WO1999006559A1 - Procedes d'identification des recepteurs des facteurs de differenciation de la croissance - Google Patents

Procedes d'identification des recepteurs des facteurs de differenciation de la croissance Download PDF

Info

Publication number
WO1999006559A1
WO1999006559A1 PCT/US1998/015598 US9815598W WO9906559A1 WO 1999006559 A1 WO1999006559 A1 WO 1999006559A1 US 9815598 W US9815598 W US 9815598W WO 9906559 A1 WO9906559 A1 WO 9906559A1
Authority
WO
WIPO (PCT)
Prior art keywords
gdf
receptor
binding
antibody
receptors
Prior art date
Application number
PCT/US1998/015598
Other languages
English (en)
Inventor
Se-Jin Lee
Alexandra 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
Priority to US09/485,046 priority Critical patent/US6696260B1/en
Application filed by The Johns Hopkins University School Of Medicine filed Critical The Johns Hopkins University School Of Medicine
Priority to AU86663/98A priority patent/AU8666398A/en
Publication of WO1999006559A1 publication Critical patent/WO1999006559A1/fr
Priority to US09/841,730 priority patent/US6891082B2/en
Priority to US11/051,267 priority patent/US20050257278A1/en
Priority to US12/360,093 priority patent/US20090186806A1/en
Priority to US12/361,512 priority patent/US8124830B2/en
Priority to US12/861,738 priority patent/US8822411B2/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • C12N15/8509Vectors or expression systems specially adapted for eukaryotic hosts for animal cells for producing genetically modified animals, e.g. transgenic
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K67/00Rearing or breeding animals, not otherwise provided for; New or modified breeds of animals
    • A01K67/027New or modified breeds of vertebrates
    • A01K67/0275Genetically modified vertebrates, e.g. transgenic
    • A01K67/0276Knock-out vertebrates
    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/71Receptors; Cell surface antigens; Cell surface determinants for growth factors; for growth regulators
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/74Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving hormones or other non-cytokine intercellular protein regulatory factors such as growth factors, including receptors to hormones and growth factors
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/07Animals genetically altered by homologous recombination
    • A01K2217/075Animals genetically altered by homologous recombination inducing loss of function, i.e. knock out
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2227/00Animals characterised by species
    • A01K2227/10Mammal
    • A01K2227/105Murine
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/02Animal zootechnically ameliorated
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; AVICULTURE; APICULTURE; PISCICULTURE; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2267/00Animals characterised by purpose
    • A01K2267/03Animal model, e.g. for test or diseases
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/475Assays involving growth factors
    • G01N2333/495Transforming growth factor [TGF]
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2500/00Screening for compounds of potential therapeutic value

Definitions

  • This invention relates generally to ligand-receptor interactions and more specifically to growth differentiation factor receptor proteins and the ligands that bind to such receptors and methods of use therefor.
  • TGF- ⁇ transforming growth factor ⁇
  • 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.
  • MIS Mullerian inhibiting substance
  • Drosophila decapentaplegic (DPP) gene product, which is required for dorsal -ventral axis formation and morphogenesis of the imaginal disks (Padgett, et al, 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.
  • DPP Drosophila decapentaplegic
  • TGF- ⁇ s can influence a variety of differentiation processes, including adipogenesis, myogenesis, chondrogenesis, hematopolesis, 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 (Cheifetz, et al, Cell, 48:409, 1987), heterodimers have also been detected, and these appear to have different biological properties than the respective homodimers.
  • the present invention provides receptors for the growth differentiation factor (GDF) growth factor family. These receptors are useful for identifying antagonists and agonists for agricultural and human therapeutic purposes.
  • GDF growth differentiation factor
  • the invention provides a recombinant cell line that expresses growth differentiation factor-8 (GDF-8) or growth differentiation factor- 11 (GDF-11) receptor polypeptide. Also included are antibodies that bind to GDF receptors, polynucleotides encoding the receptors and the GDF receptor proteins themselves. Peptide fragments of GDF receptors, such as the GDF-8 or GDF-11 receptors, are also included. Such peptides may be useful in inhibiting binding of GDF-8 or GDF-11 to either its own receptor or another GDF-receptor (e.g., GDF-8 and -11 may bind the same receptor).
  • GDF-8 growth differentiation factor-8
  • GDF-11 growth differentiation factor- 11
  • the invention provides a substantially purified GDF-8-binding agent, wherein the binding agent inhibits GDF-8 binding to GDF-8 receptor.
  • the binding agent inhibits GDF-8 binding to GDF-8 receptor.
  • agents that inhibit GDF-11 binding are also included.
  • the invention provides a method for identifying a GDF receptor polypeptide including incubating components such as GDF polypeptide and a cell expressing a receptor or a soluble receptor under conditions sufficient to allow the GDF to bind to the receptor; measuring the binding of the GDF polypeptide to the receptor; and isolating the receptor.
  • the invention also includes a method for identifying a compound that binds to GDF receptor polypeptide including incubating components comprising the compound and GDFpolypeptide under conditions sufficient to allow the components to interact and measuring the binding or effect of binding of the compound to GDF receptor polypeptide.
  • the invention also provides non-human transgenic animals that have a phenotype characterized by expression of GDF-receptor polypeptide, the phenotype being conferred by a transgene contained in the somatic and germ cells of the animal, the transgene comprising a nucleic acid sequence which encodes GDF- receptor polypeptide. Methods of producing such transgenic animals are also included.
  • the invention includes a method for inhibiting the expression of
  • GDF-receptor in a cell including contacting GDF-receptor with an inhibiting effective amount of an antisense oligonucleotide that binds to a segment of an mRNA transcribed from a GDF-receptor gene, whereby the binding of the antisense to the mRNA segment inhibits GDF-receptor expression.
  • Figure la and lb are the nucleotide and amino acid sequence of murine GDF-8.
  • Figure lc and Id are the nucleotide and amino acid sequence of human GDF-8.
  • Figures 2a-2e are the nucleotide and amino acid sequence of baboon, bovine, chicken, rat, and turkey GDF-8.
  • Figures 3a and 3b are Northern blots showing expression of GDF-8 in muscle and in various species, respectively.
  • Figures 4a and 4b show the nucleotide and amino acid sequence of murine GDF-11 and expression of GDF-11, respectively.
  • Figure 5 shows an autoradiogram showing GDF-8.
  • Figures 6 and 7 show binding studies for GDF-8.
  • Figures 8-11 show 4 myoblast cell lines that do not bind GDF-8.
  • Figure 12 shows the construction of GDF-11 null mice by homologous targeting, a) is a map of the GDF-11 locus (top line) and targeting construct (second line). The black and stippled boxes represent coding sequences for the pro-and C-terminal regions, respectively.
  • the targeting construct contains a total of 11 kb of homology with the GDF-11 gene.
  • a probe derived from the region upstream of the 3' homology fragment and downstream of the first EcoRI site shown hybridizes to a 6.5 kb EcoRI fragment in the GDF-1 1 gene and a 4.8 kb fragment in a homologously targeted gene.
  • FIG. 13 shows kidney abnormalities in GDF-11 knockout mice. Kidneys of newborn animals were examined and classified according to the number of normal sized or small kidneys as shown at the top. Numbers in the table indicate number of animals falling into each classification according to genotype.
  • Figure 14 shows homeotic transformations in GDF-11 mutant mice, a) Newborn pups with missing (first and second from left) and normal looking tails, b-j) Skeleton preparations for newborn wild-type (b, e, h), heterozygous (c, f, I) and homozygous (d, g, j) mutant mice.
  • Whole skeleton preparations (b-d), vertebral columns (e-g), vertebrosternal ribs (h-j) showing transformations and defects in homozygous and heterozygous mutant mice. Numbers indicate thoracic segments.
  • Figure 15 is a table summarizing anterior transformations in wild-type, heterozygous and homozygous GDF-11 mice.
  • the invention provides an isolated polynucleotide sequence encoding the receptors of the invention.
  • isolated includes polynucleotides substantially free of other nucleic acids, proteins, lipids, carbohydrates or other materials with which it is naturally associated.
  • Polynucleotide sequences of the invention include DNA, cDNA and RNA sequences which encode GDF receptors. It is understood that all polynucleotides encoding all or a portion of GDF receptors are also included herein, as long as they encode a polypeptide with GDF receptors activity (e.g., bind to GDF).
  • Such polynucleotides include naturally occurring, synthetic, and intentionally manipulated polynucleotides.
  • portions of the mRNA sequence may be altered due to alternate RNA splicing patterns or the use of alternate promoters for RNA transcription.
  • GDF receptor polynucleotide may be subjected to site-directed mutagenesis.
  • the polynucleotide sequence for GDF receptors 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 receptors polypeptide encoded by the nucleotide sequence is functionally unchanged.
  • nucleotide sequences which encode GDF receptors polypeptide.
  • the polynucleotide encoding GDF receptors for GDFs such as GDF-8 or 11 (shown in the figures).
  • the sequence is RNA
  • the deoxyribonucleotides A, G, C, and T are replaced by ribonucleotides A, G, C, and U, respectively.
  • fragments (portions) 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 GDF receptor.
  • “Selective hybridization” as used herein refers to hybridization under moderately stringent or highly stringent physiological conditions (See, for example, the techniques described in Maniatis et al., 1989 Molecular Cloning A Laboratory Manual, Cold Spring Harbor Laboratory, N.Y., incorporated herein by reference), which distinguishes related from unrelated nucleotide sequences.
  • nucleic acid hybridization reactions the conditions used to achieve a particular level of stringency will vary, depending on the nature of the nucleic acids being hybridized. For example, the length, degree of complementarity, nucleotide sequence composition (e.g., GC v. AT content), and nucleic acid type (e.g., RNA v. DNA) of the hybridizing regions of the nucleic acids can be considered in selecting hybridization conditions. An additional consideration is whether one of the nucleic acids is immobilized, for example, on a filter.
  • An example of progressively higher stringency conditions is as follows: 2 x SSC/0.1% SDS at about room temperature (hybridization conditions); 0.2 x SSC/0.1 % SDS at about room temperature (low stringency conditions); 0.2 x SSC/0.1% SDS at about 42°C (moderate stringency conditions); and 0.1 x SSC at about 68 °C (high stringency conditions). Washing can be carried out using only one of these conditions, e.g., high stringency conditions, or each of the conditions can be used, e.g., for 10-15 minutes each, in the order listed above, repeating any or all of the steps listed. However, as mentioned above, optimal conditions will vary, depending on the particular hybridization reaction involved, and can be determined empirically.
  • SEQ ID NO:3 represents the wild-type sequence
  • SEQ ID NO: 1 represents a cDNA which encodes GDF receptors having a conservative substitution of Leucine for Alanine at amino acid residue 127. The result of this conservative variation should not affect biological activity of GDF receptors polypetide or peptides containing the variation (see Example 5).
  • DNA sequences of the invention can be obtained by several methods.
  • the DNA can be isolated using hybridization or computer-based 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) antibody screening of expression libraries to detect cloned DNA fragments with shared structural features; 3) polymerase chain reaction (PCR) on genomic DNA or cDNA using primers capable of annealing to the DNA sequence of interest; 4) computer searches of sequence databases for similar sequences; and 5) differential screening of a subtracted DNA library.
  • hybridization or computer-based 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) antibody screening of expression libraries to detect cloned DNA fragments with shared structural features; 3) polymerase chain reaction (PCR) on genomic DNA or cDNA using primers capable of annealing to the DNA sequence of
  • the GDF receptor polynucleotide of the invention is derived from avian, bovine, ovine, piscine, murine, human or porcine. 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).
  • a subtractive library as illustrated herein is useful for elimination of non-specific cDNA clones.
  • 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., ⁇ :2325, 1983).
  • a cDNA expression library such as lambda gtl 1
  • Such antibodies can be either polyclonally or monoclonally derived and used to detect expression product indicative of the presence of GDF receptors cDNA.
  • Alterations in GDF receptors nucleic acid include intragenic mutations (e.g., point mutation, nonsense (stop), missense, splice site and frameshift) and heterozygous or homozygous deletions. Detection of such alterations can be done by standard methods known to those of skill in the art including sequence analysis, Southern blot analysis, PCR based analyses (e.g., multiplex PCR, sequence tagged sites (STSs)) and in situ hybridization. Such proteins can be analyzed by standard SDS-PAGE and/or immuno- precipitation analysis and/or Western blot analysis, for example.
  • DNA sequences encoding GDF receptors 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 receptor 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 receptors 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., Tl, metallothionein I, or polyhedrin promoters).
  • Polynucleotide sequences encoding GDF receptors 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.
  • a variety of host-expression vector systems may be utilized to express the GDF receptors coding sequence. These include but are not limited to microorganisms such as bacteria transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing the GDF receptors coding sequence; yeast transformed with recombinant yeast expression vectors containing the GDF receptors coding sequence; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing the GDF receptors coding sequence; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing the GDF receptors coding sequence; or animal cell systems infected with recombinant virus expression vectors (e.g., retroviruses, adenovirus, vaccinia virus) containing the
  • any of a number of suitable transcription and translation elements including constitutive and inducible promoters, transcription enhancer elements, transcription terminators, etc. may be used in the expression vector (see e.g., Bitter et al., 1987, Methods in Enzymology j_53_:516-544).
  • inducible promoters such as pL of bacteriophage ⁇ , plac, ptrp, ptac (ptrp-lac hybrid promoter) and the like may be used.
  • promoters derived from the genome of mammalian cells e.g., metallothionein promoter
  • mammalian viruses e.g., the retrovirus long terminal repeat; the adenovirus late promoter; the vaccinia virus 7.5K promoter
  • Promoters produced by recombinant DNA or synthetic techniques may also be used to provide for transcription of the inserted GDF receptors coding sequence.
  • yeast a number of vectors containing constitutive or inducible promoters may be used.
  • Current Protocols in Molecular Biology Vol. 2, 1988, Ed. Ausubel et al, Greene Publish. Assoc. & Wiley Interscience, Ch. 13; Grant et al., 1987, Expression and Secretion Vectors for Yeast, in Methods in Enzymology, Eds. Wu & Grossman, 31987, Acad. Press, N.Y., Vol. 153, pp.516-544; Glover, 1986, DNA Cloning, Vol. II, IRL Press, Wash., D.C., Ch.
  • yeast promoter such as ADH or LEU2 or an inducible promoter such as GAL may be used (Cloning in Yeast, Ch. 3, R. Rothstein In: DNA Cloning Vol.l 1, A Practical Approach, Ed. DM Glover, 1986, IRL Press, Wash., D.C.).
  • vectors may be used which promote integration of foreign DNA sequences into the yeast chromosome.
  • Eukaryotic systems and preferably mammalian expression systems, allow for proper post-translational modifications of expressed mammalian proteins to occur.
  • Eukaryotic cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, phosphorylation, and advantageously, plasma membrane insertion of the gene product may be used as host cells for the expression of GDF receptors.
  • Mammalian cell systems which utilize recombinant viruses or viral elements to direct expression may be engineered.
  • the GDF receptors coding sequence may be ligated to an adenovirus transcription/- translation control complex, e.g., the late promoter and tripartite leader sequence.
  • the vaccinia virus 7.5K promoter may be used, (e.g., see, Mackett et al., 1982, Proc. Natl. Acad. Sci. USA 79: 7415-7419; Mackett et al, 1984, J. Virol. 49: 857- 864; Panicali et al, 1982, Proc. Natl. Acad. Sci. USA 79: 4927-4931).
  • vectors based on bovine papilloma virus which have the ability to replicate as extrachromosomal elements (Sarver, et al, 1981, Mol. Cell. Biol. 1 : 486).
  • the plasmid Shortly after entry of this DNA into mouse cells, the plasmid replicates to about 100 to 200 copies per cell. Transcription of the inserted cDNA does not require integration of the plasmid into the host's chromosome, thereby yielding a high level of expression.
  • These vectors can be used for stable expression by including a selectable marker in the plasmid, such as, for example, the neo gene.
  • the retroviral genome can be modified for use as a vector capable of introducing and directing the expression of the GDF receptors gene in host cells (Cone & Mulligan, 1984, Proc. Natl Acad. Sci. USA 81:6349-6353). High level expression may also be achieved using inducible promoters, including, but not limited to, the metallothionine IIA promoter and heat shock promoters.
  • host cells can be transformed with the GDF receptors cDNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • appropriate expression control elements e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines.
  • engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • a number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler,et al, 1977, Cell I k 223), hypoxanthine-guanine phospho- ribosyltransferase (Szybalska & Szybalski, 1962, Proc. Natl. Acad. Sci.
  • adenine phosphoribosyltransferase genes can be employed in tk-, hgprt " or aprt " cells respectively.
  • antimetabolite resistance can be used as the basis of selection for dhfr, which confers resistance to methotrexate (Wigler, et al, 1980, Natl. Acad. Sci. USA 77: 3567; O ⁇ are, et al, 1981, Proc. Natl. Acad. Sci. USA 78: 1527); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, 1981, Proc. Natl.
  • neo which confers resistance to the aminoglycoside G-418 (Colberre-Garapin, et al, 1981, J. Mol. Biol. 150: 1); and hygro, which confers resistance to hygromycin (Santerre, et al, 1984, Gene 30: 147) genes.
  • trpB which allows cells to utilize indole in place of tryptophan
  • hisD which allows cells to utilize histinol in place of histidine
  • ODC ornithine decarboxylase
  • Eukaryotic cells can also be cotransformed with DNA sequences encoding the GDF receptors 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
  • the present invention relates to stable recombinant cell lines, the cells of which express GDF receptor polypeptides and contain DNA that encodes GDF receptors.
  • Suitable cell types include but are not limited to cells of the following types: NIH 3T3 (Murine), C2C12, L6, and P19.
  • C2C12 and L6 myoblasts will differentiate spontaneously in culture and form myotubes depending on the particular growth conditions (Yaffe and Saxel, 1977; Yaffe, 1968).
  • PI 9 is an embryonal carcinoma cell line.
  • Such cells are described, for example, in the Cell Line Catalog of the American Type Culture Collection (ATCC). These cells can be stably transformed by a method known to the skilled artisan. See, for example, Ausubel et al.
  • GDF receptors can be expressed using inducible or constituitive regulatory elements for such expression.
  • constituitive or inducible promoters for example, are known in the art.
  • the desired protein encoding sequence and an operably linked promoter may be introduced into a recipient cell either as a non-replicating DNA (or RNA) molecule, which may either be a linear molecule or, more preferably, a closed covalent circular molecule. Since such molecules are incapable of autonomous replication, the expression of the desired molecule may occur through the transient expression of the introduced sequence. Alternatively, permanent expression may occur through the integration of the introduced sequence into the host chromosome. Therefore the cells can be transformed stably or transiently.
  • An example of a vector that may be employed is one which is capable of integrating the desired gene sequences into the host cell chromosome.
  • Cells which have stably integrated the introduced DNA into their chromosomes can be selected by also introducing one or more markers which allow for selection of host cells which contain the expression vector.
  • the marker may complement an auxotrophy in the host (such as leu2, or ura3, which are common yeast auxotrophic markers), biocide resistance, e.g., antibiotics, or heavy metals, such as copper, or the like.
  • the selectable marker gene can either be directly linked to the DNA gene sequences to be expressed, or introduced into the same cell by co-transfection.
  • the introduced sequence will be incorporated into a plasmid or viral vector capable of autonomous replication in the recipient host.
  • a plasmid or viral vector capable of autonomous replication in the recipient host.
  • Any of a wide variety of vectors may be employed for this purpose. Factors of importance in selecting a particular plasmid or viral vector include: the ease with which recipient cells that contain the vector may be recognized and selected from those recipient cells which do not contain the vector; the number of copies of the vector which are desired in a particular host; and whether it is desirable to be able to "shuttle" the vector between host cells of different species.
  • vectors For a mammalian host, several possible vector systems are available for expression.
  • One class of vectors utilize DNA elements which provide autonomously replicating extra-chromosomal plasmids, derived from animal viruses such as bovine papilloma virus, polyoma virus, adenovirus, or SV40 virus.
  • a second class of vectors include vaccinia virus expression vectors.
  • a third class of vectors relies upon the integration of the desired gene sequences into the host chromosome. Cells which have stably integrated the introduced DNA into their chromosomes may be selected by also introducing one or more markers (e.g., an exogenous gene) which allow selection of host cells which contain the expression vector.
  • the marker may provide for prototropy to an auxotrophic host, biocide resistance, e.g., antibiotics, or heavy metals, such as copper or the like.
  • the selectable marker gene can either be directly linked to the DNA sequences to be expressed, or introduced into the same cell by co-transformation. Additional elements may also be needed for optimal synthesis of mRNA. These elements may include splice signals, as well as transcription promoters, enhancers, and termination signals.
  • the cDNA expression vectors incorporating such elements include those described by Okayama, H, Mol. Cell. Biol, 3:280 (1983), and others.
  • the DNA construct may be introduced (transformed) into an appropriate host.
  • Various techniques may be employed, such as protoplast fusion, calcium phosphate precipitation, electroporation or other conventional techniques.
  • the present invention relates to transgenic animals having cells that express GDF receptors.
  • Such transgenic animals may have decreased fat content and increased muscle mass.
  • the subject invention provides non-human transgenic animals which are useful as a source of food products with high muscle and protein content, and reduced fat and cholesterol content.
  • the animals have been altered chromosomally in their germ cells and somatic cells so that the production of GDF-8 may be at "normal" levels, however, the GDF-8 receptor is produced in reduced amounts, or is completely disrupted, resulting in animals with decreased binding of GDF-8 and higher than normal levels of muscle tissue, preferably without increased fat and/or cholesterol levels.
  • the present invention also includes food products provided by the animals. Such food products have increased nutritional value because of the increase in muscle tissue.
  • the transgenic non-human animals of the invention include bovine, porcine, ovine and avian animals, for example.
  • the subject invention also provides a method of producing animal food products having increased muscle content.
  • the method includes modifying the genetic makeup of the germ cells of apronuclear embryo of the animal, implanting the embryo into the oviduct of a pseudopregnant female thereby allowing the embryo to mature to full term progeny, testing the progeny for presence of the transgene to identify transgene-positive progeny, cross-breeding transgene-positive progeny to obtain further transgene-positive progeny and processing the progeny to obtain foodstuff.
  • the modification of the germ cell comprises altering the genetic composition so as to disrupt or reduce the expression of the naturally occurring gene encoding for production of GDF-8 receptor protein.
  • the transgene comprises antisense polynucleotide sequences to the GDF-8 receptor protein.
  • the transgene may comprise a non-functional sequence which replaces or intervenes in the native GDF-8 receptor gene or the transgene may encode a GDF-8 receptor antagonist.
  • the subject invention also provides a method of producing avian food products having improved muscle content.
  • the method includes modifying the genetic makeup of the germ cells of a pronuclear embryo of the avian animal, implanting the embryo into the oviduct of a pseudopregnant female into an embryo of a chicken, culturing the embryo under conditions whereby progeny are hatched, testing the progeny for presence of the genetic alteration to identify transgene-positive progeny, cross-breeding transgene- positive progeny and processing the progeny to obtain foodstuff.
  • animal here denotes all mammalian species except human. It also includes an individual animal in all stages of development, including embryonic and fetal stages. Farm animals (pigs, goats, sheep, cows, horses, rabbits and the like), rodents (such as mice), and domestic pets (for example, cats and dogs) are included within the scope of the present invention.
  • transgenic animal is any animal containing cells that bear genetic information received, directly or indirectly, by deliberate genetic manipulation at the subcellular level, such as by microinjection or infection with recombinant virus. "Transgenic” in the present context does not encompass classical crossbreeding or in vitro fertilization, but rather denotes animals in which one or more cells receive a recombinant DNA molecule.
  • transgenic animal also includes a "germ cell line” transgenic animal.
  • a germ cell line transgenic animal is a transgenic animal in which the genetic information has been taken up and incorporated into a germ line cell, therefore conferring the ability to transfer the information to offspring. If such offspring in fact possess some or all of that information, then they, too, are transgenic animals.
  • the cDNA that encodes GDF receptors can be fused in proper reading frame under the transcriptional and translational control of a vector to produce a genetic construct that is then amplified, for example, by preparation in a bacterial vector, according to conventional methods. See, for example, the standard work: Sambrook et al., MOLECULAR CLONING: A LABORATORY MANUAL (Cold Spring Harbor Press 1989), the contents of which are incorporated by reference. The amplified construct is thereafter excised from the vector and purified for use in producing transgenic animals.
  • transgenic as used herein additionally includes any organism whose genome has been altered by in vitro manipulation of the early embryo or fertilized egg or by any transgenic technology to induce a specific gene knockout.
  • gene knockout refers to the targeted disruption of a gene in vivo with complete loss of function that has been achieved by any transgenic technology familiar to those in the art.
  • transgenic animals having gene knockouts are those in which the target gene has been rendered nonfunctional by an insertion targeted to the gene to be rendered non-functional by homologous recombination.
  • transgenic includes any transgenic technology familiar to those in the art which can produce an organism carrying an introduced transgene or one in which an endogenous gene has been rendered non-functional or "knocked out.”
  • the transgene to be used in the practice of the subject invention may be a DNA sequence comprising a modified GDF receptors coding sequence.
  • the transgene to be used in the practice of the subject invention may be a DNA sequence comprising a modified GDF receptors coding sequence.
  • the GDF receptor gene is disrupted by homologous targeting in embryonic stem cells.
  • the entire mature C-terminal region of the GDF receptors gene may be deleted as described in the examples below.
  • the GDF receptors disruption or deletion may be accompanied by insertion of or replacement with other DNA sequences, such as a non-functional GDF receptors sequence.
  • the transgene comprises DNA antisense to the coding sequence for GDF receptors.
  • the transgene comprises DNA encoding an antibody or receptor peptide sequence which is able to bind to GDF receptors.
  • DNA sequences that encode proteins having GDF receptors activity but differ in nucleic acid sequence due to the degeneracy of the genetic code may also be used herein, as may truncated forms, allelic variants and interspecies homologues.
  • the present invention relates to antibodies that bind GDF receptors that block GDF binding to the receptor.
  • such antibodies may be useful for ameliorating disorders associated with muscle tissue.
  • a monoclonal antibody which binds to GDF-8 receptor may have the effect of increasing the development of skeletal muscles.
  • the GDF-8 receptor monoclonal antibody, polypeptide, or polynucleotide is administered to a patient suffering from a disorder selected from the group consisting of muscle wasting disease, neuromuscular disorder, muscle atrophy or aging.
  • the GDF-8 receptor antibody may also be administered to a patient suffering from a disorder selected from the group consisting of muscular dystrophy, spinal cord injury, traumatic injury, congestive obstructive pulmonary disease (COPD), AIDS or cachechia.
  • COPD congestive obstructive pulmonary disease
  • the GDF-8 antibody is administered to a patient with muscle wasting disease or disorder by intravenous, intramuscular or subcutaneous injection; preferably, a monoclonal antibody is administered within a dose range between about 0.1 mg/kg to about 100 mg/kg; more preferably between about 1 ug/kg to 75 mg/kg; most preferably from about 10 mg/kg to 50 mg/kg.
  • the antibody may be administered, for example, by bolus injunction or by slow infusion. Slow infusion over a period of 30 minutes to 2 hours is preferred.
  • the GDF-8 antibody may be formulated in a formulation suitable for administration to a patient. Such formulations are known in the art.
  • the dosage regimen will be determined by the attending physician considering various factors which modify the action of the GDF-8 receptor protein, e.g. amount of tissue desired to be formed, the site of tissue damage, the condition of the damaged tissue, the size of a wound, type of damaged tissue, the patient's age, sex, and diet, the severity of any infection, time of administration and other clinical factors.
  • the dosage may vary with the type of matrix used in the reconstitution and the types of agent, such as anti- GDF-8 receptor antibodies, to be used in the composition.
  • systemic or injectable administration such as intravenous (IV), intramuscular (IM) or subcutaneous (Sub-Q) injection.
  • Administration will generally be initiated at a dose which is minimally effective, and the dose will be increased over a preselected time course until a positive effect is observed. Subsequently, incremental increases in dosage will be made limiting such incremental increases to such levels that produce a corresponding increase in effect, while taking into account any adverse affects that may appear.
  • growth factors such as IGF I (insulin like growth factor I), human, bovine, or chicken growth hormone which may aid in increasing muscle mass, to the final composition, may also affect the dosage.
  • the anti-GDF-8 antibody is generally administered within a dose range of about 0.1 ug/kg to about 100 mg/kg.; more preferably between about 10 mg/kg to 50 mg/kg.
  • polyclonal antibodies The preparation of polyclonal antibodies is well-known to those skilled in the art. See, for example, Green et al , Production of Polyclonal Antisera, in IMMUNOCHEMICAL PROTOCOLS (Manson, ed.), pages 1-5 (Humana Press 1992); Coligan et al, Production of Polyclonal Antisera in Rabbits, Rats, Mice and Hamsters, in CURRENT PROTOCOLS N IMMUNOLOGY, section 2.4.1 (1992), which are hereby incorporated by reference. The preparation of monoclonal antibodies likewise is conventional.
  • monoclonal antibodies can be obtained by injecting mice with a composition comprising an antigen, verifying the presence of antibody production by removing a serum sample, removing the spleen to obtain B lymphocytes, fusing the B lymphocytes with myeloma cells to produce hybridomas, cloning the hybridomas, selecting positive clones that produce antibodies to the antigen, and isolating the antibodies from the hybridoma cultures.
  • Monoclonal antibodies can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation techniques include affinity chromatography with Protein-A Sepharose, size-exclusion chromatography, and ion- exchange chromatography.
  • Multiplication in vitro may be carried out in suitable culture media such as Dulbecco's Modified Eagle Medium or RPMI 1640 medium, optionally replenished by a mammalian serum such as fetal calf serum or trace elements and growth-sustaining supplements such as normal mouse peritoneal exudate cells, spleen cells, bone marrow macrophages.
  • suitable culture media such as Dulbecco's Modified Eagle Medium or RPMI 1640 medium
  • a mammalian serum such as fetal calf serum or trace elements
  • growth-sustaining supplements such as normal mouse peritoneal exudate cells, spleen cells, bone marrow macrophages.
  • Production in vitro provides relatively pure antibody preparations and allows scale-up to yield large amounts of the desired antibodies.
  • Large scale hybridoma cultivation can be carried out by homogenous suspension culture in an airlift reactor, in a continuous stirrer reactor, or in immobilized or entrapped cell culture.
  • Multiplication in vivo may be carried out by injecting cell clones into mammals histocompatible with the parent cells, e.g. , osyngeneic mice, to cause growth of antibody- producing tumors.
  • the animals are primed with a hydrocarbon, especially oils such as pristane (tetramethylpentadecane) prior to injection.
  • pristane tetramethylpentadecane
  • the desired monoclonal antibody is recovered from the body fluid of the animal.
  • Therapeutic applications for antibodies disclosed herein are also part of the present invention.
  • antibodies of the present invention may also be derived from subhuman primate antibody.
  • a therapeutically useful anti-GDF receptors antibody may be derived from a "humanized" monoclonal antibody.
  • Humanized monoclonal antibodies are produced by transferring mouse complementarity determining regions from heavy and light variable chains of the mouse immunoglobulin into a human variable domain, and then substituting human residues in the framework regions of the murine counterparts.
  • the use of antibody components derived from humanized monoclonal antibodies obviates potential problems associated with the immunogenicity of murine constant regions.
  • General techniques for cloning murine immunoglobulin variable domains are described, for example, by Orlandi et al, Proc. Nat'l Acad. Sci. USA 86:3833 (1989), which is hereby incorporated in its entirety by reference.
  • Antibodies of the invention also may be derived from human antibody fragments isolated from a combinatorial immunoglobulin library. See, for example, Barbas et al, METHODS: A COMPANION TO METHODS IN ENZYMOLOGY, VOL. 2, page 119 (1991); Winter etal, Ann. Rev. Immunol 12: 433 (1994), which are hereby incorporated by reference.
  • Cloning and expression vectors that are useful for producing a human immunoglobulin phage library can be obtained, for example, from STRATAGENE Cloning Systems (La Jolla, CA).
  • antibodies of the present invention may be derived from a human monoclonal antibody.
  • Such antibodies are obtained from transgenic mice that have been "engineered” to produce specific human antibodies in response to antigenic challenge.
  • elements of the human heavy and light chain loci are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy and light chain loci.
  • the transgenic mice can synthesize human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas.
  • Methods for obtaining human antibodies from transgenic mice are described by Green et al, Nature Genet. 7:13 (1994); Lonberg et al, Nature 368:856 (1994); and Taylor et al, Int. Immunol. 6:579 (1994), which are hereby incorporated by reference.
  • Antibody fragments of the present invention can be prepared by proteolytic hydrolysis of the antibody or by expression in E. coli of DNA encoding the fragment.
  • Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods.
  • antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab') 2 .
  • This fragment can be further cleaved using a thiol reducing agent, and optionally a blocking group for the sulfhydryl groups resulting from cleavage of disulfide linkages, to produce 3.5S Fab' monovalent fragments.
  • cleaving antibodies such as separation of heavy chains to form monovalent light-heavy chain fragments, further cleavage of fragments, or other enzymatic, chemical, or genetic techniques may also be used, so long as the fragments bind to the antigen that is recognized by the intact antibody.
  • Fv fragments comprise an association of V H and Y chains. This association may be noncovalent, as described in Inbar et al, Proc. Nat'l Acad. Sci. USA 69:2659 (1972).
  • the variable chains can be linked by an intermolecular disulfide bond or cross-linked by chemicals such as glutaraldehyde. See, e.g., Sandhu, supra.
  • the Fv fragments comprise V H and V L chains connected by a peptide linker.
  • These single-chain antigen binding proteins (sFv) are prepared by constructing a structural gene comprising DNA sequences encoding the V H and V L domains connected by an oligonucleotide.
  • the structural gene is inserted into an expression vector, which is subsequently introduced into a host cell such as E. coli.
  • the recombinant host cells synthesize a single polypeptide chain with a linker peptide bridging the two V domains.
  • Methods for producing sFvs are described, for example, by Whitlow et al, METHODS: A COMPANION TO METHODS IN ENZYMOLOGY, VOL. 2, page 97 (1991); Bird etal, Science 242:423-426 (1988); Ladner et al, U.S. patent No. 4,946,778; Pack et al, Bio/Technology 11 : 1271-77 (1993); and Sandhu, supra.
  • CDR peptides (“minimal recognition units") can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody-producing cells. See, for example, Larrick et al, METHODS: A COMPANION TO METHODS IN ENZYMOLOGY, VOL. 2, page 106 (1991).
  • the invention provides a method for identifying a GDF receptor polypeptide comprising incubating components comprising GDF polypeptide and a cell expressing a receptor or a soluble receptor under conditions sufficient to allow the GDF to bind to the receptor; measuring the binding of the GDF polypeptide to the receptor; and isolating the receptor.
  • the GDF may be any of the known GDFs (e.g., GDF-1-16), and preferably is GDF-8 or GDF-11. Methods of isolating the receptors are described in more detail in the Examples section below.
  • GDF receptors variant means a molecule that simulates at least part of the structure of GDF receptors. GDF receptor variants may also be useful in preventing GDF binding, thereby ameliorating symptoms of disorders described above.
  • the present invention relates to peptides and peptide derivatives that have fewer amino acid residues than GDF receptors.
  • Such peptides and peptide derivatives could represent research and diagnostic tools in the study of muscle wasting diseases and the development of more effective therapeutics.
  • the invention relates not only to peptides and peptide derivatives of naturally-occurring GDF receptors, but also to GDF receptor mutants and chemically synthesized derivatives of GDF receptors that bind GDFs.
  • changes in the amino acid sequence of GDF receptors are contemplated in the present invention.
  • GDF receptors can be altered by changing the DNA encoding the protein. Preferably, only conservative amino acid alterations are undertaken, using amino acids that have the same or similar properties.
  • Illustrative amino acid substitutions include the changes of: alanine to serine; arginine to lysine; asparagine to glutamine or histidine; aspartate to glutamate; cysteine to serine; glutamine to asparagine; glutamate to aspartate; glycine to proline; histidine to asparagine or glutamine; isoleucine to leucine or valine; leucine to valine or isoleucine; lysine to arginine, glutamine, or glutamate; methionine to leucine or isoleucine; phenylalanine to tyrosine, leucine or methionine; serine to threonine; threonine to serine; tryptophan to tyrosine; tyrosine to tryptophan or phenylalanine; valine to isoleucine or leucine.
  • Variants useful for the present invention comprise analogs, homologs, muteins and mimetics of GDF receptors that retain the ability to bind to their respective GDFs.
  • Peptides of the GDF receptors refer to portions of the amino acid sequence of GDF receptors that also retain this ability.
  • the variants can be generated directly from GDF receptors itself by chemical modification, by proteolytic enzyme digestion, or by combinations thereof. Additionally, genetic engineering techniques, as well as methods of synthesizing polypeptides directly from amino acid residues, can be employed.
  • Peptides of the invention can be synthesized by such commonly used methods as t-BOC or FMOC protection of alpha-amino groups. Both methods involve stepwise syntheses whereby a single amino acid is added at each step starting from the C terminus of the peptide (See, Coligan, et al, Current Protocols in Immunology, Wiley Interscience, 1991, Unit 9). Peptides of the invention can also be synthesized by the well known solid phase peptide synthesis methods described Merrifield, J. Am. Chem. Soc, 85:2149.
  • This can normally be purified by such techniques as gel filtration on Sephadex G-15 using 5% acetic acid as a solvent. Lyophilization of appropriate fractions of the column will yield the homogeneous peptide or peptide derivatives, which can then be characterized by such standard techniques as amino acid analysis, thin layer chromatography, high performance liquid chromatography, ultraviolet absorption spectroscopy, molar rotation, solubility, and quantitated by the solid phase Edman degradation.
  • peptides can be produced by recombinant methods as described below.
  • substantially purified refers to a molecule, such as a peptide that is substantially free of other proteins, lipids, carbohydrates, nucleic acids, and other biological materials with which it is naturally associated.
  • a substantially pure molecule such as a polypeptide, can be at least 60%, by dry weight, the molecule of interest.
  • GDF receptors peptides can be purified using standard protein purification methods and the purity of the polypeptides can be determined using standard methods including, e.g., polyacrylamide gel electrophoresis (e.g., SDS-PAGE), column chromatography (e.g., high performance liquid chromatography (HPLC)), and amino-terminal amino acid sequence analysis.
  • polyacrylamide gel electrophoresis e.g., SDS-PAGE
  • column chromatography e.g., high performance liquid chromatography (HPLC)
  • amino-terminal amino acid sequence analysis e.g., amino-terminal amino acid sequence analysis.
  • Non-peptide compounds that mimic the binding and function of GDF receptors can be produced by the approach outlined in Saragovi et al, Science 253: 792-95 (1991).
  • Mimetics are molecules which mimic elements of protein secondary structure. See, for example, Johnson et ⁇ /,”Peptide Turn Mimetics,” in BIOTECHNOLOGY AND PHARMACY, Pezzuto et al, Eds, (Chapman and Hall, New York 1993).
  • the underlying rationale behind the use of peptide mimetics is that the peptide backbone of proteins exists chiefly to orient amino acid side chains in such a way as to facilitate molecular interactions.
  • appropriate mimetics can be considered to be the equivalent of GDF receptors itself.
  • Longer peptides can be produced by the "native chemical" ligation technique which links together peptides (Dawson, et al, Science, 266:776. 1994). Variants can be created by recombinant techniques employing genomic or cDNA cloning methods. Site-specific and region-directed mutagenesis techniques can be employed. See CURRENT PROTOCOLS IN MOLECULAR BIOLOGY vol. 1, ch. 8 (Ausubel et al eds, J. Wiley & Sons 1989 & Supp. 1990-93); PROTEIN ENGINEERING (Oxender & Fox eds, A. Liss, Inc. 1987).
  • linker-scanning and PCR-mediated techniques can be employed for mutagenesis. See PCR TECHNOLOGY (Erlich ed, Stockton Press 1989); CURRENT PROTOCOLS IN MOLECULAR BIOLOGY, vols. 1 & 2, supra. Protein sequencing, structure and modeling approaches for use with any of the above techniques are disclosed in PROTEIN ENGINEERING, loc. cit, and CURRENT PROTOCOLS PN MOLECULAR BIOLOGY, vols. 1 & 2, supra.
  • the present invention relates to GDF receptor-binding agents that block binding of GDFs to their receptors.
  • GDF receptor-binding agents may represent effective therapeutics.
  • GDF receptor-binding agent denotes a naturally occurring ligand of GDF receptors such as, for example: GDF-1 -16; a synthetic ligand of GDF receptors, or appropriate derivatives of the natural or synthetic ligands. The determination and isolation of ligands is well described in the art. See, e.g., Lerner, Trends NeuroSci. 17:142-146 (1994) which is hereby incorporated in its entirety by reference.
  • the present invention relates to GDF receptor-binding agents that interfere with binding between GDF receptor and a GDF.
  • binding agents may interfere by competitive inhibition, by non-competitive inhibition or by uncompetitive inhibition. Interference with normal binding between GDF receptors and one or more GDF can result in a useful pharmacological effect.
  • the invention provides a method for identifying a composition which binds to GDF receptors.
  • the method includes incubating components comprising the composition and GDF receptors under conditions sufficient to allow the components to interact and measuring the binding of the composition to GDF receptors.
  • Compositions that bind to GDF receptors include peptides, peptidomimetics, polypeptides, chemical compounds and biologic agents as described above.
  • Incubating includes conditions which allow contact between the test composition and GDF receptors.
  • Contacting includes in solution and in solid phase.
  • the test ligand(s)/composition may optionally be a combinatorial library for screening a plurality of compositions.
  • compositions identified in the method of the invention can be further evaluated, detected, cloned, sequenced, and the like, either in solution or after binding to a solid support, by any method usually applied to the detection of a specific DNA sequence such as PCR, oligomer restriction (Saiki, et al, Bio/Technology, 3_: 1008-1012, 1985), allele-specific oligonucleotide (ASO) probe analysis (Conner, et al, Proc. Natl. Acad. Sci. USA, 80:278, 1983), oligonucleotide ligation assays (OLAs) (Landegren, et al, Science, 241: 1077, 1988), and the like. Molecular techniques for DNA analysis have been reviewed (Landegren, et al, Science, 242:229-237. 1988).
  • compositions can functionally complex with the receptor protein
  • induction of the exogenous gene is monitored by monitoring changes in the protein levels of the protein encoded for by the exogenous gene, for example.
  • this composition(s) can bind to the receptor protein coded for by the nucleic acid encoding the initial sample test composition(s).
  • exogenous gene can be monitored by a functional assay or assay for a protein product, for example.
  • the exogenous gene is therefore a gene which will provide an assayable/measurable expression product in order to allow detection of expression of the exogenous gene.
  • exogenous genes include, but are not limited to, reporter genes such as chloramphenicol acetyltransferase gene, an alkaline phosphatase gene, beta-galactosidase,a luciferase gene, a green fluorescent protein gene, guanine xanthine phosphoribosyltransferase, alkaline phosphatase, and antibiotic resistance genes (e.g., neomycin phosphotransferase).
  • reporter genes such as chloramphenicol acetyltransferase gene, an alkaline phosphatase gene, beta-galactosidase,a luciferase gene, a green fluorescent protein gene, guanine x
  • compositions of the present invention can be extracted and purified from the culture media or a cell by using known protein purification techniques commonly employed, such as extraction, precipitation, ion exchange chromatography, affinity chromatography, gel filtration and the like.
  • Compositions can be isolated by affinity chromatography using the modified receptor protein extracellular domain bound to a column matrix or by heparin chromatography.
  • screening method of the invention is combinatorial chemistry methods for identifying chemical compounds that bind to GDF receptors.
  • the screening method is also useful for identifying variants, binding or blocking agents, etc, which functionally, if not physically (e.g., sterically) act as antagonists or agonists, as desired.
  • the purified GDF-8 and GDF-11 proteins will be used primarily to assay for biological activities. In order to identify potential target cells for GDF-8 and GDF-11 action cells expressing their receptors will be searched. For this purpose, the purified protein will be radioiodinated using the chloramine T method, which has been used successfully to label other members of this superfamily, like TGF- ⁇ (Cheifetz et al, 1987), activins (Sugino et al, 1988), and BMPs (Paralkar et al, 1991), for receptor-binding studies. The mature processed forms of GDF-8 and GDF-11 each contain multiple tyrosine residues. Two different approaches will then be taken to attempt to identify receptors for these proteins.
  • One approach will be taken to determine the number, affinity, and distribution of receptors. Either whole cells grown in culture, frozen sections of embryos or adult tissues, or total membrane fractions prepared from tissues or cultured cells will be incubated with the labeled protein, and the amount or distribution of bound protein will be determined. For experiments involving cell lines or membranes, the amount of binding will be determined by measuring either the amount of radioactivity bound to cells on the dish after several washes or, in the case of membranes, the amount of radioactivity sedimented with the membranes after centrifugation or retained with the membranes on a filter. For experiments involving primary cultures, where the number of cells may be more limited, binding sites will be visualized directly by overlaying with photographic emulsion.
  • a second approach will also be taken to begin to characterize the receptor biochemically.
  • Membrane preparations or potential target cells grown in culture will be incubated with labeled ligand, and receptor/ligand complexes will be covalently cross-linked using disuccinimidyl suberate, which has been commonly used to identify receptors for a variety of ligands, including members of the TGF- ⁇ superfamily (for example, see Massague and Like, 1985).
  • Cross-linked complexes will then be electrophoresed on SDS polyacrylamide gels to look for bands labeled in the absence but not in the presence of excess unlabeled protein.
  • the molecular weight of the putative receptor will be estimated by subtracting the molecular weight of the ligand.
  • An important question that these experiments will address is whether GDF-8 and GDF-11 signal through type I and type II receptors like many other members of the TGF- ⁇ superfamily (for review, see Massague, 1996).
  • binding studies on these primary cells after various days in culture will be performed and binding sites localized by autoradiography so that the binding sites can be co-localized with various myogenic markers, such as muscle myosin (Vivarelli et al, 1988), and correlate binding with the differentiation state of the cells, such as formation of multinucleated myotubes.
  • myogenic markers such as muscle myosin (Vivarelli et al, 1988)
  • cell lines will be utilized to look for receptors.
  • the initial focus will be on three cells lines, C2C12, L6, and PI 9.
  • C2C12 and L6 myoblasts will differentiate spontaneously in culture and form myotubes depending on the particular growth conditions (Yaffe and Saxel, 1977; Yaffe, 1968).
  • PI 9 embryonal carcinoma cells can be induced to differentiate into various cell types, including skeletal muscle cells in the presence of DMSO (Rudnicki and McBurney, 1987). Receptor binding studies will be carried out on these cell lines under various growth conditions and at various stages of differentiation.
  • the dissociation constant (K d ) is 370 pM and L6 myoblasts have a high number (5,000 receptors/cell) of cell surface binding proteins (Figure 6).
  • GDF-11 also called BMP-11
  • BMP-11 BMP-11
  • Receptor binding studies were performed to determine if GDF-11 also binds the GDF-8 receptor.
  • Figure 6 shows that GDF-8 and GDF-11 do bind to the same binding proteins on L6 myoblasts. It is important to establish whether or not GDF-8 binds to the known TGF- ⁇ receptor. As shown in Figure 6, TGF- ⁇ does not compete the binding of GDF-8, indicating that the GDF-8 receptor is distinct from the TGF- ⁇ receptor.
  • the GDF-8 receptor is not expressed on all myoblast cell lines.
  • Figures 8-11 are examples of four myoblast cell lines (C2C12, G7, MLB13MYC cl4 and BC3H1) which do not bind GDF-8.
  • the first approach will be to use an expression cloning strategy. In fact, this was the strategy that was orginally used by Mathews and Vale (1991) and Lin et al. (1992) to clone the first activin and TGF- ⁇ receptors.
  • We will begin by preparing poly A-selected RNA from the tissue or cell type that expresses the highest relative number of high affinity binding sites.
  • We will then use this RNA to prepare a cDNA library in the mammalian expression vector pcDNA-1. This vector contains a CMV promoter and an SV40 origin of replication.
  • the library will be plated, and cells from each plate will be pooled into broth and frozen. Aliquots from each pool will then be grown for preparation of DNA.
  • Each individual pool will be transiently transfected into COS cells in chamber slides, and transfected cells will be incubated with iodinated GDF-8 or GDF-11. After washing away the unbound protein, the sites of ligand binding will be visualized by autoradiography. Once a positive pool is identified, the cells from that pool will be replated at lower density, and the process will be repeated. Positive pools will then be plated, and individual colonies will be picked into grids and re-analyzed as described (Wong et al, 1985).
  • TGF- ⁇ and a cloned type II receptor The coding sequence for the TGF- ⁇ type II receptor will be cloned into the pcDNA-1 vector, and bacteria transformed with this construct will be mixed with bacteria from our library at various ratios, including 1 : 1500.
  • the general strategy will be to design degenerate primers corresponding to conserved regions of the known receptors, to use these primers for PCR on cDNA prepared from the appropriate RNA samples (most likely from skeletal muscle), to subclone the PCR products, and finally to sequence individual subclones. As sequences are identified, they will be used as hybridization probes to eliminate duplicate clones from further analysis. We will then test the receptors that we identify for their ability to bind purified GDF-8 and GDF-11.
  • this screen will yield only small PCR products, we will obtain full-length cDNA clones for each receptor from cDNA libraries prepared from the appropriate tissue, insert these cDNA clones into the pcDNA-1 vector, transfect these constructs into COS cells, and assay the transfected cells for their ability to bind iodinated GDF-8 or GDF-11. Ideally, we would like to test every receptor that we identify in this screen for their ability to bind these ligands. However, the number of receptors that we identify may be large, and isolating all of the full-length cDNAs and testing them may require considerable effort.
  • GDF-8 and GDF-11 receptors are further complicated by the fact at least one member of the TGF- ⁇ superfamily, namely, GDNF, is capable of signalling through a completely different type of receptor complex involving a GPI-linked component (GDNFR-alpha) and a receptor tyrosine kinase (c-ret) (Trupp et al, 1996; Durbec et al, 1996; Treanor et al, 1996; Jing et al, 1996).
  • GDNFR-alpha GPI-linked component
  • c-ret receptor tyrosine kinase
  • GDF-8 and GDF-11 do signal through a similar receptor complex
  • our expression screening approach should be able to identify at least the GPI-linked component (indeed GDNFR-alpha was identified using an expression screening approach) of this complex.
  • identifying the analogous receptor tyrosine kinase would probably require a substantial amount of additional work, such as biochemical purification of the complex.
  • the similar phenotypes of GDNF- and c-ret-deficient mice suggested c-ret as a potential receptor for GDNF.
  • the phenotype of GDF-11 knockout mice in several respects resembles the phenotype of mice carrying a deletion of a receptor for some members of the TGF- ⁇ superfamily, the activintype HB receptor (ActRIIB).
  • ActRIIB activintype HB receptor
  • RI ES cells were transfected with the targeting construct, selected with gancyclovir (2 ⁇ M) and G418 (250 ⁇ g/ml), and analyzed by Southern analysis. Homologous targeting of the GDF-11 gene was seen in 8/155 g- ancyclovir/G418 doubly resistant ES cell clones. Following injection of several targeted clones into C57BL/6J blastocysts, we obtained chimeras from one ES clone that produced heterozygous pups when crossed to both C57BL/6J and 129/SvJ females.
  • Crosses of C57BL/6J/129/SvJ hybrid FI heterozygotes produced 49 wild-type (34%), 94 heterozygous (66%) and no homozygous mutant adult offspring. Similarly, there were no adult homozygous null animals seen in the 129/SvJ background (32 wild-type (36%>) and 56 heterozygous mutant (64%) animals).
  • thoracic segments T8, T9, T10, and in some cases even Tl 1, which all have free ribs in wild-type animals, were transformed in mutant animals to have a characteristic typical of more anterior thoracic segments, namely, the presence of ribs attached to the sternum. Consistent with this finding, the transitional spinous process and transitional articular processes which are normally found on T10 in wild-type animals were instead found on T13 in homozygous mutants (data not shown). Additional transformations within the thoracic region were also noted in certain mutant animals. For example, in wild-type mice, the ribs derived from Tl normally touch the top of the sternum.
  • T2 appeared to have been transformed to have a morphology resembling that of T 1 ; that is, in these animals, the ribs derived from T2 extended to touch the top of the sternum. In these cases, the ribs derived from Tl appeared to fuse to the second pair of ribs. Finally, in 82% of homozygous mutants, the long spinous process normally present on T2 was shifted to the position of T3. In certain other homozygous mutants, asymmetric fusion of a pair of vertebrosternal ribs was seen at other thoracic levels.
  • the anterior transformations were not restricted to the thoracic region. The anterior most transformation that we observed was at the level of the 6th cervical vertebra (C6).
  • C6 6th cervical vertebra
  • C6 is readily identifiable by the presence of two anterior tuberculi on the ventral side.
  • C7 appeared to have been partially transformed to have a morphology resembling that of C6.
  • One other homozygous mutant had 2 anterior tuberculi on C7 but retained one on C6 for a complete C7 to C6 transformation but a partial C6 to C5 transformation.
  • Transformations of the axial skeleton also extended into the lumbar region.
  • wild-type animals normally have only 6 lumbar vertebrae
  • homozygous mutants had 8-9. At least 6 of the lumbar vertebrae in the mutants must have derived from segments that would normally have given rise to sacral and caudal vertebrae as the data described above suggest that 4 to 5 lumbar segments were transformed into thoracic segments.
  • homozygous mutant mice had a total of 33-34 presacral vertebrae compared to 26 presacral vertebrae normally present in wild-type mice.
  • Heterozygous mice also showed abnormalities in the axial skeleton although the phenotype was much milder than in homozygous mice.
  • the most obvious abnormality in heterozygous mice was the presence of an additional thoracic segment with an associated pair of ribs (Figure 14(c,f)). This transformation was present in every heterozygous animal examined, and in every case, the additional pair of ribs was attached to the sternum ( Figure 14(i)).
  • T8 whose associated rib normally does not touch the sternum, appeared to have been transformed to a morphology characteristic of a more anterior thoracic vertebra, and LI appeared to have been transformed to a morphology characteristic of a posterior thoracic vertebra.
  • mutant embryos isolated at various stages of development were not readily distinguishable from corresponding wild-type embryos.
  • the number of somites present at any given developmental age was identical between mutant and wild-type embryos, suggesting that the rate of somite formation was unaltered in the mutants.
  • day 10.5-11.5 p.c mutant embryos could be easily distinguished from wild-type embryos by the posterior displacement of the hindlimb by 7-8 somites.
  • the abnormalities in tail development were also readily apparent at this stage.
  • Alterations in expression of homeobox containing genes are known to cause transformations in Drosophila and in vertebrates.
  • Hox genes the vertebrate homeobox containing genes
  • GDF-11 null mutants we determined the expression pattern of 3 representative Hox genes, Hoxc-6, Hoxc-8 and Hoxc-11, in day 12.5 p.c. wild-type, heterozygous and homozygous mutant embryos by whole mount in situ hybridization.
  • the expression pattern of Hoxc-6 in wild-type embryos spanned prevertebrae 8-15 which correspond to thoracic segments T1-T8.
  • Hoxc-6 expression pattern was shifted posteriorly and expanded to prevertebrae 9-18 (T2-T11). A similar shift was seen with the Hoxc-8 probe.
  • Hoxc-8 was expressed in prevertebrae 13-18 (T6-T11) but, in homozygous mutant embryos, Hoxc-8 was expressed in prevertebrae 14-22 (T7-T15).
  • Hoxc-11 expression was also shifted posteriorly in that the anterior boundary of expression changed from prevertebrae 28 tin wild-type embryos to prevertebrae 36 in mutant embryos.
  • GDF-11 acts early during embryogenesis as a global regulator of axial patterning.
  • GDF-1 1 acts early during embryogenesis as a global regulator of axial patterning.
  • GDF-11 acts early during embryogenesis as a global regulator of axial patterning.
  • the phenotype of GDF-11 knockout mice in several respects resembles the phenotype of mice carrying a deletion of a receptor for some members of the TGF- ⁇ superfamily, the activin type IIB receptor (ActRIIB).
  • ActRIIB activin type IIB receptor
  • the ActRHB knockout mice have extra pairs of ribs and a spectrum of kidney defects ranging from hypoplastic kidneys to complete absence of kidneys.
  • the similarity in the phenotypes of these mice raises the possibility that ActRIIB may be a receptor for GDF-11.
  • Act RJJB may not be the sole receptor for GDF-11 because the phenotype of GDF-11 knockout mice is more severe than the phenotype of ActRIIB mice.
  • the ActRIIB knockout animals have only 3 extra pairs of ribs and do not show transformations at other axial levels.
  • the data indicate that the kidney defects in the GDF-11 knockout mice are also more severe than those in ActRIIB knockout mice.
  • the ActRHB knockout mice show defects in left/right axis formation, such as lung isomerixm and a range of heart defects that we have not yet observed in GDF- 11 knockout mice.
  • ActRIIB can bind the activins and certain BMPs, although none of the knockout mice generated for these ligands show defects in left/right axis formation.
  • GDF-11 does act directly on mesodermal cells to establish positional identity, the data presented here would be consistent with either short range or morphogen models for GDF-11 action. That is, GDF-11 may act on mesodermal precursors to establish patterns of Hox gene expression as these cells are being generated at the site of GDF-11 expression, or alternatively, GDF-11 produced at the posterior end of the embryo may diffuse to form a morphogen gradient. Whatever the mechanism of action of GDF-11 may be, the fact that gross anterior/posterior patterning still does occur in GDF-11 knockout animals suggests that GDF-11 may not be the sole regulator of anterior/posterior specification. Nevertheless, it is clear that GDF-11 plays an important role as a global regulator of axial patterning and that further study of this molecule will lead to important new insights into how positional identity along the anterior/posterior axis is established in the vertebrate embryo.
  • GDF-8 knockout animals Similar phenotypes are expected in GDF-8 knockout animals.
  • GDF-8 knockout animals are expected to have increased number of ribs, kidney defects and anatomical differences when compared to wild-type.
  • TGF ⁇ signaling Receptors, transducers, and Mad proteins. Cell 85: 947-950.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Urology & Nephrology (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Physics & Mathematics (AREA)
  • Hematology (AREA)
  • Environmental Sciences (AREA)
  • Cell Biology (AREA)
  • General Engineering & Computer Science (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Plant Pathology (AREA)
  • Animal Husbandry (AREA)
  • Biodiversity & Conservation Biology (AREA)
  • Toxicology (AREA)
  • Endocrinology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Pathology (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Abstract

La présente invention se rapporte à des récepteurs de la famille des facteurs de différenciation de la croissance (GDF growth differentiation factor) qui sont des facteurs de croissance, ainsi qu'à des procédés d'identification de ces récepteurs. Elle se rapporte également à des procédés d'identification d'anticorps qui se lient à ces récepteurs, à des fragments peptidiques du récepteur qui inhibent la liaison à un GDF, aux agents de liaison aux récepteurs de GDF capables de bloquer la liaison du GDF au récepteur. Les récepteurs de cette invention permettent l'identification d'antagonistes et d'agonistes utilisables en agriculture et à des fins thérapeutiques chez l'homme.
PCT/US1998/015598 1997-08-01 1998-07-28 Procedes d'identification des recepteurs des facteurs de differenciation de la croissance WO1999006559A1 (fr)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US09/485,046 US6696260B1 (en) 1997-08-01 1997-08-01 Methods to identify growth differentiation factor (GDF) binding proteins
AU86663/98A AU8666398A (en) 1997-08-01 1998-07-28 Methods to identify growth differentiation factor (gdf) receptors
US09/841,730 US6891082B2 (en) 1997-08-01 2001-04-24 Transgenic non-human animals expressing a truncated activintype II receptor
US11/051,267 US20050257278A1 (en) 1997-08-01 2005-02-03 Transgenic non-human animals expressing a truncated activin type II receptor
US12/360,093 US20090186806A1 (en) 1997-08-01 2009-01-26 Truncated Activin Type II Receptor and Methods of Use
US12/361,512 US8124830B2 (en) 1997-08-01 2009-01-28 Transgenic non-human animals expressing a truncated activin type II receptor
US12/861,738 US8822411B2 (en) 1997-08-01 2010-08-23 Truncated activin type II receptor and methods of use

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US5446197P 1997-08-01 1997-08-01
US60/054,461 1997-08-01

Related Child Applications (4)

Application Number Title Priority Date Filing Date
US09/485,046 A-371-Of-International US6696260B1 (en) 1997-08-01 1997-08-01 Methods to identify growth differentiation factor (GDF) binding proteins
US09485046 A-371-Of-International 1998-07-28
US09/626,896 Continuation-In-Part US6656475B1 (en) 1997-08-01 2000-07-27 Growth differentiation factor receptors, agonists and antagonists thereof, and methods of using same
US10/456,852 Division US20040077053A1 (en) 1997-08-01 2003-06-06 Methods to identify growth differentiation factor (GDF) receptors

Publications (1)

Publication Number Publication Date
WO1999006559A1 true WO1999006559A1 (fr) 1999-02-11

Family

ID=21991236

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US1998/015598 WO1999006559A1 (fr) 1997-08-01 1998-07-28 Procedes d'identification des recepteurs des facteurs de differenciation de la croissance

Country Status (2)

Country Link
AU (1) AU8666398A (fr)
WO (1) WO1999006559A1 (fr)

Cited By (51)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB2333706A (en) * 1998-02-02 1999-08-04 Merck & Co Inc Method for increasing muscle mass in animals
WO2000043781A2 (fr) * 1999-01-21 2000-07-27 Metamorphix, Inc. Inhibiteurs de facteurs de differenciation de la croissance et leurs utilisations
US6103466A (en) * 1997-07-14 2000-08-15 University Of Liege Double-muscling in mammals
US6369201B1 (en) 1998-02-19 2002-04-09 Metamorphix International, Inc. Myostatin multimers
JP2002513768A (ja) * 1998-05-06 2002-05-14 メタモーフイクス・インコーポレーテツド Gdf−8の阻害による糖尿病の処置法
US6656475B1 (en) 1997-08-01 2003-12-02 The Johns Hopkins University School Of Medicine Growth differentiation factor receptors, agonists and antagonists thereof, and methods of using same
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
EP1593689A2 (fr) * 2000-01-18 2005-11-09 Ovita Limited myostatine et mimetiques de ces derniers
US7037501B2 (en) 2001-01-04 2006-05-02 Regents Of The University Of Minnesota Myostatin immnoconjugate
WO2006083182A1 (fr) * 2005-02-07 2006-08-10 Orico Limited Utilisation d'antagonistes de myostatine (gdf-8) pour ameliorer la cicatrisation et prevenir les maladies fibreuses
WO2006107611A2 (fr) * 2005-03-23 2006-10-12 Wyeth Detection d'une reponse immunitaire contre les agents de modulation gdf-8
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
US7566768B1 (en) 1995-10-26 2009-07-28 The Johns Hopkins University School Of Medicine Promyostatin peptides and methods of using same
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
US7655763B2 (en) 2002-10-22 2010-02-02 Wyeth Neutralizing antibodies against GDF-8 and uses therefor
US7709605B2 (en) 2004-07-23 2010-05-04 Acceleron Pharma Inc. ActRII receptor polypeptides, methods and compositions
US7731961B1 (en) 2001-09-26 2010-06-08 Wyeth Methods of increasing muscle mass or muscle strength using antibody inhibitors of GDF-8
US7737116B2 (en) 2001-02-08 2010-06-15 Wyeth Modified and stabilized GDF propeptides and uses thereof
US7785587B2 (en) 2003-06-02 2010-08-31 Wyeth Therapeutic methods for muscular or neuromuscular disorders
US7842663B2 (en) 2007-02-02 2010-11-30 Acceleron Pharma Inc. Variants derived from ActRIIB and uses therefor
US7888486B2 (en) 2005-08-19 2011-02-15 Wyeth Llc Antagonist antibodies against GDF-8
US7951771B2 (en) 2005-11-23 2011-05-31 Acceleron Pharma Inc. Activin-ActRIIa antagonists and uses for promoting bone growth
US7960343B2 (en) 2007-09-18 2011-06-14 Acceleron Pharma Inc. Activin-ActRIIa antagonists and uses for decreasing or inhibiting FSH secretion
US7988973B2 (en) 2006-12-18 2011-08-02 Acceleron Pharma Inc. Activin-ActRII antagonists and uses for increasing red blood cell levels
US8058229B2 (en) 2008-08-14 2011-11-15 Acceleron Pharma Inc. Method of increasing red blood cell levels or treating anemia in a patient
US8063188B2 (en) 2006-09-05 2011-11-22 Eli Lilly And Company Anti-myostatin antibodies
US8128933B2 (en) 2005-11-23 2012-03-06 Acceleron Pharma, Inc. Method of promoting bone growth by an anti-activin B antibody
US8138142B2 (en) 2009-01-13 2012-03-20 Acceleron Pharma Inc. Methods for increasing adiponectin in a patient in need thereof
US8173601B2 (en) 2007-02-09 2012-05-08 Acceleron Pharma, Inc. Activin-ActRIIa antagonists and uses for treating multiple myeloma
US8178488B2 (en) 2009-06-08 2012-05-15 Acceleron Pharma, Inc. Methods for increasing thermogenic adipocytes
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
US8293881B2 (en) 2009-06-12 2012-10-23 Acceleron Pharma Inc. Isolated nucleic acid encoding a truncated ActRIIB fusion protein
US8388968B2 (en) 2009-04-27 2013-03-05 Novartis Ag Compositions and methods for increasing muscle growth
US8895016B2 (en) 2006-12-18 2014-11-25 Acceleron Pharma, Inc. Antagonists of activin-actriia and uses for increasing red blood cell levels
US8992913B2 (en) 2012-06-15 2015-03-31 Pfizer Inc. Antagonist antibodies against GDF-8 and uses therefor
US9493556B2 (en) 2010-11-08 2016-11-15 Acceleron Pharma Inc. Actriia binding agents and uses thereof
US9526759B2 (en) 2007-02-01 2016-12-27 Acceleron Pharma Inc. Activin-actriia antagonists and uses for treating or preventing breast cancer
US9617319B2 (en) 2009-11-17 2017-04-11 Acceleron Pharma Inc. ActRIIB proteins and variants and uses therefore relating to utrophin induction for muscular dystrophy therapy
US9850298B2 (en) 2014-06-13 2017-12-26 Acceleron Pharma Inc. Methods for treating ulcers in thalassemia syndrome with an ActRIIB polypeptide
US9919030B2 (en) 2008-06-26 2018-03-20 Acceleron Pharma Inc. Follistatin fusion proteins and uses thereof
US9975934B2 (en) 2015-03-26 2018-05-22 Acceleron Pharma Inc. Follistatin-related fusion proteins
US10010498B2 (en) 2014-06-04 2018-07-03 Acceleron Pharma Inc. Methods for treatment of amyotrophic lateral sclerosis with follistatin fusion proteins
US10023621B2 (en) 2014-06-04 2018-07-17 Acceleron Pharma Inc. Follistatin-related fusion proteins
US10195249B2 (en) 2012-11-02 2019-02-05 Celgene Corporation Activin-ActRII antagonists and uses for treating bone and other disorders
US10307455B2 (en) 2016-03-10 2019-06-04 Acceleron Pharma Inc. Activin type 2 receptor antibodies
RU2708170C2 (ru) * 2014-11-24 2019-12-04 Сомалоджик, Инк. Соединения нуклеиновой кислоты для связывания ростового фактора дифференцировки 11
US11471510B2 (en) 2014-12-03 2022-10-18 Celgene Corporation Activin-ActRII antagonists and uses for treating anemia
US11813308B2 (en) 2014-10-09 2023-11-14 Celgene Corporation Treatment of cardiovascular disease using ActRII ligand traps

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5639638A (en) * 1993-05-12 1997-06-17 Genetics Institute, Inc. DNA molecules encoding bone morpogenetic protein-11

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5639638A (en) * 1993-05-12 1997-06-17 Genetics Institute, Inc. DNA molecules encoding bone morpogenetic protein-11

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
BOUIZAR Z., ET AL.: "PURIFICATION AND CHARACTERIZATION OF CALCITONIN RECEPTORS IN RAT KIDNEY MEMBRANES BY COVALENT CROSS-LINKING TECHNIQUES.", EUROPEAN JOURNAL OF BIOCHEMISTRY, WILEY-BLACKWELL PUBLISHING LTD., GB, vol. 155., 1 January 1986 (1986-01-01), GB, pages 141 - 147., XP002913509, ISSN: 0014-2956, DOI: 10.1111/j.1432-1033.1986.tb09469.x *
HANNON K., ET AL.: "DIFFERENTIALLY EXPRESSED FIBROBLAST GROWTH FACTORS REGULATE SKELETAL MUSCLE DEVELOPMENT THROUGH AUTOCRINE AND PARACRINE MECHANISMS.", THE JOURNAL OF CELL BIOLOGY : JCB, THE ROCKEFELLER UNIVERSITY PRESS, US, vol. 132., no. 06., 1 March 1996 (1996-03-01), US, pages 1151 - 1159., XP002913507, ISSN: 0021-9525, DOI: 10.1083/jcb.132.6.1151 *
MCPHERRON A. C., LAWLER A. M., LEE S.-I.: "REGULATION OF SKELETAL MUSCLE MASS IN MICE BY A NEW TGF-BETA SUPERFAMILY MEMBER.", NATURE, NATURE PUBLISHING GROUP, UNITED KINGDOM, vol. 387., no. 6628., 1 May 1997 (1997-05-01), United Kingdom, pages 83 - 90., XP002913508, ISSN: 0028-0836, DOI: 10.1038/387083a0 *

Cited By (125)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8323964B2 (en) 1993-03-19 2012-12-04 The John Hopkins University School Of Medicine Polynucleotides encoding promyostatin polypeptides
US7393682B1 (en) 1993-03-19 2008-07-01 The Johns Hopkins University School Of Medicine Polynucleotides encoding promyostatin polypeptides
US7976839B2 (en) 1994-07-08 2011-07-12 Johns Hopkins University School Of Medicine Growth differentiation factor-11
US7384753B2 (en) 1994-07-08 2008-06-10 The Johns Hopkins University School Of Medicine Growth differentiation factor-11
US7566768B1 (en) 1995-10-26 2009-07-28 The Johns Hopkins University School Of Medicine Promyostatin peptides and methods of using same
US6103466A (en) * 1997-07-14 2000-08-15 University Of Liege Double-muscling in mammals
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
US8124830B2 (en) 1997-08-01 2012-02-28 The Johns Hopkins University School Of Medicine Transgenic non-human animals expressing a truncated activin type II receptor
US6696260B1 (en) 1997-08-01 2004-02-24 The Johns Hopkins University School Of Medicine Methods to identify growth differentiation factor (GDF) binding proteins
US6656475B1 (en) 1997-08-01 2003-12-02 The Johns Hopkins University School Of Medicine Growth differentiation factor receptors, agonists and antagonists thereof, and methods of using same
US8822411B2 (en) 1997-08-01 2014-09-02 The Johns Hopkins University School Of Medicine Truncated activin type II receptor and methods of use
GB2333706A (en) * 1998-02-02 1999-08-04 Merck & Co Inc Method for increasing muscle mass in animals
US6369201B1 (en) 1998-02-19 2002-04-09 Metamorphix International, Inc. Myostatin multimers
JP2002513768A (ja) * 1998-05-06 2002-05-14 メタモーフイクス・インコーポレーテツド Gdf−8の阻害による糖尿病の処置法
WO2000043781A3 (fr) * 1999-01-21 2001-02-01 Metamorphix Inc Inhibiteurs de facteurs de differenciation de la croissance et leurs utilisations
WO2000043781A2 (fr) * 1999-01-21 2000-07-27 Metamorphix, Inc. Inhibiteurs de facteurs de differenciation de la croissance et leurs utilisations
EP1593689A3 (fr) * 2000-01-18 2006-04-05 Ovita Limited Myostatine et mimétiques de ces derniers
US7368534B2 (en) 2000-01-18 2008-05-06 Orico Limited Myostatin and mimetics thereof
EP1593689A2 (fr) * 2000-01-18 2005-11-09 Ovita Limited myostatine et mimetiques de ces derniers
CN100422212C (zh) * 2000-07-27 2008-10-01 约翰斯·霍普金斯大学医学院 生长分化因子受体、其激动剂和拮抗剂及其使用方法
EP1317485B1 (fr) * 2000-07-27 2011-03-30 The Johns Hopkins University School Of Medicine Antagonistes de la myostatine et leurs utilisations
EP2322199A1 (fr) 2000-07-27 2011-05-18 The Johns Hopkins University School Of Medicine Récepteurs de facteur de différentiation de la croissance et procédés d'utilisation correspondants
US7037501B2 (en) 2001-01-04 2006-05-02 Regents Of The University Of Minnesota Myostatin immnoconjugate
US7488480B2 (en) 2001-01-04 2009-02-10 Regents Of The University Of Minnesota Use of passive myostatin immunization in egg laying vertebrates
US8710025B2 (en) 2001-02-08 2014-04-29 Wyeth Llc Modified and stabilized GDF propeptides and uses thereof
US7737116B2 (en) 2001-02-08 2010-06-15 Wyeth Modified and stabilized GDF propeptides and uses thereof
US7731961B1 (en) 2001-09-26 2010-06-08 Wyeth Methods of increasing muscle mass or muscle strength using antibody inhibitors of GDF-8
US8710202B2 (en) 2001-09-26 2014-04-29 Wyeth Llc Isolated nucleic acid molecule encoding an antibody that reduces GDF-8 activity
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
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
US7655763B2 (en) 2002-10-22 2010-02-02 Wyeth Neutralizing antibodies against GDF-8 and uses therefor
US8940874B2 (en) 2002-10-22 2015-01-27 Wyeth Llc 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
US9138459B2 (en) 2004-07-23 2015-09-22 Acceleron Pharma Inc. ACTRIIB-FC polynucleotides, polypeptides, and compositions
US8252900B2 (en) 2004-07-23 2012-08-28 Acceleron Pharma Inc. Actriib-Fc polynucleotides, polypeptides, and compositions
US7709605B2 (en) 2004-07-23 2010-05-04 Acceleron Pharma Inc. ActRII receptor polypeptides, methods and compositions
WO2006083182A1 (fr) * 2005-02-07 2006-08-10 Orico Limited Utilisation d'antagonistes de myostatine (gdf-8) pour ameliorer la cicatrisation et prevenir les maladies fibreuses
WO2006107611A3 (fr) * 2005-03-23 2007-04-26 Wyeth Corp Detection d'une reponse immunitaire contre les agents de modulation gdf-8
WO2006107611A2 (fr) * 2005-03-23 2006-10-12 Wyeth Detection d'une reponse immunitaire contre les agents de modulation gdf-8
US7888486B2 (en) 2005-08-19 2011-02-15 Wyeth Llc Antagonist antibodies against GDF-8
US9926368B2 (en) 2005-08-19 2018-03-27 Wyeth Llc Method for treating muscle wasting syndrome using antagonist antibodies against GDF-8
US8956608B2 (en) 2005-08-19 2015-02-17 Wyeth Llc Method for treating sarcopenia using antagonist antibodies against GDF-8
US7910107B2 (en) 2005-08-19 2011-03-22 Wyeth Llc Antagonist antibodies against GDF-8 and uses in treatment of ALS and other GDF-8 associated disorders
US8349327B2 (en) 2005-08-19 2013-01-08 Wyeth Llc Method for treating muscular dystrophy using antagonist antibodies against GDF-8
US8372625B2 (en) 2005-08-19 2013-02-12 Wyeth Llc Polynucleotides encoding antagonist antibodies against GDF-8
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
US8629109B2 (en) 2005-11-23 2014-01-14 Acceleron Pharma Inc. Method for promoting bone growth using activin-actriia antagonists
US8486403B2 (en) 2005-11-23 2013-07-16 Acceleron Pharma, Inc. Method of promoting bone growth by an anti-activin A antibody
US11129873B2 (en) 2005-11-23 2021-09-28 Acceleron Pharma Inc. Method for promoting bone growth using activin-actriia antagonists
US7951771B2 (en) 2005-11-23 2011-05-31 Acceleron Pharma Inc. Activin-ActRIIa antagonists and uses for promoting bone growth
US9163075B2 (en) 2005-11-23 2015-10-20 Acceleron Pharma Inc. Isolated polynucleotide that encodes an ActRIIa-Fc fusion polypeptide
US10239940B2 (en) 2005-11-23 2019-03-26 Acceleron Pharma Inc. Method of promoting bone growth by an anti-actriia antibody
US9480742B2 (en) 2005-11-23 2016-11-01 Acceleron Pharma Inc. Method of promoting bone growth by an anti-actriia antibody
US9572865B2 (en) 2005-11-23 2017-02-21 Acceleron Pharma Inc. Activin-actriia antagonists and uses for treating multiple myeloma
US8067360B2 (en) 2005-11-23 2011-11-29 Acceleron Pharma Inc. Method for promoting bone growth using activin-ActRIIa antagonists
US10071135B2 (en) 2005-11-23 2018-09-11 Acceleron Pharma Inc. Method of identifying an agent that promotes bone growth or increases bone density
US8128933B2 (en) 2005-11-23 2012-03-06 Acceleron Pharma, Inc. Method of promoting bone growth by an anti-activin B antibody
US8063188B2 (en) 2006-09-05 2011-11-22 Eli Lilly And Company Anti-myostatin antibodies
US8007809B2 (en) 2006-12-18 2011-08-30 Acceleron Pharma Inc. Activin-actrii antagonists and uses for increasing red blood cell levels
US8895016B2 (en) 2006-12-18 2014-11-25 Acceleron Pharma, Inc. Antagonists of activin-actriia and uses for increasing red blood cell levels
US10093707B2 (en) 2006-12-18 2018-10-09 Acceleron Pharma Inc. Antagonists of activin-ActRIIa and uses for increasing red blood cell levels
US7988973B2 (en) 2006-12-18 2011-08-02 Acceleron Pharma Inc. Activin-ActRII antagonists and uses for increasing red blood cell levels
US9526759B2 (en) 2007-02-01 2016-12-27 Acceleron Pharma Inc. Activin-actriia antagonists and uses for treating or preventing breast cancer
US7842663B2 (en) 2007-02-02 2010-11-30 Acceleron Pharma Inc. Variants derived from ActRIIB and uses therefor
US10259861B2 (en) 2007-02-02 2019-04-16 Acceleron Pharma Inc. Variants derived from ActRIIB and uses therefor
US8343933B2 (en) 2007-02-02 2013-01-01 Acceleron Pharma, Inc. Variants derived from ActRIIB and uses therefor
US9399669B2 (en) 2007-02-02 2016-07-26 Acceleron Pharma Inc. Variants derived from ActRIIB
US8173601B2 (en) 2007-02-09 2012-05-08 Acceleron Pharma, Inc. Activin-ActRIIa antagonists and uses for treating multiple myeloma
US8367611B2 (en) 2007-09-18 2013-02-05 Acceleron Pharma Inc. Activin-actriia antagonists for inhibiting germ cell maturation
US7960343B2 (en) 2007-09-18 2011-06-14 Acceleron Pharma Inc. Activin-ActRIIa antagonists and uses for decreasing or inhibiting FSH secretion
US9353356B2 (en) 2007-09-18 2016-05-31 Acceleron Pharma Inc. Activin-actriia antagonists for treating a follicle-stimulating horomone-secreting pituitary tumor
US9919030B2 (en) 2008-06-26 2018-03-20 Acceleron Pharma Inc. Follistatin fusion proteins and uses thereof
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
US10829532B2 (en) 2008-08-14 2020-11-10 Acceleron Pharma Inc. Combined use of gdf traps and erythropoietin receptor activators to increase red blood cell levels
US9505813B2 (en) 2008-08-14 2016-11-29 Acceleron Pharma Inc. Use of GDF traps to treat anemia
US11162085B2 (en) 2008-08-14 2021-11-02 Acceleron Pharma Inc. Methods for treating anemia in a subject in need thereof
US11155791B2 (en) 2008-08-14 2021-10-26 Acceleron Pharma Inc. Methods for treating anemia in a subject in need thereof
US8058229B2 (en) 2008-08-14 2011-11-15 Acceleron Pharma Inc. Method of increasing red blood cell levels or treating anemia in a patient
US11168311B2 (en) 2008-08-14 2021-11-09 Acceleron Pharma Inc. Methods for treating anemia in a subject in need thereof
US10889626B2 (en) 2008-08-14 2021-01-12 Acceleron Pharma Inc. Combined use of GDF traps and erythropoietin receptor activators to increase red blood cell levels
US8361957B2 (en) 2008-08-14 2013-01-29 Acceleron Pharma, Inc. Isolated GDF trap polypeptide
US10829533B2 (en) 2008-08-14 2020-11-10 Acceleron Pharma Inc. Combined use of GDF traps and erythropoietin receptor activators to increase red blood cell levels
US10689427B2 (en) 2008-08-14 2020-06-23 Acceleron Pharma Inc. Combined use of GDF traps and erythropoietin receptor activators to increase red blood cell levels
US10377996B2 (en) 2008-08-14 2019-08-13 Acceleron Pharma Inc. Methods of identifying ActRIIB variants
US9439945B2 (en) 2008-08-14 2016-09-13 Acceleron Pharma Inc. Isolated nucleotide sequences encoding GDF traps
US9932379B2 (en) 2008-08-14 2018-04-03 Acceleron Pharma Inc. Isolated nucleotide sequences encoding GDF traps
US8138142B2 (en) 2009-01-13 2012-03-20 Acceleron Pharma Inc. Methods for increasing adiponectin in a patient in need thereof
US8551482B2 (en) 2009-04-27 2013-10-08 Novartis Ag Compositions and methods for increasing muscle growth
US8388968B2 (en) 2009-04-27 2013-03-05 Novartis Ag Compositions and methods for increasing muscle growth
US8178488B2 (en) 2009-06-08 2012-05-15 Acceleron Pharma, Inc. Methods for increasing thermogenic adipocytes
US10968282B2 (en) 2009-06-08 2021-04-06 Acceleron Pharma Inc. Methods for screening compounds for increasing thermogenic adipocytes
US9790284B2 (en) 2009-06-08 2017-10-17 Acceleron Pharma Inc. Methods for increasing thermogenic adipocytes
US9745559B2 (en) 2009-06-12 2017-08-29 Acceleron Pharma Inc. Method for decreasing the body fat content in a subject by administering an ActRIIB protein
US9181533B2 (en) 2009-06-12 2015-11-10 Acceleron Pharma, Inc. Truncated ACTRIIB-FC fusion protein
US10358633B2 (en) 2009-06-12 2019-07-23 Acceleron Pharma Inc. Method for producing an ActRIIB-Fc fusion polypeptide
US8293881B2 (en) 2009-06-12 2012-10-23 Acceleron Pharma Inc. Isolated nucleic acid encoding a truncated ActRIIB fusion protein
US11066654B2 (en) 2009-06-12 2021-07-20 Acceleron Pharma Inc. Methods and compositions for reducing serum lipids
US10968262B2 (en) 2009-11-17 2021-04-06 Acceleron Pharma Inc. Methods of increasing sarcolemmal utrophin
US9617319B2 (en) 2009-11-17 2017-04-11 Acceleron Pharma Inc. ActRIIB proteins and variants and uses therefore relating to utrophin induction for muscular dystrophy therapy
US9493556B2 (en) 2010-11-08 2016-11-15 Acceleron Pharma Inc. Actriia binding agents and uses thereof
US8992913B2 (en) 2012-06-15 2015-03-31 Pfizer Inc. Antagonist antibodies against GDF-8 and uses therefor
US9751937B2 (en) 2012-06-15 2017-09-05 Pfizer Inc. Antagonist antibodies against GDF-8 and uses therefor
US10195249B2 (en) 2012-11-02 2019-02-05 Celgene Corporation Activin-ActRII antagonists and uses for treating bone and other disorders
US11497792B2 (en) 2014-06-04 2022-11-15 Acceleron Pharma Inc. Methods for treatment of Duchenne muscular dystrophy with follistatin polypeptides
US10023621B2 (en) 2014-06-04 2018-07-17 Acceleron Pharma Inc. Follistatin-related fusion proteins
US10954279B2 (en) 2014-06-04 2021-03-23 Acceleron Pharma Inc. Methods and compositions for treatment of disorders with follistatin polypeptides
US10010498B2 (en) 2014-06-04 2018-07-03 Acceleron Pharma Inc. Methods for treatment of amyotrophic lateral sclerosis with follistatin fusion proteins
US10765626B2 (en) 2014-06-04 2020-09-08 Acceleron Pharma Inc. Methods for treatment of charcot-marie-tooth disease with follistatin polypeptides
US11260107B2 (en) 2014-06-13 2022-03-01 Acceleron Pharma Inc. Methods and compositions for treating ulcers
US10487144B2 (en) 2014-06-13 2019-11-26 Acceleron Pharma Inc. Methods for treating ulcers in a hemoglobinopathy anemia with a soluble actRIIB polypeptide
US9850298B2 (en) 2014-06-13 2017-12-26 Acceleron Pharma Inc. Methods for treating ulcers in thalassemia syndrome with an ActRIIB polypeptide
US11813308B2 (en) 2014-10-09 2023-11-14 Celgene Corporation Treatment of cardiovascular disease using ActRII ligand traps
RU2708170C2 (ru) * 2014-11-24 2019-12-04 Сомалоджик, Инк. Соединения нуклеиновой кислоты для связывания ростового фактора дифференцировки 11
US10808251B2 (en) 2014-11-24 2020-10-20 Somalogic, Inc. Nucleic acid compounds for binding growth differentiation factor 11
US11535852B2 (en) 2014-11-24 2022-12-27 Somalogic Operating Co., Inc. Nucleic acid compounds for binding growth differentiation factor 11
US11471510B2 (en) 2014-12-03 2022-10-18 Celgene Corporation Activin-ActRII antagonists and uses for treating anemia
US11001614B2 (en) 2015-03-26 2021-05-11 Acceleron Pharma Inc. Method for treating a muscle-related disorder with follistatin-related fusion proteins
US9975934B2 (en) 2015-03-26 2018-05-22 Acceleron Pharma Inc. Follistatin-related fusion proteins
US11000565B2 (en) 2016-03-10 2021-05-11 Acceleron Pharma Inc. Methods of increasing muscle mass by administration of activin type 2 receptor antibodies
US10307455B2 (en) 2016-03-10 2019-06-04 Acceleron Pharma Inc. Activin type 2 receptor antibodies
US12042524B2 (en) 2016-03-10 2024-07-23 Acceleron Pharma Inc. Activin type 2 receptor binding proteins methods of making them

Also Published As

Publication number Publication date
AU8666398A (en) 1999-02-22

Similar Documents

Publication Publication Date Title
US6696260B1 (en) Methods to identify growth differentiation factor (GDF) binding proteins
WO1999006559A1 (fr) Procedes d'identification des recepteurs des facteurs de differenciation de la croissance
US7976839B2 (en) Growth differentiation factor-11
US7399848B2 (en) Polynucleotides encoding growth differentiation factor-8
AU772694B2 (en) Growth differentiation factor inhibitors and uses therefor
US6607884B1 (en) Methods of detecting growth differentiation factor-8
JP3786909B2 (ja) ポリペプチド及び同じものをコードしている核酸
US20080213426A1 (en) Growth Differentiation Factor-8
US20020150577A1 (en) Use of antibodies specific for growth differentiation factor-11
KR20180019521A (ko) 인간화된 sirpa-il15 녹인 마우스 및 이의 이용 방법
WO1998033887A1 (fr) Facteur-8 de differenciation de croissance
US6517835B2 (en) Growth differentiation factor-11
JP2004504832A (ja) 増殖分化因子受容体、そのアゴニスト、およびアンタゴニスト、ならびにそれらの使用方法
US8236751B2 (en) Methods of increasing muscle mass using follistatin-like related gene (FLRG)
CA2807478A1 (fr) Utilisation d'expression specifique aux muscles de gene de recepteur d'activine de type ii pour augmenter la masse musculaire chez un animal non humain
WO1999040181A1 (fr) Facteur-8 de differenciation de la croissance
AU726918B2 (en) TGFbeta signal transduction proteins, genes, and uses related thereto
AU765832B2 (en) Growth differentiation factor-11
US6210964B1 (en) Avian extracellular calcium-sensing receptor
JP2001505420A (ja) 肝臓アクチビン/インヒビンのヌクレオチド配列およびタンパク質配列ならびにそれらに基づく方法
AU2003262440A1 (en) Growth differentiation factor-11
Superfamily A aturally Occurring Growth Hormone Receptor Mutation: in Vivo

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

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

AL Designated countries for regional patents

Kind code of ref document: A1

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

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

Ref country code: KR

WWE Wipo information: entry into national phase

Ref document number: 09485046

Country of ref document: US

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

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

Ref country code: CA