AU2012216342A1 - Use of neuropeptide Y (NPY) and agonists and antagonists thereof for tissue regeneration - Google Patents

Use of neuropeptide Y (NPY) and agonists and antagonists thereof for tissue regeneration Download PDF

Info

Publication number
AU2012216342A1
AU2012216342A1 AU2012216342A AU2012216342A AU2012216342A1 AU 2012216342 A1 AU2012216342 A1 AU 2012216342A1 AU 2012216342 A AU2012216342 A AU 2012216342A AU 2012216342 A AU2012216342 A AU 2012216342A AU 2012216342 A1 AU2012216342 A1 AU 2012216342A1
Authority
AU
Australia
Prior art keywords
tissue
npy
site
injury
neuropeptide
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
AU2012216342A
Inventor
Vishal Bhasin
Helder Marcel
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.)
Regenertech Pty Ltd
Original Assignee
Regenertech Pty 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 AU2006235200A external-priority patent/AU2006235200A1/en
Application filed by Regenertech Pty Ltd filed Critical Regenertech Pty Ltd
Priority to AU2012216342A priority Critical patent/AU2012216342A1/en
Publication of AU2012216342A1 publication Critical patent/AU2012216342A1/en
Abandoned legal-status Critical Current

Links

Landscapes

  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

Abstract The present invention relates to the use of Neuropeptide Y (NPY) and agonists and antagonists thereof for tissue regeneration. According to the present invention, tissue regeneration is distinguished from wound healing.

Description

WO 2006/108218 PCT/AU2006/000481 1 Tissue regeneration Technical field The invention relates to tissue regeneration and to therapeutic compositions, 5 formulations and dressings. Background Tissue regeneration is a process that typically involves the remodelling and/or replacement of tissue elements, such as cellular and extracellular elements. For many invertebrate species, the process is important for asexual reproduction. Further, for 10 these and some vertebrate species, the process is essential for maintenance of tissue structure and function and for restoration of tissue structure and function after tissue injury. Typically, a hallmark of tissue regeneration is the creation of new tissue that has structure and function that is comparable with old tissue. Tissue regeneration is distinguished from wound healing. The latter occurs in response 15 to tissue injury only and typically involves angiogenesis and fibrosis, resulting in granulation and the formation of scar tissue. A key difference between tissue regeneration and wound healing is that the scar tissue formed from wound healing of a tissue does not have structure and function that is comparable to the tissue prior to wounding. 20 Tissue regeneration, in the form of vasculogenesis is also different from angiogenesis. A hallmark of angiogenesis, whether associated with normal or abnormal physiology, is the formation of new blood vessels from an existing vascular bed. In normal physiology, angiogenesis is observed during embryonic development, during the female reproductive cycle and, as noted above, during wound repair. In these circumstances, 25 angiogenesis is tightly regulated and is limited by the metabolic demands of the tissues concerned. Angiogenesis is also observed in a number of pathologies, including tumorigenesis, inflammation and various autoimmune conditions. In these circumstances, regulation of angiogenesis appears to be lost.
WO 2006/108218 PCT/AU2006/000481 2 In comparison, tissue regeneration in the form of vasculogenesis is a series of differentiation and morphogenetic events which result in the formation of a primary capillary plexus. The process typically has three stages; 1) the in situ differentiation of mesodermal cells into angioblasts, 2) the differentiation of angioblasts into endothelial 5 cells, 3) the organization of newly formed endothelial cells into a primary plexus. The primary differentiation of mesodermal angioblasts is a process that is limited exclusively to vasculogenesis. It does not occur during angiogenesis because mesoderm does not persist into post-natal life. By definition vasculogenesis must precede angiogenesis, although the two processes continue in parallel during early development. Unlike 10 vasculogenesis which appears to be restricted to early development, angiogenesis is also required for the maintenance of functional and structural integrity of the organism in post-natal life. As noted above, tissue regeneration is an essential physiological process for many invertebrate species and some vertebrates. In some invertebrate species, such as 15 planarians, existing stem cells, known as neoblasts, are essential for regenerating tissue. Such a regenerative capacity depends on the activation, proliferation, and differentiation of the neoblasts. The axolotl is one of the few vertebrate species in which tissue regeneration has been observed during adult life. Evidence suggests that these amphibians regenerate 20 amputated limb, tail, eye, jaw, and heart structures through a process that involves the formation of a blastema and subsequent activation of a regenerative process reminiscent of the developmental program that originally functioned to specify limb formation. It is believed that the blastema consists of progenitor cells that form from the dedifferentiation of terminally differentiated cells existing at a site of amputation. 25 However, it is equally possible that multipotent adult stem cells exist in adult axolotl tissue, and indeed at a wound site, and that these cells, rather than dedifferentiated mature cells, are the progenitor cells that are required for tissue regeneration. Post-natal mammalian tissue, and especially adult tissues seem to be incapable of tissue regeneration. Dedifferentiation of terminally differentiated cells in adult mammals 30 has not been clearly and unequivocally documented, and at present, no evidence WO 2006/108218 PCT/AU2006/000481 3 directly supports transdifferentiation or dedifferentiation events as an explanation for stem cell plasticity in vivo. Approaches for potentiating the regenerative capacity of mammalian tissue are: (1) transplantation of stem or progenitor cells, or differentiated cells into a compromised 5 tissue or organ; (2) transplantation of cell-seeded scaffolds (biodegradable, biocompatible, bio-mimetic) into damaged regions; and (3) induction of endogenous regeneration through exogenous addition of compounds to activate stem cells or mobilise a pool of stem cells from the surrounding tissue and expand them through stimulation of cellular proliferation and chemotaxis. 10 Summary In certain embodiments there is provided a method for inducing regeneration of a mammalian tissue at a site of injury in the tissue, the method including the step of: providing an agent selected from the group consisting of: neuropeptide Y; 15 a fragment of neuropeptide Y; and an agonist or antagonist of a neuropeptide Y receptor; to a site of injury in a mammalian tissue to induce regeneration of the tissue at the site of injury. In other embodiments there is provided a use of an agent selected from the group 20 consisting of: -neuropeptide Y; -a fragment of neuropeptide Y; -a compound for inducing expression of the neuropeptide Y gene; and WO 2006/108218 PCT/AU2006/000481 4 -an agonist or antagonist of a neuropeptide Y receptor; in the manufacture of a medicament for inducing regeneration of a mammalian tissue at a site of injury in the tissue. In still further embodiments there is provided a dressing for inducing regeneration of a 5 mammalian tissue at a site of injury in the tissue, the dressing including: an agent selected from the group consisting of: -neuropeptide Y; -a fragment of neuropeptide Y; -a compound for inducing expression of the neuropeptide Y gene; and 10 -an agonist or antagonist of a neuropeptide Y receptor. In other embodiments there is provided a composition for inducing regeneration of a mammalian tissue at a site of injury in the tissue, the composition including: an agent selected from the group consisting of: -neuropeptide Y; 15 -a fragment of neuropeptide Y; -a compound for inducing expression of the neuropeptide Y gene; and -an agonist or antagonist of a neuropeptide Y receptor; and a pharmaceutically acceptable carrier, excipient, diluent or lubricant.
WO 2006/108218 PCT/AU2006/000481 5 Brief description of the drawings Figure 1. 2DE PAGE analysis of protein extract from Axolotl regenerating limb blastema. Detailed description of the embodiments 5 The inventors have surprisingly found that NPY is associated with regeneration of vertebrate tissue, especially mammalian tissue. NPY is a peptide typically at least 30 amino acid residues and generally less than 36 amino acid residues that is expressed in the nervous system and other peripheral tissues of a humans, mammals and other vertebrates. Examples of NPY peptides are described below. To date the major clinical 10 interest in NPY has been in relation to the treatment of eating disorders and anxiety. In certain embodiments there is provided a method for inducing regeneration of a mammalian tissue at a site of injury in the tissue, the method including the step of providing an agent selected from the group consisting of NPY, a fragment of NPY and an agonist or antagonist of a NPY receptor to a site of injury in a mammalian tissue to 15 induce regeneration of the tissue at the site of injury. Typically the NPY is a peptide having an amino acid sequence selected from the group of sequences referenced by a SwissProt reference number listed below: SwissProt reference NEUY ALLMIIP68007 20 NEUYBRAREJQ9I8P3 NEUY CARAUIP28672 NEUY CAVPOIP68008 NEUY CHICKIP28673 NEUY DICLAIQ9PTAO 25 NEUYGADMOIP80167 NEUY HUMANIP01303 NEUY ICTPUIQ9I9D3 NEUYLAMFLIP48097 NEUY MACMU IQ9XSW6 30 NEUYMOUSEjP57774 NEUY ONCMYIP29071 NEUY PIGIP01304 NEUY RABITIPO9640 NEUY RANRIIP29949 35 NEUYRATIP07808 WO 2006/108218 PCT/AU2006/000481 6 NEUY SHEEPIP14765 NEUY_TORMAIP28674 NEUY TYPNAlQ9PW68 NEUYXENLA1P33689 5 NPF AEDAEIQ8MP0O NPF ARTTRIP41334 NPF HELASIP41321 NPF MONEXIP41967 NPY CYPCAIQ9DGK7 10 NPY PAROL I Q90WF4 004906 060021 PAHOALLMI|P06305 PAHOANSANIP06304 15 PAHO BOVINIP01302 PAHO CANFAIP01299 PAHOCAVPOGP13083 PAHO CERSIlP37999 PAHO CHIBRIP41519 20 PAHOCHICKIP01306 PAHO DIDMAIP18107 PAHO EQUPRIP68010 PAHO EQUZEIP38000 PAHO ERIEUIP41335 25 PAHOFELCAIP06884 PAHO HUMANIP01298 PAHO LARARIP41337 PAHO MACMUIP33684 PAHO MELGAIP68249 30 PAHOMOUSE I P10601 PAHO_PIGIP01300 PAHO RABITjP41336 PAHO RANCAIP15427 PAHO RANTEIP31229 35 PAHORATIP06303 PAHO SHEEPIP01301 PAHO STRCAIP11967 PAHO TAPPIIP39659 PMY PETMAIP80024 40 PPY-LOPAMIP09475 PYY AMICAIP29205 PYY BOVINIP51694 PYY BRAREIQ9I8P2 PYY CANFAIP68004 45 PYY-CHICKI P29203 PYY_DICLAIQ9PT99 PYYHUMANIP10082 PYY LAMFLIP48098 PYY LEPSPIP09473 50 PYYMOUSEIQ9EPS2 PYY MYOSCIP09641 PYY ONCKIlPO9474 PYY ORENIIP81028 PYY PIGIP01305 55 PYYRABIT1Q9TR93 PYYRAJRHIP29206 PYYRANRIIP29204 PYYRATIP10631 PY DICLAIQ9PT98 60 Q27441 WO 2006/108218 PCT/AU2006/000481 7 Q66KG4 Q6DMM9 Q6EOK5 Q6FGH8 5 Q6NGBO Q6RUW3 Q6T8R7 Q6UA81 Q76CL2 10 Q7SCE6 Q7SZV5 Q7ZYZO Q8JHE7 Q8LDS2 15 Q8SPF7 Q8YK71 Q90WF2 Q90WF3 Q91XDO ?0 Q925V2 Q9GKLO Q9H365 Q9LDVO Q9NOM5 25 Q9PS46 Q9PS51 Q9SEB1 Q9TR92 Q9TSI6 30 Q9UOS8 Q9UOS9 Q9XVQO SPYYPHYBIIP80952 35 It will be understood however, that the NPY may have an amino acid sequence that is not the same as, but rather, has homology with the sequence referenced by SwissProt reference NEUYHUMANIP01303 shown above. These are described herein as an "NPY variant". In some embodiments, an NPY variant has a sequence that is at least 70% homologous to the sequence referenced by SwissProt reference 40 NEUYHUMANIP01303 shown above. In other embodiments the NPY variant sequence is at least 75% homologous to the sequence referenced by SwissProt reference NEUYHUMANIP01303 shown above. In other embodiments the NPY variant sequence is at least 80% homologous to the sequence referenced by SwissProt reference NEUYHUMANIP01303 shown above. In other embodiments the NPY variant 45 sequence is at least 85% homologous to the sequence referenced by SwissProt reference NEUYHUMANIP01303 shown above. In other embodiments the NPY variant sequence is at least 90% homologous to the sequence referenced by SwissProt WO 2006/108218 PCT/AU2006/000481 8 reference NEUYHUMANIP01303 shown above. In other embodiments the NPY variant sequence is at least 95% homologous to the sequence referenced by SwissProt reference NEUYHUMANIP01 303 shown above. In other embodiments the NPY variant sequence is at least 98% homologous to the sequence shown in SEQ ID No: 1. In other 5 embodiments the NPY variant sequence is at least 99% homologous to the sequence referenced by SwissProt reference NEUYHUMANIP01303 shown above. It will be understood that in determining homology, regard is not to be had to peptide regions that are not included in a peptide that has been subjected to post translational modifications, such as, for example a leader or signal peptide. 10 Typically a fragment of NPY is a peptide having an amino acid sequence selected from the following group of amino acid sequences: ProSerLysProAspAsn; ProGlyGluAspAlaPro; AlaGluAspMetAlaArg; TyrTyrSerAlaLeuArg; HisTyrileAsnLeulle; IleThrArgGlnArgTyr As described herein, NPY, including human and non human NPY, fragments thereof 15 and NPY variants, may be prepared by chemical synthesis methodologies or by recombinant DNA technology. For example, these agents can be prepared from monomers using a chemical synthesis methodology based on the sequential addition of amino acid residues, for example as described in Merrifield, J. Am. Chem. Soc., 85: 2149 (1963). These monomers may be naturally occurring residues, or non naturally 20 occurring residues, examples of which are described below. Alternatively, the agents, and in particular, a NPY fragment, can be prepared by enzymatically or chemically treating a peptide having, for example, a sequence referenced by SwissProt reference NEUYHUMANIP01303 shown above. Where these peptides are to be synthesised by recombinant DNA technology, they may be prepared by random or pre-determined 25 mutation (eg site directed PCR mutagenesis) of a nucleic acid molecule that encodes a NPY sequence, for example, a sequence referenced by SwissProt reference NEUYHUMANIP01303 shown above and expression of the sequence in a host cell to obtain the peptide. This is a particularly useful process for preparing NPY variants. An alternative process is de novo chemical synthesis of a nucleic acid molecule that 30 encodes a sequence -referenced by SwissProt reference NEUYHUMANIP01303 WO 2006/108218 PCT/AU2006/000481 9 shown above, or a sequence that is homologous to the sequence referenced by SwissProt reference NEUYHUMANJP01303 shown above and expression of the sequence in the host cell to obtain the peptide. The peptides that are variants of the sequence referenced by SwissProt reference 5 NEUYHUMANIP01303 shown above typically differ in terms of one or more conservative amino acid substitutions in these sequences. Examples of conservative substitutions are shown in Table 1 below. Table 1 Original Residue Exemplary Conservative Preferred Conservative Substitution Substitution Ala Val, Leu, lie Val Asn Gln Lys His Phe GIn GIn Asn Asn Gly Pro Pro Ile Leu, Val, Met, Ala, Leu Phe Leu lle, Val, Met, Ala, Phe Ile Lys Arg, GIn, Asn Arg Phe Lue, Val, lle, Ala Leu Pro Gly Gly Ser Thr Thr Val lie, Leu, Met, Phe, Ala Leu Asp Glu Glu Thr Ser Ser Trp Tyr Tyr WO 2006/108218 PCT/AU2006/000481 10 Tyr Trp Phe Thr Ser Phe As noted above, the human and non human NPY and fragments thereof, and NPY variants, may include non naturally occurring amino acid residues. Commonly encountered amino acids which are not encoded by the genetic code, include: 2-amino adipic acid (Aad) for GIu and Asp; 5 2-aminopimelic acid (Apm) for Glu and Asp; 2-aminobutyric (Abu) acid for Met, Leu, and other aliphatic amino acids; 2-aminoheptanoic acid (Ahe) for Met, Leu and other aliphatic amino acids; 2-aminoisobutyric acid (Aib) for Gly; cyclohexylalanine (Cha) for Val, and Leu and lie; 10 homoarginine (Har) for Arg and Lys; 2, 3-diaminopropionic acid (Dpr) for Lys, Arg and His; N-ethylglycine (EtGly) for Gly, Pro, and Ala; N-ethylasparigine (EtAsn) for Asn, and Gin; Hydroxyllysine (Hyl) for Lys; 15 allohydroxyllysine (AHyl) for Lys; 3-(and 4) hydroxyproline (3Hyp, 4Hyp) for Pro, Ser, and Thr; alloisoleucine (Alle) for lie, Leu, and Val; p-amidinophenylalanine for Ala; WO 2006/108218 PCT/AU2006/000481 N-methylglycine (MeGly, sarcosine) for Gly, Pro, Ala. N-methylisoleucine (Melle) for lie; Norvaline (Nva) for Met and other aliphatic amino acids; Norleucine (NIe) for Met and other aliphatic amino acids; 5 Ornithine (Orn) for Lys, Arg and His; Citrulline (Cit) and methionine sulfoxide (MSO) for Thr, Asn and Gin; N-methylphenylalanine (MePhe), trimethylphenylalanine, halo (F, Cl, Br and I) phenylalanine, triflourylphenylalanine, for Phe. A useful method for identification of a residue of the sequence referenced by SwissProt 10 reference NEUYHUMANIP01303 shown above for amino acid substitution to generate a NPY variant is called alanine scanning mutagenesis as described by Cunningham and Wells (1989) Science, 244:1081-1085. Here a residue or group of target residues are identified (eg charged residues such as Asn, Gin and Lys) and replaced by a neutral or negatively charged amino acid to affect the interaction of the amino acids with the 15 surrounding environment. Those domains demonstrating functional sensitivity to the substitution then are refined by introducing further or other variations at or for the sites of substitution. Thus while the site for introducing an amino acid sequence variation is predetermined the nature of the mutation per se need not be predetermined. For example, to optimize the performance of a mutation at a given site, Ala scanning or 20 random mutagenesis may be conducted at the target codon or region and the expressed peptide screened for the optimal combination of desired activity. Phage display of protein or peptide libraries offers another methodology for the selection of peptide with improved or altered affinity, specificity, or stability (Smith, G, P, (1991) Curr Opin Biotechnol (2:668-673). High affinity proteins, displayed in a 25 monovalent fashion as fusions with the M13 gene Ill coat protein (Clackson, T, (1994) et al, Trends Biotechnol 12:173-183), can be identified by cloning and sequencing the WO 2006/108218 PCT/AU2006/000481 12 corresponding DNA packaged in the phagemid particles after a number of rounds of binding selection. Human and non human NPY, fragments thereof and NPY variants may be prepared as the free acid or base or converted to salts of various inorganic and organic acids and 5 bases. Examples of such salts include ammonium, metal salts like sodium, potassium, calcium and magnesium; salts with organic bases like dicyclohexylamine, N-methyl-D glucamine and the like; and salts with amino acids like arginine or lysine. Salts with inorganic and organic acids may be likewise prepared, for example, using hydrochloric, hydrobromic, sulfuric, phosphoric, trifluoroacetic, methanesulfonic, malic, maleic, 10 fumaric and the like. Non-toxic and physiologically compatible salts are particularly useful, although other less desirable salts may have use in the processes of isolation and purification. In one embodiment, human or non human NPY, fragments thereof or NPY variants include at least one carbohydrate molecule and/or at least one lipid molecule. 15 In one embodiment human or non human NPY, fragments thereof or NPY variants include a N-terminal fatty acid moiety, for example a C14 to C17 fatty acid of n, iso or antieso form. In one embodiment, human or non human NPY, fragments thereof or NPY variants include at least one akyl group. 20 The human or non human NPY, fragments thereof or NPY variants may include a further peptide, for example for controlling degradation of the peptide, or for arranging the peptide on a solid phase or for binding with an antibody or receptor to purify, isolate or detect the peptide. Such peptides are otherwise known in the art as fusion proteins. Fusion proteins can be made by the chemical synthesis methods describe below, or 25 they can be made by recombinant DNA techniques, for example, wherein a nucleic acid molecule encoding the peptide having the sequence referenced by SwissProt reference NEUYHUMANIP01303 shown above is arranged in a vector with a gene encoding another protein or a fragment of another protein. Expression of the vector results in the WO 2006/108218 PCT/AU2006/000481 13 human or non human NPY, fragments thereof or NPY variants being produced as a fusion with another protein or peptide. The further protein or peptide that is fused to the human or non human NPY, fragments thereof or NPY variants may be a protein or peptide that can be secreted by a cell, 5 making it possible to isolate and purify from the culture medium and eliminating the necessity of destroying the host cells; this necessity arises when human or non human NPY, fragments thereof or NPY variants remains inside the cell. Alternatively, the fusion protein can be expressed inside the cell as a function of the further protein or peptide. It is useful to use fusion proteins that are highly expressed. 10 The use of fusion proteins, though not essential, can facilitate the expression of heterologous peptides in E. coil as well as the subsequent purification of those gene products. Harris, in Genetic Engineering, Williamson, R., Ed. (Academic Press, London, Vol. 4, 1983), p. 127; Ljunqquist et al., Eur. J. Biochem., 186: 557-561 (1989) and Liunqquist et al., Eur. J. Biochem., 186: 563-569 (1989). Protein A fusions are often 15 used because the binding of protein A, or more specifically the Z domain of protein A, to IgG provides an "affinity handle" for the purification of the fused protein. It has also been shown that many heterologous proteins are degraded when expressed directly in E. coli, but are stable when expressed as fusion proteins. Marston, Biochem J., 240: 1 (1986). 20 Fusion proteins can be cleaved using chemicals, such as cyanogen bromide, which cleaves at a methionine, or hydroxylamine, which cleaves between an Asn and Gly residue. Using standard recombinant DNA methodology, the nucleotide base pairs encoding these amino acids may be inserted just prior to the 5' end of the gene encoding the desired peptide. 25 Alternatively, one can employ proteolytic cleavage of fusion protein, see for example Carter in Protein Purification: From Molecular mechanisms to Large-Scale Processes, Ladisch et al., eds. (American Chemical Society Symposium Series No. 427, 1990), Ch 13, pages 181-193.
WO 2006/108218 PCT/AU2006/000481 14 Proteases such as Factor Xa, thrombin, and subtilisin or its mutants, and a number of others have been successfully used to cleave fusion proteins. Typically, a peptide linker that is amenable to cleavage by the protease used is inserted between the further proteins (e.g., the Z domain of protein A) and human or non human NPY, fragments 5 thereof or NPY variants. Using recombinant DNA methodology, the nucleotide base pairs encoding the linker are inserted between the genes or gene fragments coding for the other proteins. Proteolytic cleavage of the partially purified fusion protein containing the correct linker can then be carried out on either the native fusion protein, or the reduced or denatured fusion protein. 10 The human or non human NPY, fragments thereof or NPY variants may not be properly folded when expressed as a fusion protein. Also, the specific peptide linker containing the cleavage site may or may not be accessible to the protease. These factors determine whether the fusion protein must be denatured and refolded, and if so, whether these procedures are employed before or after cleavage. 15 When denaturing and refolding are needed, typically human or non human NPY, fragments thereof or NPY variants is treated with a chaotrope, such as guanidine HCI, and is then treated with a redox buffer, containing, for example, reduced and oxidized dithiothereitol or glutathione at the appropriate ratios, pH, and temperature, such that the relevant peptide is refolded to its native structure. 20 Other fusion proteins include those wherein the human or non human NPY, fragments thereof or NPY variants is fused to a protein having a long half-life such as immunoglobulin constant region or other immunoglobulin regions, albumin, or ferritin. The human or non human NPY, fragments thereof or NPY variants may be stabilized by polymerization. This may be accomplished by cross linking the human or non human 25 NPY, fragments thereof or NPY variants with polyfunctional cross linking agents, either directly or indirectly, through multi-functional polymers. For example, two substantially identical polypeptides may be cross linked at their C- or N-termini using a bifunctional cross linking agent. The agent may be used to cross link the terminal amino and/or carboxyl groups. While both terminal carboxyl groups or both terminal amino groups WO 2006/108218 PCT/AU2006/000481 15 may be cross linked to one another, other cross linking agents permit the alpha amino of one peptide to be cross linked to the terminal carboxyl group of the other peptide. To facilitate use of other reagents for cross linking, the human or non human NPY, fragments thereof or NPY variants may be substituted at their C-termini with cysteine. 5 Under conditions well known in the art a disulfide bond can be formed between the terminal cysteines, thereby cross linking peptide chains. For example, disulfide bridges are conveniently formed by metal-catalyzed oxidation of the free cysteines or by nucleophilic substitution of a suitably modified cysteine residue. Selection of the cross linking agent will depend upon the identities of the reactive side 10 chains of the amino acids present in the peptides. For example, disulfide cross linking would not be preferred if cysteine was present in the peptide at additional sides other than the C-terminus. A further approach for cross linking peptides is the use of methylene bridges. Suitable cross linking sites on the peptides, aside from the N-terminal amino and C 15 terminal carboxyl groups, include epsilon amino groups found on lysine residues, as well as amino, imino, carboxyl, sulfhydryl and hydroxyl groups located on the side chains of internal residues of the peptides or residues introduced into flanking sequences. Cross linking through externally added cross linking agents is suitably achieved, e.g., using any of a number of reagents familiar to those skilled in the art, for 20 example, via carbodiimide treatment of the peptide. Other examples of suitable multi functional (ordinarily bifunctional) cross linking agents are found in the literature. The human or non human NPY, fragments thereof or NPY variants also may be conformationally stabilized by cyclization. They may be cyclized by covalently bonding the N- and the C-terminal domains of one peptide to the corresponding domain of 25 another human or non human NPY, fragments thereof or NPY variants, so as to form cyclo-oligomers containing two or more iterated peptide sequences. Further, cyclized peptides (whether cyclo-oligomers or cyclo-monomers) may be cross linked to form 1-3 cyclic structures having from 2 to 6 peptides comprised therein. The peptides typically WO 2006/108218 PCT/AU2006/000481 16 are not covalently bonded through a-amino and main chain carboxyl groups (head to tail), but rather are crosslinked through the side chains of residues located in the N- and C-terminal domains. The linking sites thus generally will be between the side chains of the residues. 5 Many suitable methods per se are known for preparing mono- or poly-cyclized peptides as contemplated herein. Lys/Asp cyclization has been accomplished using Na-Boc amino acids on solid-phase support with Fmoc/9-fluorenylmethyl (OFm) side-chain protection for Lys/Asp; the process is completed by piperidine treatment followed by cyclization. 10 Glu and Lys side chains also have been crosslinked in preparing cyclic or bicyclic peptides: the peptide is synthesized by solid phase chemistry on a p methylbenzhydrylamine resin. The peptide is cleaved from the resin and deprotected. The cyclic peptide is formed using disphenylphosphrylazide in diluted methylformamide. For an alternative procedure, see Schiller et al., Peptide Protein Res., 25: 171-177 15 (1985). See also U.S. Pat. No. 4,547,489. Disulfide cross linked or cyclized peptides may be generated by conventional methods. The method of Pelton et al. (J. Med. Chem., 29: 2370-2375 (1986) is suitable. The same chemistry is useful for synthesis of dimers or cyclo-oliogomers or cyclo monomers. Also useful are thiomethylene bridges. Lebl and Hruby, Tetrahedron Letters, 20 25: 2067-2068 (1984). See also Cody et al., J. Med. Chem., 28: 583 (1985). The desired cyclic or polymeric peptides may be purified by gel filtration followed by reversed-phase high pressure liquid chromatography or other conventional procedures. The peptides may be sterile filtered for formulation into a therapeutic composition described further herein. 25 The human or non human NPY, fragments thereof or NPY variants described above can be made by chemical synthesis or by employing recombinant DNA technology. These methods are known in the art. Chemical synthesis, especially solid phase synthesis, is preferred for short (e.g., less than 50 residues) peptides or those WO 2006/108218 PCT/AU2006/000481 17 containing unnatural or unusual amino acids such as D-Tyr, Ornithine, amino adipic acid, and the like. Recombinant procedures are preferred for longer peptides. When recombinant procedures are selected, a synthetic gene may be constructed de novo or a natural gene may be mutated by, for example, cassette mutagenesis. These 5 procedures are described further herein. Set forth below are exemplary general procedures for chemical synthesis of human or non human NPY, fragments thereof or NIPY variants . Peptides are typically prepared using solid-phase synthesis, such as that generally described by Merrifield, J. Am. Chem. Soc., 85: 2149 (1963), although other equivalent 10 chemical syntheses known in the art are employable. Solid-phase synthesis is initiated from the C-terminus of the peptide by coupling a protected a-amino acid to a suitable resin. Such a starting material can be prepared by attaching a a-amino-protected amino acid by an ester linkage to a chloromethylated resin or a hydroxymethyl resin, or by an amide bond to a BHA resin or MBHA resin. The preparation of the hydroxymethyl resin 15 is described by Bodansky et al., Chem. Ind. (London), 38: 1597-1598 (1966). Chloromethylated resins are commercially available from BioRad Laboratories, Richmond, Calif. And from Lab. Systems, Inc. The preparation of such a resin is described by Stewart et al., "Solid Phase Peptide Synthesis" (Freeman & Co., San Francisco 1969), Chapter 1, pp. 1-6. BHA and MBHA resin supports are commercially 20 available and are generally used only when the desired polypeptide being synthesized has an unsubstituted amide at the C-terminus. The amino acids are coupled to the peptide chain using techniques well known in the art for the formation of peptide bonds. One method involves converting the amino acid to a derivative that will render the carboxyl group more susceptible to reaction with the free 25 N-terminal amino group of the peptide fragment. For example, the amino acid can be converted to a mixed anhydride by reaction of a protected amino acid with ethychloroformate, phenyl chloroformate, sec-butyl chloroformate, isobutyl chloroformate, pivaloyl chloride or like acid chlorides. Alternatively, the amino acid can be converted to an active ester such as a 2,4,5-trichlorophenyl ester, a 30 pentachlorophenyl ester, a pentafluorophenyl ester, a p-nitrophenyl ester, a N hydroxysuccinimide ester, or an ester formed from 1-hydroxybenzotriazole.
WO 2006/108218 PCT/AU2006/000481 18 Another coupling method involves use of a suitable coupling agent such as N,Nl dicyclohexylcarbodiimide or N,N-diisopropylcarbodiimide. Other appropriate coupling agents, apparent in those skilled in the art, are disclosed in E Gross & J Meienhofer, The Peptides: Analysis, Structure, Biology, Vol. I: Maior Methods of Peptide Bond 5 Formation (Academic Press, New York, 1979). It should be recognized that the a-amino group of each amino acid employed in the peptide synthesis must be protected during the coupling reaction to prevent side reactions involving their active a-amino. function. It should also be recognized that certain amino acids contain reactive side-chain functional groups (eg sulfhydryl, amino, 10 carboxyl, and hydroxyl) and that such functional groups must also be protected with suitable protecting groups to prevent a chemical reaction from occurring at that site during both the initial and subsequent coupling steps. Suitable protecting groups, known in the art, are described in Gross and Meienhofer, The Peptides: Analysis, Structure, Biology, Vol. 3: "Protection of Functional Groups in Peptide Synthesis" (Academic 15 Press, New York 1981). In the selection of a particular side-chain protecting group to be used in synthesizing the peptides, the following general rules are followed. An a-amino protecting group must render the a-amino function inert under the conditions employed in the coupling reacting, must be readily removable after the coupling reaction under conditions that will 20 not remove side-chain protecting groups and will not alter the structure of the peptide fragment, and must eliminate the possibility of racemization upon activation immediately prior to coupling. A side-chain protecting group must render the side chain functional group inert under the conditions employed in the coupling reaction, must be stable under the conditions employed in removing the a-amino protecting group, and must be 25 readily removable upon completion of the desired amino acid peptide under reaction conditions that will not alter the structure of the peptide chain. It will be apparent to those skilled in the art that the protecting groups known to be useful for peptide synthesis will vary in reactivity with the agents employed for their removal. For example, certain protecting groups such as triphenylmethyl and 2-(p 30 biphenylyl)isopropyloxycarbonyl are very labile and can be cleaved under mild acid WO 2006/108218 PCT/AU2006/000481 19 conditions. Other protecting groups, such as t-butyloxycarbonyl (BOC), t amyloxycarbonyl, adamantyloxycarbonyl, and p-methoxybenzyloxycarbonyl are less labile and require moderately strong acid, such as trifluoroacetic, hydrochloric, or boron trifluoride in acetic acid, for their removal. Still other protecting groups, such as 5 benzyloxy-carbonyl (CBZ or Z), halobenzyloxycarbonyl, p-nitrobenzyloxycarbonyl cycloalkyloxycarbonyl, and isopropyloxycarbonyl, are even less labile and require stronger acids, such as hydrogen fluoride, hydrogen bromide, or boron trifluoroacetate in trifluoroacetic acid, for their removal. Among the classes of useful amino acid protecting groups are included: 10 (1) for an a-amino group, (a) aromatic urethane-type protecting groups, such as fluorenylmethyloxycarbonyl (FMOC) CBZ, and substituted CBZ, such as, eg, p chlorobenzyloxycarbonyl, p-6-nitrobenzyloxycarbonyl, p-bromobenzyloxycarbonyl, and p-methoxybenzyloxycarbonyl, o-chlorobenzyloxycarbonyl, 2,4 dichlorobenzyloxycarbonyl, 2,6-dichlorobenzyloxycarbonyl, and the like; (b) aliphatic 15 urethane-type protecting groups, such as BOC, t-amyloxycarbonyl, isopropyloxycarbonyl, 2-(p-biphenylyl)-isopropyloxycarbonyl, allyloxycarbonyl and the like; (c) cycloalkyl urethane-type protecting groups, such as cyclopentyloxycarbonyl, adamantyloxycarbonyl, and cyclohexyloxycarbonyl; and (d) allyloxycarbonyl. The preferred a-amino protecting groups are BOX or FMOC. 20 (2) for the side chain amino group present in Lys, protection may be by any of the groups mentioned above in (1) such as BOC, p-chlorobenzyloxycarbonyl, etc. (3) for the guanidino group of Arg, protection may be by mitro, tosyl, CBZ, adamantyloxycarbonyl, 2,2,5,7,8-pentamethylchroman-6-sulfonyl or 2,3,6-trimethyl-4 methoxyphenylsulfonyl, or BOC. 25 (4) for the hydroxyl group of Ser, Thr, or Tyr, protection may be, for example, by C1-C4 alkyl, such as t-butyl; benzyl (BAL); substituted BZL, such as p-methoxybenzyl, p nitrobenzyl, p-chlorobenzyl, o-chlorobenzyl, and 2,6-dichlorobenzyl.
WO 2006/108218 PCT/AU2006/000481 20 (5) for the carboxyl group of Asp or Glu, protection may be, for example, by esterification using groups such as BZL, t-butyl, cyclohexyl, cyclopentyl, and the like. (6) for the imidazole nitrogen of His, the tosyl moiety is suitable employed. (7) for the phenolic hydroxyl group of Tyr, a protecting group such as tetrahydropyranyl, 5 tert-butyl, trityl, BZL, chlorobenzyl, 4-bromobenzyl, or 2,6-dichlorobenzyl is suitably employed. The preferred protecting group is 2,6-dichlorobenzyl. (8) for the side chain amino group of Asn or Gln, xanthyl (Xan) is preferably employed. (9) for Met, the amino acid is preferably left unprotected. (10) for the thio group of Cys, p-methoxybenzyl is typically employed. 0 The C-terminal amino acid, eg, Lys, is protected at the N-amino position by an appropriately selected protecting group, in the case of Lys, BOC. The BOC-Lys-OH can be first coupled to the benzyhydrylamine or chloromethylated resin according to the procedure set forth in Horiki et al, (Chemistry Letters, 165-168 1978) or using isopropylcarbodiimide at about 250C for 2 hours with stirring. Following the coupling of 5 the BOC-protected amino acid to the resin support, the a-amino protecting group is removed, as by using trifluoroacetic acid (TFA) in methylene chloride or TFA alone. The deprotection is carried out at a temperature between about 00C and room temperature. Other standard cleaving reagents, such as HCI in dioxane, and conditions for removal of specific a-amino protecting groups are described in the literature. 20 After removal of the a-amino protecting group, the remaining a-amino and side-chain protected amino acids are coupled stepwise within the desired order. As an alternative to adding each amino acid separately in the synthesis, some may be coupled to one another prior to addition to the solid-phase synthesizer. The selection of an appropriate coupling reagent is within the skill of the art. Particularly suitable as a coupling reagent 25 is N,Nl-dicyclohexyl carbodiimide or diisopropylcarbodiimide.
WO 2006/108218 PCT/AU2006/000481 21 Each protected amino acid or amino acid sequence. is introduced into the solid-phase reactor in excess, and the coupling is suitably carried out in a medium of dimethylformamide (DMF) or CH 2
CI
2 or mixtures thereof. If incomplete coupling occurs, the coupling procedure is repeated before removal of the N-amino protecting group piror 5 to the coupling of the next amino acid. The success of the coupling reaction at each stage of the synthesis may be monitored. A preferred method of monitoring the synthesis is by the ninhydrin reaction, as described by Kaiser et al., Anal Biochem, 34: 595 (1970). The coupling reactions can be performed automatically using well known methods, for example, a BIOSEARCH 9 5 0 0 TM peptide synthesizer. 10 Upon completion of the desired peptide sequence, the protected peptide must be cleaved from the resin support, and all protecting groups must be removed. The cleavage reaction and removal of the protecting groups is suitably accomplished simultaneously or stepwise. When the resin support is a chloromethylated polystyrene resin, the bond anchoring the peptide to the resin is an ester linkage formed between 15 the free carboxyl group of the C-terminal residue and one of the many chloromethyl groups present on the resin matrix. It will be appreciated that the anchoring bond can be cleaved by reagents that are known to be capable of breaking an ester linkage and of penetrating the resin matrix. One especially convenient method is by treatment with liquid anhydrous hydrogen 20 fluoride. This reagent not only will cleave the peptide from the resin but also will remove all protecting groups. Hence, use of this reagent will directly afford the fully deprotected peptide. When the chloromethylated resin is used, hydrogen fluoride treatment results in the formation of the free peptide acids. When the benzhydrylamine resin is used, hydrogen fluoride treatment results directly in the free peptide amines. Reaction with 25 hydrogen fluoride in the presence of anisole and dimethylsulfide at 00C for one hour will simultaneously remove the side-chain protecting groups and release the peptide from the resin. When it is desired to cleave the peptide without removing protecting groups, the protected peptide-resin can undergo methanolysis to yield the protected peptide-resin 30 can undergo methanolysis to yield the protected peptide in which the C-terminal WO 2006/108218 PCT/AU2006/000481 22 carboxyl group is methylated. The methyl ester is then hydrolysed under mild alkaline conditions to give the free C-terminal carboxyl group. The protecting groups on the peptide chain then are removed by treatment with a strong acid, such as liquid hydrogen fluoride. A particularly useful technique for methanolysis is that of Moore et al, 5 Peptides, Proc Fifth Amer Pept Symp, M Goodman and J Meienhofer, Eds, (John Wiley, N.Y., 1977), p.518-521, in which the protected peptide-resin is treated with methanol and potassium cyanide in the presence of crown ether. Another method of cleaving the protected peptide form the resin when the chloromethylated resin is employed is by ammonolysis or by treatment with hydrazine. If 10 desired, the resulting C-terminal amide or hydrazide can be hydrolysed to the free C terminal carboxyl moiety, and the protecting groups can be removed conventionally. It will also be recognized that the protecting group present on the N-terminal a-amino group may be removed preferentially either before or after the protected peptide is cleaved from the support. 15 If in the peptides being created carbon atoms bonded to four non identical substituents are asymmetric, then the compounds may exist as disastereoisomers, enantiomers or. mixtures thereof. The syntheses described above may employ racemates, enantiomers or disastereoisomers as starting materials or intermediates. Disastereomeric products resulting from such syntheses may be separated by chromatographic or crystallization 20 methods. Likewise, enantiomeric product mixtures may be separated using the same techniques or by other methods known in the art. Each of the asymmetric carbon atoms, when present, may be in one of two configurations (R or S) and both are within the scope of the present invention. Purification of the peptide is typically achieved using conventional procedures such as 25 preparative HPLC (including reversed phase HPLC) or other known chromatographic techniques such as gel permeation, ion exchange, partition chromatography, affinity chromatography (including monoclonal antibody columns) or counter-current distribution.
WO 2006/108218 PCT/AU2006/000481 23 As described above, the peptide may be prepared as salts of various inorganic and organic acids and bases. A number of methods are useful for the preparation of these salts and are known to those skilled in the art. Examples include reaction of the free acid or free base form of the peptide with one or more molar equivalents of the desired 5 acid or base in a solvent or solvent mixture in which the salt is insoluble; or in a solvent like water after which the solvent is removed by evaporation, distillation or freeze drying. Alternatively, the free acid or base form of the produce may be passed over an ion exchange resin to form the desired salt or one salt form of the product may be convened to another using the same general process. 10 The starting materials required for use in the chemical synthesis of peptides described above are known in the literature or can be prepared using known methods and known starting materials. The method of the invention may include the step of providing one or more NPY receptor agonists or antagonists. In certain embodiments, useful agonists and 15 antagonists include compounds that interact with one or more of the NPY1, 2, 4 and 5 receptors. Examples of these agonists and antagonists and their manufacture are discussed in the following US patents and patent application listed in Table 2, the contents of which are incorporated by reference in their entirety: 20 Table 2 Publication Title Assignee US6849733 Neuropeptide-Y ligands Agouron Pharmaceuticals, Inc. US20050014742A1 Use of inhibitors of the y2 receptor of neuropeptide y in the treatment of alcoholism US6841552 3a,4,5,9b-tetrahydro-1H-benz[e]indol-2-yI Ortho-McNeil amine-derived neuropeptide Y receptors Pharmaceutical, Inc. ligands useful in the treatment of obesity and other disorders US20040157918A1 Weight loss induced by reduction in None neuropeptide y level US20040127440A1 Methods and compositions for control of BAYLOR COLLEGE OF WO 2006/108218 PCT/AU2006/000481 24 bone formation via modulation of MEDICINE neuropeptide Y activity US20040053864A1 Methods and compositions for control of None bone formation via modulation of neuropeptide y activity US6696409 * Neuropeptide Y agonists Prince of Wales Medical Research Institute Limited (POWMR Ltd.) US20040029953A1 Use of neuropeptide-y antagonists in None treatment of alcoholism US6653478 Substituted benzimidazol-2-ones as Ortho-McNeil vasopressin receptor antagonists and Pharmaceutical, Inc. neuropeptide Y modulators US6649759 Neuropeptide Y antagonists Pfizer Inc. US6632828 Substituted imidazole neuropeptide Y Y5 Schering Corporation receptor antagonists US20030100546A1 Neuropeptide Y antagonists None US20030093821A1 DNA molecule encoding a mutant prepro- None neuropeptide Y, a mutant signal peptide,and uses thereof US6562862 Methods of inhibiting physiological Eli Lilly and Company conditions associated with an excess of neuropeptide Y US20030073842A1 Novel substituted benzimidazol-2-ones as None vasopressin receptor antagonists and neuropeptide Y modulators US20030050446A1 Regulation of human neuropeptide y-like g None protein-coupled receptor US6511984 Neuropeptide Y antagonists Pfizer Inc. US6426330 Satiety inducing composition comprising East Carolina University Neuropeptide Y analogues and methods of inducing satiety and treating a disease or condition associated with it US6410792 Amide derivatives and methods for using Bayer Corporation the same as selective neuropeptide Y receptor antagonists US6407120 Neuropeptide Y antagonists Pfizer Inc. US20020061897A1 Neuropeptide Y antagonists None US20020058671A1 Neuropeptide Y antagonists None US20020052356A1 Neuropeptide Y antagonists US6380224 Amine and amide derivatives as ligands for Ortho-McNeil the neuropeptide Y Y5 receptor useful in the Pharmaceutical, Inc. treatment of obesity and other disorders US20020048791A1 Human neuropeptide Y-like G protein- coupled receptor US6368824 Neuropeptide Y receptor Y5 and nucleic acid Bayer Corporation sequences US6358991 Methods of treating neuropeptide Y-related American Home Products conditions Corporation US6355478 Rhesus monkey neuropeptide Y Y2 receptor Eli Lilly and Company WO 2006/108218 PCT/AU2006/000481 25 US20020019392A1 Methods of treating neuropeptide Y-related American Home Products conditions Corporation US20020007071A1 Benzimidzolyl neuropeptide Y receptor None antagonists US6337332 Neuropeptide Y receptor antagonists Pfizer Inc. US6313128 Cosmetic composition containing a Sanofi-Synthelabo neuropeptide Y receptor antagonist US20010031758A1 Substituted 9H-pyridino[2,3-b] indole and Neurogen Corporation 9H-pyrimidino[4,5-b] indole derivatives: selective neuropeptide Y receptor ligands US20010031474A1 Chimeric neuropeptide Y receptors NEUROGEN CORPORATION US6258837 Neuropeptide Y receptor antagonist Banyu Pharmaceutical Co., Ltd. US6255494 Benzimidzolyl neuropeptide Y receptor Eli Lilly and Company antagonists US6245761 Indolyl neuropeptide Y receptor antagonists Eli Lilly and Company US6221875 Substituted 9H-pyridino [2,3-B]indole and Neurogen Corporation 9H-pyrimidino [4,5-B]indole derivatives: selective neuropeptide Y receptor ligands US6221838 Methods of treating neuropeptide Y- Eli Lilly and Company associated conditions US6207799 Neuropeptide Y receptor Y5 and nucleic acid Bayer Corporation sequences US6201025 N-aralkylaminotetralins as ligands for the Ortho-McNeil neuropeptide Y Y5 receptor Pharmaceutical, Inc. US6187778 4-aminopyrrole (3, 2-D) pyrimidines as Pfizer Inc. neuropeptide Y receptor antagonists US6140354 N-substituted aminotetralins as ligands for Ortho-McNeil the neuropeptide Y Y5 receptor useful in the Pharmaceutical, Inc. treatment of obesity and other disorders US6114336 Cosmetic composition containing a Sanofi neuropeptide Y receptor antagonist US6075009 Neuropeptide Y analogues, compositions East Carolina University and methods of lowering blood pressure US6048900 Amide derivatives and methods for using Bayer Corporation the same as selective neuropeptide Y receptor antagonists US6046317 DNA molecule encoding a mutant prepro- Hormos Medical Oy, Ltd. neuropeptide Y, a mutant signal peptide, and uses thereof US6040310 4-aminopyrrole (3,2-d) pyrimidines as Pfizer Inc. neuropeptide Y receptor antagonists US5939263 Neuropeptide Y receptor Merck & Co., Ltd. US5395823 Neuropeptide Y agonists and partial Bayer Corporation agonists US5914329 Dimesylate salts of neuropeptide Y ligands Pfizer Inc. US5827853 Cosmetic composition containing a Sanofi neuropeptide Y receptor antagonist WO 2006/108218 PCT/AU2006/000481 26 US5776931 Naphthimidazolyl neuropeptide Y receptor Eli Lilly and Company antagonists US5696093 Method of treating nasal congestion using CRC for neuropeptide Y Y2 agonist peptides Biopharmaceutical Research Pty Limited US5670482 Neuropeptide Y antagonists Glaxo Wellcome Inc. US5663192 Heterocyclic neuropeptide Y receptor Eli Lilly and Company antagonists US5621079 Neuropeptide Y receptor Merck & Co., Inc. US5576337 Method of treating anxiety by inhibiting Eli Lilly and Company physiological conditions associated with an excess of neuropeptide Y US5571695 Human neuropeptide Y-Y1 receptor Garvan Institute of Medical Research US5567715 Methods of treating depression by inhibiting Eli Lilly and Company physiological conditions associated with an excess of neuropeptide Y US5567714 Methods of treating obesity by inhibiting Eli Lilly and Company physiological conditions associated with an excess of neuropeptide Y. US5504094 Use of bengothiophenis to treat pain due to Eli Lilly and Company an excess of neuropeptide y US5395823 Neuropeptide Y agonists and partial Merrell Dow agonists Pharmaceuticals Inc. Typically the agent is provided to the site of injury by contacting the tissue with NPY, one or more fragments thereof, a NPY variant or an agonist or antagonist of a NPY receptor. It will be understood that in certain embodiments, NPY may be provided to the site of 5 injury by contacting the tissue with a molecule that induces expression of a neuropeptide Y gene such as FGF. In other embodiments, an NPY fragment is provided to the site of injury by contacting the tissue with an enzyme for converting NPY to a fragment. An example of such an enzyme is DPPIV. In still further embodiments, the NPY is provided to the site of injury by contacting the tissue with an agent for 10 preventing the degradation of NPY. Further, in certain embodiments, NPY is provided to the site of injury by providing a nucleic acid that when expressed provides NPY, or an agonist or antagonist of a NPY receptor to the site of injury.
WO 2006/108218 PCT/AU2006/000481 27 Further, in certain embodiments, NPY is provided to the site of injury by providing a cell that expresses or otherwise produces NPY to the site of injury. Examples of such cells include stem cells, progenitor cells and precursor cells. These are described further below. Other examples of cells include NPY cell transfectants that express NPY. 5 Thus in another embodiment there is provided a method for inducing regeneration of a mammalian tissue at a site of injury in the tissue, the method including the step of providing a cell that expresses NPY or a fragment thereof to a site of injury in a mammalian tissue to induce regeneration of the tissue at the site of injury. In certain embodiments, the cell is an olfactory ensheathing glial cell or an olfactory cell. 10 Typically the tissue is human tissue, although it will be understood that the agent may. be useful for inducing regeneration in other mammalian tissues. Further, the NPY, one or more fragments or agonist or antagonist of a NPY receptor thereof is typically selected to correspond to the species from which the NPY, fragment thereof, agonist or antagonist may be obtained. 15 Typically the tissue to be regenerated is selected from the group consisting of skin, muscle, fat, bone, or any tissue derived from the group of endoderm, mesoderm, ectoderm or combination thereof and including bone, cartilage, muscle, connective tissue, tendon, nerve adipose, skin, gastrointestinal tissue, heart, organs, cornea, optical tissue, exocrine and/or endocrine glands. For example if subcutaneous fatty 20 tissue were to be regenerated it would include include the regeneration of the primary cell type i.e. fat, and its blood supply (vascular tissue) nerve supply and stromal tissue (supporting structures including ECM, basil lamina etc). Similarly this concept can be used to support the regeneration of most tissues e.g. for muscle it will be myocytes, vascular supply and nerve supply and stromal tissue. 25 It will be understood that the method is particularly useful for the regeneration of tissue at a site of injury is selected from the group consisting of laceration, burns, surgical incisions and excisions/ removals/ debridements, abrasion, puncture, amputation, excision.
WO 2006/108218 PCT/AU2006/000481 28 The tissue to be regenerated may be tissue injured, lost, or atrophied by disease processes or degeneration. Such tissues could be the spinal cord (for example, multiple sclerosis), the substantia nigra in Parkinson's disease, or the olfactory mucosa or Alzheimer's disease. It will be understood that NPY may be provided in individuals 5 predisposed to multiple sclerosis, Parkinson's or Alzheimer's disease, or to individuals having symptoms of onset of these diseases for preventing or reducing the severity of these diseases. The method of the invention is particular useful for the therapy of individuals having a deficiency in wound healing. An example of individual is a diabetic. Thus in a particularly 10 preferred embodiment, the tissue is diabetic tissue. In certain embodiments, the invention is useful for the therapy of individuals requiring regeneration of spinal cord tissue. Thus the tissue may be spinal cord or neural tissue such as brain, spinal cord, peripheral nerves, optic nerves, retina, cranial nerves and autonomic nerves. 15 Where the site of injury is an external surface, in particular, skin, in certain embodiments the agent may be administered topically. Other forms of administration, such as oral,' nasal, subcutaneous, intra-peritoneal, per-rectal or intravenous administration may be selected in accordance with the location of the particular tissue injury. For example, ischaemic pancreatic tissue may best be treated by oral administration of the agent. 20 In one embodiment, the neuropeptide Y, variant or fragment thereof is provided in an amount of between about 1 to 500 ng per mm 3 of tissue to be regenerated. In certain embodiments, amounts within this range may be useful, for example from about 2 to 250 ng per mm 3 of tissue to be regenerated, 5 to 100 ng per mm 3 of tissue to be regenerated, and 10 to 50 ng per mm 3 of tissue to be regenerated. In most 25 circumstances, about 10 ng of NPY per mm 3 of tissue to be regenerated is a suitable amount for most tissue types. Appropriate amounts of NPY for tissue regeneration in given circumstances can be determined by one skilled in the art according to the pharmacokinetic properties of the delivery of the agent and the tissue to be regenerated, and the disclosures herein.
WO 2006/108218 PCT/AU2006/000481 29 In certain embodiments, and especially where tissue injury in dermis is to be treated by topical application, useful amounts of NPY would be from about 1 to 100ng per mm 2 of tissue to be regenerated, although other amounts could be used, for example from about 5 to 75ng per mm 2 of tissue to be regenerated, from about 7.5 to 50ng per mm 2 of 5 tissue to be regenerated and from about 10 to 25ng per mm 2 of tissue to be regenerated. It will be understood that the agent may be administered with other compounds, including compounds that facilitate or induce angiogenesis, neurogenesis, anti inflammatory compounds and antibiotic compounds. Compounds such as FGF's, basic 10 FGF, PDGF, IGF-1, EGF, SPARC, G-CSF, TGF-p, MCSF, IL-1, IL-IA, IL-3, IL-6, IL-7, IL-8, IL-1 1, Flt3 ligand, c-kit ligand (steel factor SF) and thrombopoietin (TPO) including sub-classifications thereof for example TGFa, TGFp1, and TGFp2 and the like may be used. It will be understood that the agent may be administered with either somatic stem cells 15 and/or other cells having plasticity such as some precursor cells, or with agents that induce plasticity in cells, or agents that promote the recruitment of stem cells to the site of injury e.g. GMSCF, SCF. This agent may be applied with the cells before, during or after the delivery of the cells to the site to be regenerated. In certain embodiments the cells express p75, otherwise known as CD271 or the low 20 affinity nerve growth factor receptor (LNGFR). Examples of these cells are olfactory ensheathing cells (OECs), olfactory stem cells, neural stem cells, neural progenitor cells, neurospheres, mesenchymal stem cells, oesophageal keratinocyte stem cells, haemopoietic stem cells and certain sub-populations of fibroblasts. The stem cells may be autologous (derived from the individual that it is desired to treat) 25 or allogeneic. Stem cells are typically identified/characterised by the presence or absence of one or more cell surface markers, which are also a useful means of isolating stem cells. For example, haemopoietic stem cells are CD34* and mesenchymal stem cells are STRO- WO 2006/108218 PCT/AU2006/000481 30 1*, SH2*, SH3* and those which express CD10, CD13, Thy-1, VCAM-1, CD29, CD49b/CD29, CD49e/CD29 and/or receptors for PDGF, EGF and IGF-1. Another marker for stem cells is p75 (also known as CD271, or LNGFR (low affinity nerve growth factor receptor). Embryonic stem cells express a variety of markers such as 5 alkaline phosphatase, SSEA-1 antigen and Oct-4. Another characteristic of stem cells is that they are relatively large, and often have indistinct cell morphology. Their size also provides a means of isolating stem cells from other cells as discussed below. Where progenitor or in other words, determined cells are used, these may be osteoblast, chrondoblast, hepatic progenitor, cardiac heamopoetic, neural progenitor or 10 neurospheres cells. Preferred cells include those which are p75 positive and/or cells which express proteins including STRO-1*, Lin-, c-kitPos, or bone marrow cells which express, CD9, CD10, CD13, Cd29, Cd34, Cd44, CD49d, CD49e, CD54, CD55, CD59, CD105, CD106, CD146 and CD166. 15 Precursor cells may also be used. These are generally understood to be cells that are determined but not differentiated, or in some instances, cells that have undergone some degree of differentiation and yet retain plasticity characteristics. Methods. for isolating and maintaining stem cells and progenitor cells have been developed in recent years and are known and available to persons skilled in the art. For 20 example, bone marrow mesenchymal stem cells (MSCs) can be isolated from bone marrow, blood, dermis, periosteum and umbilical cord blood. MSCs can be purified from these sources by, for example, flow cytometry or other cell sorting methods based on cell surface markers, Percoll density gradients, adherence to plastic surfaces or size sieving (see US patent no. 5,486,359; Hung et at., 2002, Stem Cells 20: 249-258). 25 Neural stem cells can be isolated from the central nervous system, embryonic/foetal tissue (see for example, Uchida et al., 2000, PNAS 97: 14720-14725). To prevent rejection of the cells upon introduction to the individual, it is preferred that the cells are immunocompatible with the individual, or the individual is WO 2006/108218 PCT/AU2006/000481 31 immunocompromised to reduce the risk of an immune response being mounted against the cells. For this reason, where adult cells are to be employed, it is preferable to use the individual's own stem cells, progenitor cells or precursor cells. Since stem cells are generally present in only small numbers in tissues, particularly 5 adult tissues, it is desirable to increase the number of stem cells available for treatment using in vitro expansion techniques. Suitable in vitro expansion methods for mesenchymal stem cells, using complete media (such as DMEM, F-12 (Ham) or BGJb) supplemented with FCS/FBS, or chemical defined media are described in US patent nos. 5,486,359 and 5,908,782; and Hung et 10 al., 2002, Stem Cells 20: 249-258). Suitable in vitro expansion methods for neural stem cells, via neurosphere formation, are described in, for example, Reynold and Weiss, 1992, Science 255: 1707. It will be understood that in certain embodiments, the plastic cells may be p75 negative i.e. they are cells that have little or no p75 molecules on the cell surface. 15 A further expansion method for olfactory stem cells is discussed below in the Examples. In a preferred embodiment, the stem cells, progenitor cells or precursor cells are expanded on or within a biomaterial scaffold, such as a biomaterial scaffold intended to be surgically implanted into an individual, as detailed below. Suitable biomaterials are also described below. A scaffold may also be conveniently used when dedifferentiating 20 mature cells to stem cells, progenitor cells and/or precursor cells as described above. In some embodiments of the invention, it may be preferable to proliferate the cells in vitro until the critical density of cells is reached, as discussed below. In such an embodiment, the NPY will be applied to the cells in vitro, and optionally also in vivo if necessary to promote differentiation of the cells. 25 It will be understood that the agent will typically be applied in the form of a medicament, such as a composition, formulation, dressing, suture or scaffold. Accordingly, in certain WO 2006/108218 PCT/AU2006/000481 32 embodiments, there is provided a use of an agent selected from the group consisting of neuropeptide Y, a fragment of neuropeptide Y; a compound for inducing expression of the neuropeptide Y gene; and an agonist or antagonist of a neuropeptide Y receptor, or a nucleic acid encoding anyone of these molecules, in the manufacture of a 5 medicament for inducing regeneration of a mammalian tissue at a site of injury in the tissue. The NPY peptide, fragment thereof, compound for inducing expression of NPY and agonist or antagonist of the NPY receptor, or nucleic acid encoding any one of these molecules useful in this embodiment of the invention are as described above. In certain embodiments the medicament further includes other compounds, including 10 compounds that facilitate or induce angiogenesis, neurogenesis, anti-inflammatory compounds and antibiotic compounds, examples of which are identified above. In certain embodiments, NPY is provided with a morphogen such as a retinoic acid (or variant), sonic hedgehog (or variant), or wnt protein (or variant). The medicament further comprises a pharmaceutically carrier, excipient, diluent or 15 lubricant. NPY may be associated with a mechanism which enables controlled release of the biologically active molecule, for example delayed, sustained or slow release. Suitable release mechanisms will be known to persons skilled in the art, and include microspheres or beads of materials such as Affigel TM (Bio-Rad), poly(D,L-lactic-co 20 glycolic acid) (PLGA), agarose, heparin, alginate, or gelatin. The NPY may be provided in the form of a pellet, such as those available from Innovative Research of America, Saratosa, FL, US. Typically, where the site of injury is in skin or subcutaneous tissue, the medicament is to be provided in a form for topical application, such as a liquid, for example a cream or 25 lotion, a semi- solid states, such as a gel or a solid state, such as a powder. A composition for the sustained release NPY, (discussed below) is particularly useful.
WO 2006/108218 PCT/AU2006/000481 33 In other embodiments, the medicament is provided on, or in the form of a dressing such as a bandage, gauze pad, adhesive plaster or other like surgical or therapeutic dressing. Accordingly, there is provided a dressing, suture or scaffold for inducing regeneration of 5 a mammalian tissue at a site of injury in the tissue including an agent selected from the group consisting of neuropeptide Y, a fragment of neuropeptide Y, a compound for inducing expression of the neuropeptide Y gene, an agonist or antagonist of a neuropeptide Y receptor, a nucleic acid encoding any one of these molecules, or a cell that expresses NPY. 10 Processes for the adsorption of a peptide or other compound onto a dressing or suture are known to one skilled in the art. The NPY peptide, a fragment thereof, a compound for inducing expression of NPY and an agonist or antagonist of the NPY receptor, nucleic acid for encoding any one of these compounds, or cell useful in this embodiment are as described above,. 15 In certain embodiments the medicament further includes other compounds, including compounds that facilitate or induce angiogenesis, neurogenesis, anti-inflammatory compounds and antibiotic compounds, examples of which are identified above. In one embodiment, a scaffold is employed to NPY peptide, a fragment thereof, a compound for inducing expression of NPY and an agonist or antagonist of the NPY 20 receptor, nucleic acid for encoding any one of these compounds, or cell for producing NPY. The scaffolds are preferably biocompatible, meaning that they fail to cause an acute immune reaction when introduced into the individual, and are generally three dimensional, preferably being shaped according to the desired shape of the tissue to be regenerated. For example, the tissue to be regenerated has a meniscus-like shape, the 25 scaffold preferably has a meniscus-like shape. The scaffold may not be in solid phase and maybe a gel or liquid.
WO 2006/108218 PCT/AU2006/000481 34 The scaffold may be biodegradable, or bioresorbable. Such scaffolds have the advantage that they break down and are absorbed by the body over time - preferably during, or after the desired tissue regeneration has occurred. The scaffold may be made from woven, non-woven, knitted, braided or crocheted 5 material, foam, sponge, or dendritic material. Suitable biodegradable materials for the scaffold include a polymer or copolymer such as those formed by a hydroxy acid (e. g., lactic acid), a glycolic acid, caprolactone, hydroxybutyrate, dioxanone, an orthoester, an orthocarbonate, or an aminocarbonate. Alternatively, or in addition, the material of the scaffold can include collagen, gelatin (e.g. Gelfoam), cellulose, fibrin, hyaluronic acid, 10 fibronectin, chitosan, or a devitalised graft (e. g., a devitalized xenograft or allograft). Ceramics, such as those formed with a mono-, di-, octa-, a-tri-, P-tri, or tetra-calcium phosphate, hydroxyapatite, fluoroapatite, calcium sulphate, calcium fluoride, or calcium oxide are also suitable. Bioactive silicon, whose bioresorbable characteristics can be, tailored to suit the particular application, can also be used. Non-biodegradeable 15 materials include, but are not limited to, polyesters, particularly aromatic polyesters, polyalkylene terephthalate (such as polyethylene terephthalate and polybutylene terephthalates): polyamides, polyalkenes, polyethylene and polypropylene; poly (vinyl fluoride), polytetrafluoroethylene, carbon fibres, natural or synthetic silk, ceramics and glass, or any mixture of these materials. An advantage of non-biodegradable materials 20 is that they retain their mechanical properties. Thus, their strength does not lessen over time. The scaffold may also comprise a hydrogel, to facilitate the transfer of cells and other biological material (e. g., growth factors) from the surrounding tissue into the scaffold. The hydrogel may be positively charged, negatively charged, or neutral hydrogel, and 25 saturated or unsaturated. Examples of suitable hydrogels include TetronicsTM and Poloxamines TM, which are poly (oxyethylene)-poly (oxypropylene) block copolymers of ethylene diamine; polysaccharides, chitosan, poly(vinyl amines), poly(vinyl pyridine), poly(vinyl imidazole), polyethylenimine, poly-L-lysine, growth factor binding or cell adhesion molecule binding derivatives, derivatised versions of the above (e.g., 30 polyanions, polycations, peptides, polysaccharides, lipids, nucleic acids or blends, block-copolymers or combinations of the above or copolymers of the corresponding WO 2006/108218 PCT/AU2006/000481 35 monomers); agarose, methylcellulose, hydroxyproylmethyl cellulose, xyloglucan, acetan, carrageenan, xanthan gum/locust beangum, gelatin, collagen (particularly Type 1), PluronicsTM, Poloxamers
TM
, poly (N-isopropylacrylmide) and N-isopropylacryhnide copolymers. 5 The volume/size of the scaffold will generally be slightly larger (to account for contraction and shrinkage which typically occurs) than, or will correspond to, the final size of the tissue to be regenerated. Preferably, the scaffold will have a volume/size such that it fits the selected site into which it is introduced during the appropriate surgical procedure. 10 The scaffold may be porous, or partially porous, thus allowing tissue in-growth. In a porous scaffold, cells can infiltrate most areas of the scaffold during regeneration. The pore diameter is determined by balancing the need for adequate surface area for tissue in-growth against the need for nutrients, other biological molecules, and/or water to reach the cells. The degree of porosity will depend upon the need for fast permeation of 15 cells and nutrients, and the need for mechanical integrity and strength. The scaffold can also include a shield to exclude in-growth of unwanted tissue phenotypes, such as those that are not, or do not produce, the tissue to be regenerated. Preferably, the shield is placed around the part of the scaffold adjacent to cells of the unwanted tissue type. Preferably, the shield will be sufficiently dense to prevent the 20 passage of cells, but porous enough to permit influx of nutrients and water and outflux of waste products. The shield can be removed prior to the completion of tissue regeneration. When introduced into the selected site, the scaffold may be secured within the selected site for regeneration in the preferred shape to occur, for example by suturing, pinning, 25 tacking, or stapling. The scaffold may be implanted into the selected site such that it is in contact with the native tissue of the selected site. Alternatively, the scaffold may be attached to juxtaposed tissue, for example, where the cornea is to be regenerated, the scaffold may be attached to juxtaposing endothelial or epithelial tissue. In this way, native cells of the selected site may associate with the scaffold. If such association is WO 2006/108218 PCT/AU2006/000481 36 not desired, the scaffold may be physically separated from the native tissue of the selected site. Where the scaffold is biodegradable or bioresorbable, it is preferred that it not significantly degrade until the tissue has reached the stage of regeneration where the 5 support offered by the scaffold is no longer required. Thus, it is particularly preferred that the scaffold will retain its shape and mechanical integrity until the desired tissue regeneration is substantially complete. As biodegradable and bioresorbable materials having varying in vivo degradation times are known, the degradation of the scaffold may be tailored to the requirements of the tissue to be regenerated. 10 The NPY, fragment thereof, cells for producing NPY or NPY gene can be incorporated into the scaffold using a variety of methods, which will be available to persons skilled in the art. For example, the cells can be injected directly into the scaffold. Where scaffold materials are made by polymerising components in solution to form a gel or solid material, the cells can be introduced into the solution prior to polymerisation. 15 Alternatively, in the case of spongy scaffold materials, the cells can be introduced by compressing the material, contacting the compressed material with a solution containing the cells, and allowing the material to expand, thereby taking up the cells into the material. Alternatively, the scaffold and cells will be introduced separately, in which case the cells 20 will become associated with the scaffold in vivo. In either case, the cells will preferably be associated with the scaffold by biological materials including polysaccharides, proteins, peptides genes, antigens, and antibodies, hormones, and cytokines. In certain embodiments it will be understood that NPY is provided to a site of injury in a mammalian tissue by providing NPY only. In these embodiments, further agents such 25 as cells, growth factors, cytokines and morphogens are not provided to the site of injury. Examples Example 1: Identification of NPY in an Axolotl blastema WO 2006/108218 PCT/AU2006/000481 37 Methods Animals All experiments were performed according to their guidelines. Laboratory axolotls were maintained in 60-liter tanks containing dechlorinated and filtered water at 18-20 C. 5 Water was changed weekly, and the animals were fed worms every 72hrs. The animal tanks were treated with a combination of solutions to mimic synthetic pond water: KCL 0.05g/L (2.5g) CaCl2 O.1g/L (5g) NaCL 3.4g/L (170g) exciting 10 Animals were used at the time point of 240hrs post amputation point. Animals were anesthetized with MS-222 (3-aminobenzoic acid ethyl ester, which was added to a separate litre of the water (0.1% w/v), and were subsequently killed by decapitation. Animal amputations All operations were performed on healthy animals under sterile conditions with 15 maximum time of operation being 1 Omins as follows: anaesthetic liquid composition (0.1% tricaine) record water temperature and pH weigh animal measure animal (record in millimeters) 20 wash animal with 2% bleach solution WO 2006/108218 PCT/AU2006/000481 38 Place animal in customised container with solution from step 1 to induce anaesthesia for 5min. Animal is submerged in solution to the torso level only. Regularly pinch limb in order to calculate time of amputation, ensure no reflex No sign of reflex and is flaccid, amputate hindlimb by cutting the limb bilaterally proximal 5 to the elbow under sterile conditions. Following amputation, dampened limb with 0.5% sulfamerazine for 1 min Return animal to tank for recovery Analyse and record behaviour of animal for 1 st hour and constantly monitor animal for remainder of the day. 10 Blastema tissue sample collection Tissue from axolotl regenerating blastema was collected at a time point of 240hrs (day 10), post-amputation as follows: Prepare anaesthesia liquid composition (0.5% tricaine) Prepare 2 batches of media (4 x 5ml aliquots to a total volume of 20ml) according to 15 tissue culture media composition and conditions aliquot 500pl of media from step 2 into a labeled 24well culture plate and place aside record water temperature and pH weigh animal (record to four decimal points) measure animal (record in millimeters) 20 Place animal in a customized container with solution from step 1 to induce euthanasia for 5min.
WO 2006/108218 PCT/AU2006/000481 39 pinch limbs, in order to calculate time of sacrifice, ensure no reflex sacrifice the animal by decapitation wash animal with 0.5% sulfamerazine wash animal three times in sterile PBS(-) 5 amputate hindlimb regenerating blastema by cutting just proximal of the knee with a scalpel and strip back the epidermis of the limb tissue, this will minimise contamination immediately after collection, tissue can be flash frozen in liquid nitrogen and stored at 800C. transfer tissue to labeled eppendorf tubes with fresh media, performed at 4*C or on ice 10 mince with an electronic tissue homogenizer for 1 min hand homogenized for 10-15 min Cell debris is to be removed by two centrifugation steps. spin at 2000g for 20 min aspirate supernatant and spin again at 100,000g for 60 min sonicate for 30 s, the insoluble lipid layer is aspirated and the remaining supernatant 15 filter sterilized through a 0.45m filter. Regenerating blastema sample preparation 1. Add 90 % (w/v) Tricholoracetic acid (TCA) to prepared tissue extract sample in a ratio of 1:10 (v/v). Mix samples and leave samples on ice for 3 hours (mixing well occasionally) or left overnight on ice. 20 2. Centrifuge samples for 15 min at 15,000 g, centrifuge must be refrigerated once rotor is in centrifuge WO 2006/108218 PCT/AU2006/000481 40 3. Pour off supernatant and resuspend pellet in 1 mL 40 mM acetic acid in ethanol. Use a sterile glass Pasteur pipette (tubes are narrow), and transfer the resuspended pellet into a sterile 1.5 mL eppendorf tube (sterile!).resuspend by gently pipetting up and then sonicate the tubes in the sonicating bath for about 1 min. 5 4. Centrifuge sample @ 15,000 g for 15 min. 5. Pour off supernatant and resuspend pellet in 100% ice cold acetone. Vortex and sonicate the sample for 1 min to wash protein well with 100% ice cold acetone. 6. Centrifuge pellet @ 15,000 g for 15 min to pellet protein. 7. Pour off supernatant and allow samples to air dry in laminar flow hood to ensure the 10 last couple of drops of acetone have been removed. Samples should now be a white powdery pellet. Pellets can be stored @ -80 0 C until use (RT if only left for a few days). Sample preparation for IEF - Sample Resolubilisation Samples for separation by 2D gels are solubilised in sample buffer compatible with the cell fraction and contain a combination of detergents. 15 Solubilisation Solution 8 M Urea 47 mL of 8.5M stock OR 24 g of dry Urea 100 mM DTT OR 770 mg OR 2 mM TBP 500 EL of 200 mM TBP stock 20 4% (w/v) CHAPS 2 g 0.2 % Carrier ampholytes 250 uL (40% Bio-lyte 3/10 or 4/7) WO 2006/108218 PCT/AU2006/000481 41 0.02 % Bromophenol Blue 100 uL of 0.1% Stock 40 mM Tris Base (pH 7) 242 mg IPG Strip Passive Re-hydration IPG strips (18cm) pH 3-10) were passively re-hydrated overnight. Custom made 2 mL 5 serological pipettes were used for procedure. Remove the IPG strip protective backings and place the IPG strips within the 2 mL pipettes. Pipette the 350 ml of resolublized protein samples on top of gel side of strips. Parafilm open end of pipette. Place pipettes with IPG strips inside, gel side up during rehydration. First Dimension Setup and Sample Application 10 Cooling unit was set at 12 0 C. In order to ensure uniform cooling between components, a film of low viscosity paraffin oil is poured onto the electrophoresis unit cooling plate. The strip tray is placed in position over the oil and the electrodes linked. Additional parafin oil is placed in the strip tray prior to placement of the plastic strip-aligning sheet. The re-swelling solution is drip drained from IPG strips and loaded parallel on the strip 15 aligning sheet (cathode end at top). Moist electrode wicks are positioned across the ends of the IPG strips and cathodic and anodic electrodes placed in contact with the wicks to absorb any surfeit salts. Additional paraffin oil (to fill strip tray half full) is poured into strip tray. Set the power pack (EPS 3501 XL - Amersham) as outlined below and run. 20 Preparation of IPG strips for 2 nd dimension electrophoresis After the first dimension run, proteins in the IPG strips are resolubilised, reduced and alkylated prior to loading onto the 2 nd dimension. Reduction of proteins is carried out by immersing the IPG strips in IPG equilibration solution. IPG Equilibration Solution 25 Component Purpose Amount [Final] WO 2006/108218 PCT/AU2006/000481 42 Urea -denaturation & solubilization 36 g 6 M of proteins SDS - solubilization of proteins 2 g 2 % 5x Tris/ - buffering (pH 8.8) 20 mL 1 x 5 HCI Buffer 50 % glycerol - inhibits electroendosmosis 40 mL 20% (i.e. the viscosity stops water transfer across the IPG strip) 10 DTT - reduction (breaks the 1.54 g 100 mM disulfide bonds) 25% Acrylamide - alkylation (prevents bonds from 10 mL 2.5% rejoining, compatibility for MS) 15 Second Dimension methods Gel Casting for 2DE SDS-PAGE Gel dimensions are 20 cm x 20 cm x 2mm. Gradient gel compositions are: 8% Solution Acrylamide 40% / PDA 49.62 mL WO 2006/108218 PCT/AU2006/000481 43 5x Tris / HCI Buffer 61.91 mL 10 % SDS 2.5 mL TEMED 123.8 OL 10 % APS 618.9 OL 5 Water 132.75 mL 18% Solution Acrylamide 40% / PDA 111.6 mL 5x Tris / HCI Buffer 61.91 mL 10 % SDS 2.5 mL 10 TEMED 123.8 OL 10 % APS 618.9 OL 50 % Glycerol 70.73 mL Prior to addition of TEMED the gel solution is degassed under vacuum for 30 minutes. TEMED is added (33pl) with stirring and the acrylamide solution gravity fed into the gel 15 casting cassette. The cassette is filled to 0.5cm from the top of the plates and the gels overlaid with sec-butanol for at least 3 hours. Following polymerisation the butanol is washed off and replaced by MQ water. The gels are left overnight at RT prior to loading the second dimension. Transfer of IPG Strips to the Second Dimension WO 2006/108218 PCT/AU2006/000481 44 Remove the polymerized gels from the cassettes taking care not to separate the gels from their glass plates. Wash the glass surfaces with tap water (to removed excess polymerized acrylamide gel) then assemble the cassettes to run the 2 nd dimension. Following equilibration, the 18cm IPG strips are embedded into an agarose solution. 5 Agarose Embedding Solution 0.5 % (w/v) Agarose 0.5 g 0.001 % Bromophenol Blue 1 mL of a 0.1 % Solution 1x Cathode Buffer make up to 100 mL boil the agarose embedding solution, and while very hot, pipette solution onto the 2D 10 well. Quickly insert the IPG strip through the agarose placing the strip directly on top of the polymerized gels. Have the gel side of the IPG strip facing out (front) so that it does not stick to the back plate. Load each 2D gel with hot agarose and then the IPG strip one at a time so that the agarose for each individual gel does not cool. Load a small piece of filter paper with 10 pL of MW ladder and also saturate in agarose embedding 15 solution to lock in MW ladder. Allow agarose to set for a few minutes, then load cassettes into running chamber (Ettan T M DALTsix Electrophoresis Unit - Amersham) and fill the tank with running buffer SDS-PAGE Running Buffer for Glycine Gels Glycine 144 g 20 SDS 10 g Tris Base 30 g make up to 10 L (will be enough for 2 runs) and check that the pH is 8.3 WO 2006/108218 PCT/AU2006/000481 45 Second Dimension Electrophoresis Conditions Start the 2 nd dimension at 180 V, 240 mA, 50 W. Once the gel has been running for a few hours, you will see that it will begin to slow down (proteins are encountering an increasing acrylamide gradient). After a couple hours the electrophoresis conditions 5 can be increased up to 280 V, 50 mA/gel. Be sure to calculate the W that result from the set V and mA so that the W do not become the limiting condition. Protein Detection and Visualization 2DE Gel Fixinq Following second dimensional runs, gels are fixed in a solution of 10 10 % (v/v) methanol 7 % (v/v) acetic acid for a minimum of 2 hours. This is visualised by a change in colour of bromophenol blue to a greenish/yellow colouration. Fluorescent Sypro Ruby Staining and Casting 15 After fixing all gels are stained with Sypro Ruby for 4 hours to overnight. This was achieved with 200 mL of Sypro Ruby stain per gel and placed on an orbital shaker at RT. Gels are then destained in 10% (v/v) methanol 20 7% (v/v) acetic acid WO 2006/108218 PCT/AU2006/000481 46 for 3 hours. Protein detection is achieved with a Molecular imager system with the program, at a pixel resolution of 200pm and at a wavelength of 618nm. Coomassie G-250 Staining Gels are Coomassie Brilliant Blue G-250 stained for protein visualization and spot 5 excision. Coomassie G-250 Stain (Modified Neuhoff Stain) Ammonium Sulfate 170 g Phosphoric Acid 36 mL of an 86 % solution Coomassie G-250 1 g 10 (Bio-Rad Cat# 161-0406) Methanol 340 mL Water to make up to 1 L Cover gel completely with Coomassie stain (- 200 mL per gel), and stain for -3-4 hours (proteins begin to appear) to overnight for quantitative staining. After staining, detection 15 is enhanced by placing the gel in 1 % acetic acid. Protein Identification by MALDI-ToF MS Spot Excision Protein spot was manually excised from gel using custom cut P1000 pipette tip (cut pipette tips approx 1 cm from bottom of tip). Gentle pressure onto the gel encloses the 20 protein area. Transfer spot into 1.5 mL eppendorf tube and store at 40C until clean up or in freezer for long-term storage.
WO 2006/108218 PCT/AU2006/000481 47 Protein Cleavage by in-gel digestion Gel pie6e was destained by washing twice in 120 pl 25 mM NH 4
HCO
3 containing 50 % (v/v) acetonitrile (solution can be made up and stored at RT for extended periods time just check that pH is ~ 8) for 30 min followed by drying in a Speedivac (approximately 5 15min). Gel piece is rehydrated in 16 tL trypsin solution (15 ng/ml trypsin in 25 mM
NH
4
HCO
3 ). Incubate for about 1 hr to allow trypsin to reswell gel pieces. Then add an additional 20 pL of 10 mM NH 4
HCO
3 that does not contain enzyme. Incubate @ 370C for 16 - 24 hours. Subsequent to incubation, centrifuge the eppendorf tube containing the gel piece for 2 min @ 7000 rpm and then sonicate the gel piece for 20 min. 10 Sample clean up prior to MS The trypsin extracts are then concentrated with C18 Millipore ZipTips. This procedure involves washing the ZipTip with 1 OpI of 30 % (v/v) acetonitrile (twice) then lOpI of 0.5 % (v/v) TFA (twice) 15 Extracted-in-gel-digested protein sample was loaded onto the ZipTip. Sample was loaded by pipetting. The sample was repeatedly pipetted up and down with the tip in the sample until you notice that the sample is no longer pipetting (protein has filled the column). Peptides are eluted from the ZipTip, directly onto the MALDI target plate, using 3pl of 10 mg/mL matrix (a-cyano-4-hydroxycinnamic acid) in 80% (v/v) acetonitrile 20 containing 0.08 % (v/v) TFA (solution is also stable for prolonged periods of time). Plates are then allowed to dry at RT. Peptide mass fingerprinting Digested samplewas dried at RT prior to MALDI. Peptide mass spectra was generated on a Voyager DE-STR mass spectrometer. The instrument is used with delayed 25 extraction in reflectron mode with an accelerating voltage of 20000V. Laser power varied between 1200 and 1600 over 200 shots. Peptide mass in the range 800 to 3500 WO 2006/108218 PCT/AU2006/000481 48 Da are measured. Peptide mass assignments were performed following internal calibration on trypsin peaks occurring at 842.51 and 2211.11Da. Once spectrum was generated the spectra was dragged into the data explorer and is internally calibrated with the trypsin autolysis peaks. It is important to deisotope the 5 spectrum (peaks / peak deisotoping). Export the peak list to Mascot for peptide matching (Tools / Macros / Toolbox Palette / Bottom choice / Mascot). Results Axolotl regenerating limb blastema peptide mass fingerprinting matched to: P33689 (NEUY XENLA) Neuropeptide Y precursor; C-flanking peptide of NPY (CPON)] 10 Neuropeptide Y precursor Due to limited data in bank, the most prominent and first match was within the same species of amphibian. 15 Nominal mass (Mr): 11370; Calculated pl value: 5.76 NCBI BLAST search of P33689 MQGNMRLWMSVLTLCLSMLICLGTFAEAYPSKPDNPGEDAPAEDMAKYYSALRHYIN LITRQRYGKRSSPETMLSDVWWRENTENIPRSRFEDPPMW >P33689 20 (NEUYXENLA) Neuropeptide Y precursor [Contains: Neuropeptide Y (Neuropeptide tyrosine) (NPY); C-flanking peptide of NPY Unformatted sequence string Taxonomy matched: Xenopus laevis 25 Variable modifications: Oxidation (M) Cleavage by Trypsin: cuts C-term side of KR unless next residue is P Sequence Coverage: 31% 30 Matched peptides underlined 1 MQGNMRLWMS VLTLCLSMLI CLGTFAEAYP SKPDNPGEDA PAEDMAKYYS 51 ALRHYINLIT RQRYGKRSSP ETMLSDVWWR ENTENIPRSR FEDPPMW 35 Start - End Observed Mr (expt) Mr (calc) Delta Miss Sequence 81 - 88 972.52 971.51 971.47 0.05 0 ENTENIPR 89 - 97 1180.69 1179.68 1179.50 0.18 1 SRFEDPPMW Oxidation(M) 40 68 - 80 1609.72 1608.71 1608.72 -0.01 0 SSPETMLSDVWWR Oxidation M) 67 - 80 1765.76 1764.75 1764.83 -0.08 1 RSSPETMLSDVWWR Oxidation(M) WO 2006/108218 PCT/AU2006/000481 49 S i 0 1000O 12501 1500 1750 RiS error 83 ppm Mass (Da) Protein separation in the first dimension of the 2-Dimensional electrophoresis gel is 5 isolated according to protein differences in amino acid sequences, therefore generating a difference in isoelectric point. The second dimension is run according to molecular weight (Figure 1). The excised protein spot was identified through the mascot programme in swissprot data base as Neuropeptide Y. The nominal mass (Mr): 11370; Calculated pl value: 5.76. 10 This result indicates that the process of tissue regeneration requires the influence and involvement of NPY. This result was generated at a time point of 240hrs which is the early stage of the regenerating process. At this stage of the process the cells are undergoing cellular reprogramming and are induced to obtain plasticity, thus become pluripotent. Due to the protein spot outlined in figure 1 is dark and intense, it is likely that 15 NPY has a strong influence on pluripotency capacity whilst cells undertake reprogramming. Example 2: Inducing regeneration of mammalian tissue at a site of tissue injury with NPY. 32 male Wistar rats were anaesthetized by an intramuscular injection of Ketamine and 20 Xylaxine. The dorsum was shaved and disinfected. Two full thickness skin wounds of 1cm (square shaped) were made.
WO 2006/108218 PCT/AU2006/000481 50 In each rat, in one wound a single slow release pellet of NPY containing either 0.2tg, 2pg, 20tg, or 2 00tg was placed. In the other wound a control pellet (not containing NPY) was placed. 8 rats were assigned to each dose treatment. Animals were allowed to recover and wound size was assessed every 3 days for 15 5 days. The animals were sacrificed at 6 weeks, and wounds were both grossly and histologically examined. Both wounds were excised en bloc and placed into fixative (10% buffered formalin). Multiple cross sections were cut across the wound area perpendicular to epidermis to include epidermis, dermis and subcutaneous tissue. 10 Sections were processed, embedded in paraffin and cut into 5pm thick sections. The sections were then stained with Haernatoxylin and Eosin and Masson's Trichrome. Tissue regeneration was then evaluated by assessment of the following parameters: A. Wound length was measured with an optical micrometer as the distance between intact epidermis. Re-epithelisation was calculated by the measuring new epithelium as a 15 percentage of wound length. B. Collagen deposition was assessed in dermis in the centre of the wound and graded semi quantitatively as 0 (no collagen deposition); 1+ (slight collagen deposition); 2+ (moderate collagen deposition); 3+ (heavy collagen deposition). C. Degree of angiogenesis was assessed by number of capillary sprouts in dermis. 20 D. Degree of inflammation was assessed by evaluating the number of neutrophils, lymphocytes, macrophages, plasma cells, eosinophils and foreign body type of giant cells semi quantitatively on a scale of 0 to 3 ( 0- absent, 1-mild, 2-moderate and 3 severe). E. Any necrosis (or absence thereof) was noted.
WO 2006/108218 PCT/AU2006/000481 51 F. Cell proliferation is assessed by special stains ki67 and PCNA. G. Regeneration of the epidermal appendages was noted by their reinstallation . H. Overall regeneration was scored as 1- poor regeneration;. 2 - moderate regeneration; 3 - good regeneration of epidermis and dermis; 4 - regeneration of 5 epidermis, dermis and evidence of re-installation of the epidermal appendages Example 3: Inducing regeneration of mammalian tissue at a site of tissue injury with an endothelial cell. In this example, NPY is provided to a wound site for tissue regeneration according to 10 Example 2. However, NPY is provided to the wound site by providing and endothelial cell that expresses NPY. An example of this cell is shown in Kaipio K. et al. 2005 Biochem. Biophys. Res. Comm. 337:633-640. Example 4: Inducing regeneration of mammalian tissue at a site of tissue injury with an NPY transfectant that expresses soluble NPY. 15 In this example, NPY is provided to a wound site for tissue regeneration according to Example 2. However, NPY is provided to the wound site by providing an olfactory endothelial transfectant that expresses NPY. Briefly, olfactory stem cells were harvested from male PVG/c rats and expanded in vitro as follows. 20 Cold Tissue culture medium containing: 1x 10ml (cold) and 2x 30ml -DMEM/HAM F12 -10% foetal calf serum, WO 2006/108218 PCT/AU2006/000481 52 -penicillin/streptomycin/fungizone (100 U/ml, 100 and 2.5 mg/ml) Primary tissue extraction/biopsies 1. Take medium-sized biopsies from the nasal septum in the superior region of the nasal cavity close to the cribriform plate 5 2. Excise the tissue using forceps/blade and place immediately into a sterile container in cold culture medium and all further processing is done under aseptic conditions. 3. Subsequent to extraction allow cell culture of the olfactory mucosa to rest for 2 hrs, invert tube from time to time 4. Then incubate the tissue for 45 min at 37C in 2 ml of a 2.4 U/ml dispase II solution 10 5. Separate the lamina propria from the overlying epithelium under a dissection microscope using a microspatula and cut into pieces of 0.05 mm using a tissue chopper, 6. Take the minced lamina propria and incubated at 37C in collagenase H (0.3%; Sigma) for 45 mins and mechanically triturated every 5 min using a flame polished Pasteur pipette. 15 7. Stop the enzymatic activity by adding a 0.5mM EDTA solution 8. Pellet the dissociated cells at 300g 9. Plate onto poly-L-lysine (100micograms/ml or 1 g/cm2, Sigma) pre-treated dishes and grown at 37C in 95% 02/5% C02 for 48hours subsequent to initial plating 10.Wash cells with PBS and change the serum-containing medium to DMEM/HAM F12 20 supplemented with penicillin/streptomycin and 10% FCS. 11. Change media every 2 days and passage cells at intervals when they reach confluence and plate into T75 cm2 flasks with the procedure repeated over 4 weeks to WO 2006/108218 PCT/AU2006/000481 53 build up an adequate supply of cells. Harvested by EDTA/PBS or trypsinisation after which the enzymatic activity is stopped by soybean trypsin inhibitor (Invitrogen). El-deleted adenoviral vectors were produced. Recombinant Adenovirus with NPY (Adv NPY) was created by inserting cDNA encoding recombinant human preproNPY (rh 5 NPY) into the plasmid vector pAC-cytomegalovirus using standard procedures (see Kaipio K. et al. 2005 Biochem. Biophys. Res. Comm. 337:633-640). OSC's were then transduced using Adenovirus at an multiplicity of infection (MOI) of 100 with Adv-NPY for 24 hours, to create OSC-Adv-NPY. After 24 hours, the media is changed and fresh media added for a further 48hours. NPY expression was confirmed by via 10 immunostaining, and also by testing the of supernatants from transduced cells by Western blot and ELISA assay. Biological activity of NPY was tested. OSC-Adv-NPY were then transplanted back into syngeneic PVG/c rats into bone defects. The rats were sacrificed at 6 weeks, and bones were examined radiographically and histologically for bone regeneration. 15 Example 5: Inducing regeneration of porcine tissue at a site of injury with NPY. Four healthy, male domestic pigs weighing approximately 20-30 kg were sedated with Ketamine 20 mg/kg IM +Xylazine, 2.0 mg/kg IM. An endotracheal tube was placed while maintaining spontaneous ventilation and anaesthesia maintained using isoflurane (0.5%-2.0%) administered in a mixture of 20 appropriate oxygen/nitrous oxide. Each animal underwent four aseptic dorsal skin incisions. In brief local hair was shaved to clear way for four 2cm full thickness skin incisions. Each incision was made down to a depth of 10mm. A standard sterile number 22 blade (Swann-Morton) was used. One incision was allocated per quadrant of dorsal skin as not to interfere with the other 25 incisions. All incisions were produced with a single stroke to a premarked measured line. All wounds were closed with a subcutaneous layer of 3-0 braided absorbable Vicryl suture (Ethicon Ltd, UK).
WO 2006/108218 PCT/AU2006/000481 54 1mg of Porcine NPY (Sigma, Catalogue number N3266) was dissolved in serial dilutions of 5mls of 3mg/mL pH neutral collagen gel giving 0.2mg/mL, 0.02mg/mL and 0.002mg/mL concentrations of NPY. An additional gel with no NPY was used as a control. The same solution was applied topically daily to the same wound edge for 5 5 days under Tegaderm (3M ) transparent adhesive dressings to localize the NPY-gel to the site. Within each pig there was an incision exposed to each concentration of NPY: 0-, 0.2-, 0.02- and 0.002 mg/mL of NPY. Buprenorphine, 0.075 mg/kg IMI was administered immediately after surgery for analgesia with additional doses provided as required. 10 Wound sites were observed and evaluated subjectively for wound edge approximation, progression of regeneration, evidence of inflammation or infection, and incisional dehiscence daily for the first 5 days after surgery and on a regular basis thereafter. At 28 days animals were euthanized and wound sites were harvested en bloc as a 3 x 6 cm squares and placed in 10% buffered formalin histopathologic examination. 15 Histopathologic specimens for evaluation pf regeneration and scoring were fixed in 10% buffered formalin for a minimum of 48 hours. Cross sections of each wound were processed using standard methods, embedded in paraffin, cut at 5 pm, and mounted on glass slides. Individual sections for each wound were stained and were examined under a light microscope for assessment as based on Example 2. 20 Example 6: Inducing regeneration of muscle tissue at a site of injury with NPY. 16 PVG/c rats (6-8 weeks old) were induced under 5% Isoflurane, and then maintained under 2% Isoflurane. Each rat was assigned to one of four groups: (1) Control (Pellet alone) (n=4 rats) (2) 0.2ug NPY Pellet (n=4 rats) 25 (3) 2ug NPY Pellet (n=4 rats) WO 2006/108218 PCT/AU2006/000481 55 (4) 20ug NPY Pellet (n=4 rats) (5) 200ug NPY pellet (n=4rats) A 1cm lateral skull incision was made. Superficial lateral cranial muscles were exposed (primarily temporalis muscle). A circular defect of 6mm in muscle was incised using a 5 6mm burr on a slow dental drill. A single pellet containing a dose of Human NPY (either 0 (pellet alone), 0.2- , 2- or 20-ug) designed to release over 28days (Innovative research of America) was placed in each defect. Both the muscle defect and the skin incision were closed with 5-0 Maxon absorbable sutures. The animals were returned to their cage and be allowed to move freely after recovery. 10 Post operative monitoring is initiated. Supplementary intraoperative and peri-operative Temgesic 0.02-0.05mg/kg IM or SC ie start prior to recovery was given as required. Rats were euthanased at 6 weeks. Muscle tissue was harvested en bloc and histological sections of 5 um were prepared. Muscle defects were graded for regeneration using standard histological stains as per Example 2, 15 immunohistochemistry and image analysis.

Claims (14)

1. A method for inducing regeneration of a mammalian tissue at a site of injury in the tissue including providing neuropeptide Y to a site of injury in a mammalian tissue to induce regeneration of the tissue at the site of injury. 5
2. A method for inducing regeneration of a mammalian tissue at a site of injury in the tissue including providing a fragment of neuropeptide Y to a site of injury in a mammalian tissue to induce regeneration of the tissue at the site of injury.
3. A method for inducing regeneration of a mammalian tissue at a site of injury in the tissue including providing an agonist or antagonist of a neuropeptide Y receptor to a 10 site of injury in a mammalian tissue to induce regeneration of the tissue at the site of injury.
4. A method according to claim 1 wherein the NPY is provided to the site of injury in the tissue by providing a cell that produces NPY to the site of injury in the tissue.
5. A method according to claim 1 wherein the NPY is provided to the site of injury in 15 the tissue by providing a nucleic acid encoding NPY to the site of injury in the tissue.
6. A method according to claim 1 further including providing an agent selected from the group consisting of FGF, PDGF, EGF, GMSCF and SCF to the site of injury in the tissue.
7. A method according to claim 1 further including providing a morphogen such as a 20 retinoic acid to the site of injury in the tissue.
8. A method according to claim 1 further including providing a wnt protein to the site of injury in the tissue.
9. A method according to claim 1 further including providing a stem cell, progenitor cell, or a precursor cell that is capable of differentiating to regenerate the elements of 25 the tissue. WO 2006/108218 PCT/AU2006/000481 57
10. A method according to claim 9 wherein the cell is an epithelial stem cell, olfactory stem cell, neural stem cell, neural progenitor cell, neurosphere, or mesenchymal stem cell.
11. A method according to claim 1 further including providing an olfactory 5 ensheathing cell.
12. A method according to claim 1 wherein the neuropeptide Y is provided to the site of injury in the tissue on a dressing.
13. A method according to claim 1 wherein the neuropeptide Y is provided to the site of injury in the tissue on a scaffold. 10
14. A method according to claim 1 wherein the tissue is neural tissue.
AU2012216342A 2005-04-15 2012-08-22 Use of neuropeptide Y (NPY) and agonists and antagonists thereof for tissue regeneration Abandoned AU2012216342A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2012216342A AU2012216342A1 (en) 2005-04-15 2012-08-22 Use of neuropeptide Y (NPY) and agonists and antagonists thereof for tissue regeneration

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
NZ539441 2005-04-15
NZ545383 2006-02-16
AU2006235200A AU2006235200A1 (en) 2005-04-15 2006-04-10 Use of Neuropeptide Y (NPY) and agonists and antagonists thereof for tissue regeneration
AU2012216342A AU2012216342A1 (en) 2005-04-15 2012-08-22 Use of neuropeptide Y (NPY) and agonists and antagonists thereof for tissue regeneration

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU2006235200A Division AU2006235200A1 (en) 2005-04-15 2006-04-10 Use of Neuropeptide Y (NPY) and agonists and antagonists thereof for tissue regeneration

Publications (1)

Publication Number Publication Date
AU2012216342A1 true AU2012216342A1 (en) 2012-09-06

Family

ID=46785950

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2012216342A Abandoned AU2012216342A1 (en) 2005-04-15 2012-08-22 Use of neuropeptide Y (NPY) and agonists and antagonists thereof for tissue regeneration

Country Status (1)

Country Link
AU (1) AU2012216342A1 (en)

Similar Documents

Publication Publication Date Title
AU2006235200A1 (en) Use of Neuropeptide Y (NPY) and agonists and antagonists thereof for tissue regeneration
JP2548414B2 (en) Biosynthetic bone morphogenetic protein and osteogenic device containing the protein
ES2255059T3 (en) BMP-12, BMP-13 AND YOUR INDUCTION COMPOSITIONS OF TENDON.
US8691944B2 (en) Fibronectin polypeptides and methods of use
CN105025917B (en) Peptides and compositions for treating joint damage
JP2004041236A (en) Osteogenic device
US20100292161A1 (en) Polypeptides and methods of use
EP1261365B1 (en) Product for biological anchoring of connective tissue to bone
WO2018121235A1 (en) Peptide for repairing cartilage and treating osteoarthritis
EP3250223A1 (en) Methods for the regeneration of articular cartilage in vivo
US7504099B2 (en) Methods of inducing or enhancing connective tissue repair
WO2005011722A2 (en) Use of vegf-c or vegf-d in reconstructive surgery
JP2009142287A (en) Frazzled nucleotide sequence, expression product, composition and use
EP2310035A2 (en) Fibroblast growth factor (fgf) analogs and uses thereof
US20030228292A1 (en) Methods of inducing or enhancing connective tissue repair
AU2012216342A1 (en) Use of neuropeptide Y (NPY) and agonists and antagonists thereof for tissue regeneration
US20100215722A1 (en) Tissue Regeneration II
JP2023049978A (en) Neurite elongation promoter, and pharmaceutical composition for inducing neuroregeneration
Vazquez-Portalatin The Use of Biopolymers for Tissue Engineering
CA3235565A1 (en) Truncated and fusion proteins
AU2013219428A1 (en) Tissue regeneration II
AU4586402A (en) Frazzled nucleotide sequences expression products compositions and uses

Legal Events

Date Code Title Description
MK4 Application lapsed section 142(2)(d) - no continuation fee paid for the application