WO2007048202A1 - Methods and compositions for modulating wound repair - Google Patents
Methods and compositions for modulating wound repair Download PDFInfo
- Publication number
- WO2007048202A1 WO2007048202A1 PCT/AU2006/001610 AU2006001610W WO2007048202A1 WO 2007048202 A1 WO2007048202 A1 WO 2007048202A1 AU 2006001610 W AU2006001610 W AU 2006001610W WO 2007048202 A1 WO2007048202 A1 WO 2007048202A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cell
- expression
- activity
- agent
- flightless
- Prior art date
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/70—Carbohydrates; Sugars; Derivatives thereof
- A61K31/7088—Compounds having three or more nucleosides or nucleotides
- A61K31/713—Double-stranded nucleic acids or oligonucleotides
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P17/00—Drugs for dermatological disorders
- A61P17/02—Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P43/00—Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
- C07K14/4701—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
- C07K14/4702—Regulators; Modulating activity
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/505—Medicinal preparations containing antigens or antibodies comprising antibodies
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2317/00—Immunoglobulins specific features
- C07K2317/70—Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
- C07K2317/74—Inducing cell proliferation
Definitions
- the present invention relates to methods and compositions for modulating wound repair and scar formation.
- the present invention also relates to methods and compositions for preventing and/or treating diseases, conditions and states associated with undesired or abnormal fibrosis, and to methods and compositions for modulating migration and/or proliferation of cells involved in wound repair, scar formation or fibrosis.
- the present invention further relates to isolated skin cells with altered expression of a regulator of wound repair.
- Wound healing is a complex and dynamic process that results in the restoration of cellular structures and tissue layers.
- the wound healing process can be divided into 3 distinct phases: the inflammatory phase, the proliferative phase, and the remodeling phase. Each of these phases involves a complex and coordinated series of events that includes chemotaxis, phagocytosis, neocollagenesis, collagen degradation, and collagen remodeling.
- the recruitment of a variety of specialised' cell types to the site of a wound is also a critical part of the process of wound healing.
- This process requires extracellular matrix and basement membrane deposition, angiogenesis, selective protease activity and re- epithelialisation.
- One component of the healing process in mammals is the stimulation of fibroblasts to generate the extracellular matrix.
- This extracellular matrix constitutes a major component of the connective tissue that develops to repair the wound area.
- the actin cytoskeleton is an essential network of filaments found in all cells. Reorganisation of the actin cytoskeleton is central to changes in cell adhesion and motility that underpin wound repair processes. These changes include the lammellipodial crawling of keratinocytes during wound re-epithelialisation, infiltration of inflammatory cells and migration of fibroblasts required for the deposition and remodelling of the extracellular matrix and dermal contraction at the wound site.
- Actin assembly and organisation Many regulatory proteins influence actin assembly and organisation. For example, members of the gelsolin family of proteins appear to regulate actin filaments by severing pre-existing filaments and/or capping the filament ends. After severing, the proteins remain attached to the "barbed" ends of the broken filament, thereby preventing annealing or addition of actin monomers. Actin filaments are subsequently uncapped by interaction with phosphoinositides, leading to rapid actin assembly. This is the first step in enabling cells to reorientate their cytoskeleton to drive changes in motility, adhesion and contraction.
- Fibrotic disorders are characterised by the accumulation of fibrous tissue (predominately collagens) in an abnormal fashion within the tissue. Accumulation of such fibrous tissues may result from a variety of disease processes.
- Flightless I a member of the gelsolin family
- repair of a wound may be modulated by modulating the activity and/or expression of this protein.
- the present invention arises out of studies into the role of the Flightless I protein in wound repair.
- the present studies demonstrate that modulating the activity of Flightless I regulates wound healing.
- Flightless I is a target for manipulation to improve wound healing and scar formation, and to prevent and/or treat fibrotic disorders.
- the present invention provides a method of modulating repair of a wound, the method including modulating expression and/or activity of Flightless I in cells involved in repair of the wound.
- the present invention also provides use of an agent that modulates expression and/or activity of Flightless I in the preparation of a medicament for modulating wound repair.
- the present invention also provides a pharmaceutical composition when used to modulate repair of a wound, the composition including an effective amount of an agent that modulates expression and/or activity of Flightless I.
- the present invention also provides a method of modulating scar formation in a wound of a subject, the method including the step of delivering to the wound an effective amount of an agent that modulates expression and/or activity of Flightless I.
- the present invention also provides use of an agent that modulates expression and/or activity of Flightless I in the preparation of a medicament for modulating scar formation.
- the present invention also provides a pharmaceutical composition when used to modulate scar formation, the composition including an effective amount of an agent that modulates expression and/or activity of Flightless I.
- the present invention also provides a method of preventing and/or treating a disease, condition or state associated with undesired or abnormal fibrosis in a subject, the method including the step of administering to the subject a therapeutically effective amount of an agent that modulates expression and/or activity of Flightless I.
- the present invention also provides use of an agent that modulates expression and/or activity of Flightless I in the preparation of a medicament for preventing and/or treating a disease, condition or state associated with undesired or abnormal fibrosis.
- the present invention also provides a pharmaceutical composition when used to prevent and/or treat a disease, condition or state associated with undesired or abnormal fibrosis, the composition including an effective amount of an agent that modulates expression and/or activity of Flightless I.
- the present invention also provides a method of modulating one or more of expression, secretion and/or activity of TGF- ⁇ and/or collagen, the method including the step of modulating expression and/or activity of Flightless I in a cell.
- the present invention also provides use of an agent that modulates the expression and/or activity of Flightless I in the preparation of a medicament for modulating one or more of expression, secretion and/or activity of TGF- ⁇ and/or collagen.
- the present invention also provides a pharmaceutical composition when used to modulate one or more of expression, secretion and/or activity of TGF- ⁇ and/or collagen, the composition including an effective amount of an agent that modulates expression and/or activity of Flightless I.
- the present invention also provides use of an agent that modulates expression and/or activity of Flightless I in the preparation of a medicament for modulating wound repair.
- the present invention also provides a method of modulating migration and/or proliferation of a cell involved in wound repair, scar formation or fibrosis, the method including the step of modulating the expression and/or activity of Flightless I in the cell.
- the present invention also provides use of an agent that modulates the expression and/or activity of Flightless I in the preparation of a medicament for modulating migration and/or proliferation of a cell involved in wound repair, scar formation or fibrosis.
- the present invention also provides a pharmaceutical composition when used to modulate migration and/or proliferation of a cell involved in wound repair, scar formation or fibrosis, the composition including an effective amount of an agent that modulates expression and/or activity of Flightless I.
- the present invention also provides an isolated skin cell, or a progenitor or derivative thereof, the cell having an altered expression and/or activity of Flightless I.
- the present invention also provides an isolated skin cell, or a progenitor or derivative thereof, the cell including a nucleic acid that modulates Flightless I expression and/or activity in the cell.
- Flightless I as used throughout the specification will be understood to mean a protein with at least 50% sequence identity to human Flightless I.
- a Flightless I protein may be identified, for example, by the BLAST algorithm, as described in Altschul et al. (199O) J. MoI. Biol. 215:403-410.
- module as used throughout the specification is to be understood to mean a promotion or inhibition of a process.
- an increase in rate and/or extent of wound repair may result from either an increase or decrease in the expression and/or activity of Flightless I.
- an inhibition in rate and/or extent of wound repair may also result from either an increase or decrease in the expression and/or activity of Flightless I.
- a similar situation applies to the case of modulating the rate and/or extent of scar formation, and the modulation of proliferation and/or migration of cells involved in wound healing.
- the modulation of Flightless I expression and/or activity in cells involved in repair (healing) of a wound, or in cells involved in scar formation is a modulation of the expression and/or activity in one or more cells involved in these processes, directly or indirectly, and includes cell types such as fibroblasts, keratinocytes, endothelial cells, neutrophils, macrophages, and other inflammatory cells.
- the modulation of the expression and/or activity of Flightless I includes within its scope one or more of a modulation of the Flightless I protein level, an alteration in the activity of the Flightless I protein, an alteration in the intracellular and/or extracellular localisation of the protein, and an alteration in the rate or level of secretion of the protein from a cell.
- biological system as used throughout the specification is to be understood to mean any multi-cellular system and includes isolated groups of cells to whole organisms.
- variant as used throughout the specification is to be understood to mean an amino acid sequence of a polypeptide or protein that is altered by one or more amino acids.
- the variant may have "conservative” changes, wherein a substituted amino acid has similar structural or chemical properties to the replaced amino acid (e.g., replacement of leucine with isoleucine).
- a variant may also have "non-conservative” changes (e.g., replacement of a glycine with a tryptophan) or a deletion and/or insertion of one or more amino acids.
- nucleic acid as used throughout the specification is to be understood to mean any oligonucleotide or polynucleotide.
- the nucleic acid may be DNA or RNA and may be single stranded or double stranded.
- the nucleic acid may be any type of nucleic acid, including a nucleic acid of genomic origin, cDNA origin (ie derived from a mRNA), derived from a virus, or of synthetic origin.
- an oligonucleotide or polynucleotide may be modified at the base moiety, sugar moiety, or phosphate backbone, and may include other appending groups to facilitate the function of the nucleic acid.
- the oligonucleotide or polynucleotide may be modified at any position on its structure with constituents generally known in the art.
- an oligonucleotide may include at least one modified base moiety which is selected from the group including 5-fluorouracil, 5-bromouracil, 5-chlorouracil, 5- iodouracil, hypoxanthine, xanthine, 4-acetylcytosine, 5- (carboxyliydroxylmethyl) uracil,5-carboxymethylaminomethyl-2-thiouridine, 5-carboxymethylaminomethyluracil, dihydrouracil, beta-D-galactosylqueosine, inosine, N6-isopentenyladenine, 1- methylguanine, 1-methylinosine, 2,2-dimethylguanine, 2-methyladenine, 2- methylguanine, 3-methylcytosine, 5-methylcytosine, N6-adenine, 7-methylguanine, 5- methylaminomethyluracil, 5-methoxyaminomethyl-2-thiouracil, beta D- mannos
- the oligonucleotide or polynucleotide may also include at least one modified sugar moiety selected from the group including, but not limited to, arabinose, 2- fluoroarabinose, xylulose, and hexose.
- the oligonucleotide or polynucleotide may include at least one modified phosphate backbone, such as a phosphorothioate, a phosphorodithioate, a phosphoramidothioate, a phosphoramidate, a phosphordiamidate, a methylphosphonate, an alkyl phosphotriester, and a formacetal or any analogue thereof.
- subject as used throughout the specification is to be understood to mean any multicellular organism, including an animal or human subject.
- the subject may be a human or other mammal, a primate, a livestock animal (eg. a horse, a cow, a sheep, a pig, or a goat), a companion animal (eg. a dog, cat), a laboratory test animal (eg. a mouse, a rat, a guinea pig, a bird), an animal of veterinary significance, or an animal of economic significance.
- isolated as used throughout the specification is to be understood to mean that a material is removed from its original environment, such as removed from its natural environment if it is naturally-occurring.
- a naturally-occurring polynucleotide, polypeptide or cell present in a living animal is not isolated, but the same polynucleotide, polypeptide or cell separated from some or all of the coexisting materials in the natural system, is isolated, even if subsequently reintroduced into the natural system.
- Figure 1 shows that wounding upregulates FIiI expression in human fibroblasts and keratinocytes.
- Confluent human fibroblasts and keratinocytes HaCaTs were scratch- wounded and stained for FIiI and actin by immunocytochemistry. Representative images are shown, (a) Unwounded (UnW) fibroblasts stained for FIiI (red), (b) composite of FIiI and actin (green).
- FIG. 2 shows that FIiI is upregulated in response to wounding
- Figure 3 shows that FIiI overexpression decreases cell proliferation, reduces migration and impairs wound healing.
- (b) Primary fibroblasts derived from wild- type and FIiI overexpressing mouse skin were cultured until confluent, scratch-wounded and the resultant wound area measured at various time-points post wounding. Results represent mean +/- s.e.m. (n 6, *P ⁇ 0.05 vs equivalent wild-type time point ).
- FIG. 4 shows that FIiI siRNA reduces FIiI gene and protein expression.
- RNA was extracted from human fibroblasts treated with increasing concentrations of FIiI siRNA for 24h.
- Figure 5 shows that FIiI deficiency increases cell proliferation, enhances migration and improves wound healing.
- Human fibroblasts were treated with FIiI siRNA (10OnM) for 24 hours and the resulting effect on (a) cell proliferation and (b) migration determined using a WST-I proliferation assay and a scratch wound assay respectively.
- Keratinocytes were treated with FIiI siRNA (60 nM.
- FIG. 6 shows that proliferation is impaired in FIiI overexpressing wounds
- WT WiId- type
- TgI FIiI deficient (+/-)
- PCNA stained cells within the wounds Closed arrows point to non-proliferating cells.
- FIG. 7 shows that FHI neutralising antibodies increase fibroblast proliferation.
- Fibroblasts were scratch-wounded and after 24 hours conditioned medium collected,
- Figure 8 shows the effect of FIiI Antibodies on human keratinocytes.
- Human keratinocytes HaCats
- HaCats Human keratinocytes
- FIiI neutralizing antibodies a) FIiL,
- FIiG antisera a FIiG antisera from Santa- Cruz (CA).
- Figure 9 shows the effect of FIiI Antibodies on human skin fibroblasts.
- Human skin fibroblasts were incubated with increasing concentrations of FIiI neutralizing antibodies (a) FIiL, (b) FIiG, (c) FIiG antisera, (d) FIiI antibody from Santa-Cruz (CA).
- Figure 10 shows the effect of FKL Antibody on FHI overexpressing skin fibroblasts.
- Primary skin fibroblasts cultured from FHI overexpressing skin were incubated with increasing concentrations of FHI neutralizing antibody FHL.
- FIG 11 shows that exogenous addition of FHI neutralising antibodies accelerates wound healing.
- FIG. 12 shows differential expression of FHI affects TGF- ⁇ and collagen I expression in wounds.
- Wounds from wild-type (WT), FHI deficient +/- (+/-) and FHI transgenic (TgI) mice were immunostained for TGF- ⁇ 1, TGF- ⁇ 3 and collagen I.
- (b) Fluorescence intensity of collagen I at 14 days post- wounding. * denotes significance and P ⁇ 0.05. Results represent means +/- s.e.m. n 12 for each group.
- the present invention provides a method of modulating repair of a wound, the method including modulating expression and/or activity of Flightless I in cells involved in repair of the wound.
- This present invention may be used to promote or inhibit the repair of healing of a wound present in a biological system.
- the present invention may be used to repair a wound in vivo, such as a wound in an entire human or animal subject.
- biological system as used in the various relevant embodiments of the present invention is to be understood to mean a multi-cellular system, and includes isolated groups of cells to whole organisms.
- the biological system may be a tissue or organ, or an entire human or animal subject.
- present invention may also be used to modulate wound healing of cells in vitro.
- Flightless I as used throughout the specification in the various embodiments of the present invention will be understood to mean a protein with at least 50% sequence identity to human Flightless I.
- a Flightless I protein may be identified, for example, by the BLAST algorithm which identifies local alignments between the sequences in the database and predicts the probability of the local alignment occurring by chance.
- the BLAST algorithm is as described in Altschul et al. (1990) J. MoI, Biol. 215:403-410.
- Flightless I is a member of the gelsolin family.
- the human homologue of the Flightless I gene encodes a 140 kD protein, originally identified in Drosophila where mutations in the gene cause defects in the flight muscles which, consequently, are unable to support flight.
- the Flightless I protein is highly conserved between the mouse and human, and is the most evolutionarily conserved member of the gelsolin family, suggesting that it carries out important, conserved functions.
- the amino acid sequence of the human form of Flightless I is provided in Genbank Accession No. NP_002009.
- the nucleotide sequence of the mRNA is provided in GenBank Accession No. NM_002018.
- the present invention may be used to promote or inhibit repair of a wound.
- wounds examples include acute wounds (such as penetrative injuries, burns, nerve damage and wounds resulting from elective surgery), chronic wounds (such is diabetic, venous and decubitus ulceration) or wounds in individuals with compromised wound healing capacity, such as the elderly.
- repair of the wound is promoted, hi this case, the expression and/or activity of Flightless I in cells involved in repair of the wound may be decreased.
- repair of the wound is inhibited, hi this case, the expression and/or activity of Flightless I in the cells involved in repair of the wound may be increased.
- an increase in rate and/or extent of wound repair may result from either an increase or decrease in the expression and/or activity of Flightless I.
- an inhibition in rate and/or extent of wound repair may also result from either an increase or decrease in the expression and/or activity of Flightless I.
- Flightless I activity may be associated with improved healing.
- the present invention involves the modulation of the expression and/or activity of Flightless I in cells involved in repair of a wound.
- the cells involved in repair of the wound are fibroblast cells or keratinocytes.
- the modulation of one or more of the expression, secretion and/or activity of the Flightless I protein in the various embodiments of the present invention may be accomplished by a suitable method.
- the modulation of Flightless I expression and/or activity includes delivering or exposing to the cells involved in repair of the wound an effective amount of an agent that modulates expression and/or activity of Flightless I.
- the agent may modulate the expression and/or activity of the Flightless I protein directly or indirectly.
- the agent is delivered to a wound in a subject to modulate repair of the wound.
- the agent may be applied to the wound directly and/or injected intradermally around the wound.
- the present invention provides a method modulating repair of a wound in a subject, the method including delivering to the wound an effective amount of an agent that modulates expression and/or activity of Flightless I in cells involved in repair of the wound.
- the subject may be, for example, a human or an animal subject, including a mammalian subject, a primate, a livestock animal (eg. a horse, a cow, a sheep, a pig, or a goat), a companion animal (eg. a dog, cat), a laboratory test animal (eg. a mouse, a rat, a guinea pig, a bird), an animal of veterinary significance, or an animal of economic significance.
- a mammalian subject a primate
- a livestock animal eg. a horse, a cow, a sheep, a pig, or a goat
- a companion animal eg. a dog, cat
- a laboratory test animal eg. a mouse, a rat, a guinea pig, a bird
- an animal of veterinary significance e.g. a human or an animal subject.
- the modulation of expression and/or activity of Flightless I may occur at any one or more of prior to, concurrently with, and/or after a wound has occurred.
- the present invention not only comtemplates the modulation of the expression and/or activity of Flightless I for the treatment of wounds after wounding has occurred, the present invention also contemplates modulation of the expression and/or activity of Flightless I before wounding has occurred.
- the subject in the various embodiments of the present invention may be a subject suffering from a wound in need of repair, or alternatively, be a subject susceptible to wounding.
- the agent may increase the expression and/or activity of Flightless I.
- Agents that may increase the expression and/or activity of Flightless I are as previously discussed herein, and include agents such as a nucleic acid encoding a functional part of the Flightless I protein, agents that activate transcription of the Flightless I gene, or the Flightless I protein itself (or a variant or active fragment thereof). Such agents may be used to reduce or inhibit repair of a wound, or to reduce the extent of scar formation associated with the repair of a wound.
- the agent may decrease the expression and/or activity of Flightless I.
- a decrease in the expression and/or activity of Flightless I may be achieved by a suitable method.
- a decrease in expression may be accomplished by the use of one or more of a neutralizing antibody (or an antigen binding part thereof), use of an antisense nucleic acid that binds to the mRNA and which interferes with translation, the use of a molecule that can specifically repress transcription of an endogenous mRNA such as a specific DNA or RNA binding protein, a nucleic acid capable of forming a triple helix structure, a small interfering RNA, a microRNA, a ribozyme that can cleave a specific mRNA, or an inhibitory agent that interacts with Flightless I (or a regulator of Flightless
- Such agents may be used to promote or increase repair of a wound.
- the agent that decreases the expression and/or activity of Flightless I is a neutralising antibody (or an antigen binding part thereof), including a neutralising antibody to Flightless I (or an antigen binding part thereof).
- antigen binding part is to be understood to mean the antigen-binding portion of an antibody molecule, including a Fab, Fab', F(ab') 2 , Fv, a single-chain antibody (scFv), a chimeric antibody, a diabody or any polypeptide that contains at least a portion of an immunoglobulin that is sufficient to confer specific antigen binding, such as a molecule including one or more CDRs.
- the neutralising antibody to Flightless I is an antibody that binds specifically to the leucine rich repeat domain of the Flightless I protein.
- Methods for producing antibodies, including methods for producing neutralising antibodies, are known in the art, for example as described in Antibodies: A Laboratory Manual (1988) by Ed Harlow and David Lane, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York.
- antisense nucleic acids these may be produced by a suitable method known in the art, including expression from a recombinant nucleic acid or by chemical synthesis.
- phosphorothioate oligonucleotides may be synthesized by the method as described in Stein et al. (1988) Nucl. Acids Res. 16: 3209.
- small interfering RNAs these may be produced by a method known in the art.
- methods of designing and using siRNAs to decrease the expression are essentially as described in Elbashir et al (2001) Nature AlV. 494-498, Harborth et al (2003) Antisense Nucleic Acid Drug Dev. 13: 83-106, and Semizarov et al (2003) Proc Natl Acad Sd USA 100: 6347-6352.
- ribozymes In the case of ribozymes, the functional constraints necessary for a nucleic acid to act as a ribozyme are essentially as described in Haseloff et al. (1988) Nature 334: 585-591; Koizumi et al, (1988) FEBS Lett. 228: 228-230; Koizumi et al, (1988) FEBS Lett. 239: 285-288).
- Ribozyme methods that involve inducing expression in a cell of ribozyme molecules are essentially as described in Grassi and Marini (1996) Annals of Medicine 28: 499-510 ; Gibson (1996) Cancer and Metastasis Reviews 15: 287-299.
- ribozyme action involves sequence-specific hybridization of the ribozyme molecule to complementary target RNA, followed by endonucleolytic cleavage.
- ribozymes which may be used include engineered hammerhead motif ribozyme molecules that can specifically and efficiently catalyze endonucleolytic cleavage of sequences encoding the target sequence.
- ribozyme cleavage sites within any potential RNA target are initially identified by scanning the target molecule for ribozyme cleavage sites which include the following sequences: GUA, GUU, and GUC. Once identified, short RNA sequences of between 15 and 20 ribonucleotides corresponding to the region of the target gene containing the cleavage site may be evaluated for secondary structural features which may render the oligonucleotide inoperable. The suitability of candidate targets may also be evaluated by testing accessibility to hybridization with complementary oligonucleotides using ribonuclease protection assays, as known in the art.
- the antibody in the various embodiments of the present invention may be a polyclonal antibody, a monoclonal antibody, a chimeric antibody, a single chain antibody, a Fab fragment, and fragments produced by a Fab expression library.
- various hosts including goats, rabbits, rats, mice, humans, and others, may be immunized by injection with the polypeptide or any fragment or oligopeptide thereof that has immunogenic properties.
- various adjuvants may be used to increase immunological response.
- Such adjuvants include, but are not limited to, Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol.
- Monoclonal antibodies may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture. These include, but are not limited to, the hybridoma technique, the human B-cell hybridoma technique, and the EBV-hybridoma technique (essentially as described in Kohler, G. et al. (1975)
- Antibody fragments which contain specific binding sites may also be generated.
- fragments include F(ab') 2 fragments which can be produced by pepsin digestion of the antibody molecule and the Fab fragments which can be generated by reducing the disulfide bridges of the F(ab') 2 fragments.
- Fab expression libraries may be constructed to allow rapid and easy identification of monoclonal Fab fragments with the desired specificity (for example as described in Huse, W. D. et al. (1989) Science 254:1275-1281).
- a neutralising antibody to Flightless I may be produced by immunizing an animal against the leucine rich repeat domain of the protein.
- Various features of the Flightless I protein may be found in UniProtKB/Swiss-Prot entry Q13045.
- the agent in the various embodiments of the present invention may also cause an alteration in the intracellular and/or extracellular localisation of Flightless I.
- the agent may cause re-localisation of Flightless I from the cytoplasm of the cell to the nucleus of the cell, or re-localisation of Flightless I from the nucleus to the cytoplasm.
- the agent in the various embodiments of the present invention may also modulate one or more of the expression, secretion, localisation and/or activity of a molecule upstream and/or downstream of Flightless I that is involved in wound repair.
- the agent may modulate one or more of the expression, secretion and activity of a TGF- ⁇ protein, and/or modulate one or more of the expression, secretion and/or activity of collagen.
- TGF- ⁇ is a group of multifunctional proteins that control proliferation, differentiation, and other functions in many cell types.
- Members of the TGF- ⁇ family include TGF- ⁇ 1, TGF- ⁇ 2, and TGF- ⁇ 3.
- the proteins all function through the same receptor signalling systems. Methods for identifying TGF- ⁇ proteins are known in the art.
- TGF- ⁇ as used throughout the specification will be understood to mean a protein with at least 50% sequence identity to human TGF- ⁇ 1.
- a TGF- ⁇ protein may be identified, for example, by the BLAST algorithm.
- the amino acid sequence of the human form of TGF- ⁇ 1 is provided in Genbank Accession No. NP_000651.
- the nucleotide sequence of the mRNA is provided in GenBank Accession No. NM_000660.
- the amino acid sequence of the human form of TGF- ⁇ 2 is provided in Genbank Accession No. NP_003229.
- the nucleotide sequence of the mRNA is provided in GenBank Accession No. NM_003238.
- the amino acid sequence of the human form of TGF- ⁇ 3 is provided in Genbank Accession No. NP_003230.
- the nucleotide sequence of the mRNA is provided in GenBank Accession No. NM__003239.
- the agent may decrease expression and/or activity of TGF- ⁇ l, and/or increase the expression and/or activity of TGF- ⁇ 3. In the case where wound repair is decreased, the agent may increase the expression and/or activity of TGF- ⁇ 1.
- the agent in the various embodiments of the present invention may also modulate the expression, activity and/or secretion of collagen in cells involved in repair of the wound.
- Collagen is a major component of skin, cartilage, bone, and other connective tissue.
- types of collagen including type I collagen, and type II collagen.
- the agent may modulate the expression and/or secretion of collagen I.
- the amino acid sequence of the human form of collagen, type I, alpha 1 is provided in Genbank Accession No. NPJ300079.
- the nucleotide sequence of the mRNA is provided in GenBank Accession No. NM_000088.
- the agent may decrease the expression, activity and/or secretion of collagen I.
- the agent may increase the expression, activity and/or secretion of collagen I.
- the present invention may also be used to prepare a medicament for modulating wound repair.
- the present invention provides the use of an agent that modulates expression and/or activity of Flightless I in the preparation of a medicament for modulating wound repair.
- the agent may be used in the preparation of a medicament for improving wound repair.
- the agent may decrease the expression and/or activity of Flightless I.
- An example of such an agent is a neutralising antibody to Flightless I (or an antigen binding part thereof), such as an antibody to the leucine rich repeat domain of the protein.
- the agent may be delivered to the wound by a suitable method known in the art. Methods of delivery include direct exposure of cells in the wound to the agent, such as by the use of a topical composition, intradermal injection of the agent to the wound and/or around the wound, and administration of the agent to a subject.
- the agent may be administered to the subject in a suitable form to effect modulation of the expression and/or activity of Fhightless I in cells involved in repair of the wound.
- the effective amount of agent to be delivered or administered is not particularly limited, so long as it is within such an amount and in such a form that generally exhibits a pharmacologically useful or therapeutic effect.
- an effective amount of the agent may be appropriately chosen, depending upon, for example, the type of wound, the mode of delivery, the age and body weight of the subject, the frequency of delivery or administration, and the presence of other active agents.
- the agent is delivered or administered as a pharmaceutical composition to modulate repair of a wound.
- the present invention provides a pharmaceutical composition when used to modulate repair of a wound, the composition including an effective amount of an agent that modulates expression and/or activity of Flightless I.
- a neutralising antibody to Flightless I may be formulated into a topical composition for topical administration to a wound, or the neutralising antibody may be formulated into a composition for injection into one or more regions near or surrounding the wound.
- the present invention provides a topical wound healing composition, the composition including a therapeutically effective amount of an agent that reduces expression and/or activity of Flightless I in cells involved in wound repair.
- the topical wound healing composition is a composition including a therapeutically effective amount of a neutralising antibody to Flightless I.
- Topical compositions including an antibody are generally as described in US patent 5,702,946.
- the present invention may also be used to modulate scar formation.
- the present invention provides a method of modulating scar formation in a wound of a subject, the method including the step of delivering to a wound in a subject an effective amount of an agent that modulates expression and/or activity of Flightless I.
- Flightless I may be used to modulate scar formation.
- the agent may delivered to the wound by administration of the agent to the wound and/or a region(s) near the wound.
- the agent may be formulated into a topical composition for administration to the wound, and/or the agent may be formulated into a composition for injection into a region(s) near and/or surrounding the wound.
- scar formation is reduced.
- the agent may increase the expression and/or activity of Flightless I in cells involved in repair of a wound.
- the present invention may also used in the preparation of a medicament for modulating scar formation.
- the present invention provides the use of an agent that modulates expression and/or activity of Flightless I in the preparation of a medicament for modulating scar formation.
- the medicament is used to reduce scar formation.
- the agent may increase the expression and/or activity of Flightless I in cells involved in repair of a wound.
- the agent may be prepared into a pharmaceutical composition to modulate scar formation.
- the present invention provides a pharmaceutical composition when used to modulate scar formation, the composition including an effective amount of an agent that modulates expression and/or activity of Flightless I.
- the composition is a topical composition.
- the present invention may also be used to prevent and/or treat a disease, condition or state associated with undesired or abnormal fibrosis in a subject.
- the present invention provides a method of preventing and/or treating a disease, condition or state associated with undesired or abnormal fibrosis in a subject, the method including the step of administering to the subject a therapeutically effective amount of an agent that modulates expression and/or activity of Flightless I.
- the agent modulates expression and/or activity of Flightless I in cells involved with the fibrosis.
- diseases, conditions and states associated with abnormal or undesired fibrosis include atherosclerosis; fibrotic diseases of the lung, liver, kidney and cardiovascular system; excessive wound healing; and cancer. Accordingly, the present invention may be used to prevent and/or treat such diseases, conditions or states.
- the agent increases the expression and/or activity of Flightless I. Examples of such agents are as previously described herein.
- the present invention may also be used in the preparation of a medicament for preventing and/or treating a disease, condition or state associated with undesired or abnormal fibrosis.
- the present invention provides the use of an agent that modulates expression and/or activity of Flightless I in the preparation of a medicament for preventing and/or treating a disease, condition or state associated with undesired or abnormal fibrosis.
- the agent may also be prepared into a pharmaceutical composition used for preventing and/or treating a disease, condition or state associated with undesired or abnormal fibrosis.
- the present invention provides a pharmaceutical composition when used to prevent and/or treat a disease, condition or state associated with undesired or abnormal fibrosis, the composition including an effective amount of an agent that modulates expression and/or activity of Flightless I.
- the present invention may also be used to modulate one or more of expression, secretion and/or activity of TGF- ⁇ and/or collagen.
- the present invention provides a method of modulating one or more of expression, secretion and/or activity of TGF- ⁇ and/or collagen, the method including the step of modulating expression and/or activity of Flightless I in a cell.
- the cell is a cell involved in wound repair and/or scar formation, such as a fibroblast or a keratinocyte.
- the cell is a human cell or an animal cell, such as a cell present in vivo in a human or animal.
- the cell may be a cell involved in repair of a wound in a subject, a cell involved in scar formation in a subject, or a cell involved with fibrosis in a subject.
- the method is used to reduce the extent or rate of fibrosis in the subject.
- the expression and/or activity of TGF- ⁇ l is modulated
- increasing expression and/or activity of Flightless I in the cell results in an increase in expression and/or activity of TGF- ⁇ l in the cell and/or an increase in the expression and/or secretion of collagen in the cell. Under these conditions, the extent of scarring of a wound, or the extent or rate of fibrosis, in a subject is reduced.
- the expression and/or activity of TGF- ⁇ 3 is modulated.
- the expression and/or secretion of collagen type I is modulated.
- the present invention may also be used for the preparation of a medicament for modulating expression and/or activity of TGF- ⁇ and/or collagen.
- the present invention provides the use of an agent that modulates the expression and/or activity of Flightless I in the preparation of a medicament for modulating expression and/or activity of TGF- ⁇ and/or collagen.
- the present invention may also be used in a pharmaceutical composition used to modulate expression and/or activity of TGF- ⁇ and/or collagen.
- the present invention provides a pharmaceutical composition when used to modulate expression and/or activity of TGF- ⁇ and/or collagen, the composition including an effective amount of an agent that modulates expression and/or activity of Flightless I.
- the present invention may also be used to modulate migration and/or proliferation of a cell involved in wound repair, scar formation or fibrosis.
- the present invention provides a method of modulating migration and/or proliferation of a cell involved in wound repair, scar formation or fibrosis, the method including the step of modulating the expression and/or activity of Flightless I in the cell.
- the cell is a fibroblast, a keratinocyte or an inflammatory cell.
- the cell is a human cell or an animal cell, such as a cell present in vivo in a human or animal.
- the cell may be a cell involved in repair of a wound in a subject, a cell involved in scar formation in a subject, or a cell involved with fibrosis in a subject.
- decreasing expression and/or activity of Flightless I increases migration and/or proliferation of a cell.
- increasing expression and/or activity of Flightless I decreases migration and/or proliferation of a cell.
- the modulation of expression and/or activity of Flightless I is achieved by delivering or exposing the cell to an agent that modulates the expression and/or activity of Flightless I.
- the agent may be delivered to the cells by exposing the cells to the agent, or in the case of a subject, administering the agent to the subject.
- the agent decreases the expression and/or activity of Flightless I in the cell.
- An example of such an agent is a neutralising antibody or an antigen binding portion thereof, such as a neutralising antibody to Flightless I. Examples of antibodies are previously described herein.
- the present invention may also be used for the preparation of a medicament for modulating migration and/or proliferation of a cell involved in wound repair, scar formation or fibrosis.
- the present invention provides the use of an agent that modulates the expression and/or activity of Flightless I in the preparation of a medicament for modulating migration and/or proliferation of a cell involved in wound repair, scar formation or fibrosis.
- the present invention may also be used for the preparation of a pharmaceutical composition to modulate migration and/or proliferation of a cell involved in wound repair, scar formation or fibrosis. Accordingly, in another embodiment the present invention provides a pharmaceutical composition when used to modulate migration and/or proliferation of a cell involved in wound repair, scar formation or fibrosis, the composition including an effective amount of an agent that modulates expression and/or activity of Flightless I.
- the delivery or administration of the agent in the various embodiments of the present invention may be within any time suitable to produce the desired effect of directly or indirectly modulating the expression and/or activity of Flightless I.
- the modulation of expression and/or activity of Flightless I in the cell may occur at any one or more of the time of wounding, during healing of the wound and prior to wounding.
- the modulation of expression and/or activity of Flightless I in the cell may occur at either or both of prior to fibrosis occurring, and during fibrosis.
- the agent in the various embodiments of the present invention may be delivered, exposed or administered by a suitable method.
- the agent may be administered orally, parenterally, topically, by injection, systemically or by any other suitable means, and therefore transit time of the agent must be taken into account.
- the agent may for example be administered directly to the wound and/or a region(s) near and/or surrounding the wound.
- the agent may be delivered to the site of fibrosis directly and/or administered to a subject so as to reach the site of fibrosis.
- the agent may be administered systemically.
- the delivery or administration of the agent in the various embodiments of the present invention may be delivery or administration of the agent alone, or delivery or administration of the agent formulated into a suitable pharmaceutical composition.
- the pharmaceutical composition may also include the use of one or more pharmaceutically acceptable additives, including pharmaceutically acceptable salts, amino acids, polypeptides, polymers, solvents, buffers, excipients and bulking agents, taking into consideration the particular physical and chemical characteristics of the agent to be administered.
- the agent can be prepared into a variety of pharmaceutical compositions in the form of, e.g., an aqueous solution, an oily preparation, a fatty emulsion, an emulsion, a gel, etc., and these preparations can be administered as intramuscular or subcutaneous injection or as injection to an organ, or as an embedded preparation or as a transmucosal preparation through nasal cavity, rectum, uterus, vagina, lung, etc.
- the composition may be administered in the form of oral preparations (for example solid preparations such as tablets, capsules, granules or powders; liquid preparations such as syrup, emulsions or suspensions).
- Compositions containing the agent may also contain a preservative, stabiliser, dispersing agent, pH controller or isotonic agent.
- suitable preservatives are glycerin, propylene glycol, phenol or benzyl alcohol.
- suitable stabilisers are dextran, gelatin, a-tocopherol acetate or alpha- thioglycerin.
- suitable dispersing agents include polyoxyethylene (20), sorbitan mono-oleate (T ween 80), sorbitan sesquioleate (Span 30), polyoxyethylene (160) polyoxypropylene (30) glycol (Pluronic F68) or polyoxyethylene hydrogenated castor oil 60.
- suitable pH controllers include hydrochloric acid, sodium hydroxide and the like.
- suitable isotonic agents are glucose, D-sorbitol or D-mannitol.
- the administration of the agent in the various embodiments of the present invention may also be in the form of a composition containing a pharmaceutically acceptable carrier, diluent, excipient, suspending agent, lubricating agent, adjuvant, vehicle, delivery system, emulsifier, disintegrant, absorbent, preservative, surfactant, colorant, flavorant or sweetener, taking into account the physical and chemical properties of the agent being administered.
- a pharmaceutically acceptable carrier diluent, excipient, suspending agent, lubricating agent, adjuvant, vehicle, delivery system, emulsifier, disintegrant, absorbent, preservative, surfactant, colorant, flavorant or sweetener, taking into account the physical and chemical properties of the agent being administered.
- composition may be administered orally, parenterally, by inhalation spray, adsorption, absorption, topically, rectally, nasally, bucally, vaginally, intraventricularly, via an implanted reservoir in dosage formulations containing conventional non-toxic pharmaceutically-acceptable carriers, or by any other convenient dosage form.
- parenteral as used herein includes subcutaneous, intravenous, intramuscular, intraperitoneal, intrathecal, intraventricular, intrasternal, and intracranial injection or infusion techniques.
- the composition When administered parenterally, the composition will normally be in a unit dosage, sterile injectable form (solution, suspension or emulsion) which is preferably isotonic with the blood of the recipient with a pharmaceutically acceptable carrier.
- sterile injectable forms are sterile injectable aqueous or oleaginous suspensions. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
- the sterile injectable forms may also be sterile injectable solutions or suspensions in non-toxic parenterally- acceptable diluents or solvents, for example, as solutions in 1,3-butanediol.
- any bland fixed oil may be employed including synthetic mono- or di-glycerides, corn, cottonseed, peanut, and sesame oil.
- Fatty acids such as ethyl oleate, isopropyl myristate, and oleic acid and its glyceride derivatives, including olive oil and castor oil, especially in their polyoxyethylated versions, are useful in the preparation of injectables.
- These oil solutions or suspensions may also contain long- chain alcohol diluents or dispersants.
- the carrier may contain minor amounts of additives, such as substances that enhance solubility, isotonicity, and chemical stability, for example anti-oxidants, buffers and preservatives.
- additives such as substances that enhance solubility, isotonicity, and chemical stability, for example anti-oxidants, buffers and preservatives.
- the agent When administered orally, the agent will usually be formulated into unit dosage forms such as tablets, cachets, powder, granules, beads, chewable lozenges, capsules, liquids, aqueous suspensions or solutions, or similar dosage forms, using conventional equipment and techniques known in the art.
- Such formulations typically include a solid, semisolid, or liquid carrier.
- Exemplary carriers include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, mineral oil, cocoa butter, oil of theobroma, alginates, tragacanth, gelatin, syrup, methyl cellulose, polyoxyethylene sorbitan monolaurate, methyl hydroxybenzoate, propyl hydroxybenzoate, talc, magnesium stearate, and the like.
- a tablet may be made by compressing or moulding the agent optionally with one or more accessory ingredients.
- Compressed tablets may be prepared by compressing, in a suitable machine, the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, surface active, or dispersing agent.
- Moulded tablets may be made by moulding in a suitable machine, a mixture of the powdered active ingredient and a suitable carrier moistened with an inert liquid diluent.
- the administration of the agent in the various embodiments of the present invention may also utilize controlled release technology.
- the agent may also be administered as a sustained-release pharmaceutical.
- the agent may be formulated with additional components such as vegetable oil (for example soybean oil, sesame oil, camellia oil, castor oil, peanut oil, rape seed oil); middle fatty acid triglycerides; fatty acid esters such as ethyl oleate; polysiloxane derivatives; alternatively, water-soluble high molecular weight compounds such as hyaluronic acid or salts thereof (weight average molecular weight: ca. 80,000 to 2,000,000), carboxymethylcellulose sodium (weight average molecular weight: ca.
- hydroxypropylcellulose viscosity in 2% aqueous solution: 3 to 4,000 cps
- atherocollagen weight average molecular weight: ca. 300,000
- polyethylene glycol weight average molecular weight: ca. 400 to 20,000
- polyethylene oxide weight average molecular weight: ca. 100,000 to 9,000,000
- hydroxypropylmethylcellulose viscosity in 1% aqueous solution: 4 to 100,000 cSt
- methylcellulose viscosity in 2% aqueous solution: 15 to 8,000 cSt
- polyvinyl alcohol viscosity: 2 to 100 cSt
- polyvinylpyrrolidone weight average molecular weight: 25,000 to 1,200,000.
- the agent may be incorporated into a hydrophobic polymer matrix for controlled release over a period of days.
- the agent may then be moulded into a solid implant, or externally applied patch, suitable for providing efficacious concentrations of the agent over a prolonged period of time without the need for frequent re-dosing.
- Such controlled release films are well known to the art.
- Other examples of polymers commonly employed for this purpose that may be used include nondegradable ethylene- vinyl acetate copolymer a degradable lactic acid-glycolic acid copolymers which may be used externally or internally.
- Certain hydrogels such as poly(hydroxyethylmethacrylate) or poly(vinylalcohol) also may be useful, but for shorter release cycles than the other polymer release systems, such as those mentioned above.
- the carrier may also be a solid biodegradable polymer or mixture of biodegradable polymers with appropriate time release characteristics and release kinetics.
- the agent may then be moulded into a solid implant suitable for providing efficacious concentrations of the agent over a prolonged period of time without the need for frequent re-dosing.
- the agent can be incorporated into the biodegradable polymer or polymer mixture in any suitable manner known to one of ordinary skill in the art and may form a homogeneous matrix with the biodegradable polymer, or may be encapsulated in some way within the polymer, or may be moulded into a solid implant.
- the composition of the present invention may be in the form of a solution, spray, lotion, cream (for example a non-ionic cream), gel, paste or ointment.
- the composition may be delivered via a liposome, nanosome, or nutri-diffuser vehicle.
- a cream is a formulation that contains water and oil and is stabilized with an emulsifier.
- Lipophilic creams are called water-in-oil emulsions, and hydrophilic creams oil-in- water emulsions.
- the cream base for water-in-oil emulsions are normally absorption bases such as vaseline, ceresin or lanolin.
- the bases for oil-in-water emulsions are mono-, di- and triglycerides of fatty acids or fatty alcohols with soaps, alkyl sulphates or alkyl polyglycol ethers as emulsifiers.
- a lotion is an opaque, thin, non-greasy emulsion liquid dosage form for external application to the skin, which generally contains a water-based vehicle with greater than 50% of volatiles and sufficiently low viscosity that it may be delivered by pouring. Lotions are usually hydrophilic, and contain greater than 50% of volatiles as measured by LOD (loss on drying). A lotion tends to evaporate rapidly with a cooling sensation when rubbed onto the skin.
- a paste is an opaque or translucent, viscous, greasy emulsion or suspension semisolid dosage form for external application to the skin, which generally contains greater than 50% of hydrocarbon-based or a polyethylene glycol-based vehicle and less than 20% of volatiles.
- a paste contains a large proportion (20-50%) of dispersed solids in a fatty or aqueous vehicle. An ointment tends not to evaporate or be absorbed when rubbed onto the skin.
- An ointment is an opaque or translucent, viscous, greasy emulsion or suspension semisolid dosage form for external application to the skin, which generally contains greater than 50% of hydrocarbon-based or a polyethylene glycol-based vehicle and less than 20% of volatiles.
- An ointment is usually lipophilic, and contains > 50% of hydrocarbons or polyethylene glycols as the vehicle and ⁇ 20% of volatiles as measured by LOD. An ointment tends not to evaporate or be absorbed when rubbed onto the skin.
- a gel is usually a translucent, non-greasy emulsion or suspension semisolid dosage form for external application to the skin, which contains a gelling agent in quantities sufficient to impart a three-dimensional, cross-linked matrix.
- a gel is usually hydrophilic, and contains sufficient quantities of a gelling agent such as starch, cellulose derivatives, carbomers, magnesium-aluminum silicates, xanthan gum, colloidal silica, aluminium or zinc soaps.
- composition for topical administration may further include drying agents, anti- foaming agents; buffers, neutralizing agents, agents to adjust pH; colouring agents and decolouring agents; emollients; emulsifying agents, emulsion stabilizers and viscosity builders; humectants; odorants; preservatives, antioxidants, and chemical stabilizers; solvents; and thickening, stiffening, and suspending agents, and a balance of water or solvent.
- the agent may be delivered by way of a nucleic acid or vector that allows for expression of the agent in the appropriate target cells.
- the agent may be delivered by way of a viral vector that causes expression of the agent in target cells.
- the present invention specifically contemplates gene therapy methods, and in particular, gene therapy methods for correcting defects in diseases, conditions and states as previously described herein.
- Methods for gene therapy are known in the art.
- the cell in the various relevant forms of the present invention may be a cell in vitro, or a cell in a biological system.
- the present invention also provides an isolated skin cell, or a progenitor or derivative thereof, in which the expression and/or activity of Flightless I in the skin cell is altered.
- the present invention provides an isolated skin cell, or a progenitor or derivative thereof, the cell having an altered expression and/or activity of Flightless I.
- Isolated skin cells in which the expression and/or activity of Flightless I in the skin cell is altered, may have use as therapeutic or cosmetic agents.
- the cell is a fibroblast cell or a keratinocyte cell. Methods for altering the expression and/or activity of Flightless I in the cell are as previously herein described.
- an exogenous form of the protein or a regulator of the protein may be cloned and introduced into a cell by a suitable method known in the art.
- a nucleic acid may be isolated and cloned into a suitable expression vector for use in the cell type of interest by methods known in the art. Methods for the isolation of nucleic acid sequences and their cloning into a suitable expression vector are essentially as described in Sambrook, J, Fritsch, E.F. and Maniatis, T. Molecular Cloning: A Laboratory Manual 2nd. ed. Cold Spring Harbor Laboratroy Press, New York. (1989). The recombinant molecule may then be introduced into the cell and the cloned nucleic acid expressed.
- suitable vectors include plasmid vectors and viral vectors.
- the vector may also further include regulatory elements for the expression of inserted nucleic acids, for example inducible or constitutive promoters for driving the expression of an inserted nucleic acid in a particular cell type, poly A signals for efficient polyadenylation of mRNA transcribed from inserted nucleic acids, or other regulatory elements to control translation, transcription or mRNA stability, all known in the art.
- regulatory elements for the expression of inserted nucleic acids for example inducible or constitutive promoters for driving the expression of an inserted nucleic acid in a particular cell type, poly A signals for efficient polyadenylation of mRNA transcribed from inserted nucleic acids, or other regulatory elements to control translation, transcription or mRNA stability, all known in the art.
- the expression may be achieved by a number of methods known in the art. These include transient or stable transfection of cells with a recombinant nucleic acid encoding the gene of interest under the control of a promoter that is active in the particular cell type.
- a decrease in expression may be achieved by a number of methods known in the art.
- an antisense nucleic acid that binds to an endogenous mRNA and which interferes with translation the use of a molecule that can specifically repress transcription of the endogenous mRNA such as a specific DNA or RNA binding protein, a nucleic acid capable of forming a triple helix structure, a small interfering RNA (siRNA), a microRNA, or a ribozyme that can cleave a specific mRNA.
- siRNA small interfering RNA
- microRNA a microRNA
- ribozyme that can cleave a specific mRNA.
- the introduction of exogenous nucleic acids to decrease the expression of a target gene will involve constitutive expression in the cell of the nucleic acid.
- nucleic acids may be introduced into a cell by various methods, including transformation using calcium phosphate, viral infection, electroporation, lipofection, and particle bombardment.
- Methods for introducing DNAs into cells are essentially as described in Sambrook, J, Fritsch, E.F. and Maniatis, T. Molecular Cloning: A Laboratory Manual 2nd. ed. Cold Spring Harbor Laboratory Press, New York. (1989).
- the expression and/or activity of Flightless I in the cell may be altered for example by the introduction of an exogenous nucleic acid into the cell.
- the expression and/or activity of Flightless I may be altered by a manipulation of an endogenous nucleic acid in the cell, such as by alteration of the Flightless I gene itself or a regulator of Flightless I expression.
- the present invention also provides an isolated skin cell, or a progenitor or derivative thereof, wherein the cell includes a nucleic acid that modulates Flightless I expression and/or activity in the cell.
- the altered expression and/or activity of Flightless I is due to introduction of an exogenous nucleic acid into the cell. In another embodiment the altered expression and/or activity of Flightless I is due to an alteration of an endogenous nucleic acid in the cell.
- the nucleic acid is integrated into the genome, such as a suitable transgene.
- the cell may also be, for example, a transiently transfected cell.
- the isolated cell in the various forms of the present invention also has an altered expression and/or activity of TGF- ⁇ and/or an altered expression and/or secretion of collagen.
- the cells of the present invention may also be part of a whole animal. Accordingly, in another form the present invention provides an animal including one or more skin cells in which the expression and/or activity of Flightless I is altered.
- the present invention provides an animal with one or more skin cells including a nucleic acid that modulates Flightless I expression and/or activity in the cell.
- Examples of such animals are chimeric animals or transgenic animals. Methods for producing such animals are known in the art.
- the cells of the present invention may have use in, for example, wound repair, reducing scar formation and in preventing and/or treating a disease, condition or state in a subject associated with undesired or abnormal fibrosis.
- the cells may be delivered to a desired site of action to provide a therapeutic or beneficial effect.
- the cells may be delivered to the site in an appropriate composition to exert their therapeutic or beneficial effect.
- Compositions for maintaining and delivering cells are known in the art.
- the present invention also provides methods for screening new agents that may modulate one or more of wound repair, scar formation and migration and/or proliferation of cells involved in wound repair.
- the present invention provides a method of identifying an agent that modulates wound repair, the method including identifying an agent that modulates one or more of expression, secretion and activity of Flightless I.
- the identification of the agent includes identifying an agent that modulates one or more of expression, secretion and activity of Flightless I in cells involved in wound repair. Examples of cells involved in wound repair are as previously discussed herein.
- the screening method may be used to identify an agent that promotes wound repair.
- the agent may decrease Flightless I activity.
- the screening method may be use to identify an agent that reduces scar formation.
- the agent may increase Flightless I activity.
- the screening method may be used to identify an agent that modulates migration and/or proliferation of cells involved in wound repair.
- the present invention also provides an agent identified by the screening methods, and the use of the agents to modulate for example wound repair, scar formation and fibrosis.
- the flightless I protein colocalizes with actin- and microtubule-based structures in motile Swiss 3T3 fibroblasts: evidence for the involvement of PI 3-kinase and Ras-related small GTPases. J Cell Sci l 14:549-562).
- mice with the BALB/c background All studies were performed in mice with the BALB/c background.
- FIiI deficient heterozygous null mice (FHI +/-) and mice carrying the complete human FHI gene on a cosmid transgene were as described previously (Campbell, H.D. et al. 2002. Fliih, a gelsolin-related cytoskeletal regulator essential for early mammalian embryonic development. MoI Cell Biol 22:3518-3526).
- Heterozygous transgenic mice (TgI) were made by crossing FHI +/+ with cosmid transgene +/-. These transgenic mice were intercrossed to obtain animals homozygous for the transgene (Tg2; FHI+/+, cosmid transgene +/+).
- Tg3 An independent transgenic line (Tg3) was made by excising the FUI gene from cosmid cl 10H8 (Campbell, H.D. et al. 2002. Fliih, a gelsolin-related cytoskeletal regulator essential for early mammalian embryonic development. MoI Cell Biol 22:3518-3526) as a 17.8 kb BspRl fragment. This fragment contains ⁇ 4 kb of 5 '-flanking sequence extending into the 5' end of the next identified gene, and at the 3' end contains a small portion of the overlapping 3 ' end of the LLGL gene.
- the fragment was cloned into the Ncol site of LITMUS-29 (New England Biolabs, Ipswich, USA) and was resected as an Spel-Nsil fragment for preparation of transgenic mice, (FHI +/+, 17.8 kb transgene +/-) using BALB/c ES cells as described previously (Campbell, H.D. et al. 2002. Fliih, a gelsolin-related cytoskeletal regulator essential for early mammalian embryonic development. MoI Cell Biol 22:3518-3526). ES cells were cotransfected with pMClNeoPolyA to allow co- selection with G418.
- ES cells were screened, chimaera were prepared and bred and transgenic mice were genotyped as described previously (Campbell, H.D. et al. 2002. Fliih, a gelsolin-related cytoskeletal regulator essential for early mammalian embryonic development. MoI Cell Biol 22:3518-3526).
- FHI transgenic mice TgI -3; female 16-20 weeks old
- wild-type sex and age- matched litter mates were wounded using the same protocol previously described in (Cowin, AJ. et al. 2006. Wound Healing Is Defective in Mice Lacking Tetraspanin CDl 51. J Invest Dermatol). Briefly, two equidistant lcm full thickness incisions were made through the skin and left to heal by secondary intention. Digital photographs were taken of the wounds at 0, 3, 7, 14 and 21 days post- wounding. A ruler was aligned next to the wound to allow direct wound area and wound gape (mid-point of the lcm incision) measurements to be made. Wounds were harvested at 3, 7, 14 and 21 days and were bisected.
- FIiI deficient +/- mice female 16-20 weeks old
- sex and age-matched wild-type littermates were wounded similarly to reported studies (Cowin, AJ. et al. 2001. Hepatocyte growth factor and macrophage- stimulating protein are upregulated during excisional wound repair in rats. Cell Tissue Res 306:239-250).
- Histological sections (4 ⁇ m) were cut from paraffin-embedded fixed tissue. Sections were stained with hematoxylin and eosin or subjected to immunohistochemistry following antigen retrieval according to the manufacturer's protocols (DAKO Corporation, Botany, Australia). Following blocking in 3% normal horse serum, primary antibodies against FIiI (1:400), TGF- ⁇ l (1:200), collagen I (1 :200) or gelsolin (1 :100) were applied. Species-specific, biotinylated secondary antibodies (1:200) were used and detection was by CY3 -conjugated streptavidin (1 :200) (Sigma- Aldrich, Sydney, Australia).
- Integrated fluorescence intensity was determined using AnalySIS software package (Soft Imaging System GmbH, Munster Germany).
- Cell proliferation was determined using the proliferating cell nuclear antigen (PCNA) immunostaining technique (Geier, M.S. et al. 2005. Development and resolution of experimental colitis in mice with targeted deletion of dipeptidyl peptidase IV. J Cell Physiol 204:687-692).
- PCNA proliferating cell nuclear antigen
- the number of PCNA positive cells were counted and expressed as a percentage of total cells within the wound.
- Negative controls included replacing primary antibodies with normal rabbit IgG, normal mouse or normal goat IgG. Non-specific binding was determined by omitting primary or secondary antibodies. AU control sections had negligible immunofluorescence.
- Image analysis was performed using the ImageProPlus program (MediaCybernetics Inc., Maryland, USA). Wound size was determined by manually drawing below the epidermis or clot between the wound margins. The percentage of the wound that had reepithelialised was determined by measuring the portions of the wound that were covered with epidermis as a percentage of the entire wound. Dermal gape was determined by measuring between the dermal wound margins. Blinded measurements of histological slides by two independent assessors was performed.
- HFFs human foreskin fibroblasts
- HaCaTs keratinocytes
- Primary fibroblasts were also cultured from wild-type and FIiI overexpressing (TgI) mouse skins. Briefly, punch biopsies taken from wild-type and TgI mice skin were washed with PBS, allowed to adhere to the base of culture dishes and then cultured in DMEM supplemented with 10% FBS, penicillin and streptomycin (50 U ml "1 ) for several days. After removal of biopsy, adherent cells were trypsinized and maintained in culture.
- HFFs and HaCaTs were cultured until confluent in a 37°C, 5% CO 2 incubator before seeding into 96-well plates at a density of 4 x 10 4 cells/well. After 24 hours the media was replaced with serum-free DMEM and incubated for 4 hours to synchronise cell cycle.
- Cell proliferation assays were performed using the metabolic substrate WST-I according to manufacturer's protocols (Roche Applied Science, Kunststoff, Germany). Briefly, lO ⁇ l of WST-I reagent was added to the cells and left at 37°C for 30 minutes. The presence of the formazan product is quantified using a dual absorbance of 450nm and 600nm using a plate reader.
- HFFs and HaCaTs were grown to confluence in DMEM with 10% FBS in 6- well plates then scratched with a P200 pipette tip, producing a wound of approximately 2 mm x 1 cm. The cells were photographed at 0, 3, 6, 12, 24, 27, 30 and 48 hours. Wound margins were measured using the Image Pro-Plus program (MediaCybernetics Inc., Silver Springs, Maryland, USA) and rate of closure quantified as percent of initial wound area.
- HFFs and HaCaTs were grown to confluence on glass cover slips in DMEM containing 10% FBS.
- Cells were fixed with acetone for 5 minutes and blocked with 1% BSA/PBS for 15 minutes.
- FIiI (1 :400; 1 hour incubation at room temperature
- phalloidin-FITC (1 :250 of 100 ⁇ g/ml; 30 minutes incubation at room temperature) were added to the cells.
- Biotinylated anti-mouse (1:200, Sigma-Aldrich, Sydney, Australia) was added for 1 hour and detected by C Y3 -conjugated streptavidin (1 :200). Integrated fluorescence intensity was determined as before.
- HFFs and HaCaTs were seeded into 6-well plates and cultured until 30-50% confluent at time of transfection.
- FHI siRNA Lee, Y.H. et al. 2004. Developmentally essential protein flightless I is a nuclear receptor coactivator with actin binding activity. MoI Cell Biol 24:2103-2117) was transfected into the cells using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA).
- cDNA together with specific primers were set up to a final concentration of Ix SYBR Green, Ix Amplitaq PCR Buffer, 3 mM MgCl 2 , dNTPs (200 ⁇ M each), 0.9 ⁇ M of Primers (forward and reverse), 1.25 Units AmpliTaq Gold DNA polymerase in 25 ⁇ l H 2 O.
- the primer sequences were as follows: FIiI forward, 5 '-CCTCCTACAGCTAGCAGGTTATCAAC-S ' (SEQ ID NO: 1); reverse, 5 '-GCATGTGCTGGATATATACCTGGCAG-S ' (SEQ ID NO:2). Cyclophilin A forward, 5'-GGTTGGATGGCAAGCATGTG-S ' (SEQ ID NO.3); reverse, 5 '-TGCTGGTCTTGCCATTCCTG-S ' (SEQ ID NO:4).
- Skin fibroblasts were grown to confluence in DMEM (10%FBS), scratch wounded and incubated for a further 24 hours.
- the conditioned media was removed, concentrated using Centricon®Centrifugal Filter Columns (Millipore Australia Pty Ltd, North Ryde NSW) and protein concentration determined using Pierce BCA protein assay (Quantum Scientific, Paddington, QId).
- the cells were lysed in lysis buffer (5OmM Tris pH 7.5, ImM EDTA, 5OmM NaCl, 0.5% Triton-X-100), containing CompleteTM Mini protease inhibitor cocktail tablet (Roche Diagnostics Australia Pty. Ltd. Castle Hill NSW) /1 OmIs). and protein levels determined as before . Equal amounts of protein were loaded onto 12.5% SDS-PAGE gels and subjected to Western blotting for FIiI as described above.
- FIiI deficiency on wound healing was determined using mice heterozygous for the FHI gene knockout (FHI +/-) since homozygous deletion of FHI is embryonic lethal.
- the heterozygous null mice produce half the wild-type level of FIiI (Campbell, H.D. et al. 2002. Fliih, a gelsolin-related cytoskeletal regulator essential for early mammalian embryonic development. MoI Cell Biol 22:3518-3526).
- Age-matched wild- type littermates were used in all experiments as controls.
- To determine the effect of overexpression of FIiI on wound healing we made FIiI transgenic mice carrying extra copies of the human FHI gene.
- mice showed no visible differences compared with control littermates and reached the same average lifespan.
- To confirm the different levels of FIiI in these mouse lines we extracted mRNA from unwounded skins and performed real-time qRT-PCR (Figure 2a). Significantly reduced levels of FIiI were observed in the FIiI +/- skin whilst there was almost a 4 fold increase in FIiI expression in the FIiI TgI mouse skin ( Figure 2a).
- FHI is upregulated during wound healing in vivo
- FHI overexpression decreases cell proliferation, reduces migration and impairs wound healing
- FHI deficiency increases cell proliferation, enhances migration and improves wound healing
- This method also demonstrates the general concept of being able to screen for agents that modulate wound repair.
- FIiI antibodies could have a beneficial effect on wound healing in vivo
- FIiL antibody or dose-matched rabbit IgG were next injected intradermally around incisional wounds of wild-type mice and the effect on wound healing determined after 7 days (Figure Ha).
- Macroscopic analysis revealed that treatment with FIiL had decreased the wound area and wound gape by 33% and 31% respectively compared to IgG treated controls.
- TGF- ⁇ 1 TGF- ⁇ 3 and collagen I were affected by differential expression of FIiI in wound repair
- wounds created in wild-type, FIiI deficient +/- and FHI overexpressing mice were subjected to imrnunohistochemistry and the resulting staining patterns quantified ( Figure 12a).
- TGF- ⁇ 1 was significantly elevated in FIiI overexpressing wounds with little effect of FIiI overexpression being observed on TGF- ⁇ 3 ( Figure 12a).
- No effect of differential FIiI gene expression was observed on collagen III at any time-point post- wounding (data not shown).
- FIiI overexpressing mice Three independent lines of FIiI overexpressing mice were used in this study and all three showed significant impairments in wound repair confirming that FIiI overexpression and not just the position of the chromosomal insertion of the transgene was causing the observed differences.
- the improvement in wound healing in FIiI heterozygous null mice was modest, most likely reflecting the presence of one remaining functional FIiI allele. However, it was statistically significant and it indicated the potential benefits in being able to reduce FIiI expression.
- FIiI localises to ⁇ -tubulin-based structures known to be involved in cell division suggesting a role for FIiI in cell proliferation processes. Quantification of the number of proliferating dermal cells within the wounds revealed significantly fewer proliferating cells in the FIiI overexpressing mice wounds compared to wild type wounds. This deficiency in dermal cell proliferation within the wound bed is likely to contribute to the observed impaired healing. Interestingly, no significant difference was observed in the number of proliferating dermal cells in the FIiI +/- wounds, perhaps also reflective of the remaining functional FIiI allele within the cells. Our in vitro data support our in vivo studies, revealing that reduced FUI gene expression increases both fibroblast and keratinocyte motility and proliferation.
- fibroblasts overexpressing FIiI show reduced migration and proliferation which is likely to contribute to the observed impaired healing phenotype.
- Our studies confirm that FIiI is involved in both cell proliferation and migration and that it may negatively regulate these processes, perhaps via its actin severing abilities.
- FHL Intradermal application of FHL antibody to incisional wounds significantly reduces the size of these wounds at 7 days post-injury. Therefore, FHL represents a potential effective novel therapeutic factor to improve impaired wound healing.
- Our studies also show that wounds in FHI deficient mice have reduced TGF- ⁇ l yet increased expression of TGF- ⁇ 3 and that these wounds have improved healing.
- FIiI is an important regulator of cell proliferation, migration and wound repair.
- Topical application of FIiL antibodies to incisional wounds significantly enhances wounds repair. Therefore manipulation of FIiI levels may lead to potential new therapeutic interventions by which wound healing may be improved.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Organic Chemistry (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Biochemistry (AREA)
- Molecular Biology (AREA)
- Genetics & Genomics (AREA)
- Biophysics (AREA)
- Pharmacology & Pharmacy (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Animal Behavior & Ethology (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Epidemiology (AREA)
- Engineering & Computer Science (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Gastroenterology & Hepatology (AREA)
- Immunology (AREA)
- Toxicology (AREA)
- Zoology (AREA)
- Dermatology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
Abstract
Description
Claims
Priority Applications (5)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
AU2006308431A AU2006308431B2 (en) | 2005-10-25 | 2006-10-25 | Methods and compositions for modulating wound repair |
JP2008536881A JP5153639B2 (en) | 2005-10-25 | 2006-10-25 | Methods and compositions for modulating wound repair |
CA2665717A CA2665717C (en) | 2005-10-25 | 2006-10-25 | Methods and compositions comprising a flightless i antagonist for modulating wound repair |
US12/091,146 US8574571B2 (en) | 2005-10-25 | 2006-10-25 | Methods and compositions for modulating wound repair |
EP06790437A EP1945256B1 (en) | 2005-10-25 | 2006-10-25 | Methods and compositions for improving wound repair |
Applications Claiming Priority (4)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
AU2005905891A AU2005905891A0 (en) | 2005-10-25 | Methods and compositions for modulating TBF-Beta and collagen | |
AU2005905891 | 2005-10-25 | ||
AU2006905128A AU2006905128A0 (en) | 2006-09-18 | Methods and compositions for modulating wound repair | |
AU2006905128 | 2006-09-18 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2007048202A1 true WO2007048202A1 (en) | 2007-05-03 |
Family
ID=37967354
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/AU2006/001610 WO2007048202A1 (en) | 2005-10-25 | 2006-10-25 | Methods and compositions for modulating wound repair |
Country Status (5)
Country | Link |
---|---|
US (1) | US8574571B2 (en) |
EP (1) | EP1945256B1 (en) |
JP (1) | JP5153639B2 (en) |
CA (1) | CA2665717C (en) |
WO (1) | WO2007048202A1 (en) |
Cited By (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2014100852A1 (en) * | 2012-12-24 | 2014-07-03 | Royal Melbourne Institute Of Technology | Inhibition of cancer growth and metastasis |
WO2016040987A1 (en) * | 2014-09-15 | 2016-03-24 | Abregen Pty Ltd | Treating and/or preventing psoriasis |
WO2020077397A1 (en) * | 2018-10-15 | 2020-04-23 | Abregen Pty Ltd | Methods and compositions for the treatment of mucosal lesions |
Families Citing this family (12)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
ITRM20030376A1 (en) | 2003-07-31 | 2005-02-01 | Univ Roma | PROCEDURE FOR THE ISOLATION AND EXPANSION OF CARDIOC STAMIN CELLS FROM BIOPSIA. |
US11660317B2 (en) | 2004-11-08 | 2023-05-30 | The Johns Hopkins University | Compositions comprising cardiosphere-derived cells for use in cell therapy |
US9249392B2 (en) | 2010-04-30 | 2016-02-02 | Cedars-Sinai Medical Center | Methods and compositions for maintaining genomic stability in cultured stem cells |
US9845457B2 (en) | 2010-04-30 | 2017-12-19 | Cedars-Sinai Medical Center | Maintenance of genomic stability in cultured stem cells |
WO2013184527A1 (en) | 2012-06-05 | 2013-12-12 | Capricor, Inc. | Optimized methods for generation of cardiac stem cells from cardiac tissue and their use in cardiac therapy |
EP3563859B1 (en) | 2012-08-13 | 2021-10-13 | Cedars-Sinai Medical Center | Cardiosphere-derived exosomes for tissue regeneration |
JP6878274B2 (en) | 2014-10-03 | 2021-05-26 | シーダーズ−サイナイ・メディカル・センターCedars−Sinai Medical Center | Myocardial cell-derived cells and exosomes secreted by such cells in the treatment of muscular dystrophy |
WO2017123662A1 (en) | 2016-01-11 | 2017-07-20 | Cedars-Sinai Medical Center | Cardiosphere-derived cells and exosomes secreted by such cells in the treatment of heart failure with preserved ejection fraction |
WO2017210652A1 (en) | 2016-06-03 | 2017-12-07 | Cedars-Sinai Medical Center | Cdc-derived exosomes for treatment of ventricular tachyarrythmias |
US11541078B2 (en) | 2016-09-20 | 2023-01-03 | Cedars-Sinai Medical Center | Cardiosphere-derived cells and their extracellular vesicles to retard or reverse aging and age-related disorders |
US11759482B2 (en) | 2017-04-19 | 2023-09-19 | Cedars-Sinai Medical Center | Methods and compositions for treating skeletal muscular dystrophy |
US11660355B2 (en) | 2017-12-20 | 2023-05-30 | Cedars-Sinai Medical Center | Engineered extracellular vesicles for enhanced tissue delivery |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1998024465A1 (en) * | 1996-12-04 | 1998-06-11 | The Victoria University Of Manchester | Wound healing and treatment of fibrosis |
WO1999041364A2 (en) * | 1998-02-13 | 1999-08-19 | The Wistar Institute | Compositions and methods for wound healing |
Family Cites Families (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US4828991A (en) * | 1984-01-31 | 1989-05-09 | Akzo N.V. | Tumor specific monoclonal antibodies |
GB9106678D0 (en) * | 1991-03-28 | 1991-05-15 | Ferguson Mark W J | Wound healing |
US6395282B1 (en) * | 1998-04-16 | 2002-05-28 | University Of Rochester | Immunogenic conjugates of Gram-negative bacterial autoinducer molecules |
-
2006
- 2006-10-25 US US12/091,146 patent/US8574571B2/en active Active
- 2006-10-25 WO PCT/AU2006/001610 patent/WO2007048202A1/en active Application Filing
- 2006-10-25 JP JP2008536881A patent/JP5153639B2/en not_active Expired - Fee Related
- 2006-10-25 CA CA2665717A patent/CA2665717C/en active Active
- 2006-10-25 EP EP06790437A patent/EP1945256B1/en active Active
Patent Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO1998024465A1 (en) * | 1996-12-04 | 1998-06-11 | The Victoria University Of Manchester | Wound healing and treatment of fibrosis |
WO1999041364A2 (en) * | 1998-02-13 | 1999-08-19 | The Wistar Institute | Compositions and methods for wound healing |
Non-Patent Citations (3)
Title |
---|
CZUWARA-LADYKOWSKA J. ET AL.: "Fli-1 inhibits collagen type I production in dermal fibroblasts via an Sp1-dependent pathway", J BIOL CHEM., vol. 276, no. 24, 15 June 2001 (2001-06-15), pages 20839 - 20848, XP003012485 * |
DAVY D.A. ET AL.: "The flightless I protein colocalizes with actin- and microtubule-based structures in motile Swiss 3T3 fibroblasts: evidence for the involvement of PI 3-kinase and Ras-related small GTPases", J CELL SCI., vol. 114, no. PT 3, February 2001 (2001-02-01), pages 549 - 562, XP003012486 * |
See also references of EP1945256A4 * |
Cited By (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2014100852A1 (en) * | 2012-12-24 | 2014-07-03 | Royal Melbourne Institute Of Technology | Inhibition of cancer growth and metastasis |
AU2013202668B2 (en) * | 2012-12-24 | 2014-12-18 | Adelaide Research & Innovation Pty Ltd | Inhibition of cancer growth and metastasis |
US9895414B2 (en) | 2012-12-24 | 2018-02-20 | Royal Melbourne Institute Of Technology | Inhibition of cancer growth and metastasis |
WO2016040987A1 (en) * | 2014-09-15 | 2016-03-24 | Abregen Pty Ltd | Treating and/or preventing psoriasis |
WO2020077397A1 (en) * | 2018-10-15 | 2020-04-23 | Abregen Pty Ltd | Methods and compositions for the treatment of mucosal lesions |
Also Published As
Publication number | Publication date |
---|---|
EP1945256A4 (en) | 2010-01-20 |
EP1945256B1 (en) | 2012-07-04 |
CA2665717A1 (en) | 2007-05-03 |
EP1945256A1 (en) | 2008-07-23 |
US20100203064A1 (en) | 2010-08-12 |
US8574571B2 (en) | 2013-11-05 |
JP2009512718A (en) | 2009-03-26 |
CA2665717C (en) | 2016-01-19 |
JP5153639B2 (en) | 2013-02-27 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CA2665717C (en) | Methods and compositions comprising a flightless i antagonist for modulating wound repair | |
Guo et al. | Exosomes derived from platelet-rich plasma promote the re-epithelization of chronic cutaneous wounds via activation of YAP in a diabetic rat model | |
US5824655A (en) | Anti-transforming growth factor-β gene therapy | |
Osz et al. | The thrombospondin-1 receptor CD36 is an important mediator of ovarian angiogenesis and folliculogenesis | |
US11446263B2 (en) | Caspase inhibitor-containing drug for treating or preventing disorders caused by TGF-β, and applications thereof | |
EP2473526A1 (en) | Treatment of vasculoproliferative conditions | |
AU2007353332B2 (en) | Modulation of Rhamm (CD168) for selective adipose tissue development | |
Barayan et al. | Interleukin-6 blockade, a potential adjunct therapy for post-burn hypermetabolism | |
Izumi et al. | Involvement of insulin-like growth factor-I and insulin-like growth factor binding protein-3 in corneal fibroblasts during corneal wound healing | |
EP4196491A1 (en) | Agrin polypeptide and uses thereof | |
Conradt et al. | Epidermal or Dermal Collagen VII Is Sufficient for Skin Integrity: Insights to Anchoring Fibril Homeostasis | |
JP6440107B2 (en) | Cell sheet production method, composition, cell culture aid, and cell culture method | |
AU2006308431B2 (en) | Methods and compositions for modulating wound repair | |
US20230053881A1 (en) | Neuregulin for the treatment and/or prevention of tumors of the nervous system | |
RU2672377C1 (en) | Method of treating wounds | |
KR102026138B1 (en) | Composition for Inhibition or Treatment of Keloids and Hypertrophic Scar Comprising CRIF1 Antagonist | |
US20240050529A1 (en) | Modulating lymphatic vessels in neurological disease | |
CN100551437C (en) | The purposes of antibacterial peptide Bin1b aspect spermioteleosis that epididymis is special | |
US20160304881A1 (en) | Ddr1 antagonist or an inhibitor of ddr1 gene expression for use in the prevention or treatment of crescentic glomerulonephritis | |
WO2022268644A1 (en) | Inhibitors for use in prevention- and/or alleviation of heart fibrosis | |
Westerhuis | Pathogenetic Aspects of IgA-nephropathy |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application | ||
WWE | Wipo information: entry into national phase |
Ref document number: 2006308431 Country of ref document: AU |
|
ENP | Entry into the national phase |
Ref document number: 2006308431 Country of ref document: AU Date of ref document: 20061025 Kind code of ref document: A |
|
WWP | Wipo information: published in national office |
Ref document number: 2006308431 Country of ref document: AU |
|
ENP | Entry into the national phase |
Ref document number: 2008536881 Country of ref document: JP Kind code of ref document: A |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2006790437 Country of ref document: EP |
|
WWP | Wipo information: published in national office |
Ref document number: 2006790437 Country of ref document: EP |
|
WWE | Wipo information: entry into national phase |
Ref document number: 12091146 Country of ref document: US |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2665717 Country of ref document: CA |