WO1996001845A1 - Growth differentiation factor-11 - Google Patents

Growth differentiation factor-11 Download PDF

Info

Publication number
WO1996001845A1
WO1996001845A1 PCT/US1995/008543 US9508543W WO9601845A1 WO 1996001845 A1 WO1996001845 A1 WO 1996001845A1 US 9508543 W US9508543 W US 9508543W WO 9601845 A1 WO9601845 A1 WO 9601845A1
Authority
WO
WIPO (PCT)
Prior art keywords
gdf
leu
pro
cell
gly
Prior art date
Application number
PCT/US1995/008543
Other languages
French (fr)
Inventor
Se-Jin Lee
Alexandra C. Mcpherron
Original Assignee
The Johns Hopkins University School Of Medicine
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 The Johns Hopkins University School Of Medicine filed Critical The Johns Hopkins University School Of Medicine
Priority to JP8504413A priority Critical patent/JPH10502811A/en
Priority to DE69534468T priority patent/DE69534468T2/en
Priority to EP95925559A priority patent/EP0776337B1/en
Priority to CA002194660A priority patent/CA2194660C/en
Priority to US08/765,875 priority patent/US5914234A/en
Priority to AT95925559T priority patent/ATE305036T1/en
Publication of WO1996001845A1 publication Critical patent/WO1996001845A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/51Bone morphogenetic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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
    • C07K14/475Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the invention relates generally to growth factors and specifically to a new member of the transforming growth factor beta (TGF- ⁇ ) superfamily, which is denoted, growth differentiation factor-11 (GDF-11).
  • TGF- ⁇ transforming growth factor beta
  • GDF-11 growth differentiation factor-11
  • TGF- ⁇ The transforming growth factor ⁇ (TGF- ⁇ ) superfamily encompasses a group of structurally-related proteins which affect a wide range of differentiation processes during embryonic development.
  • the family includes, Mullerian inhibiting substance (MIS), which is required for normal male sex development (Behringer, et al., Nature, 345:167, 1990), Drosophila decapentaplegic (DPP) gene product, which is required for dorsal- ventral axis formation and morphogenesis of the imaginal disks (Padgett, er a/., Nature, 325:81-84, 1987), the Xenopus Vg-1 gene product, which localizes to the vegetal pole of eggs ((Weeks, et al., Cell, 51:861-867, 1987), the activins (Mason, et al., Biochem,
  • TGF- ⁇ s can influence a variety of differentiation processes, including adipogenesis, myogenesis, chondrogenesis, hematopoiesis, and epithelial cell differentiation (for review, see Massague, Cell 49:437. 1987).
  • the proteins of the TGF- ⁇ family are initially synthesized as a large precursor protein which subsequently undergoes proteolytic cleavage at a cluster of basic residues approximately 110-140 amino acids from the C-terminus.
  • the C-terminal regions, or mature regions, of the proteins are all structurally related and the different family members can be classified into distinct subgroups based on the extent of their homology. Although the homologies within particular subgroups range from 70% to 90% amino acid sequence identity, the homologies between subgroups are significantly lower, generally ranging from only 20% to 50%. In each case, the active species appears to be a disulfide-linked dimer of C-terminal fragments.
  • the homodimeric species has been found to be biologically active, but for other family members, like the inhibins (Ling, et al., Nature, 321 :779. 1986) and the TGF- ⁇ s
  • heterodimers have also been detected, and these appear to have different biological properties than the respective homodimers.
  • the present invention provides a cell growth and differentiation factor, GDF-11, a polynucleotide sequence which encodes the factor, and antibodies which are bind to the factor.
  • GDF-11 a cell growth and differentiation factor
  • This factor appears to relate to various cell proliferative disorders, especially those involving muscle, neural, and uterine cells, as well as disorders related to the function of the immune system.
  • the invention provides a method for detecting a cell proliferative disorder of muscle, neural, uterine, spleen, or thymus origin and which is associated with GDF-11.
  • the invention provides a method for treating a cell proliferative or immunologic disorder by suppressing or enhancing GDF-11 activity.
  • FIGURE 1 shows the nucleotide and predicted amino acid sequences of murine (FIGURE 1a) and human (FIGURE 1b) GDF-11.
  • the putative proteolytic processing sites are shown by the shaded boxes.
  • the potential N-linked glycosylation signal is shown by the open box, and the consensus polyadenylation signal is underlined; the poly A tail is not shown.
  • FIGURE 2 shows Northern blots of RNA prepared from adult (FIGURE 2a) or fetal and neonatal (FIGURE 2b) tissues probed with a murine GDF-11 probe.
  • FIGURE 3 shows amino acid homologies among different members of the TGF- ⁇ superfamily. Numbers represent percent amino acid identities between each pair calculated from the first conserved cysteine to the C-terminus. Boxes represent homologies among highly-related members within particular subgroups.
  • FIGURE 4 shows an alignment of the predicted amino acid sequences of human GDF-11 (top lines) with human GDF-8 (bottom lines). Vertical lines indicate identities. Dots represent gaps introduced in order to maximize the alignment. Numbers represent amino acid positions relative to the N-terminus. The putative proteolytic processing sites are shown by the open box. The conserved cysteine residues on the C-terminal region are shown by the shaded boxes.
  • FIGURE 5 shows the expression of GDF-11 in mammalian cells.
  • Conditioned medium prepared from Chinese hamster ovary cells transfected with a hybrid GDF-8/GDF-11 gene (see text) cloned into the MSXND expression vector in either the antisense (lane 1) or sense (lane 2) orientation was dialyzed, lyophilized, and subjected to Western analysis using antibodies directed against the C-terminal portion of GDF-8 protein.
  • Arrows at right indicate the putative unprocessed (pro-GDF-8/GDF-11) or processed GDF-11 proteins. Numbers at left indicate mobilities of molecular weight standards.
  • FIGURE 6 shows the chromosomal mapping of human GDF-11.
  • DNA samples prepared from human/rodent somatic cell lines were subjected to PCR, electrophoresed on agarose gels, blotted, and probed.
  • the human chromosome contained in each of the hybrid cell lines is identified at the top of each of the first 24 lanes (1-22, X, and Y).
  • the starting DNA template was total genomic DNA from hamster, mouse, and human sources, respectively.
  • the lane marked B1 no template DNA was used. Numbers at left indicate the mobilities of DNA standards.
  • FIGURE 7 shows the FISH localization of GDF-11.
  • Metaphase chromosomes derived from peripheral blood lymphocytes were hybridized with digoxigenin-labelled human GDF-11 probe (a) or a mixture of human GDF-11 genomic and chromosome 12-specific centromere probes (b) and analyzed as described in the text.
  • a schematic showing the location of GDF-11 at position 12q13 is shown in panel (c).
  • FIGURE 8 shows the nucleotide and deduced amino acid sequence of murine GDF-8.
  • the present invention provides a growth and differentiation factor, GDF-11, and a polynucleotide sequence encoding GDF-11.
  • GDF-11 is expressed at highest levels in muscle, brain, uterus, spleen, and thymus and at lower levels in other tissues.
  • the invention provides a method for detection of a cell proliferative or immunologic disorder of muscle, neural, uterine, spleen, or thymus origin which is associated with GDF-11 expression or function.
  • the invention provides a method for treating a cell proliferative or immunologic disorder by using an agent which suppresses or enhances GDF-11 activity.
  • the TGF- ⁇ superfamily consists of multifunctional polypeptides that control proliferation, differentiation, and other functions in many cell types. Many of the peptides have regulatory, both positive and negative, effects on other peptide growth factors.
  • GDNF a potent neurotrophic factor that can promote the survival of dopaminergic neurons
  • Another family member namely dorsalin-1
  • dorsalin-1 is capable of promoting the differentiation of neural crest cells (Basler, etal., Cell, 73:687, 1993).
  • the inhibins and activins have been shown to be expressed in the brain (Meunier, et al., Proc. Natl. Acad.
  • GDF-1 nerve cell survival molecule
  • Another family member, namely GDF-1 is nervous system-specific in its expression pattern (Lee, Proc. Nat'l. Acad. Sci., USA, 88:4250, 1991), and certain other family members, such as Vgr-1 (Lyons, et al., Proc. Natl Acad.
  • GDF-11 GDF-11 may also possess activities that relate to the function of the nervous system.
  • skeletal muscle produces a factor or factors that promote the survival of motor neurons (Brown, Trends Neurosci., 7:10, 1984).
  • GDF-11 may have neurotrophic activities for other neuronal populations.
  • GDF-11 may have in vitro and in vivo applications in the treatment of neurodegenerative diseases, such as amyotrophic lateral sclerosis, or in maintaining cells or tissues in culture prior to transplantation.
  • GDF-11 may also have applications in treating disease processes involving muscle, such as in musculodegenerative diseases or in tissue repair due to trauma.
  • many other members of the TGF- ⁇ family are also important mediators of tissue repair.
  • TGF- ⁇ has been shown to have marked effects on the formation of collagen and to cause a striking angiogenic response in the newborn mouse (Roberts, et al., Proc. Natl.
  • TGF- ⁇ has also been shown to inhibit the differentiation of myoblasts in culture (Massague, et al., Proc. Natl. Acad. Sci., USA 83.: 8206, 1986).
  • myoblast cells may be used as a vehicle for delivering genes to muscle for gene therapy, the properties of GDF-11 could be exploited for maintaining cells prior to transplantation or for enhancing the efficiency of the fusion process.
  • GDF-11 may also have applications in the treatment of immunologic disorders.
  • TGF- ⁇ has been shown to have a wide range of immunoregulatory activities, including potent suppressive effects on B and T cell proliferation and function (for review, see Palladino, et al., Ann. N. Y. Acad. Sci., 593:181 , 1990).
  • the expression of GDF-11 in spleen and thymus suggests that GDF-11 may possess similar activities and therefore, may be used as an anti-inflammatory agent or as a treatment for disorders related to abnormal proliferation or function of lymphocytes.
  • substantially pure refers to GDF-11 which is substantially free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated.
  • One skilled in the art can purify GDF-11 using standard techniques for protein purification.
  • the substantially pure polypeptide will yield a single major band on a non-reducing polyacrylamide gel.
  • the purity of the GDF-11 polypeptide can also be determined by amino-terminal amino acid sequence analysis.
  • GDF-11 polypeptide includes functional fragments of the polypeptide, as long as the activity of GDF-11 remains. Smaller peptides containing the biological activity of GDF-11 are included in the invention.
  • the invention provides polynucleotides encoding the GDF-11 protein. These polynucleotides include DNA, cDNA and RNA sequences which encode GDF-11. It is understood that all polynucleotides encoding all or a portion of GDF-11 are also included herein, as long as they encode a polypeptide with GDF-11 activity. Such polynucleotides include naturally occurring, synthetic, and intentionally manipulated polynucleotides. For example, GDF-11 polynucleotide may be subjected to site-directed mutagenesis. The polynucleotide sequence for GDF-11 also includes antisense sequences.
  • the polynucleotides of the invention include sequences that are degenerate as a result of the genetic code. There are 20 natural amino acids, most of which are specified by more than one codon. Therefore, all degenerate nucleotide sequences are included in the invention as long as the amino acid sequence of GDF-11 polypeptide encoded by the nucleotide sequence is functionally unchanged.
  • a DNA sequence containing the human GDF-11 gene The sequence contains an open reading frame encoding a polypeptide 407 amino acids in length.
  • the sequence contains a putative RXXR proteolytic cleavage site at amino acids 295-298.
  • Cleavage of the precursor at this site would generate an active C- terminal fragment 109 amino acids in length with a predicted molecular weight of approximately 12,500 kD.
  • a partial murine genomic sequence Preferably, the human GDF-11 nucleotide sequence is SEQ ID NO:1 and the mouse nucleotide sequence is SEQ ID NO:3.
  • the polynucleotide encoding GDF-11 includes SEQ ID NO:1 and 3, as well as nucleic acid sequences complementary to SEQ ID NO's:1 and 3.
  • a complementary sequence may include an antisense nucleotide.
  • the sequence is RNA
  • the deoxynucieotides A, G, C, and T of SEQ ID NO:1 and 3 are replaced by ribonucleotides A, G, C, and U, respectively.
  • fragments of the above-described nucleic acid sequences that are at least 15 bases in length, which is sufficient to permit the fragment to selectively hybridize to DNA that encodes the protein of SEQ ID NO: 2 or 4 under physiological conditions.
  • GDF-11 The C-terminal region of GDF-11 following the putative proteolytic processing site shows significant homology to the known members of the TGF- ⁇ superfamily.
  • the GDF-11 sequence contains most of the residues that are highly conserved in other family members (see FIGURE 1). Like the TGF- ⁇ s and inhibin ⁇ s, GDF-11 contains an extra pair of cysteine residues in addition to the 7 cysteines found in virtually all other family members.
  • GDF-11 is most homologous to GDF-8 (92% sequence identity) (see FIGURE 3).
  • Minor modifications of the recombinant GDF-11 primary amino acid sequence may result in proteins which have substantially equivalent activity as compared to the GDF-11 polypeptide described herein. Such modifications may be deliberate, as by site-directed mutagenesis, or may be spontaneous. All of the polypeptides produced by these modifications are included herein as long as the biological activity of GDF-11 still exists. Further, deletion of one or more amino acids can also result in a modification of the structure of the resultant molecule without significantly altering its biological, activity. This can lead to the development of a smaller active molecule which would have broader utility. For example, one can remove amino or carboxy terminal amino acids which are not required for GDF-11 biological activity.
  • the nucleotide sequence encoding the GDF-11 polypeptide of the invention includes the disclosed sequence and conservative variations thereof.
  • conservative variation denotes the replacement of an amino acid residue by another, biologically similar residue. Examples of conservative variations include the substitution of one hydrophobic residue such as isoleucine, valine, leucine or methionine for another, or the substitution of one polar residue for another, such as the substitution of arginine for lysine, glutamicfor aspartic acid, or glutamine for asparagine, and the like.
  • conservative variation also includes the use of a substituted amino acid in place of an unsubstituted parent amino acid provided that antibodies raised to the substituted polypeptide also immunoreact with the unsubstituted polypeptide.
  • DNA sequences of the invention can be obtained by several methods.
  • the DNA can be isolated using hybridization techniques which are well known in the art. These include, but are not limited to: 1) hybridization of genomic or cDNA libraries with probes to detect homologous nucleotide sequences, 2) polymerase chain reaction (PCR) on genomic DNA or cDNA using primers capable of annealing to the DNA sequence of interest, and 3) antibody screening of expression libraries to detect cloned DNA fragments with shared structural features.
  • hybridization techniques which are well known in the art. These include, but are not limited to: 1) hybridization of genomic or cDNA libraries with probes to detect homologous nucleotide sequences, 2) polymerase chain reaction (PCR) on genomic DNA or cDNA using primers capable of annealing to the DNA sequence of interest, and 3) antibody screening of expression libraries to detect cloned DNA fragments with shared structural features.
  • the GDF-11 polynucleotide of the invention is derived from a mammalian organism, and most preferably from a mouse, rat, or human. Screening procedures which rely on nucleic acid hybridization make it possible to isolate any gene sequence from any organism, provided the appropriate probe is available. Oligonucleotide probes, which correspond to a part of the sequence encoding the protein in question, can be synthesized chemically. This requires that short, oligopeptide stretches of amino acid sequence must be known. The DNA sequence encoding the protein can be deduced from the genetic code, however, the degeneracy of the code must be taken into account. It is possible to perform a mixed addition reaction when the sequence is degenerate.
  • hybridization is preferably performed on either single-stranded DNA or denatured double-stranded DNA.
  • Hybridization is particularly useful in the detection of cDNA clones derived from sources where an extremely low amount of mRNA sequences relating to the polypeptide of interest are present.
  • stringent hybridization conditions directed to avoid non-specific binding, it is possible, for example, to allow the autoradiographic visualization of a specific cDNA clone by the hybridization of the target DNA to that single probe in the mixture which is its complete complement (Wallace, et al., Nucl. Acid Res., 9:879, 1981 ; Maniatis, et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, N.Y. 1989).
  • DNA sequences encoding GDF-11 can also be obtained by: 1) isolation of double-stranded DNA sequences from the genomic DNA; 2) chemical manufacture of a DNA sequence to provide the necessary codons for the polypeptide of interest; and 3) in vitro synthesis of a double-stranded DNA sequence by reverse transcription of mRNA isolated from a eukaryotic donor cell. In the latter case, a double- stranded DNA complement of mRNA is eventually formed which is generally referred to as cDNA.
  • genomic DNA isolates are the least common. This is especially true when it is desirable to obtain the microbial expression of mammalian polypeptides due to the presence of introns.
  • the synthesis of DNA sequences is frequently the method of choice when the entire sequence of amino acid residues of the desired polypeptide product is known. When the entire sequence of amino acid residues of the desired polypeptide is not known, the direct synthesis of DNA sequences is not possible and the method of choice is the synthesis of cDNA sequences.
  • plasmid- or phage-carrying cDNA libraries which are derived from reverse transcription of mRNA which is abundant in donor cells that have a high level of genetic expression.
  • phage-carrying cDNA libraries which are derived from reverse transcription of mRNA which is abundant in donor cells that have a high level of genetic expression.
  • the production of labeled single or double-stranded DNA or RNA probe sequences duplicating a sequence putatively present in the target cDNA may be employed in DNA/DNA hybridization procedures which are carried out on cloned copies of the cDNA which have been denatured into a single-stranded form (Jay, et al., Nucl. Acid Res., 11:2325, 1983).
  • a cDNA expression library such as lambda gt11
  • Such antibodies can be either polyclonally or monoclonally derived and used to detect expression product indicative of the presence of GDF-11 cDNA.
  • DNA sequences encoding GDF-11 can be expressed in vitro by DNA transfer into a suitable host cell.
  • "Host cells” are cells in which a vector can be propagated and its DNA expressed.
  • the term also includes any progeny of the subject host cell. It is understood that all progeny may not be identical to the parental cell since there may be mutations that occur during replication. However, such progeny are included when the term "host cell” is used. Methods of stable transfer, meaning that the foreign DNA is continuously maintained in the host, are known in the art.
  • the GDF-11 polynucleotide sequences may be inserted into a recombinant expression vector.
  • recombinant expression vector refers to a plasmid, virus or other vehicle known in the art that has been manipulated by insertion or incorporation of the GDF-11 genetic sequences.
  • Such expression vectors contain a promoter sequence which facilitates the efficient transcription of the inserted genetic sequence of the host.
  • the expression vector typically contains an origin of replication, a promoter, as well as specific genes which allow phenotypic selection of the transformed cells.
  • Vectors suitable for use in the present invention include, but are not limited to the T7-based expression vector for expression in bacteria (Rosenberg, et al., Gene, 56:125, 1987), the pMSXND expression vector for expression in mammalian cells
  • the DNA segment can be present in the vector operably linked to regulatory elements, for example, a promoter (e.g., T7, metallothionein I, or polyhedrin promoters).
  • a promoter e.g., T7, metallothionein I, or polyhedrin promoters.
  • Polynucleotide sequences encoding GDF-11 can be expressed in either prokaryotes or eukaryotes.
  • Hosts can include microbial, yeast, insect and mammalian organisms. Methods of expressing DNA sequences having eukaryotic or viral sequences in prokaryotes are well known in the art.
  • Biologically functional viral and plasmid DNA vectors capable of expression and replication in a host are known in the art. Such vectors are used to incorporate DNA sequences of the invention.
  • the mature sequences of the invention Preferably, the mature sequences of the invention.
  • GDF-11 is expressed from a DNA clone containing the entire coding sequence of GDF-11.
  • the C-terminal portion of GDF-11 can be expressed as a fusion protein with the pro- region of another member of the TGF- ⁇ family or co ⁇ expressed with another pro- region (see for example, Hammonds, et al., Molec. Endocrin. 5:149, 1991; Gray, A., and Mason, A., Science, 247:1328, 1990).
  • Transformation of a host cell with recombinant DNA may be carried out by conventional techniques as are well known to those skilled in the art.
  • the host is prokaryotic, such as £ coli
  • competent cells which are capable of DNA uptake can be prepared from cells harvested after exponential growth phase and subsequently treated by the CaCI 2 method using procedures well known in the art. Altematively, MgCI 2 or RbCI can be used. Transformation can also be performed after forming a protoplast of the host cell if desired.
  • Eukaryotic cells can also be cotransformed with DNA sequences encoding the GDF-11 of the invention, and a second foreign DNA molecule encoding a selectable phenotype, such as the herpes simplex thymidine kinase gene.
  • Another method is to use a eukaryotic viral vector, such as simian virus 40 (SV40) or bovine papilloma virus, to transiently infect or transform eukaryotic cells and express the protein, (see for example, Eukaryotic Viral Vectors, Cold Spring Harbor Laboratory, Gluzman ed., 1982).
  • a eukaryotic viral vector such as simian virus 40 (SV40) or bovine papilloma virus
  • Isolation and purification of microbial expressed polypeptide, or fragments thereof, provided by the invention may be carried out by conventional means including preparative chromatography and immunological separations involving monoclonal or polyclonal antibodies.
  • the GDF-11 polypeptides of the invention can also be used to produce antibodies which are immunoreactive or bind to epitopes of the GDF-11 polypeptides.
  • Antibody which consists essentially of pooled monoclonal antibodies with different epitopic specificities, as well as distinct monoclonal antibody preparations are provided.
  • Monoclonal antibodies are made from antigen containing fragments of the protein by methods well known in the art (Kohler, et al., Nature, 256:495, 1975; Current Protocols in Molecular Biology, Ausubel, et al., ed., 1989).
  • antibody as used in this invention includes intact molecules as well as fragments thereof, such as Fab, F(ab') 2 , and Fv which are capable of binding the epitopic determinant. These antibody fragments retain some ability to selectively bind with its antigen or receptor and are defined as follows:
  • Fab the fragment which contains a monovalent antigen-binding fragment of an antibody molecule can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain;
  • Fab 1 the fragment of an antibody molecule can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain; two Fab' fragments are obtained per antibody molecule;
  • Fv defined as a genetically engineered fragment containing the variable region of the light chain and the variable region of the heavy chain expressed as two chains
  • Single chain antibody defined as a genetically engineered molecule containing the variable region of the light chain, the variable region of the heavy chain, linked by a suitable polypeptide linker as a genetically fused single chain molecule.
  • epitopic determinants means any antigenic determinant on an antigen to which the paratope of an antibody binds.
  • Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
  • Antibodies which bind to the GDF-11 polypeptide of the invention can be prepared using an intact polypeptide or fragments containing small peptides of interest as the immunizing antigen.
  • the polypeptide or a peptide used to immunize an animal can be derived from translated cDNA or chemical synthesis which can be conjugated to a carrier protein, if desired.
  • Such commonly used carriers which are chemically coupled to the peptide include keyhole limpet hemocyanin (KLH), thyroglobulin, bovine serum albumin (BSA), and tetanus toxoid.
  • KLH keyhole limpet hemocyanin
  • BSA bovine serum albumin
  • the coupled peptide is then used to immunize the animal (e.g., a mouse, a rat, or a rabbit).
  • polyclonal or monoclonal antibodies can be further purified, for example, by binding to and elution from a matrix to which the polypeptide or a peptide to which the antibodies were raised is bound.
  • a matrix to which the polypeptide or a peptide to which the antibodies were raised is bound.
  • Those of skill in the art will know of various techniques common in the immunology arts for purification and/or concentration of polyclonal antibodies, as well as monoclonal antibodies (See for example, Coligan, et al., Unit 9, Current Protocols in Immunology, Wiley Interscience, 1991 , incorporated by reference).
  • an anti-idiotypic monoclonal antibody made to a first monoclonal antibody will have a binding domain in the hypervariable region which is the "image" of the epitope bound by the first monoclonal antibody.
  • cell-proliferative disorder denotes malignant as well as non-malignant cell populations which often appear to differ from the surrounding tissue both morphologically and genotypically. Malignant cells (i.e. cancer) develop as a result of a multistep process.
  • the GDF-11 polynucleotide that is an antisense molecule is useful in treating malignancies of the various organ systems, particularly, for example, cells in muscle, uterus, spleen, thymus, or neural tissue.
  • any disorder which is etiologically linked to altered expression of GDF-11 could be considered susceptible to treatment with a GDF-11 suppressing reagent.
  • One such disorder is a malignant cell proliferative disorder, for example.
  • the invention provides a method for detecting a cell proliferative disorder of muscle, uterine or neural tissue, for example, which comprises contacting an anti-GDF-11 antibody with a cell suspected of having a GDF-11 associated disorder and detecting binding to the antibody.
  • the antibody reactive with GDF-11 is labeled with a compound which allows detection of binding to GDF-11.
  • an antibody specific for GDF-11 polypeptide may be used to detect the level of GDF-11 in biological fluids and tissues. Any specimen containing a detectable amount of antigen can be used.
  • a preferred sample of this invention is muscle, uterus, spleen, thymus, or neural tissue.
  • the level of GDF-11 in the suspect cell can be compared with the level in a normal cell to determine whether the subject has a GDF-11 -associated cell proliferative disorder.
  • the subject is human.
  • the antibodies of the invention can be used in any subject in which it is desirable to administer in vitro or in vivo immunodiagnosis or immunotherapy.
  • the antibodies of the invention are suited for use, for example, in immunoassays in which they can be utilized in liquid phase or bound to a solid phase carrier.
  • the antibodies in these immunoassays can be detectably labeled in various ways.
  • types of immunoassays which can utilize antibodies of the invention are competitive and non- competitive immunoassays in either a direct or indirect format. Examples of such immunoassays are the radioimmunoassay (RIA) and the sandwich (immunometric) assay.
  • Detection of the antigens using the antibodies of the invention can be done utilizing immunoassays which are run in either the forward, reverse, or simultaneous modes, including immunohistochemical assays on physiological samples. Those of skill in the art will know, or can readily discern, other immunoassay formats without undue experimentation.
  • the antibodies of the invention can be bound to many different carriers and used to detect the presence of an antigen comprising the polypeptide of the invention.
  • carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, agaroses and magnetite.
  • the nature of the carrier can be either soluble or insoluble for purposes of the invention. Those skilled in the art will know of other suitable carriers for binding antibodies, or will be able to ascertain such, using routine experimentation.
  • labels and methods of labeling known to those of ordinary skill in the art.
  • Examples of the types of labels which can be used in the present invention include enzymes, radioisotopes, fluorescent compounds, colloidal metals, chemiluminescent compounds, phosphorescent compounds, and bioluminescent compounds.
  • Those of ordinary skill in the art will know of other suitable labels for binding to the antibody, or will be able to ascertain such, using routine experimentation.
  • Another technique which may also result in greater sensitivity consists of coupling the antibodies to low molecular weight haptens. These haptens can then be specifically detected by means of a second reaction. For example, it is common to use such haptens as biotin, which reacts with avidin, or dinitrophenyl, puridoxal, and fluorescein, which can react with specific antihapten antibodies.
  • the detectably labeled antibody is given a dose which is diagnostically effective.
  • diagnostically effective means that the amount of detectably labeled monoclonal antibody is administered in sufficient quantity to enable detection of the site having the antigen comprising a polypeptide of the invention for which the monoclonal antibodies are specific.
  • concentration of detectably labeled monoclonal antibody which is administered should be sufficient such that the binding to those cells having the polypeptide is detectable compared to the background. Further, it is desirable that the detectably labeled monoclonal antibody be rapidly cleared from the circulatory system in order to give the best target-to-background signal ratio.
  • the dosage of detectably labeled monoclonal antibody for in vivo diagnosis will vary depending on such factors as age, sex, and extent of disease of the individual. Such dosages may vary, for example, depending on whether multiple injections are given, antigenic burden, and other factors known to those of skill in the art.
  • a radioisotope used for in vivo imaging will lack a particle emission, but produce a large number of photons in the 140-250 keV range, which may readily be detected by conventional gamma cameras.
  • radioisotopes may be bound to immunoglobulin either directly or indirectly by using an intermediate functional group.
  • Intermediate functional groups which often are used to bind radioisotopes which exist as metallic ions to immunoglobulins are the bifunctional chelating agents such as diethylenetriaminepentacetic acid (DTPA) and ethylenediaminetetraacetic acid (EDTA) and similar molecules.
  • DTPA diethylenetriaminepentacetic acid
  • EDTA ethylenediaminetetraacetic acid
  • metallic ions which can be bound to the monoclonal antibodies of the invention are 11 ln, 97 Ru, 67 Ga, 68 Ga, 72 As, 89 Zr, and 201 TI.
  • the monoclonal antibodies of the invention can also be labeled with a paramagnetic isotope for purposes of in vivo diagnosis, as in magnetic resonance imaging (MRI) or electron spin resonance (ESR).
  • MRI magnetic resonance imaging
  • ESR electron spin resonance
  • any conventional method for visualizing diagnostic imaging can be utilized.
  • gamma and positron emitting radioisotopes are used for camera imaging and paramagnetic isotopes for MRI.
  • Elements which are particularly useful in such techniques include 157 Gd, 5 Mn, 162 Dy, 52 Cr, and ⁇ Fe.
  • the monoclonal antibodies of the invention can be used in vitro and in vivo to monitor the course of amelioration of a GDF-11 -associated disease in a subject.
  • ameliorate denotes a lessening of the detrimental effect of the
  • GDF-11 -associated disease in the subject receiving therapy is a GDF-11 -associated disease in the subject receiving therapy.
  • the present invention identifies a nucleotide sequence that can be expressed in an altered manner as compared to expression in a normal cell, therefore it is possible to design appropriate therapeutic or diagnostic techniques directed to this sequence.
  • a cell-proliferative disorder is associated with the expression of GDF-11
  • nucleic acid sequences that interfere with GDF-11 expression at the translational level can be used.
  • This approach utilizes, for example, antisense nucleic acid and ribozymes to block translation of a specific GDF-11 mRNA, either by masking that mRNA with an antisense nucleic acid or by cleaving it with a ribozyme.
  • disorders include neurodegenerative diseases, for example.
  • Antisense nucleic acids are DNA or RNA molecules that are complementary to at least a portion of a specific mRNA molecule (Weintraub, Scientific American, 262:40, 1990). In the cell, the antisense nucleic acids hybridize to the corresponding mRNA, forming a double-stranded molecule. The antisense nucleic acids interfere with the translation of the mRNA, since the cell will not translate a mRNA that is double-stranded. Antisense oligomers of about 15 nucleotides are preferred, since they are easily synthesized and are less likely to cause problems than larger molecules when introduced into the target GDF-11 -producing cell. The use of antisense methods to inhibit the in vitro translation of genes is well known in the art (Marcus-Sakura, Anal.Biochem., 172:289, 1988).
  • Ribozymes are RNA molecules possessing the ability to specifically cleave other single- stranded RNA in a manner analogous to DNA restriction endonucleases. Through the modification of nucleotide sequences which encode these RNAs, it is possible to engineer molecules that recognize specific nucleotide sequences in an RNA molecule and cleave it (Cech, J.Amer.Med. Assn., 260:3030. 1988). A major advantage of this approach is that, because they are sequence-specific, only mRNAs with particular sequences are inactivated.
  • ribozymes There are two basic types of ribozymes namely, tetrahymena-type (Hasselhoff, Nature,
  • Tetrahymena-type ribozymes recognize sequences which are four bases in length, while “hammerhead”-type ribozymes recognize base sequences 11-18 bases in length. The longer the recognition sequence, the greater the likelihood that the sequence will occur exclusively in the target mRNA species. Consequently, hammerhead-type ribozymes are preferable to tetrahymena- type ribozymes for inactivating a specific mRNA species and 18-based recognition sequences are preferable to shorter recognition sequences.
  • the present invention also provides gene therapy for the treatment of cell proliferative or immunologic disorders which are mediated by GDF-11 protein. Such therapy would achieve its therapeutic effect by introduction of the GDF-11 antisense polynucleotide into cells having the proliferative disorder. Delivery of antisense GDF-11 polynucleotide can be achieved using a recombinant expression vector such as a chimeric virus or a colloidal dispersion system. Especially preferred for therapeutic delivery of antisense sequences is the use of targeted liposomes.
  • RNA virus such as a retrovirus
  • retroviral vector is a derivative of a murine or avian retrovirus.
  • retroviral vectors in which a single foreign gene can be inserted include, but are not limited to: Moloney murine leukemia virus (MoMuLV), Harvey murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV), and Rous Sarcoma Virus (RSV).
  • MoMuLV Moloney murine leukemia virus
  • HaMuSV Harvey murine sarcoma virus
  • MuMTV murine mammary tumor virus
  • RSV Rous Sarcoma Virus
  • Retroviral vectors can be made target specific by attaching, for example, a sugar, a glycolipid, or a protein.
  • Preferred targeting is accomplished by using an antibody to target the retroviral vector.
  • an antibody to target the retroviral vector.
  • Those of skill in the art will know of, or can readily ascertain without undue experimenta ⁇ tion, specific polynucleotide sequences which can be inserted into the retroviral genome or attached to a viral envelope to allow target specific delivery of the retroviral vector containing the GDF-11 antisense polynucleotide.
  • helper cell lines that contain plasmids encoding all of the structural genes of the retrovirus under the control of regulatory sequences within the LTR. These plasmids are missing a nucleotide sequence which enables the packaging mechanism to recognize an RNA transcript for encapsidation.
  • Helper cell lines which have deletions of the packaging signal include, but are not limited to ⁇ 2, PA317 and PA12, for example. These cell lines produce empty virions, since no genome is packaged. If a retroviral vector is introduced into such cells in which the packaging signal is intact, but the structural genes are replaced by other genes of interest, the vector can be packaged and vector virion produced.
  • NIH 3T3 or other tissue culture cells can be directly transfected with plasmids encoding the retroviral structural genes gag, pol and env, by conventional calcium phosphate transfection. These cells are then transfected with the vector plasmid containing the genes of interest. The resulting cells release the retroviral vector into the culture medium.
  • colloidal dispersion systems include macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes.
  • the preferred colloidal system of this invention is a iiposome.
  • Liposomes are artificial membrane vesicles which are useful as delivery vehicles in vitro and in vivo. It has been shown that large unilamellar vesicles ( UV), which range in size from 0.2-4.0 m can encapsulate a substantial percentage of an aqueous buffer containing large macromolecules.
  • RNA, DNA and intact virions can be encapsulated within the aqueous interior and be delivered to cells in a biologically active form (Fraley, et al., Trends Biochem. Sci., 6:77, 1981).
  • liposomes In addition to mammalian cells, liposomes have been used for delivery of polynucleotides in plant, yeast and bacterial cells.
  • Iiposome In order for a Iiposome to be an efficient gene transfer vehicle, the following characteristics should be present: (1) encapsulation of the genes of interest at high efficiency while not compromising their biological activity; (2) preferential and substantial binding to a target cell in comparison to non-target cells; (3) delivery of the aqueous contents of the vesicle to the target cell cytoplasm at high efficiency; and (4) accurate and effective expression of genetic information (Mannino, et al., Biotechniques,
  • the composition of the Iiposome is usually a combination of phospholipids, particularly high-phase-transition-temperature phospholipids, usually in combination with steroids, especially cholesterol. Other phospholipids or other lipids may also be used.
  • the physical characteristics of liposomes depend on pH, ionic strength, and the presence of divalent cations. Examples of lipids useful in Iiposome production include phosphatidyl compounds, such as phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidyletha- nolamine, sphingolipids, cerebrosides, and gangliosides.
  • diacylphosphatidylglycerols where the lipid moiety contains from 14-18 carbon atoms, particulariy from 16-18 carbon atoms, and is saturated.
  • Illustrative phospholipids include egg phosphatidylcholine, dipalmitoylphosphatidylcholine and distearoylphos- phatidylcholine.
  • the targeting of liposomes can be classified based on anatomical and mechanistic factors.
  • Anatomical classification is based on the level of selectivity, for example, organ- specific, cell-specific, and organelle-specific.
  • Mechanistic targeting can be distinguished based upon whether it is passive or active. Passive targeting utilizes the natural tendency of liposomes to distribute to cells of the reticulo-endothelial system (RES) in organs which contain sinusoidal capillaries.
  • RES reticulo-endothelial system
  • Active targeting involves alteration of the Iiposome by coupling the Iiposome to a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein, or by changing the composition or size of the Iiposome in order to achieve targeting to organs and cell types other than the naturally occurring sites of localization.
  • a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein
  • the surface of the targeted delivery system may be modified in a variety of ways.
  • lipid groups can be incorporated into the lipid bilayer of the Iiposome in order to maintain the targeting ligand in stable association with the liposomal bilayer.
  • Various linking groups can be used for joining the lipid chains to the targeting ligand.
  • GDF-11 Due to the expression of GDF-11 in muscle, spleen, uterus, thymus, and neural tissue, there are a variety of applications using the polypeptide, polynucleotide, and antibodies of the invention, related to these tissues. Such applications include treatment of cell proliferative and immunologic disorders involving these and other tissues.
  • GDF-11 may be useful in various gene therapy procedures. The following examples are intended to illustrate but not limit the invention. While they are typical of those that might be used, other procedures known to those skilled in the art may alternatively be used.
  • a murine genomic library was screened at reduced stringency using a murine GDF-8 probe (FIGURE 8; nucleotides 865-1234) spanning the region encoding the C-terminal portion of the GDF-8 precursor protein.
  • Hybridization was carried out as described (Lee, Mol. Endocrinol., 4:1034, 1990) at 65°C, and the final wash was carried out at the same temperature in a buffer containing 0.5 M NaCI.
  • the hybridizing phage was one that could be distinguished from GDF-8-containing phage on the basis of its reduced hybridization intensity to the GDF-8 probe. Partial nucleotide sequence analysis of the genomic insert present in this weakly hybridizing phage showed that this clone contained a sequence highly related to but distinct from murine GDF-8.
  • FIGURE 1a A partial nucleotide sequence of the genomic insert present in this phage is shown in FIGURE 1a.
  • the sequence contained an open reading frame extending from nucleotides 198 to 575 that showed significant homology to the known members of the TGF- ⁇ superfamily (see below). Preceding this sequence was a 3' splice consensus sequence at precisely the same position as in the GDF-8 gene. This new TGF- ⁇ family member was given the designation GDF-11 (growth/differentiation factor-11).
  • GDF-11 growth/differentiation factor-11
  • RNA samples prepared from a variety of tissues were screened by Northern analysis. RNA isolation and Northern analysis were carried out as described previously (Lee, Mol. Endocrinol., 4:1034, 1990) except that the hybridization was carried out in 5X SSPE, 10% dextran sulfate, 50% formamide, 1 % SDS, 200 g/ml salmon DNA, and 0.1% each of bovine serum albumin, ficoll, and polyvinylpyrrolidone.
  • RNA Five micrograms of twice poly A-selected RNA prepared from each tissue (except for 2 day neonatal brain, for which only 3.3 ⁇ g RNA were used) were electrophoresed on formaldehyde gels, blotted, and probed with GDF-11. As shown in
  • FIGURE 2 the GDF-11 probe detected two RNA species, approximately 4.2 and 3.2 kb in length, in adult thymus, brain, spleen, uterus, and muscle as well as in whole embryos isolated at day 12.5 or 18.5 and in brain samples taken at various stages of development. On longer exposures of these blots, lower levels of GDF-11 RNA could also be detected in a number of other tissues.
  • a cDNA library was prepared in the lambda ZAP II vector (Stratagene) using RNA prepared from human adult spleen. From 5 ⁇ g of twice poly A-selected RNA prepared from human spleen, a cDNA library consisting of 21 million recombinant phage was constructed according to the instructions provided by Stratagene. The library was screened without amplification. Library screening and characterization of cDNA inserts were carried out as described previously (Lee, Mol. Endocrinol., 4:1034, 1990). From this library, 23 hybridizing phage were obtained.
  • the entire nucleotide sequence of the clone extending furthest toward the 5' end of the gene was determined.
  • the 1258 base pair sequence contained a single long open reading frame beginning from the 5' end of the clone and extending to a TAA stop codon. Because the open reading frame and the homology with GDF-8 (see below) extended to the very 5' end of the clone, it seemed likely that this clone was missing the coding sequence corresponding to the N-terminal portion of the GDF-11 precursor protein. In order to obtain the remaining portion of the GDF-11 sequence, several genomic clones were isolated by screening a human genomic library with the human GDF-11 cDNA probe.
  • FIGURE 1b shows the predicted sequence of GDF-11 assembled from the genomic and cDNA sequences.
  • Nucleotides 136 to 1393 represent the extent of the sequence obtained from a cDNA clone.
  • Nucleotides 1 to 135 were obtained from a genomic clone. The sequence has been arbitrarily numbered beginning with a Sac II site present in the genomic clone, but the location of the mRNA start site is not known. The sequence contains a putative initiating methionine at nucleotide 54.
  • the open reading frame extends for 407 amino acids.
  • the sequence contains one potential N-linked glycosylation site at asparagine 94.
  • the sequence contains a predicted RXXR proteolytic cleavage site at amino acids 295 to 298, and cleavage of the precursor at this site would generate an active C-terminal fragment 109 amino acids in length with a predicted molecular weight of approximately 12,500 kD.
  • the predicted murine and human GDF-11 amino acid sequences are 100% identical. The high degree of sequence conservation across species suggests that GDF-11 plays an important role in vivo.
  • GDF-11 contains most of the residues that are highly conserved in other family members, including the seven cysteine residues with their characteristic spacing. Like the TGF- ⁇ 's, the inhibin ⁇ 's, and GDF-8, GDF-11 also contains two additional cysteine residues. In the case of TGF- ⁇ 2, these additional cysteine residues are known to form an intramolecular disulfide bond (Daopin, et al., Science, 257:369. 1992; Schlunegger and Grutter, Nature, 358:430. 1992). A.
  • FIGURE 3 tabulation of the amino acid sequence homologies between GDF-11 and the other TGF- ⁇ family members is shown in FIGURE 3. Numbers represent percent amino acid identities between each pair calculated from the first conserved cysteine to the C-terminus. Boxes represent homologies among highly-related members within particular subgroups. In this region, GDF-11 is most highly related to GDF-8 (92% sequence identity).
  • GDF-8 SEQ ID NO:5
  • GDF-11 SEQ ID NO:6 amino acid sequences
  • NIS N-linked glycosylation signals
  • RSRR putative proteolytic processing sites
  • GDF-11 protein In order to express GDF-11 protein, a hybrid gene was constructed in which the N- terminal region of GDF-11 was replaced by the analogous region of GDF-8. Such hybrid constructs have been used to produce biologically-active BMP-4 (Hammonds, et al., Mol. Endocrinol., 5:149, 1991) and Vg-1 (Thomsen and Melton, Cell, 74:433, 1993). In order to ensure that the GDF-11 protein produced from the hybrid construct would represent authentic GDF-11, the hybrid gene was constructed in such a manner that the fusion of the two gene fragments would occur precisely at the predicted cleavage sites. In particular, an AvaW restriction site is present in both sequences at the location corresponding to the predicted proteolytic cleavage site. The N-terminal pro-region of
  • GDF-8 up to this AvaW site was obtained by partial digestion of the clone with AvaW and fused to the C-terminal region of GDF-11 beginning at this AvaW site.
  • the resulting hybrid construct was then inserted into the pMSXND mammalian expression vector (Lee and Nathans, J. Biol. Chem., 263:3521) and transfected into Chinese hamster ovary cells.
  • FIGURE 5 Western analysis of conditioned medium from G418- resistant cells using antibodies raised against the C-terminal portion of GDF-8 showed that these cells secreted GDF-11 protein into the medium and that at least some of the hybrid protein was proteolytically processed. Furthermore, these studies demonstrate that the antibodies directed against the C-terminal portion of GDF-8 will also react with GDF-11 protein.
  • PCR products were electrophoresed on agarose gels, blotted, and probed with oligonucleotide #104, 5'- AAATATCCGCATACCCATTT-3', (SEQ ID NO:9) which corresponds to a sequence internal to the region flanked by primer #101 and #102.
  • Filters were hybridized in 6 X SSC, 1 X Denhardt's solution, 100 g/ml yeast transfer RNA, and 0.05% sodium pyrophosphate at 50 °C.
  • the human-specific probe detected a band of the predicted size (approximately 224 base pairs) in the positive control sample (total human genomic DNA) and in a single DNA sample from the human/rodent hybrid panel.
  • This positive signal corresponds to human chromosome 12.
  • the human chromosome contained in each of the hybrid cell lines is identified at the top of each of the first 24 lanes (1-22, X, and Y). In the lanes designated CHO, M, and H, the starting DNA template was total genomic DNA from hamster, mouse, and human sources, respectively. In the lane marked B1 , no template DNA was used. Numbers at left indicate the mobilities of DNA standards. These data show that the human GDF-11 gene is located on chromosome 12.
  • FISH florescence in situ hybridization
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • ACG AAA AGG TCC CGG CGG AAC CTA GGC CTG GAC TGC GAT GAA CAC TCG 278 Thr Lys Arg Ser Arg Arg Asn Leu Gly Leu Asp Cys Asp Glu His Ser 15 20 25
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)
  • MOLECULE TYPE DNA (genomic)

Abstract

Growth differentiation factor-11 (GDF-11) is disclosed along with its polynucleotide sequence and amino acid sequence. Also disclosed are diagnostic and therapeutic methods of using the GDF-11 polypeptide and polynucleotide sequences.

Description

GROWTH DIFFERENTIATION FACTOR-11
BACKGROUND OF THE INVENTION
1. Field of the Invention
The invention relates generally to growth factors and specifically to a new member of the transforming growth factor beta (TGF-β) superfamily, which is denoted, growth differentiation factor-11 (GDF-11).
2. Description of Related Art
The transforming growth factor β (TGF-β) superfamily encompasses a group of structurally-related proteins which affect a wide range of differentiation processes during embryonic development. The family includes, Mullerian inhibiting substance (MIS), which is required for normal male sex development (Behringer, et al., Nature, 345:167, 1990), Drosophila decapentaplegic (DPP) gene product, which is required for dorsal- ventral axis formation and morphogenesis of the imaginal disks (Padgett, er a/., Nature, 325:81-84, 1987), the Xenopus Vg-1 gene product, which localizes to the vegetal pole of eggs ((Weeks, et al., Cell, 51:861-867, 1987), the activins (Mason, et al., Biochem,
Biophys. Res. Commun., 135:957-964, 1986), which can induce the formation of mesoderm and anterior structures in Xenopus embryos (Thomsen, et al., Cell, 63:485, 1990), and the bone morphogenetic proteins (BMPs, osteogenin, OP-1) which can induce de novo cartilage and bone formation (Sampath, er al., J. Biol. Chem., 265:13198. 1990). The TGF-βs can influence a variety of differentiation processes, including adipogenesis, myogenesis, chondrogenesis, hematopoiesis, and epithelial cell differentiation (for review, see Massague, Cell 49:437. 1987).
The proteins of the TGF-β family are initially synthesized as a large precursor protein which subsequently undergoes proteolytic cleavage at a cluster of basic residues approximately 110-140 amino acids from the C-terminus. The C-terminal regions, or mature regions, of the proteins are all structurally related and the different family members can be classified into distinct subgroups based on the extent of their homology. Although the homologies within particular subgroups range from 70% to 90% amino acid sequence identity, the homologies between subgroups are significantly lower, generally ranging from only 20% to 50%. In each case, the active species appears to be a disulfide-linked dimer of C-terminal fragments. Studies have shown that when the pro- region of a member of the TGF-β family is coexpressed with a mature region of another member of the TGF-β family, intracellular dimerization and secretion of biologically active homodimers occur (Gray, A., and Maston, A., Science, 247:1328. 1990). Additional studies by Hammonds, etal., (Molec. Endocrin. 5:149, 1991) showed that the use of the BMP-2 pro-region combined with the BMP-4 mature region led to dramatically improved expression of mature BMP-4. For most of the family members that have been studied, the homodimeric species has been found to be biologically active, but for other family members, like the inhibins (Ling, et al., Nature, 321 :779. 1986) and the TGF-βs
(Cheifetz, et al., Cell, 48:409, 1987), heterodimers have also been detected, and these appear to have different biological properties than the respective homodimers.
Identification of new factors that are tissue-specific in their expression pattern will provide a greater understanding of that tissue's development and function.
SUMMARY OF THE INVENTION
The present invention provides a cell growth and differentiation factor, GDF-11, a polynucleotide sequence which encodes the factor, and antibodies which are bind to the factor. This factor appears to relate to various cell proliferative disorders, especially those involving muscle, neural, and uterine cells, as well as disorders related to the function of the immune system.
Thus, in one embodiment, the invention provides a method for detecting a cell proliferative disorder of muscle, neural, uterine, spleen, or thymus origin and which is associated with GDF-11. In another embodiment, the invention provides a method for treating a cell proliferative or immunologic disorder by suppressing or enhancing GDF-11 activity.
BRIEF DESCRIPTION OF THE DRAWINGS
FIGURE 1 shows the nucleotide and predicted amino acid sequences of murine (FIGURE 1a) and human (FIGURE 1b) GDF-11. The putative proteolytic processing sites are shown by the shaded boxes. In the human sequence, the potential N-linked glycosylation signal is shown by the open box, and the consensus polyadenylation signal is underlined; the poly A tail is not shown.
FIGURE 2 shows Northern blots of RNA prepared from adult (FIGURE 2a) or fetal and neonatal (FIGURE 2b) tissues probed with a murine GDF-11 probe.
FIGURE 3 shows amino acid homologies among different members of the TGF-β superfamily. Numbers represent percent amino acid identities between each pair calculated from the first conserved cysteine to the C-terminus. Boxes represent homologies among highly-related members within particular subgroups.
FIGURE 4 shows an alignment of the predicted amino acid sequences of human GDF-11 (top lines) with human GDF-8 (bottom lines). Vertical lines indicate identities. Dots represent gaps introduced in order to maximize the alignment. Numbers represent amino acid positions relative to the N-terminus. The putative proteolytic processing sites are shown by the open box. The conserved cysteine residues on the C-terminal region are shown by the shaded boxes.
FIGURE 5 shows the expression of GDF-11 in mammalian cells. Conditioned medium prepared from Chinese hamster ovary cells transfected with a hybrid GDF-8/GDF-11 gene (see text) cloned into the MSXND expression vector in either the antisense (lane 1) or sense (lane 2) orientation was dialyzed, lyophilized, and subjected to Western analysis using antibodies directed against the C-terminal portion of GDF-8 protein. Arrows at right indicate the putative unprocessed (pro-GDF-8/GDF-11) or processed GDF-11 proteins. Numbers at left indicate mobilities of molecular weight standards. FIGURE 6 shows the chromosomal mapping of human GDF-11. DNA samples prepared from human/rodent somatic cell lines were subjected to PCR, electrophoresed on agarose gels, blotted, and probed. The human chromosome contained in each of the hybrid cell lines is identified at the top of each of the first 24 lanes (1-22, X, and Y). In the lanes designated CHO, M, and H, the starting DNA template was total genomic DNA from hamster, mouse, and human sources, respectively. In the lane marked B1 , no template DNA was used. Numbers at left indicate the mobilities of DNA standards.
FIGURE 7 shows the FISH localization of GDF-11. Metaphase chromosomes derived from peripheral blood lymphocytes were hybridized with digoxigenin-labelled human GDF-11 probe (a) or a mixture of human GDF-11 genomic and chromosome 12-specific centromere probes (b) and analyzed as described in the text. A schematic showing the location of GDF-11 at position 12q13 is shown in panel (c).
FIGURE 8 shows the nucleotide and deduced amino acid sequence of murine GDF-8.
DETAILED DESCRIPTION OF THE INVENTION
The present invention provides a growth and differentiation factor, GDF-11, and a polynucleotide sequence encoding GDF-11. GDF-11 is expressed at highest levels in muscle, brain, uterus, spleen, and thymus and at lower levels in other tissues. In one embodiment, the invention provides a method for detection of a cell proliferative or immunologic disorder of muscle, neural, uterine, spleen, or thymus origin which is associated with GDF-11 expression or function. In another embodiment, the invention provides a method for treating a cell proliferative or immunologic disorder by using an agent which suppresses or enhances GDF-11 activity.
The TGF-β superfamily consists of multifunctional polypeptides that control proliferation, differentiation, and other functions in many cell types. Many of the peptides have regulatory, both positive and negative, effects on other peptide growth factors. The structural homology between the GDF-11 protein of this invention and the members of the TGF-β family, indicates that GDF-11 is a new member of the family of growth and differentiation factors. Based on the known activities of many of the other members, it can be expected that GDF-11 will also possess biological activities that will make it useful as a diagnostic and therapeutic reagent.
Certain members of this superfamily have expression patterns or possess activities that relate to the function of the nervous system. For example, one family member, namely GDNF, has been shown to be a potent neurotrophic factor that can promote the survival of dopaminergic neurons (Lin, er al., Science, 260:1130). Another family member, namely dorsalin-1 , is capable of promoting the differentiation of neural crest cells (Basler, etal., Cell, 73:687, 1993). The inhibins and activins have been shown to be expressed in the brain (Meunier, et al., Proc. Natl. Acad. Sci., USA, 85:247, 1988; Sawchenko, et al., Nature, 334:615, 1988), and activin has been shown to be capable of functioning as a nerve cell survival molecule (Schubert, et al., Nature, 344:868. 1990). Another family member, namely GDF-1 , is nervous system-specific in its expression pattern (Lee, Proc. Nat'l. Acad. Sci., USA, 88:4250, 1991), and certain other family members, such as Vgr-1 (Lyons, et al., Proc. Natl Acad. Sci, USA, 86:4554, 1989; Jones, et al., Development, 1U:581 , 1991), OP-1 (Ozkaynak, et al., J. Biol. Chem., 267:25220, 1992), and BMP-4 (Jones, et al., [Development, 1H:531 , 1991), are also known to be expressed in the nervous system. The expression of GDF-11 in brain and muscle suggests that GDF-11 may also possess activities that relate to the function of the nervous system. In particular, it is known, for example, that skeletal muscle produces a factor or factors that promote the survival of motor neurons (Brown, Trends Neurosci., 7:10, 1984). The known neurotrophic activities of other members of this family and the expression of GDF- 11 in muscle suggest that one activity of GDF-11 may be as a trophic factor for motor neurons; indeed, GDF-11 is highly related to GDF-8, which is virtually muscle-specific in its expression pattern. Altematively, GDF-11 may have neurotrophic activities for other neuronal populations. Hence, GDF-11 may have in vitro and in vivo applications in the treatment of neurodegenerative diseases, such as amyotrophic lateral sclerosis, or in maintaining cells or tissues in culture prior to transplantation.
GDF-11 may also have applications in treating disease processes involving muscle, such as in musculodegenerative diseases or in tissue repair due to trauma. In this regard, many other members of the TGF-β family are also important mediators of tissue repair. TGF-β has been shown to have marked effects on the formation of collagen and to cause a striking angiogenic response in the newborn mouse (Roberts, et al., Proc. Natl.
Acad. Sci., USA 83:4167, 1986). TGF-β has also been shown to inhibit the differentiation of myoblasts in culture (Massague, et al., Proc. Natl. Acad. Sci., USA 83.: 8206, 1986). Moreover, because myoblast cells may be used as a vehicle for delivering genes to muscle for gene therapy, the properties of GDF-11 could be exploited for maintaining cells prior to transplantation or for enhancing the efficiency of the fusion process.
GDF-11 may also have applications in the treatment of immunologic disorders. In particular, TGF-β has been shown to have a wide range of immunoregulatory activities, including potent suppressive effects on B and T cell proliferation and function (for review, see Palladino, et al., Ann. N. Y. Acad. Sci., 593:181 , 1990). The expression of GDF-11 in spleen and thymus suggests that GDF-11 may possess similar activities and therefore, may be used as an anti-inflammatory agent or as a treatment for disorders related to abnormal proliferation or function of lymphocytes.
The temn "substantially pure" as used herein refers to GDF-11 which is substantially free of other proteins, lipids, carbohydrates or other materials with which it is naturally associated. One skilled in the art can purify GDF-11 using standard techniques for protein purification. The substantially pure polypeptide will yield a single major band on a non-reducing polyacrylamide gel. The purity of the GDF-11 polypeptide can also be determined by amino-terminal amino acid sequence analysis. GDF-11 polypeptide includes functional fragments of the polypeptide, as long as the activity of GDF-11 remains. Smaller peptides containing the biological activity of GDF-11 are included in the invention.
The invention provides polynucleotides encoding the GDF-11 protein. These polynucleotides include DNA, cDNA and RNA sequences which encode GDF-11. It is understood that all polynucleotides encoding all or a portion of GDF-11 are also included herein, as long as they encode a polypeptide with GDF-11 activity. Such polynucleotides include naturally occurring, synthetic, and intentionally manipulated polynucleotides. For example, GDF-11 polynucleotide may be subjected to site-directed mutagenesis. The polynucleotide sequence for GDF-11 also includes antisense sequences. The polynucleotides of the invention include sequences that are degenerate as a result of the genetic code. There are 20 natural amino acids, most of which are specified by more than one codon. Therefore, all degenerate nucleotide sequences are included in the invention as long as the amino acid sequence of GDF-11 polypeptide encoded by the nucleotide sequence is functionally unchanged. Specifically disclosed herein is a DNA sequence containing the human GDF-11 gene. The sequence contains an open reading frame encoding a polypeptide 407 amino acids in length. The sequence contains a putative RXXR proteolytic cleavage site at amino acids 295-298. Cleavage of the precursor at this site would generate an active C- terminal fragment 109 amino acids in length with a predicted molecular weight of approximately 12,500 kD. Also disclosed herein is a partial murine genomic sequence. Preferably, the human GDF-11 nucleotide sequence is SEQ ID NO:1 and the mouse nucleotide sequence is SEQ ID NO:3.
The polynucleotide encoding GDF-11 includes SEQ ID NO:1 and 3, as well as nucleic acid sequences complementary to SEQ ID NO's:1 and 3. A complementary sequence may include an antisense nucleotide. When the sequence is RNA, the deoxynucieotides A, G, C, and T of SEQ ID NO:1 and 3 are replaced by ribonucleotides A, G, C, and U, respectively. Also included in the invention are fragments of the above-described nucleic acid sequences that are at least 15 bases in length, which is sufficient to permit the fragment to selectively hybridize to DNA that encodes the protein of SEQ ID NO: 2 or 4 under physiological conditions.
The C-terminal region of GDF-11 following the putative proteolytic processing site shows significant homology to the known members of the TGF-β superfamily. The GDF-11 sequence contains most of the residues that are highly conserved in other family members (see FIGURE 1). Like the TGF-βs and inhibin βs, GDF-11 contains an extra pair of cysteine residues in addition to the 7 cysteines found in virtually all other family members. Among the known family members, GDF-11 is most homologous to GDF-8 (92% sequence identity) (see FIGURE 3).
Minor modifications of the recombinant GDF-11 primary amino acid sequence may result in proteins which have substantially equivalent activity as compared to the GDF-11 polypeptide described herein. Such modifications may be deliberate, as by site-directed mutagenesis, or may be spontaneous. All of the polypeptides produced by these modifications are included herein as long as the biological activity of GDF-11 still exists. Further, deletion of one or more amino acids can also result in a modification of the structure of the resultant molecule without significantly altering its biological, activity. This can lead to the development of a smaller active molecule which would have broader utility. For example, one can remove amino or carboxy terminal amino acids which are not required for GDF-11 biological activity.
The nucleotide sequence encoding the GDF-11 polypeptide of the invention includes the disclosed sequence and conservative variations thereof. The term "conservative variation" as used herein denotes the replacement of an amino acid residue by another, biologically similar residue. Examples of conservative variations include the substitution of one hydrophobic residue such as isoleucine, valine, leucine or methionine for another, or the substitution of one polar residue for another, such as the substitution of arginine for lysine, glutamicfor aspartic acid, or glutamine for asparagine, and the like. The term "conservative variation" also includes the use of a substituted amino acid in place of an unsubstituted parent amino acid provided that antibodies raised to the substituted polypeptide also immunoreact with the unsubstituted polypeptide.
DNA sequences of the invention can be obtained by several methods. For example, the DNA can be isolated using hybridization techniques which are well known in the art. These include, but are not limited to: 1) hybridization of genomic or cDNA libraries with probes to detect homologous nucleotide sequences, 2) polymerase chain reaction (PCR) on genomic DNA or cDNA using primers capable of annealing to the DNA sequence of interest, and 3) antibody screening of expression libraries to detect cloned DNA fragments with shared structural features.
Preferably the GDF-11 polynucleotide of the invention is derived from a mammalian organism, and most preferably from a mouse, rat, or human. Screening procedures which rely on nucleic acid hybridization make it possible to isolate any gene sequence from any organism, provided the appropriate probe is available. Oligonucleotide probes, which correspond to a part of the sequence encoding the protein in question, can be synthesized chemically. This requires that short, oligopeptide stretches of amino acid sequence must be known. The DNA sequence encoding the protein can be deduced from the genetic code, however, the degeneracy of the code must be taken into account. It is possible to perform a mixed addition reaction when the sequence is degenerate.
This includes a heterogeneous mixture of denatured double-stranded DNA. For such screening, hybridization is preferably performed on either single-stranded DNA or denatured double-stranded DNA. Hybridization is particularly useful in the detection of cDNA clones derived from sources where an extremely low amount of mRNA sequences relating to the polypeptide of interest are present. In other words, by using stringent hybridization conditions directed to avoid non-specific binding, it is possible, for example, to allow the autoradiographic visualization of a specific cDNA clone by the hybridization of the target DNA to that single probe in the mixture which is its complete complement (Wallace, et al., Nucl. Acid Res., 9:879, 1981 ; Maniatis, et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor, N.Y. 1989).
The development of specific DNA sequences encoding GDF-11 can also be obtained by: 1) isolation of double-stranded DNA sequences from the genomic DNA; 2) chemical manufacture of a DNA sequence to provide the necessary codons for the polypeptide of interest; and 3) in vitro synthesis of a double-stranded DNA sequence by reverse transcription of mRNA isolated from a eukaryotic donor cell. In the latter case, a double- stranded DNA complement of mRNA is eventually formed which is generally referred to as cDNA.
Of the three above-noted methods for developing specific DNA sequences for use in recombinant procedures, the isolation of genomic DNA isolates is the least common. This is especially true when it is desirable to obtain the microbial expression of mammalian polypeptides due to the presence of introns. The synthesis of DNA sequences is frequently the method of choice when the entire sequence of amino acid residues of the desired polypeptide product is known. When the entire sequence of amino acid residues of the desired polypeptide is not known, the direct synthesis of DNA sequences is not possible and the method of choice is the synthesis of cDNA sequences. Among the standard procedures for isolating cDNA sequences of interest is the formation of plasmid- or phage-carrying cDNA libraries which are derived from reverse transcription of mRNA which is abundant in donor cells that have a high level of genetic expression. When used in combination with polymerase chain reaction technology, even rare expression products can be cloned. In those cases where significant portions of the amino acid sequence of the polypeptide are known, the production of labeled single or double-stranded DNA or RNA probe sequences duplicating a sequence putatively present in the target cDNA may be employed in DNA/DNA hybridization procedures which are carried out on cloned copies of the cDNA which have been denatured into a single-stranded form (Jay, et al., Nucl. Acid Res., 11:2325, 1983).
A cDNA expression library, such as lambda gt11 , can be screened indirectly for GDF-11 peptides having at least one epitope, using antibodies specific for GDF-11. Such antibodies can be either polyclonally or monoclonally derived and used to detect expression product indicative of the presence of GDF-11 cDNA.
DNA sequences encoding GDF-11 can be expressed in vitro by DNA transfer into a suitable host cell. "Host cells" are cells in which a vector can be propagated and its DNA expressed. The term also includes any progeny of the subject host cell. It is understood that all progeny may not be identical to the parental cell since there may be mutations that occur during replication. However, such progeny are included when the term "host cell" is used. Methods of stable transfer, meaning that the foreign DNA is continuously maintained in the host, are known in the art. In the present invention, the GDF-11 polynucleotide sequences may be inserted into a recombinant expression vector. The term "recombinant expression vector" refers to a plasmid, virus or other vehicle known in the art that has been manipulated by insertion or incorporation of the GDF-11 genetic sequences. Such expression vectors contain a promoter sequence which facilitates the efficient transcription of the inserted genetic sequence of the host. The expression vector typically contains an origin of replication, a promoter, as well as specific genes which allow phenotypic selection of the transformed cells. Vectors suitable for use in the present invention include, but are not limited to the T7-based expression vector for expression in bacteria (Rosenberg, et al., Gene, 56:125, 1987), the pMSXND expression vector for expression in mammalian cells
(Lee and Nathans, J. Biol. Chem., 263:3521, 1988) and baculovirus-derived vectors for expression in insect cells. The DNA segment can be present in the vector operably linked to regulatory elements, for example, a promoter (e.g., T7, metallothionein I, or polyhedrin promoters).
Polynucleotide sequences encoding GDF-11 can be expressed in either prokaryotes or eukaryotes. Hosts can include microbial, yeast, insect and mammalian organisms. Methods of expressing DNA sequences having eukaryotic or viral sequences in prokaryotes are well known in the art. Biologically functional viral and plasmid DNA vectors capable of expression and replication in a host are known in the art. Such vectors are used to incorporate DNA sequences of the invention. Preferably, the mature
C-terminal region of GDF-11 is expressed from a DNA clone containing the entire coding sequence of GDF-11. Alternatively, the C-terminal portion of GDF-11 can be expressed as a fusion protein with the pro- region of another member of the TGF-β family or co¬ expressed with another pro- region (see for example, Hammonds, et al., Molec. Endocrin. 5:149, 1991; Gray, A., and Mason, A., Science, 247:1328, 1990).
Transformation of a host cell with recombinant DNA may be carried out by conventional techniques as are well known to those skilled in the art. Where the host is prokaryotic, such as £ coli, competent cells which are capable of DNA uptake can be prepared from cells harvested after exponential growth phase and subsequently treated by the CaCI2 method using procedures well known in the art. Altematively, MgCI2 or RbCI can be used. Transformation can also be performed after forming a protoplast of the host cell if desired.
When the host is a eukaryote, such methods of transfection of DNA as calcium phosphate co-precipitates, conventional mechanical procedures such as microinjection, electroporation, insertion of a plasmid encased in liposomes, or virus vectors may be used. Eukaryotic cells can also be cotransformed with DNA sequences encoding the GDF-11 of the invention, and a second foreign DNA molecule encoding a selectable phenotype, such as the herpes simplex thymidine kinase gene. Another method is to use a eukaryotic viral vector, such as simian virus 40 (SV40) or bovine papilloma virus, to transiently infect or transform eukaryotic cells and express the protein, (see for example, Eukaryotic Viral Vectors, Cold Spring Harbor Laboratory, Gluzman ed., 1982).
Isolation and purification of microbial expressed polypeptide, or fragments thereof, provided by the invention, may be carried out by conventional means including preparative chromatography and immunological separations involving monoclonal or polyclonal antibodies.
The GDF-11 polypeptides of the invention can also be used to produce antibodies which are immunoreactive or bind to epitopes of the GDF-11 polypeptides. Antibody which consists essentially of pooled monoclonal antibodies with different epitopic specificities, as well as distinct monoclonal antibody preparations are provided. Monoclonal antibodies are made from antigen containing fragments of the protein by methods well known in the art (Kohler, et al., Nature, 256:495, 1975; Current Protocols in Molecular Biology, Ausubel, et al., ed., 1989).
The term "antibody" as used in this invention includes intact molecules as well as fragments thereof, such as Fab, F(ab')2, and Fv which are capable of binding the epitopic determinant. These antibody fragments retain some ability to selectively bind with its antigen or receptor and are defined as follows:
(1) Fab, the fragment which contains a monovalent antigen-binding fragment of an antibody molecule can be produced by digestion of whole antibody with the enzyme papain to yield an intact light chain and a portion of one heavy chain;
(2) Fab1, the fragment of an antibody molecule can be obtained by treating whole antibody with pepsin, followed by reduction, to yield an intact light chain and a portion of the heavy chain; two Fab' fragments are obtained per antibody molecule;
(3) (Fab')2, the fragment of the antibody that can be obtained by treating whole antibody with the enzyme pepsin without subsequent reduction; F(ab')2 is a dimer of two Fab' fragments held together by two disulfide bonds;
(4) Fv, defined as a genetically engineered fragment containing the variable region of the light chain and the variable region of the heavy chain expressed as two chains; and
(5) Single chain antibody ("SCA"), defined as a genetically engineered molecule containing the variable region of the light chain, the variable region of the heavy chain, linked by a suitable polypeptide linker as a genetically fused single chain molecule.
Methods of making these fragments are known in the art. (See for example, Hariow and
Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, New York (1988), incorporated herein by reference). As used in this invention, the term "epitope" means any antigenic determinant on an antigen to which the paratope of an antibody binds. Epitopic determinants usually consist of chemically active surface groupings of molecules such as amino acids or sugar side chains and usually have specific three dimensional structural characteristics, as well as specific charge characteristics.
Antibodies which bind to the GDF-11 polypeptide of the invention can be prepared using an intact polypeptide or fragments containing small peptides of interest as the immunizing antigen. The polypeptide or a peptide used to immunize an animal can be derived from translated cDNA or chemical synthesis which can be conjugated to a carrier protein, if desired. Such commonly used carriers which are chemically coupled to the peptide include keyhole limpet hemocyanin (KLH), thyroglobulin, bovine serum albumin (BSA), and tetanus toxoid. The coupled peptide is then used to immunize the animal (e.g., a mouse, a rat, or a rabbit).
If desired, polyclonal or monoclonal antibodies can be further purified, for example, by binding to and elution from a matrix to which the polypeptide or a peptide to which the antibodies were raised is bound. Those of skill in the art will know of various techniques common in the immunology arts for purification and/or concentration of polyclonal antibodies, as well as monoclonal antibodies (See for example, Coligan, et al., Unit 9, Current Protocols in Immunology, Wiley Interscience, 1991 , incorporated by reference).
It is also possible to use the anti-idiotype technology to produce monoclonal antibodies which mimic an epitope. For example, an anti-idiotypic monoclonal antibody made to a first monoclonal antibody will have a binding domain in the hypervariable region which is the "image" of the epitope bound by the first monoclonal antibody.
The term "cell-proliferative disorder" denotes malignant as well as non-malignant cell populations which often appear to differ from the surrounding tissue both morphologically and genotypically. Malignant cells (i.e. cancer) develop as a result of a multistep process. The GDF-11 polynucleotide that is an antisense molecule is useful in treating malignancies of the various organ systems, particularly, for example, cells in muscle, uterus, spleen, thymus, or neural tissue. Essentially, any disorder which is etiologically linked to altered expression of GDF-11 could be considered susceptible to treatment with a GDF-11 suppressing reagent. One such disorder is a malignant cell proliferative disorder, for example.
The invention provides a method for detecting a cell proliferative disorder of muscle, uterine or neural tissue, for example, which comprises contacting an anti-GDF-11 antibody with a cell suspected of having a GDF-11 associated disorder and detecting binding to the antibody. The antibody reactive with GDF-11 is labeled with a compound which allows detection of binding to GDF-11. For purposes of the invention, an antibody specific for GDF-11 polypeptide may be used to detect the level of GDF-11 in biological fluids and tissues. Any specimen containing a detectable amount of antigen can be used. A preferred sample of this invention is muscle, uterus, spleen, thymus, or neural tissue. The level of GDF-11 in the suspect cell can be compared with the level in a normal cell to determine whether the subject has a GDF-11 -associated cell proliferative disorder. Preferably the subject is human.
The antibodies of the invention can be used in any subject in which it is desirable to administer in vitro or in vivo immunodiagnosis or immunotherapy. The antibodies of the invention are suited for use, for example, in immunoassays in which they can be utilized in liquid phase or bound to a solid phase carrier. In addition, the antibodies in these immunoassays can be detectably labeled in various ways. Examples of types of immunoassays which can utilize antibodies of the invention are competitive and non- competitive immunoassays in either a direct or indirect format. Examples of such immunoassays are the radioimmunoassay (RIA) and the sandwich (immunometric) assay. Detection of the antigens using the antibodies of the invention can be done utilizing immunoassays which are run in either the forward, reverse, or simultaneous modes, including immunohistochemical assays on physiological samples. Those of skill in the art will know, or can readily discern, other immunoassay formats without undue experimentation.
The antibodies of the invention can be bound to many different carriers and used to detect the presence of an antigen comprising the polypeptide of the invention. Examples of well-known carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, agaroses and magnetite. The nature of the carrier can be either soluble or insoluble for purposes of the invention. Those skilled in the art will know of other suitable carriers for binding antibodies, or will be able to ascertain such, using routine experimentation.
There are many different labels and methods of labeling known to those of ordinary skill in the art. Examples of the types of labels which can be used in the present invention include enzymes, radioisotopes, fluorescent compounds, colloidal metals, chemiluminescent compounds, phosphorescent compounds, and bioluminescent compounds. Those of ordinary skill in the art will know of other suitable labels for binding to the antibody, or will be able to ascertain such, using routine experimentation.
Another technique which may also result in greater sensitivity consists of coupling the antibodies to low molecular weight haptens. These haptens can then be specifically detected by means of a second reaction. For example, it is common to use such haptens as biotin, which reacts with avidin, or dinitrophenyl, puridoxal, and fluorescein, which can react with specific antihapten antibodies.
In using the monoclonal antibodies of the invention for the in vivo detection of antigen, the detectably labeled antibody is given a dose which is diagnostically effective. The term "diagnostically effective" means that the amount of detectably labeled monoclonal antibody is administered in sufficient quantity to enable detection of the site having the antigen comprising a polypeptide of the invention for which the monoclonal antibodies are specific. The concentration of detectably labeled monoclonal antibody which is administered should be sufficient such that the binding to those cells having the polypeptide is detectable compared to the background. Further, it is desirable that the detectably labeled monoclonal antibody be rapidly cleared from the circulatory system in order to give the best target-to-background signal ratio.
As a rule, the dosage of detectably labeled monoclonal antibody for in vivo diagnosis will vary depending on such factors as age, sex, and extent of disease of the individual. Such dosages may vary, for example, depending on whether multiple injections are given, antigenic burden, and other factors known to those of skill in the art.
For in vivo diagnostic imaging, the type of detection instrument available is a major factor in selecting a given radioisotope. The radioisotope chosen must have a type of decay which is detectable for a given type of instrument. Still another important factor in selecting a radioisotope for in vivo diagnosis is that deleterious radiation with respect to the host is minimized. Ideally, a radioisotope used for in vivo imaging will lack a particle emission, but produce a large number of photons in the 140-250 keV range, which may readily be detected by conventional gamma cameras.
For in vivo diagnosis radioisotopes may be bound to immunoglobulin either directly or indirectly by using an intermediate functional group. Intermediate functional groups which often are used to bind radioisotopes which exist as metallic ions to immunoglobulins are the bifunctional chelating agents such as diethylenetriaminepentacetic acid (DTPA) and ethylenediaminetetraacetic acid (EDTA) and similar molecules. Typical examples of metallic ions which can be bound to the monoclonal antibodies of the invention are 11 ln, 97Ru, 67Ga, 68Ga, 72As, 89Zr, and 201TI.
The monoclonal antibodies of the invention can also be labeled with a paramagnetic isotope for purposes of in vivo diagnosis, as in magnetic resonance imaging (MRI) or electron spin resonance (ESR). In general, any conventional method for visualizing diagnostic imaging can be utilized. Usually gamma and positron emitting radioisotopes are used for camera imaging and paramagnetic isotopes for MRI. Elements which are particularly useful in such techniques include 157Gd, 5 Mn, 162Dy, 52Cr, and ^Fe.
The monoclonal antibodies of the invention can be used in vitro and in vivo to monitor the course of amelioration of a GDF-11 -associated disease in a subject. Thus, for example, by measuring the increase or decrease in the number of cells expressing antigen comprising a polypeptide of the invention or changes in the concentration of such antigen present in various body fluids, it would be possible to determine whether a particular therapeutic regimen aimed at ameliorating the GDF-11 -associated disease is effective. The term "ameliorate" denotes a lessening of the detrimental effect of the
GDF-11 -associated disease in the subject receiving therapy.
The present invention identifies a nucleotide sequence that can be expressed in an altered manner as compared to expression in a normal cell, therefore it is possible to design appropriate therapeutic or diagnostic techniques directed to this sequence. Thus, where a cell-proliferative disorder is associated with the expression of GDF-11 , nucleic acid sequences that interfere with GDF-11 expression at the translational level can be used. This approach utilizes, for example, antisense nucleic acid and ribozymes to block translation of a specific GDF-11 mRNA, either by masking that mRNA with an antisense nucleic acid or by cleaving it with a ribozyme. Such disorders include neurodegenerative diseases, for example.
Antisense nucleic acids are DNA or RNA molecules that are complementary to at least a portion of a specific mRNA molecule (Weintraub, Scientific American, 262:40, 1990). In the cell, the antisense nucleic acids hybridize to the corresponding mRNA, forming a double-stranded molecule. The antisense nucleic acids interfere with the translation of the mRNA, since the cell will not translate a mRNA that is double-stranded. Antisense oligomers of about 15 nucleotides are preferred, since they are easily synthesized and are less likely to cause problems than larger molecules when introduced into the target GDF-11 -producing cell. The use of antisense methods to inhibit the in vitro translation of genes is well known in the art (Marcus-Sakura, Anal.Biochem., 172:289, 1988).
Ribozymes are RNA molecules possessing the ability to specifically cleave other single- stranded RNA in a manner analogous to DNA restriction endonucleases. Through the modification of nucleotide sequences which encode these RNAs, it is possible to engineer molecules that recognize specific nucleotide sequences in an RNA molecule and cleave it (Cech, J.Amer.Med. Assn., 260:3030. 1988). A major advantage of this approach is that, because they are sequence-specific, only mRNAs with particular sequences are inactivated.
There are two basic types of ribozymes namely, tetrahymena-type (Hasselhoff, Nature,
334:585. 1988) and "hammerhead"-type. Tetrahymena-type ribozymes recognize sequences which are four bases in length, while "hammerhead"-type ribozymes recognize base sequences 11-18 bases in length. The longer the recognition sequence, the greater the likelihood that the sequence will occur exclusively in the target mRNA species. Consequently, hammerhead-type ribozymes are preferable to tetrahymena- type ribozymes for inactivating a specific mRNA species and 18-based recognition sequences are preferable to shorter recognition sequences.
The present invention also provides gene therapy for the treatment of cell proliferative or immunologic disorders which are mediated by GDF-11 protein. Such therapy would achieve its therapeutic effect by introduction of the GDF-11 antisense polynucleotide into cells having the proliferative disorder. Delivery of antisense GDF-11 polynucleotide can be achieved using a recombinant expression vector such as a chimeric virus or a colloidal dispersion system. Especially preferred for therapeutic delivery of antisense sequences is the use of targeted liposomes.
Various viral vectors which can be utilized for gene therapy as taught herein include adenovirus, herpes virus, vaccinia, or, preferably, an RNA virus such as a retrovirus. Preferably, the retroviral vector is a derivative of a murine or avian retrovirus. Examples of retroviral vectors in which a single foreign gene can be inserted include, but are not limited to: Moloney murine leukemia virus (MoMuLV), Harvey murine sarcoma virus (HaMuSV), murine mammary tumor virus (MuMTV), and Rous Sarcoma Virus (RSV). A number of additional retroviral vectors can incorporate multiple genes. All of these vectors can transfer or incorporate a gene for a selectable marker so that transduced cells can be identified and generated. By inserting a GDF-11 sequence of interest into the viral vector, along with another gene which encodes the ligand for a receptor on a specific target cell, for example, the vector is now target specific. Retroviral vectors can be made target specific by attaching, for example, a sugar, a glycolipid, or a protein.
Preferred targeting is accomplished by using an antibody to target the retroviral vector. Those of skill in the art will know of, or can readily ascertain without undue experimenta¬ tion, specific polynucleotide sequences which can be inserted into the retroviral genome or attached to a viral envelope to allow target specific delivery of the retroviral vector containing the GDF-11 antisense polynucleotide.
Since recombinant retroviruses are defective, they require assistance in order to produce infectious vector particles. This assistance can be provided, for example, by using helper cell lines that contain plasmids encoding all of the structural genes of the retrovirus under the control of regulatory sequences within the LTR. These plasmids are missing a nucleotide sequence which enables the packaging mechanism to recognize an RNA transcript for encapsidation. Helper cell lines which have deletions of the packaging signal include, but are not limited to Ψ2, PA317 and PA12, for example. These cell lines produce empty virions, since no genome is packaged. If a retroviral vector is introduced into such cells in which the packaging signal is intact, but the structural genes are replaced by other genes of interest, the vector can be packaged and vector virion produced.
Altematively, NIH 3T3 or other tissue culture cells can be directly transfected with plasmids encoding the retroviral structural genes gag, pol and env, by conventional calcium phosphate transfection. These cells are then transfected with the vector plasmid containing the genes of interest. The resulting cells release the retroviral vector into the culture medium.
Another targeted delivery system for GDF-11 antisense polynucleotides is a colloidal dispersion system. Colloidal dispersion systems include macromolecule complexes, nanocapsules, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. The preferred colloidal system of this invention is a iiposome. Liposomes are artificial membrane vesicles which are useful as delivery vehicles in vitro and in vivo. It has been shown that large unilamellar vesicles ( UV), which range in size from 0.2-4.0 m can encapsulate a substantial percentage of an aqueous buffer containing large macromolecules. RNA, DNA and intact virions can be encapsulated within the aqueous interior and be delivered to cells in a biologically active form (Fraley, et al., Trends Biochem. Sci., 6:77, 1981). In addition to mammalian cells, liposomes have been used for delivery of polynucleotides in plant, yeast and bacterial cells. In order for a Iiposome to be an efficient gene transfer vehicle, the following characteristics should be present: (1) encapsulation of the genes of interest at high efficiency while not compromising their biological activity; (2) preferential and substantial binding to a target cell in comparison to non-target cells; (3) delivery of the aqueous contents of the vesicle to the target cell cytoplasm at high efficiency; and (4) accurate and effective expression of genetic information (Mannino, et al., Biotechniques,
6:682, 1988).
The composition of the Iiposome is usually a combination of phospholipids, particularly high-phase-transition-temperature phospholipids, usually in combination with steroids, especially cholesterol. Other phospholipids or other lipids may also be used. The physical characteristics of liposomes depend on pH, ionic strength, and the presence of divalent cations. Examples of lipids useful in Iiposome production include phosphatidyl compounds, such as phosphatidylglycerol, phosphatidylcholine, phosphatidylserine, phosphatidyletha- nolamine, sphingolipids, cerebrosides, and gangliosides. Particularly useful are diacylphosphatidylglycerols, where the lipid moiety contains from 14-18 carbon atoms, particulariy from 16-18 carbon atoms, and is saturated. Illustrative phospholipids include egg phosphatidylcholine, dipalmitoylphosphatidylcholine and distearoylphos- phatidylcholine.
The targeting of liposomes can be classified based on anatomical and mechanistic factors. Anatomical classification is based on the level of selectivity, for example, organ- specific, cell-specific, and organelle-specific. Mechanistic targeting can be distinguished based upon whether it is passive or active. Passive targeting utilizes the natural tendency of liposomes to distribute to cells of the reticulo-endothelial system (RES) in organs which contain sinusoidal capillaries. Active targeting, on the other hand, involves alteration of the Iiposome by coupling the Iiposome to a specific ligand such as a monoclonal antibody, sugar, glycolipid, or protein, or by changing the composition or size of the Iiposome in order to achieve targeting to organs and cell types other than the naturally occurring sites of localization.
The surface of the targeted delivery system may be modified in a variety of ways. In the case of a liposomal targeted delivery system, lipid groups can be incorporated into the lipid bilayer of the Iiposome in order to maintain the targeting ligand in stable association with the liposomal bilayer. Various linking groups can be used for joining the lipid chains to the targeting ligand.
Due to the expression of GDF-11 in muscle, spleen, uterus, thymus, and neural tissue, there are a variety of applications using the polypeptide, polynucleotide, and antibodies of the invention, related to these tissues. Such applications include treatment of cell proliferative and immunologic disorders involving these and other tissues. In addition, GDF-11 may be useful in various gene therapy procedures. The following examples are intended to illustrate but not limit the invention. While they are typical of those that might be used, other procedures known to those skilled in the art may alternatively be used.
EXAMPLE 1 IDENTIFICATION AND ISOLATION
OF A NOVEL TGF-β FAMILY MEMBER
To identify novel members of the TGF-β superfamily, a murine genomic library was screened at reduced stringency using a murine GDF-8 probe (FIGURE 8; nucleotides 865-1234) spanning the region encoding the C-terminal portion of the GDF-8 precursor protein. Hybridization was carried out as described (Lee, Mol. Endocrinol., 4:1034, 1990) at 65°C, and the final wash was carried out at the same temperature in a buffer containing 0.5 M NaCI. Among the hybridizing phage was one that could be distinguished from GDF-8-containing phage on the basis of its reduced hybridization intensity to the GDF-8 probe. Partial nucleotide sequence analysis of the genomic insert present in this weakly hybridizing phage showed that this clone contained a sequence highly related to but distinct from murine GDF-8.
A partial nucleotide sequence of the genomic insert present in this phage is shown in FIGURE 1a. The sequence contained an open reading frame extending from nucleotides 198 to 575 that showed significant homology to the known members of the TGF-β superfamily (see below). Preceding this sequence was a 3' splice consensus sequence at precisely the same position as in the GDF-8 gene. This new TGF-β family member was given the designation GDF-11 (growth/differentiation factor-11). EXAMPLE 2 EXPRESSION OF GDF-11
To determine the expression pattem of GDF-11, RNA samples prepared from a variety of tissues were screened by Northern analysis. RNA isolation and Northern analysis were carried out as described previously (Lee, Mol. Endocrinol., 4:1034, 1990) except that the hybridization was carried out in 5X SSPE, 10% dextran sulfate, 50% formamide, 1 % SDS, 200 g/ml salmon DNA, and 0.1% each of bovine serum albumin, ficoll, and polyvinylpyrrolidone. Five micrograms of twice poly A-selected RNA prepared from each tissue (except for 2 day neonatal brain, for which only 3.3 μg RNA were used) were electrophoresed on formaldehyde gels, blotted, and probed with GDF-11. As shown in
FIGURE 2, the GDF-11 probe detected two RNA species, approximately 4.2 and 3.2 kb in length, in adult thymus, brain, spleen, uterus, and muscle as well as in whole embryos isolated at day 12.5 or 18.5 and in brain samples taken at various stages of development. On longer exposures of these blots, lower levels of GDF-11 RNA could also be detected in a number of other tissues.
EXAMPLE 3 ISOLATION OF cDNA CLONES ENCODING GDF-11
In order to isolate cDNA clones encoding GDF-11 , a cDNA library was prepared in the lambda ZAP II vector (Stratagene) using RNA prepared from human adult spleen. From 5 μg of twice poly A-selected RNA prepared from human spleen, a cDNA library consisting of 21 million recombinant phage was constructed according to the instructions provided by Stratagene. The library was screened without amplification. Library screening and characterization of cDNA inserts were carried out as described previously (Lee, Mol. Endocrinol., 4:1034, 1990). From this library, 23 hybridizing phage were obtained. The entire nucleotide sequence of the clone extending furthest toward the 5' end of the gene was determined. The 1258 base pair sequence contained a single long open reading frame beginning from the 5' end of the clone and extending to a TAA stop codon. Because the open reading frame and the homology with GDF-8 (see below) extended to the very 5' end of the clone, it seemed likely that this clone was missing the coding sequence corresponding to the N-terminal portion of the GDF-11 precursor protein. In order to obtain the remaining portion of the GDF-11 sequence, several genomic clones were isolated by screening a human genomic library with the human GDF-11 cDNA probe. Partial sequence analysis of one of these genomic clones showed that this clone contained the GDF-11 gene. From this clone, the remaining GDF-11 coding sequence was obtained. FIGURE 1b shows the predicted sequence of GDF-11 assembled from the genomic and cDNA sequences. Nucleotides 136 to 1393 represent the extent of the sequence obtained from a cDNA clone. Nucleotides 1 to 135 were obtained from a genomic clone. The sequence has been arbitrarily numbered beginning with a Sac II site present in the genomic clone, but the location of the mRNA start site is not known. The sequence contains a putative initiating methionine at nucleotide 54. Whether the sequence upstream of this methionine codon is all present in the mRNA is not known. Beginning with this methionine codon, the open reading frame extends for 407 amino acids. The sequence contains one potential N-linked glycosylation site at asparagine 94. The sequence contains a predicted RXXR proteolytic cleavage site at amino acids 295 to 298, and cleavage of the precursor at this site would generate an active C-terminal fragment 109 amino acids in length with a predicted molecular weight of approximately 12,500 kD. In this region, the predicted murine and human GDF-11 amino acid sequences are 100% identical. The high degree of sequence conservation across species suggests that GDF-11 plays an important role in vivo.
The C-terminal region following the predicted cleavage site contains all the hallmarks present in other TGF-β family members. GDF-11 contains most of the residues that are highly conserved in other family members, including the seven cysteine residues with their characteristic spacing. Like the TGF-β's, the inhibin β's, and GDF-8, GDF-11 also contains two additional cysteine residues. In the case of TGF-β2, these additional cysteine residues are known to form an intramolecular disulfide bond (Daopin, et al., Science, 257:369. 1992; Schlunegger and Grutter, Nature, 358:430. 1992). A. tabulation of the amino acid sequence homologies between GDF-11 and the other TGF-β family members is shown in FIGURE 3. Numbers represent percent amino acid identities between each pair calculated from the first conserved cysteine to the C-terminus. Boxes represent homologies among highly-related members within particular subgroups. In this region, GDF-11 is most highly related to GDF-8 (92% sequence identity).
An alignment of GDF-8 (SEQ ID NO:5) and GDF-11 (SEQ ID NO:6) amino acid sequences is shown in FIGURE 4. The two sequences contain potential N-linked glycosylation signals (NIS) and putative proteolytic processing sites (RSRR) at analogous positions. The two sequences are related not only in the C-terminal region following the putative cleavage site (90% amino acid sequence identity), but also in the pro-region of the molecules (45% a ino acid sequence identity).
EXAMPLE 4
CONSTRUCTION OF A HYBRID GDF-8/GDF11 GENE
In order to express GDF-11 protein, a hybrid gene was constructed in which the N- terminal region of GDF-11 was replaced by the analogous region of GDF-8. Such hybrid constructs have been used to produce biologically-active BMP-4 (Hammonds, et al., Mol. Endocrinol., 5:149, 1991) and Vg-1 (Thomsen and Melton, Cell, 74:433, 1993). In order to ensure that the GDF-11 protein produced from the hybrid construct would represent authentic GDF-11, the hybrid gene was constructed in such a manner that the fusion of the two gene fragments would occur precisely at the predicted cleavage sites. In particular, an AvaW restriction site is present in both sequences at the location corresponding to the predicted proteolytic cleavage site. The N-terminal pro-region of
GDF-8 up to this AvaW site was obtained by partial digestion of the clone with AvaW and fused to the C-terminal region of GDF-11 beginning at this AvaW site. The resulting hybrid construct was then inserted into the pMSXND mammalian expression vector (Lee and Nathans, J. Biol. Chem., 263:3521) and transfected into Chinese hamster ovary cells. As shown in FIGURE 5, Western analysis of conditioned medium from G418- resistant cells using antibodies raised against the C-terminal portion of GDF-8 showed that these cells secreted GDF-11 protein into the medium and that at least some of the hybrid protein was proteolytically processed. Furthermore, these studies demonstrate that the antibodies directed against the C-terminal portion of GDF-8 will also react with GDF-11 protein.
EXAMPLE 5
CHROMOSOMAL LOCALIZATION OF GDF-11
In order to map the chromosomal location of GDF-11 , DNA samples from human/rodent somatic cell hybrids (Drwinga, et al., Genomics, 16:311-313, 1993; Dubois and Naylor, Genomics, 16:315-319, 1993) were analyzed by polymerase chain reaction followed by Southern blotting. Polymerase chain reaction was carried out using primer #101 , 5'-
GAGTCCCGCTGCTGCCGATATCC-3', (SEQ ID NO:7) and primer #102, 5'- TAGAGCATGTTGATTGGGGACAT-3', (SEQ ID NO:8) for 35 cycles at 94°C for 2 minutes, 58°C for 1 minutes, and 72°C for 1 minute. These primers correspond to nucleotides 981 to 1003 and the reverse complement of nucleotides 1182 to 1204, respectively, in the human GDF-11 sequence. PCR products were electrophoresed on agarose gels, blotted, and probed with oligonucleotide #104, 5'- AAATATCCGCATACCCATTT-3', (SEQ ID NO:9) which corresponds to a sequence internal to the region flanked by primer #101 and #102. Filters were hybridized in 6 X SSC, 1 X Denhardt's solution, 100 g/ml yeast transfer RNA, and 0.05% sodium pyrophosphate at 50 °C. As shown in FIGURE 6, the human-specific probe detected a band of the predicted size (approximately 224 base pairs) in the positive control sample (total human genomic DNA) and in a single DNA sample from the human/rodent hybrid panel. This positive signal corresponds to human chromosome 12. The human chromosome contained in each of the hybrid cell lines is identified at the top of each of the first 24 lanes (1-22, X, and Y). In the lanes designated CHO, M, and H, the starting DNA template was total genomic DNA from hamster, mouse, and human sources, respectively. In the lane marked B1 , no template DNA was used. Numbers at left indicate the mobilities of DNA standards. These data show that the human GDF-11 gene is located on chromosome 12.
In order to determine the more precise location of GDF-11 on chromosome 12, the GDF- 11 gene was localized by florescence in situ hybridization (FISH). These FISH localization studies were carried out by contract to BIOS laboratories (New Haven, Connecticut). Purified DNA from a human GDF-11 genomic clone was labelled with digoxigenin dUTP by nick translation. Labelled probe was combined with sheared human DNA and hybridized to normal metaphase chromosomes derived from PHA stimulated peripheral blood lymphocytes in a solution containing 50% formamide, 10% dextran sulfate and 2xSSC. Specific hybridization signals were detected by incubating the hybridized slides in fluorescein-conjugated sheep antidigoxigenin antibodies. Slides were then counterstained with propidium iodide and analyzed. As shown in FIGURE 7a, this experiment resulted in the specific labelling of the proximal long arm of a group C chromosome, the size and morphology of which were consistent with chromosome 12. In order to confirm the identity of the specifically labelled chromosome, a second experiment was conducted in which a chromosome 12- specific centromere probe was cohybridized with GDF-11. As shown in FIGURE 7b, this experiment clearly demonstrated that GDF-11 is located at a position which is 23% of the distance from the centromere to the telomere of the long arm of chromosome 12, an area which corresponds to band 12q13 (FIGURE 7c). A total of 85 metaphase cells were analyzed and 80 exhibited specific labelling. Although the invention has been described with reference to the presently preferred embodiment, it should be understood that various modifications can be made without departing from the spirit of the invention. Accordingly, the invention is limited only by the following claims.
SEQUENCE LISTING
(1) GENERAL INFORMATION:
(i) APPLICANT: The Johns Hopkins University School of Medicine
(ii) TITLE OF INVENTION: GROWTH DIFFERENTIATION FACTOR-11
(iii) NUMBER OF SEQUENCES: 9
(iv) CORRESPONDENCE ADDRESS:
(A) ADDRESSEE: Fish & Richardson P.C.
(B) STREET: 4225 Executive Square, Suite 1400 (C) CITY: La Jolla
(D) STATE: California
(E) COUNTRY: US
(F) ZIP: 92037
(v) COMPUTER READABLE FORM: (A) MEDIUM TYPE: Floppy disk
(B) COMPUTER: IBM PC compatible
(C) OPERATING SYSTEM: PC-DOS/MS-DOS
(D) SOFTWARE: Patentin Release #1.0, Version #1.25
( i) CURRENT APPLICATION DATA: (A) APPLICATION NUMBER: PCT/US95/
(B) FILING DATE: 07-JUL-1995
(C) CLASSIFICATION:
(viii) ATTORNEY/AGENT INFORMATION:
(A) NAME: HAILE, PH.D., LISA A. (B) REGISTRATION NUMBER: 38,347
(C) REFERENCE/DOCKET NUMBER: 07265/036WO1
(ix) TELECOMMUNICATION INFORMATION:
(A) TELEPHONE: 619/678-5070
(B) TELEFAX: 619/678-5099 (2) INFORMATION FOR SEQ ID NO:l:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 1393 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(vii) IMMEDIATE SOURCE:
(B) CLONE: HUMAN GDF-11
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 54..1274
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:l:
CCGCGGGACT CCGGCGTCCC CGCCCCCCAG TCCTCCCTCC CCTCCCCTCC AGC ATG 56 Met
1
GTG CTC GCG GCC CCG CTG CTG CTG GGC TTC CTG CTC CTC GCC CTG GAG 104 Val Leu Ala Ala Pro Leu Leu Leu Gly Phe Leu Leu Leu Ala Leu Glu 5 10 15
CTG CGG CCC CGG GGG GAG GCG GCC GAG GGC CCC GCG GCG GCG GCG GCG 152 Leu Arg Pro Arg Gly Glu Ala Ala Glu Gly Pro Ala Ala Ala Ala Ala 20 25 30
GCG GCG GCG GCG GCG GCA GCG GCG GGG GTC GGG GGG GAG CGC TCC AGC 200 Ala Ala Ala Ala Ala Ala Ala Ala Gly Val Gly Gly Glu Arg Ser Ser 35 40 45
CGG CCA GCC CCG TCC GTG GCG CCC GAG CCG GAC GGC TGC CCC GTG TGC 248 Arg Pro Ala Pro Ser Val Ala Pro Glu Pro Asp Gly Cys Pro Val Cys 50 55 60 65
GTT TGG CGG CAG CAC AGC CGC GAG CTG CGC CTA GAG AGC ATC AAG TCG 296 Val Trp Arg Gin His Ser Arg Glu Leu Arg Leu Glu Ser lie Lys Ser
70 75 80 CAG ATC TTG AGC AAA CTG CGG CTC AAG GAG GCG CCC AAC ATC AGC CGC 344 Gin lie Leu Ser Lys Leu Arg Leu Lys Glu Ala Pro Asn lie Ser Arg 85 90 95
GAG GTG GTG AAG CAG CTG CTG CCC AAG GCG CCG CCG CTG CAG CAG ATC 392 Glu Val Val Lys Gin Leu Leu Pro Lys Ala Pro Pro Leu Gin Gin lie 100 105 110
CTG GAC CTA CAC GAC TTC CAG GGC GAC GCG CTG CAG CCC GAG GAC TTC 440 Leu Asp Leu His Asp Phe Gin Gly Asp Ala Leu Gin Pro Glu Asp Phe 115 120 125
CTG GAG GAG GAC GAG TAC CAC GCC ACC ACC GAG ACC GTC ATT AGC ATG 488 Leu Glu Glu Asp Glu Tyr His Ala Thr Thr Glu Thr Val lie Ser Met 13C 135 140 145
GCC CAG GAG ACG GAC CCA GCA GTA CAG ACA GAT GGC AGC CCT CTC TGC 536 Ala Gin Glu Thr Asp Pro Ala Val Gin Thr Asp Gly Ser Pro Leu Cys 150 155 160
TGC CAT TTT CAC TTC AGC CCC AAG GTG ATG TTC ACA AAG GTA CTG AAG 584 Cys His Phe His Phe Ser Pro Lys Val Met Phe Thr Lys Val Leu Lys 165 170 175
GCC CAG CTG TGG GTG TAC CTA CGG CCT GTA CCC CGC CCA GCC ACA GTC 632 Ala Gin Leu Trp Val Tyr Leu Arg Pro Val Pro Arg Pro Ala Thr Val 180 185 190
TAC CTG CAG ATC TTG CGA CTA AAA CCC CTA ACT GGG GAA GGG ACC GCA 680 Tyr Leu Gin lie Leu Arg Leu Lys Pro Leu Thr Gly Glu Gly Thr Ala 195 200 205
GGG GGA GGG GGC GGA GGC CGG CGT CAC ATC CGT ATC CGC TCA CTG AAG 728 Gly Gly Gly Gly Gly Gly Arg Arg His lie Arg lie Arg Ser Leu Lys 210 215 220 225
ATT GAG CTG CAC TCA CGC TCA GGC CAT TGG CAG AGC ATC GAC TTC AAG 776 lie Glu Leu His Ser Arg Ser Gly His Trp Gin Ser lie Asp Phe Lys 230 235 240
CAA GTG CTA CAC AGC TGG TTC CGC CAG CCA CAG AGC AAC TGG GGC ATC 824 Glr. Val Leu His Ser Trp Phe Arg Gin Pro Gin Ser Asn Trp Gly lie 245 250 255 GAG ATC AAC GCC TTT GAT CCC AGT GGC ACA GAC CTG GCT GTC ACC TCC 872 Glu He Asn Ala Phe Asp Pro Ser Gly Thr Asp Leu Ala Val Thr Ser 260 265 270
CTG GGG CCG GGA GCC GAG GGG CTG CAT CCA TTC ATG GAG CTT CGA GTC 920 Leu Gly Pro Gly Ala Glu Gly Leu His Pro Phe Met Glu Leu Arg Val 275 280 285
CTA GAG AAC ACA AAA CGT TCC CGG CGG AAC CTG GGT CTG GAC TGC GAC 968 Leu Glu Asn Thr Lys Arg Ser Arg Arg Asn Leu Gly Leu Asp Cys Asp 290 295 300 305
GAG CAC TCA AGC GAG TCC CGC TGC TGC CGA TAT CCC CTC ACA GTG GAC 1016 Glu His Ser Ser Glu Ser Arg Cys Cys Arg Tyr Pro Leu Thr Val Asp 310 315 320
TTT GAG GCT TTC GGC TGG GAC TGG ATC ATC GCA CCT AAG CGC TAC AAG 1064 Phe Glu Ala Phe Gly Trp Asp Trp He He Ala Pro Lys Arg Tyr Lys 325 330 335
GCC AAC TAC TGC TCC GGC CAG TGC GAG TAC ATG TTC ATG CAA AAA TAT 1112 Ala Asn Tyr Cys Ser Gly Gin Cys Glu Tyr Met Phe Met Gin Lys Tyr 340 345 350
CCG CAT ACC CAT TTG GTG CAG CAG GCC AAT CCA AGA GGC TCT GCT GGG 1160 Pro His Thr His Leu Val Gin Gin Ala Asn Pro Arg Gly Ser Ala Gly 355 360 365
CCC TGT TGT ACC CCC ACC AAG ATG TCC CCA ATC AAC ATG CTC TAC TTC 1208 Pro Cys Cys Thr Pro Thr Lys Met Ser Pro He Asn Met Leu Tyr Phe 370 375 380 385
AAT GAC AAG CAG CAG ATT ATC TAC GGC AAG ATC CCT GGC ATG GTG GTG 1256 Asn Asp Lys Gin Gin He He Tyr Gly Lys He Pro Gly Met Val Val 390 395 400
GAT CGC TGT GGC TGC TCT TAAGTGGGTC ACTACAAGCT GCTGGAGCAA 1304
Asp Arg Cys Gly Cys Ser 405
AGACTTGGTG GGTGGGTAAC TTAACCTCTT CACAGAGGAT AAAAAATGCT TGTGAGTATG 1364
ACAGAAGGGA ATAAACAGGC TTAAAGGGT 1393 (2) INFORMATION FOR SEQ ID NO:2:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 407 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(Xi) SEQUENCE DESCRIPTION: SEQ ID NO:2:
Met Val Leu Ala Ala Pro Leu Leu Leu Gly Phe Leu Leu Leu Ala Leu 1 5 10 15
Glu Leu Arg Pro Arg Gly Glu Ala Ala Glu Gly Pro Ala Ala Ala Ala
20 25 30
Ala Ala Ala Ala Ala Ala Ala Ala Ala Gly Val Gly Gly Glu Arg Ser 35 40 45
Ser Arg Pro Ala Pro Ser Val Ala Pro Glu Pro Asp Gly Cys Pro Val 50 55 60
Cys Val Trp Arg Gin His Ser Arg Glu Leu Arg Leu Glu Ser He Lys 65 70 75 80
Ser Gin He Leu Ser Lys Leu Arg Leu Lys Glu Ala Pro Asn He Ser 85 90 95
Arg Glu Val Val Lys Gin Leu Leu Pro Lys Ala Pro Pro Leu Gin Gin
100 105 110
He Leu Asp Leu His Asp Phe Gin Gly Asp Ala Leu Gin Pro Glu Asp 115 120 125
Phe Leu Glu Glu Asp Glu Tyr His Ala Thr Thr Glu Thr Val He Ser 130 135 140
Met Ala Gin Glu Thr Asp Pro Ala Val Gin Thr Asp Gly Ser Pro Leu 145 150 155 160
Cys Cys His Phe His Phe Ser Pro Lys Val Met Phe Thr Lys Val Leu 165 170 175
Lys Ala Gin Leu Trp Val Tyr Leu Arg Pro Val Pro Arg Pro Ala Thr 180 185 190
Val Tyr Leu Gin He Leu Arg Leu Lys Pro Leu Thr Gly Glu Gly Thr 195 200 205
Ala Gly Gly Gly Gly Gly Gly Arg Arg His He Arg He Arg Ser Leu 210 215 220
Lys He Glu Leu His Ser Arg Ser Gly His Trp Gin Ser He Asp Phe 225 230 235 240
Lys Gin Val Leu His Ser Trp Phe Arg Gin Pro Gin Ser Asn Trp Gly 245 250 255
He Glu He Asn Ala Phe Asp Pro Ser Gly Thr Asp Leu Ala Val Thr
260 265 270
Ser Leu Gly Pro Gly Ala Glu Gly Leu His Pro Phe Met Glu Leu Arg 275 280 285
Val Leu Glu Asn Thr Lys Arg Ser Arg Arg Asn Leu Gly Leu Asp Cys 290 295 300
Asp Glu His Ser Ser Glu Ser Arg Cys Cys Arg Tyr Pro Leu Thr Val 305 310 315 320
Asp Phe Glu Ala Phe Gly Trp Asp Trp He He Ala Pro Lys Arg Tyr 325 330 335
Lys Ala Asn Tyr Cys Ser Gly Gin Cys Glu Tyr Met Phe Met Gin Lys
340 345 350
Tyr Pro His Thr His Leu Val Gin Gin Ala Asn Pro Arg Gly Ser Ala 355 360 365
Gly Pro Cys Cys Thr Pro Thr Lys Met Ser Pro He Asn Met Leu Tyr 370 375 380
Phe Asn Asp Lys Gin Gin He He Tyr Gly Lys He Pro Gly Met Val 385 390 395 400
Val Asp Arg Cys Gly Cys Ser 405 (2) INFORMATION FOR SEQ ID NO:3:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 630 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(vii) IMMEDIATE SOURCE:
(B) CLONE: MOUSE GDF-11
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 198..575
( i) SEQUENCE DESCRIPTION: SEQ ID NO: 3:
TCTAGATGTC AAGAGAAGTG GTCACAATGT CTGGGTGGGA GCCGTAAACA AGCCAAGAGG 60
TTATGGTTTC TGGTCTGATG CTCCTGTTGA GATCAGGAAA TGTTCAGGAA ATCCCCTGTT 120
GAGATGTAGG AAAGTAAGAG GTAAGAGACA TTGTTGAGGG TCATGTCACA TCTCTTTCCC 180
CTCTCCCTGA CCCTCAG CAT CCT TTC ATG GAG CTT CGA GTC CTA GAG AAC 230
His Pro Phe Met Glu Leu Arg Val Leu Glu Asn 1 5 10
ACG AAA AGG TCC CGG CGG AAC CTA GGC CTG GAC TGC GAT GAA CAC TCG 278 Thr Lys Arg Ser Arg Arg Asn Leu Gly Leu Asp Cys Asp Glu His Ser 15 20 25
AGT GAG TCC CGC TGC TGC CGA TAT CCT CTC ACA GTG GAC TTT GAG GCT 326 Ser Glu Ser Arg Cys Cys Arg Tyr Pro Leu Thr Val Asp Phe Glu Ala 30 35 40
TTT GGC TGG GAC TGG ATC ATC GCA CCT AAG CGC TAC AAG GCC AAC TAC 374 Phe Gly Trp Asp Trp He He Ala Pro Lys Arg Tyr Lys Ala Asn Tyr 45 50 55
TGC TCC GGC CAG TGC GAA TAC ATG TTC ATG CAA AAG TAT CCA CAC ACC 422 Cys Ser Gly Gin Cys Glu Tyr Met Phe Met Gin Lys Tyr Pro His Thr 60 65 70 75
CAC TTG GTG CAA CAG GCC AAC CCA AGA GGC TCT GCT GGG CCC TGC TGC 470 His- Leu Val Gin Gin Ala Asn Pro Arg Gly Ser Ala Gly Pro Cys Cys 80 85 90
ACC CCT ACC AAG ATG TCC CCA ATC AAC ATG CTC TAC TTC AAT GAC AAG 518 Thr Pro Thr Lys Met Ser Pro He Asn Met Leu Tyr Phe Asn Asp Lys 95 100 105
CAG CAG ATT ATC TAC GGC AAG ATC CCT GGC ATG GTG GTG GAT CGA TGT 566 Gin Gin He He Tyr Gly Lys He Pro Gly Met Val Val Asp Arg Cys 110 115 120
GGC TGC TCC TAAGTTGTGG GCTACAGTGG ATGCCTCCCT CAGACCCTAC 615
Gly Cys Ser 125
CCCAAGAACC CCAGC 630
(2) INFORMATION FOR SEQ ID NO:4:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 126 amino acids
(B) TYPE: amino acid (D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(xi) SEQUENCE DESCRIPTION: SEQ ID NO:4:
His Pro Phe Met Glu Leu Arg Val Leu Glu Asn Thr Lys Arg Ser Arg 1 5 10 15
Arg Asn Leu Gly Leu Asp Cys Asp Glu His Ser Ser Glu Ser Arg Cys 20 25 30
Cys Arg Tyr Pro Leu Thr Val Asp Phe Glu Ala Phe Gly Trp Asp Trp 35 40 45
He He Ala Pro Lys Arg Tyr Lys Ala Asn Tyr Cys Ser Gly Gin Cys 50 55 60 Glu Tyr Met Phe Met Gin Lys Tyr Pro His Thr His Leu Val Gin Gin 65 70 75 80
Ala Asn Pro Arg Gly Ser Ala Gly Pro Cys Cys Thr Pro Thr Lys Met 85 90 95
Ser Pro He Asn Met Leu Tyr Phe Asn Asp Lys Gin Gin He He Tyr
100 105 110
Gly Lys He Pro Gly Met Val Val Asp Arg Cys Gly Cys Ser 115 120 125
(2) INFORMATION FOR SEQ ID NO:5:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 375 amino acids
(B) TYPE: amino acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(vii) IMMEDIATE SOURCE: (B) CLONE: GDF-8
(ix) FEATURE:
(A) NAME/KEY: Protein (B) LOCATION: 1..375
( i) SEQUENCE DESCRIPTION: SEQ ID NO:5:
Met Gin Lys Leu Gin Leu Cys Val Tyr He Tyr Leu Phe Met Leu He 1 5 10 15
Val Ala Gly Pro Val Asp Leu Asn Glu Asn Ser Glu Gin Lys Glu Asn 20 25 30
Val Glu Lys Glu Gly Leu Cys Asn Ala Cys Thr Trp Arg Gin Asn Thr 35 40 45
Lys Ser Ser Arg He Glu Ala He Lys He Gin He Leu Ser Lys Leu 50 55 60 Arg Leu Glu Thr Ala Pro Asn He Ser Lys Asp Val He Arg Gin Leu 65 70 75 80
Leu Pro Lys Ala Pro Pro Leu Arg Glu Leu He Asp Gin Tyr .Asp Val 85 90 95
Gin Arg Asp Asp Ser Ser Asp Gly Ser Leu Glu Asp Asp Asp Tyr His
100 105 110
Ala Thr Thr Glu Thr He He Thr Met Pro Thr Glu Ser Asp Phe Leu 115 120 125
Met Gin Val Asp Gly Lys Pro Lys Cys Cys Phe Phe Lys Phe Ser Ser 130 135 140
Lys He Gin Tyr Asn Lys Val Val Lys Ala Gin Leu Trp He Tyr Leu 145 150 155 160
Arg Pro Val Glu Thr Pro Thr Thr Val Phe Val Gin He Leu Arg Leu 165 170 175
He Lys Pro Met Lys Asp Gly Thr Arg Tyr Thr Gly He Arg Ser Leu
180 185 190
Lys Leu Asp Met Asn Pro Gly Thr Gly He Trp Gin Ser He Asp Val 195 200 205
Lys Thr Val Leu Gin Asn Trp Leu Lys Gin Pro Glu Ser Asn Leu Gly 210 215 220
He Glu He Lys Ala Leu Asp Glu Asn Gly His Asp Leu Ala Val Thr 225 230 235 240
Phe Pro Gly Pro Gly Glu Asp Gly Leu Asn Pro Phe Leu Glu Val Lys 245 250 255
Val Thr Asp Thr Pro Lys Arg Ser Arg Arg Asp Phe Gly Leu Asp Cys
260 265 270
Asp Glu His Ser Thr Glu Ser Arg Cys Cys Arg Tyr Pro Leu Thr Val 275 280 285
Asp Phe Glu Ala Phe Gly Trp Asp Trp He He Ala Pro Lys Arg Tyr 290 295 300 Lys Ala Asn Tyr Cys Ser Gly Glu Cys Glu Phe Val Phe Leu Gin Lys 305 310 315 320
■ Tyr Pro His Thr His Leu Val His Gin Ala Asn Pro Arg Gly .Ser Ala 325 330 335
Gly Pro Cys Cys Thr Pro Thr Lys Met Ser Pro He Asn Met Leu Tyr
340 345 350
Phe Asn Gly Lys Glu Gin He He Tyr Gly Lys He Pro Ala Met Val 355 360 365
Val Asp Arg Cys Gly Cys Ser 370 375
(2) INFORMATION FOR SEQ ID NO:6:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 407 amino acids
(B) TYPE: amino acid (C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: protein
(vii) IMMEDIATE SOURCE: (B) CLONE: GDF-11
(ix) FEATURE:
(A) NAME/KEY: Protein
(B) LOCATION: 1..407
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 6:
Met Val Leu Ala Ala Pro Leu Leu Leu Gly Phe Leu Leu Leu Ala Leu 1 5 10 15
Glu Leu Arg Pro Arg Gly Glu Ala Ala Glu Gly Pro Ala Ala Ala Ala 20 25 30
Ala Ala Ala Ala Ala Ala Ala Ala Ala Gly Val Gly Gly Glu Arg Ser 35 40 45 Ser Arg Pro Ala Pro Ser Val Ala Pro Glu Pro Asp Gly Cys Pro Val 50 55 60
• Cys Val Trp Arg Gin His Ser Arg Glu Leu Arg Leu Glu Ser He Lys 65 70 75 80
Ser Gin He Leu Ser Lys Leu Arg Leu Lys Glu Ala Pro Asn He Ser
85 90 95
Arg Glu Val Val Lys Gin Leu Leu Pro Lys Ala Pro Pro Leu Gin Gin 100 105 110
He Leu Asp Leu His Asp Phe Gin Gly Asp Ala Leu Gin Pro Glu Asp 115 120 125
Phe Leu Glu Glu Asp Glu Tyr His Ala Thr Thr Glu Thr Val He Ser 130 135 140
Met Ala Gin Glu Thr Asp Pro Ala Val Gin Thr Asp Gly Ser Pro Leu 145 150 155 160
Cys Cys His Phe His Phe Ser Pro Lys Val Met Phe Thr Lys Val Leu
165 170 175
Lys Ala Gin Leu Trp Val Tyr Leu Arg Pro Val Pro Arg Pro Ala Thr 180 185 190
Val Tyr Leu Gin He Leu Arg Leu Lys Pro Leu Thr Gly Glu Gly Thr 195 200 205
Ala Gly Gly Gly Gly Gly Gly Arg Arg His He Arg He Arg Ser Leu 210 215 220
Lys He Glu Leu His Ser Arg Ser Gly His Trp Gin Ser He Asp Phe 225 230 235 240
Lys Gin Val Leu His Ser Trp Phe Arg Gin Pro Gin Ser Asn Trp Gly
245 250 255
He Glu He Asn Ala Phe Asp Pro Ser Gly Thr Asp Leu Ala Val Thr 260 265 270
Ser Leu Gly Pro Gly Ala Glu Gly Leu His Pro Phe Met Glu Leu Arg 275 280 285 Val Leu Glu Asn Thr Lys Arg Ser Arg Arg Asn Leu Gly Leu Asp Cys 290 295 300
Asp Glu His Ser Ser Glu Ser Arg Cys Cys Arg Tyr Pro Leu Thr Val 305 310 315 320
Asp Phe Glu Ala Phe Gly Trp Asp Trp He He Ala Pro Lys Arg Tyr
325 330 335
Lys Ala Asn Tyr Cys Ser Gly Gin Cys Glu Tyr Met Phe Met Gin Lys 340 345 350
Tyr Pro His Thr His Leu Val Gin Gin Ala Asn Pro Arg Gly Ser Ala 355 360 365
Gly Pro Cys Cys Thr Pro Thr Lys Met Ser Pro He Asn Met Leu Tyr 370 375 380
Phe Asn Asp Lys Gin Gin He He Tyr Gly Lys He Pro Gly Met Val 385 390 395 400
Val Asp Arg Cys Gly Cys Ser
405
(2) INFORMATION FOR SEQ ID NO:7:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 23 base pairs (B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(ix) FEATURE: (A) NAME/KEY: CDS
(B) LOCATION: 1..23
( i) SEQUENCE DESCRIPTION: SEQ ID NO:7:
GAGTCCCGCT GCTGCCGATA TCC 23 (2) INFORMATION FOR SEQ ID NO: 8:
(i) SEQUENCE CHARACTERISTICS:
(A) LENGTH: 23 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(ix) FEATURE:
(A) NAME/KEY: CDS (B) LOCATION: 1..23
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 8:
TAGAGCATGT TGATTGGGGA CAT 23
(2) INFORMATION FOR SEQ ID NO: 9:
(i) SEQUENCE CHARACTERISTICS: (A) LENGTH: 20 base pairs
(B) TYPE: nucleic acid
(C) STRANDEDNESS: single
(D) TOPOLOGY: linear
(ii) MOLECULE TYPE: DNA (genomic)
(ix) FEATURE:
(A) NAME/KEY: CDS
(B) LOCATION: 1..20
(xi) SEQUENCE DESCRIPTION: SEQ ID NO: 9:
AAATATCCGC ATACCCATTT 20

Claims

1. Substantially pure growth differentiation factor-11 (GDF-11).
2. An isolated polynucleotide sequence encoding the GDF-11 polypeptide of claim 1.
3. The polynucleotide of claim 2, wherein the GDF-11 nucleotide sequence is selected from the group consisting of: a. SEQ ID NO: 1, wherein T can also be U; b. SEQ ID NO:3, wherein T can also be U; c. nucleic acid sequences complementary to SEQ ID NO:1; d. nucleic acid sequences complementary to SEQ ID NO:3; e. fragments of a. or c. that are at least 15 bases in length and that will selectively hybridize to DNA which encodes the GDF-11 protein of SEQ ID NO:2; and f. fragments of b. or d. that are at least 15 bases in length and that will selectively hybridize to DNA which encodes the GDF-11 protein of SEQ ID NO:4.
4. The polynucleotide sequence of claim 2, wherein the polynucleotide is isolated from a mammalian cell.
5. The polynucleotide of claim 4, wherein the mammalian cell is selected from the group consisting of mouse, rat, and human cell.
6. An expression vector including the polynucleotide of claim 2.
7. The vector of claim 6, wherein the vector is a plasmid.
8. The vector of claim 6, wherein the vector is a virus.
9. A host cell stably transformed with the vector of claim 6.
10. The host cell of claim 9, wherein the cell is prokaryotic.
11. The host cell of claim 9, wherein the cell is eukaryotic.
12. Antibodies that bind to the polypeptide of claim 1 or fragments thereof.
13. The antibodies of claim 12, wherein the antibodies are polyclonal.
14. The antibodies of claim 12, wherein the antibodies are monoclonal.
15. A method of detecting a cell proliferative disorder comprising contacting the antibody of claim 12 with a specimen of a subject suspected of having a GDF-11 associated disorder and detecting binding of the antibody.
16. The method of claim 15, wherein the cell is a muscle cell.
17. The method of claim 15, wherein the detecting is in vivo.
18. The method of claim 17, wherein the antibody is detectably labeled.
19. The method of claim 18, wherein the detectable label is selected from the group consisting of a radioisotope, a fluorescent compound, a bioluminescent compound and a chemiluminescent compound.
20. The method of claim 15, wherein the detection is in vitro.
21. The method of claim 20, wherein the antibody is detectably labeled.
22. The method of claim 21, wherein the label is selected from the group -consisting of a radioisotope, a fluorescent compound, a bioluminescent compound, a chemoluminescent compound and an enzyme.
23. A method of treating a cell proliferative disorder associated with expression of GDF-11 , comprising contacting the cells with a reagent which suppresses the GDF-11 activity.
24. The method of claim 23, wherein the reagent is an anti-GDF-11 antibody.
25. The method of claim 23, wherein the reagent is a GDF-11 antisense sequence.
26. The method of claim 23, wherein the cell is a muscle cell.
27. The method of claim 23, wherein the reagent which suppresses GDF-11 activity is introduced to a cell using a vector.
28. The method of claim 27, wherein the vector is a colloidal dispersion system.
29. The method of claim 28, wherein the colloidal dispersion system is a Iiposome.
30. The method of claim 29, wherein the Iiposome is essentially target specific.
31. The method of claim 30, wherein the Iiposome is anatomically targeted.
32. The method of claim 31 , wherein the Iiposome is mechanistically targeted.
33. The method of claim 32, wherein the mechanistic targeting is passive.
34. The method of claim 32, wherein the mechanistic targeting is active..
35. The method of claim 34, wherein the Iiposome is actively targeted by coupling with a moiety selected from the group consisting of a sugar, a glycolipid, and a protein.
36. The method of claim 35, wherein the protein moiety is an antibody.
37. The method of claim 36, wherein the vector is a virus.
38. The method of claim 37, wherein the virus is an RNA virus.
39. The method of claim 38, wherein the RNA virus is a retrovirus.
40. The method of claim 39, wherein the retrovirus is essentially target specific.
41. The method of claim 40, wherein a moiety for target specificity is encoded by a polynucleotide inserted into the retroviral genome.
42. The method of claim 40, wherein a moiety for target specificity is selected from the group consisting of a sugar, a glycolipid, and a protein.
43. The method of claim 42, wherein the protein is an antibody.
PCT/US1995/008543 1994-07-08 1995-07-07 Growth differentiation factor-11 WO1996001845A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
JP8504413A JPH10502811A (en) 1994-07-08 1995-07-07 Growth differentiation factor-11
DE69534468T DE69534468T2 (en) 1994-07-08 1995-07-07 GROWTH DIFFERENTIATION FACTOR 11
EP95925559A EP0776337B1 (en) 1994-07-08 1995-07-07 Growth differentiation factor-11
CA002194660A CA2194660C (en) 1994-07-08 1995-07-07 Growth differentiation factor-11
US08/765,875 US5914234A (en) 1994-07-08 1995-07-07 Methods of detecting growth differentiation factor-11
AT95925559T ATE305036T1 (en) 1994-07-08 1995-07-07 GROWTH DIFFERENTIATION FACTOR-11

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US27276394A 1994-07-08 1994-07-08
US08/272,763 1994-07-08

Publications (1)

Publication Number Publication Date
WO1996001845A1 true WO1996001845A1 (en) 1996-01-25

Family

ID=23041174

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1995/008543 WO1996001845A1 (en) 1994-07-08 1995-07-07 Growth differentiation factor-11

Country Status (9)

Country Link
US (2) US5914234A (en)
EP (2) EP0776337B1 (en)
JP (1) JPH10502811A (en)
AT (1) ATE305036T1 (en)
CA (1) CA2194660C (en)
DE (1) DE69534468T2 (en)
DK (1) DK0776337T3 (en)
ES (1) ES2251721T3 (en)
WO (1) WO1996001845A1 (en)

Cited By (70)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5846770A (en) * 1994-11-22 1998-12-08 Genetics Institute, Inc. DNA molecules encoding human chordin
EP0904292A1 (en) * 1996-03-29 1999-03-31 The Johns Hopkins University School Of Medicine Growth differentiation factor-14
WO1999024058A2 (en) * 1997-11-07 1999-05-20 Genetics Institute, Inc. Neuronal uses of bmp-11
WO1999040181A1 (en) * 1998-02-05 1999-08-12 The Johns Hopkins University School Of Medicine Growth differentiation factor-8
WO1999042573A1 (en) * 1998-02-19 1999-08-26 Biostar Inc. Immunological methods to modulate myostatin in vertebrate subjects
US5965403A (en) * 1996-09-18 1999-10-12 Genetics Institute, Inc. Nucleic acids encoding bone morphogenic protein-16 (BMP-16)
US6027917A (en) * 1997-12-10 2000-02-22 Genetics Institute, Inc. Bone morphogenetic protein (BMP)-17 and BMP-18 compositions
WO2000043781A2 (en) * 1999-01-21 2000-07-27 Metamorphix, Inc. Growth differentiation factor inhibitors and uses therefor
EP1100887A1 (en) * 1998-07-28 2001-05-23 The Johns Hopkins University School Of Medicine Growth differentiation factor-11
US6340668B1 (en) 1993-05-12 2002-01-22 Genetics Institute, Inc. Neuronal uses of BMP-11
US6465239B1 (en) 1993-03-19 2002-10-15 The John Hopkins University School Of Medicine Growth differentiation factor-8 nucleic acid and polypeptides from aquatic species and non-human transgenic aquatic species
WO2002100331A2 (en) 2001-06-08 2002-12-19 Wyeth Calcium phosphate delivery vehicles for osteoinductive proteins
US6500664B1 (en) 1993-03-19 2002-12-31 The Johns Hopkins University School Of Medicine Growth differentiation factor-8 family nucleic acid sequences
US6517835B2 (en) 1994-07-08 2003-02-11 The Johns Hopkins University Growth differentiation factor-11
US6593106B1 (en) 1997-10-08 2003-07-15 Genetics Institute Llc Methods of refolding proteins
WO2003066081A2 (en) * 2002-02-05 2003-08-14 K.U.Leuven Research And Development Bmp inhibitors for the treatment of spondyloarthropathies
US6607884B1 (en) 1993-03-19 2003-08-19 The Johns Hopkins University School Of Medicine Methods of detecting growth differentiation factor-8
WO2005089829A2 (en) 2004-03-10 2005-09-29 Scil Technology Gmbh Coated implants, their manufacturing and use thereof
EP1719531A2 (en) 1997-03-20 2006-11-08 Stryker Corporation Osteogenic devices and methods of use thereof for repair of bones
US7147839B2 (en) 1998-05-29 2006-12-12 Curis, Inc. Methods for evaluating tissue morphogenesis and activity
US7192717B2 (en) 2002-02-21 2007-03-20 Wyeth GASP1: a follistatin domain containing protein
US7202210B2 (en) 2001-02-08 2007-04-10 Wyeth Modified and stabilized GDF propeptides and uses thereof
WO2007067561A2 (en) 2005-12-06 2007-06-14 Etex Corporation Porous calcium phosphate bone material
US7261893B2 (en) 2002-10-22 2007-08-28 Wyeth Neutralizing antibodies against GDF-8 and uses therefor
US7320789B2 (en) 2001-09-26 2008-01-22 Wyeth Antibody inhibitors of GDF-8 and uses thereof
US7332575B2 (en) 1994-03-18 2008-02-19 The Johns Hopkins University School Of Medicine Growth differentiation factor-8 nucleic acid and polypeptide from aquatic species, and transgenic aquatic species
US7384753B2 (en) 1994-07-08 2008-06-10 The Johns Hopkins University School Of Medicine Growth differentiation factor-11
US7393682B1 (en) 1993-03-19 2008-07-01 The Johns Hopkins University School Of Medicine Polynucleotides encoding promyostatin polypeptides
WO2008082563A2 (en) 2006-12-21 2008-07-10 Stryker Corporation Sustained-release formulations comprising crystals, macromolecular gels, and particulate suspensions of biologic agents
EP1988395A1 (en) 1997-05-30 2008-11-05 Curis, Inc. Methods for evaluating tissue morphogenesis and morphogenic activity
US7566768B1 (en) 1995-10-26 2009-07-28 The Johns Hopkins University School Of Medicine Promyostatin peptides and methods of using same
US7572763B2 (en) 2002-02-21 2009-08-11 Wyeth Follistatin domain containing proteins
US7575751B2 (en) 2004-04-27 2009-08-18 Research Development Foundation Activin-A mutants
US7632499B2 (en) 2005-10-12 2009-12-15 Eli Lilly And Company Anti-myostatin antibodies
US7635760B2 (en) 2005-10-06 2009-12-22 Eli Lilly And Company Anti-myostatin antibodies
EP2174674A1 (en) 1999-02-01 2010-04-14 Genetics Institute, LLC Methods and compositions for healing and repair of articular cartilage
WO2010093925A2 (en) 2009-02-12 2010-08-19 Stryker Corporation PERIPHERAL ADMINISTRATION OF PROTEINS INCLUDING TGF-β SUPERFAMILY MEMBERS FOR TREATMENT OF SYSTEMIC DISORDERS AND DISEASE
WO2010093941A2 (en) 2009-02-12 2010-08-19 Stryker Corporation COMPOSITIONS AND METHODS FOR MINIMALLY-INVASIVE SYSTEMIC DELIVERY OF PROTEINS INCLUDING TGF-β SUPERFAMILY MEMBERS
US7785587B2 (en) 2003-06-02 2010-08-31 Wyeth Therapeutic methods for muscular or neuromuscular disorders
EP2223698A1 (en) 2004-10-14 2010-09-01 Biomimetic Therapeutics, Inc. Platelet-derived growth factor compositions and method of use thereof
WO2010110974A1 (en) 2009-03-24 2010-09-30 Stryker Corporation Methods and compositions for tissue engineering
WO2010115132A2 (en) 2009-04-03 2010-10-07 Warsaw Orthopedic, Inc. Medical implant with bioactive material and method of making the medical implant
WO2010144696A1 (en) 2009-06-11 2010-12-16 Burnham Institute For Medical Research Directed differentiation of stem cells
WO2011031856A1 (en) 2009-09-09 2011-03-17 Stryker Corporation Bmp -7 for use in treating pain induced by injuries and diseases of an articular joint
EP2298335A1 (en) 2004-05-25 2011-03-23 Stryker Corporation Use of morphogenic proteins for treating cartilage defects
WO2011035094A1 (en) 2009-09-17 2011-03-24 Stryker Corporation Buffers for controlling the ph of bone morphogenetic proteins
US7947649B2 (en) 2008-04-14 2011-05-24 Advanced Technologies And Regenerative Medicine, Llc Liquid buffered GDF-5 formulations
US7956028B2 (en) 2006-12-14 2011-06-07 Johnson & Johnson Regenerative Therapeutics, Llc Protein stabilization formulations
US7964561B2 (en) 2007-06-29 2011-06-21 Advanced Technologies And Regenerative Medicine, Llc Protein formulations for use at elevated temperatures
WO2011087768A1 (en) 2009-12-22 2011-07-21 Stryker Corporation Bmp-7 variants with reduced immunogenicity
EP2374471A1 (en) 2003-09-12 2011-10-12 Wyeth LLC Injectable hardenable calcium phosphate pastes for delivery of osteogenic proteins
US8058237B2 (en) 2007-08-07 2011-11-15 Advanced Technologies & Regenerative Medicine, LLC Stable composition of GDF-5 and method of storage
US8063188B2 (en) 2006-09-05 2011-11-22 Eli Lilly And Company Anti-myostatin antibodies
EP2428233A1 (en) 2006-08-17 2012-03-14 Warsaw Orthopedic, Inc. Medical implant sheets useful for tissue regeneration
EP2540310A1 (en) 2006-05-17 2013-01-02 Stryker Corporation Methods of treating cartilage defects using a soluble morphogenic protein complex
US8372805B1 (en) 1997-03-20 2013-02-12 Stryker Corporation Osteogenic devices and methods of use thereof for repair of endochondral bone, osteochondral and chondral defects
US8741840B2 (en) 2008-02-13 2014-06-03 Washington University BMP-7 for use in treating neointimal hyperplasia
US8870954B2 (en) 2008-09-09 2014-10-28 Biomimetic Therapeutics, Llc Platelet-derived growth factor compositions and methods for the treatment of tendon and ligament injuries
US9161967B2 (en) 2006-06-30 2015-10-20 Biomimetic Therapeutics, Llc Compositions and methods for treating the vertebral column
WO2017015622A3 (en) * 2015-07-22 2017-03-02 Scholar Rock, Inc Gdf11 binding proteins and uses thereof
US9642891B2 (en) 2006-06-30 2017-05-09 Biomimetic Therapeutics, Llc Compositions and methods for treating rotator cuff injuries
US10034945B2 (en) 2012-07-13 2018-07-31 Trustees Of Tufts College Silk powder compaction for production of constructs with high mechanical strength and stiffness
US10258566B2 (en) 2004-10-14 2019-04-16 Biomimetic Therapeutics, Llc Compositions and methods for treating bone
US10751413B2 (en) 2015-09-15 2020-08-25 Scholar Rock, Inc. Anti-pro/latent-Myostatin antibodies and uses thereof
US10882904B2 (en) 2016-01-08 2021-01-05 Scholar Rock, Inc. Methods for inhibiting myostatin activation by administering anti-pro/latent myostatin antibodies
US10946036B2 (en) 2016-06-13 2021-03-16 Scholar Rock, Inc. Use of myostatin inhibitors and combination therapies
EP3821880A1 (en) 2015-10-26 2021-05-19 President and Fellows of Harvard College Oxidized polysaccharides and methods of use thereof
US11135291B2 (en) 2014-11-06 2021-10-05 Scholar Rock, Inc. Methods for making and using anti-myostatin antibodies
US11155611B2 (en) 2017-01-06 2021-10-26 Scholar Rock, Inc. Compositions and methods for making and using anti-myostatin antibodies
US11235030B2 (en) 2010-02-22 2022-02-01 Biomimetic Therapeutics, Llc Platelet-derived growth factor compositions and methods for the treatment of tendinopathies

Families Citing this family (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6696260B1 (en) * 1997-08-01 2004-02-24 The Johns Hopkins University School Of Medicine Methods to identify growth differentiation factor (GDF) binding proteins
US6891082B2 (en) * 1997-08-01 2005-05-10 The Johns Hopkins University School Of Medicine Transgenic non-human animals expressing a truncated activintype II receptor
US6555374B1 (en) * 1999-08-19 2003-04-29 Artecel Sciences, Inc. Multiple mesodermal lineage differentiation potentials for adipose tissue-derived stromal cells and uses thereof
KR100420798B1 (en) * 2001-02-10 2004-03-02 (주)알에이싸이언스 Peptide vector
US6798160B2 (en) * 2001-11-02 2004-09-28 Honda Giken Kogyo Kabushiki Kaisha Electric working machine
US7527966B2 (en) 2002-06-26 2009-05-05 Transgenrx, Inc. Gene regulation in transgenic animals using a transposon-based vector
US20040172667A1 (en) 2002-06-26 2004-09-02 Cooper Richard K. Administration of transposon-based vectors to reproductive organs
WO2004024092A2 (en) * 2002-09-16 2004-03-25 Wyeth Metalloprotease activation of myostatin, and methods of modulating myostatin activity
US20040223966A1 (en) * 2002-10-25 2004-11-11 Wolfman Neil M. ActRIIB fusion polypeptides and uses therefor
US8071364B2 (en) 2003-12-24 2011-12-06 Transgenrx, Inc. Gene therapy using transposon-based vectors
US20060034831A1 (en) * 2004-08-12 2006-02-16 Wyeth Combination therapy for diabetes, obesity and cardiovascular diseases using GDF-8 inhibitors
MX2007011400A (en) * 2005-03-23 2007-10-11 Wyeth Corp Detection of gdf-8 modulating agents.
CN101137906A (en) * 2005-03-23 2008-03-05 惠氏公司 Detection of an immune response to gdf-8 modulating agents
JP5219359B2 (en) 2006-02-21 2013-06-26 日東電工株式会社 Reflective and / or light-blocking adhesive tape or sheet and liquid crystal display device
PE20091163A1 (en) * 2007-11-01 2009-08-09 Wyeth Corp ANTIBODIES FOR GDF8
US8216997B2 (en) 2008-08-14 2012-07-10 Acceleron Pharma, Inc. Methods for increasing red blood cell levels and treating anemia using a combination of GDF traps and erythropoietin receptor activators
US9157097B2 (en) 2008-09-25 2015-10-13 Proteovec Holding, L.L.C. Vectors for production of growth hormone
US9150880B2 (en) 2008-09-25 2015-10-06 Proteovec Holding, L.L.C. Vectors for production of antibodies
US9150881B2 (en) 2009-04-09 2015-10-06 Proteovec Holding, L.L.C. Production of proteins using transposon-based vectors
AU2010258931B2 (en) 2009-06-08 2015-04-23 Acceleron Pharma Inc. Methods for increasing thermogenic adipocytes
MX2011013364A (en) 2009-06-12 2012-03-16 Acceleron Pharma Inc Truncated actriib-fc fusion proteins.
AU2013334660B2 (en) * 2012-10-24 2018-08-09 Celgene Corporation Methods for treating anemia
EP3608419A1 (en) * 2012-10-24 2020-02-12 Celgene Corporation Biomarker for use in treating anemia
KR102279522B1 (en) 2012-11-02 2021-07-19 셀진 코포레이션 Activin-actrii antagonists and uses for treating bone and other disorders
EP2956545A4 (en) * 2013-02-15 2016-07-20 Joslin Diabetes Ct Thymic regeneration
JP2016518357A (en) 2013-04-08 2016-06-23 プレジデント アンド フェローズ オブ ハーバード カレッジ Methods and compositions for rejuvenating skeletal muscle stem cells
US20160287667A1 (en) * 2013-11-08 2016-10-06 President And Fellows Of Harvard College Methods and compositions for rejuvenating neuromuscular junctions
US10670611B2 (en) 2014-09-26 2020-06-02 Somalogic, Inc. Cardiovascular risk event prediction and uses thereof
CN109310742B (en) 2016-01-06 2022-12-02 哈佛学院校长同事会 Treatment with GDF11 prevents weight gain, increases glucose tolerance and reduces fatty liver

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994021681A1 (en) * 1993-03-19 1994-09-29 Johns Hopkins University School Of Medicine Growth differentiation factor-8

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1992000382A1 (en) * 1990-06-15 1992-01-09 Carnegie Institution Of Washington Gdf-1
US5650276A (en) * 1991-03-11 1997-07-22 Creative Biomolecules, Inc. Morphogenic protein screening method
DK0698094T3 (en) * 1993-05-12 2004-05-03 Inst Genetics Llc BMP-11 compositions

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994021681A1 (en) * 1993-03-19 1994-09-29 Johns Hopkins University School Of Medicine Growth differentiation factor-8

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
PROC. NATL. ACAD. SCI. U.S.A., Volume 90, issued July 1993, T.K. SAMPATH et al., "Drosophila Transforming Growth Factor Beta Superfamily Proteins Induce Endochondral Bone Formation in Mammals", pages 6004-6008. *

Cited By (130)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7179884B2 (en) 1993-03-19 2007-02-20 The Johns Hopkins University School Of Medicine Growth differentiation factor-8
US6607884B1 (en) 1993-03-19 2003-08-19 The Johns Hopkins University School Of Medicine Methods of detecting growth differentiation factor-8
US6465239B1 (en) 1993-03-19 2002-10-15 The John Hopkins University School Of Medicine Growth differentiation factor-8 nucleic acid and polypeptides from aquatic species and non-human transgenic aquatic species
US8323964B2 (en) 1993-03-19 2012-12-04 The John Hopkins University School Of Medicine Polynucleotides encoding promyostatin polypeptides
US7534432B2 (en) 1993-03-19 2009-05-19 The Johns Hopkins University School Of Medicine Methods of treating musculodegenerative disease with an antibody that binds growth differentiation factor-8
US7393682B1 (en) 1993-03-19 2008-07-01 The Johns Hopkins University School Of Medicine Polynucleotides encoding promyostatin polypeptides
US6500664B1 (en) 1993-03-19 2002-12-31 The Johns Hopkins University School Of Medicine Growth differentiation factor-8 family nucleic acid sequences
US6340668B1 (en) 1993-05-12 2002-01-22 Genetics Institute, Inc. Neuronal uses of BMP-11
EP1770161A2 (en) * 1993-05-12 2007-04-04 Genetics Institute, LLC BMP-11 compositions
EP1770161A3 (en) * 1993-05-12 2008-04-23 Genetics Institute, LLC BMP-11 compositions
US7332575B2 (en) 1994-03-18 2008-02-19 The Johns Hopkins University School Of Medicine Growth differentiation factor-8 nucleic acid and polypeptide from aquatic species, and transgenic aquatic species
US7384753B2 (en) 1994-07-08 2008-06-10 The Johns Hopkins University School Of Medicine Growth differentiation factor-11
US7976839B2 (en) 1994-07-08 2011-07-12 Johns Hopkins University School Of Medicine Growth differentiation factor-11
US6517835B2 (en) 1994-07-08 2003-02-11 The Johns Hopkins University Growth differentiation factor-11
US5986056A (en) * 1994-11-22 1999-11-16 Lavallie; Edward R. Chordin compositions
US5846770A (en) * 1994-11-22 1998-12-08 Genetics Institute, Inc. DNA molecules encoding human chordin
US7566768B1 (en) 1995-10-26 2009-07-28 The Johns Hopkins University School Of Medicine Promyostatin peptides and methods of using same
EP0904292A1 (en) * 1996-03-29 1999-03-31 The Johns Hopkins University School Of Medicine Growth differentiation factor-14
EP0904292A4 (en) * 1996-03-29 2000-04-19 Univ Johns Hopkins Med Growth differentiation factor-14
US6524802B1 (en) 1996-03-29 2003-02-25 The Johns Hopkins University School Of Medicine Methods of detecting growth differentiation factor-14
US6623934B2 (en) 1996-09-18 2003-09-23 Genetics Institute, Llc. Bone morphogenetic protein-16 (BMP-16)antibodies
US5965403A (en) * 1996-09-18 1999-10-12 Genetics Institute, Inc. Nucleic acids encoding bone morphogenic protein-16 (BMP-16)
US6331612B1 (en) 1996-09-18 2001-12-18 Genetics Institute, Inc. Bone morphogenic protein-16 (BMP-16) compositions
EP1719531A2 (en) 1997-03-20 2006-11-08 Stryker Corporation Osteogenic devices and methods of use thereof for repair of bones
US8372805B1 (en) 1997-03-20 2013-02-12 Stryker Corporation Osteogenic devices and methods of use thereof for repair of endochondral bone, osteochondral and chondral defects
EP1719532A2 (en) 1997-03-20 2006-11-08 Stryker Corporation Osteogenic devices and methods of use thereof for repair of bones
US8354376B2 (en) 1997-03-20 2013-01-15 Stryker Corporation Osteogenic devices and methods of use thereof for repair of endochondral bone, osteochondral and chondral defects
US8802626B2 (en) 1997-03-20 2014-08-12 Stryker Corporation Osteogenic devices and methods of use thereof for repair of endochondral bone, osteochondral and chondral defects
EP1988395A1 (en) 1997-05-30 2008-11-05 Curis, Inc. Methods for evaluating tissue morphogenesis and morphogenic activity
EP2309261A1 (en) 1997-05-30 2011-04-13 Stryker Corporation Methods for evaluating tissue morphogenesis and morphogenic activity
US6593106B1 (en) 1997-10-08 2003-07-15 Genetics Institute Llc Methods of refolding proteins
US6632635B1 (en) 1997-10-08 2003-10-14 Genetics Institute Methods of refolding proteins
US6596511B1 (en) 1997-10-08 2003-07-22 Genetics Institute Llc Methods of refolding proteins
US6593107B1 (en) 1997-10-08 2003-07-15 Genetics Institute Llc Methods of refolding proteins
WO1999024058A3 (en) * 1997-11-07 1999-08-19 Genetics Inst Neuronal uses of bmp-11
AU761940B2 (en) * 1997-11-07 2003-06-12 Genetics Institute, Llc Neuronal uses of BMP-11
WO1999024058A2 (en) * 1997-11-07 1999-05-20 Genetics Institute, Inc. Neuronal uses of bmp-11
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
US6027917A (en) * 1997-12-10 2000-02-22 Genetics Institute, Inc. Bone morphogenetic protein (BMP)-17 and BMP-18 compositions
WO1999040181A1 (en) * 1998-02-05 1999-08-12 The Johns Hopkins University School Of Medicine Growth differentiation factor-8
EP1770162A1 (en) * 1998-02-19 2007-04-04 MetaMorphix International, Inc. Immunological compositions to modulate myostatin in vertebrate subjects
WO1999042573A1 (en) * 1998-02-19 1999-08-26 Biostar Inc. Immunological methods to modulate myostatin in vertebrate subjects
US6369201B1 (en) 1998-02-19 2002-04-09 Metamorphix International, Inc. Myostatin multimers
US7147839B2 (en) 1998-05-29 2006-12-12 Curis, Inc. Methods for evaluating tissue morphogenesis and activity
EP1100887A1 (en) * 1998-07-28 2001-05-23 The Johns Hopkins University School Of Medicine Growth differentiation factor-11
EP1100887A4 (en) * 1998-07-28 2003-01-15 Univ Johns Hopkins Med Growth differentiation factor-11
WO2000043781A2 (en) * 1999-01-21 2000-07-27 Metamorphix, Inc. Growth differentiation factor inhibitors and uses therefor
WO2000043781A3 (en) * 1999-01-21 2001-02-01 Metamorphix Inc Growth differentiation factor inhibitors and uses therefor
EP2174674A1 (en) 1999-02-01 2010-04-14 Genetics Institute, LLC Methods and compositions for healing and repair of articular cartilage
US7737116B2 (en) 2001-02-08 2010-06-15 Wyeth Modified and stabilized GDF propeptides and uses thereof
US8222384B2 (en) 2001-02-08 2012-07-17 Wyeth Llc Modified and stabilized GDF propeptides and uses thereof
US7202210B2 (en) 2001-02-08 2007-04-10 Wyeth Modified and stabilized GDF propeptides and uses thereof
US7560441B2 (en) 2001-02-08 2009-07-14 Wyeth Modified and stabilized GDF propeptides and uses thereof
EP2260860A1 (en) 2001-06-08 2010-12-15 Wyeth LLC Calcium phosphate delivery vehicles for osteoinduktive proteins
WO2002100331A2 (en) 2001-06-08 2002-12-19 Wyeth Calcium phosphate delivery vehicles for osteoinductive proteins
US7731961B1 (en) 2001-09-26 2010-06-08 Wyeth Methods of increasing muscle mass or muscle strength using antibody inhibitors of GDF-8
US8092798B2 (en) 2001-09-26 2012-01-10 Wyeth Llc Method of increasing trabecular bone density in a patient in need thereof by an antibody against GDF-8
US7320789B2 (en) 2001-09-26 2008-01-22 Wyeth Antibody inhibitors of GDF-8 and uses thereof
US9505831B2 (en) 2001-09-26 2016-11-29 Wyeth Llc Isolated cell comprising a nucleic acid encoding antibody inhibitors of gdf-8 and uses thereof
US8710202B2 (en) 2001-09-26 2014-04-29 Wyeth Llc Isolated nucleic acid molecule encoding an antibody that reduces GDF-8 activity
US7371725B2 (en) 2002-02-05 2008-05-13 K.U. Leuven Research & Development Spondyloarthropathies
WO2003066081A2 (en) * 2002-02-05 2003-08-14 K.U.Leuven Research And Development Bmp inhibitors for the treatment of spondyloarthropathies
WO2003066081A3 (en) * 2002-02-05 2004-03-18 Leuven K U Res & Dev Bmp inhibitors for the treatment of spondyloarthropathies
US7572763B2 (en) 2002-02-21 2009-08-11 Wyeth Follistatin domain containing proteins
US7956038B2 (en) 2002-02-21 2011-06-07 Wyeth Llc GASP1: a follistatin domain containing protein
US7585835B2 (en) 2002-02-21 2009-09-08 Wyeth GASP1: a follistatin domain containing protein
US7192717B2 (en) 2002-02-21 2007-03-20 Wyeth GASP1: a follistatin domain containing protein
US7541154B2 (en) 2002-02-21 2009-06-02 Wyeth GASP1: a follistatin domain containing protein
US8940874B2 (en) 2002-10-22 2015-01-27 Wyeth Llc Neutralizing antibodies against GDF-8 and uses therefor
US7261893B2 (en) 2002-10-22 2007-08-28 Wyeth Neutralizing antibodies against GDF-8 and uses therefor
US7655763B2 (en) 2002-10-22 2010-02-02 Wyeth Neutralizing antibodies against GDF-8 and uses therefor
US8420082B2 (en) 2002-10-22 2013-04-16 Wyeth Llc Neutralizing antibodies against GDF-8 and uses therefor
US7785587B2 (en) 2003-06-02 2010-08-31 Wyeth Therapeutic methods for muscular or neuromuscular disorders
EP2374471A1 (en) 2003-09-12 2011-10-12 Wyeth LLC Injectable hardenable calcium phosphate pastes for delivery of osteogenic proteins
US8372419B2 (en) 2004-03-10 2013-02-12 Scil Technology Gmbh Coated implants, their manufacturing and use thereof
WO2005089829A2 (en) 2004-03-10 2005-09-29 Scil Technology Gmbh Coated implants, their manufacturing and use thereof
US7575751B2 (en) 2004-04-27 2009-08-18 Research Development Foundation Activin-A mutants
EP2298335A1 (en) 2004-05-25 2011-03-23 Stryker Corporation Use of morphogenic proteins for treating cartilage defects
US11364325B2 (en) 2004-10-14 2022-06-21 Biomimetic Therapeutics, Llc Platelet-derived growth factor compositions and methods of use thereof
EP2223698A1 (en) 2004-10-14 2010-09-01 Biomimetic Therapeutics, Inc. Platelet-derived growth factor compositions and method of use thereof
EP2308501A1 (en) 2004-10-14 2011-04-13 Biomimetic Therapeutics, Inc. Platelet-derived growth factor compositions and methods of use thereof
US9545377B2 (en) 2004-10-14 2017-01-17 Biomimetic Therapeutics, Llc Platelet-derived growth factor compositions and methods of use thereof
EP2308500A1 (en) 2004-10-14 2011-04-13 Biomimetic Therapeutics, Inc. Platelet-derived growth factor compositions and methods of use thereof
US10258566B2 (en) 2004-10-14 2019-04-16 Biomimetic Therapeutics, Llc Compositions and methods for treating bone
US11571497B2 (en) 2004-10-14 2023-02-07 Biomimetic Therapeutics, Llc Platelet-derived growth factor compositions and methods of use thereof
US11318230B2 (en) 2004-10-14 2022-05-03 Biomimetic Therapeutics, Llc Platelet-derived growth factor compositions and methods of use thereof
EP3170505A1 (en) 2004-10-14 2017-05-24 BioMimetic Therapeutics, LLC Platelet-derived growth factor compositions and method of use thereof
US7745583B2 (en) 2005-10-06 2010-06-29 Eli Lilly And Company Anti-myostatin antibodies
US7635760B2 (en) 2005-10-06 2009-12-22 Eli Lilly And Company Anti-myostatin antibodies
US8066995B2 (en) 2005-10-12 2011-11-29 Eli Lilly And Company Anti-myostatin antibodies
US7632499B2 (en) 2005-10-12 2009-12-15 Eli Lilly And Company Anti-myostatin antibodies
EP2992855A1 (en) 2005-12-06 2016-03-09 Etex Corporation Porous calcium phosphate bone material
WO2007067561A2 (en) 2005-12-06 2007-06-14 Etex Corporation Porous calcium phosphate bone material
EP2540310A1 (en) 2006-05-17 2013-01-02 Stryker Corporation Methods of treating cartilage defects using a soluble morphogenic protein complex
US9642891B2 (en) 2006-06-30 2017-05-09 Biomimetic Therapeutics, Llc Compositions and methods for treating rotator cuff injuries
US11058801B2 (en) 2006-06-30 2021-07-13 Biomimetic Therapeutics, Llc Compositions and methods for treating the vertebral column
US10456450B2 (en) 2006-06-30 2019-10-29 Biomimetic Therapeutics, Llc Compositions and methods for treating rotator cuff injuries
US9161967B2 (en) 2006-06-30 2015-10-20 Biomimetic Therapeutics, Llc Compositions and methods for treating the vertebral column
EP2428233A1 (en) 2006-08-17 2012-03-14 Warsaw Orthopedic, Inc. Medical implant sheets useful for tissue regeneration
US8063188B2 (en) 2006-09-05 2011-11-22 Eli Lilly And Company Anti-myostatin antibodies
US8895506B2 (en) 2006-12-14 2014-11-25 DePuy Synthes Products, LLC Protein stabilization formulations
US7956028B2 (en) 2006-12-14 2011-06-07 Johnson & Johnson Regenerative Therapeutics, Llc Protein stabilization formulations
US8435943B2 (en) 2006-12-14 2013-05-07 Advanced Technogies And Regenerative Medicine, Llc Protein stabilization formulations
WO2008082563A2 (en) 2006-12-21 2008-07-10 Stryker Corporation Sustained-release formulations comprising crystals, macromolecular gels, and particulate suspensions of biologic agents
US7964561B2 (en) 2007-06-29 2011-06-21 Advanced Technologies And Regenerative Medicine, Llc Protein formulations for use at elevated temperatures
US8058237B2 (en) 2007-08-07 2011-11-15 Advanced Technologies & Regenerative Medicine, LLC Stable composition of GDF-5 and method of storage
US8741840B2 (en) 2008-02-13 2014-06-03 Washington University BMP-7 for use in treating neointimal hyperplasia
US7947649B2 (en) 2008-04-14 2011-05-24 Advanced Technologies And Regenerative Medicine, Llc Liquid buffered GDF-5 formulations
US8870954B2 (en) 2008-09-09 2014-10-28 Biomimetic Therapeutics, Llc Platelet-derived growth factor compositions and methods for the treatment of tendon and ligament injuries
US11135341B2 (en) 2008-09-09 2021-10-05 Biomimetic Therapeutics, Llc Platelet-derived growth factor composition and methods for the treatment of tendon and ligament injuries
WO2010093941A2 (en) 2009-02-12 2010-08-19 Stryker Corporation COMPOSITIONS AND METHODS FOR MINIMALLY-INVASIVE SYSTEMIC DELIVERY OF PROTEINS INCLUDING TGF-β SUPERFAMILY MEMBERS
WO2010093925A2 (en) 2009-02-12 2010-08-19 Stryker Corporation PERIPHERAL ADMINISTRATION OF PROTEINS INCLUDING TGF-β SUPERFAMILY MEMBERS FOR TREATMENT OF SYSTEMIC DISORDERS AND DISEASE
WO2010110974A1 (en) 2009-03-24 2010-09-30 Stryker Corporation Methods and compositions for tissue engineering
WO2010115132A2 (en) 2009-04-03 2010-10-07 Warsaw Orthopedic, Inc. Medical implant with bioactive material and method of making the medical implant
WO2010144696A1 (en) 2009-06-11 2010-12-16 Burnham Institute For Medical Research Directed differentiation of stem cells
WO2011031856A1 (en) 2009-09-09 2011-03-17 Stryker Corporation Bmp -7 for use in treating pain induced by injuries and diseases of an articular joint
WO2011035094A1 (en) 2009-09-17 2011-03-24 Stryker Corporation Buffers for controlling the ph of bone morphogenetic proteins
WO2011087768A1 (en) 2009-12-22 2011-07-21 Stryker Corporation Bmp-7 variants with reduced immunogenicity
US11235030B2 (en) 2010-02-22 2022-02-01 Biomimetic Therapeutics, Llc Platelet-derived growth factor compositions and methods for the treatment of tendinopathies
US10034945B2 (en) 2012-07-13 2018-07-31 Trustees Of Tufts College Silk powder compaction for production of constructs with high mechanical strength and stiffness
US11925683B2 (en) 2014-11-06 2024-03-12 Scholar Rock, Inc. Compositions for making and using anti-Myostatin antibodies
US11135291B2 (en) 2014-11-06 2021-10-05 Scholar Rock, Inc. Methods for making and using anti-myostatin antibodies
WO2017015622A3 (en) * 2015-07-22 2017-03-02 Scholar Rock, Inc Gdf11 binding proteins and uses thereof
US10751413B2 (en) 2015-09-15 2020-08-25 Scholar Rock, Inc. Anti-pro/latent-Myostatin antibodies and uses thereof
US11439704B2 (en) 2015-09-15 2022-09-13 Scholar Rock, Inc. Methods for using anti-myostatin antibodies
EP3821880A1 (en) 2015-10-26 2021-05-19 President and Fellows of Harvard College Oxidized polysaccharides and methods of use thereof
US10882904B2 (en) 2016-01-08 2021-01-05 Scholar Rock, Inc. Methods for inhibiting myostatin activation by administering anti-pro/latent myostatin antibodies
US10946036B2 (en) 2016-06-13 2021-03-16 Scholar Rock, Inc. Use of myostatin inhibitors and combination therapies
US11155611B2 (en) 2017-01-06 2021-10-26 Scholar Rock, Inc. Compositions and methods for making and using anti-myostatin antibodies

Also Published As

Publication number Publication date
EP0776337B1 (en) 2005-09-21
EP1574577A3 (en) 2006-06-14
JPH10502811A (en) 1998-03-17
EP0776337A4 (en) 1998-05-06
DE69534468D1 (en) 2006-02-02
ES2251721T3 (en) 2006-05-01
US5914234A (en) 1999-06-22
DE69534468T2 (en) 2006-07-13
ATE305036T1 (en) 2005-10-15
EP0776337A1 (en) 1997-06-04
CA2194660C (en) 2009-09-29
CA2194660A1 (en) 1996-01-25
DK0776337T3 (en) 2005-12-12
EP1574577A2 (en) 2005-09-14
US20020150577A1 (en) 2002-10-17

Similar Documents

Publication Publication Date Title
EP0776337B1 (en) Growth differentiation factor-11
EP0690873B1 (en) Growth differentiation factor-8
US7074574B2 (en) Method of detecting growth differentiation factor-7 (GDF-7) using GDF-7 antibodies
US5808007A (en) Growth differentiation factor-3
WO1995010539A1 (en) Growth differentiation factor-10
WO1994015966A1 (en) Growth differentiation factor-9
AU7359394A (en) Growth differentiation factor-6
EP0770089B1 (en) Growth differentiation factor-12
US20030211541A1 (en) Growth differentiation factor-14
US20030109684A1 (en) Growth differentiation factor-3
US6130050A (en) Method for detection of growth differentiation factor-12 polypeptide
US20040067556A1 (en) Growth differentiation factor-6
US20020107369A1 (en) Growth differentiation factor-10

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): CA JP US

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): AT BE CH DE DK ES FR GB GR IE IT LU MC NL PT SE

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
WWE Wipo information: entry into national phase

Ref document number: 2194660

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: 1995925559

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 08765875

Country of ref document: US

WWP Wipo information: published in national office

Ref document number: 1995925559

Country of ref document: EP

WWG Wipo information: grant in national office

Ref document number: 1995925559

Country of ref document: EP