-transferase fusion proteins are typically soluble, and easily purifiable from E. coli lysates on immobilized glutathione columns.
  • a Zntr2 fusion protein comprising a maltose binding protein polypeptide can be isolated with an amylose resin column, while a fusion protein comprising the C-terminal end of a truncated Protein A gene can be purified using IgG-Sepharose.
  • Established techniques for expressing a heterologous polypeptide as a fusion protein in a bacterial cell are described, for example, by Williams et al. , "Expression of Foreign Proteins in E. coli Using Plasmid Vectors and Purification of Specific Polyclonal Antibodies," in DNA Cloning 2: A Practical Approach, 2 nd Edition, Glover and Hames (Eds.), pages 15-58 (Oxford University Press 1995).
  • the PINPOINT Xa protein purification system provides a method for isolating a fusion protein comprising a polypeptide that becomes biotinylated during expression with a resin that comprises avidin.
  • Peptide tags that are useful for isolating heterologous polypeptides expressed by either prokaryotic or eukaryotic cells include polyHistidine tags (which have an affinity for nickel-chelating resin), c-myc tags, calmodulin binding protein (isolated with calmodulin affinity chromatography), substance P, the RYIRS tag (which binds with anti-RYIRS antibodies), the Glu-Glu tag, and the FLAG tag (which binds with anti-FLAG antibodies). See, for example, Luo et al, Arch. Biochem. Biophys. 329:215 (1996), Morganti et al, Biotechnol. Appl. Biochem. 23:67 (1996), and Zheng et al, Gene 186:55 (1997). Nucleic acid molecules encoding such peptide tags are available, for example, from Sigma- Aldrich Corporation (St. Louis, MO).
  • the present invention also contemplates that the use of the secretory signal sequence contained in the Zntr2 polypeptides of the present invention to direct other polypeptides into the secretory pathway.
  • a signal fusion polypeptide can be made wherein a secretory signal sequence derived from amino acid residues 1 to 30, or 9 to 30, of SEQ ID NO:2 is operably linked to another polypeptide using methods known in the art and disclosed herein.
  • the secretory signal sequence contained in the fusion polypeptides of the present invention is preferably fused amino-terminally to an additional peptide to direct the additional peptide into the secretory pathway.
  • Such constructs have numerous applications known in the art.
  • these novel secretory signal sequence fusion constructs can direct the secretion of an active component of a normally non-secreted protein, such as a receptor.
  • a normally non-secreted protein such as a receptor.
  • Such fusions may be used in a transgenic animal or in a cultured recombinant host to direct peptides through the secretory pathway.
  • exemplary polypeptides include pharmaceutically active molecules such as Factor Vila, proinsulin, insulin, follicle stimulating hormone, tissue type plasminogen activator, tumor necrosis factor, interleukins (e.g., interleukin-1 (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, and IL-15), colony stimulating factors (e.g., granulocyte-colony stimulating factor (G-CSF) and granulocyte macrophage-colony stimulating factor (GM-CSF)), interferons (e.g., interferons- ⁇ , - ⁇ , - ⁇ , - ⁇ , and - ⁇ ), the stem cell growth factor designated "SI factor,” erythropoietin, and thrombopoietin.
  • interleukins e.g., interleukin-1 (IL
  • the Zntr2 secretory signal sequence contained in the fusion polypeptides of the present invention is preferably fused amino-terminally to an additional peptide to direct the additional peptide into the secretory pathway. Fusion proteins comprising a Zntr2 secretory signal sequence can be constructed using standard techniques.
  • fusion protein comprises a Zntr2 polypeptide and an immunoglobulin heavy chain constant region, typically an F c fragment, which contains two constant region domains and a hinge region but lacks the variable region.
  • an immunoglobulin heavy chain constant region typically an F c fragment
  • F c fragment an immunoglobulin heavy chain constant region
  • Chang et al, U.S. Patent No. 5,723,125 describe a fusion protein comprising a human interferon and a human immunoglobulin Fc fragment.
  • the C- terminal of the interferon is linked to the N-terminal of the Fc fragment by a peptide linker moiety.
  • An example of a peptide linker is a peptide comprising primarily a T cell inert sequence, which is immunologically inert.
  • An exemplary peptide linker has the amino acid sequence: GGSGG SGGGG SGGGG S (SEQ ID NO: 10).
  • a preferred Fc moiety is a human ⁇ 4 chain, which is stable in solution and has little or no complement activating activity.
  • the present invention contemplates a Zntr2 fusion protein that comprises a Zntr2 extracellular domain moiety and a human Fc fragment, wherein the C-terminus of the Zntr2 extracellular domain moiety is attached to the N-terminus of the Fc fragment via a peptide linker, such as a peptide consisting of the amino acid sequence of SEQ ID NO: 10.
  • the Zntr2 extracellular domain moiety can be a soluble Zntr2 receptor, as described above.
  • a Zntr2 fusion protein comprises an IgG sequence, a soluble Zntr2 receptor moiety covalently joined to the amino terminal end of the IgG sequence, and a signal peptide that is covalently joined to the amino terminal of the soluble Zntr2 receptor moiety, wherein the IgG sequence consists of the following elements in the following order: a hinge region, a CH 2 domain, and a CH 3 domain. Accordingly, the IgG sequence lacks a CH, domain.
  • the soluble Zntr2 receptor moiety displays a Zntr2 activity, such as the ability to bind with a Zntr2 ligand.
  • Fusion proteins comprising a soluble Zntr2 receptor moiety and an Fc moiety can be used, for example, as an in vitro assay tool.
  • the presence of a Zntr2 ligand in a biological sample can be detected using these Zntr2-antibody fusion proteins, in which the Zntr2 moiety is used to target the cognate ligand, and a macromolecule, such as Protein A or anti-Fc antibody, is used to detect the bound fusion protein-ligand complex.
  • antibody-Zntr2 fusion proteins comprising antibody variable domains, are useful as therapeutic proteins, in which the antibody moiety binds with a target antigen, such as a tumor associated antigen.
  • Fusion proteins can be prepared by methods known to those skilled in the art by preparing each component of the fusion protein and chemically conjugating them.
  • a polynucleotide encoding both components of the fusion protein in the proper reading frame can be generated using known techniques and expressed by the methods described herein.
  • part or all of a domain(s) conferring a biological function can be exchanged between Zntr2 of the present invention with the functionally equivalent domain(s) from another transmembrane protein.
  • domains include, but are not limited to, the secretory signal sequence, extracellular domain, transmembrane domain, and intracellular domain.
  • General methods for enzymatic and chemical cleavage of fusion proteins are described, for example, by Ausubel (1995) at pages 16-19 to 16-25.
  • polypeptides of the present invention can be produced in recombinant host cells following conventional techniques.
  • a nucleic acid molecule encoding the polypeptide must be operably linked to regulatory sequences that control transcriptional expression in an expression vector and then, introduced into a host cell.
  • expression vectors can include translational regulatory sequences and a marker gene which is suitable for selection of cells that carry the expression vector.
  • Expression vectors that are suitable for production of a foreign protein in eukaryotic cells typically contain (1) prokaryotic DNA elements coding for a bacterial replication origin and an antibiotic resistance marker to provide for the growth and selection of the expression vector in a bacterial host; (2) eukaryotic DNA elements that control initiation of transcription, such as a promoter; and (3) DNA elements that control the processing of transcripts, such as a transcription termination/polyadenylation sequence.
  • expression vectors can also include nucleotide sequences encoding a secretory sequence that directs the heterologous polypeptide into the secretory pathway of a host cell.
  • a Zntr2 expression vector may comprise a Zntr2 gene and a secretory sequence derived from a Zntr2 gene or another secreted gene.
  • Zntr2 proteins of the present invention may be expressed in mammalian cells.
  • suitable mammalian host cells include African green monkey kidney cells (Vero; ATCC CRL 1587), human embryonic kidney cells (293-HEK; ATCC CRL 1573), baby hamster kidney cells (BHK-21, BHK-570; ATCC CRL 8544, ATCC CRL 10314), canine kidney cells (MDCK; ATCC CCL 34), Chinese hamster ovary cells (CHO-K1 ; ATCC CCL61; CHO DG44 [Chasin et al, Som. Cell Molec. Genet.
  • rat pituitary cells GH1; ATCC CCL82
  • HeLa S3 cells ATCC CCL2.2
  • rat hepatoma cells H-4-II-E
  • SV40-transformed monkey kidney cells COS-1 ; ATCC CRL 1650
  • murine embryonic cells NIH-3T3; ATCC CRL 1658.
  • the transcriptional and translational regulatory signals may be derived from viral sources, such as adenovirus, bovine papilloma virus, simian virus, or the like, in which the regulatory signals are associated with a par- ticular gene which has a high level of expression.
  • viral sources such as adenovirus, bovine papilloma virus, simian virus, or the like, in which the regulatory signals are associated with a par- ticular gene which has a high level of expression.
  • Suitable transcriptional and translational regulatory sequences also can be obtained from mammalian genes, such as actin, collagen, myosin, and metallothionein genes.
  • Transcriptional regulatory sequences include a promoter region sufficient to direct the initiation of RNA synthesis.
  • Suitable eukaryotic promoters include the promoter of the mouse metallothionein I gene (Hamer et al, J. Molec. Appl. Genet. 1:273 (1982)), the TK promoter of Herpes virus (McKnight, Cell 31:355 (1982)), the SV40 early promoter (Benoist et al, Nature 290:304 (1981)), the Rous sarcoma virus promoter (Gorman et al, Proc. Nat'l Acad. Sci.
  • a prokaryotic promoter such as the bacteriophage T3 RNA polymerase promoter, can be used to control Zntr2 gene expression in mammalian cells if the prokaryotic promoter is regulated by a eukaryotic promoter (Zhou et al, Mol. Cell. Biol. 10:4529 (1990), and Kaufman et al, Nucl Acids Res. 19:4485 (1991)).
  • An expression vector can be introduced into host cells using a variety of standard techniques including calcium phosphate transfection, liposome-mediated transfection, microprojectile-mediated delivery, electroporation, and the like.
  • the transfected cells can be selected and propagated to provide recombinant host cells that comprise the expression vector stably integrated in the host cell genome.
  • Techniques for introducing vectors into eukaryotic cells and techniques for selecting such stable transformants using a dominant selectable marker are described, for example, by Ausubel (1995) and by Murray (ed.), Gene Transfer and Expression Protocols (Humana Press 1991).
  • one suitable selectable marker is a gene that provides resistance to the antibiotic neomycin.
  • selection is carried out in the presence of a neomycin-type drug, such as G-418 or the like.
  • Selection systems can also be used to increase the expression level of the gene of interest, a process referred to as "amplification.” Amplification is carried out by culturing transfectants in the presence of a low level of the selective agent and then increasing the amount of selective agent to select for cells that produce high levels of the products of the introduced genes.
  • a suitable amplifiable selectable marker is dihydrofolate reductase, which confers resistance to methotrexate.
  • drugs resistance genes e.g., hygromycin resistance, multi-drug resistance, puromycin acetyltransferase
  • markers that introduce an altered phenotype such as green fluorescent protein, or cell surface proteins such as CD4, CD8, Class I MHC, placental alkaline phosphatase may be used to sort transfected cells from untransfected cells by such means as FACS sorting or magnetic bead separation technology.
  • Zntr2 polypeptides can also be produced by cultured mammalian cells using a viral delivery system.
  • viruses for this purpose include adenovirus, herpesvirus, vaccinia virus and adeno-associated virus (AAV).
  • Adenovirus a double- stranded DNA virus, is currently the best studied gene transfer vector for delivery of heterologous nucleic acid (for a review, see Becker et al, Meth. Cell Biol. 43:161 (1994), and Douglas and Curiel, Science & Medicine 4:44 (1997)).
  • Advantages of the adenovirus system include the accommodation of relatively large DNA inserts, the ability to grow to high-titer, the ability to infect a broad range of mammalian cell types, and flexibility that allows use with a large number of available vectors containing different promoters.
  • By deleting portions of the adenovirus genome larger inserts (up to 7 kb) of heterologous DNA can be accommodated. These inserts can be incorporated into the viral DNA by direct ligation or by homologous recombination with a co- transfected plasmid.
  • An option is to delete the essential El gene from the viral vector, which results in the inability to replicate unless the El gene is provided by the host cell.
  • Adenovirus vector-infected human 293 cells (ATCC Nos. CRL- 1573, 45504, 45505), for example, can be grown as adherent cells or in suspension culture at relatively high cell density to produce significant amounts of protein (see Gamier et al, Cytotechnol 15: 45 (1994)).
  • Zntr2 genes may also be expressed in other higher eukaryotic cells, such as avian, fungal, insect, yeast, or plant cells.
  • the baculovirus system provides an efficient means to introduce cloned Zntr2 genes into insect cells.
  • Suitable expression vectors are based upon the Autographa californica multiple nuclear polyhedrosis virus (AcMNPV), and contain well-known promoters such as Drosophila heat shock protein (hsp) 70 promoter, Autographa californica nuclear polyhedrosis virus immediate-early gene promoter (ie-1) and the delayed early 39K promoter, baculovirus plO promoter, and the Drosophila metallothionein promoter.
  • hsp Drosophila heat shock protein
  • ie-1 Autographa californica nuclear polyhedrosis virus immediate-early gene promoter
  • baculovirus plO promoter the delayed early 39K promoter
  • baculovirus plO promoter the Drosophila metallothionein promoter.
  • a second method of making recombinant baculovirus utilizes a transposon-based system described by Luckow (Luckow, et al, J. Virol. 67:4566 (1993)
  • This system which utilizes transfer vectors, is sold in the BAC-to-BAC kit (Life Technologies, Rockville, MD).
  • This system utilizes a transfer vector, PFASTBAC (Life Technologies) containing a Tn7 transposon to move the DNA encoding the Zntr2 polypeptide into a baculovirus genome maintained in E. coli as a large plasmid called a "bacmid.” See, Hill-Perkins and Possee, J. Gen. Virol. 71:97 (1990), Bonning, et al, J. Gen. Virol. 75:1551 (1994), and Chazenbalk, and Rapoport, J. Biol. Chem. 270:1543 (1995).
  • transfer vectors can include an in-frame fusion with DNA encoding an epitope tag at the C- or N-terminus of the expressed Zntr2 polypeptide, for example, a Glu-Glu epitope tag (Grussenmeyer et al, Proc. Nat 'I Acad. Sci. 82:7952 (1985)).
  • a transfer vector containing a Zntr2 gene is transformed into E. coli, and screened for bacmids which contain an interrupted lacZ gene indicative of recombinant baculovirus.
  • the bacmid DNA containing the recombinant baculovirus genome is then isolated using common techniques.
  • the illustrative PFASTBAC vector can be modified to a considerable degree.
  • the polyhedrin promoter can be removed and substituted with the baculovirus basic protein promoter (also known as Pcor, p6.9 or MP promoter) which is expressed earlier in the baculovirus infection, and has been shown to be advantageous for expressing secreted proteins (see, for example, Hill-Perkins and Possee, J. Gen. Virol 71:971 (1990), Bonning, et al, J. Gen. Virol. 75:1551 (1994), and Chazenbalk and Rapoport, J. Biol. Chem. 270:1543 (1995).
  • a short or long version of the basic protein promoter can be used.
  • transfer vectors can be constructed which replace the native Zntr2 secretory signal sequences with secretory signal sequences derived from insect proteins.
  • a secretory signal sequence from Ecdysteroid Glucosyltransferase (EGT), honey bee Melittin (Invitrogen Corporation; Carlsbad, CA), or baculovirus gp67 (PharMingen: San Diego, CA) can be used in constructs to replace the native Zntr2 secretory signal sequence.
  • the recombinant virus or bacmid is used to transfect host cells.
  • suitable insect host cells include cell lines derived from IPLB-S -21, a Spodoptera frugiperda pupal ovarian cell line, such as S/9 (ATCC CRL 1711), S/21AE, and S 21 (Invitrogen Corporation; San Diego, CA), as well as Drosophila Schneider-2 cells, and the HIGH FIVEO cell line (Invitrogen) derived from Trichoplusia ni (U.S. Patent No. 5,300,435).
  • S/9 ATCC CRL 1711
  • S/21AE S/21AE
  • S 21 Invitrogen Corporation
  • Drosophila Schneider-2 cells Drosophila Schneider-2 cells
  • HIGH FIVEO cell line Invitrogen
  • Commercially available serum-free media can be used to grow and to maintain the cells.
  • Suitable media are Sf900 IITM (Life Technologies) or ESF 921TM (Expression Systems) for the Sf9 cells; and Ex-cellO405TM (JRH Biosciences, Lenexa, KS) or Express FiveOTM (Life Technologies) for the T. ni cells.
  • the cells are typically grown up from an inoculation density of approximately 2-5 x 10 5 cells to a density of 1-2 x 10 6 cells at which time a recombinant viral stock is added at a multiplicity of infection (MOI) of 0.1 to 10, more typically near 3.
  • MOI multiplicity of infection
  • yeast cells can also be used to express the genes described herein.
  • Yeast species of particular interest in this regard include Saccharomyces cerevisiae, Pichia pastoris, and Pichia methanolica.
  • Suitable promoters for expression in yeast include promoters from GALl (galactose), PGK (phosphogly cerate kinase), ADH (alcohol dehydrogenase), AOX1 (alcohol oxidase), ⁇ IS4 (histidinol dehydrogenase), and the like.
  • GALl galactose
  • PGK phosphogly cerate kinase
  • ADH alcohol dehydrogenase
  • AOX1 alcohol oxidase
  • ⁇ IS4 histidinol dehydrogenase
  • These vectors include Yip-based vectors, such as YIp5, YRp vectors, such as YRpl7, Y ⁇ p vectors such as Y ⁇ pl3 and YCp vectors, such as YCp 19.
  • Methods for transforming S. cerevisiae cells with exogenous DNA and producing recombinant polypeptides therefrom are disclosed by, for example, Kawasaki, U.S. Patent No. 4,599,311, Kawasaki et al, U.S. Patent No. 4,931,373, Brake, U.S. Patent No. 4,870,008, Welch et al, U.S. Patent No. 5,037,743, and Murray et al, U.S. Patent No. 4,845,075.
  • Transformed cells are selected by phenotype determined by the selectable marker, commonly drug resistance or the ability to grow in the absence of a particular nutrient (e.g., leucine).
  • a suitable vector system for use in Saccharomyces cerevisiae is the POT1 vector system disclosed by Kawasaki et al. (U.S. Patent No. 4,931,373), which allows transformed cells to be selected by growth in glucose-containing media. Additional suitable promoters and terminators for use in yeast include those from glycolytic enzyme genes (see, e.g., Kawasaki, U.S. Patent No. 4,599,311, Kingsman et al, U.S. Patent No. 4,615,974, and Bitter, U.S. Patent No.
  • Aspergillus cells may be utilized according to the methods of McKnight et al, U.S. Patent No. 4,935,349. Methods for transforming Acremonium chrysogenum are disclosed by Sumino et al, U.S. Patent No. 5,162,228. Methods for transforming Neurospora are disclosed by Lambowitz, U.S. Patent No. 4,486,533.
  • Pichia methanolica as host for the production of recombinant proteins is disclosed by Raymond, U.S. Patent No. 5,716,808, Raymond, U.S. Patent No. 5,736,383, Raymond et al, Yeast 14: -23 (1998), and in international publication Nos. WO 97/17450, WO 97/17451, WO 98/02536, and WO 98/02565.
  • DNA molecules for use in transforming P. methanolica will commonly be prepared as double-stranded, circular plasmids, which are preferably linearized prior to transformation.
  • the promoter and terminator in the plasmid be that of a P.
  • methanolica gene such as a P. methanolica alcohol utilization gene (AUGl or AUGl).
  • Other useful promoters include those of the dihydroxyacetone synthase (DHAS), formate dehydrogenase (FMD), and catalase (CAT) genes.
  • DHAS dihydroxyacetone synthase
  • FMD formate dehydrogenase
  • CAT catalase
  • a suitable selectable marker for use in Pichia methanolica is a P. methanolica ADE2 gene, which encodes phosphoribosyl-5- aminoimidazole carboxylase (AIRC; EC 4.1.1.21), and which allows ade2 host cells to grow in the absence of adenine.
  • methanolica cells can be transformed by electroporation using an exponentially decaying, pulsed electric field having a field strength of from 2.5 to 4.5 kV/cm, preferably about 3.75 kV/cm, and a time constant (t) of from 1 to 40 milliseconds, most preferably about 20 milliseconds.
  • Expression vectors can also be introduced into plant protoplasts, intact plant tissues, or isolated plant cells.
  • Methods for introducing expression vectors into plant tissue include the direct infection or co-cultivation of plant tissue with Agrobacterium tumefaciens, microprojectile-mediated delivery, DNA injection, electroporation, and the like. See, for example, Horsch et al, Science 227:1229 (1985), Klein et al, Biotechnology 10:268 (1992), and Miki et al, "Procedures for Introducing Foreign DNA into Plants," in Methods in Plant Molecular Biology and Biotechnology, Glick et al. (eds.), pages 67-88 (CRC Press, 1993).
  • Zntr2 genes can be expressed in prokaryotic host cells.
  • Suitable promoters that can be used to express Zntr2 polypeptides in a prokaryotic host are well-known to those of skill in the art and include promoters capable of recognizing the T4, T3, Sp6 and T7 polymerases, the P R and P L promoters of bacteriophage lambda, the trp, recA, heat shock, lacUV5, tac, Ipp-lacSpr, phoA, and lacZ promoters of E. coli, promoters of B.
  • subtilis the promoters of the bacteriophages of Bacillus, Streptomyces promoters, the int promoter of bacteriophage lambda, the bla promoter of pBR322, and the CAT promoter of the chloramphenicol acetyl trans- ferase gene.
  • Prokaryotic promoters have been reviewed by Glick, J Ind. Microbiol. 7:277 (1987), Watson et al, Molecular Biology of the Gene, 4th Ed. (Benjamin Cummins 1987), and by Ausubel et al. (1995/ Suitable prokaryotic hosts include E. coli and Bacillus subtilus. Suitable strains of E.
  • coli include BL21(D ⁇ 3), BL21(DE3)pLysS, BL21(DE3)pLysE, DH1, DH4I, DH5, DH5I, DH5IF, DH5IMCR, DH10B, DH10B/p3, DH11S, C600, HB101, JM101, JM105, JM109, JM110, K38, RR1, Y1088, Y1089, CSH18, ER1451, and ER1647 (see, for example, Brown (ed.), Molecular Biology Labfax (Academic Press 1991)).
  • Suitable strains of Bacillus subtilus include BR151, YB886, Mil 19, MI 120, and B170 (see, for example, Hardy, "Bacillus Cloning Methods,” in DNA Cloning: A Practical Approach, Glover (ed.) (IRL Press 1985)).
  • the polypeptide When expressing a Zntr2 polypeptide in bacteria such as E. coli, the polypeptide may be retained in the cytoplasm, typically as insoluble granules, or may be directed to the periplasmic space by a bacterial secretion sequence. In the former case, the cells are lysed, and the granules are recovered and denatured using, for example, guanidine isothiocyanate or urea. The denatured polypeptide can then be refolded and dimerized by diluting the denaturant, such as by dialysis against a solution of urea and a combination of reduced and oxidized glutathione, followed by dialysis against a buffered saline solution.
  • the denaturant such as by dialysis against a solution of urea and a combination of reduced and oxidized glutathione
  • the polypeptide can be recovered from the periplasmic space in a soluble and functional form by disrupting the cells (by, for example, sonication or osmotic shock) to release the contents of the periplasmic space and recovering the protein, thereby obviating the need for denaturation and refolding.
  • polypeptides of the present invention can be synthesized by exclusive solid phase synthesis, partial solid phase methods, fragment condensation or classical solution synthesis. These synthesis methods are well-known to those of skill in the art (see, for example, Merrifield, J. Am. Chem. Soc. 55:2149 (1963), Stewart et al, "Solid Phase Peptide Synthesis” (2nd Edition), (Pierce Chemical Co. 1984), Bayer and Rapp, Chem. Pept. Prot.
  • polypeptides and polypeptides of the present invention comprise at least six, at least nine, or at least 15 contiguous amino acid residues of SEQ ID NO:2.
  • polypeptides can comprise at least 15 contiguous amino acid residues of any of the following amino acid sequences of SEQ ID NO:2: amino acid residues 1 to 220, amino acid residues 31 to 220, amino acid residues 1 to 241, amino acid residues 31 to 241, amino acid residues 1 to 507, amino acid residues 9 to 507, amino acid residues 508 to 533, and amino acid residues 534 to 602.
  • the polypeptides comprise 20, 30, 40, 50, or more contiguous residues of these amino acid sequences.
  • polypeptides can comprise at least 30 contiguous amino acid residues of any of the following amino acid sequences of SEQ ID NO:2: amino acid residues 1 to 235, amino acid residues 31 to 235, amino acid residues 1 to 256, amino acid residues 31 to 256, amino acid residues 1 to 507, amino acid residues 9 to 507, amino acid residues 508 to 533, and amino acid residues 534 to 602.
  • Nucleic acid molecules encoding such peptides and polypeptides are useful as polymerase chain reaction primers and probes. 7. Isolation ofZntr2 Polypeptides
  • polypeptides of the present invention can be purified to at least about 80% purity, to at least about 90% purity, to at least about 95%) purity, or even greater than 95% purity with respect to contaminating macromolecules, particularly other proteins and nucleic acids, and free of infectious and pyrogenic agents.
  • the polypeptides of the present invention may also be purified to a pharmaceutically pure state, which is greater than 99.9% pure.
  • Certain preparations comprise a purified polypeptide that is substantially free of other polypeptides, particularly other polypeptides of animal origin.
  • Fractionation and/or conventional purification methods can be used to obtain preparations of Zntr2 purified from natural sources (e.g., peripheral blood leukocytes), and recombinant Zntr2 polypeptides and fusion Zntr2 polypeptides purified from recombinant host cells.
  • natural sources e.g., peripheral blood leukocytes
  • ammonium sulfate precipitation and acid or chaotrope extraction may be used for fractionation of samples.
  • Exemplary purification steps may include hydroxyapatite, size exclusion, FPLC and reverse- phase high performance liquid chromatography. Suitable chromatographic media include derivatized dextrans, agarose, cellulose, polyacrylamide, specialty silicas, and the like. PEI, DEAE, QAE and Q derivatives are preferred.
  • Exemplary chromatographic media include those media derivatized with phenyl, butyl, or octyl groups, such as Phenyl-Sepharose FF (Pharmacia), Toyopearl butyl 650 (Toso Haas, Montgomeryville, PA), Octyl-Sepharose (Pharmacia) and the like; or polyacrylic resins, such as Amberchrom CG 71 (Toso Haas) and the like.
  • Suitable solid supports include glass beads, silica-based resins, cellulosic resins, agarose beads, cross-linked agarose beads, polystyrene beads, cross-linked polyacrylamide resins and the like that are insoluble under the conditions in which they are to be used. These supports may be modified with reactive groups that allow attachment of proteins by amino groups, carboxyl groups, sulfhydryl groups, hydroxyl groups and/or carbohydrate moieties.
  • Examples of coupling chemistries include cyanogen bromide activation, N-hydroxysuccinimide activation, epoxide activation, sulfhydryl activation, hydrazide activation, and carboxyl and amino derivatives for carbodiimide coupling chemistries. These and other solid media are well known and widely used in the art, and are available from commercial suppliers. Selection of a particular method for polypeptide isolation and purification is a matter of routine design and is determined in part by the properties of the chosen support. See, for example, Affinity Chromatography: Principles & Methods (Pharmacia LKB Biotechnology 1988), and Doonan, Protein Purification Protocols (The Humana Press 1996).
  • Zntr2 isolation and purification can be devised by those of skill in the art.
  • anti-Zntr2 antibodies obtained as described below, can be used to isolate large quantities of protein by immunoaffinity purification.
  • methods for binding receptors, such as Zntr2 to ligand polypeptides bound to support media are well known in the art.
  • the polypeptides of the present invention can also be isolated by exploitation of particular properties.
  • immobilized metal ion adsorption (IMAC) chromatography can be used to purify histidine-rich proteins, including those comprising polyhistidine tags. Briefly, a gel is first charged with divalent metal ions to form a chelate (Sulkowski, Trends in Biochem. 3: (1985)). Histidine-rich proteins will be adsorbed to this matrix with differing affinities, depending upon the metal ion used, and will be eluted by competitive elution, lowering the pH, or use of strong chelating agents.
  • IMAC immobilized metal ion adsorption
  • a fusion of the polypeptide of interest and an affinity tag may be constructed to facilitate purification.
  • an affinity tag e.g., maltose- binding protein, an immunoglobulin domain
  • Zntr2 polypeptides or fragments thereof may also be prepared through chemical synthesis, as described below.
  • Zntr2 polypeptides may be monomers or multimers; glycosylated or non-glycosylated; phosphorylated or non-phosphorylated; pegylated or non-pegylated; and may or may not include an initial methionine amino acid residue.
  • the disclosed polypeptides can be used to construct Zntr2 variants. These polypeptides can be used to identify natural ligands of the Zntr2 receptor polypeptide, and analogs of the natural ligand (a "Zntr2 ligand").
  • Zntr2 ligand One type of Zntr2 ligand analog mimics the natural ligand of Zntr2 by binding with a Zntr2 polypeptide, such as a soluble Zntr2 receptor.
  • Such an analog is considered to be a Zntr2 ligand agonist if the binding of the analog with a Zntr2 polypeptide produces a response by a cell that expresses the receptor.
  • a Zntr2 ligand analog that binds with a Zntr2 polypeptide, but does not stimulate a cellular response may be a Zntr2 ligand antagonist.
  • Such an antagonist may diminish ligand or agonist activity, for example, by a competitive or non-competitive binding of the antagonist to the Zntr2 polypeptide.
  • Zntr2 ligand analog One approach to producing a Zntr2 ligand analog is to generate an antibody that binds with the extracellular domain of Zntr2. Studies have shown that an antibody that binds with an extracellular domain can act as an agonist or an antagonist of the natural ligand (see, for example, Kita et al, Biochem. Biophys. Res. Commun. 226:59 (1996), and Alia et al, J. Biol. Chem. 277:1748 (1996)). Another approach to obtaining Zntr2 ligand analogs is provided by the use of combinatorial libraries.
  • a Zntr2 ligand or an agonist can be used as a component of defined cell culture media, alone or in combination with other bioactive agents, to replace serum that is commonly used in cell culture.
  • Antagonists are also useful as research reagents for characterizing sites of interaction between a Zntr2 ligand and its receptor.
  • pharmaceutical compositions comprising Zntr2 ligand antagonists can be used to inhibit Zntr2 ligand activity.
  • Zntr2 receptor polypeptides can be used to identify and to isolate Zntr2 ligands. Fragments of Zntr2, such as a Zntr2 extracellular domain (e.g., amino acids 31 to 507 of SEQ ID NO:2) and other forms of a soluble Zntr2 receptor, are particularly useful for these methods.
  • proteins and peptides of the present invention can be immobilized on a column and used to bind ligands from a biological sample that is run over the column (Hermanson et al (eds.), Immobilized Affinity Ligand Techniques, pages 195-202 (Academic Press 1992)).
  • the activity of a Zntr2 polypeptide can be observed by a silicon-based biosensor microphysiometer, which measures the extracellular acidification rate or proton excretion associated with receptor binding and subsequent physiologic cellular responses.
  • An exemplary device is the CYTOSENSOR Microphysiometer manufactured by Molecular Devices, Sunnyvale, CA.
  • a variety of cellular responses, such as cell proliferation, ion transport, energy production, inflammatory response, regulatory and receptor activation, and the like, can be measured by this method (see, for example, McConnell et al, Science 257:1906 (1992), Pitchford et al, Meth. Enzymol 228:84 (1997), Arimilli et al, J. Immunol. Meth.
  • the microphysiometer can be used for assaying eukaryotic, prokaryotic, adherent, or non-adherent cells. By measuring extracellular acidification changes in cell media over time, the microphysiometer directly measures cellular responses to various stimuli, including agonists, ligands, or antagonists of the Zntr2 receptor.
  • the microphysiometer can be used to measure responses of a Zntr2- expressing eukaryotic cell, compared to a control eukaryotic cell that does not express Zntr2 polypeptide.
  • Suitable cells responsive to Zntr2-modulating stimuli include recombinant host cells comprising a Zntr2 expression vector, and cells that naturally express Zntr2, such as peripheral blood lymphocytes or spleen cells. Extracellular acidification provides one measure for a Zntr2-modulated cellular response.
  • this approach can be used to identify ligands, agonists, and antagonists of the Zntr2 receptor polypeptide.
  • a compound can be identified as an agonist of the Zntr2 polypeptide by providing cells that express a Zntr2 polypeptide, culturing a first portion of the cells in the absence of the test compound, culturing a second portion of the cells in the presence of the test compound, and determining whether the second portion exhibits a cellular response, in comparison with the first portion.
  • a solid phase system can be used to identify a ligand, agonist, or antagonist of the Zntr2 receptor polypeptide.
  • a Zntr2 polypeptide or Zntr2 fusion protein can be immobilized onto the surface of a receptor chip of a commercially available biosensor instrument (BIACORE, Biacore AB; Uppsala, Sweden).
  • BIACORE Biacore AB
  • the use of this instrument is disclosed, for example, by Karlsson, Immunol Methods 145:229 (1991), and Cunningham and Wells, J Mol. Biol. 234:554 (1993).
  • a Zntr2 polypeptide or fusion protein is covalently attached, using amine or sulfhydryl chemistry, to dextran fibers that are attached to gold film within a flow cell.
  • a test sample is then passed through the cell. If a ligand is present in the sample, it will bind to the immobilized polypeptide or fusion protein, causing a change in the refractive index of the medium, which is detected as a change in surface plasmon resonance of the gold film.
  • This system allows the determination of on- and off-rates, from which binding affinity can be calculated, and assessment of stoichiometry of binding. This system can also be used to examine antibody-antigen interactions, and the interactions of other complement/anti-complement pairs.
  • Zntr2 receptor binding domains can be further characterized by physical analysis of structure, as determined by such techniques as nuclear magnetic resonance, crystallography, electron diffraction or photoaffinity labeling, in conjunction with mutation of putative contact site amino acids of Zntr2 ligand agonists. See, for example, de Vos et al, Science 255:306 (1992), Smith et al, J. Mol. Biol. 224:899 (1992), and Wlodaver et al, FEBS Lett. 309:59 (1992).
  • compositions comprising a peptide or polypeptide described herein.
  • Such compositions can further comprise a carrier.
  • the carrier can be a conventional organic or inorganic carrier.
  • carriers include water, buffer solution, alcohol, propylene glycol, macrogol, sesame oil, com oil, and the like.
  • Antibodies to Zntr2 can be obtained, for example, using the product of a Zntr2 expression vector or Zntr2 isolated from a natural source as an antigen. Particularly useful anti-Zntr2 antibodies "bind specifically" with Zntr2. Antibodies are considered to be specifically binding if the antibodies exhibit at least one of the following two properties: (1) antibodies bind to Zntr2 with a threshold level of binding activity, and (2) antibodies do not significantly cross-react with polypeptides related to Zntr2.
  • Suitable antibodies include antibodies that bind with Zntr2 in particular domains, such as the Zntr2 secretory signal sequence, the Zntr2 extracellular domain, the Zntr2 transmembrane domain, and the Zntr2 intracellular domain.
  • antibodies specifically bind if they bind to a Zntr2 polypeptide, peptide or epitope with a binding affinity (K ) of 10 6 M “1 or greater, preferably 10 7 M “1 or greater, more preferably 10 8 M “1 or greater, and most preferably 10 9 M “! or greater.
  • K binding affinity
  • the binding affinity of an antibody can be readily determined by one of ordinary skill in the art, for example, by Scatchard analysis (Scatchard, Ann. NY Acad. Sci. 51:660 (1949)).
  • antibodies do not significantly cross-react with related polypeptide molecules, for example, if they detect Zntr2, but not known related polypeptides using a standard Western blot analysis.
  • anti-Zntr2 antibodies bind with Zntr2, but not with other polypeptides that contain an epidermal growth factor-like domain, such as epidermal growth factor, coagulation factors VII, IX, X, and XII, fibulin-1, and the like.
  • Anti-Zntr2 antibodies can be produced using antigenic Zntr2 epitope- bearing peptides and polypeptides.
  • Antigenic epitope-bearing peptides and polypeptides of the present invention contain a sequence of at least four, or between 15 to about 30 amino acids contained within SEQ ID NO:2.
  • peptides or polypeptides comprising a larger portion of an amino acid sequence of the invention, containing from 30 to 50 amino acids, or any length up to and including the entire amino acid sequence of a polypeptide of the invention, also are useful for inducing antibodies that bind with Zntr2. It is desirable that the amino acid sequence of the epitope-bearing peptide is selected to provide substantial solubility in aqueous solvents (i.e., the sequence includes relatively hydrophilic residues, while hydrophobic residues are preferably avoided). Moreover, amino acid sequences containing proline residues may be also be desirable for antibody production.
  • Zntr2 potential antigenic sites in Zntr2 were identified using the Jameson-Wolf method, Jameson and Wolf, CABIOS 4:181, (1988), as implemented by the PROTEAN program (version 3.14) of LASERGENE (DNASTAR; Madison, WI). Default parameters were used in this analysis.
  • amino acids 78 to 86 amino acids 78 to 86
  • amino acids 78 to 83 amino acids 79 to 84
  • amino acids 80 to 85 amino acids 81 to 86
  • amino acids 98 to 103 amino acids 131 to 138
  • amino acids 131 to 136 amino acids 131 to 136
  • amino acids 132 to 137 amino acids 133 to 138
  • amino acids 171 to 182 amino acids 171 to 176
  • amino acids 173 to 178 amino acids 173 to 178
  • antigenic peptide fragments amino acids 330 to 335 (“antigenic peptide 66"), amino acids 554 to 559 (“antigenic peptide 67”), amino acids 329 to 334 (“antigenic peptide 68”), and amino acids 539 to 544 ("antigenic peptide 69").
  • Particularly suitable fragments include antigenic peptides 61, 66, 67, 68, and 69.
  • the present invention contemplates the use of any one of antigenic peptides 1 to 69 to generate antibodies to Zntr2.
  • the present invention also contemplates polypeptides comprising at least one of antigenic peptides 1 to 69.
  • Polyclonal antibodies to recombinant Zntr2 protein or to Zntr2 isolated from natural sources can be prepared using methods well-known to those of skill in the art. See, for example, Green et al, "Production of Polyclonal Antisera,” in Immunochemical Protocols (Manson, ed.), pages 1-5 (Humana Press 1992), and Williams et al, "Expression of foreign proteins in E. coli using plasmid vectors and purification of specific polyclonal antibodies," in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), page 15 (Oxford University Press 1995).
  • the immunogenicity of a Zntr2 polypeptide can be increased through the use of an adjuvant, such as alum (aluminum hydroxide) or Freund's complete or incomplete adjuvant.
  • an adjuvant such as alum (aluminum hydroxide) or Freund's complete or incomplete adjuvant.
  • Polypeptides useful for immunization also include fusion polypeptides, such as fusions of Zntr2 or a portion thereof with an immunoglobulin polypeptide or with maltose binding protein.
  • the polypeptide immunogen may be a full-length molecule or a portion thereof.
  • polypeptide portion is "hapten-like,” such portion may be advantageously joined or linked to a macromolecular carrier (such as keyhole limpet hemocyanin (KLH), bovine serum albumin (BSA) or tetanus toxoid) for immunization.
  • a macromolecular carrier such as keyhole limpet hemocyanin (KLH), bovine serum albumin (BSA) or tetanus toxoid
  • an anti- Zntr2 antibody of the present invention may also be derived from a subhuman primate antibody.
  • General techniques for raising diagnostically and therapeutically useful antibodies in baboons may be found, for example, in Goldenberg et al, international patent publication No. WO 91/11465, and in Losman et al, Int. J. Cancer 46:3 0 (1990).
  • monoclonal anti-Zntr2 antibodies can be generated.
  • Rodent monoclonal antibodies to specific antigens may be obtained by methods known to those skilled in the art (see, for example, Kohler et al, Nature 256:495 (1975), Coligan et al. (eds.), Current Protocols in Immunology, Vol. 1, pages 2.5.1- 2.6.7 (John Wiley & Sons 1991) ["Coligan”], Picksley et al, "Production of monoclonal antibodies against proteins expressed in E. coli," in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), page 93 (Oxford University Press 1995)).
  • monoclonal antibodies can be obtained by injecting mice with a composition comprising a Zntr2 gene product, verifying the presence of antibody production by removing a serum sample, removing the spleen to obtain B- lymphocytes, fusing the B-lymphocytes with myeloma cells to produce hybridomas, cloning the hybridomas, selecting positive clones which produce antibodies to the antigen, culturing the clones that produce antibodies to the antigen, and isolating the antibodies from the hybridoma cultures.
  • an anti-Zntr2 antibody of the present invention may be derived from a human monoclonal antibody.
  • Human monoclonal antibodies are obtained from transgenic mice that have been engineered to produce specific human antibodies in response to antigenic challenge.
  • elements of the human heavy and light chain locus are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci.
  • the transgenic mice can synthesize human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas.
  • Methods for obtaining human antibodies from transgenic mice are described, for example, by Green et al, Nature Genet. 7:13 (1994), Lonberg et al, Nature 368:856 (1994), and Taylor et al, Int. Immun. 6:579 (1994).
  • Monoclonal antibodies can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography (see, for example, Coligan at pages 2.7.1-2.7.12 and pages 2.9.1-2.9.3; Baines et al, "Purification of Immunoglobulin G (IgG),” in Methods in Molecular Biology, Vol. 10, pages 79-104 (The Humana Press, Inc. 1992)).
  • antibody fragments can be obtained, for example, by proteolytic hydrolysis of the antibody.
  • Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods.
  • antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab') 2 .
  • This fragment can be further cleaved using a thiol reducing agent to produce 3.5S Fab' monovalent fragments.
  • the cleavage reaction can be performed using a blocking group for the sulfhydryl groups that result from cleavage of disulfide linkages.
  • an enzymatic cleavage using pepsin produces two monovalent Fab fragments and an Fc fragment directly.
  • These methods are described, for example, by Goldenberg, U.S. patent No. 4,331,647, Nisonoff et al, Arch Biochem. Biophys. 59:230 (1960), Porter, Biochem. J 73: 9 (1959), Edelman et al, in Methods in Enzymology Vol. 1, page 422 (Academic Press 1967), and by Coligan at pages 2.8.1-2.8.10 and 2.10.-2.10.4.
  • cleaving antibodies such as separation of heavy chains to form monovalent light-heavy chain fragments, further cleavage of fragments, or other enzymatic, chemical or genetic techniques may also be used, so long as the fragments bind to the antigen that is recognized by the intact antibody.
  • Fv fragments comprise an association of V H and V L chains.
  • This association can be noncovalent, as described by Inbar et al, Proc. Nat'l Acad. Sci. USA 69:2659 (1972).
  • the variable chains can be linked by an intermolecular disulfide bond or cross-linked by chemicals such as glutaraldehyde (see, for example, Sandhu, Crit. Rev. Biotech. 12:437 (1992)).
  • the Fv fragments may comprise V H and V L chains which are connected by a peptide linker.
  • These single-chain antigen binding proteins are prepared by constructing a structural gene comprising DNA sequences encoding the V H and V L domains which are connected by an oligonucleotide. The structural gene is inserted into an expression vector which is subsequently introduced into a host cell, such as E. coli. The recombinant host cells synthesize a single polypeptide chain with a linker peptide bridging the two V domains.
  • scFv Methods for producing scFvs are described, for example, by Whitlow et al, Methods: A Companion to Methods in Enzymology 2:97 (1991) (also see, Bird et al, Science 242:423 (1988), Ladner et al, U.S. Patent No. 4,946,778, Pack et al, Bio/Technology 77:1271 (1993), and Sandhu, supra).
  • a scFV can be obtained by exposing lymphocytes to Zntr2 polypeptide in vitro, and selecting antibody display libraries in phage or similar vectors (for instance, through use of immobilized or labeled Zntr2 protein or peptide).
  • Genes encoding polypeptides having potential Zntr2 polypeptide binding domains can be obtained by screening random peptide libraries displayed on phage (phage display) or on bacteria, such as E. coli. Nucleotide sequences encoding the polypeptides can be obtained in a number of ways, such as through random mutagenesis and random polynucleotide synthesis. These random peptide display libraries can be used to screen for peptides which interact with a known target which can be a protein or polypeptide, such as a ligand or receptor, a biological or synthetic macromolecule, or organic or inorganic substances. Techniques for creating and screening such random peptide display libraries are known in the art (Ladner et al, U.S.
  • Patent No. 5,223,409 Ladner et al, U.S. Patent No. 4,946,778, Ladner et al, U.S. Patent No. 5,403,484, Ladner et al, U.S. Patent No. 5,571,698, and Kay et al, Phage Display of Peptides and Proteins (Academic Press, Inc. 1996)) and random peptide display libraries and kits for screening such libraries are available commercially, for instance from CLONT ⁇ CH Laboratories, Inc. (Palo Alto, CA), Invitrogen Inc. (San Diego, CA), New England Biolabs, Inc. (Beverly, MA), and Pharmacia LKB Biotechnology Inc. (Piscataway, NJ). Random peptide display libraries can be screened using the Zntr2 sequences disclosed herein to identify proteins which bind to Zntr2.
  • CDR peptides (“minimal recognition units") can be obtained by constmcting genes encoding the CDR of an antibody of interest. Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody-producing cells (see, for example, Larrick et al, Methods: A Companion to Methods in Enzymology 2:106 (1991), Courtenay-Luck, "Genetic Manipulation of Monoclonal Antibodies," in Monoclonal Antibodies: Production, Engineering and Clinical Application, Ritter et al.
  • an anti-Zntr2 antibody may be derived from a "humanized" monoclonal antibody.
  • Humanized monoclonal antibodies are produced by transferring mouse complementary determining regions from heavy and light variable chains of the mouse immunoglobulin into a human variable domain. Typical residues of human antibodies are then substituted in the framework regions of the murine counterparts.
  • the use of antibody components derived from humanized monoclonal antibodies obviates potential problems associated with the immunogenicity of murine constant regions. General techniques for cloning murine immunoglobulin variable domains are described, for example, by Orlandi et al, Proc. Nat'l Acad. Sci. USA 5(5:3833 (1989).
  • Polyclonal anti-idiotype antibodies can be prepared by immunizing animals with anti-Zntr2 antibodies or antibody fragments, using standard techniques. See, for example, Green et al, "Production of Polyclonal Antisera,” in Methods In Molecular Biology: Immunochemical Protocols, Manson (ed.), pages 1-12 (Humana Press 1992). Also, see Coligan at pages 2.4.1-2.4.7.
  • monoclonal anti- idiotype antibodies can be prepared using anti-Zntr2 antibodies or antibody fragments as immunogens with the techniques, described above.
  • humanized anti-idiotype antibodies or subhuman primate anti-idiotype antibodies can be prepared using the above-described techniques.
  • Nucleic acid molecules can be used to detect the expression of a Zntr2 gene in a biological sample.
  • probe molecules include double-stranded nucleic acid molecules comprising the nucleotide sequence of SEQ ID NO: 1 , or a fragment thereof, as well as single-stranded nucleic acid molecules having the complement of the nucleotide sequence of SEQ ID NO: l, or a fragment thereof.
  • Probe molecules may be DNA, RNA, oligonucleotides, and the like.
  • Suitable probes comprise a portion of the nucleotide sequence of nucleotides 103 to 1908 of SEQ ID NO:l, or complement thereof.
  • portion refers to at least eight nucleotides to at least 20 or more nucleotides.
  • suitable probes can comprise a portion of the nucleotide sequence of nucleotides 103 to 720, or nucleotides 103 to 783, of SEQ ID NO:l .
  • Additional illustrative probes bind to certain functional domains of Zntr2 Such probes would have a nucleotide sequence, or its complement, that encodes a portion of a signal sequence, extracellular domain, transmembrane domain, or intracellular domain.
  • RNA isolated from a biological sample
  • RNA isolated from a biological sample
  • RNA detection includes northern analysis and dot/slot blot hybridization (see, for example, Ausubel (1995) at pages 4-1 to 4-27, and Wu et al. (eds.), "Analysis of Gene Expression at the RNA Level," in Methods in Gene Biotechnology, pages 225-239 (CRC Press, Inc. 1997)).
  • Nucleic acid probes can be detectably labeled with radioisotopes such as 32 P or 35 S.
  • Zntr2 RNA can be detected with a nonradioactive hybridization method (see, for example, Isaac (ed.), Protocols for Nucleic Acid Analysis by Nonradioactive Probes (Humana Press, Inc. 1993)).
  • nonradioactive detection is achieved by enzymatic conversion of chromogenic or chemiluminescent substrates.
  • Illustrative nonradioactive moieties include biotin, fluorescein, and digoxigenin.
  • Zntr2 oligonucleotide probes are also useful for in vivo diagnosis.
  • 18 F-labeled oligonucleotides can be administered to a subject and visualized by positron emission tomography (Tavitian et al, Nature Medicine 4:467 (1998)).
  • PCR polymerase chain reaction
  • Standard techniques for performing PCR are well-known (see, generally, Mathew (ed.), Protocols in Human Molecular Genetics (Humana Press, Inc. 1991), White (ed.), PCR Protocols: Current Methods and Applications (Humana Press, Inc. 1993), Cotter (ed.), Molecular Diagnosis of Cancer (Humana Press, Inc. 1996), Hanausek and Walaszek (eds.), Tumor Marker Protocols (Humana Press, Inc. 1998), Lo (ed.), Clinical Applications of PCR (Humana Press, Inc. 1998), and Meltzer (ed.), PCR in Bioanalysis (Humana Press, Inc. 1998)).
  • RNA is isolated from a biological sample, reverse transcribed to cDNA, and the cDNA is incubated with Zntr2 primers (see, for example, Wu et al. (eds.), "Rapid Isolation of Specific cDNAs or Genes by PCR,” in Methods in Gene Biotechnology, pages 15-28 (CRC Press, Inc. 1997)). PCR is then performed and the products are analyzed using standard techniques.
  • RNA is isolated from biological sample using, for example, the guanidinium-thiocyanate cell lysis procedure described above.
  • a solid-phase technique can be used to isolate mRNA from a cell lysate.
  • a reverse transcription reaction can be primed with the isolated RNA using random oligonucleotides, short homopolymers of dT, or Zntr2 anti-sense oligomers.
  • Oligo-dT primers offer the advantage that various mRNA nucleotide sequences are amplified that can provide control target sequences.
  • Zntr2 sequences are amplified by the polymerase chain reaction using two flanking oligonucleotide primers that are typically 20 bases in length.
  • PCR amplification products can be detected using a variety of approaches.
  • PCR products can be fractionated by gel electrophoresis, and visualized by ethidium bromide staining.
  • fractionated PCR products can be transferred to a membrane, hybridized with a detectably-labeled Zntr2 probe, and examined by autoradiography.
  • Additional alternative approaches include the use of digoxigenin-labeled deoxyribonucleic acid triphosphates to provide chemiluminescence detection, and the C-TRAK colorimetric assay.
  • Another approach for detection of Zntr2 expression is cycling probe technology, in which a single-stranded DNA target binds with an excess of DNA- RNA-DNA chimeric probe to form a complex, the RNA portion is cleaved with RNAase H, and the presence of cleaved chimeric probe is detected (see, for example, Beggs et al, J. Clin. Microbiol 34:2985 (1996), Bekkaoui et al, Biotechniques 20:240 (1996)).
  • Alternative methods for detection of Zntr2 sequences can utilize approaches such as nucleic acid sequence-based amplification, cooperative amplification of templates by cross-hybridization, and the ligase chain reaction (see, for example, Marshall et al, U.S. Patent No. 5,686,272 (1997), Dyer et al, J. Virol Methods 60: 6 (1996), Ehricht et al, Eur. J. Biochem. 243:358 (1997), and Chadwick et al, J. Virol. Methods 70:59 (1998)).
  • Other standard methods are known to those of skill in the art.
  • Zntr2 probes and primers can also be used to detect and to localize Zntr2 gene expression in tissue samples.
  • Methods for such in situ hybridization are well-known to those of skill in the art (see, for example, Choo (ed.), In Situ Hybridization Protocols (Humana Press, Inc. 1994), Wu et al. (eds.), "Analysis of Cellular DNA or Abundance of mRNA by Radioactive In Situ Hybridization (RISH),” in Methods in Gene Biotechnology, pages 259-278 (CRC Press, Inc. 1997), and Wu et al.
  • the Zntr2 gene has been localized to human chromosome 9q33. This region is also associated with genes linked to familial dysautonomia, limb-girdle muscular dystrophy 2H, and lethal congenital contracture syndrome (Blumenfeld et al, Nature Genet. 4:160 (1993); Makela-Bengs et al, Am. J. Hum. Genet. r57(Suppl.):A30 (1997); Makela-Bengs et al, Am. J. Hum. Genet. 63:506 (1998); Weiler et al, Am. J. Hum. Genet. 63:140 (1998)).
  • Nucleic acid molecules comprising Zntr2 nucleotide sequences can be used to determine whether a subject's chromosomes contain a mutation in the Zntr2 gene. Detectable chromosomal aberrations at the Zntr2 gene locus include, but are not limited to, aneuploidy, gene copy number changes, insertions, deletions, restriction site changes and rearrangements. Of particular interest are genetic alterations that inactivate the Zntr2 gene.
  • Aberrations associated with the Zntr2 locus can be detected using nucleic acid molecules of the present invention by employing molecular genetic techniques, such as restriction fragment length polymorphism analysis, short tandem repeat analysis employing PCR techniques, amplification-refractory mutation system analysis, single-strand conformation polymorphism detection, RNase cleavage methods, denaturing gradient gel electrophoresis, fluorescence-assisted mismatch analysis, and other genetic analysis techniques known in the art (see, for example, Mathew (ed.), Protocols in Human Molecular Genetics (Humana Press, Inc. 1991), Marian, Chest 108:255 (1995), Coleman and Tsongalis, Molecular Diagnostics (Human Press, Inc.
  • molecular genetic techniques such as restriction fragment length polymorphism analysis, short tandem repeat analysis employing PCR techniques, amplification-refractory mutation system analysis, single-strand conformation polymorphism detection, RNase cleavage methods, denaturing gradient gel electrophoresis, fluorescence-assisted
  • RNA is isolated from a biological sample, and used to synthesize cDNA. PCR is then used to amplify the Zntr2 target sequence and to introduce an RNA polymerase promoter, a translation initiation sequence, and an in- frame ATG triplet. PCR products are transcribed using an RNA polymerase, and the transcripts are translated in vitro with a T7-coupled reticulocyte lysate system.
  • the translation products are then fractionated by SDS-PAGE to determine the lengths of the translation products.
  • the protein truncation test is described, for example, by Dracopoli et al. (eds.), Current Protocols in Human Genetics, pages 9.11.1 - 9.11.18 (John Wiley & Sons 1998).
  • kits for performing a diagnostic assay for Zntr2 gene expression or to examine the Zntr2 locus comprise nucleic acid probes, such as double-stranded nucleic acid molecules comprising the nucleotide sequence of SEQ ID NO:l, or a fragment thereof, as well as single-stranded nucleic acid molecules having the complement of the nucleotide sequence of SEQ ID NO:l, or a fragment thereof.
  • Probe molecules may be DNA, RNA, oligonucleotides, and the like.
  • Kits may comprise nucleic acid primers for performing PCR. Such a kit can contain all the necessary elements to perform a nucleic acid diagnostic assay described above.
  • a kit will comprise at least one container comprising a Zntr2 probe or primer.
  • the kit may also comprise a second container comprising one or more reagents capable of indicating the presence of Zntr2 sequences.
  • indicator reagents include detectable labels such as radioactive labels, fluorochromes, chemiluminescent agents, and the like.
  • a kit may also comprise a means for conveying to the user that the Zntr2 probes and primers are used to detect Zntr2 gene expression.
  • written instmctions may state that the enclosed nucleic acid molecules can be used to detect either a nucleic acid molecule that encodes Zntr2, or a nucleic acid molecule having a nucleotide sequence that is complementary to a Zntr2-encoding nucleotide sequence.
  • the written material can be applied directly to a container, or the written material can be provided in the form of a packaging insert.
  • anti-Zntr2 antibodies to screen biological samples in vitro for the presence of Zntr2.
  • anti-Zntr2 antibodies are used in liquid phase.
  • the presence of Zntr2 in a biological sample can be tested by mixing the biological sample with a trace amount of labeled Zntr2 and an anti-Zntr2 antibody under conditions that promote binding between Zntr2 and its antibody.
  • Complexes of Zntr2 and anti-Zntr2 in the sample can be separated from the reaction mixture by contacting the complex with an immobilized protein which binds with the antibody, such as an Fc antibody or Staphylococcus protein A.
  • concentration of Zntr2 in the biological sample will be inversely proportional to the amount of labeled Zntr2 bound to the antibody and directly related to the amount of free labeled Zntr2.
  • in vitro assays can be performed in which anti-Zntr2 antibody is bound to a solid-phase carrier.
  • antibody can be attached to a polymer, such as aminodextran, in order to link the antibody to an insoluble support such as a polymer-coated bead, a plate or a tube.
  • a polymer such as aminodextran
  • an insoluble support such as a polymer-coated bead, a plate or a tube.
  • anti-Zntr2 antibodies can be used to detect Zntr2 in tissue sections prepared from a biopsy specimen. Such immunochemical detection can be used to determine the relative abundance of Zntr2 and to determine the distribution of Zntr2 in the examined tissue.
  • Immunochemical detection can be performed by contacting a biological sample with an anti-Zntr2 antibody, and then contacting the biological sample with a detectably labeled molecule which binds to the antibody.
  • the detectably labeled molecule can comprise an antibody moiety that binds to anti-Zntr2 antibody.
  • the anti-Zntr2 antibody can be conjugated with avidin/streptavidin (or biotin) and the detectably labeled molecule can comprise biotin (or avidin/streptavidin). Numerous variations of this basic technique are well-known to those of skill in the art.
  • an anti-Zntr2 antibody can be conjugated with a detectable label to form an anti-Zntr2 immunoconjugate.
  • Suitable detectable labels include, for example, a radioisotope, a fluorescent label, a chemiluminescent label, an enzyme label, a bioluminescent label or colloidal gold. Methods of making and detecting such detectably-labeled immunoconjugates are well-known to those of ordinary skill in the art, and are described in more detail below.
  • the detectable label can be a radioisotope that is detected by autoradiography. Isotopes that are particularly useful for the purpose of the present invention are 3 H, 125 I, ,3, 1, 35 S and 14 C.
  • Anti-Zntr2 immunoconjugates can also be labeled with a fluorescent compound.
  • the presence of a fluorescently-labeled antibody is determined by exposing the immunoconjugate to light of the proper wavelength and detecting the resultant fluorescence.
  • Fluorescent labeling compounds include fluorescein isothiocyanate, rhodamine, phycoerytherin, phycocyanin, allophycocyanin, o-phthaldehyde and fluorescamine.
  • anti-Zntr2 immunoconjugates can be detectably labeled by coupling an antibody component to a chemiluminescent compound.
  • the presence of the chemiluminescent-tagged immunoconjugate is determined by detecting the presence of luminescence that arises during the course of a chemical reaction.
  • chemiluminescent labeling compounds include luminol, isoluminol, an aromatic acridinium ester, an imidazole, an acridinium salt and an oxalate ester.
  • Bioluminescent compound can be used to label anti-Zntr2 immunoconjugates of the present invention.
  • Bioluminescence is a type of chemiluminescence found in biological systems in which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence.
  • Bioluminescent compounds that are useful for labeling include luciferin, luciferase and aequorin.
  • anti-Zntr2 immunoconjugates can be detectably labeled by linking an anti-Zntr2 antibody component to an enzyme.
  • the enzyme moiety reacts with the substrate to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorometric or visual means.
  • enzymes that can be used to detectably label polyspecific immunoconjugates include ⁇ -galac- tosidase, glucose oxidase, peroxidase and alkaline phosphatase.
  • the convenience and versatility of immunochemical detection can be enhanced by using anti-Zntr2 antibodies that have been conjugated with avidin, streptavidin, and biotin (see, for example, Wilchek et al. (eds.), “Avidin-Biotin Technology,” Methods In Enzymology, Vol. 184 (Academic Press 1990), and Bayer et al, "Immunochemical Applications of Avidin-Biotin Technology,” in Methods In Molecular Biology, Vol. 10, Manson (ed.), pages 149-162 (The Humana Press, Inc. 1992).
  • biotin- or FITC-labeled Zntr2 can be used to identify cells that bind Zntr2. Such can binding can be detected, for example, using flow cytometry.
  • kits for performing an immunological diagnostic assay for Zntr2 gene expression comprise at least one container comprising an anti-Zntr2 antibody, or antibody fragment.
  • a kit may also comprise a second container comprising one or more reagents capable of indicating the presence of Zntr2 antibody or antibody fragments. Examples of such indicator reagents include detectable labels such as a radioactive label, a fluorescent label, a chemiluminescent label, an enzyme label, a bioluminescent label, colloidal gold, and the like.
  • a kit may also comprise a means for conveying to the user that Zntr2 antibodies or antibody fragments are used to detect Zntr2 protein. For example, written instructions may state that the enclosed antibody or antibody fragment can be used to detect Zntr2. The written material can be applied directly to a container, or the written material can be provided in the form of a packaging insert.
  • the present invention includes the use of proteins, polypeptides, and peptides having Zntr2 activity (such as Zntr2 polypeptides, anti-idiotype anti-Zntr2 antibodies, and Zntr2 fusion proteins).
  • the activity of the endogenous Zntr2 ligand can be inhibited by administration of polypeptides that bind the ligand, such as Zntr2 soluble receptor polypeptides or anti-idiotype Zntr2 antibodies, or by the administration of polypeptides that inhibit the effective binding of the Zntr2 ligand with the cognate Zntr2 receptor, such as an antibody that binds with the Zntr2 extracellular domain.
  • a Zntr2 polypeptide e.g., a soluble Zntr2 receptor
  • vascular smooth muscle cell proliferation can be used to regulate vascular smooth muscle cell proliferation, to restore normal neurological functioning after trauma, to treat ocular disorders, to treat kidney and liver disorders, to promote hair follicular development, to stimulate growth and differentiation of various epidermal and epithelial cells in vivo and in vitro, for the treatment of bums, ulcers and corneal incisions, and to stimulate embryogenesis in vitro.
  • Zntr2 polypeptides can also be used to stimulate wound healing (e.g., cutaneous wounds, comeal wounds, injuries to the epithelial-lined hollow organs of the body, etc.), which, for example, result from trauma such as bums, abrasions and cuts, from surgical procedures, such as surgical incisions and skin grafting, and the like.
  • wound healing e.g., cutaneous wounds, comeal wounds, injuries to the epithelial-lined hollow organs of the body, etc.
  • trauma such as bums, abrasions and cuts
  • surgical procedures such as surgical incisions and skin grafting, and the like.
  • the dosage of administered polypeptide, protein or peptide will vary depending upon such factors as the subject's age, weight, height, sex, general medical condition and previous medical history. Typically, it is desirable to provide the recipient with a dosage of a molecule having Zntr2 activity which is in the range of from about 1 pg/kg to 10 mg/kg (amount of agent/body weight of subject), although a lower or higher dosage also may be administered as circumstances dictate. Suitable subjects include mammalian subjects, such as humans.
  • Administration of a molecule having Zntr2 activity to a subject can be intravenous, intraarterial, intraperitoneal, intramuscular, subcutaneous, intrapleural, intrathecal, by perfusion through a regional catheter, or by direct intralesional injection.
  • the administration may be by continuous infusion or by single or multiple boluses.
  • a pharmaceutical composition comprising a protein, polypeptide, or peptide having Zntr2 activity can be formulated according to known methods to prepare pharmaceutically useful compositions, whereby the therapeutic proteins are combined in a mixture with a pharmaceutically acceptable carrier.
  • a composition is said to be a "pharmaceutically acceptable carrier" if its administration can be tolerated by a recipient patient.
  • Sterile phosphate-buffered saline is one example of a pharmaceutically acceptable carrier.
  • suitable carriers are well-known to those in the art. See, for example, Gennaro (ed.), Remington's Pharmaceutical Sciences, 19th Edition (Mack Publishing Company 1995).
  • molecules having Zntr2 activity and a pharmaceutically acceptable carrier are administered to a patient in a therapeutically effective amount.
  • a combination of a protein, polypeptide, or peptide having Zntr2 activity and a pharmaceutically acceptable carrier is said to be administered in a "therapeutically effective amount" if the amount administered is physiologically significant.
  • An agent is physiologically significant if its presence results in a detectable change in the physiology of a recipient patient.
  • a pharmaceutical composition comprising molecules having Zntr2 activity can be furnished in liquid form, or in solid form. Liquid forms, including liposome-encapsulated formulations, are illustrated by injectable solutions and oral suspensions.
  • Exemplary solid forms include capsules, tablets, and controlled-release forms, such as a miniosmotic pump or an implant.
  • Other dosage forms can be devised by those skilled in the art, as shown, for example, by Ansel and Popovich, Pharmaceutical Dosage Forms and Drug Delivery Systems, 5 th Edition (Lea & Febiger 1990), Gennaro (ed.), Remington's Pharmaceutical Sciences, 19 th Edition (Mack Publishing Company 1995), and by Ranade and Hollinger, Drug Delivery Systems (CRC Press 1996).
  • Zntr2 pharmaceutical compositions may be supplied as a kit comprising a container that comprises Zntr2.
  • Zntr2 can be provided in the form of an injectable solution for single or multiple doses, or as a sterile powder that will be reconstituted before injection.
  • a kit may further comprise written information on indications and usage of the pharmaceutical composition.
  • such information may include a statement that the Zntr2 composition is contraindicated in patients with known hypersensitivity to Zntr2.
  • the present invention includes the use of Zntr2 nucleotide sequences to provide Zntr2 polypeptides to a subject in need of such treatment.
  • a therapeutic expression vector can be provided that inhibits Zntr2 gene expression, such as an anti-sense molecule, a ribozyme, or an external guide sequence molecule.
  • an expression vector is constructed in which a nucleotide sequence encoding a Zntr2 gene is operably linked to a core promoter, and optionally a regulatory element, to control gene transcription.
  • a core promoter and optionally a regulatory element
  • a Zntr2 gene can be delivered using recombinant viral vectors, including for example, adenoviral vectors (e.g., Kass-Eisler et al, Proc. Nat'l Acad. Sci. USA 90:11498 (1993), Kolls et al, Proc. Nat'l Acad. Sci. USA 97:215 (1994), Li et al, Hum. Gene Ther. 4:403 (1993), Vincent et al, Nat. Genet. 5:130 (1993), and Zabner et al, Cell 75:207 (1993)), adenovirus-associated viral vectors (Flotte et al, Proc. Nat'l Acad. Sci.
  • adenoviral vectors e.g., Kass-Eisler et al, Proc. Nat'l Acad. Sci. USA 90:11498 (1993), Kolls et al, Proc. Nat'l Acad. Sci. USA 97:215 (19
  • alphaviruses such as Semliki Forest Vims and Sindbis Vims (Hertz and Huang, J. Vir. 66:857 (1992), Raju and Huang, J. Vir. 65:2501 (1991), and Xiong et al, Science 243:1188 (1989)), herpes viral vectors (e.g., U.S. Patent Nos. 4,769,331, 4,859,587, 5,288,641 and 5,328,688), parvo virus vectors (Koering et al, Hum. Gene Therap. 5:457 (1994)), pox vims vectors (Ozaki et al, Biochem. Biophys. Res. Comm.
  • pox vimses such as canary pox vims or vaccinia vims (Fisher-Hoch et al, Proc. Nat'l Acad. Sci. USA 86:3 7 (1989), and Flexner et al, Ann. N Y. Acad. Sci. 569:86 (1989)), and retroviruses (e.g., Baba et al, J. Neurosurg 79:729 (1993), Ram et al, Cancer Res. 53:83 (1993), Takamiya et al, J. Neurosci.
  • the viral vector itself, or a viral particle which contains the viral vector may be utilized in the methods and compositions described below.
  • adenovirus a double-stranded DNA vims
  • the adenovims system offers several advantages including: (i) the ability to accommodate relatively large DNA inserts, (ii) the ability to be grown to high-titer, (iii) the ability to infect a broad range of mammalian cell types, and (iv) the ability to be used with many different promoters including ubiquitous, tissue specific, and regulatable promoters.
  • adenovimses can be administered by intravenous injection, because the vimses are stable in the bloodstream.
  • inserts are incorporated into the viral DNA by direct ligation or by homologous recombination with a co-transfected plasmid.
  • the essential El gene is deleted from the viral vector, and the vims will not replicate unless the El gene is provided by the host cell.
  • adenovims When intravenously administered to intact animals, adenovims primarily targets the liver. Although an adenoviral delivery system with an El gene deletion cannot replicate in the host cells, the host's tissue will express and process an encoded heterologous protein. Host cells will also secrete the heterologous protein if the corresponding gene includes a secretory signal sequence. Secreted proteins will enter the circulation from tissue that expresses the heterologous gene (e.g., the highly vascularized liver).
  • adenoviral vectors containing various deletions of viral genes can be used to reduce or eliminate immune responses to the vector.
  • Such adenovimses are El -deleted, and in addition, contain deletions of E2A or E4 (Lusky et al, J. Virol 72:2022 (1998); Raper et al, Human Gene Therapy 9:671 (1998)).
  • the deletion of E2b has also been reported to reduce immune responses (Amalfitano et al, J. Virol. 72:926 (1998)). By deleting the entire adenovims genome, very large inserts of heterologous DNA can be accommodated.
  • High titer stocks of recombinant vimses capable of expressing a therapeutic gene can be obtained from infected mammalian cells using standard methods.
  • recombinant HSV can be prepared in Vero cells, as described by Brandt et al, J. Gen. Virol. 72:2043 (1991), Herold et al, J. Gen. Virol. 75:1211 (1994), Visalli and Brandt, Virology 755:419 (1991), Grau et al, Invest. Ophthalmol Vis. Sci. 30:2474 (1989), Brandt et al, J. Virol. Meth. 36:209 (1992), and by Brown and MacLean (eds.), HSV Virus Protocols (Humana Press 1997).
  • an expression vector comprising a Zntr2 gene can be introduced into a subject's cells by lipofection in vivo using liposomes.
  • Synthetic cationic lipids can be used to prepare liposomes for in vivo transfection of a gene encoding a marker (Feigner et al, Proc. Nat'l Acad. Sci. USA 54:7413 (1987); Mackey et al, Proc. Nat'l Acad. Sci. USA 55:8027 (1988)).
  • the use of lipofection to introduce exogenous genes into specific organs in vivo has certain practical advantages.
  • Liposomes can be used to direct transfection to particular cell types, which is particularly advantageous in a tissue with cellular heterogeneity, such as the pancreas, liver, kidney, and brain.
  • Lipids may be chemically coupled to other molecules for the purpose of targeting.
  • Targeted peptides e.g., hormones or neurotransmitters
  • proteins such as antibodies
  • non-peptide molecules can be coupled to liposomes chemically.
  • Electroporation is another alternative mode of administration of a Zntr2 nucleic acid molecules. For example, Aihara and Miyazaki, Nature Biotechnology 16:867 (1998), have demonstrated the use of in vivo electroporation for gene transfer into muscle.
  • a therapeutic gene may encode a Zntr2 anti-sense RNA that inhibits the expression of Zntr2.
  • Suitable sequences for Zntr2 anti-sense molecules can be derived from the nucleotide sequences of Zntr2 disclosed herein.
  • an expression vector can be constructed in which a regulatory element is operably linked to a nucleotide sequence that encodes a ribozyme.
  • Ribozymes can be designed to express endonuclease activity that is directed to a certain target sequence in a mRNA molecule (see, for example, Draper and Macejak, U.S. Patent No. 5,496,698, McSwiggen, U.S. Patent No. 5,525,468, Chowrira and McSwiggen, U.S. Patent No. 5,631,359, and Robertson and Goldberg, U.S. Patent No. 5,225,337).
  • ribozymes include nucleotide sequences that bind with Zntr2 mRNA.
  • expression vectors can be constmcted in which a regulatory element directs the production of RNA transcripts capable of promoting RNase P-mediated cleavage of mRNA molecules that encode a Zntr2 gene.
  • an external guide sequence can be constmcted for directing the endogenous ribozyme, RNase P, to a particular species of intracellular mRNA, which is subsequently cleaved by the cellular ribozyme (see, for example, Airman et al, U.S. Patent No. 5,168,053, Yuan et al, Science 263:1269 (1994), Pace et al, international publication No. WO 96/18733, George et al, international publication No.
  • the external guide sequence comprises a ten to fifteen nucleotide sequence complementary to Zntr2 mRNA, and a 3'-NCCA nucleotide sequence, wherein N is preferably a purine.
  • the external guide sequence transcripts bind to the targeted mRNA species by the formation of base pairs between the mRNA and the complementary external guide sequences, thus promoting cleavage of mRNA by RNase P at the nucleotide located at the 5'-side of the base-paired region.
  • the dosage of a composition comprising a therapeutic vector having a Zntr2 nucleotide acid sequence, such as a recombinant vims will vary depending upon such factors as the subject's age, weight, height, sex, general medical condition and previous medical history.
  • Suitable routes of administration of therapeutic vectors include intravenous injection, intraarterial injection, intraperitoneal injection, intramuscular injection, intratumoral injection, and injection into a cavity that contains a tumor.
  • a composition comprising viral vectors, non-viral vectors, or a combination of viral and non- viral vectors of the present invention can be formulated according to known methods to prepare pharmaceutically useful compositions, whereby vectors or vimses are combined in a mixture with a pharmaceutically acceptable carrier.
  • a composition such as phosphate-buffered saline is said to be a "pharmaceutically acceptable carrier” if its administration can be tolerated by a recipient subject.
  • suitable carriers are well-known to those in the art (see, for example, Remington 's Pharmaceutical Sciences, 19th Ed. (Mack Publishing Co. 1995), and Gilman 's the Pharmacological Basis of Therapeutics, 7th Ed. (MacMillan Publishing Co. 1985)).
  • a therapeutic gene expression vector, or a recombinant vims comprising such a vector, and a pharmaceutically acceptable carrier are administered to a subject in a therapeutically effective amount.
  • a combination of an expression vector (or vims) and a pharmaceutically acceptable carrier is said to be administered in a "therapeutically effective amount" if the amount administered is physiologically significant.
  • An agent is physiologically significant if its presence results in a detectable change in the physiology of a recipient subject.
  • the therapy is preferably somatic cell gene therapy.
  • the preferred treatment of a human with a therapeutic gene expression vector or a recombinant vims does not entail introducing into cells a nucleic acid molecule that can form part of a human germ line and be passed onto successive generations (i.e., human germ line gene therapy).
  • Transgenic mice can be engineered to over-express the Zntr2 gene in all tissues or under the control of a tissue-specific or tissue-preferred regulatory element. These over-producers of Zntr2 can be used to characterize the phenotype that results from over-expression, and the transgenic animals can serve as models for human disease caused by excess Zntr2. Transgenic mice that over-express Zntr2 also provide model bioreactors for production of Zntr2, such as soluble Zntr2, in the milk or blood of larger animals.
  • a method for producing a transgenic mouse that expresses a Zntr2 gene can begin with adult, fertile males (studs) (B6C3fl , 2-8 months of age (Taconic Farms, Germantown, NY)), vasectomized males (duds) (B6D2fl, 2-8 months, (Taconic Farms)), prepubescent fertile females (donors) (B6C3fl, 4-5 weeks, (Taconic Farms)) and adult fertile females (recipients) (B6D2fl, 2-4 months, (Taconic Farms)).
  • the donors are acclimated for one week and then injected with approximately 8 IU/mouse of Pregnant Mare's Semm gonadotrophin (Sigma Chemical Company; St. Louis, MO) I. P., and 46-47 hours later, 8 IU/mouse of human Chorionic Gonadotropin (hCG (Sigma)) I.P. to induce superovulation.
  • Donors are mated with studs subsequent to hormone injections. Ovulation generally occurs within 13 hours of hCG injection. Copulation is confirmed by the presence of a vaginal plug the morning following mating.
  • Fertilized eggs are collected under a surgical scope.
  • the oviducts are collected and eggs are released into urinanalysis slides containing hyaluronidase (Sigma).
  • Eggs are washed once in hyaluronidase, and twice in Whitten's W640 medium (described, for example, by Menino and O'Claray, Biol. Reprod. 77:159 (1986), and Dienhart and Downs, Zygote 4:129 (1996)) that has been incubated with 5% CO 2 , 5% O 2 , and 90% N at 37°C.
  • the eggs are then stored in a 37°C/5% CO 2 incubator until microinjection.
  • plasmid DNA containing a Zntr2 encoding sequence is linearized, gel-purified, and resuspended in 10 mM Tris-HCl (pH 7.4), 0.25 mM EDTA (pH 8.0), at a final concentration of 5-10 nanograms per microliter for microinjection.
  • the Zntr2 encoding sequences can encode a polypeptide comprising amino acid resides 31 to 507 of SEQ ID NO:2. Plasmid DNA is microinjected into harvested eggs contained in a drop of W640 medium overlaid by warm, CO -equilibrated mineral oil.
  • the DNA is drawn into an injection needle (pulled from a 0.75mm ID, 1mm OD borosilicate glass capillary), and injected into individual eggs. Each egg is penetrated with the injection needle, into one or both of the haploid pronuclei.
  • Picoliters of DNA are injected into the pronuclei, and the injection needle withdrawn without coming into contact with the nucleoli. The procedure is repeated until all the eggs are injected. Successfully microinjected eggs are transferred into an organ tissue-culture dish with pre-gassed W640 medium for storage overnig °ht in a 37°C/5% CO 2 incubator.
  • two-cell embryos are transferred into pseudopregnant recipients.
  • the recipients are identified by the presence of copulation plugs, after copulating with vasectomized duds.
  • Recipients are anesthetized and shaved on the dorsal left side and transferred to a surgical microscope.
  • a small incision is made in the skin and through the muscle wall in the middle of the abdominal area outlined by the ribcage, the saddle, and the hind leg, midway between knee and spleen.
  • the reproductive organs are exteriorized onto a small surgical drape.
  • the fat pad is stretched out over the surgical drape, and a baby serrefine (Roboz, Rockville, MD) is attached to the fat pad and left hanging over the back of the mouse, preventing the organs from sliding back in.
  • a baby serrefine Robot, Rockville, MD
  • the pipette is transferred into the nick in the oviduct, and the embryos are blown in, allowing the first air bubble to escape the pipette.
  • the fat pad is gently pushed into the peritoneum, and the reproductive organs allowed to slide in.
  • the peritoneal wall is closed with one suture and the skin closed with a wound clip.
  • the mice recuperate on a 37°C slide warmer for a minimum of four hours.
  • the recipients are returned to cages in pairs, and allowed 19-21 days gestation. After birth, 19-21 days postpartum is allowed before weaning.
  • the weanlings are sexed and placed into separate sex cages, and a 0.5 cm biopsy (used for genotyping) is snipped off the tail with clean scissors.
  • Genomic DNA is prepared from the tail snips using, for example, a QIAGEN DNEASY kit following the manufacturer's instructions. Genomic DNA is analyzed by PCR using primers designed to amplify a Zntr2 gene or a selectable marker gene that was introduced in the same plasmid. After animals are confirmed to be transgenic, they are back-crossed into an inbred strain by placing a transgenic female with a wild-type male, or a transgenic male with one or two wild-type female(s). As pups are bom and weaned, the sexes are separated, and their tails snipped for genotyping. To check for expression of a transgene in a live animal, a partial hepatectomy is performed.
  • a surgical prep is made of the upper abdomen directly below the zyphoid process. Using sterile technique, a small 1.5-2 cm incision is made below the sternum and the left lateral lobe of the liver exteriorized. Using 4-0 silk, a tie is made around the lower lobe securing it outside the body cavity. An atraumatic clamp is used to hold the tie while a second loop of absorbable Dexon (American Cyanamid; Wayne, NJ.) is placed proximal to the first tie. A distal cut is made from the Dexon tie and approximately 100 mg of the excised liver tissue is placed in a sterile petri dish.
  • the excised liver section is transferred to a 14 ml polypropylene round bottom tube and snap frozen in liquid nitrogen and then stored on dry ice.
  • the surgical site is closed with suture and wound clips, and the animal's cage placed on a 37°C heating pad for 24 hours post operatively.
  • the animal is checked daily post operatively and the wound clips removed 7-10 days after surgery.
  • the expression level of Zntr2 mRNA is examined for each transgenic mouse using an RNA solution hybridization assay or polymerase chain reaction.
  • Such transgenic mice provide useful models for diseases associated with a lack of Zntr2.
  • Zntr2 gene expression can be inhibited using anti-sense genes, ribozyme genes, or external guide sequence genes.
  • anti-sense genes ribozyme genes, or external guide sequence genes.
  • inhibitory sequences are targeted to Zntr2 mRNA.
  • Methods for producing transgenic mice that have abnormally low expression of a particular gene are known to those in the art (see, for example, Wu et al. , “Gene Underexpression in Cultured Cells and Animals by Antisense DNA and RNA Strategies," in Methods in Gene Biotechnology, pages 205-224 (CRC Press 1997)).
  • An alternative approach to producing transgenic mice that have little or no Zntr2 gene expression is to generate mice having at least one normal Zntr2 allele replaced by a nonfunctional Zntr2 gene.
  • One method of designing a nonfunctional Zntr2 gene is to insert another gene, such as a selectable marker gene, within a nucleic acid molecule that encodes Zntr2. Standard methods for producing these so-called “knockout mice” are known to those skilled in the art (see, for example, Jacob, "Expression and Knockout of Interferons in Transgenic Mice," in Overexpression and Knockout of Cytokines in Transgenic Mice, Jacob (ed.), pages 111-124 (Academic Press, Ltd. 1994), and Wu et al, "New Strategies for Gene Knockout,” in Methods in Gene Biotechnology, pages 339-365 (CRC Press 1997)).
  • a full-length Zntr2 clone was obtained by screening a fetal brain arrayed library, containing 80 pools of 12,000 colonies each, with an expressed sequence tag (EST). Twenty picomoles each of oligonucleotide primers ZC18,638 (5' CTC TGT GGT TGG AAG CCT GAA TGT G 3'; SEQ ID NO:4) and ZC 18,639 (5' AGC CCT CTT TGC AGG TTT CAC AGT G 3'; SEQ ID NO:5) and 5 ⁇ ls of each pool were used in each reaction.
  • EST expressed sequence tag
  • the conditions used were as follows: 94°C for 1 minute, then mn for 30 cycles of 94°C, 20 seconds; 68°C, 1 minute; and ended with a 7 minute incubation at 72°C. There were three strong positives and four weak positives. The three strong positives were followed to the next level (transfer plate level). Twelve pools containing 1,000 clones each for each working plate positive was screened by PCR as above. Each of the three working plate positives had one positive in the corresponding transfer plate level. Pool HI 1 was analyzed further.
  • the DNA was affixed to the filter using a STRATALINKER UV crosslinker (STRATAGENE, La Jolla, CA ) at 1200 joules and prewashed at 65°C in prewash buffer consisting of 0.25 x SSC, 0.25% SDS and 1 mM EDTA, changing solution three times for a total of 45 minutes to remove cell debris.
  • Lifts were prehybridized overnight at 65°C in 10 mis EXPRESSHYB Hybridization Solution (Clontech, Palo Alto, CA) mixed with 1 mg salmon sperm DNA, which had been boiled 5 minutes, and then stored on ice for one minute.
  • the probe was generated by reamplification of the subcloned PCR fragment as described below with the primers ZC14,064 (5' CTC CAG AGT ATG TAT GTA ACT GC 3'; SEQ ID NO:6) and ZC 14,065 (5' CAC ATG GCT GAC AGA CCC ACA 3'; SEQ ID NO:7).
  • the PCR product was fractionated by electrophoresis on a 1.5% TBE gel, the fragment excised with a razor blade, and the DNA extracted from the agarose with the QIAquick Gel Extraction Kit (QIAGEN, Chats worth, CA).
  • Radiolabeled probe (8 x 10 ⁇ cpms) and 1 mg of salmon sperm DNA were boiled 5 minutes, cooled on ice for 1 minute, mixed with 8 mis of EXPRESSHYB hybridization solution and added to the filter lifts. Hybridization was carried out overnight at 65°C. Lifts were washed in prewash solution until radioactivity was below 200 cpm. Blots were exposed to film overnight at -80°C. One of the positives was sequenced, and found to contain a full-length sequence with a 1,805 base pair open reading frame.
  • Northern analyses were performed using Human Multiple Tissue Blots I, II and III and Human RNA Master Blot (CLONTECH Laboratories, Inc.; Palo Alto, CA) to determine the tissue distribution of Zntr2.
  • a 187 base pair subcloned PCR fragment which had been subcloned into the pCR2.1 vector (Invitrogen, Carlsbad, CA) was used as the probe.
  • the PCR fragment was generated using 20 pms each of primers ZG 14,064 and ZG 14,065 and 5 ⁇ ls of a utems cDNA marathon library prepared from utems RNA using the Marathon cDNA Amplification Kit (CLONTECH).
  • the reactions conditions were as follows: a initial denaturation at 94°C for 1 minute, followed by 5 cycles of 94°C, 20 seconds; 59°C, 30 seconds; 72°C, 30 seconds, followed by 30 cycles of 94°C, 20 seconds; 55°C, 30 seconds; 72°C, 30 seconds and then a final extension for 7 minutes at 72°C. Due to the low level of material, reamplification was performed for subcloning at the following conditions with 2 ⁇ ls of the first reaction: initial 94°C denaturation followed by 25 cycles of 94°C, 20 seconds; 59°C, 30 seconds; 72°C, 30 seconds and then final extension at 72°C for 7 minutes.
  • Radiolabeled probe (7 x 10" cpm) and 1 mg of salmon sperm DNA were boiled for 1 minute, stored on ice for 1 minute, mixed with 7 mis of the same EXPRESSHYB solution, and then added to the blots mentioned above. Hybridization was carried out overnight at 65°C. Blots were washed in 2 x SSC, 0.1 %> SDS at room temperature for 40 minutes with several solution changes, followed by a 40 minute wash at 50°C in 0.1 x SSC, 0.1%) SDS with one solution change. After an overnight -80°C exposure, the blots were additionally washed in 0.1 x SSC, 0.1 % SDS at 60°C to remove general background, and the overnight exposure was repeated.
  • RNA Master Blot showed strong expression in fetal brain.
  • Zntr2 was mapped to chromosome 9 using the commercially available version of the "Stanford G3 Radiation Hybrid Mapping Panel" (Research Genetics, Inc., Huntsville, AL).
  • the Stanford G3 RH Panel contains amplifiable DNA molecules from each of 83 radiation hybrid clones of the whole human genome, plus two control DNAs (the RM donor and the A3 recipient).
  • a WWW server http://shgc- www.stanford.edu) allows chromosomal localization of markers.
  • Each of the 85 PCR reactions consisted of 2 ⁇ l lOx KlenTaq PCR reaction buffer (CLONTECH Laboratories, Inc., Palo Alto, CA), 1.6 ⁇ l dNTPs mix (2.5 mM each, PERKIN-ELMER, Foster City, CA), 1 ⁇ l sense primer, ZC 21,097 (5' CCC GAG AGT GGT GAG TGC 3'; SEQ ID NO:8), 1 ⁇ l antisense primer, ZC 21,098 (5 * CTC GAG GTC GTG AAC ATA 3'; SEQ ID NO:9), 2 ⁇ l "RediLoad” (Research Genetics, Inc., Huntsville, AL), 0.4 ⁇ l 50x Advantage KlenTaq Polymerase Mix (CLONTECH), 25 ng of DNA from an individual hybrid clone or control and ddH2O for a total volume of 20 ⁇ l.
  • the reactions were overlaid with an equal amount of mineral oil and sealed.
  • the PCR cycler conditions were as follows: an initial 1 cycle 5 minute denaturation at 94°C, 35 cycles of a 45 second denaturation at 94°C, 45 seconds annealing at 64°C and 1 minute, and 15 seconds extension at 72°C, followed by a final 1 cycle extension of 7 minutes at 72°C.
  • the reactions were separated by electrophoresis on a 2% agarose gel (Life Technologies, Gaithersburg, MD).

Landscapes

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

Abstract

Les domaines du type facteur de croissance épidermique sont présents dans de nombreuses protéines remplissant des fonctions biologiques importantes, comme la régulation de la différentiation cellulaire. Zntr2 est un nouveau membre de ce groupe de protéines.
PCT/US2000/001419 1999-01-25 2000-01-20 Polypeptide humain avec multiples domaines du type facteur de croissance epidermique (egf), denomme zntr2 WO2000043512A1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CA002358873A CA2358873A1 (fr) 1999-01-25 2000-01-20 Polypeptide humain avec multiples domaines du type facteur de croissance epidermique (egf), denomme zntr2
AU28546/00A AU2854600A (en) 1999-01-25 2000-01-20 Human polypeptide having multiple epidermal growth factor (egf) -like domains, zntr2

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US23707499A 1999-01-25 1999-01-25
US09/237,074 1999-01-25

Publications (1)

Publication Number Publication Date
WO2000043512A1 true WO2000043512A1 (fr) 2000-07-27

Family

ID=22892233

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2000/001419 WO2000043512A1 (fr) 1999-01-25 2000-01-20 Polypeptide humain avec multiples domaines du type facteur de croissance epidermique (egf), denomme zntr2

Country Status (3)

Country Link
AU (1) AU2854600A (fr)
CA (1) CA2358873A1 (fr)
WO (1) WO2000043512A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002004599A2 (fr) * 2000-07-12 2002-01-17 Zymogenetics, Inc. Proteine placentaire possedant des domaines multiples semblables a egf
WO2022217037A1 (fr) * 2021-04-09 2022-10-13 Board Of Regents, The University Of Texas System Compositions et méthodes de traitement de maladies pulmonaires chroniques

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999038972A2 (fr) * 1998-01-28 1999-08-05 Chiron Corporation Genes humains et produits ii d'expression genique
WO2000012708A2 (fr) * 1998-09-01 2000-03-09 Genentech, Inc. Nouveaux pro-polypeptides et sequences correspondantes

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999038972A2 (fr) * 1998-01-28 1999-08-05 Chiron Corporation Genes humains et produits ii d'expression genique
WO2000012708A2 (fr) * 1998-09-01 2000-03-09 Genentech, Inc. Nouveaux pro-polypeptides et sequences correspondantes

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
NAKAYAMA M ET AL: "Identification of high-molecular-weight proteins with multiple EGF-like motifs by motif-trap screening", GENOMICS,US,ACADEMIC PRESS, SAN DIEGO, vol. 51, no. 1, 1 July 1998 (1998-07-01), pages 27 - 34, XP002097998, ISSN: 0888-7543 *

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002004599A2 (fr) * 2000-07-12 2002-01-17 Zymogenetics, Inc. Proteine placentaire possedant des domaines multiples semblables a egf
WO2002004599A3 (fr) * 2000-07-12 2003-01-30 Zymogenetics Inc Proteine placentaire possedant des domaines multiples semblables a egf
WO2022217037A1 (fr) * 2021-04-09 2022-10-13 Board Of Regents, The University Of Texas System Compositions et méthodes de traitement de maladies pulmonaires chroniques

Also Published As

Publication number Publication date
CA2358873A1 (fr) 2000-07-27
AU2854600A (en) 2000-08-07

Similar Documents

Publication Publication Date Title
CA2392128A1 (fr) Proteines humaines zven
US7169897B2 (en) Adipocyte complement related protein ZACRP3X2
US20030017980A1 (en) Mammalian Wnt polypeptide-5
US20020037551A1 (en) New member of the lectin superfamily
US20030077751A1 (en) Zvwf1: a member of the von Willebrand factor type a domain superfamily
US20030166049A1 (en) Human secreted protein, Zsig47
WO2001038501A2 (fr) Serine protease humaine
WO2000043512A1 (fr) Polypeptide humain avec multiples domaines du type facteur de croissance epidermique (egf), denomme zntr2
US6403783B1 (en) Nucleic acid which encodes the tumor marker ZSIG62
US6423526B1 (en) Human serine protease
US6524822B1 (en) Polynucleotide encoding human serpin
WO2000042184A1 (fr) Zlrr3: proteine humaine repetee riche en leucine
US20020151029A1 (en) Human serine protease
US20020150991A1 (en) Insulin homolog polypeptide Zins5
US20020091239A1 (en) Human chemokine
US20030143678A1 (en) Zlrr3: a human leucine-rich repeat protein
US20030108995A1 (en) Human proteoglycan
US20020150974A1 (en) Placental protein having multiple EGF-like domains
WO2001032707A1 (fr) Semaphorine humaine
US20030170824A1 (en) Human chemokine
EP1117793A2 (fr) Proteine secretoire 48
WO2002012479A2 (fr) Zcys9: un membre de la superfamille des cystatines
WO2000042183A2 (fr) Marqueur tumoral zsig62
WO2001066749A2 (fr) Nouvel element de la superfamille de la lectine
WO2001038534A2 (fr) Serpine humaine

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW SD SL SZ TZ 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
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
ENP Entry into the national phase

Ref document number: 2358873

Country of ref document: CA

Ref country code: CA

Ref document number: 2358873

Kind code of ref document: A

Format of ref document f/p: F

WWE Wipo information: entry into national phase

Ref document number: PA/a/2001/007521

Country of ref document: MX

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase