US20080118487A1 - Myostatin Isoform - Google Patents

Myostatin Isoform Download PDF

Info

Publication number
US20080118487A1
US20080118487A1 US11/576,449 US57644905A US2008118487A1 US 20080118487 A1 US20080118487 A1 US 20080118487A1 US 57644905 A US57644905 A US 57644905A US 2008118487 A1 US2008118487 A1 US 2008118487A1
Authority
US
United States
Prior art keywords
polynucleotide
polypeptide
seq
muscle
msv
Prior art date
Legal status (The legal status 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 status listed.)
Abandoned
Application number
US11/576,449
Other languages
English (en)
Inventor
Ferenc Jeanplong
Christopher David McMahon
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Orico Ltd
Original Assignee
Orico Ltd
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 claimed from NZ53569604A external-priority patent/NZ535696A/en
Application filed by Orico Ltd filed Critical Orico Ltd
Publication of US20080118487A1 publication Critical patent/US20080118487A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/02Muscle relaxants, e.g. for tetanus or cramps
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07HSUGARS; DERIVATIVES THEREOF; NUCLEOSIDES; NUCLEOTIDES; NUCLEIC ACIDS
    • C07H21/00Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids
    • C07H21/04Compounds containing two or more mononucleotide units having separate phosphate or polyphosphate groups linked by saccharide radicals of nucleoside groups, e.g. nucleic acids with deoxyribosyl as saccharide radical
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention is based upon the identification of a novel splice variant of myostatin.
  • the present invention is based on the use of the myostatin splice variant to modulate or regulate muscle growth and myostatin activity.
  • Myostatin (or GDF-8) is a negative regulator of muscle growth and is structurally related to the transforming growth factor ⁇ (TGF- ⁇ ) superfamily (McPherron et al 1997a). More particularly, myostatin is a potent negative regulator of skeletal muscle during development, and in adult life, in a wide range of species from fish to mammals (McPherron and Lee, 1997). Myostatin is known to regulate its own expression via a mechanism that is incompletely understood at present (Berry et al. 2002, Spiller et al. 2002, Rebbapragada et al. 2003).
  • the myostatin protein is initially translated as a 375 amino acid precursor molecule having a secretory signal sequence at the N-terminus, a proteolytic processing signal (RSRR) of the furin endoprotease, and nine conserved cysteine residues in the C-terminal region to facilitate the formation of a “cysteine knot” structure.
  • Myostatin is activated by furin endoprotease cleavage at Arg 266 releasing the N-terminal, or “latency-associated peptide” (LAP) and the mature, C-terminal domain, which dimerises to form the active myostatin molecule.
  • a homodimer of the LAP peptide remains non-covalently bound to the homodimer of mature myostatin in an inactive complex (Lee et al. 2001).
  • Other, proteins for example, follistatin, titin cap, GDFP1, follistatin related gene and hSGT are also known to bind to and regulate the secretion and activation of the latent myostatin complex (Lee et al. 2001, Nicolas et al. 2002, Hill et al. 2002, Hill et al. 2003, Wang et al. 2003).
  • myostatin inhibits myoblast proliferation and differentiation without inducing apoptosis or stimulating muscle protein breakdown (Thomas et al. 2000, Langley et al. 2002, Rios et al. 2001, Taylor et al. 2001).
  • Knock-out mice for myostatin have greatly increased muscle mass over their entire body.
  • Myostatin-null mice have approximately 30% greater body weight than normal mice, and exhibit a 2-3-fold increase in individual muscle weights due to muscle fibre hyperplasia and hypertrophy.
  • Myostatin has also been linked with many other biological processes. For example, knockout transgenic mice have altered cortical bone structure indicating a role in osteogenesis (Hamrick 2003). Furthermore, myostatin has been shown to be involved in regulating glucose and fat metabolism, thus it may be implicated in type 2 diabetes and obesity (McPherron and Lee 2002).
  • myostatin has been implicated in a number of disorders associated with muscle wasting, or muscle atrophy, such as that seen in individuals affected by HIV, cancer, prolonged bed rest, or muscular dystrophy (Gonzalez-Cadavid et al. 1998, Langley et al 2004, Zachwieja et al 1999, Bogdanovich et al. 2002). It was demonstrated that in vivo administration of myostatin induces cachexia, a severe form of muscle wasting associated with cancer and sepsis (Zimmers et al. 2002). Furthermore, up-regulation of myostatin in glucocorticoid-induced muscle atrophy has been observed (Ma et al. 2003). Changes in myostatin expression have been shown in other conditions, for example, up-regulated in cardiomyocytes after heart damage, and down regulated in regenerating muscle (Sharma et al. 1999).
  • the present invention fulfils these needs, in part by providing a novel, biologically active myostatin splice variant, and also offers other related advantages.
  • the present invention provides for an isolated polypeptide comprising an amino acid sequence having the formula
  • X 1 I F L E X 2 X 3 X 4 Q X 5 C S I L X 6 X 7 X 8 X 9 X 10
  • X 1 is I or L
  • X 2 is V or L
  • X 3 is Y, C, G or S
  • X 4 is I or F
  • X 5 is F or L
  • X 6 is G or E
  • X 7 is E or V
  • Xs is A or T
  • X 9 is A or V
  • X 10 is absent, F or L.
  • the isolated polypeptide can also be selected from:
  • the isolated polypeptide according to the present invention also includes an amino acid sequence of any one of SEQ ID NOS: 48-95 or a variant thereof having at least 80% sequence identity, wherein the peptide is capable of promoting myoblast cell growth.
  • the invention also provides for an isolated polynucleotide comprising a nucleotide sequence that encodes a polypeptide according to any one of claims 1 to 3 , or a complementary sequence thereto.
  • An isolated polynucleotide according to the present invention can be selected from:
  • the invention also provides an isolated polynucleotide consisting of SEQ ID NO: 6 or SEQ ID NO: 27.
  • the isolated polynucleotides includes a nucleotide sequence of any one of SEQ ID NOS: 1 to 47 or 96 or a variant thereof having 80% sequence identity that encodes a peptide that is capable of promoting myoblast cell growth.
  • the invention also provides for a vector comprising the polynucleotide sequences according to the present invention.
  • the vector can be an expression vector, and can comprising in operable linkage:
  • the polynucleotide can either be in a sense orientation or in an antisense orientation.
  • Host cell comprising a such vectors are also included, along with host animals comprising one or a plurality of cells which contain a vector according to the present invention.
  • the invention also provides for a composition for regulating muscle growth, comprising a compound of any one of:
  • the composition can include an anti-sense polynucleotide, including an interfering RNA molecule such as an RNAi or siRNA.
  • an anti-sense polynucleotide including an interfering RNA molecule such as an RNAi or siRNA.
  • the invention also provides for a method for regulating muscle growth, comprising administering a composition that is selected from the group consisting of.
  • the invention also provides for a method for treating a disease associated with muscle tissue, comprising administering a composition that is selected from the group consisting of:
  • the method can be to treat a disease associated with muscle tissue comprises a condition that is associated with muscle atrophy and may be selected from the group consisting of muscular dystrophy, muscle cachexia, atrophy, hypertrophy, disease-associated muscle atrophy and amyotrophic lateral sclerosis (ALS).
  • the method also includes the treatment of a disease-associated muscle wasting associated with cancer or HIV/AIDS.
  • the disease also includes a disease associated with cardiac muscle, such as comprises infarct.
  • the present invention also provides for a modulator of MSV gene expression comprising a composition that is able to specifically bind to a polynucleotide selected from the group consisting of:
  • the modulator of MSV gene can comprises an anti-sense polynucleotide, such as an RNAi or siRNA molecule.
  • the present invention also provides for a method for treating a disease associated with muscle tissue in a patient, comprising administering to said patient a modulator according to the present invention.
  • the disease can be muscle atrophy, and may be selected from the group consisting of muscular dystrophy, muscle cachexia, atrophy, hypertrophy, disease-associated muscle atrophy, and amyotrophic lateral sclerosis (ALS),
  • the disease may also be a disease-associated muscle atrophy comprising muscle wasting associated with cancer or HIV/AIDS.
  • the disease can also be associated with cardiac muscle, and includes infarct.
  • the present invention also provides for a method for modulating MSV activity, comprising contacting a MSV propeptide with a propeptide convertase, under conditions and for a time sufficient for the convertase to alter proteolytic processing of the MSV propeptide into an active MSV peptide, and thereby modulating MSV activity.
  • the contacting step can filter comprise contacting the MSV propeptide with all agonist or antagonist of the convertase.
  • An example of a propeptide convertase is a furin endoprotease.
  • the present invention also provides for a method for treating a disease associated with muscle tissue, comprising administering a propeptide convertase to a subject having a disease associated with muscle tissue, said subject comprising a MSV propeptide, under conditions and for a time sufficient for the convertase to alter proteolytic processing of the MSV propeptide into an active MSV peptide, and thereby treating the disease.
  • the administering step can further comprise contacting the MSV propeptide with an agonist or antagonist of the convertase,
  • An example of a propeptide convertase is a furin endoprotease.
  • the method can be for the treatment of a disease associated with muscle atrophy, and can be selected from any one muscular dystrophy, muscle cachexia, atrophy, hypertrophy, disease-associated muscle atrophy and amyotrophic lateral sclerosis (ALS).
  • the method can also be for the treatment of a disease-associated muscle atrophy comprising muscle wasting associated with cancer or HIV/AIDS.
  • the method can also be for the treatment of a disease associated with cardiac muscle, including infarct.
  • the present invention also provides for a method of regulating muscle growth of an animal comprising administering to said animal any one of: a composition according to the present invention; a modulator of MSV gene expression according to the present invention; an MSV propeptide that has been contacted with propeptide convertase or a agonist or antagonist of a propeptide convertase.
  • a propeptide convertase is furan endoprotease.
  • the method may also be used to produce an animal having an increased muscle mass.
  • the present invention also provides for a method of predicting muscle mass in an animal, comprising the steps of:
  • the level of gene expression can be determined by a method comprising nucleic acid hybridization under stringent conditions to the polynucleotide, including RT-PCR or northern analysis.
  • the present invention also provides for a method of predicting muscle mass in an animal, including the steps of:
  • the amount of polypeptide can be determined by a method comprising detection of the polypeptide with an antibody that specifically binds to said polypeptide, including ELISA or western blot analysis.
  • the present invention also provides for a method of increasing the muscle mass of one or more offspring of an animal comprising the steps of:
  • the method can be preformed on an selected from a sheep, cattle, deer, poultry, turkey, pig, horse, mouse, rat or human,
  • the present invention also provides for a mammalian antibody that specifically binds a polypeptide having
  • the antibody may comprise a mammalian antibody, or a fragment or derivative thereof that is able to bind polypeptide having a sequence of any one of SEQ ID NOS: 48 to 95 or a polypeptide having 95%, 90%, 80% or 70% sequence identity to any one of SEQ ID NOS: 48 to 95.
  • FIG. 1 shows a schematic representation of the myostatin gene and the RNA splicing giving rise to the canonical mRNA and the variant mRNAs (oMSV, bMSVh/f and bMSVbb).
  • FIG. 2 shows northern blots detecting the presence of canonical ovine myostatin (Mstn) and MSV mRNAs using a probe extending across the boundary of Exons 1 and 2 (Ex1/2), and probes complementary to the 3′-UTR of ovine myostatin gene beyond the polyadenylation sites of canonical myostatin (designated MSV 1 and MSV 2).
  • FIGS. 3A & B show the effect of recombinant ovine MSV65 protein on the proliferation of murine C 2 C 12 (3A) and ovine primary myoblasts (3B).
  • Myoblasts were grown in the presence of increasing concentrations of roMSV65 (0 to 10 ⁇ g/ml) for 72 h.
  • Cell proliferation was determined by methylene blue assay (optical density at 655 nm). Values are mean ⁇ SEM (*p ⁇ 0.05, **p ⁇ 0.01, ***p ⁇ 0.001).
  • FIG. 4 shows the effect of recombinant ovine MSV65 protein on the amount (Mean ⁇ SEM) of myogenin protein in differentiated C 2 Cl 2 myotubes.
  • Total protein was extracted from the cells and the abundance of myogenin was determined by Western blot analysis ( ⁇ p ⁇ 0.1,**p ⁇ 0.01).
  • FIG. 5 shows the effect of recombinant ovine MSV65 on the amount of developmental myosin heavy chain (dMHC) protein expression in C 2 Cl 2 myotubes.
  • FIG. 6 shows the effect of recombinant ovine MSV65 on the amount (Mean ⁇ SEM) of p21 protein levels in proliferating C 2 C 12 myoblasts.
  • Total protein was extracted from the cells and p21 levels were determined by Western blot analysis (* p ⁇ 0.05, ** p ⁇ 0.01).
  • FIG. 7 shows the effect of recombinant ovine MSV65 on the amount (Mean ⁇ SEM) of proliferating cell nuclear antigen (PCNA) protein levels in proliferating C 2 C 12 myoblasts.
  • Total protein was extracted from the cells and PCNA levels were determined by Western blot analysis ( ⁇ p ⁇ 0.1, * p ⁇ 0.05).
  • FIG. 8 shows the identification of MSV proteins in tissues of different species using Western immunoblots. Protein samples were separated by 10% ( 8 A) or 15% ( 8 B, C) SDS-PAGE and transferred to nitrocellulose membranes. Blots were probed with propeptide-specific ( 8 A) or mature MSV-specific polyclonal antibodies ( 8 B & C). Expected sizes of precursor (37 kDa), propeptide (29 kDa) and putative mature dimmer (11 kDa) MSV proteins in sheep are indicated by arrows.
  • hypoth hypothalamus
  • hu human
  • sh seep
  • mo mo—mouse
  • 8 A, B mo—month
  • UN undernourished
  • y year
  • female
  • male.
  • FIG. 9 shows mean ( ⁇ SEM) proliferation of murine C 2 C 12 myoblasts treated with and without myostatin (Mstn) and roMSV65 as indicated for 48 h.
  • Mstn myostatin
  • roMSV65 firstly, stimulated proliferation of murine myoblasts and, secondly overcame myostatin-induced inhibition of proliferation of primary human myoblasts and stimulated proliferation beyond restoration.
  • FIG. 10 shows the effect of roMSV47 protein on the proliferation of murine C 2 C 12 myoblasts.
  • Myoblasts were grown in the presence of increasing concentrations of roMSV47 (1-10,000 ng/ml) for 60 h.
  • Cell proliferation was determined by methylene blue assay (absorbance at 655 ⁇ m). The dagger and asterisks indicate significant differences from time 0 ( ⁇ P ⁇ 0.1, ***P ⁇ 0.001).
  • FIG. 11 shows mean ( ⁇ SEM) proliferation of murine C 2 C 12 myoblasts treated with and without myostatin (Mstn) and roMSV47 as indicated for 69 h.
  • FIGS. 12A & B A shows the effect of roMSV47 and rhMSV38 on proliferation of murine C 2 C 12 myoblasts over 80h.
  • Cell proliferation was determined by methylene blue assay (absorbance at 655 nm).
  • FIG. 13 shows the mean ( ⁇ SEM) proliferation of primary human myoblasts treated with and without myostatin (Mstn) and rhMSV38 as indicated for 48 h.
  • Doses of rhMSV38 are in molar amounts (x) relative to the 1.5 ⁇ g/ml Mstn used in the second part, or the 3 ⁇ g/ml Mstn used in the third part.
  • Unlike letters indicate significance (a,bP ⁇ 0.1, a,cP ⁇ 0.01).
  • Asterisks indicate significant differences from controls (Con, * P ⁇ 0.05, *** P ⁇ 0.001).
  • FIG. 15 shows the putative bioactive domains of MSV.
  • GCG software Accelrys Inc., San Diego Calif. USA
  • bovine Bos taurus
  • porcine MSV sequences In contrast, the peptide sequence is terminated C-terminal to the first alpha helix in the remaining species shown.
  • a putative proteolytic cleavage site R ⁇ (R/K) ⁇ X ⁇ I is indicated below the consensus sequence that was consistent with a site recognised by the proprotein convertase SK-I which cleaves peptides C-terminal to the arrow (Seidah and Chretien 1999).
  • the ionic charge of the amino acids in the helices shows that the ⁇ -helix1 has a consistent pattern for all sequences listed and where amino acids differ, they have the same charge.
  • Cell proliferation was determined by methylene blue assay (absorbance at 655 nm).
  • Asterisks indicate significance from controls (no added roMSV ⁇ 1, ** P ⁇ 0.01,*** P ⁇ 0.001).
  • Cell proliferation was determined by methylene blue assay (absorbance at 655 nm).
  • Asterisks indicate significance from controls (no added peptide, * P ⁇ 0.05, ** P ⁇ 0.01,*** P ⁇ 0.001).
  • FIG. 18 shows the effect of recombinant ovine (ro) MSV47 protein on the extent of muscle wasting in rats injected with the AH130 tumour.
  • Muscle mass from the combined wet mass of biceps femoris, gastrocnemius, tibialis anterior, quadriceps femoris, plantaris and soleus is expressed as percent of initial bodymass relative to non-cancer controls. Asterisks indicates significant difference from the AH130 cancer controls (*P ⁇ 0.05).
  • FIG. 19 shows that mean concentrations of creatine kinase (CK) were reduced at 6 d in rats bearing the AH130 tumour compared to the control rats. In contrast, concentrations of CK were not as markedly reduced in rats bearing the AH130 tumour, but injected twice daily with roMSV47 (MSV) (1 ⁇ g/g bodymass, s.c.) for 6 d. Unlike letters indicate significance (a,bP ⁇ 0.01, a,cP ⁇ 0.01, b,cP ⁇ 0.05).
  • the present invention relates in certain embodiments to the surprising discovery of a functional and biologically active myostatin variant mRNA molecule, which is the unexpected product of alternative splicing in the course of myostatin gene expression.
  • flanking PCR primers specific for nucleotide sequences in and adjacent to a myostatin-encoding open reading frame (ORF) were used in a reverse transcription-polymerase chain reaction (RT-PCR).
  • oMSV ovine myostatin splice variant
  • polynucleotide is to be understood as meaning a polymer of deoxyribonucleic acids or ribonucleic acids, and includes both single stranded and double stranded polymers, including DNA, RNA, cDNA, genomic DNA, recombinant DNA, nucleic acid molecules prepared from natural or artificial nucleosides or nucleotides, and all other known forms of polynucleotides.
  • the polynucleotide may be isolated from a naturally occurring source, produced using recombinant or molecular biological techniques, or produced synthetically.
  • a polynucleotide may include a whole gene or any pat thereof, and does not have to include an open reading frame.
  • Open reading frames can be established using known techniques in the art. These techniques include the analysis of polynucleotide sequences to identify known start and stop codons. Many computer software programmes that can perform this function are known in the art.
  • a “polypeptide” is to be understood as meaning a polymer of naturally occurring and/or artificial amino acids covalently linked via peptide bonds.
  • a polypeptide includes a polypeptide that has been isolated from a naturally occurring source, a polypeptide that has been produced using recombinant techniques, or a polypeptide that has been produced synthetically. It is to be appreciated that a polypeptide that includes a leader or pro-sequence, or a polypeptide that undergoes a post translational modification is intended to come within the definition of a polypeptide.
  • fragment or variant is to be understood to mean any polynucleotide or polypeptide sequence or partial sequence that has been modified by substitution, insertion or deletion of one or more nucleotides or one or more amino acids, but that has substantially the same activity or function as the MSV sequence or partial sequence of the invention
  • Polynucleotide or polypeptide variants have at least 70% similarity (preferably a 70% sequence identity), 80% similarity (preferably a 80% sequence identity), 85% similarity (preferably a 85% sequence identity), and more preferably 90% similarity (more preferably a 90% sequence identity) to he reported polynucleotides or polypeptides, more preferably 95% similarity (more preferably a 95% sequence identity), and still more preferably a 98% similarity (still more preferably a 98% sequence identity) to the reported polynucleotides or polypeptides.
  • % identity refers to the percentage of identical amino acids situated at corresponding amino acid residue positions in a sequence when two or more polypeptide are aligned and their sequences analyzed using a gapped BLAST algorithm (e.g., Altschul et al., Nucleic Acids Res.
  • a variant contains conservative substitutions.
  • a “conservative substitution” is one in which an amino acid is substituted for another amino acid that has similar properties, such that one skilled in the art of peptide chemistry would expect the secondary structure and hydropathic nature of the polypeptide to be substantially unchanged.
  • Amino acid substitutions may generally be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity and/or the amphipathic nature of the residues.
  • negatively charged amino acids include aspartic acid and glutamic acid; positively charged amino acids include lysine and arginine; and amino acids with uncharged polar head groups having similar hydrophilicity values include leucine, isoleucine, and valine; glycine, and alanine; asparagine and glutamine; and serine, threonine, phenylalanine, and tyrosine.
  • amino acids that may represent conservative changes include (1) ala, pro, gly, glu, asp, gln, asn, ser, thr; (2) cys, ser, tyr, thr; (3) val, ile, leu, met, ala, phe; (4) lys, arg, his; and (5) phe, tyr, trp, his.
  • Amino acids may be classified according to the nature of their side groups.
  • Amino acids with nonpolar alkyl side groups include glycine, alanine, valine, leucine, and isoleucine.
  • Serine and threonine have hydroxyl groups on their side chains, and because hydroxyl groups are polar and capable of hydrogen bonding, these amino acids are hydrophilic.
  • Sulfur groups may be found in methionine and cysteine.
  • Carboxylic acid groups are part of the side chain of aspartic acid and glutamic acid, which because of the acidity of the carboxylic acid group, the amino acids are not only polar but can become negatively charged in solution.
  • Glutamine and asparagine are similar to glutamic acid and aspartic acid except the side chains contain amide groups. Lysine, arginine, and histidine have one or more amino groups in their side chains, which can accept protons, and thus these amino acids act as bases. Aromatic groups may be found on the side chains of phenylalanine, tyrosine, and tryptophan. Tyrosine is polar because of its hydroxyl group, but tryptophan and phenylalanine are non-polar. A variant may also, or alternatively, contain nonconservative changes.
  • a MSV variant with at least one substitution, addition, insertion, or deletion may be made according to mutagenesis methods described herein and known in the art. Such modifications in a polynucleotide sequence that encodes a MSV variant or derivative may be introduced using standard mutagenesis techniques, such as oligonucleotide-directed site-specific mutagenesis. Alterations of the native amino acid sequence may be accomplished by any of a number of conventional methods. Mutations can be introduced at particular loci by synthesizing oligonucleotides containing a mutant sequence, flanked by restriction sites enabling ligation to fragments of the native sequence. Following ligation, the resulting reconstructed sequence encodes an analog having the desired amino acid insertion, substitution, or deletion.
  • a MSV polypeptide for use in the present invention may be a portion or fragment of a full-length MSV polypeptide or a truncated MSV polypeptide as described herein.
  • a truncated MSV polypeptide as described in greater detail herein may be any MSV polypeptide molecule that comprises less than a full-length version of the MSV polypeptide.
  • deletion has its common meaning as understood by those familiar with the art, and may refer to molecules that lack one or more of a portion of a sequence from either terminus or from a non-terminal region, relative to a corresponding full length molecule, for example, as in the case of truncated molecules provided herein.
  • a portion or fragment or a truncated form of a MSV polypeptide may have any number of amino acids fewer than about 320, 300, 275, 250, 225, 200, 175, 170, 160, 155, 150, 149, 148, 147, 146, 145, 144, 143, 142, 141, 140, 139, 138, 137, 135, 130, 125, 120, 115, 100, 95, 90, 85, 80, 75, 70, 65, 60, 65, 50, 45, 40, 35, 35, 25, 20, or 15 amino acids.
  • a polynucleotide fragment also includes a polynucleotide fragment of sufficient length and specificity to hybridise under stringent conditions to a sequence of any one of SEQ ID NOS: 1-7.
  • stringent conditions involves pre-hybridisation with 5 ⁇ SSC, 0.2% SDS at 65° C.; performing the hybridisation overnight in 5 ⁇ SSC, 0.2% SDS at 65° C.; two washes of 1 ⁇ SSC, 0.1% SDS at 65° C. for 30 min each; followed by a further two washes of 0.2 ⁇ SSC, 0.1% SDS at 65° C., also for 30 min each.
  • a polypeptide fragment also includes a fragment that retains the activity of promoting muscle growth.
  • This fragment may have altered activity, i.e., activity that is increased or decreased in a statistically significant manner relative to an appropriate control polypeptide (e.g., a full length polypeptide), for example, enhanced activity such that the fragment, when introduced or expressed in a cell, results in an increased (e.g., with statistical significance) ability to promote muscle growth.
  • the fragment may have altered activity that is manifest as a statistically significant decrease in function, for instance, a dominant negative effect.
  • a suitable cell line could be movine C 2 C 12 myoblasts (ATCC NO: CRL-1772), however, it will be appreciated that any suitable myoblast call line could be used, such as primary ovine or human myoblasts as described herein.
  • isolated refers to removal of a molecule such as a MSV polypeptide or encoding polynucleotide from its natural source, environment or milieu (e.g., removal of a protein from an intact cell source), and the term “purified” as used herein means that the MSV polypeptide or encoding polynucleotide is essentially free of association with other polynucleotides, proteins or polypeptides, for example, as a purification product of recombinant host cell culture, or as a purified product from a non-recombinant source.
  • An “isolated” polypeptide therefore is one that is removed from its original environment.
  • such polypeptides are at least about 70%, 75%, 80%, 85% or 90% pure, at least about 95% pure, or at least about 99% pure, for example, where such a degree of purity refers to the percentage of detectable MSV polypeptide or encoding polynucleotide that is present in a preparation relative to other detectable polynucleotides and/or polypeptides.
  • substantially purified or “substantially isolated” as used herein means a mixture that contains a molecule such as a MSV polypeptide or encoding polynucleotide that is essentially free of association with other polynucleotides, proteins or polypeptides, but for the presence of known proteins that can be removed using conventional methods, such as by affinity chromatography with a specific antibody or ligand, and which substantially purified or substantially isolated MSV polypeptide or encoding polynucleotide retains its biochemical characteristics as described herein or retains at least one of its detectable functional biological activities.
  • a molecule such as a MSV polypeptide or encoding polynucleotide that is essentially free of association with other polynucleotides, proteins or polypeptides, but for the presence of known proteins that can be removed using conventional methods, such as by affinity chromatography with a specific antibody or ligand, and which substantially purified or substantially isolated MSV polypeptide or encoding polynucleotide
  • Gene expression is to be understood as meaning the initiation of transcription, the transcription of a section of DNA into mRNA, and the translation of the mRNA into a polypeptide.
  • a modulator of gene expression is defined as any compound that is able to cause, in a statistically significant fashion, an increase or decrease in gene expression, and may act at any point in the gene expression pathway.
  • Muscle growth is to be understood as meaning the division and/or differentiation of muscle cells and includes the division and/or differentiation of any precursor cell, fusion of such cells with each other and/or with existing muscle fibres, and it also includes increased protein synthesis in myofibers leading to higher protein content and greater muscle fibre volume (muscle fibre hypertrophy).
  • a modulator of protein activity is to be understood as meaning an agent, compound or composition that is able to increase or decrease protein activity or function (e.g., a myostatin biological activity) in a statistically significant fashion.
  • the presently disclosed myostatin splice variant is believed to be the result of an alternative splicing event.
  • the precursor canonical myostatin mRNA may be transcribed so as to include polynucleotide sequences of all three exons of the myostatin gene.
  • the three exons are bounded by splice donor and acceptor sites, SD1 and SA1, and SD2 and SA2 respectively. Removal of the two introns, one between SD1 and SA1, and the other between SD2 and SA2, produces the canonical myostatin mRNA.
  • the translation start site is located in exon 1, and the stop codon is located about one third of the 5 way into exon 3, meaning that normally there is a large 3′ untranslated region within exon 3.
  • an alternative splicing event may also take place in the course of myostatin gene expression by a process in which an extra splicing event occurs between SD3 and SA3 or between SD2 and SA3 ( FIG. 1 ), causing the majority of the normally translated portion of the canonical exon 3 to be excised and replaced with the normally 3′ untranslated section of exon 3.
  • This unexpected splicing event creates a new open reading frame from which the MSV protein may be translated.
  • the resulting MSV polypeptide, ovine MSV (oMSV; SEQ ID NO: 48) and bovine MSV (bMSV; SEQ ID NO: 52) shares the first 257 amino acids with native myostatin propeptide, but has a unique 64 amino acid C-terminal end (ovine oMSV65, SEQ ID NO:49 and bovine bMSV, SEQ ID NO: 52). It is important to note that at the mRNA level, the 3′ unique end of the MSV ORF differs by 195 nucleotides.
  • valine residue at position 257 in MSV protein is the same in the canonical myostatin sequence by pure coincidence because the splicing site exactly precedes the nucleotide triplet coding for a valine residue, which is part of the translated sequence of MSV. Therefore, the splice actually results in a 65 amino acid fragment, only 64 amino acids of which differ to the canonical myostatin sequence.
  • bovine(bt) Bos taurus ; b1(bt)MSV65, SEQ ID NO: 6 and 53; b2(bt)MSV65, SEQ ID NO: 9 and 56; b3(bt)MSV65, SEQ ID NO: a2 and 59 and b4(bt) MSV65 SEQ ID NO: 15 and 62), bovine(bi) ( Bos indicus , b(bi)MSV65, SEQ ID NO: 18 and 65), bovine(bg) ( Bos grunniens , Yak, b(bg)MSV65, SEQ ID NO: 21 and 68), porcine ( Sus scrofa , pMSV68, SEQ ID NO: 24 and 71), human ( Homo sapiens , h1MSV38, SEQ ID NO: 27 and 74 and h2MSV38, SEQ ID NO:
  • porcine sequence (pMSV68, SEQ ID NO: 71) has an extra three amino acid residues, totaling 68.
  • human, chimp, and cat are all truncated, and provide for only 38 amino acid residues.
  • the sequence for the dog is even shorter, being only 36 amino acids long.
  • Analysis of the various sequences has also identified a number of polymorphisms. Two human variations, have been identified, h1MSV38 (SEQ ID NO: 74), which has a glycine residue at position 1, and h2MSV38 (SEQ ID NO: 77) which has valine residue at position 1.
  • d1MSV36 SEQ ID NO: 83
  • d2MSV36 SEQ ID NO: 86
  • d3MSV36 SEQ ID NO: 89
  • bovine ( Bos taurus ) b1 (bt)MSV65 (SEQ ID NO: 53) has a phenylalanine at position 21, a glutamic acid at position 34 and glutamine at position 48; b2(bt)MSV65 (SEQ ID NO: 56) has a leucine at position 21, a valine at position 34 and a glutamine at position 48; b3(bt)MSV65 (SEQ ID NO: 59) has a leucine at position 21, a glutamic acid at position 34 and a glutamine at position 48; and b4(bt)MSV65 (SEQ ID NO: 62) has a phenylalanine at position 21, a glutamic acid at position 34 and a serine at position 48.
  • the invention therefore includes the MSV polynucleotide and polypeptide sequences (including polymorphisus) for bovine, ovine, porcine, human, chimp, dog and cat as set out below:
  • the polynucleotides of the present invention may be incorporated into vectors.
  • Vectors are intended to include the incorporation of a sequence according to the present invention into a plasmid and/or virus to aid in the introduction and/or maintenance of the sequence in a host cell.
  • Suitable vectors are known in the art and includes expression vectors.
  • An expression vector includes a vector specifically for expressing the protein of interest in a particular host cell.
  • Such vectors include a promoter sequence, the polynucleotide of interest and a gene termination in operable linkage, meaning that the promoter will effect gene expression of the polynucleotide of interest, while the gene termination will terminate transcription.
  • Suitable promoters to effect gene expression are well known in the art and may include, but are not limited to, the myostatin promoter.
  • the host cell may include, either, a prokaryotic or a eukaryotic cell.
  • the eukaryotic cell may be in vivo, or may be a primary or transformed cell line.
  • compositions based on the novel polynucleotide and/or polypeptides of the present invention are also contemplated.
  • One or more compositions may be used to regulate myostatin or to regulate muscle growth.
  • the regulation of muscle growth is intended to include any change in the rate of muscle growth and/or development and includes the growth and/or differentiation of any muscle precursor cell. This includes any change in the rate at which precursor muscle cells divide, and any change in the rate at which precursor muscle cells differentiate. The change may be either an increase or a decrease.
  • the composition may comprise a polynucleotide sequence according to the present invention, including any one of SEQ ID NOS: 1 to 47 or 96, a polynucleotide having 95%, 90%, 80% or 75% identity to any one of the polynucleotides, or a fragment or variant thereof.
  • the sequence may be introduced into a cell by incorporation into a suitable vector under the regulation of a promoter, either the myostatin promoter or any other suitable promoter.
  • the promoter may be used to cause expression of a MSV protein according to the present invention, thereby both increasing gene expression and MSV activity within the cell.
  • the composition may also include compliments, reverse compliments, or anti-sense polynucleotides of the polynucleotides according to the present invention.
  • the composition may also include a polypeptide sequence according to the present invention, including any one of SEQ ID NOS: 48 TO 95 or a polypeptide having 95%, 90%, 80% or 75% identity to any one of the polypeptides.
  • the peptide can be directly incorporated into a composition suitable for administration to a subject.
  • a suitable composition can include compositions for oral or topical administration, inhalation, administration by injection or any other suitable form.
  • Sequence identity may be determined by aligning the sequences and determining the number of identical residues.
  • Many computer algorithms are known in the art for determining the sequence identity, for example BLASTN for determining the identity between polynucleotide sequences, and BLASTP for determining the identity between polypeptide sequences.
  • the composition may also include a modulator of MSV activity. This may comprise a modulator of MSV gene expression or a modulator of MSV protein activity.
  • the modulator of MSV expression may be a compound that can specifically bind to a polynucleotide according to the present invention and affect the rate of MSV gene expression. An alteration in gene expression can be determined by either an increase or decrease in MSV protein in a cell, or subject.
  • the modulator of MSV expression could bind to the MSV promoter, thereby affecting the rate at which gene transcription is initiated.
  • the modulator of MSV gene expression may also bind to the MSV gene or MSV mRNA directly affecting the rate at which the gene is expressed.
  • the modulator of MSV expression may also bind to the MSV gene and introduce alterations into the sequence, for example, by homologous recombination, which may either affect the rate at which the gene is expressed, or may alter the MSV protein activity. Alterations of a sequence include a nucleotide change, insertion or deletion, which may or may not result in an amino acid change, insertion or deletion in the resulting polypeptide.
  • alterations can include the insertion of a termination codon, such that a truncated polypeptide is produced, or the alteration of one or more codons such that one or more amino acid residues are altered.
  • the variations could be to delete a section of the wild-type MSV gene, or introduce a section into the MSV gene. Techniques are well known in the art to make such alterations.
  • the modulator of MSV expression may act by altering the RNA processing step.
  • RNA processing would alter the ratio of myostatin to MSV.
  • Both a modulator of RNA transcription could be used in conjunction with a modulator of RNA processing to control both the rate at which the native RNA molecule is transcribed and the amount of resulting MSV.
  • the ratio of canonical myostatin and MSV can be altered. In doing so, for example, the inhibitory effect of myostatin on muscle growth can be reduced, and at the same time the MSV muscle growth enhancing effect can be increased.
  • the effect of an interfering RNA molecule can be established by a reduction in the amount of the MSV peptide.
  • the MSV gene expression may also be altered by introducing polynucleotides that interfere with transcription and/or translation.
  • anti-sense polynucleotides could be introduced, which may include; an anti-sense expression vector, anti-sense oligodeoxyribonucleotides, anti-sense phosphorothioate oligodeoxyribonucleotides, anti-sense oligoribonucleotides, anti-sense phosphorothioate oligonucleotides, or any other means that is known in the art, which includes the use of chemical modifications to enhance the efficiency of anti-sense polynucleotides.
  • any anti-sense polynucleotide need not be 100% complementary to the polynucleotides in question, but only needs to have sufficient identity to allow the anti-sense polynucleotide to bind to the gene, or mRNA to disrupt gene expression, without substantially disrupting the expression of other genes.
  • polynucleotides that are complementary to the gene, including 5′ untranslated regions may also be used to disrupt translation of the MSV protein.
  • these complementary polynucleotides need not have 100% complementary, but be sufficient to bind the mRNA and disrupt translation, without substantially disrupting the translation of other genes.
  • the modulation of gene expression may also comprise the use of an interfering RNA molecule as is known in the art, and includes RNA interference (RNAi) and small interfering RNA (siRNA).
  • interfering RNA's Use of interfering RNA's are now well known in the art and suitable interfering RNA's could be designed and tested given the sequences disclosed in the present invention.
  • Use of therapeutic RNA interference is known in the art (Uprichard 2005) as is the use of exon specific interference RNA to alter alternative splicing (Celolto 2002).
  • Modulation of gene expression may also be achieved by the use of catalytic RNA molecules or ribozymes. It is known in the art that such ribozymes can be designed to pair with a specifically targeted RNA molecule. The ribozymes bind to and cleave the targeted RNA.
  • the composition may also include a modulator of MSV activity.
  • a modulator of MSV may include a dominant negative mutant of the polypeptides according to the present invention. A dominant negative effect arises where a mutant acts to block the physiological activity of a wild type protein. This may occur by the dominant negative protein binding to, but not activating, a receptor, while also preventing the wild type protein from binding. Alternatively the dominant negative may act by binding directly to, and inactivating, the wild type protein.
  • the polynucleotides of the present invention can be used to make suitable compositions, or be used to design suitable compositions that regulate MSV gene expression, and thereby regulate muscle growth.
  • Such techniques could be used to regulate MSV gene expression within a cell, for example within a primary or transformed cell line, or to regulate muscle growth within an animal.
  • the modulator of MSV activity may also include a modulator of proteolytic processing of the propeptide.
  • Such a modulator could include propeptide convertase, for example furin endopeptidase, or an agonist or antagonist of propeptide convertases.
  • the muscle cell can be either a primary or transformed cell line, or the cell can be an in vivo cell of a host animal. Suitable host animals may include sheep, cattle, deer, poultry, pigs, fish, horses, mice, rats or humans.
  • One or more compositions of the present invention may also be used for the treatment of diseases associated with muscle tissue.
  • a disease associated with muscle tissue includes any disease or medical condition that involves a change in muscle tissue compared to normal muscle tissue. Such changes may indicate an increase or decrease in muscle mass, or an increase or decrease in muscle fibres.
  • Such diseases may include muscular dystrophy, muscle cachexia, atrophy, hypertrophy and muscle wasting associated with diseases such as cancer or HIV, or amyotrophic lateral sclerosis (ALS).
  • ALS amyotrophic lateral sclerosis
  • Diseases associated with cardiac muscle growth, including infarct are also contemplated. Suitable methods for diagnosing such diseases is well known in the art and can involve both physical examination of a subject or more detailed analysis of a muscle, or other body sample. Such diseases as listed above, or similar such diseases could be diagnosed by a suitable clinician.
  • composition can be administered in any suitable form, which may include oral, topical, inhalation, injection or any other suitable form of administration as known in the art.
  • the composition can be applied for a sufficient time and amount to effect an improvement in the condition.
  • compositions could be used to produce transgenic animals.
  • the compositions could be used to produce transgenic animals having an increase in muscle mass.
  • Suitable animals may include sheep, cattle, deer, poultry, pigs, fish, horses, mice, rats or humans. Many techniques are known in the art for producing transgenic animals, and any suitable method could be used.
  • Another application of the present invention may be to predict the muscle mass of an animal.
  • a sample is obtained from an animal.
  • Any body sample containing a representative amount of MSV will be suitable and can include a blood sample, a biopsy sample, or a sample of muscle tissue.
  • the sample is then analysed for the level of MSV gene expression, or a MSV protein.
  • Many techniques are known in the art for measuring gene expression or protein amount.
  • gene expression can be analysed using quantitative RT-PCR or northern analysis.
  • Protein content can be determined using ELISA [Enzyme-linked Immunosorbant Assay] or Western blot analysis. Any suitable method can be used, for example as set out in any text, for example Maniatis (Molecular Cloning, 2 nd edition, Cold spring Harbor Laboratory Press, 1989).
  • the level of MSV gene expression, or amount of the MSV protein is then compared to an average.
  • An average level of MSV gene expression is the average level obtained from a sample of animals of average muscle mass.
  • the average amount of MSV protein is the amount of protein observed in a sample of animals of average muscle mass.
  • An increased level of MSV gene expression or MSV protein, compared to the average means that the muscle mass of the animal will be predicted to have an above average muscle mass.
  • a decreased level of MSV gene expression or MSV protein, compared to the average means that the muscle mass will be predicted to be less than average.
  • the method may be used to pick animals to be involved in a breeding programme to produce offspring with increased or decreased muscle mass.
  • the invention also provides for polypeptides that are able to preferentially bind any one of the polypeptides according to the present invention.
  • Many such polypeptides are known in the art, which includes but is not limited to antibodies; a non-mammalian antibodies, for example the IgNAR class of antibodies from sharks; bacterial immunity proteins, for example a IMM7 immunity protein from E. coli , or any other class of binding protein known in the art. Given the sequences disclosed in the present specification, a person skilled in the art would be able to produce such a polypeptide or screen a library of known binding polypeptides to obtain a polypeptide that preferentially binds to a polypeptide of the present invention.
  • a polypeptide includes a fragment from such a polypeptide that that confers the preferential binding activity, for example, the variable domain of an antibody.
  • the 65 amino acid peptide (SEQ ID NO: 49) was expressed as a recombinant protein in E. coli (called recombinant ovine myostatin splice variant 65, (roMSV65) and its function was tested 20 in a muscle cell culture system.
  • Northern blot analysis was preformed to identify MSV mRNA species in total or poly(A)+ RNA isolated from tissue. Because ovine MSV was first identified in sheep skeletal muscle via RT-PCR, total RNA was isolated from M. semitendinosus for Northern analysis and subsequently poly(A)+ RNA was purified to eliminate ribosomal RNA associated non-specific binding of radio-labelled DNA probes and to enrich mRNA content. First, a probe complementary to Exon1/2 was used. As shown in FIG. 2 this Ex1/2 probe detected two bands corresponding to canonical myostatin mRNA at 2.9 kb and larger, 4.5 kb mRNA species, which identifies MSV.
  • the 65 amino acid peptide (roMSV65; SEQ ID NO: 49) when added to C 2 Cl 2 myoblasts ( FIG. 3 a ) and primary ovine myoblasts ( FIG. 3B ), is able to stimulate myoblast cell growth in a dose dependent manner. This confirms that MSV is acting as a promoter of muscle cell growth, or an antagonist to myostatin.
  • C 2 Cl 2 myoblasts were treated with roMSV65 during the course of differentiation, and then the levels of myogenin and myosin heavy chain protein, molecular markers of early and late myoblast differentiation were measured by Western immunoblotting.
  • roMSV65 dose-dependently increases myogenin protein levels in C 2 Cl 2 myotubes ( FIG. 4 ). This shows that roMSV65 also induces myoblast differentiation by up-regulating myogenin, a basic helix-loop-helix transcription factor, responsible for the induction of muscle specific genes involved in the terminal differentiation program (Montarras et al. 1991).
  • Myostatin inhibits myoblast differentiation by down-regulating myogenin, MyoD and p21 through a Smad 3 mediated mechanism (McCroskery et al. 2003, Joulia et al. 2003, Langley et al. 2002).
  • MSV65 counteracts myostatin by oppositely regulating myogenin levels in C 2 Cl 2 myotubes.
  • Myosin heavy chain (MHC) proteins are essential structural components of sarcomeres, the contractile units of muscle fibers. These large proteins of 230 kDa are abundantly expressed in myotubes, formed by fusion of terminally differentiated mononucleated myoblasts, and in myofibers of the muscle tissue. As shown in FIG. 5 developmental myosin heavy chain (dMHC) protein expression is greatly up-regulated in response to roMSV treatment in C 2 C 12 myotubes. Interestingly, there is a 18- to 20-fold increase in dMHC abundance at 0.1 and 1 ⁇ g/ml roMSV65 concentrations compared to untreated controls, respectively.
  • dMHC developmental myosin heavy chain
  • myoblasts initially undergo cell division during myogenesis, before withdrawing from the cell cycle. On withdrawing from the cell cycle the myoblasts begin to differentiate into myotubes. Progression through, and arresting from, the cell cycle is controlled by cyclin-dependent kinase and cyclin-dependent kinase inhibitor (CDK/CKI) complexes.
  • CDK/CKI cyclin-dependent kinase and cyclin-dependent kinase inhibitor
  • myostatin As well as down regulating expression of Cdk2, myostatin also upregulates the cyclin-dependent kinase inhibitor p21 thereby inactivating the cyclin/CDK complex, which stimulates progression from G1 to S phase (Thomas et al 2000).
  • PCNA proliferating cell nuclear antigen
  • the fragment excised from normal myostatin mRNA during alternative splicing includes the normal catalytic cleavage site at Arg 266.
  • a potential (KERK/RXXR) cleavage site exists at position 271 to 274 by propeptide convertases (PC1-7) including furin endopeptidase (Steiner 1998).
  • R ⁇ X ⁇ K/R—R motif More common to the TGF- ⁇ family is an R ⁇ X ⁇ K/R—R motif, where K and R are interchangeable, but an R ⁇ X ⁇ K/R—R sequence is optimal (Dubois 2001). Cleavage at position 274, would release a 47 amino acid C-terminal mature MSV fragment from ovine and bovine (ovine: oMSV47, SEQ ID NO: 50 and bovine: bMSV47, SEQ ID NO: 54).
  • Cleavage of the porcine sequence result in the release of 50 amino acid fragments (pMSV50, SEQ ID NO: 72), a 20 amino fragment for human, chimp and cat (h 1-2 MSV20, SEQ ID NOS: 75 and 78; chMSV20, SEQ ID NO: 81; and caMSV20, SEQ ID NO: 93), and a 18 amino acid peptide is released from the dog sequence (d 1-3 MSV18, SEQ ID NOS: 84, 87 and 90).
  • the existence of the mature C-terminal peptide was confirmed using an antibody against the MSV47 peptide ( FIG. 8C ).
  • recombinant ovine MSV47 stimulates murine C 2 C 12 myoblast proliferation.
  • the cell replication curve of FIG. 10 indicates a characteristic dose-response to roMSV47 protein reaching maximum proliferation enhancing activity at about 0.1-0.5 ⁇ g/ml concentration. roMSV47 treatment results in a more than 2-fold increase in myoblast proliferation.
  • roMSV47 The ability of roMSV47 to out-compete myostatin in a C 2 C 12 proliferation assay is shown in FIG. 11 .
  • roMSV47 was able to rescue myostatin inhibited myoblast proliferation at 1:1 (canonical myostatin:roMSV47) molar ratio at 1.5 ⁇ g/ml myostatin concentration. This recovery is greater than that seen for roMSV65, where rescue from myostatin inhibited proliferation required 1:20 molar ratio of myostatin:roMSV65.
  • roMSV47 is a longer peptide than rhMSV38 and has two putative alpha helices (as opposed to the one of rhMSV38) and it is possible that the longer peptide cannot antagonise the actions of myostatin as effectively at higher concentrations due to steric hindrance at cognate receptors.
  • FIG. 12 The ability of recombinant ovine MSV47 (roMSV47) and recombinant human MSV38 (rhMSV38) to stimulate myoblast proliferation is shown in FIG. 12 .
  • FIG. 12A shows the effect of roMSV47 and rhMSV38 on proliferation of murine C 2 C 12 myoblasts over 80 hours
  • FIG. 12B shows the effect on proliferation of human myoblasts over 96 hours.
  • FIG. 13 The ability of rhMSV38 to out-compete myostatin in a human primary myoblast proliferation assay is shown in FIG. 13 .
  • the first of four columns show that rhMSV38 is able to stimulate proliferation of human primary myoblasts in a dose dependent manner.
  • the middle five columns show that rhMSV38 is able to overcome the inhibitory effect of myostatin on the proliferation of the human myoblasts, beyond restoration to the control treatment receiving no myostatin.
  • Restoration was also achieved at double the concentration of myostatin at a 1 ⁇ molar ratio of rhMSV38: myostatin as shown in the final two columns of FIG. 13 .
  • FIG. 14 shows that MSV (roMSV47) suppresses the expression of myostatin both chronically (from seeding to 48 h) and acutely (6 h from 48 to 54 h) in proliferating murine C 2 C 12 myoblasts ( FIG. 14 ).
  • FIG. 15 A comparison of the different sequences is shown in FIG. 15 , with the various fragments shown.
  • Analysis of the sequences using In Silico analysis (GCG software) predicted the presence of two alpha helices separated by a six amino acid residue linker in the mature peptide.
  • human, chimp cat and dog all only contain the first alpha helix, while bovine, ovine and porcine all contain both alpha helices. Because human, chimp cat and dog all only contain the first alpha helix, this implies that the first alpha helix may be responsible for the growth promoting activity of the MSV peptide.
  • a third linker peptide (MSVL18, SEQ ID NO: 100) was also constructed comprising the six C-terminal residues of the first alpha helix, the six linker residues, and the six N-terminal residues of the second alpha helix.
  • MSV ⁇ 1 is able to stimulate muscle proliferation
  • murine C 2 C 1-2 myoblasts were grown in the presence of the peptide.
  • MSV ⁇ 1 greatly stimulated the growth of the C 2 Cl 2 cells in a dose dependent manner.
  • a comparison of the effect of MSV ⁇ 1, MSV ⁇ 2, MSVL18 and roMSV47 on C 2 C 12 proliferation is shown in FIG. 17 . Both roMSV47 and MSV ⁇ 1 were able to stimulate C 2 C 12 cell growth in a dose dependent manner, while MSV ⁇ 2 and MSVL18 had no effect.
  • X 1 is I or L
  • X 2 is V or L
  • X 3 is Y, C, G or S
  • X 4 is I or F
  • X 5 is For L
  • X 6 is G or E
  • X 7 is E or V
  • X 8 is A or T
  • X 9 is A or V
  • X 10 is absent, For L.
  • the ionic charge of all of the amino acid residues in the first alpha helix have a consistent pattern for all of the sequences. That is, where there is a substitution, the different amino acid has been found to be similar in charge. The exception being residue 17 where both ovine and bovine have a hydrophobic residue rather than a polar residue. It will therefore be possible to make further changes in the sequence, retaining the ionic charge of that position, without substantially altering the activity of the peptide. It will be appreciated that such changes are incorporated within the scope of the present invention.
  • the ionic charges for the 19 amino acid alpha helix are as follows:
  • MSV is effective in antagonising that action of myostatin and promoting muscle growth in vitro. Therefore, MSV appears to be a good candidate for the treatment or prophylaxis for muscle wasting conditions.
  • roMSV47 the effect of roMSV47 on muscle wasting in a rat model of cancer cachexia was tested.
  • 21 male rats three months of age were allocated at random into three groups of seven.
  • Group one was injected i.p. with sterile saline (non-cancer controls), groups two and three were inoculated i.p. with 100 ⁇ l of the AH130 tumour (Baracos et al 1995).
  • Groups one and two were injected twice daily s.c.
  • Rats were killed at day six and, at death, skeletal muscles (biceps femoris, gastrocnemius, tibialis anterior, quadriceps femoris, plantaris and soleus) were excised from the right hind limb of each rat and the wet mass expressed as a percent of the initial body mass on day 0.
  • Creatine kinase catalyses the reversible transfer of a high-energy phosphate group between ATP and creatine and is an important energy generating enzyme in skeletal and heart muscle in situations of increased metabolic demand (Wyss and Kaddurah-Daouk 2000).
  • CK When muscle is damaged CK is often released into blood and, therefore, increased concentrations of CK have been used as markers of injury as occurs after myocardial infarct and in muscular dystrophy (Wyss and Kaddurah-Daouk 2000).
  • CK Creatine kinase catalyses the reversible transfer of a high-energy phosphate group between ATP and creatine and is an important energy generating enzyme in skeletal and heart muscle in situations of increased metabolic demand (Wyss and Kaddurah-Daouk 2000).
  • increased concentrations of CK have been used as markers of injury as occurs after myocardial infarct and in muscular dystrophy (Wyss and Kaddurah
  • MSV can act to prevent muscle wasting in pathological conditions. It should also be kept in mind that AH130 is an extremely aggressive cancer model, which induces severe cachexia in a very short period of time (days). It is very surprising that MSV was able to suppress such rapid muscle wasting and therefore MSV is likely to be effective in other less aggressive cancers and muscle wasting conditions.
  • a deletion in myostatin has been found to be the cause of the muscular hypertrophy, commonly referred to as double muscling, observed in the Belgian Blue cattle breed.
  • Belgian Blue cattle have been shown to have an 11-bp deletion in the coding region of mature myostatin causing a frameshift and a premature stop codon in exon 3, which results in the abolition of myostatin activity.
  • the MSV of Belgian Blue cattle (bMSVbb) has also been isolated and characterised (SEQ ID NOS: 96 and 95).
  • the bMSVbb gene contains a 21-nucleotides deletion between nucleotides 749-770 of bMSV (SEQ ID NO: 5).
  • the bMSVbb protein (SEQ ID NO: 95) is 314 amino acids long, seven amino acids shorter than bMSV (SEQ ID NO: 52). However, the deleted seven amino acids does not occur within the unique 65 amino acid C-terminal peptide and therefore the bMSV65 (SEQ ID NO: 53) and bMSV47 (SEQ ID NO: 54) are identical between normal cattle and the Belgian Blue breed. This conservation of the unique 65 amino acid C-terminal peptide indicates a functional importance of the MSV protein.
  • MSV-specific polyclonal antibodies were developed in rabbits using oligopeptides specific to the propeptide (N-terminal) and mature (C-terminal) region of ovine MSV.
  • the localization of oligopeptides is based on the proposed processing of the precursor MSV at the KERK amino acid motif by propeptide convertases (PC1-7) including furin endopeptidase (Steiner 1998).
  • the MSV propeptide-specific antibody (MPSA) can identify MSV precursor (37 kDa) and propeptide (29 kDa).
  • the mature MSV-specific antibody (MMSA) is able to detect precursor (37 kDa) and mature (5.4 kDa monomer or a putative 11 kDa homo-dimer) MSV.
  • Western immunoblotting employing MPSA consistently detects the precursor and propeptide of MSV in ovine skeletal muscle and brain but their ratio was different in these tissues ( FIG. 8A ). This may suggest that the efficiency of propeptide processing is vastly different in muscle and brain.
  • the PSA also identified MSV orthologs in human, pig, mouse and rat muscle and/or brain. There are other immunoreactive bands on the Western blot, which may be cleavage products of MSV precursor or covalently linked complexes of MSV proteins (Jiang et al. 2004). MMSA identifies the precursor protein in ovine muscle and brain but either the matured monomer (5.4 kDa) or homo-dimer (11 kDa) is undetectable ( FIG. 8B ).
  • FIG. 8B also shows immunoreactive bands at 11-12 kDa corresponding to the homo-dimers of mature MSV in human and rat muscle. This shows that MSV protein expression and processing also occurs in species other than sheep and cattle. MMSA identified mature MSV protein in a range of sheep serum samples ( FIG. 8 c ). These data show that:
  • MSV has been shown to be a potent antagonist of mature myostatin in myoblast replication and differentiation in vitro and able to reduce cancer muscle cachexia in vivo.
  • MSV proteins have been detected in muscle of sheep and other species (cattle, human, pig, rat and mouse) and their abundance differ in genders and different physiological states. Mature MSV is also present in circulation in sheep.
  • both the oMSV and hMSV have been shown to be active in cells from different species.
  • Ovine myostatin spice variant was first identified by reverse transcription polymerase chain reaction (RT-PCR) using flanking PCR primers around the open reading frame (ORF) of myostatin cDNA (complementary DNA sequence to myostatin messenger RNA).
  • the forward primer (5′-TCAGACTGGGCAGGCATTAACG-3′, SEQ ID NO: 101) located in the 5′ untranslated region (UTR) and the reverse primer (5′-GCATATGGAGTTTTAAGACCA-3′, SEQ ID NO: 102) in the distant 3′UTR.
  • the PCR reaction was carried out with 2 ⁇ l of ovine muscle cDNA (reverse transcribed from mRNA of Texel sheep Muscle semitendinosus) as a template at 94° C.
  • PCR product was gel-purified using the Perfect Prep kit (Eppendorf, Germany) and cloned into the pGEM-T Easy E. coli plasmid vector according to the manufacturer's instructions (Promega, USA).
  • the plasmid clone was given the name of pFJ106/3 and the insert was analysed at the Waikato DNA Sequencing Facility (Hamilton, New Zealand) by bi-directional sequencing from the T7 and Sp6 primers.
  • the complete insert of pFJ106/3 was assembled in GSG/SeqLab software (Accelrys Inc., USA, SEQ ID NO: 1).
  • the completed sequence was then aligned with the canonical cDNA sequence of myostatin (GenBank accession number: AF019622).
  • the alignment revealed 100% DNA sequence homology from nucleotide 1 (translational start site ATG in exon 1) to nucleotide 770 (exon 3) with the canonical myostatin cDNA, except a single silent mutation at position of 435 nucleotide (G ⁇ A transition).
  • the alignment identified a novel translated DNA region of 195 nucleotides and a novel translational stop site (TAA).
  • oMSV ovine myostatin splice variant
  • the ORF of oMSV encodes for a 321 amino acid protein (SEQ ID NO: 48).
  • Amino acids 1 to 257 are identical with the canonical myostatin propeptide or LAP sequence.
  • amino acids 257 to 321 (65aa) forms a novel C-terminal polypeptide sequence of oMSV dissimilar to canonical myostatin protein.
  • Protein sequence and Western blot analysis using oMSV-specific antibody suggest that the 321aa full length oMSV protein may be processed at amino acid 275 following a KERK motif giving rise to a 274aa oMSV propeptide or LAP and a 47aa mature oMSV polypeptide.
  • Bovine myostatin splice variants were identified with RT-PCR, and subsequent cloning and sequencing of the full-length ORF.
  • the forward primer (5′-ACCATGGAAAAACTCCAAATCTTT-3′, SEQ ID NO: 103) located at the translational start site of bovine myostatin and the reverse primer (5′-GTCATCGTCATCTTTCATCCTAAAAGCTGCAGT-3′, SEQ ID NO: 104) at the predicted translational stop site of bMSV.
  • the PCR reaction was carried out with 211 of bovine muscle cDNA (reverse transcribed from mRNA of fetal Muscle semitendinosus of Hereford/Friesian cross or the double-muscled Belgian Blue cattle) as a template at 94° C. for 2 min for pre-amplification denaturation, and then at 94° C. for 30 sec, 55° C. for 30 sec, and 68° C. for 1 min for 45 cycles.
  • the PCR products were gel-purified using the Perfect Prep kit (Eppendorf, Germany) and cloned into the pMTNV5-His-TOPO plasmid vector according to the manufacturer's instructions (Invitrogen, California).
  • the inserts were analysed at the Waikato DNA Sequencing Facility (Hamilton, New Zealand) by bi-directional sequencing from the MTF1 and BGH primers. The complete insert sequences were assembled in GSG/SeqLab software (SEQ ID NO: 4).
  • the completed sequence bMSV (SEQ ID NO: 4) was aligned with the canonical cDNA sequence of myostatin (GenBank accession number: AF019620).
  • the alignment revealed 100% DNA sequence homology from nucleotide 1 (translational start site ATG in exon 1) to nucleotide 770 (exon 3) with the canonical myostatin cDNA for the Hereford/Friesian cross, and 100% DNA sequence homology from nucleotide 1 (translational start site ATG in exon 1) to nucleotide 749 (exon 3) with the canonical myostatin cDNA for Belgian Blue cattle. It also identified a novel translated DNA region of 195 nucleotides and a novel translational stop site (TAA) in both cattle breeds.
  • TAA novel translational stop site
  • the ORF of bMSVh/f encodes for a 321 amino acid protein (SEQ ID NO: 52), but the ORF of bMSVbb encodes for a 7aa shorter 314aa protein (SEQ ID NO: 95) but the rest of the protein sequence shows complete homology in the two breeds examined. Interestingly, the unique 65aa C-terminal peptide (SEQ ID NO: 53) is conserved in bMSVbb, which may indicate functional importance of this protein sequence region. Amino acids 1 to 257 in bMSVh/f racket SEQ ID NO: 52) and 1 to 250 in bbMSVbb (SEQ ID NO: 95) are identical with the canonical myostatin propeptide or LAP sequence.
  • amino acids 257 to 321 (65aa) in bMSVh/f (SEQ ID NO: 52) and 250 to 314 in bMSVbb (SEQ ID NO: 95) represent a novel C-terminal polypeptide sequence of bMSV dissimilar to canonical bovine myostatin protein.
  • Protein sequence and Western blot analysis using oMSV-specific antibody suggest that the 321 aa full length bMSVh/f (314aa full length bMSVbb) protein may be processed at amino acid 275 (268) following a RERK motif giving rise to a 274aa bMSVh/f (267aa bMSVbb) propeptide or LAP and the same 47aa mature bMSVh/f or bMSVbb polypeptide (SEQ ID NO: 54).
  • RNA Northern blot analysis was employed. Frozen semitendinosus muscles of six adult male Romney sheep were homogenized on ice in Trizol Reagent (Invitrogen, California) for 30 seconds at 13 500 rpm using an Ultra Turrax homogenizer (Janke & Kunkel GmbH, Germany). Debris was removed by centrifugation for 10 minutes at 10,000g and total RNA was isolated following the manufacturer's protocol (Invitrogen, California). RNA was re-suspended in diethyl pyrocarbonate-treated water and the total RNA concentration determined by measuring absorbance at 260 nm.
  • RNA About 600 microgram of total RNA were pooled and poly(A)+ RNA was purified with an mRNA Purification kit (Amersham Biosciences, UK) according to the manufacturer's instructions. Ten microgram of total and five microgram of poly(A)+ RNA were separated on a 1.2% formaldehyde-agarose gel, then transferred to uncharged nylon membrane (Hybond-N, Amersham Biosciences, UK) by capillary action. Membranes were cross-linked using ultra-violet radiation and stained with methylene blue to verify the integrity of RNA and the uniformity of transfer.
  • RT-PCR reverse transcription polymerase chain reaction
  • 5 microgram of total RNA from ovine skeletal muscle was reverse transcribed using a Superscript II Pre-Amplification kit (Invitrogen, California) according to the manufacturer's protocol.
  • PCR was carried out with 2 ul of the reverse transcriptase reaction (94° C. for 30 sec, 55° C. for 1 min and 72° C. for 1 min) for 35 cycles and a final extension of 5 min at 72° C.
  • Oligonucleotide primers homologous to exon1/2 of ovine myostatin were: 5′-ATGCAAAAACTGCAAATCTCTG-3′ (SEQ ID NO: 116) (forward, nt 1-22) and 5′-ATCAATGCTCTGCCAAATACC-3′ (SEQ ID NO: 117) (reverse, nt 601-621).
  • Two primer pairs were used to identify mRNA species having complementary sequences to the far 3′ untranslated region (3′UTR) of ovine myostatin gene. These probes were called MSV1 and MSV2 could not hybridise to the canonical ovine myostatin mRNA (2.9 kb).
  • PCR primers were the following: 5′-GCAAGGGTATATGGTCCTAGAG-3′ Exon1/2 oMSV P1 (SEQ ID NO: 118) (forward, nt 2911-2932, MSV1), 5′-CACCAGAGAGAATTAGTCACTG-3′ (SEQ ID NO: 119) (reverse, nt 3177-3198, MSV1), 5′-TAAAAGTCTGGGTCAGCAG-3′ (SEQ ID NO: 120) (forward, nt 3461-3479, MSV2) and 5′-GCAAAATAGGGGGGGAAATG-3′ (SEQ ID NO: 121) (reverse, nt 3731-3750, MSV2).
  • Radio-labeled cDNA probes were prepared using [ ⁇ -32P]dCTP and the Rediprime II Labeling Kit (Amersham Biosciences, UK) according to the manufacturer's instructions.
  • the membrane was pre-hybridized in Church and Gilbert buffer for two hours (0.5 M Na2HPO4 pH 7.2, 7% SDS, 1 mM EDTA) at 55° C.
  • the membrane was then hybridized at 55° C. overnight in fresh Church and Gilbert buffer with the appropriate radio-labeled probe. Following hybridization, they were washed at 55° C.
  • ovine MSV65 (aa 257-321), ovine MSV47 (aa 275-321), ovine MSV ⁇ 1 helix (aa 275-293), ovine MSV interlinking sequence (aa 287-305), ovine MSV ⁇ 2 helix (aa 300-321), human MSV38 (aa 257-298).
  • the appropriate DNA inserts for each were cloned, expressed and purified as a recombinant protein in E. coli . Pooled cDNA of human or sheep (Romney) skeletal muscle was used as a template to amplify MSV DNA inserts by polymerase chain reaction (PCR).
  • PCR polymerase chain reaction
  • ovine MSV65 (aa 257-321) 5′-CACCGTGCATTTTTACACTCCTCCCT-3′ (SEQ ID NO: 122), 5′-TTATTTCATCCTAAAAGCTGCAG-3′ (SEQ ID NO: 123); ovine MSV47 (aa 275-321) 5′-CACCATCATTTTTCTAGAGGTCTAC-3′ (SEQ ID NO: 124), 5′-TTATTTCATCCTAAAAGCTGCAG-3′ (SEQ ID NO: 125); ovine MSV ⁇ 1 helix (aa 275-297) 5′-CACCATCATTTTTCTAGAGGTCTAC-3′ (SEQ ID NO:106), 5′-TTATGACTGCCTTTTAAACACAGC-3′ (SEQ ID NO:107); ovine MSV interlinking sequence (aa 287-306) 5′-CACCATACTTGGAGAAGCTGTGTTT-3′ (SEQ ID NO: 126), 5′-TTAGAAATTTTGACAAAAATGA
  • PCR was carried out with 2 ⁇ l ovine muscle cDNA at 94° C. for 2 min as a pre-amplification denaturation, and then at 94° C. for 30 sec, 55° C. for 30 sec, and 72° C. for 30 sec for 35-45 cycles.
  • the PCR product was gel-purified using the Perfect Prep kit (Eppendorf, Germany) and cloned into pET100/D-TOPO (Invitrogen, California) E. coli protein expression vector according to the manufacturer's instructions.
  • the protein expression construct contains a 36 amino acid N-terminal tag including an Enterokinase cleavage site, Xpress epitope and a poly-His sequence.
  • the resulting plasmid DNA construct was sequenced at the Waikato DNA Sequencing Facility (Hamilton, New Zealand) to confirm sequence identity. Twenty-five nanograms of plasmid DNA was transformed into BL21 A1 or BL21 Star E. coli chemically competent protein expression E.
  • Protein expressing cells were collected by centrifugation of the culture at 6000 ⁇ g for 15 min at 4° C., and washed with distilled water. The E. coli cells were then frozen at ⁇ 80° C. The following day the cells (5g) were lysed in 25 ml of lysis buffer (25 mM NaHPO 4 pH 8.0, 500 mM NaCl, 250 ⁇ l protease inhibitor [P8849, Sigma, Missouri]) and mixed for 30 min at 4° C. on a rotating wheel. The lysate was then freeze/thawed in a dry ice/ethanol bath.
  • lysis buffer 25 mM NaHPO 4 pH 8.0, 500 mM NaCl, 250 ⁇ l protease inhibitor [P8849, Sigma, Missouri
  • the lysate was sonicated 10 times at an amplitude of 80 with 1 sec bursts for 1 min with 1 min breaks in between on wet ice and then passed through an 18-gauge needle to break genomic DNA.
  • Cell debris was removed by centrifugation at 16000 ⁇ g for 20 min at 4° C. and the supernatant incubated with 3 ml of Ni—NTA agarose resin with gentle shaking for 1 hour at 4° C.
  • the lysate/resin mixture was applied to a column and the lysate allowed to flow through. The remaining resin was washed four times with 10 ml of wash buffer (25mM NaHPO 4 pH 7.0, 500 M NaCl, 20 mM imidazole).
  • the recombinant protein was eluted from the resin with 5 ml elution buffer (25 mM NaHPO 4 pH 7.0, 500 mM NaCl, 250 mM imidazole) and dialysed in a SnakeSkin dialysis tube (MWCO 3,500, Pierce, Illinois) against 500 ml of dialysis buffer (20 mM TRIS-HCl pH 7.0, 150 mM NaCl) at 4° C. overnight. To remove endotoxins, the protein was then mixed with 25 ⁇ l of polymixin (BioRad) and incubated overnight on a rotating wheel.
  • 5 elution buffer 25 mM NaHPO 4 pH 7.0, 500 mM NaCl, 250 mM imidazole
  • MWCO 3500, Pierce, Illinois 500 ml of dialysis buffer (20 mM TRIS-HCl pH 7.0, 150 mM NaCl
  • the polymixin was removed from the sample using a column and the resulting protein was dialyzed further two times in dialysis buffer (20 mM TRIS-HCl pH 7.0, 150 mM NaCl).
  • the endotoxin-free recombinant protein solution was filter sterilized by passing it through a 0.22cm filter unit (MillexGS, Millipore, Ireland).
  • the protein concentration was determined using the bicinchoninic acid (BCA, Sigma, Missouri) protein assay. Size, purity and identity of the recombinant protein were verified by Western blot analysis and SDS-PAGE followed by Coomassie Brillant Blue staining, and its bioactivity was tested in a murine C 2 C 12 or human primary myoblast proliferation assay.
  • C 2 Cl 2 murine and primary ovine myoblasts were employed to demonstrate that MSV promotes the proliferation of muscle cells.
  • Myoblasts were seeded in 96-well tissue culture plates (Nunc, Roskilde, Denmark) at a cell density of 1000 cells per well in 200,l of Dulbecco's modified Eagle's medium (DMEM, Invitrogen, California) supplemented with foetal bovine serum (FBS, 10% v/v, Invitrogen), penicillin (1 ⁇ 10 5 IU/l, Sigma, St Louis, Mo., USA) and streptomycin (100 mg/l, Sigma) buffered with NaHCO 3 containing phenol red (7.22 nmol/l) as a pH indicator, and incubated overnight at 37° C.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS foetal bovine serum
  • penicillin 1 ⁇ 10 5 IU/l
  • Sigma St Louis, Mo., USA
  • streptomycin 100 mg/l
  • p21 is a cycle-dependent kinase inhibitor, which regulates cell cycle progression by inducing G1 arrest and block entry into S phase by inactivating Cdks or by inhibiting activity of proliferating cell nuclear antigen (PCNA).
  • PCNA proliferating cell nuclear antigen
  • p21 protein expression may indicate an increased cell proliferation rate.
  • PCNA is a positive marker of cell proliferation. It is a subunit of DNA polymerase-delta during DNA replication in the cell cycle. Higher level of PCNA protein expression is associated with higher number of cells entering the DNA replication phase of the cell cycle.
  • C 2 C 12 myoblasts were seeded in 10 cm diameter tissue culture plates (Nunc, Roskilde, Denmark) at a cell density of 3000 cells per cm 2 in 10 ml of Dulbecco's modified Eagle's medium (DMEM, Invitrogen, California) supplemented with foetal bovine serum (FBS, 10% v/v, Invitrogen), penicillin (1 ⁇ 105 IU/l, Sigma, St Louis, Mo., USA) and streptomycin (100 mg/l, Sigma) buffered with NaHCO 3 containing phenol red (7.22 nmol/l) as a pH indicator, and incubated overnight at 37° C. with 5% CO 2 .
  • DMEM Dulbecco's modified Eagle's medium
  • FBS foetal bovine serum
  • penicillin 1 ⁇ 105 IU/l
  • Sigma St Louis, Mo., USA
  • streptomycin 100 mg/l, Sigma
  • the plates were then incubated at 37° C. with 5% CO 2 for 48 hours. Cells were removed from the plate by trypsin treatment, washed with phosphate buffered saline (PBS, Oxoid, UK) and resuspended in 100 ⁇ l of lysis buffer (10 mM Hepes pH 7.9, 1.5 mM MgCl 2 , 10 mM KCl, 0.5% NP40, one Complete protease inhibitor tablet [Roche Diagnostics, USA] per 50 ml buffer).
  • lysis buffer 10 mM Hepes pH 7.9, 1.5 mM MgCl 2 , 10 mM KCl, 0.5% NP40, one Complete protease inhibitor tablet [Roche Diagnostics, USA] per 50 ml buffer.
  • the protein concentration was estimated using the bicinchoninic acid (BCA, Sigma, Missouri) protein assay. Twenty micrograms of total protein was separated by SDS-PAGE (12%) and transferred to nitrocellulose membrane by electroblotting. The blots were stained with Ponceau S stain to check equal loading. After washing the blot in TBST buffer they were blocked in TBST/5% non-fat milk for at least one hour and then incubated with 1:1000 dilution of mouse monoclonal anti-p21 (BD Pharmigen) or 1:500 dilution of rabbit polyclonal anti-PCNA (sc-7907, Santa Cruz) primary antibodies at 4° C. overnight.
  • BCA bicinchoninic acid
  • the membranes were washed with TBST (5 ⁇ 5 min) and incubated with either 1:2000 dilution of rabbit anti-mouse IgG-HRP (P0161, DAKO) or 1:2000 dilution of goat anti-rabbit IgG-HRP (P0448, DAKO) secondary antibodies at room temperature for 1 hour.
  • the membranes were washed again with TBST (5 ⁇ 5 min) and developed with enhanced chemiluminescence. Band intensities were measured with a GS800 densitometer (BioRad, USA).
  • C 2 C 12 myoblast cultures were employed. Levels of well-established early and late molecular markers of myogenic differentiation like myogenin and dMHC were measured in the presence and absence of roMSV65 protein.
  • C 2 C 12 myoblasts were seeded in 10 cm diameter tissue culture plates (Nunc, Roskilde, Denmark) at a cell density of 25,000 cells per cm 2 in 10 ml of Dulbecco's modified Eagle's medium (DMEM, Invitrogen, California) supplemented with foetal bovine serum (FBS, 10% v/v, Invitrogen), penicillin (1 ⁇ 10 5 IU/l, Sigma, St Louis, Mo., USA) and streptomycin (100 mg/l, Sigma) buffered with NaHCO 3 containing phenol red (7.22 nmol/l) as a pH indicator, and incubated overnight at 37° C. with 5% CO 2 .
  • DMEM Dulbecco's modified Eagle's medium
  • FBS foetal bovine serum
  • penicillin 1 ⁇ 10 5 IU/l
  • Sigma St Louis, Mo., USA
  • streptomycin 100 mg/l, Sigma
  • the blots were stained with Ponceau S stain to check equal loading. After washing the blot in TBST buffer they were blocked in TBST/5% non-fat milk for myogenin, or in TBS buffer supplemented with 1% PVP-10, 1% PEG 4000, 0.3% BSA, 0.1% Tween 20 for dMHC and MHC for at least one hour and then incubated with the following primary antibodies: 1:1000 dilution of rabbit polyclonal anti-myogenin (sc-576, Santa Cruz Biotechnology, Calif.), 1:500 dilution of mouse monoclonal anti-rat dMHC (Novocastra Laboratories, Newcastle upon Tyne, UK) at 4° C. overnight.
  • rabbit polyclonal anti-myogenin sc-576, Santa Cruz Biotechnology, Calif.
  • mouse monoclonal anti-rat dMHC Novocastra Laboratories, Newcastle upon Tyne, UK
  • the membranes were washed with TBST (5 ⁇ 5 min) and incubated with either 1:2000 dilution of rabbit anti-mouse IgG-HRP (P0161, DAKO) or 1:2000 dilution of goat anti-rabbit IgG-HRP (P0448, DAKO) secondary antibodies at room temperature for 1 hour.
  • the membranes were washed again with TBST (5 ⁇ 5 min) and developed with enhanced chemiluminescence. Band intensities were measured with a GS800 densitometer (Bio-Rad, CA).
  • roMSV65 was carried out with C 2 Cl 2 murine myoblasts.
  • Myoblasts were seeded in 96-well tissue culture plates (Nunc, Roskilde, Denmark) at a cell density of 1000 cells per well in 2001p of Dulbecco's modified Eagle's medium (DMEM, Invitrogen, California) supplemented with foetal bovine serum (FBS, 10% v/v, Invitrogen), penicillin (1 ⁇ 10 5 IU/l, Sigma, St Louis, Mo., USA) and streptomycin (100 mg/l, Sigma) buffered with NaHCO 3 containing phenol red (7.22 mmol/l) as a pH indicator, and incubated overnight at 37° C.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS foetal bovine serum
  • penicillin 1 ⁇ 10 5 IU/l
  • Sigma St Louis, Mo., USA
  • streptomycin 100 mg/l, Sigma
  • PBS phosphate buffered saline
  • the fixative was then removed and 100 ⁇ l of 1% methylene blue stain in 0.01 M borate buffer (pH 8.5) was added to each well and incubated for 30 min at room temperature. Excess stain was removed by four sequential washes in borate buffer. Methylene blue was eluted from the cells by the addition of 100 ⁇ l 1:1 (v/v) ethanol/0.1HCl. The plates were gently shaken and the absorbance was measured at 655 nm using a microplate photometer (VersaMax, Molecular Devices, CA).
  • MSV-Specific Antibodies and its Use for Detection of MSV Proteins in Tissues and Blood
  • Oligopeptides located at C-terminus of ovine MSV were synthesized, purified and conjugated to keyhole limpet haemocyanin (KLH) by Auspep (Parkville, Australia) for immunizations.
  • KLH keyhole limpet haemocyanin
  • the amino acid sequences of the oligopeptides were: CYTPPYGQWIFHKERK (aa 260-274 (SEQ ID NO: 1, MSV-FJ1) and CKRQSKSIHFGQNFK (aa 294-307 (SEQ ID NO: 1, MSV-FJ3).
  • Immunoglobulin-G was purified in two steps; first it was enriched by ammonium sulphate precipitation and further purified using a Protein A column.
  • the protein pellet was re-solubilised in 10 ml of 0.1 M Tris pH 8.0 and loaded on to an equilibrated Protein A column (P9424, Sigma, USA).
  • the column was washed with 20 ml of 0.1 M glycine pH 8.0 and the IgG was eluted from the column with 10 ml of 0.1 M glycine pH 4.0, followed by 10 ml of 0.1 M glycine pH 3.0.
  • 0.5 ml fractions were collected and mixed with 100 ⁇ l of 1 M Tris pH 8.0 on ice.
  • Protein concentration was estimated in each fraction using the BCA protein assay (Sigma, USA). Protein containing fractions were pooled and dialysed against 2 litres of 0.1 M NaHCO 3 , 0.5 M NaCl. After dialysis, protein concentration was determined with the BCA protein assay (Sigma, USA) and the antibody solution was mixed 1:1 with glycerol to protect IgG from freeze damage, and stored at ⁇ 20° C. The purified IgG was tested using a range of dilutions (1:300 to 1:30,000) as a primary antibody in Western immunoblotting.
  • MSV-specific antibodies allow the detection of precursor (37 kDa), pro-peptide (29 kDa) and mature (5.4 kDa) MSV proteins in ovine, bovine and possibly in other species.
  • the antibodies can be used for Western blotting, immunoprecipitation, hystochemistry, cytochemistry and ELISA assays to comparatively measure, localise and quantify MSV proteins in complex protein extracts, cells, tissues and blood samples.
  • tissue for example muscle or brain
  • lysis buffer 10 mM Hepes pH 7.9, 1.5 mM MgCl2, 10 mM KCl, 0.5% NP40, one Complete [Roche Diagnostics, USA] protease inhibitor tablet per 50 ml buffer
  • the homogenate was centrifuged at 10,000 ⁇ g for 5 min at 4° C. to remove tissue debris.
  • the protein concentration of the supernatant was estimated using the bicinchoninic acid (BCA, Sigma, Missouri) protein assay with BSA standard.
  • Protein extracts were mixed 2:1 with 3 ⁇ Laemmli sample (6% SDS, 15% 2-mercaptoethanol, 30% glycerol, 187.5 mM Tris pH 6.8, 0.05% bromophenol blue buffer and incubated in boiling water for 5 min. 20 ⁇ g of total protein was separated by SDS-PAGE (15% for mature MSV or 10% for MSV propeptide) and transferred to nitrocellulose membrane by electroblotting.
  • serum was mixed 1:1:1 with sterile water and 3 ⁇ Laemmli sample buffer and incubated in boiling water for 5 min. Serum proteins equivalent to 2 ⁇ l neat serum was separated by 15% SDS-PAGE and transferred to nitrocellulose membrane by electroblotting.
  • the blots were stained with Ponceau S stain to check equal loading. After washing the blots in TBST buffer they were blocked in TBS buffer supplemented with 1% PVP-10, 1% PEG 4000, 0.3% BSA, 0.1% Tween 20 for at least one hour and then incubated with MSV-specific primary antibodies (MPSA or MMSA) at 1:1000 dilution in the above blocking buffer at 4° C. overnight. The membranes were washed with TBST (5 ⁇ 5 min) and incubated with either 1:5000 dilution of goat anti-rabbit IgG-HRP (P0448, DAKO) secondary antibody at room temperature for 2 hour. The membranes were washed again with TBST (5 ⁇ 5 min) and developed with enhanced chemiluminescence.
  • MSV-specific primary antibodies MPSA or MMSA
  • oMSV47 To test the biological effect of oMSV47 on myoblats proliferation, it was cloned, expressed and purified as a recombinant protein in E. coli .
  • Pooled sheep (Romney) skeletal muscle cDNA was used as a template to amplify oMSV47 by polymerase chain reaction (PCR).
  • the PCR product was obtained with the following forward and reverse primers: 5′-CACCATCATTTTTCTAGAGGTCTAC-3′ (SEQ ID NO: 106) and 5′-TTATTTCATCCTAAAAGCTGCAG-3 (SEQ ID NO: 107). PCR was carried out with 2 ⁇ l ovine muscle cDNA at 94° C.
  • the PCR product was gel-purified using the Perfect Prep kit (Eppendorf, Germany) and cloned into pET100/D-TOPO (Invitrogen, California) E. coli protein expression vector according to the manufacturer's instructions.
  • the protein expression construct contains a 36 amino acid N-terminal tag including an Enterokinase cleavage site, Xpress epitope and a hexa-His sequence.
  • the resulting pFJMSV47.3/8 plasmid DNA construct was sequenced at the Waikato DNA Sequencing Facility (Hamilton, New Zealand) to confirm sequence identity. 46 ng of pFJMSV47.3/8 plasmid DNA was transformed into BL21 Star chemically competent protein expression E.
  • the E. coli cells were lysed in 100 ml of lysis buffer (6M guanidine HCl, 20 mM NaHPO 4 pH 7.8, 500 mM NaCl, 5 mM 2-mercaptoethanol, Complete protease inhibitor [Roche Diagnostics, USA]) and sonicated on ice to complete cell lysis.
  • the E. coli cell lysate was passed through an 18-gauge needle five times to break genomic DNA. Cell debris was removed by centrifugation at 3000 ⁇ g for 15 min at 4° C. The lysate was then incubated with 5 ml of Ni—NTA agarose resin with gentle shaking for 1 hour at room temperature.
  • the resin was separated from the cell lysate and washed twice with 10 ml of denaturing binding buffer (8M urea, 20 mM NaHPO 4 pH 7.8, 500 mM NaCl, 5 mM 2-mercaptoethanol). The resin was washed a further four times with denaturing wash buffer (8M urea, 20 mM NaHPO4 pH 6.0, 500 mM NaCl, 5 mM 2-mercaptoethanol).
  • denaturing binding buffer 8M urea, 20 mM NaHPO 4 pH 7.8, 500 mM NaCl, 5 mM 2-mercaptoethanol.
  • the recombinant protein was eluted from the resin with 10 ml denaturing elution buffer (8M urea, 20 mM NaHPO 4 pH 4.0, 500 mM NaCl, 5 mM beta-mercaptoethanol) and dialysed in a SnakeSkin dialysis tube (MWCO 3,500, Pierce, Ill.) against 500 volume of dialysis buffer (20 mM TRIS-HCl pH 8.5, 150 mM NaCl) at 4° C. for 24 h.
  • the protein concentration of the dialysed protein was determined using the bicinchoninic acid (BCA, Sigma, Missouri) protein assay. Size, purity and identity of the recombinant protein were verified by Western blot analysis, and its bioactivity was tested in a murine C 2 Cl 2 myoblast proliferation assay.
  • C 2 C 12 murine and primary ovine myoblasts were employed to demonstrate that roMSV47 promotes the proliferation of muscle cells.
  • Myoblasts were seeded in 96-well tissue culture plates (Nunc, Roskilde, Denmark) at a cell density of 1000 cells per well in 200 ⁇ l of Dulbecco's modified Eagle's medium (DMEM, Invitrogen, California) supplemented with foetal bovine serum (FBS, 10% v/v, Invitrogen), penicillin (1 ⁇ 10 5 IU/l, Sigma, St Louis, Mo., USA) and streptomycin (100 mg/l, Sigma) buffered with NaHCO 3 containing phenol red (7.22 nmol/l) as a pH indicator, and incubated overnight at 37° C.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS foetal bovine serum
  • penicillin 1 ⁇ 10 5 IU/l
  • Sigma St Louis, Mo., USA
  • roMSV47 was carried out with C 2 C 12 murine myoblasts.
  • Myoblasts were seeded in 96-well tissue culture plates (Nunc, Roskilde, Denmark) at a cell density of 1000 cells per well in 200 ⁇ l of Dulbecco's modified Eagle's medium (DMEM, Invitrogen, California) supplemented with foetal bovine serum (FBS, 10% v/v, Invitrogen), penicillin (1 ⁇ 10 5 IU/l, Sigma, St Louis, Mo., USA) and streptomycin (100 mg/l, Sigma) buffered with NaHCO 3 containing phenol red (7.22 nmol/l) as a pH indicator, and incubated overnight at 37° C.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS foetal bovine serum
  • penicillin 1 ⁇ 10 5 IU/l
  • Sigma St Louis, Mo., USA
  • streptomycin 100 mg/l, Sigma
  • PBS phosphate buffered saline
  • the fixative was then removed and 100 ⁇ l of 1% methylene blue stain in 0.01 M borate buffer (pH 8.5) was added to each well and incubated for 30 min at room temperature. Excess stain was removed by four sequential washes in borate buffer. Methylene blue was eluted from the cells by the addition of 100 ⁇ l 1:1 (v/v) ethanol/0.1HCl. The plates were gently shaken and the absorbance was measured at 655 nm using a microplate photometer (VersaMax, Molecular Devices, CA).
  • C 2 C 12 murine myoblasts were employed to demonstrate that the respective MSV peptides promote the proliferation of muscle cells.
  • Myoblasts were seeded in 96-well tissue culture plates (Nunc, Roskilde, Denmark) at a cell density of 1000 cells per well in 200 ⁇ l of Dulbecco's modified Eagle's medium (DMEM, Invitrogen, California) supplemented with foetal bovine serum (FBS, 10% v/v, Invitrogen), penicillin (1 ⁇ 10 5 IU/l, Sigma, St Louis, Mo., USA) and streptomycin (100 mg/l, Sigma) buffered with NaHCO 3 containing phenol red (7.22 nmol/l) as a pH indicator, and incubated overnight at 37° C.
  • DMEM Dulbecco's modified Eagle's medium
  • FBS foetal bovine serum
  • penicillin 1 ⁇ 10 5 IU/l
  • Sigma St Louis, Mo., USA
  • streptomycin 100 mg/
  • the fixative was then removed and 100 ⁇ l of 1% methylene blue stain in 0.01 M borate buffer (pH 8.5) was added to each well and incubated for 30 min at room temperature. Excess stain was removed by four sequential washes in borate buffer. Methylene blue was eluted from the cells by the addition of 100 ⁇ l 1:1 (v/v) ethanol/0.1HCl. The plates were gently shaken and the absorbance was measured at 655 nm using a microplate photometer (VersaMax, Molecular Devices, CA).
  • rhMSV38 and roMSV47 Stimulate the Proliferation of Human Primary Myoblasts.
  • rhMSV38 and roMSV47 are able to stimulate the replication of human skeletal muscle cells, they were tested in a proliferation assay.
  • Human skeletal muscle myoblasts (Cambrex, Australia) were seeded in 96-well tissue culture plates (Nunc, Roskilde, Denmark) at a cell density of 2000 cells per well in 2001p of Skeletal Muscle Basal Medium (Cambrex, Australia) supplemented with foetal bovine serum (FBS, 10% v/v, Cambrex, Australia), and incubated overnight at 37° C. with 5% CO 2 .
  • FBS foetal bovine serum
  • DMEM Dulbecco's modified Eagle's medium
  • FBS foetal bovine serum
  • penicillin 1 ⁇ 10 5 IU/l
  • streptomycin 100 mg/l, Sigma
  • NaHCO 3 containing phenol red (7.
  • the wells were washed with 200 ⁇ l of phosphate buffered saline (PBS, Oxoid, UK) buffer and fixed with 100 ⁇ l of 10% formaldehyde, 0.9% NaCl solution for at least one hour. The fixative was then removed and 100 ⁇ l of 1% methylene blue stain in 0.01 M borate buffer (pH 8.5) was added to each well and incubated for 30 min at room temperature. Excess stain was removed by four sequential washes in borate buffer. Methylene blue was eluted from the cells by the addition of 100 ⁇ l 1:1 (v/v) ethanol/0.1HCl. The plates were gently shaken and the absorbance was measured at 655 nm using a microplate photometer (VersaMax, Molecular Devices, CA).
  • roMSV47 Outcompetes Mature Myostatin in a Proliferation Assay of Murine C 2 C 12 myoblasts.
  • Myoblasts were seeded in 96-well tissue culture plates (Nunc, Roskilde, Denmark) at a cell density of 1000 cells per well in 200 ⁇ l of Dulbecco's modified Eagle's medium (DMEM, Invitrogen, California) supplemented with foetal bovine serum (FBS, 10% v/v, Invitrogen), penicillin (1 ⁇ 10 5 IU/l, Sigma, St Louis, Mo., USA) and streptomycin (100 mg/l, Sigma) buffered with NaHCO 3 containing phenol red (7.22 nmol/l) as a pH indicator, and incubated overnight at 37° C. with 5% CO 2 .
  • DMEM Dulbecco's modified Eagle's medium
  • FBS foetal bovine serum
  • penicillin 1 ⁇ 10 5 IU/l
  • streptomycin 100 mg/l, Sigma
  • PBS phosphate buffered saline
  • the fixative was then removed and 100 ⁇ l of 1% methylene blue stain in 0.01 M borate buffer (pH 8.5) was added to each well and incubated for 30 min at room temperature. Excess stain was removed by four sequential washes in borate buffer. Methylene blue was eluted from the cells by the addition of 100 ⁇ l 1:1 (v/v) ethanol/0.1HCl. The plates were gently shaken and the absorbance was measured at 655 nm using a microplate photometer (VersaMax, Molecular Devices, CA).
  • Human skeletal muscle myoblasts (Cambrex, Australia) were seeded in 96-well tissue culture plates (Nunc, Roskilde, Denmark) at a cell density of 2000 cells per well in 200 ⁇ l of Skeletal Muscle Basal Medium (Cambrex, Australia) supplemented with foetal bovine serum (FBS, 10% v/v, Cambrex, Australia), and incubated overnight at 37° C. with 5% CO 2 .
  • FBS foetal bovine serum
  • DMEM Dulbecco's modified Eagle's medium
  • FBS Invitrogen, California
  • penicillin 1 ⁇ 10 5 IU/l
  • streptomycin 100 mg/l,
  • roMSV47 Acutely and Chronically Down-Regulates Myostatin mRNA Expression in C 2 C 12 Murine Myoblasts.
  • C 2 C 12 myoblasts were seeded in 10 cm diameter tissue culture plates (Nunc, Roskilde, Denmark) at a cell density of 3000 cells per cm 2 in 10 ml of Dulbecco's modified Eagle's medium (DMEM, Invitrogen, California) supplemented with foetal bovine serum (FBS, 10% v/v, Invitrogen), penicillin (1 ⁇ 10 5 IU/l, Sigma, St Louis, Mo., USA) and streptomycin (100 mg/l, Sigma) buffered with NaHCO 3 containing phenol red (7.22 nmol/l) as a pH indicator, and incubated overnight at 37° C. with 5% CO 2 .
  • DMEM Dulbecco's modified Eagle's medium
  • FBS foetal bovine serum
  • penicillin 1 ⁇ 10 5 IU/l
  • Sigma St Louis, Mo., USA
  • streptomycin 100 mg/l, Sigma
  • chronic treatment designated “A”
  • the plates “A” were then incubated at 37° C. with 5% CO 2 for 48 hours.
  • acute treatment designated “B”
  • the plates “B” were then incubated at 37° C.
  • Oligonucleotide primers were used that span across the exon2/3 boundary of mouse myostatin (nt 715-796): forward primer 5′-GCTGTAACCTTCCCAGGACC-3′ (SEQ ID NO: 132) and reverse primer 5′-GGGACCTCTTGGGTGTGTCT-3′ (SEQ ID NO: 133).
  • PCR was carried out with 2.5 ⁇ l of the reverse transcriptase reaction with following master mix for each LightCycler reaction: 4.5 ⁇ l water, 0.5 ⁇ l of 10 ⁇ M primers and 2.0 ⁇ l LightCycler FastStart DNA Master plus SYBR Green I reagent (Roche Diagnostics). A dilution series of mouse muscle reverse transcriptase reaction was used as a standard.
  • the following experimental run protocol was used: denaturation (95° C. for 5 min), amplification (95° C. for 5sec, 62° C. for 10 sec, 72° C. for 20sec, with a single fluorescence measurement, 50 cycles), melt curve (60-95° C. with a heating rate of 0.1° C. per sec with continuous fluorescence measurement) on a LightCycler 2.0 PCR machine (Roche Diagnostics). Arbitrary concentrations were calculated the Lightcycler software using a standard curve (Roche Diagnostics).
  • the ascites hepatoma 130 (AH130) was obtained as a gift from Dr Vicki Baracos (University of Alberta, Edmonton, Alberta, Canada) and stored in cryovials in liquid nitrogen in a solution of 50% DMSO and 10% BSA. An aliquot was retrieved and thawed and 1 ml was injected i.p. into each of three donor rats and allowed to grow for seven days. The rats were killed by CO 2 asphyxiation followed by cervical dislocation. The ascites was harvested and 100 ⁇ l was injected i.p. into each of 14 recipient male rats (280 ⁇ 6 g). An equal volume of saline was injected i.p. into seven control rats.
  • CK creatine kinase
  • Mass of skeletal muscles in the AH130 experiment are presented pooled as a percent of the initial body mass (d0) and data are expressed relative to the muscle mass of control rats (those not inoculated with the tumour).
  • CK-NAC was assayed in a commercial kit (Randox laboratories Ltd, UK). Reaction volumes were scaled down proportionately so that the assays could be performed in 96-well microtitre plates with samples assayed in triplicate (CK-NAC) and read as a change in U/L per min as per manufacturer's instructions.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Medicinal Chemistry (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Genetics & Genomics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Neurology (AREA)
  • Biotechnology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Zoology (AREA)
  • Toxicology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Cardiology (AREA)
  • Pain & Pain Management (AREA)
  • Urology & Nephrology (AREA)
  • Vascular Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Peptides Or Proteins (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
US11/576,449 2004-09-30 2005-09-30 Myostatin Isoform Abandoned US20080118487A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
NZNZ535696 2004-09-30
NZ53569604A NZ535696A (en) 2004-09-30 2004-09-30 Myostatin splice variants ( MSV )
NZ53839605 2005-02-22
NZNZ538396 2005-02-22
PCT/NZ2005/000250 WO2006036074A1 (fr) 2004-09-30 2005-09-30 Isoforme de myostatine

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/NZ2005/000250 A-371-Of-International WO2006036074A1 (fr) 2004-09-30 2005-09-30 Isoforme de myostatine

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/085,263 Continuation US8455448B2 (en) 2004-09-30 2011-04-12 Myostatin isoform

Publications (1)

Publication Number Publication Date
US20080118487A1 true US20080118487A1 (en) 2008-05-22

Family

ID=36119161

Family Applications (2)

Application Number Title Priority Date Filing Date
US11/576,449 Abandoned US20080118487A1 (en) 2004-09-30 2005-09-30 Myostatin Isoform
US13/085,263 Active US8455448B2 (en) 2004-09-30 2011-04-12 Myostatin isoform

Family Applications After (1)

Application Number Title Priority Date Filing Date
US13/085,263 Active US8455448B2 (en) 2004-09-30 2011-04-12 Myostatin isoform

Country Status (6)

Country Link
US (2) US20080118487A1 (fr)
EP (1) EP1799709B1 (fr)
JP (1) JP5335239B2 (fr)
AT (1) ATE552272T1 (fr)
CA (1) CA2582940A1 (fr)
WO (1) WO2006036074A1 (fr)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR100898149B1 (ko) 2007-04-05 2009-05-19 주식회사 에스엔피 제네틱스 소의 근내지방도와 연관되어 있는 티틴-캡 다형성
LT2981822T (lt) 2013-05-06 2020-12-28 Scholar Rock, Inc. Kompozicijos ir būdai, skirti augimo faktoriaus moduliacijai
WO2019195679A1 (fr) * 2018-04-06 2019-10-10 Intrexon Corporation Tilapia à caractéristiques de croissance améliorées
US20220135637A1 (en) 2019-03-01 2022-05-05 Knc Laboratories Co., Ltd. Inhibition of myostatin signal by myostatin splice variant-derived protein and utilization thereof
CN115201474A (zh) * 2022-08-12 2022-10-18 万华普曼生物工程有限公司 一种新型冠状病毒抗体的快速检测试剂盒及其制备方法

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6207160B1 (en) * 1995-06-07 2001-03-27 La Jolla Pharmaceutical Company aPL immunoreactive peptides, conjugates thereof and methods of treatment for aPL antibody-mediated pathologies

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US4487325A (en) 1982-07-06 1984-12-11 Owens-Illinois, Inc. Crown closures and containers
US7393682B1 (en) * 1993-03-19 2008-07-01 The Johns Hopkins University School Of Medicine Polynucleotides encoding promyostatin polypeptides
AU6274298A (en) 1997-02-05 1998-08-25 Johns Hopkins University School Of Medicine, The Growth differentiation factor-8
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
US6369201B1 (en) 1998-02-19 2002-04-09 Metamorphix International, Inc. Myostatin multimers
KR100750695B1 (ko) 1999-07-20 2007-08-22 파멕사 에이/에스 Gdf-8 활성을 하향-조절하는 방법
EP1593689A3 (fr) * 2000-01-18 2006-04-05 Ovita Limited Myostatine et mimétiques de ces derniers
CA2484941A1 (fr) 2002-07-24 2004-02-05 Intercell Ag Antigenes a phase de lecture alternante a partir de virus
EP2192129A1 (fr) * 2002-09-16 2010-06-02 Johns Hopkins University Activation de métalloprotéase de myostatine, et procédés de modulation d'activité de myostatine
NZ529860A (en) * 2003-11-28 2006-10-27 Ovita Ltd Muscle growth regulator mighty and use in promoting muscle mass and treating muscle wasting diseases
AU2004312411B8 (en) * 2003-12-31 2011-11-24 Schering-Plough Pty. Limited Neutralizing epitope-based growth enhancing vaccine
NZ538097A (en) 2005-02-07 2006-07-28 Ovita Ltd Method and compositions for improving wound healing
US20070190056A1 (en) 2006-02-07 2007-08-16 Ravi Kambadur Muscle regeneration compositions and uses therefor
AU2007279456A1 (en) * 2006-08-03 2008-02-07 Myostin Therapeutics Pty Ltd Myostatin antagonists

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6207160B1 (en) * 1995-06-07 2001-03-27 La Jolla Pharmaceutical Company aPL immunoreactive peptides, conjugates thereof and methods of treatment for aPL antibody-mediated pathologies

Also Published As

Publication number Publication date
EP1799709A1 (fr) 2007-06-27
JP2008514227A (ja) 2008-05-08
CA2582940A1 (fr) 2006-04-06
US8455448B2 (en) 2013-06-04
US20110258712A1 (en) 2011-10-20
WO2006036074A1 (fr) 2006-04-06
EP1799709A4 (fr) 2008-11-12
JP5335239B2 (ja) 2013-11-06
ATE552272T1 (de) 2012-04-15
EP1799709B1 (fr) 2012-04-04

Similar Documents

Publication Publication Date Title
KR100728405B1 (ko) Nogo 유전자의 뉴클레오티드와 단백질 서열 및 이들에기초한 방법
RU2329058C2 (ru) Новые факторы роста фибробластов
US8455448B2 (en) Myostatin isoform
US7550435B2 (en) Method of modifying glucose activity using polypeptides selectively expressed in fat tissue
PT1003545E (pt) Polipéptidos de factor de crescimento que se liga à heparina (hbgf)
WO2006137597A9 (fr) Substance physiologique innovante nesfatine, substance qui lui est liée et utilisation des substances
JPH09509305A (ja) 神経軸索発芽後生長モジュレーター
Eib et al. A Novel Transmembrane Protein with Epidermal Growth Factor and Follistatin Domains Expressed in the Hypothalamo‐Hypophysial Axis of Xenopus laevis
US20060127397A1 (en) RAG polypeptides, nucleic acids, and their use
KR20130141395A (ko) Slit-Robo 시스템을 이용한 골절 또는 골다공증의 예방 또는 치료용 조성물
EP1967210A1 (fr) Nouvelle application de l'apéline
ES2397327T3 (es) Compuestos asociados a un dominio único de TDF y análogos de los mismos
US6635616B2 (en) Laminin 15
US20080221057A1 (en) Secreted protein ccdc80 regulates adipocyte differentiation
Miyabe et al. GSP-37, a novel goldfish scale matrix protein: identification, localization and functional analysis
NZ535696A (en) Myostatin splice variants ( MSV )
JP2004008027A (ja) cAMPの産生活性を有する新規ペプチド
EP2289908B1 (fr) Utilisation de produits des protéines DG177 sécretées pour la prévention et le traitement de maladies du pancreas et/ou de l'obésité et/ou du syndrome métabolique
US20030125253A1 (en) Novel protein, process for producing the same and use therof
JP2008538111A (ja) Limミネラル化タンパク質−1(lmp−1)による骨誘導の機構
WO2004020465A2 (fr) Proteines intervenant dans la regulation de l'homeostasie energetique
WO2006010619A1 (fr) Methode pour diagnostiquer et pour traiter des maladies associees aux os
WO2001023424A1 (fr) Proteine liant la phosphatidylethanolamine (pebp-2) et utilisations de cette derniere
JP2005151826A (ja) C1qtnf5の用途
WO2005025590A2 (fr) Utilisation d'un produit proteique dg280 dans la prevention et le traitement de troubles metaboliques

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION