WO1999009198A1 - Homologues humains nodal et lefty - Google Patents

Homologues humains nodal et lefty Download PDF

Info

Publication number
WO1999009198A1
WO1999009198A1 PCT/US1998/017211 US9817211W WO9909198A1 WO 1999009198 A1 WO1999009198 A1 WO 1999009198A1 US 9817211 W US9817211 W US 9817211W WO 9909198 A1 WO9909198 A1 WO 9909198A1
Authority
WO
WIPO (PCT)
Prior art keywords
amino acid
polypeptide
seq
lefty
nodal
Prior art date
Application number
PCT/US1998/017211
Other languages
English (en)
Inventor
Steven M. Ruben
Daniel R. Soppet
Reinhard Ebner
Original Assignee
Human Genome Sciences, 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 Human Genome Sciences, Inc. filed Critical Human Genome Sciences, Inc.
Priority to AU91086/98A priority Critical patent/AU9108698A/en
Publication of WO1999009198A1 publication Critical patent/WO1999009198A1/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
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to two novel human genes encoding polypeptides which are members of the transforming growth factor-beta (TGF- ⁇ ) superfamily. More specifically, isolated nucleic acid molecules are provided encoding human polypeptides designated the Nodal and Lefty homologues, hereinafter referred to as "Nodal” and “Lefty”, respectively. Nodal and Lefty polypeptides are also provided, as are vectors, host cells and recombinant methods for producing the same. Also provided are diagnostic methods for detecting disorders related to the regulation of cell growth and differentiation and therapeutic methods for treating such disorders. The invention further relates to screening methods for identifying agonists and antagonists of Nodal and Lefty activity.
  • TGF- ⁇ transforming growth factor-beta
  • the TGF- ⁇ family of peptide growth factors includes at least five members (TGF- ⁇ 1 through TGF- ⁇ 5) all of which form homodimers of approximately 25 kd.
  • the TGF- ⁇ family belongs to a larger, extended super family of peptide signaling molecules that includes the Muellerian inhibiting substance (Cate, R. L., et al, Cell 45:685-698 (1986)), decapentaplegic (Padgett, R. W., et al, Nature 325:81-84 (1987)), bone morphogenic factors (Wozney, J. M., et al, Science 242:1528-1534 (1988)), vgl (Weeks, D. L. and Melton, D.
  • TGF- ⁇ was originally described as a factor that induced normal rat kidney fibroblasts to proliferate in soft agar in the presence of epidermal growth factor (Roberts, A. B., et al, Proc. Natl. Acad. Sci. USA 78:5339-5343 (1981)). TGF- ⁇ has subsequently been shown to exert a number of different effects in a variety of cells. For example, TGF- ⁇ can inhibit the differentiation of certain cells of mesodermal origin (Florini, J. R., et al, J. Biol. Chem. 261: 1659-16513 (1986)), induced the differentiation of others (Seyedine, S. M. et al, Proc. Natl. Acad.
  • retinoblastoma lack detectable TGF- ⁇ receptors at their cell surface and fail to respond to TGF- ⁇ , while their normal counterparts express self-surface receptors in their growth is potently inhibited by TGF- ⁇ (Kim Chi, A., et al, Science 240:196-198 (1988)). More specifically, TGF- ⁇ 1 stimulates the anchorage-independent growth of normal rat kidney fibroblasts (Robert et al, Proc. Natl. Acad. Sci. USA 78:5339-5343 (1981)).
  • TGF- ⁇ 1 stimulates formation of extracellular matrix molecules in the liver and lung.
  • levels of TGF- ⁇ 1 are higher than normal, formation of fiber occurs in the extracellular matrix of the liver and lung which can be fatal.
  • High levels of TGF- ⁇ 1 occur due to chemotherapy and bone marrow transplant as an attempt to treat cancers such as breast cancer.
  • TGF- ⁇ 2 A second protein termed TGF- ⁇ 2 was isolated from several sources including demineralized bone, a human prostatic adenocarcinoma cell line (Ikeda, et al, J. Bio. Chem. 26:2406-2410 (1987)). TGF- ⁇ 2 shared several functional similarities with TGF- ⁇ 1. These proteins are now known to be members of a family of related growth modulatory proteins including TGF- ⁇ 3 (Ten-Dijke, et al, Proc. Natl. Acad. Sci. USA 85:471-4719 (1988)), Muellerian inhibitory substance and the inhibins.
  • the present invention provides isolated nucleic acid molecules comprising polynucleotides encoding at least a portion of the Nodal polypeptide having the complete amino acid sequence shown in SEQ ID NO:2 or the complete amino acid sequence encoded by the cDNA clone deposited as plasmid DNA as ATCC Deposit Number 209092, on June 5, 1997 or the complete amino acid sequence encoded by the cDNA clone deposited as plasmid DNA as ATCC Deposit Number 209135, on July 2, 1997.
  • nucleotide sequence determined by sequencing the deposited Nodal clone, which is shown in Figures 1 A and B (SEQ ID NO:l), and contains a single open reading frame encoding a complete polypeptide of 283 amino acid residues initiating with a codon encoding an N-terminal aspartic acid residue at nucleotide positions 1-3 with a predicted molecular weight of about 32.5 kDa.
  • Nucleic acid molecules of the invention include those encoding the complete amino acid sequence shown in SEQ ID NO:2, the complete amino acid sequence encoded by the cDNA clone in ATCC
  • the present invention also provides isolated nucleic acid molecules comprising polynucleotides encoding at least a portion of the Lefty polypeptide having the complete amino acid sequence shown in SEQ ID NO:4 or the complete amino acid sequence encoded by the cDNA clone deposited as plasmid DNA as ATCC Deposit Number 209091 on June 5, 1997.
  • nucleotide sequences determined by sequencing the deposited Lefty clone, which is shown in Figures 2 A and B (SEQ ID NO: 3), and contains a single open reading frame encoding a complete polypeptide of 366 amino acid residues with an initiation codon encoding an N-terminal methionine at nucleotide positions 53-55, and a predicted molecular weight of about 40.9 kDa.
  • Nucleic acid molecules of the invention include those encoding the complete amino acid sequence shown in SEQ ID NO:4, those encoding the complete amino acid sequence shown in SEQ ID NO:4 excluding the N-terminal methionine, the complete amino acid sequences encoded by the cDNA clone in ATCC Deposit Numbers 209091, or the complete amino acid sequences excepting the N-terminal methionine encoded by the cDNA clone in ATCC Deposit Number 209091, which molecules also can encode additional amino acids fused to the N-terminus of the Lefty amino acid sequence.
  • the Nodal protein of the present invention shares sequence homology with the translation product of the murine mRNA for Nodal ( Figure 3; SEQ ID NO:5), including the conserved predicted active domain of about 110 amino acids.
  • Murine Nodal is thought to be essential for mesoderm formation and subsequent organization of axial structures in early mouse development.
  • the homology between murine Nodal and the human Nodal homologue of the present invention indicates that the human Nodal homologue of the present invention may also be involved in a developmental process such as the correct formation of various structures or in one or more post-developmental capacities including sexual development, pituitary hormone production, and the creation of bone and cartilage, as are many of the other members of the TGF- ⁇ superfamily.
  • the Lefty protein of the present invention shares sequence homology with the translation product of the murine mRNA for Lefty ( Figure 4; SEQ ID NO:6), including the conserved predicted active domain of about 110 amino acids.
  • Murine Lefty is thought to be important in left/right handedness of the developing organism.
  • the homology between murine Lefty and the novel human Lefty homologue of the present invention indicates that the novel human Lefty homologue of the present invention may also be involved in correct formation of various structures with respect to the rest of the developing organism or Lefty may also be involved in one or more post-developmental capacities including sexual development, pituitary hormone production, and the creation of bone and cartilage, as are many of the other members of the TGF- ⁇ superfamily.
  • Nodal and Lefty polypeptides of the present invention are useful for enhancing or enriching the growth and/or differentiation of specific cell populations, e.g., embryonic cells or stem cells.
  • compositions which contain a therapeutically effective amount of human Nodal and/or Lefty polypeptide, in a pharmaceutically acceptable vehicle or carrier.
  • These compositions of the invention may be useful in the therapeutic modulation or diagnosis of bone, cartilage, or other connective cell or tissue growth and/or differentiation.
  • These compositions may be used to treat such conditions as osteoarthritis, osteoporosis, and other abnormalities of bone, cartilage, muscle, tendon, ligament and/or other connective tissues and/or organs such as liver, lung, cardiac, pancreas, kidney, and other tissues.
  • compositions may also be useful in the growth and/or formation of cartilage, tendon, ligament, meniscus, and other connective tissues or any combination of the above (e.g., therapeutic modulation of the tendon-to-bone attachment apparatus). These compositions may also be useful in treating periodontal disease and modulating wound healing and tissue repair of such tissues as epidermis, nerve, muscle, cardiac muscle, liver, lung, cardiac, pancreas, kidney, and other tissues and/or organs.
  • Pharmaceutical compositions containing Nodal and/or Lefty of the invention may include one or more other therapeutically useful component such as BMP-1, BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, and/or BMP-7 (See, for example, U. S. Patent Nos.
  • BMP-8 See, for example, PCT publication WO91/18098
  • BMP-9 See, for example, PCT publication WO93/00432
  • BMP- 10 See, for example, PCT publication WO94/26893
  • BMP-11 See, for example, PCT publication WO94/26892
  • BMP-12 and/or BMP-13 See, for example, PCT publication WO95/16035
  • BIP growth and differentiation factors
  • GDFs growth and differentiation factors
  • VGR-2 epidermal growth factor
  • FGF fibroblast growth factor
  • TGF-alpha TGF-beta
  • IGF insulin-like growth factor
  • the encoded Lefty polypeptide has a predicted leader sequence of 18 amino acids underlined in Figure 2A; and the amino acid sequence of the predicted secreted Lefty protein is also shown in Figures 2A-B, as amino acid residues 19-366 and as residues 1-348 in SEQ ID NO:4.
  • one embodiment of the invention provides an isolated nucleic acid molecule comprising a polynucleotide having a nucleotide sequence selected from the group consisting of: (a) a nucleotide sequence encoding the Nodal polypeptide having the complete amino acid sequence in SEQ ID NO:2 (i.e., positions 1 to 283 of SEQ ID NO:2); (b) a nucleotide sequence encoding the predicted active Nodal polypeptide having the amino acid sequence at positions 173 to 283 of SEQ ID NO:2; (c) a nucleotide sequence encoding the Nodal polypeptide having the complete amino acid sequence encoded by the cDNA clone contained in ATCC Deposit No.
  • Another embodiment of the invention provides an isolated nucleic acid molecule comprising a polynucleotide having a nucleotide sequence selected from the group consisting of: (a) a nucleotide sequence encoding the Lefty polypeptide having the complete amino acid sequence in SEQ ID NO:4 (i.e., positions -18 to 348 of SEQ ID NO:4); (b) a nucleotide sequence encoding the Lefty polypeptide having the complete amino acid sequence in SEQ ID NO:4 excepting the N-terminal methionine (i.e., positions -17 to 348 of SEQ ID NO:4); (c) a nucleotide sequence encoding the predicted active domain of the Lefty polypeptide having the amino acid sequence at positions 60 to 348 of SEQ ID NO:4; (d) a nucleotide sequence encoding the predicted active domain of the Lefty polypeptide having the amino acid sequence at positions 118 to 348 of SEQ ID NO:4; (e) a nu
  • nucleic acid molecules that comprise a polynucleotide having a nucleotide sequence at least 90% identical, and more preferably at least 95%, 96%, 97%, 98% or 99% identical, to any of the nucleotide sequences in (a), (b), (c), (d) or (e), above, with regard to Nodal, to any of the nucleotide sequences in (a), (b), (c), (d), (e), (f), (g), (h) or (i), above, with regard to Lefty, or a polynucleotide which hybridizes, preferably under stringent hybridization conditions, to a polynucleotide in (a), (b), (c), (d) or (e), above, with regard to Nodal, or any of the nucleotide sequences in (a), (b), (c), (d), (e), (f), (g), (h) or (i), above, with regard to Lefty, listed above
  • An additional nucleic acid embodiment of the invention relates to an isolated nucleic acid molecule comprising a polynucleotide which encodes the amino acid sequence of an epitope-bearing portion of a Nodal polypeptide having an amino acid sequence in (a), (b), (c), (d) or (e), with regard to Nodal, above.
  • a further nucleic acid embodiment of the invention relates to an isolated nucleic acid molecule comprising a polynucleotide which encodes the amino acid sequence of an epitope-bearing portion of a Lefty polypeptide having an amino acid sequence in (a), (b), (c), (d), (e), (f), (g), (h) or (i), with regard to Lefty, above.
  • a further embodiment of the invention relates to an isolated nucleic acid molecule comprising a polynucleotide which encodes the amino acid sequences of a Nodal or Lefty polypeptide having an amino acid sequence which contains at least one amino acid substitution, but not more than 50 amino acid substitutions, even more preferably, not more than 40 amino acid substitutions, still more preferably, not more than 30 amino acid substitutions, and still even more preferably, not more than 20 amino acid substitutions.
  • a polynucleotide which encodes the amino acid sequence of a Nodal or Lefty polypeptide to have an amino acid sequence which contains not more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid substitutions. Conservative substitutions are preferable.
  • the present invention also relates to recombinant vectors, which include the isolated nucleic acid molecules of the present invention, and to host cells containing the recombinant vectors, as well as to methods of making such vectors and host cells and for using them for production of Nodal or Lefty polypeptides or peptides by recombinant techniques.
  • a process for producing such polypeptide by recombinant techniques comprising culturing recombinant prokaryotic and/or eukaryotic host cells, containing a human Nodal or Lefty nucleic acid sequence, under conditions promoting expression of said protein and subsequent recovery of said protein.
  • the invention further provides an isolated Nodal or Lefty polypeptide comprising an amino acid sequence selected from the group consisting of: (a) the amino acid sequence of the full-length Nodal polypeptide having the complete amino acid sequence shown in SEQ ID NO:2 (i.e., positions 1 to 283 of SEQ ID NO:2); (b) the amino acid sequence of the predicted active Nodal polypeptide having the amino acid sequence at positions 173 to 283 of SEQ ID NO:2; (c) the amino acid sequence of the Nodal polypeptide having the complete amino acid sequence encoded by the cDNA clone contained in ATCC Deposit No.
  • polypeptides of the present invention also include polypeptides having an amino acid sequence at least 80% identical, more preferably at least 90% identical, and still more preferably 95%, 96%, 97%, 98% or 99% identical to those described in (a) through (1) above, as well as polypeptides having an amino acid sequence with at least 90% similarity, and more preferably at least 95% similarity, to those above.
  • An additional embodiment of the invention relates to a peptide or polypeptide which comprises the amino acid sequence of an epitope-bearing portion of a Nodal or Lefty polypeptide having an amino acid sequence described in (a) through (1), above.
  • Peptides or polypeptides having the amino acid sequence of an epitope-bearing portion of a Nodal or Lefty polypeptide of the invention include portions of such polypeptides with at least six or seven, preferably at least nine, and more preferably at least about 30 amino acids to about 50 amino acids, although epitope-bearing polypeptides of any length up to and including the entire amino acid sequence of a polypeptide of the invention described above also are included in the invention.
  • a further embodiment of the invention relates to a polypeptide which comprises the amino acid sequence of a Nodal or Lefty polypeptide having an amino acid sequence which contains at least one amino acid substitution, but not more than 50 amino acid substitutions, even more preferably, not more than 40 amino acid substitutions, still more preferably, not more than 30 amino acid substitutions, and still even more preferably, not more than 20 amino acid substitutions.
  • a peptide or polypeptide it is highly preferable for a peptide or polypeptide to have an amino acid sequence which comprises the amino acid sequence of a TNF-gamma polypeptide, which contains at least one, but not more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 amino acid substitutions.
  • the number of additions, substitutions, and/or deletions in the amino acid sequence of Figures IA and IB, Figures 2A and 2B, or fragments thereof is 1-5, 5-10, 5-25, 5-50, 10-50 or 50-150, conservative amino acid substitutions are preferable.
  • the invention provides an isolated antibody that binds specifically to a Nodal and Lefty polypeptide having an amino acid sequence described in (a) through (1) above.
  • the invention further provides methods for isolating antibodies that bind specifically to a Nodal or Lefty polypeptide having an amino acid sequence as described herein. Such antibodies are useful diagnostically or therapeutically as described below.
  • the invention also provides for pharmaceutical compositions comprising
  • Nodal and Lefty polypeptides particularly human Nodal and Lefty polypeptides, which may be employed, for instance, to treat cellular growth and differentiation disorders. Methods of treating individuals in need of Nodal and Lefty polypeptides are also provided.
  • the invention further provides compositions comprising a Nodal or Lefty polynucleotide or a Nodal or Lefty polypeptide for administration to cells in vitro, to cells ex vivo and to cells in vivo, or to a multicellular organism.
  • the compositions comprise a Nodal or Lefty polynucleotide for expression of a Nodal or Lefty polypeptide in a host organism for treatment of disease. Particularly preferred in this regard is expression in a human patient for treatment of a dysfunction associated with aberrant endogenous activity of Nodal or Lefty.
  • the present invention also provides a screening method for identifying compounds capable of enhancing or inhibiting a biological activities of the Nodal and Lefty polypeptides, which involves contacting a receptor which is enhanced by the Nodal or Lefty polypeptides with the candidate compound in the presence of a Nodal or Lefty polypeptide, assaying receptor activation in the presence of the candidate compound and of Nodal or Lefty polypeptide, and comparing the receptor activity to a standard level of activity, the standard being assayed when contact is made between the receptor and in the presence of the Nodal or Lefty polypeptide and the absence of the candidate compound
  • an increase in receptor activation over the standard indicates that the candidate compound is an agonist of Nodal or Lefty activity and a decrease in receptor activation compared to the standard indicates that the compound is an antagonist of Nodal or Lefty activity.
  • a screening assay for agonists and antagonists involves determining the effect a candidate compound has on Nodal or Lefty binding to a receptor.
  • the method involves contacting the receptor with a Nodal or Lefty polypeptide and a candidate compound and determining whether Nodal or Lefty polypeptide binding to the receptor is increased or decreased due to the presence of the candidate compound.
  • an increase in binding of Nodal or Lefty over the standard binding indicates that the candidate compound is an agonist of Nodal or Lefty binding activity and a decrease in Nodal or Lefty binding compared to the standard indicates that the compound is an antagonist of Nodal or Lefty binding activity.
  • nucleic acids of the invention are useful as hybridization probes for differential identification of the tissue(s) or cell type(s) present in a biological sample.
  • polypeptides and antibodies directed to those polypeptides are useful to provide immunological probes for differential identification of the tissue(s) or cell type(s).
  • Nodal or Lefty gene expression may be detected in certain tissues (e.g., cancerous and wounded tissues) or bodily fluids (e.g., serum, plasma, urine, synovial fluid or spinal fluid) taken from an individual having such a disorder, relative to a "standard" Nodal or Lefty gene expression level, i.e., the Nodal and Lefty expression levels in healthy tissue from an individual not having the cell growth and differentiation disorder.
  • tissues e.g., cancerous and wounded tissues
  • bodily fluids e.g., serum, plasma, urine, synovial fluid or spinal fluid
  • the invention provides a diagnostic method useful during diagnosis of such a disorder, which involves: (a) assaying Nodal and Lefty gene expression level in cells or body fluid of an individual; (b) comparing the Nodal and Lefty gene expression levels with standard Nodal and Lefty gene expression levels, whereby an increase or decrease in the assayed Nodal and Lefty gene expression level compared to the standard expression level is indicative of disorder in the regulation of cell growth and differentiation.
  • An additional embodiment of the invention is related to a method for treating an individual in need of an increased level of Nodal or Lefty activity in the body comprising administering to such an individual a composition comprising a therapeutically effective amount of an isolated Nodal or Lefty polypeptide of the invention or an agonist thereof.
  • a still further embodiment of the invention is related to a method for treating an individual in need of a decreased level of Nodal or Lefty activity in the body comprising, administering to such an individual a composition comprising a therapeutically effective amount of a Nodal or Lefty antagonist.
  • Preferred antagonists for use in the present invention are Nodal- or Lefty-specific antibodies.
  • Figures IA and IB show the nucleotide sequence (SEQ ID NO:l) and deduced amino acid sequence (SEQ ID NO:2) of the human Nodal homologue of the present invention.
  • TGF- ⁇ consensus cleavage sequences (arginine-X-X- arginine (RXXR); where X is any amino acid) of the human Nodal homologue is double underlined in Figures IA and IB.
  • the TGF- ⁇ consensus cleavage sequence appears once in the amino acid sequence of Nodal.
  • Cleavage of the precursor form of human Nodal is predicted to occur immediately after the C-terminal arginine in the abovementioned consensus sequence in the amino acid sequence of Nodal.
  • N-linked glycosylation sites are marked in Figures IA and IB with a bolded asparagine symbol (N) in the Nodal amino acid sequence and a bolded pound sign (#) above the first nucleotide encoding that asparagine residue in the Nodal nucleotide sequence.
  • Potential N-linked glycosylation sequences are found at the following locations in the Nodal amino acid sequence: N-8 through F-l l (N-8, W-9, T-10, F-l l) and N-135 through Q-138 (N-135, L-136, S-137, Q-138).
  • a potential Protein Kinase C (PKC) phosphorylation site is also marked in Figures 1 A and IB with a bolded serine symbol (S) in the Nodal amino acid sequence and an asterisk (*) above the first nucleotide encoding that serine residue in the Nodal nucleotide sequence.
  • S serine symbol
  • * asterisk
  • the potential PKC phosphorylation sequence is found in the Nodal amino acid sequence from residue S-155 through residue R-157 (S-155, W-156, R-157).
  • CK2 phosphorylation sites are also marked in Figures IA and IB with a bolded serine symbol (S) in the Nodal amino acid sequence and an asterisk (*) above the first nucleotide encoding the appropriate serine residue in the Nodal nucleotide sequence.
  • Potential CK2 phosphorylation sequences are found at the following locations in the Nodal amino acid sequence: S-19 through E-22 (S-19, Q-20, Q-21, E-22); S-35 through D-38 (S-35, P-36, V-37, D-38); and S-63 through E-66 (S-63, C-64, L-65, E-66).
  • a potential myristylation site is found in the Nodal amino acid sequence in Figures IA and IB from residue G-6 through F-l l (G-6, Q-7, N-8, W-9, T-10, F-l l).
  • a potential amidation site is found in the Nodal amino acid sequence in Figures IA and IB from residue W-167 through R-170 (W-167, G-168, K-169, R-170).
  • a TGF-beta family signature is found in the Nodal amino acid sequence in Figures 1 A and IB from residue 1-201 through C-216 (1-201, 1-202, Y-203, P-204, K-205, Q-206, Y-207, N-208, A-209, Y-210, R-211, C-212, E-213, G-214, E-215, C-216).
  • This sequence is denoted in Figures IA and IB with a dotted underline shown under the amino acid sequence from residue 1-201 through C-216.
  • Figures 2A and 2B show the nucleotide sequence (SEQ ID NO:3) and deduced amino acid sequence (SEQ ID NO:4) of the Lefty homologue of the present invention.
  • the predicted leader cleavage sequence of the human Lefty homologue of about 18 amino acids is underlined in Figure 2 A. Note that the methionine residue at the beginning of the leader sequence in Figure 2A is shown in position number (positive or "+") 1, whereas the leader positions in the corresponding sequence of SEQ ID NO:2 are designated with negative position numbers. Thus, the leader sequence positions 1 to 18 in Figure 2A correspond to positions -18 to -1 in SEQ ID NO:2.
  • the predicted consensus sequences (arginine-X-X-arginine (RXXR); where X is any amino acid) of the human Lefty homologue is double underlined in Figures 2 A and 2B.
  • the TGF- ⁇ consensus cleavage sequence appears three times in the amino acid sequence of Lefty. Cleavage of the precursor forms of human Lefty is predicted to occur immediately after the C-terminal arginine in the abovementioned consensus sequence in the amino acid sequence of Lefty.
  • a potential asparagine-linked glycosylation site is marked in Figures 2A and 2B with a bolded asparagine symbol (N) in the Nodal amino acid sequence and a bolded pound sign (#) above the first nucleotide encoding that asparagine residue in the Lefty nucleotide sequence.
  • the potential N-linked glycosylation sequence is found in the Lefty amino acid sequence from residue N-158 through S-161 (N-158, R-159, T-160, S-161).
  • a potential cAMP- and cGMP-dependent protein kinase (CPK) phosphorylation site is marked in Figures 2A and 2B with a bolded serine symbol (S) in the Lefty amino acid sequence and an asterisk (*) above the first nucleotide encoding that serine residue in the Lefty nucleotide sequence.
  • the potential CPK phosphorylation sequence is found in the Lefty amino acid sequence from residue K-76 through residue S-79 (K-76, R-77, F-78, S-79).
  • PKC Protein Kinase C
  • the potential PKC phosphorylation sequences are found in the Lefty amino acid sequence from residue S-81 through residue R-83 (S-81, F-82, R-83); S-137 through R-139 (S-137, P-138, R-139); S-140 through R-142 (S-140, A-141, R-142); S-157 through R-159 (S-157, N-158, R-159); T-296 through R-298 (T-296, C-297, R-298); and S-329 through K-331 (S-329, 1-330, K-331).
  • CK2 phosphorylation sites are also marked in Figures 2A and 2B with a bolded serine symbol (S) in the Nodal amino acid sequence and an asterisk (*) above the first nucleotide encoding the appropriate serine residue in the Lefty nucleotide sequence.
  • CK2 phosphorylation sequences are found at the following locations in the Lefty amino acid sequence: S-68 through D-71 (S-68, H-69, G-70, D-71); S-81 through E-84 (S-81, F-82, R-83, E-84); S-161 through D-164 (S-161, L-162, 1-163, D-164); S-169 through E-172 (S-169, V-170, H-171, E-172); S-319 through D-322 (S-319, E-320, T-321, D-322); and S-329 through E-332 (S-329, 1-330, K-331, E-332).
  • a potential amidation site is found in the Lefty amino acid sequence in Figures 2 A and 2B from residue R-74 through R-77 (R-74, G-75, K-76, R-77).
  • a TGF-beta family signature is found in the Lefty amino acid sequence in Figures 2A and 2B from residue V-282 through C-297 (V-282, L-283, E-284, P-285, P-286, G-287, F-288, L-289, A-290, Y-291, E-292, C-293, V-294, G-295, T-296, C-297).
  • This sequence is denoted in Figures 2 A and 2B with a dotted underline shown under the amino acid sequence from residue 1-282 through C-297.
  • Figures 3 and 4 show the regions of identity between the amino acid sequences of the Nodal and Lefty proteins and translation product of the murine mRNAs for Nodal and Lefty, respectively, (SEQ ID NO:5 and SEQ ID NO:6, respectively), determined by the computer program Bestfit (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 5371 1) using the default parameters.
  • Figures 5 and 6 show computer analyses of the Nodal and Lefty amino acid sequences depicted in Figures IA and IB (SEQ ID NO:2) and 2 A and 2B (SEQ ID NO:4), respectively.
  • Alpha, beta, turn and coil regions; hydrophilicity and hydrophobicity; amphipathic regions; flexible regions; antigenic index and surface probability, as predicted using the default parameters of the recited programs, are shown.
  • the positive peaks indicate locations of the highly antigenic regions of the Nodal and Lefty proteins, i.e., regions from which epitope-bearing peptides of the invention can be obtained.
  • Non-limiting examples of antigenic polypeptides or peptides that can be used to generate Nodal-specific antibodies include: a polypeptide comprising amino acid residues from about Lys-54 to about Asp-62, from about Val-91 to about Leu-99, from about Lys-100 to about Gln-108, from about Cys-1 16 to about Pro-124, from about Gln-140 to about Leu-148, from about Trp- 156 to about Ser- 164, from about Arg- 170, to about Gln-181, from about Cys-212 to about Phe-224, from about Tyr-239, to about Thr-247, from about Pro-251, to about Met-259, and from about Asp-263, to about His-271.
  • Non-limiting examples of antigenic polypeptides or peptides that can be used to generate Lefty-specific antibodies include: a polypeptide comprising amino acid residues from about Asp-71 to about Ser-79, from about Arg- 106 to about Val-114, from about Leu-136 to about Arg-144, from about Asp-154 to about Asp- 164, from about His-171 to about Asp- 179, from about Gin- 189 to about Leu-197, from about Pro-227 to about Glu-236, from about Gly-246 to about Glu-254, from about Pro-256 to about Gln-266, from about Cys-297 to about Ala-305, from about Ile-317 to about Pro-325, from about Ile-330 to about Val-340, and from about Val-348 to about Pro-366.
  • the present invention provides isolated nucleic acid molecules comprising polynucleotides encoding a Nodal or Lefty polypeptide having the amino acid sequences shown in SEQ ID NO:2 and SEQ ID NO:4, respectively, which were determined by sequencing cloned cDNAs.
  • the nucleotide sequences shown in Figures IA and B and 2A and B (SEQ ID NO:l and SEQ ID NO:3, respectively) were obtained by sequencing the HNGEF08 and HUKEJ46 clones, which were deposited on June 5, 1997 at the American Type Culture Collection, 10801 University Boulevard, Manassas, Virginia 201 10-2209, and given accession numbers ATCC 209092 and 209135, and 209091, respectively.
  • the deposited clones are contained in the pBluescript SK(-) plasmid (Stratagene, La Jolla, CA).
  • the Nodal and Lefty proteins of the present invention share sequence homology with the translation products of the murine mRNAs for Nodal and Lefty ( Figures 3 and 4).
  • Murine Nodal is thought to be an important TGF- ⁇ superfamily member involved in mesoderm formation during gastrulation (Zhou, X., et al, Nature 361:543-547 (1993)).
  • gastrulation the three germ layers of the embryo are formed and organized along the anterior-posterior body axis.
  • ectodermal cells of the primitive streak differentiate into the mesoderm.
  • Murine Nodal was identified in mice which were homozygously mutated in the Nodal gene. A mutation in Nodal is prenatally lethal presumably due to the resulting gross developmental abnormalities.
  • Murine Lefty is involved in the developmental processes which establish lateral symmetry or handedness of the maturing embryonic organism (Meno, C, et al, Nature 381:151-155 (1996)). Lefty is believed to be a diffusable morphogen, the expression of which may result in the initiation of determination of symmetrical development in the mouse embryo. Lefty is transiently expressed in the left half of the gastrulating embryo just before the initiation of lateral symmetry.
  • nucleotide sequences determined by sequencing a DNA molecule herein were determined using an automated DNA sequencer (such as the Model 373 from Applied Biosystems, Inc., Foster City,
  • nucleotide sequences determined herein were predicted by translation of a DNA sequence determined as above. Therefore, as is known in the art for any DNA sequence determined by this automated approach, any nucleotide sequence determined herein may contain some errors. Nucleotide sequences determined by automation are typically at least about 90% identical, more typically at least about 95% to at least about
  • a single insertion or deletion in a determined nucleotide sequence compared to the actual sequence will cause a frame shift in translation of the nucleotide sequence such that the predicted amino acid sequence encoded by a determined nucleotide sequence will be completely different from the amino acid sequence actually encoded by the sequenced DNA molecule, beginning at the point of such an insertion or deletion.
  • nucleotide sequence of a nucleic acid molecule or polynucleotide is intended, for a DNA molecule or polynucleotide, a sequence of deoxyribonucleotides, and for an RNA molecule or polynucleotide, the corresponding sequence of ribonucleotides (A, G, C and U), where each thymidine deoxyribonucleotide (T) in the specified deoxyribonucleotide sequence is replaced by the ribonucleotide uridine (U).
  • nucleic acid molecules of the present invention encoding a Nodal and Lefty polypeptide may be obtained using standard cloning and screening procedures, such as those for cloning cDNAs using mRNA as starting material.
  • nucleic acid molecules described in Figures IA and B and 2 A and B were discovered in cDNA libraries derived from neutrophils and uterine cancer, respectively. An additional clone of the Nodal gene was found in testis tissue.
  • Additional clones of the Lefty gene were also identified in cDNA libraries from the following cell and tissue types: colon cancer, apoptotic T-cells, fetal heart, Wilm's Tumor tissue, frontal lobe of the brain from a patient with dementia, neutrophils, salivary gland, small intestine, 7, 8, and 12 week old human embryos, frontal cortex and hypothalamus from a patient with schizophrenia, brain from a patient with Alzheimer's Disease, adipose tissue, brown fat, TNF- and LPS-induced and uninduced bone marrow stroma, activated monocytes and macrophages, rhabdomyosarcoma, cycloheximide-treated Raji cells, breast lymph nodes, hemangiopericytoma, testes, fetal epithelium (skin), and IL-5-induced eosinophils.
  • Each of the determined nucleotide sequences of the Nodal and Lefty cDNAs shown in Figures IA and B and 2A and B contains an open reading frame.
  • the open reading frame found in Figures 1A-B encodes a protein of 283 amino acid residues, with an initiating aspartic acid codon at nucleotide positions 1-3 of the nucleotide sequence in Figure IA (SEQ ID NO:l), and a deduced molecular weight of about 32.5 kDa.
  • the open reading frame found in Figures 2A-B encodes a protein of 366 amino acid residues, with an initiating methionine codon at nucleotide positions 53-55 of the nucleotide sequence in Figure 2A (SEQ ID NO:3), and a deduced molecular weight of about 40.9 kDa.
  • the amino acid sequence of the Nodal and Lefty proteins shown in SEQ ID NO:2 and SEQ ID NO:4, respectively, is about 80.9% and 82.0% identical to the murine mRNAs for Nodal and Lefty, respectively ( Figures 3 and 4).
  • the open reading frame of the Nodal gene shares sequence homology with the translation product of the murine mRNA for Nodal; Figure 3; SEQ ID NO:3), particularly in the conserved active domain of about 110 amino acids.
  • the open reading frame of the Lefty gene shares sequence homology with the translation product of the murine mRNA for Lefty; Figure 4; SEQ ID NO:4), particularly in the conserved active domain of about 288 amino acids.
  • Murine Nodal is thought to be important in correct mesoderm formation in the developing mouse embryo.
  • Murine Lefty is thought to be important in the initiation of lateral a symmetry in the developing mouse embryo.
  • the homologies between the murine Nodal and Lefty mRNAs and the novel human homologues of Nodal and Lefty indicate that the novel human homologues of Nodal and Lefty are involved in developmental roles as well as in the regulation of cell growth and differentiation. Further, it is likely that aberrant expression of Nodal and Lefty is a characteristic of cancer.
  • novel human genes of the instant application also function in the regulation of immune and hematopoietic cell growth and differentiation.
  • the actual complete Nodal and Lefty polypeptides encoded by the deposited cDNAs which comprise about 283 and 348 amino acids, respectively, may be somewhat longer or shorter. More generally, the actual open reading frame may be anywhere in the range of ⁇ 20 amino acids, more likely in the range of ⁇ 10 amino acids, of that predicted from either the codon at the N-terminus shown in Figures IA and B and 2A and B (SEQ ID NO:l and SEQ ID NO:3, respectively). It will further be appreciated that, depending on the analytical criteria used for identifying various functional domains, the exact "address" of the active domains of the Nodal and Lefty polypeptides may differ slightly from the predicted positions above.
  • Methods for predicting whether a protein has a secretory leader as well as the cleavage point for that leader sequence are known in the art and may routinely be applied to identify the leader sequence of the polynucleotides of the invention.
  • McGeoch Virus Res. 3:271-286 (1985)
  • the method of von Heinje uses the information from the residues surrounding the cleavage site, typically residues -13 to +2 where +1 indicates the amino terminus of the mature protein.
  • the computation analysis above predicted a single N-terminal signal sequence within the complete amino acid sequence shown in SEQ ID NO:4.
  • the amino acid sequence of the complete Lefty protein includes a leader sequence and a mature protein, as shown in Figures 2A and 2B and SEQ ID NO:4.
  • the amino acid sequence of the complete Nodal protein predicts a leader sequence and a mature protein, by comparison to the full-length murine Nodal ORF as shown in Figure 3.
  • the present invention provides nucleic acid molecules encoding a mature form of the Lefty protein.
  • proteins secreted by mammalian cells have a signal or secretory leader sequence which is cleaved from the complete polypeptide to produce a secreted "mature" form of the protein.
  • Most mammalian cells and even insect cells cleave secreted proteins with the same specificity.
  • cleavage of a secreted protein is not entirely uniform, which results in two or more mature species of the protein.
  • the cleavage specificity of a secreted protein is ultimately determined by the primary structure of the complete protein, that is, it is inherent in the amino acid sequence of the polypeptide.
  • the present invention provides a nucleotide sequence encoding the mature Lefty polypeptide having the amino acid sequence encoded by the cDNA clone contained in the host identified as ATCC Deposit No. 209091.
  • the mature Lefty polypeptide having the amino acid sequence encoded by the cDNA clone in ATCC Deposit No. 209091 is meant the mature form(s) of the Lefty protein produced by expression in a mammalian cell (e.g., COS cells, as described below) of the complete open reading frame encoded by the human DNA sequence of the clone contained in the vector in the deposited host.
  • Nucleic acid molecules of the present invention may be in the form of RNA, such as mRNA, or in the form of DNA, including, for instance, cDNA and genomic DNA obtained by cloning or produced synthetically.
  • the DNA may be double-stranded or single-stranded.
  • Single-stranded DNA or RNA may be the coding strand, also known as the sense strand, or it may be the non-coding strand, also referred to as the anti-sense strand or complementary strand.
  • the polynucleotides of the invention are less than 300 kb, 200 kb, 100 kb, 50 kb, 15 kb, 10 kb or 7.5 kb in length.
  • polynucleotides of the invention comprise at least 15 contiguous nucleotides of Human Nodal or Human Lefty coding sequence, but do not comprise all or a portion of any Human Nodal or Human Lefty intron.
  • the nucleic acid comprising Human Nodal or Human Lefty coding sequence does not contain coding sequences of a genomic flanking gene (i.e., 5' or 3' to the Human Nodal or Human Lefty coding sequences in the genome).
  • isolated nucleic acid molecule(s) is intended a nucleic acid molecule, DNA or RNA, which has been removed from its native environment
  • recombinant DNA molecules contained in a vector are considered isolated for the purposes of the present invention.
  • Further examples of isolated DNA molecules include recombinant DNA molecules maintained in heterologous host cells or purified (partially or substantially) DNA molecules in solution.
  • a nucleic acid contained in a clone that is a member of a library e.g., a genomic or cDNA library
  • a chromosome preparation e.g., a chromosome spread
  • Isolated RNA molecules include in vivo or in vitro RNA transcripts of the DNA molecules of the present invention.
  • Isolated nucleic acid molecules according to the present invention further include such molecules produced synthetically.
  • Isolated nucleic acid molecules of the present invention include DNA molecules comprising an open reading frame (ORF) with an initiating codon at positions 1-3 of the nucleotide sequence shown in Figure 1 A (SEQ ID NO:l) and DNA molecules comprising an open reading frame (ORF) with an initiation codon at positions 53-55 of the nucleotide sequence shown in Figure 2A (SEQ ID NO:3).
  • DNA molecules comprising the coding sequence for the predicted mature Lefty protein shown at positions 1-366 of SEQ ID NO:4.
  • isolated nucleic acid molecules of the invention include DNA molecules which comprise a sequence substantially different from those described above, but, which, due to the degeneracy of the genetic code, still encode the Nodal or Lefty proteins.
  • the genetic code and species-specific codon preferences are well known in the art.
  • the invention provides isolated nucleic acid molecules encoding the Nodal and Lefty polypeptides having amino acid sequences encoded by the cDNA clones contained in the plasmid deposited as ATCC Deposit Nos. 209092 and 209091 on June 5, 1997 and the plasmid deposited as ATCC Deposit No. 209135 on July 2, 1997.
  • these nucleic acid molecules will encode the mature polypeptides encoded by the above-described deposited cDNA clones.
  • the invention further provides an isolated nucleic acid molecule having the nucleotide sequence shown in Figures 1A-B (SEQ ID NO:l) and an isolated nucleic acid molecule having the nucleotide sequence shown in Figures 2A-B (SEQ ID NO:3) or the nucleotide sequences of the Nodal and Lefty cDNAs contained in the above-described deposited clones, or a nucleic acid molecule having a sequence complementary to one of the above sequences.
  • Such isolated molecules, particularly DNA molecules are useful as probes for gene mapping, by in situ hybridization with chromosomes, and for detecting expression of the Nodal and Lefty genes in human tissue, for instance, by Northern blot analysis.
  • the present invention is further directed to nucleic acid molecules encoding portions of the nucleotide sequences described herein as well as to fragments of the isolated nucleic acid molecules described herein.
  • the invention provides a polynucleotide having a nucleotide sequence representing the portion of SEQ ID NO:l which consists of positions 1-852 of SEQ ID NO:l and a polynucleotide having a nucleotide sequence representing the portion of SEQ ID NO:3 which consists of positions 1-1153 of SEQ ID NO:3.
  • fragments of an isolated nucleic acid molecule having the nucleotide sequence of the deposited cDNAs (HTLFA20, HNGEF08, and HUKEJ46), or the nucleotide sequence shown in Figures IA and B (SEQ ID NO:l), Figures 2 A and B (SEQ ID NO:3), or the complementary strand thereto is intended fragments at least 15 nt, and more preferably at least 20 nt, still more preferably at least 25 or 30 nt, and even more preferably, at least 40, 50, 60, 70, 80, 90, 100, 150, 200, 250, 300, 400, or 500 nt in length.
  • fragments have numerous uses which include, but are not limited to, diagnostic probes and primers as discussed herein.
  • larger fragments 50-1500 nt in length are also useful according to the present invention as are fragments corresponding to most, if not all, of the nucleotide sequence of the deposited cDNA clone HTLFA20, the deposited cDNA clone HNGEF08, the deposited cDNA clone HUKEJ46, the nucleotide sequence depicted in Figures IA and B (SEQ ID NO:l), or the nucleotide sequence depicted in Figures 2 A and B (SEQ ID NO:4).
  • fragments at least 20 nt in length are intended fragments which include 20 or more contiguous bases from the nucleotide sequence of the deposited cDNA clones (HTLFA20, HNGEF08, and HUKEJ46), the nucleotide sequence as shown in Figures 1 A and B (SEQ ID NO:l) or the nucleotide sequence as shown in Figures 2 A and B (SEQ ID NO:4).
  • No. 209091 which encodes the Human Lefty Homologue of the present invention, contains a cDNA insert which is represented by nucleotides 1-1596 of the sequence shown in Figures 2 A and 2B.
  • the invention provides nucleic acid molecules having nucleotide sequences related to extensive portions of SEQ ID NO: 3 which have been determined from the following related cDNA clones: HUKFN65R (SEQ ID NO:7) and HUKEJ46R (SEQ ID NO:8).
  • the invention includes a polynucleotide comprising any portion of at least about 30 nucleotides, preferably at least about 50 nucleotides, of SEQ ID NO:l from nucleotide 1-1130. More preferably, the invention includes a polynucleotide comprising nucleotides 250-1130, 500-1130, 750-1130, 1000-1130, 1-1000, 250-1000, 500-1000, 750-1000, 1-750, 250-750, 500-750, 1-500, 250-500, and 1-250 of SEQ ID NO:l .
  • the invention includes a polynucleotide comprising any portion of at least about 30 nucleotides, preferably at least about 50 nucleotides, of SEQ ID NO: 3 from residue 1 to 950 and 1150 to 1688. More preferably, the invention includes a polynucleotide comprising nucleotides 250-1688, 500-1688, 750-1688, 1000-1688, 1250-1688, 1500-1688, 1-1500, 250-1500, 500-1500, 750-1500, 1000-1500, 1250-1500, 1-1250, 250-1250, 500-1250, 750-1250, 1000-1250, 1-1000, 250-1000, 500-1000, 750-1000, 1-750, 250-750, 500-750, 1-500, and 250-500 of SEQ ID NO:3.
  • nucleotides corresponding to nucleotides 1-125, 1-90, 1-60, 1-30, 30-125, 30-90, 30-60, 60-125, 60-90, 90-125, 310-930, 350-930, 400-930, 450-930, 500-930, 550-930, 600-930, 650-930, 700-930, 750-930, 800-930, 850-930, 900-930, 310-900, 350-900, 400-900, 450-900, 500-900, 550-900, 600-900, 650-900, 700-900, 750-900, 800-900, 850-900, 310-850, 350-850, 400-850, 450-850, 500-850, 550-850, 600-850, 650-850, 700-850, 750-850, 800-850, 310-800, 350-800, 400-850, 450-850, 500-850, 550-850, 600-850, 650-850, 700-850, 750-850, 800-850,
  • fragments of an isolated nucleic acid molecule having the nucleotide sequence of the deposited cDNAs or the nucleotide sequences shown in Figures IA and B and 2A and B are intended fragments at least about 15 nt, and more preferably at least about 20 nt, still more preferably at least about 25 nt or about 30 nt, and even more preferably, at least about 40 nt or about 45 nt in length which are useful as diagnostic probes and primers as discussed herein.
  • fragments 50-300 nt in length are also useful according to the present invention as are fragments corresponding to most, if not all, of the nucleotide sequence of the deposited cDNAs or as shown in Figures IA and B and 2A and B (SEQ ID NO:l and SEQ ID NO:3, respectively).
  • a fragment at least 20 nt in length for example, is intended fragments which include 20 or more contiguous bases from the nucleotide sequences of the deposited cDNAs or the nucleotide sequences as shown in Figures IA and B and 2A and B (SEQ ID NO:l and SEQ ID NO:3, respectively).
  • nucleic acid fragments of the present invention include nucleic acid molecules encoding epitope-bearing portions of the Nodal and Lefty polypeptides as identified in Figures 5 and 6 and described in more detail below.
  • the polynucleotide fragments of the invention encode a polypeptide which demonstrates a functional activity.
  • a polypeptide demonstrating "functional activity” is meant, a polypeptide capable of displaying one or more known functional activities associated with a complete, mature or TGF- ⁇ -like active forms of the Nodal or Lefty polypeptides.
  • Such functional activities include, but are not limited to, biological activity ((e.g., the modulation of growth, development, and differentiation of a number of cell, tissue, and organ types (e.g., fibroblasts, keratinocytes, T- and B-lymphocytes, bone, cartilage, and other connective tissues, kidney, lung, and heart)), antigenicity [ability to bind (or compete with a Nodal or Lefty polypeptide for binding) to an anti-Nodal or anti-Lefty antibody], immunogenicity (ability to generate antibody which binds to a Nodal or Lefty polypeptide), the ability to form polymers (e.g., dimers) with other Nodal or Lefty or TGF- ⁇ polypeptides, and ability to bind to a receptor or ligand for a Nodal or Lefty polypeptide.
  • biological activity e.g., the modulation of growth, development, and differentiation of a number of cell, tissue, and organ types (e.g.,
  • Preferred nucleic acid fragments of the present invention also include nucleic acid molecules encoding one or more of the following domains of Nodal: amino acid residues 174-283 of SEQ ID NO:2 (i.e., the TGF- ⁇ -like domain of Nodal) and amino acid residues 1-27, 30-58, 64-82, 85-110, and 130-283 of SEQ ID NO:2.
  • Preferred nucleic acid fragments of the present invention also include nucleic acid molecules encoding one or more of the following domains of Lefty: amino acid residues 1-348 of SEQ ID NO:4 (i.e., the mature domain of Lefty), amino acid residues 60-348 of SEQ ID NO:4 (i.e., the first predicted TGF- ⁇ -like domain of Lefty), amino acid residues 118-348 of SEQ ID NO:4 (i.e., the second predicted TGF- ⁇ -like domain of Lefty), amino acid residues 125-348 of SEQ ID NO:4 (i.e., the third predicted TGF- ⁇ -like domain of Lefty), and (-15)-(-2), 3-19, 34-51, 54-72, 75-114, 117-192, 198-209, 211-286, 290-302, and 305-348 of SEQ ID NO:4.
  • the polynucleotide fragments of the invention encode antigenic regions.
  • antigenic polypeptides or peptides that can be used to generate Nodal-specific antibodies include: a polypeptide comprising amino acid residues from about Lys-54 to about Asp-62, from about Val-91 to about Leu-99, from about Lys-100 to about Gln-108, from about Cys-116 to about Pro- 124, from about Gin- 140 to about Leu- 148, from about Trp- 156 to about Ser- 164, from about Arg- 170, to about Gln-181, from about Cys-212 to about Phe-224, from about Tyr-239, to about Thr-247, from about Pro-251, to about Met-259, and from about Asp-263, to about His-271.
  • Non-limiting examples of antigenic polypeptides or peptides that can be used to generate Lefty-specific antibodies include: a polypeptide comprising amino acid residues from about Asp-71 to about Ser-79, from about Arg-106 to about Val-114, from about Leu-136 to about Arg-144, from about Asp-154 to about Asp- 164, from about His-171 to about Asp- 179, from about Gin- 189 to about Leu-197, from about Pro-227 to about Glu-236, from about Gly-246 to about Glu-254, from about Pro-256 to about Gln-266, from about Cys-297 to about Ala-305, from about Ile-317 to about Pro-325, from about Ile-330 to about Val-340, and from about Val-348 to about Pro-366.
  • polynucleotide fragments of the invention encode functional attributes of Human Nodal or Human Lefty.
  • Preferred embodiments of the invention in this regard include fragments that comprise alpha-helix and alpha-helix forming regions ("alpha-regions"), beta-sheet and beta-sheet forming regions ("beta-regions"), turn and turn-forming regions ("turn-regions”), coil and coil-forming regions ("coil-regions”), hydrophilic regions, hydrophobic regions, alpha amphipathic regions, beta amphipathic regions, flexible regions, surface-forming regions and high antigenic index regions of Human Nodal or Human Lefty.
  • the data presented in columns VIII, IX, XIII, and XIV of Tables I and II can be used to determine regions of Nodal or Lefty which exhibit a high degree of potential for antigenicity. Regions of high antigenicity are determined from the data presented in columns VIII, IX, XIII, and/or IV by choosing values which represent regions of the polypeptide which are likely to be exposed on the surface of the polypeptide in an environment in which antigen recognition may occur in the process of initiation of an immune response.
  • the above-mentioned preferred regions set out in Figures 5 and 6 and in Tables I and II include, but are not limited to, regions of the aforementioned types identified by analysis of the amino acid sequence set out in Figures IA and B and 2A and B.
  • such preferred regions include Garnier-Robson alpha-regions, beta-regions, turn-regions, and coil-regions, Chou-Fasman alpha-regions, beta-regions, and coil-regions, Kyte-Doolittle hydrophilic regions and hydrophobic regions, Eisenberg alpha- and beta-amphipathic regions, Karplus-Schulz flexible regions, Emini surface-forming regions and Jameson-Wolf regions of high antigenic index (generated using the amino acid sequences set out in Figures 1 and 2, and using the default parameters of the recited computer programs).
  • Trp 26 A A 0 52 0 29 -0 30 029
  • Trp 150 A A 009 027 0 30 0 50
  • Trp 5 A T -0 77 1 60 -0 20 0 14 Leu 6 A B -0 98 1 70 -0 60 0 09
  • Trp 11 A B -1 83 1 77 -0 60 0 19 Val 12 A B -1 76 1 54 -0 60 0 16
  • Trp 175 A T 1 29 0 01 F 1 37 0 56
  • Lys 276 A A 091 -0 14 * 0 30 0 83
  • Trp 277 A A 1 43 -0 14 * 0 45 1 34
  • Leu 304 A A 0 14 -0 10 0 60 0 58
  • Trp 308 A B -0 91 1 10 * -0 60 046
  • fragments in this regard are those that comprise regions of Human Nodal or Human Lefty that combine several structural features, such as, two, three, four, five or more of the features set out above.
  • the invention provides isolated nucleic acid molecules comprising polynucleotides which hybridize under stringent hybridization conditions to a portion of the polynucleotide in a nucleic acid molecule of the inventions described above, for instance, the cDNA clones contained in ATCC Deposit Nos. 209092, 209135, and 209091 and/or a polynucleotide fragment described above.
  • stringent hybridization conditions is intended overnight incubation at 42°C in a solution comprising: 50% formamide, 5x SSC (750 mM NaCl, 75 mM trisodium citrate), 50 mM sodium phosphate (pH 7.6), 5x Denhardt's solution, 10% dextran sulfate, and 20 ⁇ g/ml denatured, sheared salmon sperm DNA, followed by washing the filters in O.lx SSC at about 65°C.
  • nucleotides corresponding to nucleotides 1-125, 1-90, 1-60, 1-30, 30-125, 30-90, 30-60, 60-125, 60-90, 90-125, 310-930, 350-930, 400-930, 450-930, 500-930, 550-930, 600-930, 650-930, 700-930, 750-930, 800-930, 850-930, 900-930, 310-900, 350-900, 400-900, 450-900, 500-900, 550-900, 600-900, 650-900, 700-900, 750-900, 800-900, 850-900, 310-850, 350-850, 400-850, 450-850, 500-850, 550-850, 600-850, 650-850, 700-850, 750-850, 800-850, 310-800, 350-800, 400-850, 450-850, 500-850, 550-850, 600-850, 650-850, 700-850, 750-850, 800-850,
  • a polynucleotide which hybridizes to a "portion" of a polynucleotide is intended a polynucleotide (either DNA or RNA) hybridizing to at least about 15 nucleotides (nt), and more preferably at least about 20 nt, still more preferably at least about 30 nt, and even more preferably about 30-70 (e.g., 50) nt of the reference polynucleotide. These are useful as diagnostic probes and primers as discussed above and in more detail below.
  • a polynucleotide which hybridizes only to a poly A sequence such as the 3' terminal poly(A) tract of the Nodal and Lefty cDNAs shown in Figures IA and B and 2A and B (SEQ ID NO:l and SEQ ID NO:3, respectively)
  • a polynucleotide of the invention used to hybridize to a portion of a nucleic acid of the invention, since such a polynucleotide would hybridize to any nucleic acid molecule containing a poly (A) stretch or the complement thereof (e.g., practically any double-stranded cDNA clone generated using oligo dT as a primer).
  • polynucleotides which hybridize to the reference polynucleotides disclosed herein encode polypeptides which either retain substantially the same biological function or activity as the mature form or TGF- ⁇ -like active form of the Nodal polypeptide encoded by the polynucleotide sequences depicted in Figures IA and IB (SEQ ID NO:l) and/or substantially the same biological function or activity as the mature form or TGF- ⁇ -like active forms of the Lefty polypeptide encoded by the polynucleotide sequences depicted in Figures 2A and 2B (SEQ ID NO: l) depicted in Figures 2A and 2B (SEQ ID NO:3), or the cDNAs contained in the deposit (HTLFA20, HNGEF08, and HUKEJ46).
  • Alternative embodiments are directed to polynucleotides which hybridize to the reference polynucleotide (i.e., a polynucleotide sequence disclosed herein), but do not retain biological activity. While these polynucleotides do not retain biological activity, they have uses, such as, for example, as probes for the polynucleotides of SEQ ID NO:l or SEQ ID NO:3, for recovery of the polynucleotides, as diagnostic probes, and as PCR primers.
  • nucleic acid molecules of the present invention which encode a Lefty polypeptide may include, but are not limited to those encoding the amino acid sequence of the mature form of the polypeptide, by itself; and the coding sequence for the mature form of the polypeptide and additional sequences, such as those encoding the about 18 amino acid leader or secretory sequence, such as a pre-, or pro- or prepro- protein sequence; the coding sequence of the mature polypeptide, with or without the aforementioned additional coding sequences.
  • nucleic acid molecules of the present invention which encode a Nodal polypeptide may include, but are not limited to, those encoding the amino acid sequence of the complete polypeptide, by itself; and the coding sequence for the complete polypeptide and additional sequences, such as those encoding an added secretory leader sequence, such as a pre-, or pro- or prepro- protein sequence.
  • nucleic acids of the invention are the above protein sequences together with additional, non-coding sequences, including for example, but not limited to introns and non-coding 5' and 3' sequences, such as the transcribed, non-translated sequences that play a role in transcription, mRNA processing, including splicing and polyadenylation signals, for example - ribosome binding and stability of mRNA; an additional coding sequence which codes for additional amino acids, such as those which provide additional functionalities.
  • additional, non-coding sequences including for example, but not limited to introns and non-coding 5' and 3' sequences, such as the transcribed, non-translated sequences that play a role in transcription, mRNA processing, including splicing and polyadenylation signals, for example - ribosome binding and stability of mRNA; an additional coding sequence which codes for additional amino acids, such as those which provide additional functionalities.
  • the sequences encoding the polypeptides may be fused to a marker sequence, such as a sequence encoding a peptide which facilitates purification of the fused polypeptide.
  • the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311), among others, many of which are commercially available.
  • pQE vector QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, CA, 91311
  • hexa-histidine provides for convenient purification of the fusion protein.
  • the "HA” tag is another peptide useful for purification which corresponds to an epitope derived from the influenza hemagglutinin protein, which has been described by Wilson and coworkers (Cell 37:767 (1984)).
  • other such fusion proteins include the Nodal and Lefty fused to Fc at the N- or C-terminus.
  • the present invention further relates to variants of the nucleic acid molecules of the present invention, which encode portions, analogs or derivatives of the Nodal and Lefty proteins.
  • Variants may occur naturally, such as a natural allelic variant.
  • allelic variant is intended one of several alternate forms of a gene occupying a given locus on a chromosome of an organism (Genes II, Lewin, B., ed., John Wiley & Sons, New York (1985)).
  • Non-naturally occurring variants may be produced using art-known mutagenesis techniques.
  • variants include those produced by nucleotide substitutions, deletions or additions.
  • the substitutions, deletions or additions may involve one or more nucleotides.
  • the variants may be altered in coding regions, non-coding regions, or both. Alterations in the coding regions may produce conservative or non-conservative amino acid substitutions, deletions or additions. Especially preferred among these are silent substitutions, additions and deletions, which do not alter the properties and activities of the Nodal and Lefty proteins or portions thereof. Also especially preferred in this regard are conservative substitutions.
  • nucleic acid molecules encoding the mature form of the protein having the amino acid sequence shown in SEQ ID NO:4 or the mature Lefty amino acid sequence encoded by the deposited cDNA clone.
  • nucleic acid molecules encoding the active domain of the proteins having the amino acid sequence shown in SEQ ID NO: 2 or SEQ ID NO:4 or the active domains of the Nodal and Lefty amino acid sequences encoded by the deposited cDNA clones.
  • active domain is meant the C-terminal region of a Nodal or Lefty polypeptide, or fragment thereof, which has been processed either in vitro or in vivo such that the C-terminal region has been cleaved from the remainder of the molecule just C-terminal to one or more of the TGF- ⁇ cleavage consensus sites as indicated in Figures IA and IB and 2 A and 2B.
  • inventions include an isolated nucleic acid molecule comprising a polynucleotide having a nucleotide sequence at least 90% identical, and more preferably at least 95%, 96%, 97%, 98% or 99% identical to a polynucleotide selected from the group consisting of: (a) a nucleotide sequence encoding the Nodal polypeptide having the complete amino acid sequence in SEQ ID NO:2 (i.e., positions 1 to 283 of SEQ ID NO:2); (b) a nucleotide sequence encoding the predicted active Nodal polypeptide having the amino acid sequence at positions 173 to 283 of SEQ ID NO:2; (c) a nucleotide sequence encoding the Nodal polypeptide having the complete amino acid sequence encoded by the cDNA clone contained in ATCC Deposit No.
  • nucleic acid molecules that comprise a polynucleotide having a nucleotide sequence at least 90% identical, and more preferably at least 95%, 96%, 97%, 98% or 99% identical, to any of the nucleotide sequences in (a) through (m) above, or a polynucleotide which hybridizes under stringent hybridization conditions to a polynucleotide in (a) through (m) above.
  • This polynucleotide which hybridizes does not hybridize under stringent hybridization conditions to a polynucleotide having a nucleotide sequence consisting of only A residues or of only T residues.
  • An additional nucleic acid embodiment of the invention relates to an isolated nucleic acid molecule comprising a polynucleotide which encodes the amino acid sequence of an epitope-bearing portion of a Nodal and Lefty polypeptide having an amino acid sequence in (a) through (1) above.
  • a further nucleic acid embodiment of the invention relates to an isolated nucleic acid molecule comprising a polynucleotide which encodes the amino acid sequence of a Human Nodal or Human Lefty polypeptide having an amino acid sequence which contains at least one conservative amino acid substitution, but not more than 50 conservative amino acid substitutions, even more preferably, not more than 40 conservative amino acid substitutions, still more preferably not more than 30 conservative amino acid substitutions, and still even more preferably not more than 20 conservative amino acid substitutions.
  • a polynucleotide which encodes the amino acid sequence of a Human Nodal or Human Lefty polypeptide to have an amino acid sequence which contains not more than 7-10, 5-10, 3-7, 3- 5, 2-5, 1-5, 1-3, 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 conservative amino acid substitutions.
  • nucleotide sequence having a nucleotide sequence at least, for example, 95% ⁇ "identical" to a reference nucleotide sequence encoding a Nodal or Lefty polypeptide is intended that the nucleotide sequence of the polynucleotide is identical to the reference sequence except that the polynucleotide sequence may include up to five point mutations per each 100 nucleotides of the reference nucleotide sequences encoding the Nodal and Lefty polypeptides.
  • a polynucleotide having a nucleotide sequence at least 95% identical to a reference nucleotide sequence up to 5% of the nucleotides in the reference sequence may be deleted or substituted with another nucleotide, or a number of nucleotides up to 5% of the total nucleotides in the reference sequence may be inserted into the reference sequence.
  • These mutations of the reference sequence may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence.
  • nucleic acid molecule is at least 90%, 95%, 96%, 97%, 98% or 99% identical to, for instance, the nucleotide sequences shown in Figures IA and B and 2A and B or to the nucleotides sequence of the deposited cDNA clones can be determined conventionally using known computer programs such as the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 5371 1). Bestfit uses the local homology algorithm of Smith and Waterman to find the best segment of homology between two sequences (Advances in Applied Mathematics 2:482-489 (1981)).
  • the parameters are set, of course, such that the percentage of identity is calculated over the full length of the reference nucleotide sequence and that gaps in homology of up to 5% of the total number of nucleotides in the reference sequence are allowed.
  • a preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence, also referred to as a global sequence alignment can be determined using the FASTDB computer program based on the algorithm of Brutlag and colleagues (Comp. App. Biosci. 6:237-245 (1990)).
  • This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score.
  • This corrected score is what is used for the purposes of the present invention. Only bases outside the 5' and 3' bases of the subject sequence, as displayed by the FASTDB alignment, which are not matched/aligned with the query sequence, are calculated for the purposes of manually adjusting the percent identity score.
  • a 90 base subject sequence is aligned to a 100 base query sequence to determine percent identity.
  • the deletions occur at the 5' end of the subject sequence and therefore, the FASTDB alignment does not show a matched/alignment of the first 10 bases at 5' end.
  • the 10 unpaired bases represent 10% of the sequence (number of bases at the 5' and 3' ends not matched/total number of bases in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 bases were perfectly matched the final percent identity would be 90%.
  • a 90 base subject sequence is compared with a 100 base query sequence.
  • deletions are internal deletions so that there are no bases on the 5' or 3' of the subject sequence which are not matched/aligned with the query.
  • percent identity calculated by FASTDB is not manually corrected.
  • bases 5' and 3' of the subject sequence which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
  • the present application is directed to nucleic acid molecules at least 90%, 95%, 96%, 97%, 98%o or 99% identical to the nucleic acid sequences shown in Figures IA and B and 2A and B (SEQ ID NO:l and SEQ ID NO:3, respectively) or to the nucleic acid sequences of the deposited cDNAs, irrespective of whether they encode a polypeptide having Nodal or Lefty activity. This is because even where a particular nucleic acid molecule does not encode a polypeptide having Nodal or Lefty activity, one of skill in the art would still know how to use the nucleic acid molecule, for instance, as a hybridization probe or a polymerase chain reaction (PCR) primer.
  • PCR polymerase chain reaction
  • nucleic acid molecules of the present invention that do not encode a polypeptide having Nodal or Lefty activity include, inter alia, (1) isolating the Nodal or Lefty genes or allelic variants thereof in a cDNA library; (2) in situ hybridization (e.g., "FISH") to metaphase chromosomal spreads to provide precise chromosomal location of the Nodal or Lefty genes, as described by Verma and colleagues (Human Chromosomes: A Manual of Basic Techniques, Pergamon Press, New York (1988)); and Northern Blot analysis for detecting Nodal or Lefty mRNA expression in specific tissues.
  • FISH in situ hybridization
  • nucleic acid molecules having sequences at least 90%, 95%, 96%, 97%, 98% or 99% identical to the nucleic acid sequences shown in Figures IA and B and 2A and B (SEQ ID NO:l and SEQ ID NO:3, respectively) or to the nucleic acid sequences of the deposited cDNAs or to fragments of these polynucleotides as described herein, which do, in fact, encode polypeptides having Nodal or Lefty activity.
  • a polypeptide having Nodal or Lefty activity is intended polypeptides exhibiting activity similar, but not necessarily identical, to an activity of the active forms of Nodal or Lefty proteins of the invention, as measured in a particular biological assay.
  • the Nodal and Lefty proteins of the present invention are involved in the regulation of cell growth and differentiation.
  • Other TGF- ⁇ -like molecules have the capacity to stimulate the proliferation of human endothelial cells in the presence of the comitogen concanavalin A (conA).
  • ConA concanavalin A
  • Endothelial cells are obtained and cultured in 96 well flat-bottomed culture dishes (Costar, Cambridge, MA) in RPMI 1640 medium supplemented with 10% heat-inactivated fetal bovine serum (HyClone Labs, Logan, UT), 1% L-glutamine, 100 U/mL penicillin, 100 ⁇ g/mL streptomycin, 0.1% gentamicin (Life Technologies, Inc., Rockville, MD) in the presence of 2 ⁇ g/mL conA (Calbiochem, La Jolla, CA). ConA and the polypeptide to be analyzed are added to a final volume of medium of 0.2 mL.
  • compositions of the invention need not regulate cellular proliferation and differentiation in a dose-dependent manner, it is preferred that "a polypeptide having Nodal or Lefty activity" includes polypeptides that also exhibit any of the same cellular proliferation and differentiation regulatory activities in the above- described assays in a dose-dependent manner.
  • a polypeptide having Nodal or Lefty protein activity will exhibit substantially similar dose-dependence in a given activity as compared to the Nodal or Lefty proteins (i.e., the candidate polypeptide will exhibit greater activity or not more than about 25-fold less and, preferably, not more than about tenfold less activity relative to the reference Nodal and Lefty proteins).
  • polypeptides of the invention may regulate cellular growth or differentiation of a particular cell type through the use of an in vitro colony forming assay to measure the extent of inhibition of myeloid progenitor cells (Youn, et al, J. Immunol. 155:2661 -2667 (1995)).
  • this assay involves collecting human or mouse bone marrow cells and plating the same on agar, adding one or more growth factors and either (1) transfected host cell-supernatant containing Nodal or Lefty protein (or a candidate polypeptide) or (2) nontransfected host cell-supernatant control, and measuring the effect on colony formation by murine and human CFU- granulocyte-macrophages (CFU-GM), by human burst-forming unit-erythroid (BFU-E), or by human CFU granulocyte-erythroid-macrophage-megakaryocyte (CFU-GEMM).
  • CFU-GM murine and human CFU- granulocyte-macrophages
  • BFU-E human burst-forming unit-erythroid
  • CFU-GEMM human CFU granulocyte-erythroid-macrophage-megakaryocyte
  • Nodal and Lefty may exhibit an activity on leukocytes including, for example, monocytes, lymphocytes and neutrophils. For this reason, Nodal and Lefty are active in directing the proliferation and differentiation of these cell types. Such activity is useful, for example, for immune enhancement or suppression, myeloprotection, stem cell mobilization, acute and chronic inflammatory control and treatment of leukemia. Assays for measuring such activity are well known in the art (Peters, et al, Immun. Today 17:273 (1996); Young, et al, J. Exp. Med. 182:1111 (1995); Caux, et al, Nature 390:258 (1992); and Santiago-Schwarz, et al, Adv. Exp. Med. Biol. 378:7 (1995).
  • nucleic acid molecules having a sequence at least 90%, 95%, 96%, 97%, 98%, or 99% identical to the nucleic acid sequence of the deposited cDNA or the nucleic acid sequences shown in Figures IA and B and 2A and B (SEQ ID NO: l and SEQ ID NO:3, respectively), or fragments thereof, will encode polypeptides "having Nodal or Lefty protein activity.”
  • degenerate variants of these nucleotide sequences all encode the same polypeptides, this will be clear to the skilled artisan even without performing the above described comparison assay.
  • nucleic acid molecules that are not degenerate variants, a reasonable number will also encode a polypeptide having Nodal or Lefty activity. This is because the skilled artisan is fully aware of amino acid substitutions that are either less likely or not likely to significantly effect protein function (e.g., replacing one aliphatic amino acid with a second aliphatic amino acid), as further described below.
  • Polynucleotide Assays also encompasses the use of Nodal and Lefty polynucleotides to detect complementary polynucleotides, such as, for example, as a diagnostic reagent for detecting diseases or susceptibility to diseases related to the presence of mutated Nodal and Lefty.
  • diseases are related to an under-expression of Nodal and Lefty, such as, for example, abnormal cellular proliferation such as tumors and cancers.
  • Nucleic acids for diagnosis may be obtained from a patient's cells, such as from blood, urine, saliva, tissue biopsy and autopsy material.
  • the genomic DNA may be used directly for detection or may be amplified enzymatically by using PCR (Saiki et al , Nature 324: 163-166 (1986)) prior to analysis.
  • RNA or cDNA may also be used for the same purpose.
  • PCR primers complementary to the nucleic acid encoding Nodal or Lefty can be used to identify and analyze Nodal or Lefty mutations.
  • deletions and insertions can be detected by a change in size of the amplified product in comparison to the normal genotype.
  • Point mutations can be identified by hybridizing amplified DNA to radiolabeled Nodal or Lefty RNA or alternatively, radiolabeled Nodal or Lefty antisense DNA sequences. Perfectly matched sequences can be distinguished from mismatched duplexes by RNase A digestion or by differences in melting temperatures.
  • DNA sequence differences may be achieved by detection of alteration in electrophoretic mobility of DNA fragments in gels with or without denaturing agents. Small sequence deletions and insertions can be visualized by high resolution gel electrophoresis. DNA fragments of different sequences may be distinguished on denaturing formamide gradient gels in which the mobilities of different DNA fragments are retarded in the gel at different positions according to their specific melting or partial melting temperatures (see, e.g., Myers et al, Science 230:1242 (1985)). Sequence changes at specific locations may also be revealed by nuclease protection assays, such as RNase and SI protection or the chemical cleavage method (e.g., Cotton et al, Proc. Natl. Acad. Sci., USA, 85:4397-4401 (1985)).
  • the detection of a specific DNA sequence may be achieved by methods such as hybridization, RNase protection, chemical cleavage, direct DNA sequencing or the use of restriction enzymes, (e.g., Restriction Fragment Length Polymorphisms (RFLP)) and Southern blotting of genomic DNA.
  • restriction enzymes e.g., Restriction Fragment Length Polymorphisms (RFLP)
  • mutations can also be detected by in situ analysis.
  • Lefty and Nodal polypeptides (including fragments, variants derivatives, and analogs) of the invention can be chemically synthesized (e.g., see Creighton, 1983, Proteins: Structures and Molecular Principles, W.H. Freeman & Co., N.Y.), Lefty and Nodal polypeptides may advantageously be produced by recombinant DNA technology using techniques well known in the art for expressing gene sequences and/or nucleic acid coding sequences. Such methods can be used to construct expression vectors containing the polynucleotides of the invention and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination.
  • RNA capable of Lefty or Nodal sequences may be chemically synthesized using, for example, synthesizers. See, for example, the techniques described in "Oligonucleotide Synthesis", 1984, Gait, M.J. ed., IRL Press, Oxford, which is incorporated by reference herein in its entirety.
  • the present invention relates to vectors which include the isolated DNA molecules (i.e., polynucleotides) of the present invention, host cells which are genetically engineered with the recombinant vectors, and the production of Nodal or Lefty polypeptides or fragments thereof by recombinant techniques using these host cells or host cells that have otherwise been genetically engineered using techniques known in art to express a polypeptide of the invention.
  • the vector may be, for example, a phage, plasmid, viral or retroviral vector. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
  • the polynucleotides may be joined to a vector containing a selectable marker for propagation in a host.
  • a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • the polynucleotide of the invention is operatively associated with an appropriate heterologous regulatory element (e.g., a promoter or enhancer or both), such as the phage lambda PL promoter, the E. coli lac, trp, phoA and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few.
  • a heterologous regulatory element e.g., a promoter or enhancer or both
  • a promoter or enhancer or both such as the phage lambda PL promoter, the E. coli lac, trp, phoA and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few.
  • a promoter or enhancer or both such as the phage lambda PL promoter, the E. coli lac, trp, phoA and tac promoters, the SV40 early and late
  • these constructs will further contain sites for transcription initiation, termination and, in the transcribed region, a ribosome binding site for translation.
  • the coding portion of the transcripts expressed by the constructs will preferably include a translation initiating codon at the beginning and a termination codon (UAA, UGA or UAG) appropriately positioned at the end of the polypeptide to be translated.
  • the expression vectors will preferably include at least one selectable marker.
  • markers include dihydrofolate reductase, G418 or neomycin resistance for eukaryotic cell culture and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria.
  • Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, 293 and Bowes melanoma cells; and plant cells. Appropriate culture mediums and conditions for the above-described host cells are known in the art.
  • Vectors preferred for use in bacteria include pHE4-5, pQE70, pQE60 and pQE-9 (QIAGEN, Inc., supra); pBS vectors, Phagescript vectors, Bluescript vectors, pNH8A, pNH16a, pNH18A, pNH46A (Stratagene); and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 (Pharmacia).
  • eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXTl , and pSG (Stratagene); and pSVK3, pBPV, pMSG and pSVL (Pharmacia).
  • Other suitable vectors will be readily apparent to the skilled artisan.
  • Introduction of the construct into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection or other methods. Such methods are described in many standard laboratory manuals (for example, Davis, et al, Basic Methods In Molecular Biology (1986)).
  • the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly those of mammalian origin, that have been engineered to delete or replace endogenous genetic material (e.g., Human Nodal or Human Lefty coding sequence), and/or to include genetic material (e.g. heterologous polynucleotide sequences) that is operably associated with Human Nodal or Human Lefty polynucleotides of the invention, and which activates, alters, and/or amplifies endogenous Human Nodal or Human Lefty polynucleotides.
  • genetic material e.g. heterologous polynucleotide sequences
  • heterologous polynucleotide sequences that is operably associated with Human Nodal or Human Lefty polynucleotides of the invention, and which activates, alters, and/or amplifies endogenous Human Nodal or Human Lefty polynucleotides.
  • heterologous control regions e.g.
  • the polypeptide may be expressed in a modified form, such as a fusion protein, and may include not only secretion signals, but also additional heterologous functional regions. For instance, a region of additional amino acids, particularly charged amino acids, may be added to the N-terminus of the polypeptide to improve stability and persistence in the host cell, during purification, or during subsequent handling and storage. Also, peptide moieties may be added to the polypeptide to facilitate purification. Such regions may be removed prior to final preparation of the polypeptide. The addition of peptide moieties to polypeptides to engender secretion or excretion, to improve stability and to facilitate purification, among others, are familiar and routine techniques in the art.
  • a preferred fusion protein comprises a heterologous region from immunoglobulin that is useful to stabilize and purify proteins.
  • EP-A-O 464 533 (Canadian counterpart 2045869) discloses fusion proteins comprising various portions of constant region of immunoglobulin molecules together with another human protein or part thereof.
  • the Fc part in a fusion protein is thoroughly advantageous for use in therapy and diagnosis and thus results, for example, in improved pharmacokinetic properties (EP-A 0232 262).
  • Fc portion proves to be a hindrance to use in therapy and diagnosis, for example when the fusion protein is to be used as antigen for immunizations.
  • human proteins such as hIL-5
  • Fc portions have been fused with Fc portions for the purpose of high-throughput screening assays to identify antagonists of hIL-5 (Bennett, D., et al, J. Molecular Recognition 8:52-58 (1995); Johanson, K., et al, J. Biol. Chem. 270:9459-9471 (1995)).
  • the Nodal and Lefty proteins can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification.
  • HPLC high performance liquid chromatography
  • Polypeptides of the present invention include: products purified from natural sources, including bodily fluids, tissues and cells, whether directly isolated or cultured; products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. In addition, polypeptides of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes.
  • N-terminal methionine encoded by the translation initiation codon generally is removed with high efficiency from any protein after translation in all eukaryotic cells. While the N-terminal methionine on most proteins also is efficiently removed in most prokaryotes, for some proteins this prokaryotic removal process is inefficient, depending on the nature of the amino acid to which the N-terminal methionine is covalently linked.
  • Lefty and Nodal polypeptides include fragments, variants, derivatives and analogs which are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc.
  • compositions of the invention are conjugated to other molecules to increase their water-solubility (e.g., polyethylene glycol), half-life, or ability to bind targeted tissue (e.g., bisphosphonates and fluorochromes to target the proteins to bony sites).
  • water-solubility e.g., polyethylene glycol
  • targeted tissue e.g., bisphosphonates and fluorochromes to target the proteins to bony sites.
  • the invention further provides isolated Nodal and Lefty polypeptides having the amino acid sequences encoded by the deposited cDNAs, or the amino acid sequences in SEQ ID NO:2 and SEQ ID NO:4, respectively, or a peptide or polypeptide comprising a fragment (i.e., a portion) of the above polypeptides.
  • the polypeptides and polynucleotides of the present invention are preferably provided in an isolated form, and preferably are purified to a point within the range of near complete (e.g., >90% pure) to complete (e.g., >99% pure) homogeneity.
  • isolated means that the material is removed from its original environment (e.g., the natural environment if it is naturally occurring).
  • a naturally-occurring polynucleotide or polypeptide present in a living animal is not isolated, but the same polynucleotide or polypeptide, separated from some or all of the coexisting materials in the natural system, is isolated.
  • isolated polypeptide are polypeptides that have been purified partially or substantially from a recombinant host cell.
  • a recombinantly produced version of a Nodal or Lefty polypeptide can be substantially purified by the one-step method described by Smith and Johnson (Gene 67:31-40 (1988)).
  • polynucleotides could be part of a vector and/or such polynucleotides or polypeptides could be part of a composition, and still be isolated in that such vector or composition is not part of its natural environment.
  • Isolated polypeptides and polynucleotides according to the present invention also include such molecules produced naturally or synthetically.
  • Polypeptides and polynucleotides of the invention also can be purified from natural or recombinant sources using anti-Nodal or anti-Lefty antibodies of the invention which may routinely be generated and utilized using methods known in the art.
  • polypeptides may be produced using Recombinant DNA technology known to those skilled in the art. Recombinant DNA technology known to those skilled in the art can be used to create novel mutant proteins or muteins including single or multiple amino acid substitutions, deletions, additions or fusion proteins. Such modified polypeptides can show, e.g., enhanced activity or increased stability. In addition, they may be purified in higher yields and show better solubility than the corresponding natural polypeptide, at least under certain purification and storage conditions.
  • polypeptide fragments of the present invention include polypeptides comprising an amino acid sequence contained in SEQ ID NO:2, SEQ ID NO:4, encoded by the cDNA contained in the deposited clones (HTLFA20 and HNGEF08, (encoding Nodal) and HUKEJ46 (encoding Lefty)), or encoded by nucleic acids which hybridize (e.g., under stringent hybridization conditions) to the nucleotide sequence contained in the deposited clones, that shown in Figures 1 A and IB (SEQ ID NOT) and/or Figures 2 A and 2B (SEQ ID NO:3), or the complementary strand thereto.
  • Polypeptide fragments may be "free-standing" or comprised within a larger polypeptide of which the fragment forms a part or region, most preferably as a single continuous region.
  • Representative examples of polypeptide fragments of the invention included, for example, fragments that comprise or alternatively, consist of, from about amino acid residues, 1 to 20, 21 to 40, 41 to 60, 61 to 83, 84 to 100, 101 to 120, 121 to 140, 141 to 160, 161 to 180, 181 to 200, 201 to 220, 201 to 224, 210 to 231, 221 to 240, 241 to 260, 261 to 280, 261 to 283, 281 to 289, 281 to 300, 301 to 320, 321 to 340, 341 to 348, 341 to 360, and 341 to 366 of SEQ ID NO:2 and/or SEQ ID NO:4.
  • polypeptide fragments can be at least about 20, 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 210, 220, 230, 240, 250, 260, 270, 280, 290, 300, 310, 320, 330, 340, 350 or 360 amino acids in length.
  • “about” includes the particularly recited ranges, larger or smaller by several (i.e. 5, 4, 3, 2 or 1) amino acids, at either extreme or at both extremes.
  • the fragments or polypeptides of the invention are not larger than 325, 300, 250, 225, 200, 185, 175, 170, 165, 160, 155, 150, 145, 140, 135, 130, 125, 120, 115, 110, 105, 100, 90, 80, 75, 60, 50, 40, 30 or 25 amino acids residues in length.
  • Additional embodiments encompass polypeptide fragments comprising one or more functional regions of Nodal or Lefty polypeptides of the invention, such as, one or more Garnier-Robson alpha-regions, beta-regions, turn-regions, and coil-regions, Chou-Fasman alpha-regions, beta-regions, and coil-regions, Kyte-Doolittle hydrophilic regions and hydrophobic regions, Eisenberg alpha- and beta-amphipathic regions, Karplus-Schulz flexible regions, Emini surface-forming regions and Jameson- Wolf regions of high antigenic index, or any combination thereof, as disclosed in Figures 5 and 6 and in Tables I and II and as described herein.
  • polypeptide fragments comprising, or alternatively consisting of, the TGF- ⁇ -like domain of Nodal (amino acid residues 174-283 of SEQ ID NO:2).
  • Additional preferred embodiments encompass polypeptide fragments comprising, or alternatively consisting of, the mature domain of Lefty (amino acid residues 1-348 of SEQ ID NO:4), the first predicted TGF- ⁇ -like domain of Lefty (amino acid residues 60-348 of SEQ ID NO:4), the second predicted TGF- ⁇ -like domain of Lefty (amino acid residues 1 18-348 of SEQ ID NO:4), and/or the third predicted TGF- ⁇ -like domain of Lefty (amino acid residues 125-348 of SEQ ID NO:4).
  • polypeptide fragments of the invention comprise, or alternatively, consist of, amino acid residues aspartic acid-1 to alanine-27, arginine-30 to glutamic acid-58, cysteine-64 to phenylalanine-82, glycine-85 to serine-110, and leucine-130 to leucine-283 of the Nodal sequence recited in SEQ ID NO:2.
  • polypeptide fragments of the invention comprise, or alternatively, consist of, amino acid residues leucine-(-15) to serine-(-2), alanine-3 to leucine- 19, valine-34 to histidine-51 , arginine-54 to leucine-72, glutamic acid-75 to arginine- 114, arginine-117 to proline-192, histidine-198 to proline-209, glycine-211 to leucine-286, tryptophan-290 to glutamic acid-302, and serine-305 to proline-348 of the Lefty amino acid sequence recited in SEQ ID NO:4.
  • polypeptides of the invention comprise, or alternatively consist of, amino acid residues 19 to 25, 84 to 104, 105-125, 126 to 150, 151 to 170, 171 to 200, 201-250, 251 to 270, 271 to 297, 329 to 339, and/or 340 363 of the Lefty amino acid sequence depicted in Figures 2A and 2B.
  • Polynucleotides encoding these polypeptides are also encompassed by the invention, as are polynucleotides that hybridize to the complementary strand of these encoding polynucleotides under high stringency conditions (e.g., as described herein) and polypeptides encoded by these hybridizing polynucleotides.
  • the polypeptides of the present invention have uses which include, but are not limited to, a molecular weight marker on SDS-PAGE gels or on molecular sieve gel filtration columns using methods well known to those of skill in the art.
  • polypeptides of the present invention can also be used to raise polyclonal and monoclonal antibodies, which are useful in assays for detecting Nodal or Lefty protein expression as described below or as agonists and antagonists capable of enhancing or inhibiting Nodal or Lefty protein function.
  • polypeptides can be used in the yeast two-hybrid system to "capture" Nodal or Lefty protein binding proteins which are also candidate agonists and antagonists according to the present invention.
  • the yeast two hybrid system is described by Fields and Song (Nature 340:245-246 (1989)).
  • the invention provides peptides or polypeptides comprising epitope-bearing portions of a polypeptide of the invention.
  • the epitope of this polypeptide portion is an immunogenic or antigenic epitope of a polypeptide of the invention.
  • An "immunogenic epitope” is defined as a part of a protein that elicits an antibody response when the whole protein is the immunogen.
  • a region of a protein molecule to which an antibody can bind is defined as an "antigenic epitope”.
  • the number of immunogenic epitopes of a protein generally is less than the number of antigenic epitopes (see, for instance, Geysen, et al, Proc. Natl. Acad. Sci. USA 81:3998-4002 (1983)).
  • peptides or polypeptides bearing an antigenic epitope i.e., that contain a region of a protein molecule to which an antibody can bind
  • relatively short synthetic peptides that mimic part of a protein sequence are routinely capable of eliciting an antiserum that reacts with the partially mimicked protein (see, for instance, Sutcliffe, J. G., et al, Science 219:660-666 (1983)).
  • Peptides capable of eliciting protein-reactive sera are frequently represented in the primary sequence of a protein, can be characterized by a set of simple chemical rules, and are confined neither to immunodominant regions of intact proteins (i.e., immunogenic epitopes) nor to the amino or carboxyl terminals.
  • Antigenic epitope-bearing peptides and polypeptides of the invention are therefore useful to raise antibodies, including monoclonal antibodies, that bind specifically to a polypeptide of the invention (see, for instance, Wilson, et al, Cell 37:767-778 (1984)).
  • Antigenic epitope-bearing peptides and polypeptides of the invention preferably contain a sequence of at least seven, more preferably at least nine and most preferably between about 15 to about 30 amino acids contained within the amino acid sequence of a polypeptide of the invention.
  • Non-limiting examples of antigenic polypeptides or peptides that can be used to generate Nodal-specific antibodies include: a polypeptide comprising amino acid residues from about Lys-54 to about Asp-62, from about Val-91 to about Leu-99, from about Lys-100 to about Gln-108, from about Cys-116 to about Pro-124, from about Gln-140 to about Leu-148, from about Trp-156 to about Ser-164, from about Arg- 170, to about Gln-181, from about Cys-212 to about Phe-224, from about Tyr-239, to about Thr-247, from about Pro-251, to about Met-259, and from about Asp-263, to about His-271.
  • Non-limiting examples of antigenic polypeptides or peptides that can be used to generate Lefty-specific antibodies include: a polypeptide comprising amino acid residues from about Asp-71 to about Ser-79, from about Arg- 106 to about Val-1 14, from about Leu-136 to about Arg- 144, from about Asp- 154 to about Asp- 164, from about His-171 to about Asp- 179, from about Gin- 189 to about Leu- 197, from about Pro-227 to about Glu-236, from about Gly-246 to about Glu-254, from about Pro-256 to about Gln-266, from about Cys-297 to about Ala-305, from about Ile-317 to about Pro-325, from about Ile-330 to about Val-340, and from about Val-348 to about Pro-366.
  • These polypeptide fragments have been determined to bear antigenic epitopes of the Nodal and Lefty proteins by the analysis of the Jameson- Wolf antigenic index, as shown in Figures 5 and
  • the epitope-bearing peptides and polypeptides of the invention may be produced by any conventional means (see, for example, Houghten, R. A., et al, Proc. Natl. Acad. Sci. USA 52:5131-5135 (1985); and U.S. Patent No. 4,631,21 1 to Houghten, et al. (1986)).
  • Epitope-bearing peptides and polypeptides of the invention are used to induce antibodies according to methods well known in the art (see, for instance, Sutcliffe, et al, supra; Wilson, et al, supra; Chow, M. , et al, Proc. Natl. Acad. Sci.
  • Immunogenic epitope-bearing peptides of the invention i.e., those parts of a protein that elicit an antibody response when the whole protein is the immunogen, are identified according to methods known in the art (see, for instance, Geysen, et al, supra). Further still, U.S. Patent No.
  • Nodal and Lefty proteins of the invention are members of the TGF- ⁇ polypeptide superfamily
  • deletions of N-terminal amino acids up to the N-terminal-most cysteine of the predicted active form of the proteins at positions 183 and 233 of SEQ ID NO:2 and SEQ ID NO:4, respectively, may retain some biological activity such as receptor binding or modulation of target cell activities.
  • Polypeptides having further N-terminal deletions including the Cys-183 and Cys-233 residues in SEQ ID NO:2 and SEQ ID NO:4, respectively, would not be expected to retain such biological activities because it is known that this residue in a TGF- ⁇ -related polypeptide is required for forming an integral part of the "cysteine knot motif required for biological activities of the active form of TGF- ⁇ family members (McDonald, N. Q. and Hendrickson, W. A. Cell 73:303-304 (1993)).
  • the present invention further provides polypeptides having one or more residues deleted from the amino terminus of the amino acid sequence of Nodal shown in SEQ ID NO:2, up to the cysteine residue at position number 183, and polynucleotides encoding such polypeptides.
  • the present invention provides polypeptides comprising the amino acid sequence of residues n'-283 of SEQ ID NO:2, where n 1 is an integer in the range of 173-183, and 183 is the position of the first residue from the N-terminus of the complete Nodal polypeptide (shown in SEQ ID NO:2) believed to be required for receptor binding activity of the Nodal protein.
  • the invention provides polynucleotides encoding polypeptides having the amino acid sequence of residues of 173-283, 174-283, 175-283, 176-283, 177-283, 178-283, 179-283, 180-283, 181-283, 182-283, and 183-283 of SEQ ID NO:2. Polynucleotides encoding these polypeptides also are provided.
  • the present invention also provides polypeptides having one or more residues deleted from the amino terminus of the amino acid sequence of Lefty shown in SEQ ID NO:4, up to the cysteine residue at position number 233, and polynucleotides encoding such polypeptides.
  • the present invention provides polypeptides comprising the amino acid sequence of residues n 2 -348 of SEQ ID NO:4, where n 2 is an integer in the range of 125-233, and 233 is the position of the first residue from the N-terminus of the complete Nodal polypeptide (shown in SEQ ID NO:4) believed to be required for receptor binding activity of the Lefty protein.
  • the invention provides polynucleotides encoding polypeptides having the amino acid sequence of residues of 125-348, 126-348, 127-348, 128-348, 129-348, 130-348, 131-348, 132-348, 133-348, 134-348, 135-348, 136-348, 137-348, 138-348, 139-348, 140-348, 141-348, 142-348, 143-348, 144-348, 145-348, 146-348, 147-348, 148-348, 149-348, 150-348, 151-348, 152-348, 153-348, 154-348, 155-348, 156-348, 157-348, 158-348, 159-348, 160-348, 161-348, 162-348, 163-348, 164-348, 165-348, 166-348, 167-348, 168-348, 169-348, 170-348, 171-348, 172-348
  • Polypeptides having further C-terminal deletions including Cys-249 and Cys-335 of SEQ ID NO:2 and SEQ ID NO:4, respectively, would not be expected to retain such biological activities because it is known that this residue in a TGF- ⁇ -related polypeptide is required for forming an integral part of the "cysteine knot motif required for biological activities of the active form of TGF- ⁇ family members (McDonald, N. Q. and Hendrickson, W. A. Cell 73:303-304 (1993)).
  • the present invention further provides polypeptides having one or more residues from the carboxy terminus of the amino acid sequence of Nodal shown in SEQ ID NO:2, up to the cysteine residue at position 249 of SEQ ID NO:2, and polynucleotides encoding such polypeptides.
  • the present invention provides polypeptides having the amino acid sequence of residues 1-m 1 of the amino acid sequence in SEQ ID NO:2, where m 1 is any integer in the range of 249 to 283, and residue 249 is the position of the first residue from the C- terminus of the complete Nodal polypeptide (shown in SEQ ID NO:2) believed to be required for receptor binding or modulation of cellular growth and differentiation activities of the Nodal protein.
  • the invention provides polynucleotides encoding polypeptides having the amino acid sequence of residues 1-249, 1-250, 1-251, 1-252, 1-253, 1-254, 1-255, 1-256, 1-257, 1-258, 1-259, 1-260, 1-261, 1-262, 1-263, 1-264, 1-265, 1-266, 1-267, 1-268, 1-269, 1-270, 1-271, 1-272, 1-273, 1-274, 1-275, 1-276, 1-277, 1-278, 1-279, 1-280, 1-281, 1-282, and 1-283 of SEQ ID NO:2. Polynucleotides encoding these polypeptides also are provided.
  • the present invention also provides polypeptides having one or more residues from the carboxy terminus of the amino acid sequence of Lefty shown in SEQ ID NO:4, up to the cysteine residue at position 335 of SEQ ID NO:4, and polynucleotides encoding such polypeptides.
  • the present invention provides polypeptides having the amino acid sequence of residues 1-m 2 of the amino acid sequence in SEQ ID NO:4, where m 2 is any integer in the range of 335 to 348, and residue 335 is the position of the first residue from the C-terminus of the complete Lefty polypeptide (shown in SEQ ID NO:4) believed to be required for receptor binding or modulation of cellular growth and differentiation activities of the Lefty protein.
  • the invention provides polynucleotides encoding polypeptides having the amino acid sequence of residues 1-335, 1-336, 1-337,
  • the invention also provides polypeptides having one or more amino acids deleted from both the amino and the carboxyl termini, which may be described generally as having residues n'-m 1 of SEQ ID NO:2 or n 2 -m 2 SEQ ID NO:4, where n 1 , m 1 , n 2 , and m 2 are integers as described above.
  • nucleotide sequence encoding a polypeptide consisting of a portion of the complete Nodal amino acid sequence encoded by the cDNA clone contained in ATCC Deposit No. 209092 and/or 209135, where this portion excludes from 1 to about 183 amino acids from the amino terminus of the complete amino acid sequence encoded by the cDNA clone contained in ATCC Deposit No. 209092 and/or 209135, or from 1 to about 34 amino acids from the carboxy terminus, or any combination of the above amino terminal and carboxy terminal deletions, of the complete amino acid sequence encoded by the cDNA clone contained in ATCC Deposit No. 209092 and/or 209135.
  • nucleotide sequence encoding a polypeptide consisting of a portion of the complete Lefty amino acid sequence encoded by the cDNA clone contained in ATCC Deposit No. 209091 is included, where this portion excludes from 1 to about 250 amino acids from the amino terminus of the complete amino acid sequence encoded by the cDNA clone contained in ATCC Deposit No. 209091, or from 1 to about 12 amino acids from the carboxy terminus, or any combination of the above amino terminal and carboxy terminal deletions, of the complete amino acid sequence encoded by the cDNA clone contained in ATCC Deposit No. 209091.
  • Polynucleotides encoding all of the above deletion mutant polypeptide forms also are provided.
  • the present invention further provides polypeptides having one or more residues deleted from the amino terminus of the Human Nodal amino acid sequence shown in SEQ ID NO:2, up to the glutamic acid residue at position number 278 and polynucleotides encoding such polypeptides.
  • the present invention provides polypeptides comprising the amino acid sequence of residues n 3 -283 of Figures IA and B (SEQ ID NO:2), where n 3 is an integer in the range of 2 to 278, and 279 is the position of the first residue from the N-terminus of the complete Human Nodal polypeptide believed to be required for at least immunogenic activity of the Human Nodal protein.
  • the invention provides polynucleotides encoding polypeptides comprising, or alternatively consisting of, the amino acid sequence of residues of V-2 to L-283; A-3 to L-283; V-4 to L-283; D-5 to L-283; G-6 to L-283; Q-7 to L-283; N-8 to L-283; W-9 to L-283; T-10 to L-283; F-l 1 to L-283; A-12 to L-283; F-13 to L-283; D-14 to L-283; F-15 to L-283; S-16 to L-283; F-17 to L-283; L-18 to L-283; S-19 to L-283; Q-20 to L-283; Q-21 to L-283; E-22 to L-283; D-23 to L-283; L-24 to L-283; A-25 to L-283; W-26 to L-283; A-27 to L-283; E-28 to L-283; L-29 to L-283; R-30 to L-283; L-31 to L-2
  • the present invention further provides polypeptides having one or more residues deleted from the carboxy terminus of the amino acid sequence of the Human Nodal shown in SEQ ID NO:2, up to the glycine residue at position number 6, and polynucleotides encoding such polypeptides.
  • the present invention provides polypeptides comprising the amino acid sequence of residues 1-m 3 of SEQ ID NO:2, where m 3 is an integer in the range of 6 to 283, and 6 is the position of the first residue from the C-terminus of the complete Human Nodal polypeptide believed to be required for at least immunogenic activity of the Human Nodal protein.
  • the invention provides polynucleotides encoding polypeptides comprising, or alternatively consisting of, the amino acid sequence of residues D-l to C-282; D-l to G-281 ; D-l to C-280; D-l to E-279; D-l to E-278; D-l to V-277; D-l to 1-276; D-l to M-275; D-l to D-274; D-l to K-273; D-l to H-272; D-l to H-271 ; D-l to D-270; D-l to L-269; D-l to L-268; D-l to V-267; D-l to R-266; D-l to G-265; D-l to N-264; D-l to D-263; D-l to V-262; D-l to Y-261 ; D-l to L-260; D-l to M-259; D-l to S-258; D-l to L-257; D-l to P
  • the invention also provides polypeptides having one or more amino acids deleted from both the amino and the carboxyl termini of a Human Nodal polypeptide, which may be described generally as having residues n 3 -m 3 of Figures IA and B (SEQ ID NO:2), where n 3 and m 3 are integers as described above.
  • the present invention further provides polypeptides having one or more residues deleted from the amino terminus of the Human Lefty amino acid sequence shown in SEQ ID NO:4, up to the proline residue at position number 361 and polynucleotides encoding such polypeptides.
  • the present invention provides polypeptides comprising the amino acid sequence of residues n 4 -180 of Figures 2A and B (SEQ ID NO:4), where n 4 is an integer in the range of 2 to 361, and 362 is the position of the first residue from the N-terminus of the complete Human Lefty polypeptide believed to be required for at least immunogenic activity of the Human Lefty protein.
  • the invention provides polynucleotides encoding polypeptides comprising, or alternatively consisting of, the amino acid sequence of residues of Q-2 to P-366; P-3 to P-366; L-4 to P-366; W-5 to P-366; L-6 to
  • the present invention further provides polypeptides having one or more residues deleted from the carboxy terminus of the amino acid sequence of the Human Lefty shown in SEQ ID NO:4, up to the leucine residue at position number 6, and polynucleotides encoding such polypeptides.
  • the present invention provides polypeptides comprising the amino acid sequence of residues 1-m 4 of SEQ ID NO:4, where m 4 is an integer in the range of 6 to 366, and 6 is the position of the first residue from the C-terminus of the complete Human Lefty polypeptide believed to be required for at least immunogenic activity of the Human Lefty protein.
  • the invention provides polynucleotides encoding polypeptides comprising, or alternatively consisting of, the amino acid sequence of residues M-l to Q-365; M-l to L-364; M-l to R-363; M-l to R-362 P-361 ; M-l to V-360; M-l to L-359; M-l to A-358; M-l to G-357 D-356; M-l to S-355; M-l to A-354; M-l to C-353; M-l to S-352 C-351 ; M-l to K-350; M-l to Q-349; M-l to V-348; M-l to R-347 M-346; M-l to N-345; M-l to P-344; M-l to L-343; M-l to S-342 V-341 ; M-l to V-340; M-l to Q-339; M-l to P-338; M-l to R-337 T-336; M-l to
  • polypeptides having one or more amino acids deleted from both the amino and the carboxyl termini of a Human Lefty polypeptide, which may be described generally as having residues n 4 -m 4 of
  • Figures 2 A and B (SEQ ID NO:4), where n 4 and m 4 are integers as described above.
  • the invention further includes variations of the Nodal and Lefty polypeptides which show substantial Nodal or Lefty polypeptide activity or which include regions of Nodal or Lefty proteins such as the protein portions discussed below.
  • Such mutants include deletions, insertions, inversions, repeats, and type substitutions selected according to general rules known in the art so as have little effect on activity.
  • guidance concerning how to make phenotypically silent amino acid substitutions is provided wherein the authors indicate that there are two main approaches for studying the tolerance of an amino acid sequence to change (Bowie, J. U., et al, Science 247:1306-1310 (1990)),.
  • the first method relies on the process of evolution, in which mutations are either accepted or rejected by natural selection.
  • the second approach uses genetic engineering to introduce amino acid changes at specific positions of a cloned gene and selections or screens to identify sequences that maintain functionality.
  • amino acid residues may be (i) one in which one or more of the amino acid residues are substituted with a conserved or non-conserved amino acid residue (preferably a conserved amino acid residue) and such substituted amino acid residue may or may not be one encoded by the genetic code, or (ii) one in which one or more of the amino acid residues includes a substituent group, or (iii) one in which the active form of the polypeptide is fused with another compound, such as a compound to increase the half-life of the polypeptide (for example, polyethylene glycol), or (iv) one in which the additional amino acids are fused to the above form of the polypeptide, such as an IgG Fc fusion region peptide or leader or secretory sequence or a sequence which is employed for purification of the above form of the polypeptide or a proprotein sequence.
  • a conserved or non-conserved amino acid residue preferably a conserved amino acid residue
  • substituted amino acid residue may or may not be one encoded
  • the Nodal and Lefty proteins of the present invention may include one or more amino acid substitutions, deletions or additions, either from natural mutations or human manipulation. As indicated, changes are preferably of a minor nature, such as conservative amino acid substitutions that do not significantly affect the folding or activity of the protein (see Table 1). TABLE 1. Conservative Amino Acid Substitutions.
  • Embodiments of the invention are directed to polypeptides which comprise the amino acid sequence of a Nodal or Lefty polypeptide described herein, but having an amino acid sequence which contains at least one conservative amino acid substitution, but not more than 50 conservative amino acid substitutions, even more preferably, not more than 40 conservative amino acid substitutions, still more preferably, not more than 30 conservative amino acid substitutions, and still even more preferably, not more than 20 conservative amino acid substitutions, when compared with the Nodal or Lefty polynucleotide sequence described herein.
  • a peptide or polypeptide in order of ever-increasing preference, it is highly preferable for a peptide or polypeptide to have an amino acid sequence which comprises the amino acid sequence of a Nodal or Lefty polypeptide, which contains at least one, but not more than 10, 9, 8, 7, 6, 5, 4, 3, 2 or 1 conservative amino acid substitutions.
  • the number of substitutions, additions or deletions in the amino acid sequence of Figures IA and B (SEQ ID NO:2), Figures 2 A and B (SEQ ID NO:4), a polypeptide sequence encoded by the deposited clones, and/or any of the polypeptide fragments described herein (e.g., the mature forms or the active TGF- ⁇ consensus cleavage domains) is 75, 70, 60, 50, 40, 35, 30, 25, 20, 15, 10, 9, 8, 7, 6, 5, 4, 3, 2, 1 or 150-50, 100-50, 50-20, 30-20, 20-15, 20-10, 15-10, 10-1, 5-10, 1-5, 1-3 or 1-2.
  • polypeptides may be produced using Recombinant DNA technology known to those skilled in the art.
  • Recombinant DNA technology known to those skilled in the art can be used to create novel mutant polypeptides or muteins including single or multiple amino acid substitutions, deletions, additions or fusion proteins.
  • modified polypeptides can show, e.g., enhanced activity or increased stability.
  • they may be purified in higher yields and show better solubility than the corresponding natural polypeptide, at least under certain purification and storage conditions.
  • the invention also encompasses Nodal and Lefty derivatives and analogs that have one or more amino acid residues deleted, added, or substituted to generate Nodal and Lefty polypeptides that are better suited for expression, scale up, etc., in the host cells chosen.
  • cysteine residues can be deleted or substituted with another amino acid residue in order to eliminate disulfide bridges; N-linked glycosylation sites can be altered or eliminated to achieve, for example, expression of a homogeneous product that is more easily recovered and purified from yeast hosts which are known to hyperglycosylate N- linked sites.
  • Amino acids in the Nodal and Lefty polypeptides of the present invention that are essential for function can be identified by methods known in the art, such as site-directed mutagenesis or alanine-scanning mutagenesis (Cunningham and Wells, Science 244:1081 -1085 (1989)). The latter procedure introduces single alanine mutations at every residue in the molecule. The resulting mutant molecules are then tested for biological activity such as receptor binding or in vitro proliferative activity.
  • Replacement of amino acids can also change the selectivity of the binding of a ligand to cell surface receptors (for example, Ostade, et al, Nature 361:266-268 (1993)) describes certain mutations resulting in selective binding of TNF- ⁇ to only one of the two known types of TNF receptors.
  • Sites that are critical for ligand-receptor binding can also be determined by structural analysis such as crystallization, nuclear magnetic resonance or photoaffinity labeling (Smith, et al, J. Mol. Biol. 224:899-904 (1992); de Vos, et al. Science 255:306- 312 (1992)).
  • Nodal and Lefty are members of the TGF- ⁇ -related protein family, to modulate rather than completely eliminate biological activities of Nodal and Lefty preferably mutations are made in sequences encoding amino acids in the Nodal and Lefty conserved domain, i.e., in positions 173 to 283 or SEQ ID NO:2 or positions 125 to 348 of SEQ ID NO:4, more preferably in residues within this region which are not conserved in all members of the TGF- ⁇ -related protein family.
  • mutations to the Nodal and Lefty polypeptides are mad in positions other than the conserved cysteine residues comprising the "cysteine knot" motif characteristic of TGF- ⁇ -related protein family members.
  • isolated polynucleotides comprising nucleic acid sequences which encode the above Nodal and Lefty mutants.
  • polypeptides of the present invention are preferably provided in an isolated form, and preferably are substantially purified.
  • Recombinantly produced versions of the Nodal and Lefty polypeptides can be substantially purified by the one-step method described by Smith and Johnson (Gene 67:31-40 (1988)).
  • Polypeptides of the invention also can be purified from natural or recombinant sources using anti-Nodal or anti-Lefty antibodies of the invention in methods which are well known in the art of protein purification.
  • the invention further provides isolated Nodal and Lefty polypeptides comprising an amino acid sequence selected from the group consisting of: (a) the amino acid sequence of the full-length Nodal polypeptide having the complete amino acid sequence shown in SEQ ID NO:2 (i.e., positions 1 to 283 of SEQ ID NO:2); (b) the amino acid sequence of the predicted active Nodal polypeptide having the amino acid sequence at positions 173 to 283 of SEQ ID NO:2; (c) the amino acid sequence of the Nodal polypeptide having the complete amino acid sequence encoded by the cDNA clone contained in ATCC Deposit No.
  • polypeptides of the present invention include polypeptides which have at least 90% similarity, more preferably at least 95% similarity, and still more preferably at least 96%, 97%, 98% or 99% similarity to those described above.
  • the polypeptides of the invention also comprise those which are at least 80% identical, more preferably at least 90% or 95% identical, still more preferably at least 96%, 97%, 98% or 99% identical to the polypeptide encoded by the deposited cDNAs or to the polypeptides of SEQ ID NO:2 or SEQ ID NO:4, and also include portions of such polypeptides with at least 30 amino acids and more preferably at least 50 amino acids.
  • % similarity for two polypeptides is intended a similarity score produced by comparing the amino acid sequences of the two polypeptides using the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 5371 1) and the default settings for determining similarity. Bestfit uses the local homology algorithm of Smith and Waterman (Advances in Applied Mathematics 2:482-489, 1981) to find the best segment of similarity between two sequences.
  • polypeptide having an amino acid sequence at least, for example, 95% "identical" to a reference amino acid sequence of a Nodal or Lefty polypeptide is intended that the amino acid sequence of the polypeptide is identical to the reference sequence except that the polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the reference amino acid of the Nodal or Lefty polypeptide.
  • up to 5% of the amino acid residues in the reference sequence may be deleted or substituted with another amino acid, or a number of amino acids up to 5% of the total amino acid residues in the reference sequence may be inserted into the reference sequence.
  • These alterations of the reference sequence may occur at the amino or carboxy terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.
  • any particular polypeptide is at least 90%, 95%, 96%), 97%), 98%) or 99% identical to, for instance, the amino acid sequence shown in Figures IA and B (SEQ ID NO:2), the amino acid sequence shown in Figures 2A and B (SEQ ID NO:4), the amino acid sequence encoded by deposited cDNA clones HTLFA20, HNGEF08, and HUKEJ46, or fragments thereof, can be determined conventionally using known computer programs such the Bestfit program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, 575 Science Drive, Madison, WI 53711).
  • the parameters are set, of course, such that the percentage of identity is calculated over the full length of the reference amino acid sequence and that gaps in homology of up to 5% of the total number of amino acid residues in the reference sequence are allowed.
  • the identity between a reference (query) sequence (a sequence of the present invention) and a subject sequence is determined using the FASTDB computer program based on the algorithm of Brutlag et al. (Comp. App. Biosci. 6:237-245 (1990)).
  • the percent identity is corrected by calculating the number of residues of the query sequence that are N- and C-terminal of the subject sequence, which are not matched/aligned with a corresponding subject residue, as a percent of the total bases of the query sequence.
  • a determination of whether a residue is matched/aligned is determined by results of the FASTDB sequence alignment. This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score. This final percent identity score is what is used for the purposes of this embodiment. Only residues to the N- and C-termini of the subject sequence, which are not matched/aligned with the query sequence, are considered for the purposes of manually adjusting the percent identity score. That is, only query residue positions outside the farthest N- and C-terminal residues of the subject sequence. For example, a 90 amino acid residue subject sequence is aligned with a 100 residue query sequence to determine percent identity.
  • the deletion occurs at the N-terminus of the subject sequence and therefore, the FASTDB alignment does not show a matching/alignment of the first 10 residues at the N-terminus.
  • the 10 unpaired residues represent 10% of the sequence (number of residues at the N- and C- termini not matched/total number of residues in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched the final percent identity would be 90%.
  • a 90 residue subject sequence is compared with a 100 residue query sequence. This time the deletions are internal deletions so there are no residues at the N- or C-termini of the subject sequence which are not matched/aligned with the query.
  • the invention also encompasses fusion proteins in which the full-length Nodal or Lefty polypeptide or fragment, variant, derivative, or analog thereof is fused to an unrelated protein. These fusion proteins can be routinely designed on the basis of the Nodal or Lefty nucleotide and polypeptide sequences disclosed herein.
  • Nodal and/or Lefty polypeptides and fragments (including epitope-bearing fragments) thereof described herein can be combined with parts of the constant domain of immunoglobulins (IgG), resulting in chimeric (fusion) polypeptides.
  • IgG immunoglobulins
  • fusion proteins facilitate purification and show an increased half-life in vivo. This has been shown, e.g., for chimeric proteins consisting of the first two domains of the human CD4-polypeptide and various domains of the constant regions of the heavy or light chains of mammalian immunoglobulins (EP A 394,827; Traunecker, et aL, Nature 331:84-86 (1988)).
  • Fusion proteins that have a disulfide-linked dimeric structure due to the IgG part can also be more efficient in binding and neutralizing other molecules than the monomeric Nodal or Lefty proteins or protein fragments alone (Fountoulakis, et al, J. Biochem. 270:3958-3964 (1995)).
  • Nodal and Lefty fusion proteins that are encompassed by the invention include, but are not limited to, fusion of the Nodal or Lefty polypeptide sequences to any amino acid sequence that allows the fusion proteins to be displayed on the cell surface (e.g. the IgG Fc domain); or fusions to an enzyme, fluorescent protein, or luminescent protein which provides a marker function.
  • Antibodies Nodal or Lefty polypeptide-specific antibodies for use in the present invention can be raised against the intact Nodal or Lefty protein or an antigenic polypeptide fragment thereof, which may be presented together with a carrier protein, such as an albumin, to an animal system (such as rabbit or mouse) or, if it is long enough (at least about 25 amino acids), without a carrier.
  • a carrier protein such as an albumin
  • antibody As used herein, the term "antibody” (Ab) or “monoclonal antibody” (Mab) is meant to include intact molecules as well as antibody fragments (such as, for example, Fab and F(ab')2 fragments) which are capable of specifically binding to Nodal or Lefty protein. Fab and F(ab')2 fragments lack the Fc fragment of intact antibody, clear more rapidly from the circulation, and may have less non-specific tissue binding of an intact antibody (Wahl, et al, J. Nucl. Med. 24:316-325 (1983)). Thus, these fragments are preferred.
  • the antibodies of the present invention may be prepared by any of a variety of methods.
  • cells expressing the Nodal or Lefty protein or an antigenic fragment thereof can be administered to an animal in order to induce the production of sera containing polyclonal antibodies.
  • a preparation of Nodal and Lefty protein is prepared and purified to render it substantially free of natural contaminants. Such a preparation is then introduced into an animal in order to produce polyclonal antisera of greater specific activity.
  • the antibodies of the present invention are monoclonal antibodies (or Nodal or Lefty protein binding fragments thereof).
  • monoclonal antibodies can be prepared using hybridoma technology (Kohler, et al, Nature 256:495 (1975); Kohler, et al, Eur. J. Immunol. 6:511 (1976); Kohler, et al, Eur. J. Immunol. 6:292 (1976); Hammerling, et al, in: Monoclonal Antibodies and T-Cell Hybridomas, Elsevier, N.Y., (1981) pp. 563-681)).
  • such procedures involve immunizing an animal (preferably a mouse) with a Nodal or Lefty protein antigen or. more preferably, with a Nodal or Lefty protein-expressing cell.
  • Suitable cells can be recognized by their capacity to bind anti -Nodal or anti-Lefty protein antibody.
  • Such cells may be cultured in any suitable tissue culture medium; however, it is preferable to culture cells in Earle's modified Eagle's medium supplemented with 10% fetal bovine serum (inactivated at about 56° C), and supplemented with about 10 ⁇ g/1 of nonessential amino acids, about 1 ,000 U/ml of penicillin, and about 100 ⁇ g/ml of streptomycin.
  • the splenocytes of such mice are extracted and fused with a suitable myeloma cell line.
  • a suitable myeloma cell line may be employed in accordance with the present invention; however, it is preferable to employ the parent myeloma cell line (SP2O), available from the American Type Culture Collection, Rockville, Maryland.
  • SP2O parent myeloma cell line
  • the resulting hybridoma cells are selectively maintained in HAT medium, and then cloned by limiting dilution as described by Wands and colleagues (Gastroenterology 80:225-232 (1981)).
  • the hybridoma cells obtained through such a selection are then assayed to identify clones which secrete antibodies capable of binding the Nodal or Lefty protein antigen.
  • additional antibodies capable of binding to the Nodal or Lefty protein antigens may be produced in a two-step procedure through the use of anti-idiotypic antibodies.
  • Such a method makes use of the fact that antibodies are themselves antigens, and that, therefore, it is possible to obtain an antibody which binds to a second antibody.
  • Nodal or Lefty protein-specific antibodies are used to immunize an animal, preferably a mouse.
  • the splenocytes of such an animal are then used to produce hybridoma cells, and the hybridoma cells are screened to identify clones which produce an antibody whose ability to bind to the Nodal or Lefty protein-specific antibody can be blocked by the Nodal or Lefty protein antigen.
  • Such antibodies comprise anti-idiotypic antibodies to the Nodal or Lefty protein-specific antibodies and can be used to immunize an animal to induce formation of further Nodal or Lefty protein-specific antibodies.
  • Fab and F(ab')2 and other fragments of the antibodies of the present invention may be used according to the methods disclosed herein.
  • Such fragments are typically produced by proteolytic cleavage, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab')2 fragments).
  • Nodal or Lefty protein-binding fragments can be produced through the application of recombinant DNA technology or through synthetic chemistry.
  • anti-Nodal and anti-Lefty it may be preferable to use "humanized" chimeric monoclonal antibodies.
  • Such antibodies can be produced using genetic constructs derived from hybridoma cells producing the monoclonal antibodies described above.
  • the present inventors have discovered that Nodal is expressed in neutrophils and testes.
  • the present inventors have discovered that Lefty is expressed in uterine cancer, colon cancer, apoptotic T-cells, fetal heart, Wilm's Tumor tissue, frontal lobe of the brain from a patient with dementia, neutrophils, salivary gland, small intestine, 7, 8, and 12 week old human embryos, frontal cortex and hypothalamus from a patient with schizophrenia, brain from a patient with Alzheimer's Disease, adipose tissue, brown fat, TNF- and LPS-induced and uninduced bone marrow stroma, activated monocytes and macrophages, rhabdomyosarcoma, cycloheximide-treated Raji cells, breast lymph nodes, hemangiopericytoma, testes, fetal epithelium (skin), and IL-5-induced eosinophils.
  • substantially altered (increased or decreased) levels of Nodal or Lefty gene expression can be detected in affected tissues, cells, or bodily fluids (e.g., sera, plasma, urine, synovial fluid or spinal fluid) taken from an individual having such a disorder, relative to a "standard" Nodal or Lefty gene expression level, that is, the Nodal and Lefty expression level in affected tissues or bodily fluids from an individual not having the cell growth and differentiation disorder.
  • bodily fluids e.g., sera, plasma, urine, synovial fluid or spinal fluid
  • the invention provides a diagnostic method useful during diagnosis of a cell growth and differentiation disorder, which involves measuring the expression level of the gene encoding the Nodal or Lefty proteins in affected tissues, cells, or body fluids from an individual and comparing the measured gene expression level with a standard Nodal or Lefty gene expression level, whereby an increase or decrease in the gene expression level compared to the standard is indicative of a cell growth and differentiation disorder.
  • tissue in mammals with cancer of the immune or reproductive systems express significantly reduced levels of the Nodal or Lefty proteins and mRNA encoding the Nodal or Lefty proteins when compared to corresponding "standard” levels.
  • enhanced levels of the Nodal or Lefty proteins can be detected in certain body fluids (e.g., sera, plasma, urine, and spinal fluid) from mammals with such a cancer when compared to sera from mammals of the same species not having the cancer.
  • the invention provides a diagnostic method useful during diagnosis of a cellular growth and differentiation disorder, including cancers, which involves measuring the expression level of the genes encoding the Nodal and Lefty proteins in tissues, cells, or body fluids from an individual and comparing the measured gene expression levels with standard Nodal and Lefty gene expression levels, whereby an increase or decrease in the gene expression level compared to the standard is indicative of a cell growth and differentiation disorder.
  • a diagnosis of a disorder in the regulation of cell growth and differentiation including diagnosis of a tumor, has already been made according to conventional methods
  • the present invention is useful as a prognostic indicator, whereby patients exhibiting depressed Nodal or Lefty gene expression will experience a worse clinical outcome relative to patients expressing the gene at a level nearer the standard level.
  • test the expression level of the genes encoding the Nodal and Lefty polypeptides is intended qualitatively or quantitatively measuring or estimating the level of the Nodal and Lefty polypeptides or the level of the mRNA encoding the Nodal and Lefty polypeptides in a first biological sample either directly (e.g., by determining or estimating absolute protein level or mRNA level) or relatively (e.g., by comparing to the Nodal and Lefty polypeptides levels or mRNA level in a second biological sample).
  • the Nodal and Lefty polypeptides levels or mRNA levels in the first biological sample is measured or estimated and compared to a standard Nodal and Lefty polypeptide level or mRNA level, the standard being taken from a second biological sample obtained from an individual not having the disorder or being determined by averaging levels from a population of individuals not having a disorder of cellular growth and differentiation.
  • a standard Nodal and Lefty polypeptide level or mRNA level the standard being taken from a second biological sample obtained from an individual not having the disorder or being determined by averaging levels from a population of individuals not having a disorder of cellular growth and differentiation.
  • biological sample any biological sample obtained from an individual, body fluid, cell line, tissue culture, or other source which contains Nodal and Lefty protein or mRNA.
  • biological samples include body fluids (such as sera, plasma, urine, synovial fluid and spinal fluid) which contain free active forms of Nodal or Lefty protein, tissues exhibiting the effects of abnormally regulated cell growth or differentiation, and other tissue sources found to express complete, mature, or active forms of the Nodal or Lefty proteins or a Nodal or Lefty receptor.
  • body fluids such as sera, plasma, urine, synovial fluid and spinal fluid
  • tissue sources found to express complete, mature, or active forms of the Nodal or Lefty proteins or a Nodal or Lefty receptor.
  • the present invention is useful for diagnosis or treatment of various cell growth and differentiation-related disorders in mammals, preferably humans.
  • disorders include tumors, cancers, interstitial lung disease, and any disregulation of the growth and differentiation patterns of cell function including, but not limited to, autoimmunity, arthritis, leukemias, lymphomas, immunosuppression, immunity, humoral immunity, inflammatory bowel disease, myelosuppression, and the like.
  • Total cellular RNA can be isolated from a biological sample using any suitable technique such as the single-step guanidinium-thiocyanate-phenol- chloroform method described by Chomczynski and Sacchi (Anal. Biochem. 162: 156-159 (1987)). Levels of mRNA encoding the Nodal and Lefty polypeptides are then assayed using any appropriate method. These include Northern blot analysis, SI nuclease mapping, the polymerase chain reaction (PCR), reverse transcription in combination with the polymerase chain reaction (RT-PCR), and reverse transcription in combination with the ligase chain reaction (RT-LCR). Assaying Nodal and Lefty polypeptides levels in a biological sample can occur using antibody-based techniques.
  • any suitable technique such as the single-step guanidinium-thiocyanate-phenol- chloroform method described by Chomczynski and Sacchi (Anal. Biochem. 162: 156-159 (1987)).
  • Nodal and Lefty protein expression in tissues can be studied with classical immunohistological methods (Jalkanen, M., et al, J. Cell. Biol. 101:976-985 (1985); Jalkanen, M., et al, J. Cell. Biol. 105:3087-3096 (1987)).
  • Other antibody-based methods useful for detecting Nodal and Lefty polypeptides gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA).
  • ELISA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • Suitable antibody assay labels include enzyme labels, such as, glucose oxidase, and radioisotopes, such as iodine ( 125 I, 121 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( 112 In), and technetium ( 99m Tc), and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • enzyme labels such as, glucose oxidase, and radioisotopes, such as iodine ( 125 I, 121 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( 112 In), and technetium ( 99m Tc)
  • fluorescent labels such as fluorescein and rhodamine, and biotin.
  • Nodal and Lefty polypeptides can also be detected in vivo by imaging.
  • Antibody labels or markers for in vivo imaging of Nodal or Lefty protein include those detectable by X-radiography, NMR or ESR.
  • suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but are not overtly harmful to the subject.
  • Suitable markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the antibody by labeling of nutrients for the relevant hybridoma.
  • a Nodal or Lefty polypeptide-specific antibody or antibody fragment which has been labeled with an appropriate detectable imaging moiety such as a radioisotope (for example, 131 I, 112 In, 99m Tc), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is introduced (for example, parenterally, subcutaneously or intraperitoneally) into the mammal to be examined for immune system disorder.
  • a radioisotope for example, 131 I, 112 In, 99m Tc
  • a radio-opaque substance for example, parenterally, subcutaneously or intraperitoneally
  • the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99m Tc.
  • the labeled antibody or antibody fragment will then preferentially accumulate at the location of cells which contain Nodal and Lefty protein, in vivo tumor imaging is described by Burchiel and coworkers (Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, Burchiel, S. W. and Rhodes, B. A., eds., Masson Publishing Inc. (1982)). Treatment
  • Nodal and Lefty polynucleotides and polypeptides are useful for diagnosis of conditions involving abnormally high or low expression of Nodal and Lefty activities. Given the cells and tissues where Nodal and Lefty are expressed as well as the activities modulated by Nodal and Lefty, it is readily apparent that a substantially altered (increased or decreased) level of expression of Nodal and Lefty in an individual compared to the standard or "normal" level produces pathological conditions related to the bodily system(s) in which Nodal and Lefty are expressed and/or are active.
  • Nodal and Lefty proteins of the invention are members of the TGF- ⁇ superfamily the active domains of the proteins may be released in soluble form from the cells which express the Nodal and Lefty by proteolytic cleavage. Therefore, when Nodal or Lefty active domain is added from an exogenous source to cells, tissues or the body of an individual, the protein will exert its physiological activities on its target cells of that individual.
  • the invention also provides a method of treatment of an individual in need of an increased level of Nodal or Lefty activity comprising administering to such an individual a pharmaceutical composition comprising an amount of an isolated Nodal or Lefty polypeptide of the invention, particularly the active form of the Nodal and Lefty protein of the invention, effective to increase the Nodal and Lefty activity level in such an individual. no -
  • compositions e.g., polynucleotides, polypeptides, and fragments variants, derivatives and analogs thereof, and antibodies thereto, and angonists and antagonists thereto
  • Nodal and Lefty compositions may also be employed to inhibit T-cell proliferation by the inhibition of IL-2 biosynthesis for the treatment of T-cell mediated auto-immune diseases and lymphocytic leukemias.
  • compositions corresponding to Nodal and Lefty regulate T H ]/T H2 cytokine production may be used as anti-inflammatories.
  • Nodal and Lefty compositions may also be administered to treat or prevent inflammation, allergy, and infectious diseases or as an adjuvant for immunotherapy of tumors.
  • Nodal and Lefty compositions may also be employed to stimulate wound healing.
  • Nodal and Lefty compounds may also be employed to regulate hematopoiesis, by regulating the activation and differentiation of various hematopoietic progenitor cells, such as for example, to stimulate erythropoiesis or to stimulate the release of mature leukocytes from the bone marrow following chemotherapy, i.e., in stem cell mobilization.
  • Nodal is essential for mesoderm formation and subsequent organization of axial structures in early mouse development
  • the human Nodal homologue of the present invention is also likely involved developmental processes such as the correct formation of various structures or in one or more post-developmental capacities including sexual development, pituitary hormone production, and the creation of bone and cartilage, as are many of the other members of the TGF- ⁇ superfamily.
  • the invention encompasses the use of Nodal compositions to regulate these processes, such as, for example, in stimulating bone and/or cartilage formation, and stimulating the production of pituitary hormone.
  • murine Lefty is important in left/right handedness of the developing organism.
  • the homology between murine Lefty and the novel human Lefty homologue of the present invention indicates that the novel human Lefty homologue of the present invention may also be involved in correct formation of various structures with respect to the rest of the developing organism or Lefty may also be involved in one or more post-developmental capacities including sexual development, pituitary hormone production, and the creation of bone and cartilage, as are many of the other members of the TGF- ⁇ superfamily. Accordingly, the invention encompasses the use of Nodal compositions to regulate these processes, such as, for example, in stimulating bone and/or cartilage formation, and stimulating the production of hormones in the pituitary.
  • Nodal and Lefty compounds may also be administered regulate or modulate cell growth and differentiation which is not necessarily associated with endogenously high or low levels of Nodal and/or Lefty.
  • Nodal and Lefty polypeptides of the present invention are useful for enhancing or enriching the growth and/or differentiation of specific cell populations, e.g., embryonic cells or stem cells.
  • the Nodal and/or Lefty polypeptide composition will be formulated and dosed in a fashion consistent with good medical practice, taking into account the clinical condition of the individual patient (especially the side effects of treatment with Nodal and/or Lefty polypeptide alone), the site of delivery of the Nodal and/or Lefty polypeptide composition, the method of administration, the scheduling of administration, and other factors known to practitioners.
  • the "effective amount" of Nodal and/or Lefty polypeptide for purposes herein is thus determined by such considerations.
  • the total pharmaceutically effective amount of Nodal and/or Lefty polypeptide administered parenterally per dose will be in the range of about 1 ⁇ g/kg/day to 10 mg/kg/day of patient body weight, although, as noted above, this will be subject to therapeutic discretion. More preferably, this dose is at least 0.01 mg/kg/day, and most preferably for humans between about 0.01 and 1 mg/kg/day for the hormone. If given continuously, the Nodal and/or Lefty polypeptide is typically administered at a dose rate of about 1 ⁇ g/kg/hour to about 50 ⁇ g/kg/hour, either by 1-4 injections per day or by continuous subcutaneous infusions, for example, using a mini-pump.
  • compositions containing the Nodal and Lefty proteins of the invention may be administered orally, rectally, parenterally, intracistemally, intravaginally, intraperitoneally, topically (as by powders, ointments, drops or transdermal patch), bucally, or as an oral or nasal spray.
  • pharmaceutically acceptable carrier is meant a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • parenteral refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion.
  • the Nodal and Lefty polypeptides are also suitably administered by sustained-release systems.
  • sustained-release compositions include semi-permeable polymer matrices in the form of shaped articles, e.g., films, or mirocapsules.
  • Sustained-release matrices include polylactides (U.S. Pat. No. 3,773,919, EP 58,481), copolymers of L-glutamic acid and gamma-ethyl-L- glutamate (Sidman, U., et al, Biopolymers 22:547-556 (1983)), poly (2- hydroxyethyl methacrylate; Langer, R., et al, J. Biomed. Mater. Res.
  • Sustained-release Nodal and Lefty polypeptide compositions also include liposomally entrapped Nodal and Lefty polypeptides. Liposomes containing Nodal and Lefty polypeptides are prepared by methods known in the art (DE 3,218,121 ; Epstein, et al, Proc. Natl. Acad. Sci. (USA) 82:3688-3692 (1985); Hwang, et al, Proc.
  • the liposomes are of the small (about 200-800 Angstroms) unilamellar type in which the lipid content is greater than about 30 mol. percent cholesterol, the selected proportion being adjusted for the optimal Nodal and Lefty polypeptide therapy.
  • the Nodal and/or Lefty polypeptide is formulated generally by mixing it at the desired degree of purity, in a unit dosage injectable form (solution, suspension, or emulsion), with a pharmaceutically acceptable carrier, i.e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation.
  • a pharmaceutically acceptable carrier i.e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation.
  • the formulation preferably does not include oxidizing agents and other compounds that are known to be deleterious to polypeptides.
  • the formulations are prepared by contacting the Nodal and Lefty polypeptide uniformly and intimately with liquid carriers or finely divided solid carriers or both. Then, if necessary, the product is shaped into the desired formulation.
  • the carrier is a parenteral carrier, more preferably a solution that is isotonic with the blood of the recipient. Examples of such carrier vehicles include water, saline, Ringer's solution, and dextrose solution. Non-aqueous vehicles such as fixed oils and ethyl oleate are also useful herein, as well as liposomes.
  • the carrier suitably contains minor amounts of additives such as substances that enhance isotonicity and chemical stability.
  • additives such as substances that enhance isotonicity and chemical stability.
  • Such materials are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, succinate, acetic acid, and other organic acids or their salts; antioxidants such as ascorbic acid; low molecular weight (less than about ten residues) polypeptides, e.g., polyarginine or tripeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids, such as glycine, glutamic acid, aspartic acid, or arginine; monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, manose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbi
  • compositions which contain a therapeutically effective amount of human Nodal and/or Lefty polypeptide, in a pharmaceutically acceptable vehicle or carrier.
  • These compositions of the invention may be useful in the therapeutic modulation or diagnosis of bone, cartilage, or other connective cell or tissue growth and/or differentiation.
  • These compositions may be used to treat such conditions as osteoarthritis, osteoporosis, and other abnormalities of bone, cartilage, muscle, tendon, ligament and/or other connective tissues and/or organs such as liver, lung, cardiac, pancreas, kidney, and other tissues.
  • compositions may also be useful in the growth and/or formation of cartilage, tendon, ligament, meniscus, and other connective tissues or any combination of the above (e.g., therapeutic modulation of the tendon-to-bone attachment apparatus). These compositions may also be useful in treating periodontal disease and modulating wound healing and tissue repair of such tissues as epidermis, nerve, muscle, cardiac muscle, liver, lung, cardiac, pancreas, kidney, and other tissues and/or organs.
  • Pharmaceutical compositions containing Nodal and/or Lefty of the invention may include one or more other therapeutically useful component such as BMP-1, BMP-2, BMP-3, BMP-4, BMP-5, BMP-6, and/or BMP-7 (See, for example, U. S. Patent Nos.
  • BMP-8 See, for example, PCT publication WO91/18098
  • BMP-9 See, for example, PCT publication WO93/00432
  • BMP- 10 See, for example, PCT publication WO94/26893
  • BMP-1 1 See, for example, PCT publication WO94/26892
  • BMP- 12 and/or BMP- 13 See, for example, PCT publication WO95/ 16035
  • BIP growth and differentiation factors
  • GDFs growth and differentiation factors
  • VGR-2 epidermal growth factor
  • FGF fibroblast growth factor
  • TGF-alpha TGF-beta
  • activins inhibins
  • IGF insulin-like growth factor
  • Nodal and Lefty polypeptides are typically formulated in such vehicles at a concentration of about 0.1 mg/ml to 100 mg/ml, preferably 1-10 mg/ml, at a pH of about 3 to 8. It will be understood that the use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of Nodal and Lefty polypeptide salts.
  • Nodal and Lefty polypeptides to be used for therapeutic administration must be sterile. Sterility is readily accomplished by filtration through sterile filtration membranes (e.g., 0.2 micron membranes).
  • Therapeutic Nodal and Lefty polypeptide compositions generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
  • Nodal and Lefty polypeptides ordinarily will be stored in unit or multi-dose containers, for example, sealed ampoules or vials, as an aqueous solution or as a lyophilized formulation for reconstitution.
  • a lyophilized formulation 10-ml vials are filled with 5 ml of sterile-filtered 1% (w/v) aqueous Nodal and Lefty polypeptide solution, and the resulting mixture is lyophilized.
  • the infusion solution is prepared by reconstituting the lyophilized Nodal and Lefty polypeptide using bacteriostatic water- for-injection (WFI).
  • WFI bacteriostatic water- for-injection
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions of the invention.
  • Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • the polypeptides of the present invention may be employed in conjunction with other therapeutic compounds.
  • the invention also provides a method of screening compounds to identify those which enhance or block the action of Nodal and Lefty on cells, such as their interactions with Nodal- or Lefty-binding molecules such as receptor molecules.
  • An agonist is a compound which increases the natural biological functions of Nodal or Lefty or which functions in a manner similar to Nodal or Lefty, while antagonists decrease or eliminate such functions.
  • the invention provides a method for identifying a receptor protein or other ligand-binding protein which binds specifically to a Nodal or Lefty polypeptide.
  • a cellular compartment such as a membrane or a preparation thereof, may be prepared from a cell that expresses a molecule that binds Nodal or Lefty.
  • the preparation is incubated with labeled Nodal or Lefty and complexes of Nodal or Lefty bound to the receptor or other binding protein are isolated and characterized according to routine methods known in the art.
  • the Nodal or Lefty polypeptides may be bound to a solid support so that binding molecules solubilized from cells are bound to the column and then eluted and characterized according to routine methods.
  • a cellular compartment such as a membrane or a preparation thereof, may be prepared from a cell that expresses a molecule that binds Nodal or Lefty, such as a molecule of a signaling or regulatory pathway modulated by Nodal or Lefty.
  • the preparation is incubated with labeled Nodal or Lefty in the absence or the presence of a candidate molecule which may be a Nodal or Lefty agonist or antagonist.
  • the ability of the candidate molecule to bind the binding molecule is reflected in decreased binding of the labeled ligand.
  • Molecules which bind gratuitously, i.e., without inducing the effects of Nodal or Lefty on binding the Nodal or Lefty binding molecule are most likely to be good antagonists.
  • Molecules that bind well and elicit effects that are the same as or closely related to Nodal or Lefty are agonists.
  • Nodal or Lefty-like effects of potential agonists and antagonists may by measured, for instance, by determining activity of a second messenger system following interaction of the candidate molecule with a cell or appropriate cell preparation, and comparing the effect with that of Nodal or Lefty or molecules that elicit the same effects as Nodal or Lefty.
  • Second messenger systems that may be useful in this regard include but are not limited to AMP guanylate cyclase, ion channel or phosphoinositide hydrolysis second messenger systems.
  • an assay for Nodal and Lefty antagonists is a competitive assay that combines Nodal or Lefty and a potential antagonist with membrane-bound Nodal or Lefty receptor molecules or recombinant Nodal or
  • Nodal and Lefty can be labeled, such as by radioactivity, such that the number of Nodal or Lefty molecules bound to a receptor molecule can be determined accurately to assess the effectiveness of the potential antagonist.
  • Potential antagonists include small organic molecules, peptides, polypeptides and antibodies that bind to a polypeptide of the invention and thereby inhibit or extinguish its activity. Potential antagonists also may be small organic molecules, a peptide, a polypeptide such as a closely related protein or antibody that binds the same sites on a binding molecule, such as a receptor molecule, without inducing Nodal- or Lefty-induced activities, thereby preventing the action of Nodal or Lefty by excluding Nodal or Lefty from binding.
  • Antisense technology can be used to control gene expression through antisense DNA or RNA or through triple-helix formation.
  • Antisense techniques are discussed in a number of studies (for example, Okano, J. Neurochem. 56:560 (1991); "Oligodeoxynucleotides as Antisense Inhibitors of Gene Expression.” CRC Press, Boca Raton, FL (1988)).
  • Triple helix formation is discussed in a number of studies, as well (for instance, Lee, et al, Nucleic Acids Research 10-1573 (1979); Cooney, et al, Science 241:456 (1988); Dervan, et al, Science 251: 1360 (1991)).
  • the methods are based on binding of a polynucleotide to a complementary DNA or RNA.
  • the 5' coding portion of a polynucleotide that encodes the mature polypeptide of the present invention may be used to design an antisense RNA oligonucleotide of from about 10 to 40 base pairs in length.
  • a DNA oligonucleotide is designed to be complementary to a region of the gene involved in transcription thereby preventing transcription and the production of Nodal or Lefty.
  • the antisense RNA oligonucleotide hybridizes to the mRNA in vivo and blocks translation of the mRNA molecule into Nodal and Lefty polypeptide.
  • the oligonucleotides described above can also be delivered to cells such that the antisense RNA or DNA may be expressed in vivo to inhibit production of Nodal or Lefty protein.
  • the agonists and antagonists may be employed in a composition with a pharmaceutically acceptable carrier, e.g., as described above.
  • the antagonists may be employed for instance to inhibit the activation of macrophages and their precursors, and of neutrophils, basophils, B lymphocytes and some T-cell subsets, e.g., activated and CD8 cytotoxic T cells and natural killer cells, in certain autoimmune and chronic inflammatory and infective diseases.
  • autoimmune diseases include multiple sclerosis, and insulin-dependent diabetes.
  • the antagonists may also be employed to treat infectious diseases including silicosis, sarcoidosis, idiopathic pulmonary fibrosis by preventing the recruitment and activation of mononuclear phagocytes. They may also be employed to treat idiopathic hyper-eosinophilic syndrome by preventing eosinophil production and stimulation.
  • Endotoxic shock may also be treated by the antagonists by preventing the stimulation of macrophages and their production of the human chemokine polypeptides of the present invention.
  • the antagonists may also be employed to treat histamine-mediated allergic reactions and immunological disorders including late phase allergic reactions, chronic urticaria, and atopic dermatitis by inhibiting mast cell and basophil degranulation and release of histamine.
  • IgE-mediated allergic reactions such as allergic asthma, rhinitis, and eczema may also be treated.
  • the antagonists may also be employed to treat chronic and acute inflammation by preventing the activation of monocytes in a wound area.
  • Antagonists may also be employed to treat rheumatoid arthritis by preventing the activation of monocytes in the synovial fluid in the joints of patients.
  • Monocyte activation plays a significant role in the pathogenesis of both degenerative and inflammatory arthropathies.
  • the antagonists may be employed to interfere with the deleterious cascades attributed primarily to IL-1 and TNF, which prevents the biosynthesis of other inflammatory cytokines. In this way, the antagonists may be employed to prevent inflammation.
  • the antagonists may also be employed to treat cases of bone marrow failure, for example, aplastic anemia and myelodysplastic syndrome. Any of the above antagonists may be employed in a composition with a pharmaceutically acceptable carrier, e.g., as hereinafter described.
  • the nucleic acid molecules of the present invention are also valuable for chromosome identification.
  • the sequence is specifically targeted to and can hybridize with a particular location on an individual human chromosome.
  • Few chromosome marking reagents based on actual sequence data (repeat polymorphisms) are presently available for marking chromosomal location.
  • the mapping of DNAs to chromosomes according to the present invention is an important first step in correlating those sequences with genes associated with disease.
  • the cDNAs herein disclosed are used to clone genomic DNAs of Nodal and Lefty protein genes. This can be accomplished using a variety of well known techniques and libraries, which generally are available commercially. The genomic DNAs then are used for in situ chromosome mapping using well known techniques for this purpose.
  • sequences can be mapped to chromosomes by preparing PCR primers (preferably 15-25 bp) from the cDNA. Computer analysis of the 3' untranslated region of the gene is used to rapidly select primers that do not span more than one exon in the genomic DNA, thus complicating the amplification process. These primers are then used for PCR screening of somatic cell hybrids containing individual human chromosomes. Fluorescence in situ hybridization ("FISH”) of a cDNA clone to a metaphase chromosomal spread can be used to provide a precise chromosomal location in one step.
  • FISH Fluorescence in situ hybridization
  • the bacterial expression vector pQE9 (pDIO) is used for bacterial expression in this example. (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth,
  • pQE9 encodes ampicillin antibiotic resistance (“Ampr”) and contains a bacterial origin of replication (“ori”), an IPTG inducible promoter, a ribosome binding site (“RBS”), six codons encoding histidine residues that allow affinity purification using nickel-nitrilo-tri-acetic acid (“Ni-NTA”) affinity resin sold by QIAGEN, Inc., supra, and suitable single restriction enzyme cleavage sites. These elements are arranged such that an inserted DNA fragment encoding a polypeptide expresses that polypeptide with the six His residues (i.e., a "6 X His tag”) covalently linked to the amino terminus of that polypeptide.

Landscapes

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

Abstract

L'invention concerne les nouvelles protéines Nodal et Lefty lesquelles sont des membres de la famille du facteur de croissance transformant TGF-β. En particulier, l'invention concerne des molécules d'acide nucléique codant les protéines humaines Nodal et Lefty. L'invention concerne également des polypeptides Nodal et Lefty de même que des vecteurs, des cellules hôtes et des méthodes de recombinaison permettant leur production. De plus, l'invention concerne des méthodes de criblage permettant d'identifier des agonistes et des antagonistes de l'activité Nodal et Lefty. En outre, l'invention concerne des méthodes diagnostiques permettant de détecter la croissance cellulaire ainsi que des troubles relatifs à la différentiation, et des méthodes thérapeutiques de traitement de la croissance cellulaire ainsi que des troubles relatifs à la différentiation.
PCT/US1998/017211 1997-08-21 1998-08-20 Homologues humains nodal et lefty WO1999009198A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU91086/98A AU9108698A (en) 1997-08-21 1998-08-20 Human nodal and lefty homologues

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US5656597P 1997-08-21 1997-08-21
US60/056,565 1997-08-21

Publications (1)

Publication Number Publication Date
WO1999009198A1 true WO1999009198A1 (fr) 1999-02-25

Family

ID=22005262

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/017211 WO1999009198A1 (fr) 1997-08-21 1998-08-20 Homologues humains nodal et lefty

Country Status (2)

Country Link
AU (1) AU9108698A (fr)
WO (1) WO1999009198A1 (fr)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6027917A (en) * 1997-12-10 2000-02-22 Genetics Institute, Inc. Bone morphogenetic protein (BMP)-17 and BMP-18 compositions
WO2002014358A2 (fr) * 2000-08-11 2002-02-21 Eli Lilly And Company Nouvelles proteines secretees et leurs utilisations
WO2002083914A2 (fr) * 2001-04-17 2002-10-24 Pe Corporation (Ny) Proteines humaines secretees isolees, molecules d'acide nucleique codant des proteines humaines secretees et utilisations associees

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
MENO C., ET AL.: "LEFT-RIGHT ASYMMETRIC EXPRESSION OF THE TGFBETA-FAMILY MEMBER LEFTY IN MOUSE EMBRYOS.", NATURE, NATURE PUBLISHING GROUP, UNITED KINGDOM, vol. 381., 9 May 1996 (1996-05-09), United Kingdom, pages 151 - 155., XP002914732, ISSN: 0028-0836, DOI: 10.1038/381151a0 *

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6027917A (en) * 1997-12-10 2000-02-22 Genetics Institute, Inc. Bone morphogenetic protein (BMP)-17 and BMP-18 compositions
US6492493B2 (en) 1997-12-10 2002-12-10 Genetics Institute, Llc Bone morphogenetic protein (BMP)-17 and BMP-18 compositions
US7151086B2 (en) 1997-12-10 2006-12-19 Genetics Institute, Llc Bone morphogenetic protein (BMP)-17 and BMP-18 compositions
WO2002014358A2 (fr) * 2000-08-11 2002-02-21 Eli Lilly And Company Nouvelles proteines secretees et leurs utilisations
WO2002014358A3 (fr) * 2000-08-11 2003-03-13 Lilly Co Eli Nouvelles proteines secretees et leurs utilisations
WO2002083914A2 (fr) * 2001-04-17 2002-10-24 Pe Corporation (Ny) Proteines humaines secretees isolees, molecules d'acide nucleique codant des proteines humaines secretees et utilisations associees
US6482936B1 (en) * 2001-04-17 2002-11-19 Pe Corporation (Ny) Isolated human secreted proteins, nucleic acid molecules encoding human secreted proteins, and uses thereof
WO2002083914A3 (fr) * 2001-04-17 2003-06-19 Pe Corp Ny Proteines humaines secretees isolees, molecules d'acide nucleique codant des proteines humaines secretees et utilisations associees

Also Published As

Publication number Publication date
AU9108698A (en) 1999-03-08

Similar Documents

Publication Publication Date Title
US6372454B2 (en) Nucleic acid molecules encoding Follistatin-3
EP1015488B1 (fr) Proteine de type recepteur d'interleukine 17
US6413933B1 (en) Growth factor HTTER36
AU714484B2 (en) Human vascular endothelial growth factor 2
US20080242604A1 (en) Human Nodal and Lefty Homologues
US20100041136A1 (en) Connective Tissue Growth Factor-2
WO1999003982A1 (fr) Interleukine-20
WO1998007749A1 (fr) Nouveaux facteurs de croissance humains
WO1996039421A1 (fr) Facteur de croissance 3 de l'endothelium vasculaire humain
JP2002508166A (ja) ヒトDendriacおよびBrainiac−3
WO1999009198A1 (fr) Homologues humains nodal et lefty
EP0973888A1 (fr) Facteur de croissance derive de cellules dendritiques
EP0990000A1 (fr) Facteur de croissance 3 du tissu conjonctif
WO1999036565A1 (fr) Ependymine humaine
US7994284B2 (en) Connective tissue growth factor (CTGF-3) polypeptides
EP0891372B1 (fr) Facteur de croissance htter36
AU716100B2 (en) Human vascular endothelial growth factor 3
EP0797592A1 (fr) Stanniocalcine-alpha humaine
WO1998028420A1 (fr) Proteine secretrice de parotide humaine

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
121 Ep: the epo has been informed by wipo that ep was designated in this application
NENP Non-entry into the national phase

Ref country code: KR

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

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

Ref country code: CA