EP1009424A2 - Regulierung von muskelgewebe mittels hedgehog-ähnlichen polypeptiden und dazu geeignete formulierungen und anwendungen - Google Patents

Regulierung von muskelgewebe mittels hedgehog-ähnlichen polypeptiden und dazu geeignete formulierungen und anwendungen

Info

Publication number
EP1009424A2
EP1009424A2 EP98943462A EP98943462A EP1009424A2 EP 1009424 A2 EP1009424 A2 EP 1009424A2 EP 98943462 A EP98943462 A EP 98943462A EP 98943462 A EP98943462 A EP 98943462A EP 1009424 A2 EP1009424 A2 EP 1009424A2
Authority
EP
European Patent Office
Prior art keywords
hedgehog
muscle
protein
cells
cell
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.)
Withdrawn
Application number
EP98943462A
Other languages
English (en)
French (fr)
Inventor
Chris S. Bladgen
Peter D. Currie
Philip W. Ingham
Simon M. Hughes
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.)
Curis Inc
Original Assignee
Ontogeny Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ontogeny Inc filed Critical Ontogeny Inc
Publication of EP1009424A2 publication Critical patent/EP1009424A2/de
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/553Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having at least one nitrogen and one oxygen as ring hetero atoms, e.g. loxapine, staurosporine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals

Definitions

  • myoblasts skeletal muscle precursor cells
  • myoblasts In adults, myoblasts remain as a mitotically quiescent reserve precursor population which can, upon muscle injury, re-enter the cell cycle, undergo several rounds of proliferation, and subsequently differentiate and permanently exit the from the cell cycle.
  • muscle cells Upon differentiation, differentiated myoblasts (“myocytes") acquire the ability to fuse with one another or with muscle fibers, and also commence coordinate expression of a large set of muscle-specific myofibrillar and contractile proteins (e.g., muscle myosins and actin, troponin, tropomyosin, etc.).
  • Muscle tissue can grow by several different mechanisms which are controlled by different trophic factors. Muscle tissue can grow by hypertrophy, an enlargement of or increase in mass or size of muscle fibers, or by hyperplasia, an increase in the numbers of fibers or in the numbers of muscle nuclei, or by a combination of these two processes. Growth factors that act on skeletal muscle tissue can be divided into two broad groups.
  • the factors that stimulate proliferation of myoblasts usually inhibit differentiation of myoblasts and inhibit the expression and action of the muscle regulatory transcription factors (MRFs). Conversely, the factors that stimulate differentiation of myoblasts usually stimulate expression of the MRFs and can contribute to muscle hypertrophy.
  • MRFs muscle regulatory transcription factors
  • hypertrophic factors include, for example, growth hormone (GH) or insulin-like growth factor-I (IGF-I).
  • GH growth hormone
  • IGF-I insulin-like growth factor-I
  • Muscle hypertrophy can be assessed by the measurement of muscle fiber diameter in vivo or in vitro, or by the measurement of the accretion of the muscle-specific myofibrillar and contractile proteins.
  • a decline in such skeletal muscle tissue mass, or muscle atrophy is an important contributor to frailty in older individuals.
  • muscle mass declines by one-third between the ages of 50 and 80.
  • extended hospitalization can result in further disuse atrophy leading to a potential loss of the ability for independent living and to a cascade of physical decline.
  • the physical aging process profoundly affects body composition, including significant reductions in lean body mass and increases in central adiposity.
  • the changes in overall adiposity and fat distribution appear to be important factors in many common "age-related" disorders such as hypertension, glucose intolerance and diabetes, dyslipidemia, and atherosclerotic cardiovascular disease.
  • One aspect of the present application relates to a method for regulating the formation and/or maintenance of muscle tissue by ectopically contacting muscle cells, especially muscle stem/progenitor cells, in vitro or in vivo, with a hedgehog therapeutic or ptc therapeutic in an amount effective to alter the growth state the treated cells, e.g., relative to the absence of administeration of the hedgehog therapeutic or ptc therapeutic.
  • the hedgehog therapeutic preferably a polypeptide including a hedgehog portion comprising at least a bioactive extracellular portion of a hedgehog protein, e.g., the hedgehog portion includes at least 50, 100 or 150 (contiguous) amino acid residues of an N-terminal half of a hedgehog protein.
  • the hedgehog portion includes at least a portion of the hedgehog protein corresponding to a 19kd fragment of the extracellular domain of a hedgehog protein.
  • the hedgehog portion has an amino acid sequence at least 60, 75, 85, or 95 percent identical with a hedgehog protein of any of SEQ ID Nos.
  • the hedgehog portion can be encoded by a nucleic acid which hybridizes under stringent conditions to a nucleic acid sequence of any of SEQ ID Nos. 1-9 or 19, e.g., the hedgehog portion can be encoded by a vertebrate hedgehog gene, especially a human hedgehog gene.
  • the subject method can be carried out by administering a gene activation construct, wherein the gene activation construct is deigned to recombine with a genomic hedgehog gene of the patient to provide a heterologous transcriptional regulatory sequence operatively linked to a coding sequence of ' the hedgehog gene.
  • the subject method can be practiced with the administration of a gene therapy construct encoding a hedgehog polypeptide.
  • the gene therapy construct can be provided in a composition selected from a group consisting of a recombinant viral particle, a liposome, and a poly-cationic nucleic acid binding agent,
  • the subject method can be carried out using a ptc therapeutic.
  • An exemplary ptc therapeutic is a small organic molecule which binds to a patched protein and derepresses /? ⁇ tc/ze ⁇ i-mediated inhibition of mitosis, e.g., a molecule which binds to patched and mimics hedgehog-mediated patched signal transduction, which binds to patched and regulates pate he d-dependent gene expression.
  • the binding of the ptc therapeutic to patched may result in upregulation of patched and/or gli expression.
  • the ptc therapeutic can be a small organic molecule which interacts with muscle cells to induce hedgehog-mediated patched signal transduction, such as by altering the localization, protein-protein binding and/or enzymatic activity of an intracellular protein involved in a patched signal pathway.
  • the ptc therapeutic may alter the level of expression of a hedgehog protein, a patched protein or a protein involved in the intracellular signal transduction pathway of patched.
  • the ptc therapeutic is an antisense construct which inhibits the expression of a protein which is involved in the signal transduction pathway of patched and the expression of which antagonizes hedgehog-mediated signals.
  • the antisense construct is perferably an oligonucleotide of about 20-30 nucleotides in length and having a GC content of at least 50 percent.
  • the ptc therapeutic is an inhibitor of protein kinase A (PKA), such as a 5-isoquinolinesulfonamide.
  • PKA protein kinase A
  • the PKA inhibitor can be a cyclic AMP analog.
  • Exemplary PKA inhibitors include N-[2-((p-bromocinnamyl)amino)ethyl]-5- isoquinolinesulfonamide, l-(5-isoquinoline-sulfonyl)-2-methylpiperazine, KT5720, 8- bromo-cAMP, dibutyryl-cAMP and PKA Heat Stable Inhibitor isoform ⁇ .
  • Another exemplary PKA inhibitor is represented in the general formula: wherein,
  • R j and R 2 each can independently represent hydrogen, and as valence and stability permit a lower alkyl, a lower alkenyl, a lower alkynyl, a carbonyl (such as a carboxyl, an ester, a formate, or a ketone), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an amino, an acylamino, an amido, a cyano, a nitro, an azido, a sulfate, a sulfonate.
  • a lower alkyl such as a lower alkenyl, a lower alkynyl, a carbonyl (such as a carboxyl, an ester, a formate, or a ketone), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an amino, an
  • a sulfonamido -(CH 2 ) m -Rg, -(CH 2 ) m -OH, -(CH 2 ) m -O-lower alkyl, -(CH 2 ) m -O-lower alkenyl, -(CH 2 ) n -O-(CH 2 ) m -R 8 , -(CH 2 ) m -SH, -(CH 2 ) m -S-lower alkyl, -(CH 2 ) m -S-lower alkenyl, -(CH 2 ) n -S-(CH 2 ) m -R 8 , or R j and R 2 taken together with N form a heterocycle (substituted or unsubstituted);
  • R 3 is absent or represents one or more substitutions to the isoquinoline ring such as a lower alkyl, a lower alkenyl, a lower alkynyl, a carbonyl (such as a carboxyl, an ester, a formate, or a ketone), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate).
  • a lower alkyl such as a lower alkenyl, a lower alkynyl, a carbonyl (such as a carboxyl, an ester, a formate, or a ketone), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate).
  • R 8 represents a substituted or unsubstituted aryl, aralkyl, cycloalkyl, cycloalkenyl, or heterocycle; and n and m are independently for each occurrence zero or an integer in the range of 1 to 6.
  • the present application is directed to the discovery that hedgehog gene products are involved in controlling the formation and/or maintenance of muscle tissue, especially slow (red) muscle.
  • Certain aspects of the invention are directed to a preparations of hedgehog polypeptides, or other molecules which regulate patched or smoothened signalling, and their uses in stimulating muscle growth or differentiation in mammals.
  • the invention is directed to the use of hedgehog polypeptides to stimulate muscle growth, differentiation or hypertrophy.
  • hedgehog proteins are implicated in the proliferation and/or differentiation of myoblastic/myocytic cells and may provide early signals that regulate the differentiation of these or other precursor (stem) cells into muscle tissues.
  • the method of the present invention comprises contacting a muscle cells (collectively, muscle stem cells (myoblasts), and myocytic or other differentiated muscle cells), with an amount of a hedgehog therapeutic (defined infra) which produces a non-toxic response by the cell of (i) induction of of muscle tissue formation or maintenance of existing muscle tissue, or (ii) inhibition of muscle tissue formation, depending on the whether the hedgehog therapeutic is a sufficient hedgehog agonist or hedgehog antagonist.
  • the subject method can be carried out on muscle cells which may be either dispersed in culture or a part of an intact tissue or organ. Moreover, the method can be performed on cells which are provided in culture (in vitro), or on cells in a whole animal (in vivo).
  • the present invention provides pharmaceutical preparations and methods for controlling the formation of myoblastic-derived tissue utilizing, as an active ingredient, a hedgehog polypeptide or a mimetic thereof.
  • the invention also relates to methods of controlling the functional performance of an muscle-derived tissue by use of the pharmaceutical preparations of the invention.
  • the hedgehog formulations of the present invention may be used as part of regimens in the treatment or prevention of disorders of, or surgical or cosmetic repair of, such muscle tissues.
  • the subject method can be used for treating atrophy, or wasting, in particular, skeletal muscle atrophy and cardiac muscle atrophy.
  • certain diseases wherein the muscle tissue is damaged, is abnormal or has atrophied are treatable using the invention, such as, for example, normal aging, disuse atrophy, wasting or cachexia, and various secondary disorders associated with age and the loss of muscle mass, such as hypertension, glucose intolerance and diabetes, dyslipidemia and atherosclerotic cardiovascular disease.
  • the therapeutic preparatios of the present invention may be used to treat rhabdomyosarcomas by regulating myoblast differentiation.
  • the invention also is directed to the treatment of certain cardiac insufficiencies, such as congestive heart failure.
  • the treatment of muscular myopathies such as muscular dystrophies is also embodied in the invention.
  • the subject compositions can be used to inhibit, rather than promote, growth of myoblastic-derived tissue.
  • certain of the compositions disclosed herein may be applied to the treatment or prevention of a variety hyperplastic or neoplastic conditions affecting muscle tissue.
  • the method can find application for the treatment or prophylaxis of, e.g., invasive muscle tumors and myoblastic sarcomas.
  • the subject hedgehog treatments are effective on both human and animal subjects afflicted with these conditions.
  • Animal subjects to which the invention is applicable extend to both domestic animals and livestock, raised either as pets or for commercial purposes. Examples are dogs, cats, cattle, horses, sheep, hogs and goats.
  • Still another aspect of the present invention provides a method of stimulating the growth and regulating the differentiation of muscle cells and tissues in culture.
  • the induction of muscle formation by hedgehog proteins may be due at least in part to the ability of these proteins to antagonize (directly or indirectly) /? ⁇ tc/zed-mediated regulation of gene expression and other physiological effects mediated by that protein.
  • the patched gene product a cell surface protein, is understood to signal through a pathway which causes transcriptional repression of members of the Wnt and Dpp/BMP families of morphogens, proteins which impart positional information.
  • hedgehog relieves (derepresses) this inhibition conferred by patched, allowing expression of particular gene programs.
  • the present invention contemplates the use of other agents which are capable of mimicking the effect of the hedgehog protein on patched signalling, e.g., as may be identified from the drug screening assays described below.
  • hedgehog therapeutic refers to various forms of hedgehog polypeptides, as well as peptidomimetics, which can modulate the proliferation/differentiation state of muscle cells by, as will be clear from the context of individual examples, mimicing or potentiating (agonizing) or inhibiting (antagonizing) the effects of a naturally-occurring hedgehog protein.
  • a hedgehog therapeutic which mimics or potentiates the activity of a wild-type hedgehog protein is a "hedgehog agonist”.
  • a hedgehog therapeutic which inhibits the activity of a wild-type hedgehog protein is a "hedgehog antagonist".
  • hedgehog polypeptide encompasses preparations of hedgehog proteins and peptidyl fragments thereof, both agonist and antagonist forms as the specific context will make clear.
  • bioactive fragment of a hedgehog protein refers to a fragment of a full-length hedgehog polypeptide, wherein the fragment specifically agonizes or antagonizes inductive events mediated by wild-type hedgehog proteins.
  • the hedgehog biactive fragment preferably is a soluble extracellular portion of a hedgehog protein, where solubility is with reference to physiologically compatible solutions. Exemplary bioactive fragments are described in PCT publications WO 95/18856 and WO 96/17924.
  • ptc therapeutic refers to agents which either (i) mimic the effect of hedgehog proteins on patched signalling, e.g., which antagonize the cell-cycle inhibitory activity of patched, or (ii) activate or potentiate patched signalling.
  • the ptc therapeutic can be a hedgehog antagonist.
  • the ptc therapeutic can be, e.g., a peptide, a nucleic acid, a carbohydrate, a small organic molecule, or natural product extract (or fraction thereof).
  • a "proliferative" form of a hedgehog or ptc therapeutic is one which induces proliferation of muscle cells, particularly muscle stem cells.
  • antiproliferative form of a hedgehog or ptc therapeutic is one which inhibits proliferation of an muscle cells, preferably in a non-toxic manner, e.g., by promoting or maintaining a differentiated phenotype or otherwise promoting quiescence.
  • myoblast cultures refers to cultures that contain cycling skeletal muscle precursors, and are considered distinct from “muscle fiber cultures” which are derived from myoblast cultures that are allowed to undergo differentiation and fusion to form multinucleated muscle fibers.
  • myogenic culture is a generic term that refers to both kinds of cultures.
  • myocyte refers to a differentiated, post- mitotic, muscle cell that has not yet undergone fusion, and thus represents, in general, a transient cell type under most conditions.
  • proliferating and “proliferation” refer to cells undergoing mitosis.
  • Transformed cells refers to cells which have spontaneously converted to a state of unrestrained growth, i.e., they have acquired the ability to grow through an indefinite number of divisions in culture. Transformed cells may be characterized by such terms as neoplastic, anaplastic and/or hyperplastic, with respect to their loss of growth control.
  • immortalized cells refers to cells which have been altered via chemical and/or recombinant means such that the cells have the ability to grow through an indefinite number of divisions in culture.
  • a "patient” or “subject” to be treated by the subject method can mean either a human or non-human animal.
  • an "effective amount" of, e.g., a hedgehog therapeutic refers to an amount of, e.g., a hedgehog polypeptide in a preparation which, when applied as part of a desired dosage regimen brings about a change in the rate of cell proliferation and/or the state of differentiation of a cell so as to produce an amount of muscle cell proliferation or differentiation according to clinically acceptable standards for the disorder to be treated or the cosmetic purpose.
  • the "growth state" of a cell refers to the rate of proliferation of the cell and the state of differentiation of the cell.
  • Homology and identity each refer to sequence similarity between two polypeptide sequences, with identity being a more strict comparison. Homology and identity can each be determined by comparing a position in each sequence which may be aligned for purposes of comparison. When a position in the compared sequence is occupied by the same amino acid residue, then the polypeptides can be referred to as identical at that position; when the equivalent site is occupied by the same amino acid (e.g., identical) or a similar amino acid (e.g., similar in steric and/or electronic nature), then the molecules can be refered to as homologous at that position. A percentage of homology or identity between sequences is a function of the number of matching or homologous positions shared by the sequences. An "unrelated" or “non-homologous" sequence shares less than 40 percent identity, though preferably less than 25 percent identity, with an hedgeog sequence of the present invention.
  • recombinant protein refers to a polypeptide which is produced by recombinant DNA techniques, wherein generally, DNA encoding the polypeptide is inserted into a suitable expression construct which is in turn used to transform a host cell to produce the heterologous protein. That is, the polypeptide is expressed from a heterologous nucleic acid.
  • a “chimeric protein” or “fusion protein” is a fusion of a first amino acid sequence encoding a hedgehog polypeptide with a second amino acid sequence defining a domain foreign to and not substantially homologous with any domain of hh protein.
  • a chimeric protein may present a foreign domain which is found (albeit in a different protein) in an organism which also expresses the first protein, or it may be an "interspecies", “intergenic”, etc. fusion of protein structures expressed by different kinds of organisms.
  • a fusion protein can be represented by the general formula (X) n -(/z/z) m -(Y) n , wherein hh represents all or a portion of the hedgehog protein, X and Y each independently represent an amino acid sequences which are not naturally found as a polypeptide chain contiguous with the hedgehog sequence, m is an integer greater than or equal to 1 , and each occurrence of n is, independently, 0 or an integer greater than or equal to 1 (n and m are preferably no greater than 5 or 10).
  • the subject method has wide applicability to the treatment or prophylaxis of disorders afflicting muscle tissue.
  • the method can be characterized as including a step of administering to an animal an amount of a ptc or hedgehog therapeutic effective to alter the proliferative state of a treated muscle tissue.
  • the mode of administration and dosage regimens will vary depending on the muscle tissue(s) which is to be treated.
  • the use of a particular ptc or hedgehog therapeutic e.g., an agonist or antagonist, will depend on whether proliferation of cells of the treated tissue is desired or intended to be prevented.
  • the invention is directed to a muscle-trophic factor, and its use in stimulating muscle growth or differentiation in mammals.
  • Such stimulation of muscle growth is useful for treating atrophy, or wasting, in particular, skeletal muscle atrophy and cardiac muscle atrophy.
  • certain diseases wherein the muscle tissue is damaged, is abnormal or has atrophied are treatable using the invention, such as, for example, normal aging, disuse atrophy, wasting or cachexia, and various secondary disorders associated with age and the loss of muscle mass, such as hypertension, glucose intolerance and diabetes, dyslipidemia and atherosclerotic cardiovascular disease.
  • the treatment of muscular myopathies such as muscular dystrophies is also embodied in the invention.
  • compositions comprising the subject ptc and hedgehog therapeutics can be used for inhibiting muscle degeration, e.g., for decreasing the loss of muscle mass, such as part of a treatment for such muscle wasting disorders.
  • pharmaceutical compositions according to the invention are administered to patients suffering from a disorder, i.e., an abnormal physical condition, a disease or pathophysiological condition associated with abnormal and/or aberrant regulation of muscle tissue.
  • the disorders for which the compositions of the invention are administered are preferably those which directly or indirectly produce a wasting (i.e., loss) of muscle mass, that is, a muscle wasting disorder.
  • a wasting disorder i.e., loss
  • These include muscular dystrophies, cardiac cachexia, emphysema, leprosy, malnutrition, osteomalacia, child acute leukemia, AIDS cachexia and cancer cachexia.
  • the muscular dystrophies are genetic diseases which are characterized by progressive weakness and degeneration of muscle fibers without evidence of neural degeneration.
  • DMD Duchenne muscular dystrophy
  • myotonic dystrophy fractional muscle protein synthesis has been shown to be decreased by an average of 28%, without any corresponding decrease in non-muscle protein synthesis (possibly due to impaired end- organ response to anabolic hormones or substrates). Accelerated protein degradation has been demonstrated in the muscles of DMD patients.
  • the subject method can be used as part of a therapeutic strategy for preventing, and in some instance reversing, the muscle wasting conditions associated with such dystrophies.
  • Severe congestive heart failure is characterized by a "cardiac cachexia,” i.e., a muscle protein wasting of both the cardiac and skeletal muscles, with an average
  • the cardiac cachexia is caused by an increased rate of myofibrillar protein breakdown.
  • the subject method can be used as part of a treatment for cardiac cachexia.
  • Emphysema is a chronic obstructive pulmonary disease, defined by an enlargement of the air spaces distal to the terminal non-respiratory bronchioles, accompanied by destructive changes of the alveolar walls.
  • Clinical manifestations of reduced pulmonary functioning include coughing, wheezing, recurrent respiratory infections, edema, and functional impairment and shortened life-span.
  • the efflux of tyrosine is increased by 47% in emphysematous patients.
  • whole body leucine flux remains normal, whole-body leucine oxidation is increased, and whole-body protein synthesis is decreased. The result is a decrease in muscle protein synthesis, accompanied by a decrease in whole body protein turnover and skeletal muscle mass.
  • the subject ptc and hedgehog therapeutics may be used to prevent and/or reverse, the muscle wasting conditions associated with such diseases.
  • diabetes mellitus there is a generalized wasting of small muscle of the hands, which is due to chronic partial denervation (neuropathy). This is most evident and worsens with long term disease progression and severity.
  • the subject method can be used as part of a therapeutic strategy for treatement of diabetes mellitus.
  • Leprosy is associated with a muscular wasting which occurs between the metacarpals of the thumb and index finger. Severe malnutrition is characterized by, inter alia, severe muscle wasting.
  • the subject method can be used to treat muscle wasting effects of leprosy.
  • Osteomalacia is a nutritional disorder caused by a deficiency of vitamin D and calcium. It is referred to as “rickets” in children, and “osteomalacia” in adults. It is marked by a softening of the bones (due to impaired mineralization, with excess accumulation of osteoid), pain, tenderness, muscle wasting and weakness, anorexia, and overall weight loss. It can result from malnutrition, repeated pregnancies and lactation (exhausting or depleting vitamin D and calcium stores), and vitamin D resistance. The subject method can be used as part of a therapeutic strategy for treatment of osteomalacia.
  • Cancer cachexia is a complex syndrome which occurs with variable incidence in patients with solid tumors and hematological malignancies. Clinically, cancer cachexia is manifested as weight loss with massive depletion of both adipose tissue and lean muscle mass, and is one cause of death which results from cancer. Cancer cachexia patients have shorter survival times, and decreased response to chemotherapy. In addition to disorders which produce muscle wasting, other circumstances and conditions appear to be linked in some fashion with a decrease in muscle mass. Such afflictions include muscle wasting due to chronic back pain, advanced age, long term hospitalization due to illness or injury, alcoholism and corticosteroid therapy. The subject method can be used as part of a therapeutic strategy for preventing, and in some instance reversing, the muscle wasting conditions associated with such cancers.
  • a course of treatment for disorder can include administration of a therapeutic amount of ptc or hedgehog therapeutics. It is also believed that general weakness in old age is due to muscle wasting. As the body ages, an increasing proportion of skeletal muscle is replaced by fibrous tissue. The result is a significant reduction in muscle power, but only a marginal reduction in fat-free mass.
  • the subject method can be used as part of a treatment and preventive strategies for preventing/reversing muscle wasting in elderly patients.
  • the compounds of the invention can be used to alleviate the muscle mass loss resulting from the foregoing conditions, as well as others.
  • the ptc and hedgehog therapeutics of the present invention are useful in veterinary and animal husbandry applications to counter weight loss in animals, or to promote growth.
  • the invention may also find use for increasing the efficiency of animal meat production.
  • animals may be fed or injected with a ptc or hedgehog therapeutic in order to increase overall skeletal muscle mass, e.g., to increase the weight of such farm animals as cows, pigs, sheep, chickens and salmon.
  • Tissue replacement therapy is well established in the treatment of human disease.
  • muscle cells especially muscle stem cells
  • transplantation of normal myoblasts may be useful to treat Duchenne muscular dystrophy and other muscle degeneration and wasting diseases. See, for example, Partridge (1991) Muscle & Nerve 14:197-212.
  • myoblasts they may be injected at various sites to treat muscle wasting diseases.
  • the subject method can be used to regulate the growth of muscle cells and tissue in vitro, as well as to accelerate the grafting of impanted muscle tissue to an animal host
  • the present invention also concerns myoblast cultures which have been expanded by treatment with a hedgehog or other ptc therapeutic.
  • a method comprises obtaining a muscle sample, preferably one including myoblasts; optionally treating the cell sample enzymically to separate the cells; culturing, in the presence of a hedgehog or ptc therapeutic.
  • the hedgehog therapeutic compositions of the subject method can be generated by any of a variety of techniques, including purification of naturally occurring proteins, recombinantly produced proteins and synthetic chemistry.
  • Polypeptide forms of the hedgehog therapeutics are preferably derived from vertebrate hedgehog proteins, e.g., have sequences corresponding to naturally occurring hedgehog proteins, or fragments thereof, from vertebrate organisms.
  • the hedgehog polypeptide can correspond to a hedgehog protein (or fragment thereof) which occurs in any metazoan organism.
  • the various naturally-occurring hedgehog proteins from which the subject therapeutics can be derived are characterized by a signal peptide, a highly conserved N- terminal region, and a more divergent C-terminal domain.
  • hedgehog precursor proteins naturally undergo an internal autoproteolytic cleavage which depends on conserved sequences in the C-terminal portion (Lee et al. (1994) Science 266:1528-1537; Porter et al. (1995) Nature 374:363-366).
  • N-terminal peptide stays tightly associated with the surface of cells in which it was synthesized, while the C-terminal peptide is freely diffusible both in vitro and in vivo (Lee et al. (1994) supra; Bumcrot et al. (1995) supra; Mart', E. et al. (1995) Development 121 :2537-2547; Roelink, H. et al. (1995) Cell 81 :445-455).
  • the vertebrate family of hedgehog genes includes at least four members, e.g., paralogs of the single drosophila hedgehog gene (SEQ ID No. 19). Three of these members, herein referred to as Desert hedgehog (Dhh), Sonic hedgehog (Shh) and Indian hedgehog (Ihh), apparently exist in all vertebrates, including fish, birds, and mammals. A fourth member, herein referred to as tiggie-winkle hedgehog (Thh), appears specific to fish.
  • Dhh Desert hedgehog
  • Sonic hedgehog Sonic hedgehog
  • Ihh Indian hedgehog
  • Thh tiggie-winkle hedgehog
  • a chicken Shh polypeptide is encoded by SEQ ID No:l; a mouse Dhh polypeptide is encoded by SEQ ID No:2; a mouse Ihh polypeptide is encoded by SEQ ID No:3; a mouse Shh polypeptide is encoded by SEQ ID No:4 a zebrafish Shh polypeptide is encoded by SEQ ID No:5; a human Shh polypeptide is encoded by SEQ ID No:6; a human Ihh polypeptide is encoded by SEQ ID No:7; a human Dhh polypeptide is encoded by SEQ ID No. 8; and a zebrafish Thh is encoded by SEQ ID No. 9.
  • the hedgehog proteins are apparently present naturally in a number of different forms, including a pro-form, a full-length mature form, and several processed fragments thereof.
  • the pro-form includes an N-terminal signal peptide for directed secretion of the extracellular domain, while the full-length mature form lacks this signal sequence.
  • sonic hedgehog undergoes additional proteolytic processing to yield two peptides of approximately 19 kDa and 27 kDa, the 19kDa fragment corresponding to an proteolytic
  • the vertebrate hedgehog proteins can also be modified post-translationally, such as by glycosylation and/or addition of lipophilic moieties, such as stents, fatty acids, etc., though bacterially produced (e.g. unmodified) forms of the proteins still maintain certain of the bioactivities of the native protein.
  • Bioactive fragments of hedgehog polypeptides of the present invention have been generated and are described in great detail in, e.g., PCT publications WO 95/18856 and WO 96/17924. There are a wide range of lipophilic moieties with which hedgehog polypeptides can be derivatived.
  • lipophilic group in the context of being attached to a hedgehog polypeptide, refers to a group having high hydrocarbon content thereby giving the group high affinity to lipid phases.
  • a lipophilic group can be, for example, a relatively long chain alkyl or cycloalkyl (preferably n-alkyl) group having approximately 7 to 30 carbons.
  • the alkyl group may terminate with a hydroxy or primary amine "tail".
  • lipophilic molecules include naturally-occurring and synthetic aromatic and non-aromatic moieties such as fatty acids, sterols, esters and alcohols, other lipid molecules, cage structures such as adamantane and buckminsterfullerenes, and aromatic hydrocarbons such as benzene, perylene, phenanthrene, anthracene, naphthalene, pyrene, chrysene, and naphthacene.
  • aromatic hydrocarbons such as benzene, perylene, phenanthrene, anthracene, naphthalene, pyrene, chrysene, and naphthacene.
  • the hedgehog polypeptide is modified with one or more sterol moieties, such as cholesterol. See, for example, PCT publication WO 96/17924.
  • the cholesterol is preferably added to the C-terminal glycine were the hedgehog polypeptide corresponds to the naturally-occurring N-terminal proteolytic fragment.
  • the hedgehog polypeptide can be modified with a fatty acid moiety, such as a myrostoyl, palmitoyl, stearoyl, or arachidoyl moiety. See, e.g., Pepinsky et al. (1998) J Biol. Chem 273: 14037.
  • At least certain of the biological activities of the hedgehog gene products are unexpectedly potentiated by derivativation of the protein with lipophilic moieties at other sites on the protein and/or by moieties other than cholesterol or fatty acids.
  • Certain aspects of the invention are directed to the use of preparations of hedgehog polypeptides which are modified at sites other than N-terminal or C-terminal residues of the natural processed form of the protein, and/or which are modified at such terminal residues with lipophilic moieties other than a sterol at the C-terminus or fatty acid at the N-terminus.
  • lipophilic molecules are alicyclic hydrocarbons, saturated and unsaturated fatty acids and other lipid and phospholipid moieties, waxes, cholesterol, isoprenoids, terpenes and polyalicyclic hydrocarbons including adamantane and buckminsterfullerenes, vitamins, polyethylene glycol or oligoethylene glycol, (C1-C18)- alkyl phosphate diesters, -O-CH2-CH(OH)-O-(C12-C18)-alkyl, and in particular conjugates with pyrene derivatives.
  • the lipophilic moiety can be a lipophilic dye suitable for use in the invention include, but are not limited to, diphenylhexatriene, Nile Red, N-phenyl-1-naphthylamine, Prodan, Laurodan, Pyrene, Perylene, rhodamine, rhodamine B, tetramethylrhodamine, Texas Red, sulforhodamine, l,l'-didodecyl- 3,3,3', 3'tetramethylindocarbocyanine perchlorate, octadecyl rhodamine B and the BODIPY dyes available from Molecular Probes Inc.
  • exemplary lipophilic moietites include aliphatic carbonyl radical groups include 1- or 2-adamantylacetyl, 3-methyladamant-l-ylacetyl, 3-methyl-3-bromo-l- adamantylacetyl, 1-decalinacetyl, camphoracetyl, camphaneacetyl, noradamantylacetyl, norbornaneacetyl, bicyclo[2.2.2.]-oct-5-eneacetyl, l-methoxybicyclo[2.2.2.]-oct-5-ene-2- carbonyl, cis-5-norbornene-endo-2,3-dicarbonyl, 5-norbornen-2-ylacetyl, (lR)-( - )- myrtentaneacetyl, 2-norbornaneacetyl, anti-3-oxo-tricyclo[2.2.1.0 ⁇ 2,6> ]-heptane-7- carbonyl
  • the hedgehog polypeptide can be linked to the hydrophobic moiety in a number of ways including by chemical coupling means, or by genetic engineering.
  • the preferred cross-linking agents are heterobifunctional cross-linkers, which can be used to link the hedgehog polypeptide and hydrophobic moiety in a stepwise manner.
  • Heterobifunctional cross-linkers provide the ability to design more specific coupling methods for conjugating to proteins, thereby reducing the occurrences of unwanted side reactions such as homo-protein polymers.
  • a wide variety of heterobifunctional cross-linkers are known in the art.
  • SMCC succini
  • cross-linking agents having N-hydroxysuccinimide moieties can be obtained as the N-hydroxysulfosuccinimide analogs, which generally have greater water solubility.
  • those cross-linking agents having disulfide bridges within the linking chain can be synthesized instead as the alkyl derivatives so as to reduce the amount of linker cleavage in vivo.
  • DSS Disuccinimidyl suberate
  • BMH bismaleimidohexane
  • DMP dimethylpimelimidate-2 HC1
  • BASED bis-[ ⁇ -(4-azidosalicylamido)ethyl]disulfide
  • BASED bis-[ ⁇ -(4-azidosalicylamido)ethyl]disulfide
  • SANPAH N- succinimidyl-6(4'-azido-2'-nitrophenyl- amino)hexanoate
  • heterobifunctional cross-linkers contain the primary amine reactive group, N-hydroxysuccinimide (NHS), or its water soluble analog N-hydroxysulfosuccinimide (sulfo-NHS).
  • NHS N-hydroxysuccinimide
  • sulfo-NHS water soluble analog N-hydroxysulfosuccinimide
  • thiol reactive group Another reactive group useful as part of a heterobifunctional cross-linker is a thiol reactive group.
  • Common thiol reactive groups include maleimides, halogens, and pyridyl disulfides. Maleimides react specifically with free sulfhydryls (cysteine residues) in minutes, under slightly acidic to neutral (pH 6.5-7.5) conditions. Halogens (iodoacetyl functions) react with -SH groups at physiological pH's. Both of these reactive groups result in the formation of stable thioether bonds.
  • the third component of the heterobifunctional cross-linker is the spacer arm or bridge.
  • the bridge is the structure that connects the two reactive ends.
  • the most apparent attribute of the bridge is its effect on steric hindrance.
  • a longer bridge can more easily span the distance necessary to link two complex biomolecules.
  • SMPB has a span of 14.5 angstroms.
  • Preparing protein-protein conjugates using heterobifunctional reagents is a two- step process involving the amine reaction and the sulfhydryl reaction.
  • the protein chosen should contain a primary amine. This can be lysine epsilon amines or a primary alpha amine found at the N-terminus of most proteins.
  • the protein should not contain free sulfhydryl groups. In cases where both proteins to be conjugated contain free sulfhydryl groups, one protein can be modified so that all sulfhydryls are blocked using for instance, N-ethylmaleimide (see Partis et al. (1983) J. Pro. Chem.
  • Ellman's Reagent can be used to calculate the quantity of sulfhydryls in a particular protein (see for example Ellman et al. (1958) Arch. Biochem. Biophys. 74:443 and Riddles et al. (1979) Anal. Biochem. 94:75, incorporated by reference herein).
  • the reaction buffer should be free of extraneous amines and sulfhydryls.
  • the pH of the reaction buffer should be 7.0-7.5. This pH range prevents maleimide groups from reacting with amines, preserving the maleimide group for the second reaction with sulfhydryls.
  • the NHS-ester containing cross-linkers have limited water solubility. They should be dissolved in a minimal amount of organic solvent (DMF or DMSO) before introducing the cross-linker into the reaction mixture.
  • the cross-linker/solvent forms an emulsion which will allow the reaction to occur.
  • the sulfo-NHS ester analogs are more water soluble, and can be added directly to the reaction buffer. Buffers of high ionic strength should be avoided, as they have a tendency to "salt out" the sulfo-NHS esters. To avoid loss of reactivity due to hydrolysis, the cross-linker is added to the reaction mixture immediately after dissolving the protein solution.
  • the reactions can be more efficient in concentrated protein solutions.
  • the rate of hydrolysis of the NHS and sulfo-NHS esters will also increase with increasing pH. Higher temperatures will increase the reaction rates for both hydrolysis and acylation.
  • the first protein is now activated, with a sulfhydryl reactive moiety.
  • the activated protein may be isolated from the reaction mixture by simple gel filtration or dialysis.
  • the lipophilic group chosen for reaction with maleimides, activated halogens, or pyridyl disulfides must contain a free sulfhydryl.
  • a primary amine may be modified with to add a sulfhydryl
  • the buffer should be degassed to prevent oxidation of sulfhydryl groups.
  • EDTA may be added to chelate any oxidizing metals that may be present in the buffer.
  • Buffers should be free of any sulfhydryl containing compounds.
  • Maleimides react specifically with -SH groups at slightly acidic to neutral pH ranges (6.5-7.5). A neutral pH is sufficient for reactions involving halogens and pyridyl disulfides. Under these conditions, maleimides generally react with -SH groups within a matter of minutes. Longer reaction times are required for halogens and pyridyl disulfides.
  • the first sulfhydryl reactive-protein prepared in the amine reaction step is mixed with the sulfhydryl-containing lipophilic group under the appropriate buffer conditions.
  • the conjugates can be isolated from the reaction mixture by methods such as gel filtration or by dialysis.
  • activated lipophilic moieties for conjugation include: N-(l- pyrene)maleimide; 2,5-dimethoxystilbene-4'-maleimide, eosin-5-maleimide; fluorescein- 5-maleimide; N-(4-(6-dimethylamino- 2-benzofuranyl)phenyl)maleimide; benzophenone-4-maleimide; 4-dimethylaminophenylazophenyl- 4'-maleimide (DABMI), tetramethylrhodamine-5-maleimide, tetramethylrhodamine-6-maleimide, Rhodamine RedTM C2 maleimide, N-(5-aminopentyl)maleimide, trifluoroacetic acid salt, N-(2- aminoethyl)maleimide, trifluoroacetic acid salt, Oregon GreenTM 488 maleimide, N-(2- ((2-(((4-azido- 2,3,5,6-male
  • TPM-SS1 2-(l-(3-dimethylaminopropyl) -indol-3-yl)-3-(indol-3-yl) maleimide (bisindolylmaleimide; GF 109203X), BODIPY® FL N-(2-aminoethyl)maleimide, N-(7- dimethylamino- 4-methylcoumarin-3-yl)maleimide (DACM), AlexaTM 488 C5 maleimide, AlexaTM 594 C5 maleimide, sodium saltN-(l-pyrene)maleimide, 2,5- dimethoxystilbene-4'-maleimide, eosin-5 -maleimide, fluorescein-5-maleimide, N-(4-(6- dimethylamino- 2-benzofuranyl)phenyl)maleimide, benzophenone-4-maleimide, 4- dimethylaminophenylazophenyl- 4'-maleimide, l
  • the hedgehog polypeptide can be derivatived using pyrene maleimide, which can be purchased from Molecular Probes (Eugene, Oreg.), e.g., N-(l- pyrene)maleimide or 1-pyrenemethyl iodoacetate (PMIA ester).
  • pyrene maleimide which can be purchased from Molecular Probes (Eugene, Oreg.), e.g., N-(l- pyrene)maleimide or 1-pyrenemethyl iodoacetate (PMIA ester).
  • the modified hedgehog polypeptide of this invention can be constructed as a fusion protein, containing the hedgehog polypeptide and the hydrophobic moiety as one contiguous polypeptide chain.
  • the lipophilic moiety is an amphipathic polypeptide.
  • amphipathic polypeptide such as magainin, cecropin, attacin, melittin, gramicidin S, alpha-toxin of Staph. aureus, alamethicin or a synthetic amphipathic polypeptide.
  • Fusogenic coat proteins from viral particles can also be a convenient source of amphipathic sequences for the subject hedgehog proteins
  • modified hh polypeptides e.g., for such purposes as enhancing therapeutic or prophylactic efficacy, or stability (e.g., ex vivo shelf life and resistance to proteolytic degradation in vivo).
  • modified peptides can be produced, for instance, by amino acid substitution, deletion, or addition.
  • Modified hedgehog polypeptides can also include those with altered post-translational processing relative to a naturally occurring hedgehog protein, e.g., altered glycosylation, cholesterolization, prenylation and the like.
  • the hedgehog therapeutic is a polypeptide encodable by a nucleotide sequence that hybridizes under stringent conditions to a hedgehog coding sequence represented in one or more of SEQ ID Nos: 1-7.
  • Appropriate stringency conditions which promote DNA hybridization for example, 6.0 x sodium chloride/sodium citrate (SSC) at about 45°C, followed by a wash of 2.0 x SSC at 50°C, are known to those skilled in the art or can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1-6.3.6.
  • the salt concentration in the wash step can be selected from a low stringency of about 2.0 x SSC at 50°C to a high stringency of about 0.2 x SSC at 50°C.
  • the temperature in the wash step can be increased from low stringency conditions at room temperature, about 22°C, to high stringency conditions at about 65°C.
  • genes for other hedgehog proteins can be obtained from mRNA or genomic DNA samples using techniques well known in the art.
  • a cDNA encoding a hedgehog protein can be obtained by isolating total mRNA from a cell, e.g. a mammalian cell, e.g. a human cell, including embryonic cells. Double stranded cDNAs can then be prepared from the total mRNA, and subsequently inserted into a suitable plasmid or bacteriophage vector using any one of a number of known techniques.
  • the gene encoding a hedgehog protein can also be cloned using established polymerase chain reaction techniques.
  • Preferred nucleic acids encode a hedgehog polypeptide comprising an amino acid sequence at least 60% homologous or identical, more preferably 70% homologous or identical, and most preferably 80% homologous or identical with an amino acid sequence selected from the group consisting of SEQ ID Nos:8-14.
  • Nucleic acids which encode polypeptides at least about 90%, more preferably at least about 95%, and most preferably at least about 98-99% homology or identity with an amino acid sequence represented in one of SEQ ID Nos: 8- 14 are also within the scope of the invention.
  • hedgehog polypeptides preferred by the present invention are at least 60% homologous or identical, more preferably 70%) homologous or identical and most preferably 80% homologous or identical with an amino acid sequence represented by any of SEQ ID Nos: 8- 14.
  • Polypeptides which are at least 90%, more preferably at least 95%, and most preferably at least about 98-99% homologous or identical with a sequence selected from the group consisting of SEQ ID Nos:8-14 are also within the scope of the invention. The only prerequisite is that the hedgehog polypeptide is capable of modulating the growth of muscle cells.
  • recombinant protein refers to a polypeptide of the present invention which is produced by recombinant DNA techniques, wherein generally, DNA encoding a hedgehog polypeptide is inserted into a suitable expression vector which is in turn used to transform a host cell to produce the heterologous protein.
  • the phrase "derived from”, with respect to a recombinant hedgehog gene is meant to include within the meaning of "recombinant protein” those proteins having an amino acid sequence of a native hedgehog protein, or an amino acid sequence similar thereto which is generated by mutations including substitutions and deletions (including truncation) of a naturally occurring form of the protein.
  • the method of the present invention can also be carried out using variant forms of the naturally occurring hedgehog polypeptides, e.g., mutational variants.
  • hedgehog polypeptides can be produced by standard biological techniques or by chemical synthesis.
  • a host cell transfected with a nucleic acid vector directing expression of a nucleotide sequence encoding the subject polypeptides can be cultured under appropriate conditions to allow expression of the peptide to occur.
  • the polypeptide hedgehog may be secreted and isolated from a mixture of cells and medium containing the recombinant hedgehog polypeptide.
  • the peptide may be retained cytoplasmically by removing the signal peptide sequence from the recombinant hedgehog gene and the cells harvested, lysed and the protein isolated.
  • a cell culture includes host cells, media and other byproducts. Suitable media for cell culture are well known in the art.
  • the recombinant hedgehog polypeptide can be isolated from cell culture medium, host cells, or both using techniques known in the art for purifying proteins including ion-exchange chromatography, gel filtration chromatography, ultrafiltration, electrophoresis, and immunoaffinity purification with antibodies specific for such peptide.
  • the recombinant hedgehog polypeptide is a fusion protein containing a domain which facilitates its purification, such as an hedgehoglGST fusion protein.
  • the host cell may be any prokaryotic or eukaryotic cell.
  • Recombinant hedgehog genes can be produced by ligating nucleic acid encoding an hedgehog protein, or a portion thereof, into a vector suitable for expression in either prokaryotic cells, eukaryotic cells, or both.
  • Expression vectors for production of recombinant forms of the subject hedgehog polypeptides include plasmids and other vectors.
  • suitable vectors for the expression of a hedgehog polypeptide include plasmids of the types: pBR322-derived plasmids, pEMBL-derived plasmids, pEX-derived plasmids, pBTac-derived plasmids and pUC-derived plasmids for expression in prokaryotic cells, such as E. coli.
  • YEP24, YIP5, YEP51, YEP52, pYES2, and YRP17 are cloning and expression vehicles useful in the introduction of genetic constructs into S. cerevisiae (see, for example, Broach et al. (1983) in Experimental Manipulation of Gene Expression, ed. M. Inouye Academic Press, p. 83, incorporated by reference herein).
  • These vectors can replicate in E. coli due to the presence of the pBR322 ori, and in S. cerevisiae due to the replication determinant of the yeast 2 micron plasmid.
  • drug resistance markers such as ampicillin can be used.
  • an hedgehog polypeptide is produced recombinantly utilizing an expression vector generated by sub- cloning the coding sequence of one of the hedgehog genes represented in SEQ ID Nos:l- 7.
  • the preferred mammalian expression vectors contain both prokaryotic sequences, to facilitate the propagation of the vector in bacteria, and one or more eukaryotic transcription units that are expressed in eukaryotic cells.
  • the pcDNAI/amp, pcDNAI/neo, pRc/CMV, pSV2gpt, pSV2neo, pSV2-dhfr, pTk2, pRSVneo, pMSG, pSVT7, pko-neo and pHyg derived vectors are examples of mammalian expression vectors suitable for transfection of eukaryotic cells.
  • vectors are modified with sequences from bacterial plasmids, such as pBR322, to facilitate replication and drug resistance selection in both prokaryotic and eukaryotic cells.
  • derivatives of viruses such as the bovine papillomavirus (BPV-1), or Epstein-Barr virus (pHEBo, pREP-derived and p205) can be used for transient expression of proteins in eukaryotic cells.
  • BBV-1 bovine papillomavirus
  • pHEBo Epstein-Barr virus
  • the various methods employed in the preparation of the plasmids and transformation of host organisms are well known in the art.
  • suitable expression systems for both prokaryotic and eukaryotic cells, as well as general recombinant procedures see Molecular Cloning A laboratory Manual, 2nd Ed., ed.
  • baculovirus expression systems include pVL-derived vectors (such as pVL1392, pVL1393 and pVL941), pAcUW-derived vectors (such as pAcUWl), and pBlueBac-derived vectors (such as the ⁇ -gal containing pBlueBac III).
  • pVL-derived vectors such as pVL1392, pVL1393 and pVL941
  • pAcUW-derived vectors such as pAcUWl
  • pBlueBac-derived vectors such as the ⁇ -gal containing pBlueBac III.
  • MAP methionine aminopeptidase
  • removal of an N-terminal methionine can be achieved either in vivo by expressing hedgehog-de ⁇ ved polypeptides in a host which produces MAP (e.g., E. coli or CM89 or S. cerevisiae), or in vitro by use of purified MAP (e.g., procedure of Miller et al., supra).
  • MAP e.g., E. coli or CM89 or S. cerevisiae
  • the coding sequences for the polypeptide can be incorporated as a part of a fusion gene including a nucleotide sequence encoding a different polypeptide.
  • fusion proteins can also facilitate the expression of proteins, and accordingly, can be used in the expression of the hedgehog polypeptides of the present invention.
  • hedgehog polypeptides can be generated as glutathione- S-transferase (GST-fusion) proteins.
  • GST-fusion proteins can enable easy purification of the hedgehog polypeptide, as for example by the use of glutathione- derivatized matrices (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al.
  • a fusion gene coding for a purification leader sequence such as a poly-(His)/enterokinase cleavage site sequence, can be used to replace the signal sequence which naturally occurs at the N- terminus of the hedgehog protein (e.g.of the pro-form, in order to permit purification of the po ⁇ y ⁇ is)-hedgehog protein by affinity chromatography using a Ni 2+ metal resin.
  • the purification leader sequence can then be subsequently removed by treatment with enterokinase (e.g., see Hochuli et al. (1987) J Chromatography 411 :177; and Janknecht et al. PN S 88:8972).
  • fusion genes are known to those skilled in the art. Essentially, the joining of various DNA fragments coding for different polypeptide sequences is performed in accordance with conventional techniques, employing blunt- ended or stagger-ended termini for ligation, restriction enzyme digestion to provide for appropriate termini, filling-in of cohesive ends as appropriate, alkaline phosphatase treatment to avoid undesirable joining, and enzymatic ligation.
  • the fusion gene can be synthesized by conventional techniques including automated DNA synthesizers.
  • PCR amplification of gene fragments can be carried out using anchor primers which give rise to complementary overhangs between two consecutive gene fragments which can subsequently be annealed to generate a chimeric gene sequence (see, for example, Current Protocols in Molecular Biology, eds. Ausubel et al. John Wiley & Sons: 1992).
  • Hedgehog polypeptides may also be chemically modified to create hedgehog derivatives by forming covalent or aggregate conjugates with other chemical moieties, such as glycosyl groups, cholesterol, isoprenoids, lipids, phosphate, acetyl groups and the like.
  • Covalent derivatives of hedgehog proteins can be prepared by linking the chemical moieties to functional groups on amino acid sidechains of the protein or at the N- terminus or at the C-terminus of the polypeptide.
  • hedgehog proteins can be generated to include a moiety, other than sequence naturally associated with the protein, that binds a component of the extracellular matrix and enhances localization of the analog to cell surfaces.
  • sequences derived from the fibronectin "type-Ill repeat”, such as a tetrapeptide sequence R-G-D-S (Pierschbacher et al. (1984) Nature 309:30-3; and Kornblihtt et al. (1985) EMBO 4:1755-9) can be added to the hedgehog polypeptide to support attachment of the chimeric molecule to a cell through binding ECM components (Ruoslahti et al. (1987) Science 238:491-497; Pierschbacheret al. (1987) J. Biol. Chem. 262:17294-8.; Hynes (1987) Cell 48:549-54; and Hynes (1992) Cell 69:11-25).
  • the hedgehog polypeptide is isolated from, or is otherwise substantially free of, other cellular proteins, especially other extracellular or cell surface associated proteins which may normally be associated with the hedgehog polypeptide, unless provided in the form of fusion protein with the hedgehog polypeptide.
  • the term "substantially free of other cellular or extracellular proteins" (also referred to herein as “contaminating proteins") or “substantially pure preparations” or “purified preparations” are defined as encompassing preparations of hedgehog polypeptides having less than 20%) (by dry weight) contaminating protein, and preferably having less than 5% contaminating protein.
  • purified it is meant that the indicated molecule is present in the substantial absence of other biological macromolecules, such as other proteins.
  • isolated hedgehog polypeptides can include all or a portion of the amino acid sequences represented in any of SEQ ID Nos: 10-18 or 20, or a homologous sequence thereto.
  • Preferred fragments of the subject hedgehog proteins correspond to the N-terminal and C-terminal proteolytic fragments of the mature protein.
  • Bioactive fragments of hedgehog polypeptides are described in great detail in PCT publications WO 95/18856 and WO 96/17924.
  • preferred hedgehog therapeutics include at least 50 (contiguous) amino acid residues of a hedgehog polypeptide, more preferably at least 100 (contiguous), and even more preferably at least 150 (contiguous) residues.
  • Another preferred hedgehog polypeptide which can be included in the hedgehog therapeutic is an N-terminal fragment of the mature protein having a molecular weight of approximately 19 kDa.
  • Preferred human hedgehog proteins include N-terminal fragments corresponding approximately to residues 24-197 of SEQ ID No. 15, 28-202 of SEQ ID No. 16, and 23- 198 of SEQ ID No. 17.
  • corresponding approximately it is meant that the sequence of interest is at most 20 amino acid residues different in length to the reference sequence, though more preferably at most 5, 10 or 15 amino acid different in length.
  • isolated hedgehog polypeptides can include all or a portion of the amino acid sequences represented in SEQ ID No:8, SEQ ID No:9, SEQ ID No:10, SEQ ID No:l 1, SEQ ID No:12, SEQ ID No:13 or SEQ ID No:14, or a homologous sequence thereto.
  • Preferred fragments of the subject hedgehog proteins correspond to the N-terminal and C-terminal proteolytic fragments of the mature protein. Bioactive fragments of hedgehog polypeptides are described in great detail in PCT publications WO 95/18856 and WO 96/17924.
  • Still other preferred hedgehog polypeptides includes an amino acid sequence represented by the formula A-B wherein: (i) A represents all or the portion of the amino acid sequence designated by residues 1-168 of SEQ ID No:21; and B represents at least one amino acid residue of the amino acid sequence designated by residues 169-221 of SEQ ID No:21; (ii) A represents all or the portion of the amino acid sequence designated by residues 24-193 of SEQ ID No: 15; and B represents at least one amino acid residue of the amino acid sequence designated by residues 194-250 of SEQ ID No: 15; (iii) A represents all or the portion of the amino acid sequence designated by residues 25-193 of SEQ ID No: 13; and B represents at least one amino acid residue of the amino acid sequence designated by residues 194-250 of SEQ ID No: 13; (iv) A represents all or the portion of the amino acid sequence designated by residues 23-193 of SEQ ID No: 11 ; and B represents at least one amino acid residue of the amino acid sequence designated by residues 194
  • a and B together represent a contiguous polypeptide sequence designated sequence
  • A represents at least 25, 50, 75, 100, 125 or 150 (contiguous) amino acids of the designated sequence
  • B represents at least 5, 10, or 20 (contiguous) amino acid residues of the amino acid sequence designated by corresponding entry in the sequence listing
  • a and B together preferably represent a contiguous sequence corresponding to the sequence listing entry.
  • Similar fragments from other hedgehog also contemplated, e.g., fragments which correspond to the preferred fragments from the sequence listing entries which are enumerated above.
  • the hedgehog polypeptide includes a C-terminal glycine (or other appropriate residue) which is derivatized with a cholesterol.
  • Isolated peptidyl portions of hedgehog proteins can be obtained by screening peptides recombinantly produced from the corresponding fragment of the nucleic acid encoding such peptides.
  • fragments can be chemically synthesized using techniques known in the art such as conventional Merrifield solid phase f-Moc or t-Boc chemistry.
  • a hedgehog polypeptide of the present invention may be arbitrarily divided into fragments of desired length with no overlap of the fragments, or preferably divided into overlapping fragments of a desired length.
  • the fragments can be produced (recombinantly or by chemical synthesis) and tested to identify those peptidyl fragments which can function as either agonists or antagonists of a wild-type (e.g., "authentic") hedgehog protein.
  • a wild-type e.g., "authentic” hedgehog protein.
  • the recombinant hedgehog polypeptides of the present invention also include homologs of the authentic hedgehog proteins, such as versions of those protein which are resistant to proteolytic cleavage, as for example, due to mutations which alter potential cleavage sequences or which inactivate an enzymatic activity associated with the protein.
  • Hedgehog homologs of the present invention also include proteins which have been post- translationally modified in a manner different than the authentic protein.
  • Exemplary derivatives of hedgehog proteins include polypeptides which lack N-glycosylation sites (e.g. to produce an unglycosylated protein), which lack sites for cholesterolization, and/or which lack N-terminal and/or C-terminal sequences.
  • Modification of the structure of the subject hedgehog polypeptides can also be for such purposes as enhancing therapeutic or prophylactic efficacy, or stability (e.g., ex vivo shelf life and resistance to proteolytic degradation in vivo).
  • Such modified peptides when designed to retain at least one activity of the naturally-occurring form of the protein, are considered functional equivalents of the hedgehog polypeptides described in more detail herein.
  • Such modified peptides can be produced, for instance, by amino acid substitution, deletion, or addition.
  • Whether a change in the amino acid sequence of a peptide results in a functional hedgehog homolog can be readily determined by assessing the ability of the variant peptide to produce a response in cells in a fashion similar to the wild-type protein, or competitively inhibit such a response.
  • Polypeptides in which more than one replacement has taken place can readily be tested in the same manner. It is specifically contemplated that the methods of the present invention can be carried using homologs of naturally occurring hedgehog proteins. In one embodiment, the invention contemplates using hedgehog polypeptides generated by combinatorial mutagenesis.
  • Such methods are convenient for generating both point and truncation mutants, and can be especially useful for identifying potential variant sequences (e.g. homologs) that are functional in binding to a receptor for hedgehog proteins.
  • the purpose of screening such combinatorial libraries is to generate, for example, novel hedgehog homologs which can act as either agonists or antagonist.
  • hedgehog homologs can be engineered by the present method to provide more efficient binding to a cognate receptor, such as patched, yet still retain at least a portion of an activity associated with hedgehog.
  • combinatorially-derived homologs can be generated to have an increased potency relative to a naturally occurring form of the protein.
  • hedgehog homologs can be generated by the present combinatorial approach to act as antagonists, in that they are able to mimic, for example, binding to other extracellular matrix components (such as receptors), yet not induce any biological response, thereby inhibiting the action of authentic hedgehog or hedgehog agonists.
  • manipulation of certain domains of hedgehog by the present method can provide domains more suitable for use in fusion proteins, such as one that incorporates portions of other proteins which are derived from the extracellular matrix and/or which bind extracellular matrix components.
  • PCT publication WO92/15679 illustrate specific techniques which one skilled in the art could utilize to generate libraries of hedgehog variants which can be rapidly screened to identify variants/fragments which retained a particular activity of the hedgehog polypeptides. These techniques are exemplary of the art and demonstrate that large libraries of related variants/truncants can be generated and assayed to isolate particular variants without undue experimentation. Gustin et al. (1993) Virology 193:653, and Bass et al. (1990) Proteins: Structure, Function and Genetics 8:309-314 also describe other exemplary techniques from the art which can be adapted as means for generating mutagenic variants of hedgehog polypeptides.
  • the amino acid sequences for a population of hedgehog homologs or other related proteins are aligned, preferably to promote the highest homology possible.
  • a population of variants can include, for example, hedgehog homologs from one or more species.
  • Amino acids which appear at each position of the aligned sequences are selected to create a degenerate set of combinatorial sequences.
  • the variegated library of hedgehog variants is generated by combinatorial mutagenesis at the nucleic acid level, and is encoded by a variegated gene library.
  • a mixture of synthetic oligonucleotides can be enzymatically ligated into gene sequences such that the degenerate set of potential hedgehog sequences are expressible as individual polypeptides, or alternatively, as a set of larger fusion proteins (e.g. for phage display) containing the set oi hedgehog sequences therein.
  • the amino acid sequences of interest can be aligned relative to sequence homology.
  • the presence or absence of amino acids from an aligned sequence of a particular variant is relative to a chosen consensus length of a reference sequence, which can be real or artificial.
  • alignment of exons 1 , 2 and a portion of exon 3 encoded sequences e.g.
  • Shh polypeptides represented by the general formula: C-G-P-G-R-G-X(l)-G-X(2)-R-R-H-P-K-K-L-T-P-L-A-Y-K-Q-F-I-P-N-
  • Xaa(l) represents Gly, Ala, Val, Leu, He, Phe, Tyr or Trp ;
  • Xaa(2) represents Arg, His or Lys;
  • Xaa(3) represents Gly, Ala, Val, Leu, He, Ser or Thr;
  • Xaa(4) represents Gly, Ala, Val, Leu, He, Ser or Thr;
  • Xaa(5) represents Lys, Arg, His, Asn or Gin;
  • Xaa(6) represents Lys, Arg or His;
  • Xaa(7) represents Ser, Thr, Tyr, Trp or Phe;
  • Xaa(8) represents Lys, Arg or His;
  • Xaa(9) represents Met, Cys, Ser or Thr;
  • Xaa(lO) represents Gly, Ala, Val, Leu, He, Ser or Thr;
  • Xaa(l l) represents Leu, Val, Met, Thr or Ser;
  • Xaa(12) represents
  • Xaa(l) represents Gly, Ala, Val, Leu, He, Pro, Phe or Tyr;
  • Xaa(2) represents Gly, Ala, Val, Leu or He;
  • Xaa(3) represents Gly, Ala, Val, Leu, He, Lys, His or Arg;
  • Xaa(4) represents Lys, Arg or His;
  • Xaa(5) represents Phe, Trp, Tyr or an amino acid gap;
  • Xaa(6) represents Gly, Ala, Val, Leu, He or an amino acid gap;
  • Xaa(7) represents Asn, Gin, His, Arg or Lys;
  • Xaa(8) represents Gly, Ala, Val, Leu, He, Ser or Thr;
  • Xaa(9) represents Gly, Ala, Val, Leu, He, Ser or Thr;
  • Xaa(lO) represents Gly, Ala, Val, Leu, He, Ser or Thr;
  • Xaa(l 1) represents
  • the library of potential hedgehog homologs can be generated from a degenerate oligonucleotide sequence.
  • Chemical synthesis of a degenerate gene sequence can be carried out in an automatic DNA synthesizer, and the synthetic genes then ligated into an appropriate expression vector.
  • the purpose of a degenerate set of genes is to provide, in one mixture, all of the sequences encoding the desired set of potential hedgehog sequences.
  • the synthesis of degenerate oligonucleotides is well known in the art (see for example, Narang, SA (1983) Tetrahedron 39:3; Itakura et al. (1981) Recombinant DNA, Proc 3rd Cleveland Sympos. Macromolecules, ed.
  • a wide range of techniques are known in the art for screening gene products of combinatorial libraries made by point mutations, and for screening cDNA libraries for gene products having a certain property. Such techniques will be generally adaptable for rapid screening of the gene libraries generated by the combinatorial mutagenesis of hedgehog homologs.
  • the most widely used techniques for screening large gene libraries typically comprises cloning the gene library into replicable expression vectors, transforming appropriate cells with the resulting library of vectors, and expressing the combinatorial genes under conditions in which detection of a desired activity facilitates relatively easy isolation of the vector encoding the gene whose product was detected.
  • Each of the illustrative assays described below are amenable to high through-put analysis as necessary to screen large numbers of degenerate hedgehog sequences created by combinatorial mutagenesis techniques.
  • the combinatorial library is designed to be secreted (e.g. the polypeptides of the library all include a signal sequence but no transmembrane or cytoplasmic domains), and is used to transfect a eukaryotic cell that can be co-cultured with muscle stem cells.
  • a functional hedgehog protein secreted by the cells expressing the combinatorial library will diffuse to neighboring muscle cells and induce a particular biological response, such as proliferation or differentiation. The pattern of detection of such a change in phenotype will resemble a gradient function, and will allow the isolation (generally after several repetitive rounds of selection) of cells producing hedgehog homologs active as muscle-trophic agents.
  • hedgehog antagonists can be selected in similar fashion by the ability of the cell producing a functional antagonist to protect neighboring cells (e.g., to inhibit proliferation) from the effect of wild-type hedgehog added to the culture media.
  • target muscle cells are cultured in 24-well microtitre plates.
  • Other eukaryotic cells are transfected with the combinatorial hedgehog gene library and cultured in cell culture inserts (e.g. Collaborative Biomedical Products, Catalog #40446) that are able to fit into the wells of the microtitre plate.
  • the cell culture inserts are placed in the wells such that recombinant hedgehog homologs secreted by the cells in the insert can diffuse through the porous bottom of the insert and contact the target cells in the microtitre plate wells.
  • After a period of time sufficient for functional forms of a hedgehog protein to produce a measurable response in the target cells, such as growth state the inserts are removed and the effect of the variant hedgehog proteins on the target cells determined.
  • Cells from the inserts corresponding to wells which score positive for activity can be split and re-cultured on several inserts, the process being repeated until the active clones are identified.
  • the candidate hedgehog gene products are displayed on the surface of a cell or viral particle, and the ability of particular cells or viral particles to associate with a hedgehog-binding moiety (such as the patched protein or other hedgehog receptor) via this gene product is detected in a "panning assay".
  • a hedgehog-binding moiety such as the patched protein or other hedgehog receptor
  • panning steps can be carried out on cells cultured from embryos.
  • the gene library can be cloned into the gene for a surface membrane protein of a bacterial cell, and the resulting fusion protein detected by panning (Ladner et al., WO 88/06630; Fuchs et al. (1991) Bio/Technology 9:1370-1371; and Goward et al.
  • TIBS 18:136-140 fluorescently labeled molecules which bind hedgehog can be used to score for potentially functional hedgehog homologs.
  • Cells can be visually inspected and separated under a fluorescence microscope, or, where the morphology of the cell permits, separated by a fluorescence-activated cell sorter.
  • the gene library is expressed as a fusion protein on the surface of a viral particle.
  • foreign peptide sequences can be expressed on the surface of infectious phage, thereby conferring two significant benefits.
  • E.coli filamentous phages Ml 3, fd, and fl are most often used in phage display libraries, as either of the phage gill or gVIII coat proteins can be used to generate fusion proteins without disrupting the ultimate packaging of the viral particle (Ladner et al. PCT publication WO 90/02909; Garrard et al., PCT publication WO 92/09690; Marks et al. (1992) J. Biol. Chem. 267:16007-16010; Griffths et al. (1993) EMBO J 12:725-734; Clackson et al. (1991) Nature 352:624-628; and Barbas et al. (1992) PNAS 89:4457- 4461).
  • the recombinant phage antibody system (RPAS, Pharamacia Catalog number 27-9400-01) can be easily modified for use in expressing and screening hedgehog combinatorial libraries.
  • the pCANTAB 5 phagemid of the RPAS kit contains the gene which encodes the phage gill coat protein.
  • the hedgehog combinatorial gene library can be cloned into the phagemid adjacent to the gill signal sequence such that it will be expressed as a gill fusion protein.
  • the phagemid is used to transform competent E. coli TGI cells. Transformed cells are subsequently infected with M13KO7 helper phage to rescue the phagemid and its candidate hedgehog gene insert.
  • the resulting recombinant phage contain phagemid DNA encoding a specific candidate hedgehog, and display one or more copies of the corresponding fusion coat protein.
  • the phage-displayed candidate hedgehog proteins which are capable of binding an hedgehog receptor are selected or enriched by panning.
  • the phage library can be applied to cells which express the patched protein and unbound phage washed away from the cells.
  • the bound phage is then isolated, and if the recombinant phage express at least one copy of the wild type gill coat protein, they will retain their ability to infect E. coli. Thus, successive rounds of reinfection of E.
  • Combinatorial mutagenesis has a potential to generate very large libraries of mutant proteins, e.g., in the order of 10 26 molecules. Combinatorial libraries of this size may be technically challenging to screen even with high throughput screening assays such as phage display.
  • REM recursive ensemble mutagenesis
  • REM is an algorithm which enhances the frequency of functional mutants in a library when an appropriate selection or screening method is employed (Arkin and Yourvan, 1992, PNAS USA 89:7811-7815; Yourvan et al., 1992, Parallel Problem Solving from Nature, 2., In Maenner and Manderick, eds., Elsevir Publishing Co., Amsterdam, pp. 401-410; Delgrave et al., 1993, Protein Engineering 6(3):327-331).
  • the invention also provides for reduction of the hedgehog protein to generate mimetics, e.g. peptide or non-peptide agents, which are able to disrupt binding of a hedgehog polypeptide of the present invention with an hedgehog receptor.
  • mimetics e.g. peptide or non-peptide agents
  • mutagenic techniques as described above are also useful to map the determinants of the hedgehog proteins which participate in protein-protein interactions involved in, for example, binding of the subject hedgehog polypeptide to other extracellular matrix components.
  • the critical residues of a subject hedgehog polypeptide which are involved in molecular recognition of an hedgehog receptor such as patched can be determined and used to generate hedgehog-derived peptidomimetics which competitively inhibit binding of the authentic hedgehog protein with that moiety.
  • peptidomimetic compounds By employing, for example, scanning mutagenesis to map the amino acid residues of each of the subject hedgehog proteins which are involved in binding other extracellular proteins, peptidomimetic compounds can be generated which mimic those residues of the hedgehog protein which facilitate the interaction. Such mimetics may then be used to interfere with the normal function of a hedgehog protein.
  • non-hydrolyzable peptide analogs of such residues can be generated using benzodiazepine (e.g., see Freidinger et al. in Peptides: Chemistry and Biology, G.R. Marshall ed., ESCOM Publisher: Leiden, Netherlands, 1988), azepine (e.g., see Huffman et al. in Peptides: Chemistry and Biology, G.R.
  • Recombinantly produced forms of the hedgehog proteins can be produced using, e.g, expression vectors containing a nucleic acid encoding a hedgehog polypeptide, operably linked to at least one transcriptional regulatory sequence. Operably linked is intended to mean that the nucleotide sequence is linked to a regulatory sequence in a manner which allows expression of the nucleotide sequence.
  • transcriptional regulatory sequence includes promoters, enhancers and other expression control elements.
  • Such regulatory sequences are described in Goeddel; Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, CA (1990).
  • any of a wide variety of expression control sequences, sequences that control the expression of a DNA sequence when operatively linked to it, may be used in these vectors to express DNA sequences encoding hedgehog polypeptide.
  • Such useful expression control sequences include, for example, a viral LTR, such as the LTR of the Moloney murine leukemia virus, the early and late promoters of SV40, adenovirus or cytomegalovirus immediate early promoter, the lac system, the trp system, the TAC or TRC system, T7 promoter whose expression is directed by T7 RNA polymerase, the major operator and promoter regions of phage ⁇ , the control regions for fd coat protein, the promoter for 3-phosphoglycerate kinase or other glycolytic enzymes, the promoters of acid phosphatase, e.g., Pho5, the promoters of the yeast ⁇ -mating factors, the polyhedron promoter of the baculovirus system and other sequences known to control the expression of genes of prokaryotic or eukaryotic cells or their viruses, and various combinations thereof.
  • a viral LTR such as the LTR of the Moloney murine leukemia virus, the early
  • the design of the expression vector may depend on such factors as the choice of the host cell to be transformed and/or the type of protein desired to be expressed. Moreover, the vector's copy number, the ability to control that copy number and the expression of any other proteins encoded by the vector, such as antibiotic markers, should also be considered.
  • the gene constructs of the present invention can also be used as a part of a gene therapy protocol to deliver nucleic acids encoding either an agonistic or antagonistic form of a hedgehog polypeptide.
  • another aspect of the invention features expression vectors for in vivo transfection of a hedgehog polypeptide in particular cell types so as cause ectopic expression of a hedgehog polypeptide in an muscle tissue.
  • Formulations of such expression constructs may be administered in any biologically effective carrier, e.g. any formulation or composition capable of effectively delivering the recombinant gene to cells in vivo.
  • Approaches include insertion of the hedgehog coding sequence in viral vectors including recombinant retroviruses, adenovirus, adeno-associated virus, and herpes simplex virus- 1, or recombinant bacterial or eukaryotic plasmids.
  • Viral vectors transfect cells directly; plasmid DNA can be delivered with the help of, for example, cationic liposomes (lipofectin) or derivatized (e.g.
  • transduction of appropriate target cells represents the critical first step in gene therapy, choice of the particular gene delivery system will depend on such factors as the phenotype of the intended target and the route of administration, e.g. locally or systemically.
  • the particular gene construct provided for in vivo transduction of hedgehog expression are also useful for in vitro transduction of cells, such as for use in the ex vivo tissue culture systems described below.
  • a preferred approach for in vivo introduction of nucleic acid into a cell is by use of a viral vector containing nucleic acid, e.g. a cDNA, encoding the particular form of the hedgehog polypeptide desired.
  • a viral vector containing nucleic acid e.g. a cDNA
  • Infection of cells with a viral vector has the advantage that a large proportion of the targeted cells can receive the nucleic acid.
  • molecules encoded within the viral vector e.g., by a cDNA contained in the viral vector, are expressed efficiently in cells which have taken up viral vector nucleic acid.
  • Retrovirus vectors and adeno-associated virus vectors are generally understood to be the recombinant gene delivery system of choice for the transfer of exogenous genes in vivo, particularly into humans. These vectors provide efficient delivery of genes into cells, and the transferred nucleic acids are stably integrated into the chromosomal DNA of the host.
  • a major prerequisite for the use of retroviruses is to ensure the safety of their use, particularly with regard to the possibility of the spread of wild-type virus in the cell population.
  • the development of specialized cell lines (termed "packaging cells") which produce only replication-defective retroviruses has increased the utility of retroviruses for gene therapy, and defective retroviruses are well characterized for use in gene transfer for gene therapy purposes (for a review see Miller, A.D.
  • recombinant retrovirus can be constructed in which part of the retroviral coding sequence (gag, pol, env) has been replaced by nucleic acid encoding a hedgehog polypeptide and renders the retrovirus replication defective.
  • the replication defective retrovirus is then packaged into virions which can be used to infect a target cell through the use of a helper virus by standard techniques. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausubel, F.M. et al. (eds.) Greene Publishing Associates, (1989), Sections 9.10-9.14 and other standard laboratory manuals.
  • retroviruses examples include pLJ, pZIP, pWE and pEM which are well known to those skilled in the art.
  • suitable packaging virus lines for preparing both ecotropic and amphotropic retroviral systems include Crip, Cre, 2 and Am. Retroviruses have been used to introduce a variety of genes into many different cell types, including muscle cells, in vitro and/or in vivo (see for example Eglitis, et al. (1985) Science 230:1395- 1398; Danos and Mulligan (1988) Proc. Natl. Acad. Sci. USA 85:6460-6464; Wilson et al. (1988) Proc. Natl. Acad. Sci.
  • WO93/25234 and WO94/06920 are examples of retroviral vectors.
  • strategies for the modification of the infection spectrum of retroviral vectors include: coupling antibodies specific for cell surface antigens to the viral env protein (Roux et al. (1989) PNAS 86:9079-9083; Julan et al. (1992) J. Gen Virol 73:3251-3255; and Goud et al. (1983) Virology 163:251-254); or coupling cell surface receptor ligands to the viral env proteins (Neda et al. (1991) J Biol
  • Coupling can be in the form of the chemical cross-linking with a protein or other variety (e.g. lactose to convert the env protein to an asialoglycoprotein), as well as by generating fusion proteins (e.g. single-chain antibody/em' fusion proteins).
  • a protein or other variety e.g. lactose to convert the env protein to an asialoglycoprotein
  • fusion proteins e.g. single-chain antibody/em' fusion proteins
  • retroviral gene delivery can be further enhanced by the use of tissue- or cell-specific transcriptional regulatory sequences which control expression of the hedgehog gene of the retroviral vector.
  • adenovirus-derived vectors The genome of an adenovirus can be manipulated such that it encodes and expresses a gene product of interest but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. See for example Berkner et al. (1988) BioTechniques 6:616; Rosenfeld et al. (1991) Science 252:431-434; and Rosenfeld et al. (1992) Cell 68:143-155.
  • adenoviral vectors derived from the adenovirus strain Ad type 5 dl324 or other strains of adenovirus are well known to those skilled in the art.
  • Recombinant adenoviruses can be advantageous in certain circumstances in that they can be used to infect a wide variety of cell types, including muscle cells.
  • the virus particle is relatively stable and amenable to purification and concentration, and as above, can be modified so as to affect the spectrum of infectivity.
  • introduced adenoviral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced DNA becomes integrated into the host genome (e.g., retroviral DNA).
  • the carrying capacity of the adenoviral genome for foreign DNA is large (up to 8 kilobases) relative to other gene delivery vectors (Berkner et al. cited supra; Haj-Ahmand and Graham (1986) J. Virol. 57:267).
  • adenoviral vectors currently in use and therefore favored by the present invention are deleted for all or parts of the viral El and E3 genes but retain as much as 80% of the adenoviral genetic material (see, e.g., Jones et al. (1979) Cell 16:683; Berkner et al., supra; and Graham et al. in Methods in Molecular Biology, E.J. Murray, Ed. (Humana, Clifton, NJ, 1991) vol. 7. pp. 109-127).
  • Expression of the inserted hedgehog gene can be under control of, for example, the El A promoter, the major late promoter (MLP) and associated leader sequences, the E3 promoter, or exogenously added promoter sequences.
  • MLP major late promoter
  • non-viral methods can also be employed to cause expression of a hedgehog polypeptide in the tissue of an animal.
  • Most nonviral methods of gene transfer rely on normal mechanisms used by mammalian cells for the uptake and intracellular transport of macromolecules.
  • non-viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the hedgehog polypeptide gene by the targeted cell.
  • Exemplary gene delivery systems of this type include liposomal derived systems, poly-lysine conjugates, and artificial viral envelopes.
  • the gene delivery systems for the therapeutic hedgehog gene can be introduced into a patient by any of a number of methods, each of which is familiar in the art.
  • a pharmaceutical preparation of the gene delivery system can be introduced systemically, e.g. by intravenous injection, and specific transduction of the protein in the target cells occurs predominantly from specificity of transfection provided by the gene delivery vehicle, cell-type or tissue-type expression due to the transcriptional regulatory sequences controlling expression of the receptor gene, or a combination thereof.
  • initial delivery of the recombinant gene is more limited with introduction into the animal being quite localized.
  • the gene delivery vehicle can be introduced by catheter (see U.S. Patent 5,328,470) or by stereotactic injection (e.g.
  • a hedgehog expression construct can be delivered in a gene therapy construct to dermal cells by, e.g., electroporation using techniques described, for example, by Dev et al. ((1994) Cancer Treat Rev 20:105-115).
  • the pharmaceutical preparation of the gene therapy construct can consist essentially of the gene delivery system in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded.
  • the pharmaceutical preparation can comprise one or more cells which produce the gene delivery system.
  • the hedgehog or ptc therapeutic can be a "gene activation" construct which, by homologous recombination with a genomic DNA, alters the transcriptional regulatory sequences of an endogenous gene.
  • the gene activation construct can replace the endogenous promoter of a hedgehog gene with a heterologous promoter, e.g., one which causes consitutive expression of the hedgehog gene or which causes inducible expression of the gene under conditions different from the normal expression pattern of the gene.
  • Other genes in the patched signaling pathway can be similarly targeted.
  • a vareity of different formats for the gene activation constructs are available. See, for example, the Transkaryotic Therapies, Inc PCT publications WO93/09222. WO95/31560, WO96/2941 1, WO95/31560 and WO94/12650.
  • the nucleotide sequence used as the gene activation construct can be comprised of (1) DNA from some portion of the endogenous hedgehog gene (exon sequence, intron sequence, promoter sequences, etc.) which direct recombination and (2) heterologous transcriptional regulatory sequence(s) which is to be operably linked to the coding sequence for the genomic hedgehog gene upon recombination of the gene activation construct.
  • the construct may further include a reporter gene to detect the presence of the knockout construct in the cell.
  • the gene activation construct is inserted into a cell, and integrates with the genomic DNA of the cell in such a position so as to provide the heterologous regulatory sequences in operative association with the native hedgehog gene.
  • Such insertion occurs by homologous recombination, i.e., recombination regions of the activation construct that are homologous to the endogenous hedgehog gene sequence hybridize to the genomic DNA and recombine with the genomic sequences so that the construct is incorporated into the corresponding position of the genomic DNA.
  • recombination region or “targeting sequence” refer to a segment (i.e., a portion) of a gene activation construct having a sequence that is substantially identical to or substantially complementary to a genomic gene sequence, e.g., including 5' flanking sequences of the genomic gene, and can facilitate homologous recombination between the genomic sequence and the targeting transgene construct.
  • replacement region refers to a portion of a activation construct which becomes integrated into an endogenous chromosomal location following homologous recombination between a recombination region and a genomic sequence.
  • heterologous regulatory sequences e.g., which are provided in the replacement region, can include one or more of a variety elements, including: promoters (such as constitutive or inducible promoters), enhancers, negative regualtory elements, locus control regions, transcription factor binding sites, or combinations thereof.
  • Promoters/enhancers which may be used to control the expression of the targeted gene in vivo include, but are not limited to. the cytomegalovirus (CMV) promoter/enhancer (Karasuyama et al., 1989, J. Exp. Med., 169:13), the human ⁇ -actin promoter (Gunning et al. (1987) PNAS 84:4831-4835), the glucocorticoid-inducible promoter present in the mouse mammary tumor virus long terminal repeat (MMTV LTR) (Klessig et al. (1984) Mol. Cell Biol.
  • CMV cytomegalovirus
  • MMTV LTR mouse mammary tumor virus long terminal repeat
  • MoLV LTR Moloney murine leukemia virus
  • SV40 early or late region promoter Moloney murine leukemia virus
  • primer 1 includes a 5' non-coding region of the human Shh gene and is flanked by an AsuII and Clal restriction sites.
  • Primer 2 includes a portion of the 5' non- coding region immediately 3' to that present in primer 1.
  • the hedgehog gene sequence is flanked by XhoII and BamHI restriction sites. The purified amplimers are cut with each of the enzymes as appropriate.
  • the vector pCDNAl.l (Invitrogen) includes a CMV promoter.
  • the plasmid is cut with with AsuII, which cleaves just 3' to the CMV promoter sequence.
  • AsuII/Clal fragment of primer 1 is ligated to the AsuII cleavage site of the pcDNA vector.
  • the Clal/AsuII ligation destroys the AsuII site at the 3' end of a properly inserted primer 1.
  • the vector is then cut with BamHI. and an XhoII/BamHI fragment of primer 2 is ligated to the BamHI cleavage site.
  • the BamHI/XhoII ligation destroys the BamHI site at the 5' end of a properly inserted primer 2.
  • Individual colonies are selected, cut with AsuII and BamHI, and the size of the
  • the replacement region merely deletes a negative transcriptional control element of the native gene, e.g., to activate expression, or ablates a positive control element, e.g., to inhibit expression of the targeted gene.
  • compositions can be generated with, for example, compounds which bind to patched and alter its signal transduction activity, compounds which alter the binding and/or enzymatic activity of a protein (e.g., intracellular) involved in patched signal pathway, and compounds which alter the level of expression of a hedgehog protein, a patched protein or a protein involved in the intracellular signal transduction pathway of patched.
  • a protein e.g., intracellular
  • the availability of purified and recombinant hedgehog polypeptides facilitates the generation of assay systems which can be used to screen for drugs, such as small organic molecules, which are either agonists or antagonists of the normal cellular function of a hedgehog and/or patched protein, particularly their role in the pathogenesis of muscle cell proliferation and/or differentiation.
  • the assay evaluates the ability of a compound to modulate binding between a hedgehog polypeptide and a hedgehog receptor such as patched.
  • the assay merely scores for the ability of a test compound to alter the signal transduction acitity of the patched protein.
  • the compound of interest is contacted with a mixture including a hedgehog receptor protein (e.g., a cell expressing the patched receptor) and a hedgehog protein under conditions in which it is ordinarily capable of binding the hedgehog protein.
  • a composition containing a test compound e.g., a test compound.
  • Detection and quantification of receptor/hedgehog complexes provides a means for determining the test compound's efficacy at inhibiting (or potentiating) complex formation between the receptor protein and the hedgehog polypeptide.
  • the efficacy of the compound can be assessed by generating dose response curves from data obtained using various concentrations of the test compound.
  • a control assay can also be performed to provide a baseline for comparison. In the control assay, isolated and purified hedgehog polypeptide is added to the receptor protein, and the formation of receptor/ hedgehog complex is quantitated in the absence of the test compound.
  • a ptc therapeutic of the present invention is one which disrupts the association of patched with smoothened.
  • Agonist and antagonists of muscle cell growth can be distinguished, and the efficacy of the compound can be assessed, by subsequent testing with muscle cells, e.g., in culture.
  • the polypeptide utilized as a hedgehog receptor can be generated from the patched protein.
  • an exemplary screening assay includes all or a suitable portion of the patched protein which can be obtained from, for example, the human patched gene (GenBank U43148) or other vertebrate sources (see GenBank Accession numbers U40074 for chicken patched and U46155 for mouse patched), as well as from drosophila (GenBank Accession number M28999) or other invertebrate sources.
  • the patched protein can be provided in the screening assay as a whole protein (preferably expressed on the surface of a cell), or alternatively as a fragment of the full length protein which binds to hedgehog polypeptides, e.g., as one or both of the substantial extracellular domains (e.g. corresponding to residues Asnl20- Ser438 and/or Arg770-Trpl027 of the human patched protein - which are also potential antagonists of hedgehog-dependent signal transduction).
  • the patched protein can be provided in soluble form, as for example a preparation of one of the extracellular domains, or a preparation of both of the extracellular domains which are covalently connected by an unstructured linker (see, for example, Huston et al. (1988) PNAS 85:4879; and U.S. Patent No. 5,091,513).
  • the protein can be provided as part of a liposomal preparation or expressed on the surface of a cell.
  • the patched protein can derived from a recombinant gene, e.g., being ectopically expressed in a heterologous cell.
  • the protein can be expressed on oocytes, mammalian cells (e.g., COS, CHO, 3T3 or the like), or yeast cell by standard recombinant DNA techniques. These recombinant cells can be used for receptor binding, signal transduction or gene expression assays.
  • Marigo et al. (1996) Development 122:1225- 1233 illustrates a binding assay of human hedgehog to chick patched protein ectopically expressed in Xenopus laevis oocytes.
  • the assay system of Marigo et al. can be adapted to the present drug screening assays.
  • Shh binds to the patched protein in a selective, saturable, dose-dependent manner, thus demonstrating that patched is a receptor for Shh.
  • Complex formation between the hedgehog polypeptide and a hedgehog receptor may be detected by a variety of techniques. For instance, modulation of the formation of complexes can be quantitated using, for example, detectably labelled proteins such as radiolabelled, fluorescently labelled, or enzymatically labelled hedgehog polypeptides, by immunoassay, or by chromatographic detection.
  • detectably labelled proteins such as radiolabelled, fluorescently labelled, or enzymatically labelled hedgehog polypeptides
  • a fusion protein can be provided which adds a domain that allows the protein to be bound to a matrix.
  • glutathione-S-transferase/receptor (GST/receptor) fusion proteins can be adsorbed onto glutathione sepharose beads (Sigma Chemical, St. Louis, MO) or glutathione derivatized microtitre plates, which are then combined with the hedgehog polypeptide, e.g.
  • the test compound incubated under conditions conducive to complex formation, e.g. at physiological conditions for salt and pH, though slightly more stringent conditions may be desired.
  • the beads are washed to remove any unbound hedgehog polypeptide, and the matrix bead-bound radiolabel determined directly (e.g. beads placed in scintillant), or in the supernatant after the receptor y 'hedgehog complexes are dissociated.
  • the complexes can be dissociated from the bead, separated by SDS-PAGE gel, and the level of hedgehog polypeptide found in the bead fraction quantitated from the gel using standard electrophoretic techniques.
  • soluble portions of the hedgehog receptor protein can be immobilized utilizing conjugation of biotin and streptavidin.
  • biotinylated receptor molecules can be prepared from biotin-NHS (N-hydroxy- succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
  • biotinylated receptor molecules can be prepared from biotin-NHS (N-hydroxy- succinimide) using techniques well known in the art (e.g., biotinylation kit, Pierce Chemicals, Rockford, IL), and immobilized in the wells of streptavidin-coated 96 well plates (Pierce Chemical).
  • antibodies reactive with the hedgehog receptor but which do not interfere with hedgehog binding can be derivatized to the wells of the plate, and the receptor trapped in the wells by antibody conjugation.
  • preparations of a hedgehog polypeptide and a test compound are incubated in the receptor-presenting wells of the plate, and the amount of receptor v 'hedgehog complex trapped in the well can be quantitated.
  • Exemplary methods for detecting such complexes include immunodetection of complexes using antibodies reactive with the hedgehog polypeptide, or which are reactive with the receptor protein and compete for binding with the hedgehog polypeptide; as well as enzyme-linked assays which rely on detecting an enzymatic activity associated with the hedgehog polypeptide.
  • the enzyme can be chemically conjugated or provided as a fusion protein with the hedgehog polypeptide.
  • the hedgehog polypeptide can be chemically cross- linked or genetically fused with alkaline phosphatase, and the amount of hedgehog polypeptide trapped in the complex can be assessed with a chromogenic substrate of the enzyme, e.g. paranitrophenylphosphate.
  • a fusion protein comprising the hedgehog polypeptide and glutathione-S-transferase can be provided, and complex formation quantitated by detecting the GST activity using l-chloro-2,4-dinitrobenzene (Habig et al ( ⁇ 974) JBiol Chem 249:7130).
  • the protein to be detected in the complex can be "epitope tagged" in the form of a fusion protein which includes, in addition to the hedgehog polypeptide or hedgehog receptor sequence, a second polypeptide for which antibodies are readily available (e.g. from commercial sources).
  • the GST fusion proteins described above can also be used for quantification of binding using antibodies against the GST moiety.
  • Other useful epitope tags include myc-epitopes (e.g., see Ellison et al.
  • liposomal vesicles can be used to provide manipulatable and isolatable sources of the receptor.
  • both authentic and recombinant forms of the patched protein can be reconstituted in artificial lipid vesicles (e.g.
  • the readily available source of hedgehog proteins provided by the art also facilitates the generation of cell- based assays for identifying small molecule agonists/antagonists and the like.
  • cells which are sensitive to hedgehog induction e.g. /r ⁇ tc ⁇ e -expressing cells or other myoblast-derived cells sensitive to hedgehog induction
  • a hedgehog protein and a test agent of interest can be contacted with a hedgehog protein and a test agent of interest, with the assay scoring for anything from simple binding to the cell to modulation in hedgehog inductive responses by the target cell in the presence and absence of the test agent.
  • agents which produce a statistically significant change in hedgehog activities can be identified.
  • the cell-based assay scores for agents which disrupt association of patched and smoothened proteins, e.g., in the cell surface membrane or liposomal preparation.
  • cells which have been genetically engineered to ectopically express patched can be utilized for drug screening assays.
  • cells which either express low levels or lack expression of the patched protein e.g. Xenopus laevis oocytes, COS cells or yeast cells, can be genetically modified using standard techniques to ectopically express the patched protein, (see Marigo et al., supra).
  • the resulting recombinant cells can be utilized in receptor binding assays to identify agonist or anatagonsts of hedgehog binding. Binding assays can be performed using whole cells.
  • the recombinant cells of the present invention can be engineered to include other heterolgous genes encoding proteins involved in hedgehog-dependent siganl pathways.
  • the gene products of one or more of smoothened, costal-2 and/or fused can be co- expressed with patched in the reagent cell, with assays being sensitive to the functional reconstituion of the hedgehog signal transduction cascade.
  • liposomal preparations using reconstituted patched protein can be utilized.
  • Patched protein purified from detergent extracts from both authentic and recombinant origins can be reconstituted in in artificial lipid vesicles (e.g. phosphatidylcholine liposomes) or in cell membrane-derived vesicles (see, for example, Bear et al. (1992) Cell 68:809-818; Newton et al. (1983) Biochemistry 22:6110-6117; and Reber et al. (1987) J Biol Chem 262:11369-11374).
  • the lamellar structure and size of the resulting liposomes can be characterized using electron microscopy. External orientation of the patched protein in the reconstituted membranes can be demonstrated, for example, by immunoelectron microscopy.
  • the hedgehog protein binding activity of liposomes containing patched and liposomes without the protein in the presence of candidate agents can be compared in order to identify potential modulators of the hedgehog-patched interaction.
  • the hedgehog protein used in these cell-based assays can be provided as a purified source (natural or recombinant in origin), or in the form of cells/tissue which express the protein and which are co-cultured with the target cells.
  • the protein can be labelled by any of the above-mentioned techniques, e.g., fluorescently, enzymatically or radioactively, or detected by immunoassay.
  • functional assays can be used to identified modulators, i.e., agonists or antagonists, of hedgehog or patched activities.
  • modulators i.e., agonists or antagonists
  • a number of gene products have been implicated in /? ⁇ tc/2e ⁇ i-mediated signal transduction, including patched, the transcription factor cubitus interruptus (ci), the serine/threonine kinase fused (fu) and the gene products of costal-2, smoothened and suppressor of fused.
  • the G l genes encode putative transcription factors having zinc finger DNA binding domains (Orenic et al. (1990) Genes & Dev 4:1053-1067; Kinzler et al. (1990) Mol Cell Biol 10:634-642). Transcription of the G l gene has been reported to be upregulated in response to hedgehog in limb buds, while transcription of the GIB gene is downregulated in response to hedgehog induction (Marigo et al. (1996) Development 122:1225-1233). By selecting transcriptional regulatory sequences from such target genes, e.g.
  • Reporter gene based assays of this invention measure the end stage of the above described cascade of events, e.g., transcriptional modulation. Accordingly, in practicing one embodiment of the assay, a reporter gene construct is inserted into the reagent cell in order to generate a detection signal dependent on ptc signaling. To identify potential regulatory elements responsive to ptc signaling present in the transcriptional regulatory sequence of a target gene, nested deletions of genomic clones of the target gene can be constructed using standard techniques. See, for example, Current Protocols in Molecular
  • Cytokine 7:1-7 A nested set of DNA fragments from the gene's 5'-flanking region are placed upstream of a reporter gene, such as the luciferase gene, and assayed for their ability to direct reporter gene expression in patched expressing cells.
  • a reporter gene such as the luciferase gene
  • Host cells transiently transfected with reporter gene constructs can be scored for the induction of expression of the reporter gene in the presence and absence of hedgehog to determine regulatory sequences which are responsice to/? ⁇ tc/ ⁇ e ⁇ ?-dependent signalling.
  • a reporter gene construct is inserted into the reagent cell in order to generate a detection signal dependent on second messengers generated by induction with hedgehog protein.
  • the reporter gene construct will include a reporter gene in operative linkage with one or more transcriptional regulatory elements responsive to the hedgehog activity, with the level of expression of the reporter gene providing the /2eG?ge 2og-dependent detection signal.
  • the amount of transcription from the reporter gene may be measured using any method known to those of skill in the art to be suitable. For example, mRNA expression from the reporter gene may be detected using RNAse protection or RNA-based PCR, or the protein product of the reporter gene may be identified by a characteristic stain or an intrinsic activity.
  • the amount of expression from the reporter gene is then compared to the amount of expression in either the same cell in the absence of the test compound (or hedgehog) or it may be compared with the amount of transcription in a substantially identical cell that lacks the target receptor protein. Any statistically or otherwise significant difference in the amount of transcription indicates that the test compound has in some manner altered the signal transduction of the patched protein, e.g., the test compound is a potential ptc therapeutic.
  • the gene product of the reporter is detected by an intrinsic activity associated with that product.
  • the reporter gene may encode a gene product that, by enzymatic activity, gives rise to a detection signal based on color, fluorescence, or luminescence.
  • the reporter or marker gene provides a selective growth advantage, e.g., the reporter gene may enhance cell viability, relieve a cell nutritional requirement, and/or provide resistance to a drug.
  • reporter genes are those that are readily detectable.
  • the reporter gene may also be included in the construct in the form of a fusion gene with a gene that includes desired transcriptional regulatory sequences or exhibits other desirable properties.
  • reporter genes include, but are not limited to CAT (chloramphenicol acetyl transferase) (Alton and Vapnek (1979), Nature 282: 864-869) luciferase, and other enzyme detection systems, such as beta-galactosidase; firefly luciferase (deWet et al. (1987), Mol. Cell. Biol.
  • Transcriptional control elements which may be included in a reporter gene construct include, but are not limited to, promoters, enhancers, and repressor and activator binding sites. Suitable transcriptional regulatory elements may be derived from the transcriptional regulatory regions of genes whose expression is induced after modulation of a patched signal transduction pathway. The characteristics of preferred genes from which the transcriptional control elements are derived include, but are not limited to, low or undetectable expression in quiescent cells, rapid induction at the transcriptional level within minutes of extracellular simulation, induction that is transient and independent of new protein synthesis, subsequent shut-off of transcription requires new protein synthesis, and mRNAs transcribed from these genes have a short half-life. It is not necessary for all of these properties to be present.
  • second messenger generation can be measured directly in the detection step, such as mobilization of intracellular calcium, phospholipid metabolism or adenylate cyclase activity are quantitated, for instance, the products of phospholipid hydrolysis IP3, DAG or cAMP could be measured
  • PKA protein kinase A
  • High PKA activity has been shown to antagonize hedgehog signaling in these systems.
  • PKA acts directly downstream or in parallel with hedgehog signaling, it is possible that hedgehog signalling occurs via inhibition of PKA activity.
  • detection of PKA activity provides a potential readout for the instant assays.
  • the ptc therapeutic is a PKA inhibitor.
  • PKA inhibitors are known in the art, including both peptidyl and organic compounds.
  • the ptc therapeutic can be a 5-isoquinolinesulfonamide, such as represented in the general formula:
  • R j and R 2 each can independently represent hydrogen, and as valence and stability permit a lower alkyl, a lower alkenyl, a lower alkynyl, a carbonyl (such as a carboxyl, an ester, a formate, or a ketone), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an amino, an acylamino, an amido, a cyano, a nitro, an azido, a sulfate, a sulfonate, a sulfonamido, -(CH 2 ) m -R 8 , -(CH ) m -OH, -(CH 2 ) m -O-lower alkyl, -(CH 2 ) m -O-lower alkenyl, -(CH 2 ) n -O-(CH 2 ) m
  • R3 is absent or represents one or more substitutions to the isoquinoline ring such as a lower alkyl, a lower alkenyl, a lower alkynyl, a carbonyl (such as a carboxyl, an ester, a formate, or a ketone), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an amino, an acylamino, an amido, a cyano, a nitro, an azido, a sulfate, a sulfonate, a sulfonamido, -(CH 2 ) m -R 8 , -(CH 2 ) m -OH, -(CH 2 ) m -O-lower alkyl, -(CH 2 )
  • R 8 represents a substituted or unsubstituted aryl, aralkyl, cycloalkyl, cycloalkenyl, or heterocycle; and n and m are independently for each occurrence zero or an integer in the range of 1 to 6.
  • the PKA inhibitor is N-[2-((p-bromocinnamyl)amino)ethyl]- 5-isoquinolinesulfonamide (H-89; Calbiochem Cat. No. 371963), e.g., having the formula:
  • the PICA inhibitor is l-(5-isoquinolinesulfonyl)-2- methylpiperazine (H-7; Calbiochem Cat. No. 371955), e.g., having the formula:
  • the PKA inhibitor is KT5720 (Calbiochem Cat. No. 420315), having the structure
  • nucleoside analogs are also useful as PKA inhibitors.
  • the subject method can be carried out cyclic AMP analogs which inhibit the kinase activity of PKA, as for example, 8-bromo-cAMP or dibutyryl-cAMP
  • Exemplary peptidyl inhibitors of PKA activity include the PKA Heat Stable Inhibitor (isoform ⁇ ; see, for example, Calbiochem Cat. No. 539488, and Wen et al. (1995) J Biol Chem 270:2041).
  • Certain hedehog receptors may stimulate the activity of phospholipases.
  • Inositol lipids can be extracted and analyzed using standard lipid extraction techniques.
  • Water soluble derivatives of all three inositol lipids IPj, IP 2 , IP3 can also be quantitated using radiolabelling techniques or HPLC.
  • the mobilization of intracellular calcium or the influx of calcium from outside the cell may be a response to hedgehog stimulation or lack there of. Calcium flux in the reagent cell can be measured using standard techniques.
  • the choice of the appropriate calcium indicator, fluorescent, bioluminescent, metallochromic, or Ca ++ -sensitive microelectrodes depends on the cell type and the magnitude and time constant of the event under study (Borle (1990) Environ Health Perspect 84:45-56).
  • As an exemplary method of Ca ++ detection cells could be loaded with the Ca ++ sensitive fluorescent dye fura-2 or indo-1, using standard methods, and any change in Ca ++ measured using a fluorometer. In certain embodiments of the assay, it may be desirable to screen for changes in cellular phosphorylation.
  • the drosophila gene fused (fu) which encodes a serine/threonine kinase has been identified as a potential downstream target in hedgehog signaling. (Preat et al., 1990 Nature 347, 87-89; Therond et al. 1993, Mech. Dev. 44. 65- 80).
  • the ability of compounds to modulate serine/threonine kinase activation could be screened using colony immunoblotting (Lyons and Nelson (1984) Proc. Natl. Acad. Sci. USA 81 :7426-7430) using antibodies against phosphorylated serine or threonine residues.
  • the ptc therapeutic is an antisense molecule which inhibits expression of a protein involved in a patched-mediated signal transduction pathway.
  • a protein which are involved in patched signals such as fused, costal-2, smoothened and/or Gli genes
  • the ability of the patched signal pathway(s) to inhibit proliferation of a cell can be altered, e.g., potentiated or repressed.
  • antisense therapy refers to administration or in situ generation of oligonucleotide probes or their derivatives which specifically hybridize (e.g. bind) under cellular conditions with cellular mRNA and/or genomic DNA encoding a hedgehog protein, patched, or a protein involved in patched-mediated signal transduction.
  • the hybridization should inhibit expression of that protein, e.g. by inhibiting transcription and/or translation.
  • the binding may be by conventional base pair complementarity, or, for example, in the case of binding to DNA duplexes, through specific interactions in the major groove of the double helix.
  • antisense refers to the range of techniques generally employed in the art, and includes any therapy which relies on specific binding to oligonucleotide sequences.
  • an antisense construct of the present invention can be delivered, for example, as an expression plasmid which, when transcribed in the cell, produces RNA which is complementary to at least a unique portion of the target cellular mRNA.
  • the antisense construct is an oligonucleotide probe which is generated ex vivo and which, when introduced into the cell causes inhibition of expression by hybridizing with the mRNA and/or genomic sequences of a target gene.
  • oligonucleotide probes are preferably modified oligonucleotide which are resistant to endogenous nucleases, e.g. exonucleases and/or endonucleases, and is therefore stable in vivo.
  • nucleic acid molecules for use as antisense oligonucleotides are phosphoramidate, phosphothioate and methylphosphonate analogs of DNA (see also U.S. Patents 5,176,996; 5,264,564; and 5,256,775). Additionally, general approaches to constructing oligomers useful in antisense therapy have been reviewed, for example, by Van der Krol et al. (1988) Biotechniques 6:958-976; and Stein et al. (1988) Cancer Res 48:2659-2668.
  • antisense oligonucleotides for the use in the methods of the invention: (1) oligos should have a GC content of 50% or more; (2) avoid sequences with stretches of 3 or more G's; and (3) oligonucleotides should not be longer than 25-26 mers.
  • a mismatched control can be constructed. The controls can be generated by reversing the sequence order of the corresponding antisense oligonucleotide in order to conserve the same ratio of bases.
  • the ptc therapeutic can be an antisense construct for inhibiting the expression of patched, e.g., to mimic the inhibition of patched by hedgehog.
  • antisense constructs include:
  • the source of the hedgehog and ptc therapeutics to be formulated will depend on the particular form of the agent. Small organic molecules and peptidyl fragments can be chemically synthesized and provided in a pure form suitable for pharmaceutical/cosmetic usage. Products of natural extracts can be purified according to techniques known in the art. For example, the Cox et al. U.S. Patent 5,286,654 describes a method for purifying naturally occurring forms of a secreted protein and can be adapted for purification of hedgehog polypeptides. Recombinant sources of hedgehog polypeptides are also available. For example, the gene encoding hedgehog polypeptides, are known, ter alia, from PCT publications WO 95/18856 and WO 96/17924. Those of skill in treating muscle tissues can determine the effective amount of an hedgehog or ptc therapeutic to be formulated in a pharmaceutical or cosmetic preparation.
  • the hedgehog or ptc therapeutic formulations used in the method of the invention are most preferably applied in the form of appropriate compositions.
  • appropriate compositions there may be cited all compositions usually employed for systemically or topically administering drugs.
  • the pharmaceutically acceptable carrier should be substantially inert, so as not to act with the active component. Suitable inert carriers include water, alcohol polyethylene glycol, mineral oil or petroleum gel, propylene glycol and the like.
  • compositions of this invention an effective amount of the particular hedgehog or ptc therapeutic as the active ingredient is combined in intimate admixture with a pharmaceutically acceptable carrier, which carrier may take a wide variety of forms depending on the form of preparation desired for administration.
  • a pharmaceutically acceptable carrier which carrier may take a wide variety of forms depending on the form of preparation desired for administration.
  • These pharmaceutical compositions are desirable in unitary dosage form suitable, particularly, for administration orally, rectally, percutaneously, or by parenteral injection.
  • any of the usual pharmaceutical media may be employed such as, for example, water, glycols, oils, alcohols and the like in the case of oral liquid preparations such as suspensions, syrups, elixirs and solutions; or solid carriers such as starches, sugars, kaolin, lubricants, binders, disintegrating agents and the like in the case of powders, pills, capsules, and tablets. Because of their ease in administration, tablets and capsules represents the most advantageous oral dosage unit form, in which case solid pharmaceutical carriers are obviously employed.
  • the carrier will usually comprise sterile water, at least in large part, though other ingredients, for example, to aid solubility, may be included.
  • Injectable solutions may be prepared in which the carrier comprises saline solution, glucose solution or a mixture of saline and glucose solution.
  • Injectable suspensions may also be prepared in which case appropriate liquid carriers, suspending agents and the like may be employed. Also included are solid form preparations which are intended to be converted, shortly before use, to liquid form preparations.
  • the carrier optionally comprises a penetration enhancing agent and/or a suitable wetting agent, optionally combined with suitable additives of any nature in minor proportions, which additives do not introduce a significant deleterious effect on the skin.
  • preparations can be topically administered by other methods, for example, encapsulated in a temperature and/or pressure sensitive matrix or in film or solid carrier which is soluble in body fluids and the like for subsequent release, preferably sustained-release of the active component.
  • compositions usually employed for topically administering therapeuitcs e.g., creams, gellies, dressings, shampoos, tinctures, pastes, ointments, salves, powders, liquid or semiliquid formulation and the like.
  • Application of said compositions may be by aerosol e.g. with a propellent such as nitrogen carbon dioxide, a freon, or without a propellent such as a pump spray, drops, lotions, or a semisolid such as a thickened composition which can be applied by a swab.
  • a propellent such as nitrogen carbon dioxide, a freon
  • a propellent such as a pump spray
  • drops lotions
  • a semisolid compositions such as salves, creams, pastes, gellies, ointments and the like will conveniently be used.
  • Dosage unit form as used in the specification and claims herein refers to physically discreate units suitable as unitary dosages, each unit containing a predetermined quantity of active ingredient calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • dosage unit forms are tablets (including scored or coated tablets), capsules, pills, powders packets, wafers, injectable solutions or suspensions, teaspoonfuls, tablespoonfuls and the like, and segregated multiples thereof.
  • compositions known in the art preferably hypoallergic and pH controlled are especially preferred, and include toilet waters, packs, lotions, skin milks or milky lotions.
  • the preparations contain, besides the hedgehog or ptc therapeutic, components usually employed in such preparations. Examples of such components are oils, fats, waxes, surfactants, humectants, thickening agents, antioxidants, viscosity stabilizers, chelating agents, buffers, preservatives, perfumes, dyestuffs, lower alkanols, and the like. If desired, further ingredients may be incorporated in the compositions, e.g.
  • oils comprise fats and oils such as olive oil and hydrogenated oils; waxes such as beeswax and lanolin; hydrocarbons such as liquid paraffin, ceresin, and squalane; fatty acids such as stearic acid and oleic acid; alcohols such as cetyl alcohol, stearyl alcohol, lanolin alcohol, and hexadecanol; and esters such as isopropyl myristate, isopropyl palmitate and butyl stearate.
  • oils comprise fats and oils such as olive oil and hydrogenated oils; waxes such as beeswax and lanolin; hydrocarbons such as liquid paraffin, ceresin, and squalane; fatty acids such as stearic acid and oleic acid; alcohols such as cetyl alcohol, stearyl alcohol, lanolin alcohol, and hexadecanol; and esters such as isopropyl myristate, isopropyl palmitate and butyl
  • anionic surfactants such as sodium stearate, sodium cetylsulfate, polyoxyethylene laurylether phosphate, sodium N-acyl glutamate; cationic surfactants such as stearyldimethylbenzylammonium chloride and stearyltrimethylammonium chloride; ampholytic surfactants such as alkylaminoethylglycine hydrocloride solutions and lecithin; and nonionic surfactants such as glycerin monostearate, sorbitan monostearate, sucrose fatty acid esters, propylene glycol monostearate, polyoxyethylene oleylether, polyethylene glycol monostearate, polyoxyethylene sorbitan monopalmitate, polyoxyethylene coconut fatty acid monoethanolamide, polyoxypropylene glycol (e.g.
  • humectants include glycerin, 1,3-butylene glycol, and propylene glycol
  • examples of lower alcohols include ethanol and isopropanol
  • examples of thickening agents include xanthan gum, hydroxypropyl cellulose, hydroxypropyl methyl cellulose, polyethylene glycol and sodium carboxymethyl cellulose
  • examples of antioxidants comprise butylated hydroxytoluene, butylated hydroxyanisole, propyl gallate, citric acid and ethoxyquin
  • examples of chelating agents include disodium edetate and ethanehydroxy diphosphate
  • examples of buffers comprise citric acid, sodium citrate, boric acid, borax, and disodium hydrogen phosphate
  • examples of preservatives are methyl parahydroxybenzoate, ethyl parahydroxybenzoate, dehydroacetic acid, salicylic acid and benzo
  • compositions typically from 0.01 to 10%o in particular from 0.1 to 5% and more in particular from 0.2 to 2.5% of the active ingredient, e.g., of the hedgehog or ptc therapeutic, will be incorporated in the compositions.
  • active ingredient e.g., of the hedgehog or ptc therapeutic
  • the carrier for example consists of 1 to 20%, in particular 5 to 15%> of a humectant, 0.1 to 10%) in particular from 0.5 to 5% of a thickener and water; or said carrier may consist of 70 to 99%, in particular 20 to 95% of a surfactant, and 0 to 20%), in particular 2.5 to 15%> of a fat; or 80 to 99.9% in particular 90 to 99% of a thickener; or 5 to 15%> of a surfactant, 2-15% of a humectant, 0 to 80% of an oil, very small ( ⁇ 2%) amounts of preservative, coloring agent and/or perfume, and water.
  • the carrier for example consists of 2 to 10% of a lower alcohol,
  • the carrier typically consists of 10-50%> of oil, 1 to 10% of surfactant, 50-80%> of water and 0 to 3% of preservative and/or perfume.
  • all % symbols refer to weight by weight percentage.
  • compositions for use in the method of the present invention are those wherein the hedgehog or ptc therapeutic is formulated in liposome-containing compositions.
  • Liposomes are artificial vesicles formed by amphiphatic molecules such as polar lipids, for example, phosphatidyl cholines, ethanolamines and serines, sphingomyelins, cardiolipins, plasmalogens, phosphatidic acids and cerebiosides. Liposomes are formed when suitable amphiphathic molecules are allowed to swell in water or aqueous solutions to form liquid crystals usually of multilayer structure comprised of many bilayers separated from each other by aqueous material (also referred to as coarse liposomes).
  • a unilamellar vesicle Another type of liposome known to be consisting of a single bilayer encapsulating aqueous material is referred to as a unilamellar vesicle.
  • water-soluble materials are included in the aqueous phase during the swelling of the lipids they become entrapped in the aqueous layer between the lipid bilayers.
  • Water-soluble active ingredients such as, for example, various salt forms of a hedgehog polypeptide, are encapsulated in the aqueous spaces between the molecular layers.
  • the lipid soluble active ingredient of hedgehog or ptc therapeutic such as an organic mimetic, is predominantly incorporated into the lipid layers, although polar head groups may protude from the layer into the aqueous space.
  • the encapsulation of these compounds can be achieved by a number of methods.
  • the method most commonly used involves casting a thin film of phospholipid onto the walls of a flask by evaporation from an organic solvent. When this film is dispersed in a suitable aqueous medium, multilamellar liposomes are formed. Upon suitable sonication, the coarse liposomes form smaller similarly closed vesicles.
  • Water-soluble active ingredients are usually incorporated by dispersing the cast film with an aqueous solution of the compound. The unencapsulated compound is then removed by centrifugation, chromatography, dialysis or other art-known suitable procedures.
  • the lipid-soluble active ingredient is usually incorporated by dissolving it in the organic solvent with the phospholipid prior to casting the film. If the solubility of the material in the lipid phase is not exceeded or the amount present is not in excess of that which can be bound to the lipid, liposomes prepared by the above method usually contain most of the material bound in the lipid bilayers; separation of the liposomes from unencapsulated material is not required.
  • a particularly convenient method for preparing liposome formulated forms of hedgehog and ptc therapeutics is the method described in EP-A-253,619, incorporated herein by reference.
  • single bilayered liposomes containing encapsulated active ingredients are prepared by dissolving the lipid component in an organic medium, injecting the organic solution of the lipid component under pressure into an aqueous component while simultaneously mixing the organic and aqueous components with a high speed homogenizer or mixing means, whereupon the liposomes are formed spontaneously.
  • the single bilayered liposomes containing the encapsulated hedgehog or ptc therapeutic can be employed directly or they can be employed in a suitable pharmaceutically acceptable carrier for topical administration.
  • the viscosity of the liposomes can be increased by the addition of one or more suitable thickening agents such as, for example xanthan gum, hydroxypropyl cellulose, hydroxypropyl methylcellulose and mixtures thereof.
  • suitable thickening agents such as, for example xanthan gum, hydroxypropyl cellulose, hydroxypropyl methylcellulose and mixtures thereof.
  • the aqueous component may consist of water alone or it may contain electrolytes, buffered systems and other ingredients, such as, for example, preservatives.
  • Suitable electrolytes which can be employed include metal salts such as alkali metal and alkaline earth metal salts.
  • the preferred metal salts are calcium chloride, sodium chloride and potassium chloride.
  • the concentration of the electrolyte may vary from zero to 260 mM, preferably from 5 mM to 160 mM.
  • the aqueous component is placed in a suitable vessel which can be adapted to effect homogenization by effecting great turbulence during the injection of the organic component. Homogenization of the two components can be accomplished within the vessel, or, alternatively, the aqueous and organic components may be injected separately into a mixing means which is located outside the vessel. In the latter case, the liposomes are formed in the mixing means and then transferred to another vessel for collection purpose.
  • the organic component consists of a suitable non-toxic, pharmaceutically acceptable solvent such as, for example ethanol, glycerol, propylene glycol and polyethylene glycol, and a suitable phospholipid which is soluble in the solvent.
  • suitable phospholipids which can be employed include lecithin, phosphatidylcholine, phosphatydylserine, phosphatidylethanol-amine, phosphatidylinositol, lysophosphatidyl- choline and phospha-tidyl glycerol, for example.
  • Other lipophilic additives may be employed in order to selectively modify the characteristics of the liposomes. Examples of such other additives include stearylamine, phosphatidic acid, tocopherol, cholesterol and lanolin extracts.
  • ingredients which can prevent oxidation of the phospholipids may be added to the organic component.
  • examples of such other ingredients include tocopherol, butylated hydroxyanisole, butylated hydroxytoluene, ascorbyl palmitate and ascorbyl oleate.
  • Preservatives such a benzoic acid, methyl paraben and propyl paraben may also be added.
  • covers e.g. plasters, bandages, dressings, gauze pads and the like, containing an appropriate amount of a hedgehog or ptc therapeutic.
  • plasters, bandages, dressings, gauze pads and the like which have been impregnated with a topical formulation containing the therapeutic formulation.
  • Muscles are formed by the differentiation of mononucleate proliferative myoblasts into post-mitotic myocytes which subsequently fuse to form multinucleate muscle fibers.
  • muscle fibers differentiate in two waves: the first-formed primary fibers are generally slow, whereas later secondary fibers, which form in close association with primary fibers, are fast (Kelly and Rubinstein, 1980). These two fiber types are not spatially separated and, as they are formed over a considerable time period, the fate of individual cells as they mature has not been followed.
  • zebrafish by contrast, somitic muscle fibers form in two temporally-separated waves.
  • the early differentiating cells are formed medially near the notochord and migrate laterally during late somitogenesis to become slow muscle (Devoto et al., 1996; van Raamsdonk et al.. 1978). However, most somitic cells differentiate later and become fast muscle.
  • the differentiation of somites is central to vertebrate mesoderm development. Somites are epithelial balls of mesoderm that arise from a mesenchymal mass of proliferative paraxial tissue in a rostro-caudal order. Once formed, somites rapidly differentiate into a ventral sclerotomal mesenchymal compartment and a dorsal epithelial structure, the dermomyotome. In lower vertebrates, such as fish, in which the sclerotome is small (Morin-Kensicki and Eisen, 1997), the somite mainly gives rise to muscle. In amniotes, the dermomyotome contributes to trunk dermis and to several distinct populations of muscle cells. The dorsomedial lip of the dermomyotome, which is located next to the neural tube, forms the differentiated muscle of the myotome that arises between the sclerotome and dermomyotome.
  • Axial structures are important as their removal leads to cell death and somite regression (Rong et al., 1992; Detaillet and Le Douarin, 1983), and they can enhance both myogenesis and chondrogenesis (Kenny-Mobbs and Thorogood, 1987).
  • Notochord can induce myogenesis in some assays of myogenic induction (Buffinger and Stockdale, 1995; Gamel et al., 1995; Stern et al., 1995; Pownall et al., 1996), although ectopically- positioned notochords in chick embryos can induce sclerotome at the expense of myogenic tissue (Pourquie et al., 1993; Bober et al., 1994; Fan and Tessier-Lavigne, 1994; Goulding et al., 1994).
  • SHH is a signaling molecule expressed in notochord at all times when this tissue can influence muscle differentiation (Echelard et al., 1993; Krauss et al., 1993; Johnson et al., 1994; Roelink et al., 1994). SHH can substitute for notochord in various assays of both sclerotome and muscle induction (Fan et al., 1995; Munsterberg et al., 1995), and to induce ectopic muscle markers in vivo (Johnson et al., 1994; Concordet et al., 1996; Weinberg et al., 1996; Hammerschmidt et al., 1996).
  • mice homozygous for a targeted deletion of the shh gene have deficits in sclerotome and myotome precursor cell markers (Chiang et al., 1996). These data suggest that SHH may mediate notochord-dependent signals that induce myogenesis.
  • SHH may mediate notochord-dependent signals that induce myogenesis.
  • two lines of evidence argue against this simple view. First, both the MyoD and Myf-5 muscle specific transcription factors are still expressed in shh ⁇ ⁇ ⁇ mouse somites, although Myf-5 mRNA is reduced (Chiang et al., 1996). As Myf-5 and MyoD are myoblast markers in amniotes, this suggests that the myogenic program can be initiated in the absence of SHH.
  • mice with null mutations in members of the MyoD family of myogenic regulatory transcription factors suggest that several distinct populations of myogenic cells exist in different parts of the developing murine dermomyotome (Rudnicki et al., 1993; Tajbakhsh et al., 1997) and these populations appear to differ in their sensitivity to loss of SHH (Chiang et al., 1996).
  • neural tube In addition to notochord, neural tube also contains inductive signals that can support somitic myogenesis (Buffinger and Stockdale, 1995; Goulding et al., 1994; Rong et al., 1992; Stern and Hauschka, 1995; Generallet and Le Douarin, 1983), and dorsal neural tube can induce myogenesis, an effect that can be mimicked by some Wnt proteins (Gamel et al., 1995; Munsterberg et al., 1995; Stern et al., 1995). Moreover, inhibitory signals from lateral plate mesoderm and surface ectoderm have been suggested to influence myogenesis (Fan and Tessier-Lavigne, 1994; Pourquie et al., 1996).
  • the majority of the somite forms the third muscle cell population that both expresses myoD and differentiates later (Devoto et al., 1996; Weinberg et al., 1996).
  • Wild-type and heterozygote mutant breeding fish were maintained at 28.5 C on a 14-hour/ 10-hour light cycle.
  • floating head 1 flh
  • notaifol ⁇ O nl
  • boxozok ⁇ bo ⁇
  • boz' ⁇ fish exhibit a variable phenotype with defects ranging from reduced notochords to a severe lack of axial mesoderm at all rostro-caudal levels.
  • RNA injections were performed as described (Currie and Ingham, 1996). Immunohistochemistry
  • EB165 monoclonal ascites was used at 1 :5000 (Gardahaut et al., 1992).
  • First antibodies were detected with biotin-conjugated horse-derived anti- mouse IgG (Vector), Vectastain ABC Elite Peroxidase kit (Vector) and visualized using 0.5 mg/ml diaminobenzidine with (black stains) or without (brown stains) 0.03%> CoCl enhancement.
  • Cryosections for dual immunofluorescence had IgG first antibodies detected with Cappell goat anti-mouse IgG (y-specific) Texas red.
  • biotinylated BA-D5 prepared using Pierce NHS-Biotin reagent, was detected with Dako streptavidin-FITC. Sections were mounted in 150 mg/ml polyvinyl alcohol 30% glycerol PBS with DABCO antifade, and photographed by confocal microscopy.
  • Embryos were dechorinated, deyolked and homogenized manually on ice for 10 minutes in 63 mM Tris-HCl pH 6.8. 10% glycerol, 5% B-mercaptoethanol, 3.5% SDS. 0.2 mM PMSF, 0.5 M aprotinin, 0.5 M leupeptin. Samples were microfuged for 5 minutes at 4 C, 0.01% bromophenol blue added to the supernatant, the equivalent of 10 embryos run on each lane of a 7.5% acrylamide denaturing gel at 200mV for 30 mins and electroblotted onto nitrocellulose (Amersham). Purified bovine cardiac myosin was a kind gift of Dr. John Sleep.
  • Nitrocellulose strips were blocked in 5%> milk powder PBS/Az overnight, washed, and incubated with A4.0125 (1 :10), BA-D5 (1 :10), F1.652 (1 :10 (Webster, et al. 1988)) or EB 165 (1 :250) for 2h at RT. After washing, primary antibody was detected with horseradish peroxidase-conjugated sheep anti-mouse IgGF(ab)2 and an ECL kit (Amersham).
  • BA-D5 an antibody that specifically detects slow MyHC in muscle fibers of all ages of mammals and chicks examined (Schiaffino et al. (1989) and our unpublished observations), detects a single layer of cells in the superficial region of 24 hour zebrafish somites at all antero-posterior positions within the body axis.
  • EB 165 an antibody that detects fast fibers in embryonic and adult chicken muscle (Gardahaut et al.,
  • a third monoclonal antibody, A4.1025 which reacts with a conserved epitope near the ATP-binding site of all striated muscle MyHC isoforms examined in a wide variety of species (Dan-Goor et al., 1990), detects both the BA-D5+ and EB165+ populations of cells. All three antibodies reacted with muscle fibers in a striated pattern typical of sarcomeric myosin and Western analysis of 24 hour zebrafish extracts separated by SDS PAGE demonstrated that all three antibodies detect protein bands at or just under M r 200 000, the size of MyHC osoforms. Thus, these anti-MyHC antibodies distinguish slow and fast differentiated muscle cells in the zebrafish embryo.
  • the adaxial slow MyHC+ cells of the most anterior somites appear to migrate laterally, through the undifferentiated somitic mesoderm. Although it is possible that this apparent migration represents a wave of fiber type conversion, we think this unlikely from the earlier findings of Devoto et al. (1996) that early adaxial cells migrate and form slow muscle.
  • the wave of migration sweeps rapidly along the embryo from anterior to posterior so that by the 21 somite stage slow muscle cells of the anterior somites are located at the lateral edge of the somite under the epidermis, whereas slow muscle cells of mid-body somites are found in the center of the somite and the most posterior slow muscle cells are still in the adaxial position.
  • boz gene does not prevent muscle differentiation per se because a single fused somite of differentiated muscle is present beneath the neural tube, and this expresses fast MyHC.
  • Normal boz function is required for formation of slow muscle, rather than maintenance, as both severely and more mildly affected embryos from a boz heterozygote cross failed to express detectable slow MyHC at the 15 somite stage, whereas morphologically normal siblings showed normal slow MyHC expression (data not shown).
  • the absence of notochord in the boz mutant is accompanied by the specific loss of slow muscle.
  • ntl no tail mutant embryos in which midline mesodermal cells are present but fail to differentiate into mature notochord cells. Previous studies have shown that ntl embryos also lack muscle pioneer cells, a sub- population of the adaxial cells (Halpern et al., 1993).
  • ntl is the zebrafish homologue of the Brachyury transcription factor and is expressed in notochord but not in adaxial cells at the time of their differentiation and hence muscle defects are unlikely to be due to a cell-autonomous action of ntl in paraxial mesoderm (Schulte-Merker et al., 1994).
  • notochord precursors are present in anterior regions but absent posteriorly in the region beyond the yolk tube which is severely truncated (Halpern et al, 1993; Odenthal et al., 1996).
  • tfil ⁇ O f ls h for slow MyHC expression anticipating that the loss of notochordal maturation might prevent slow muscle formation.
  • ntl embryos demonstrate that although mature notochord is not necessary for slow muscle formation, severe defects in notochord establishment in the tail correlate with loss of slow muscle differentiation.
  • Sonic hedgehog induces ectopic slow muscle differentiation
  • SHH is a notochord- derived signal capable of inducing slow muscle at the expense of fast.
  • SHH could induce somitic cells to differentiate as slow muscle prematurely.
  • SHH might not affect the decision of when to differentiate, but simply determine what type of muscle is formed.
  • SHH ectopic SHH on earlier stage zebrafish embryos. In 15 somite zebrafish embryos, SHH induces a wide region of ectopic lateral differentiated muscle within the somite (46/53 unselected injected embryos). Ectopic slow muscle differentiation occurred without premature induction of fast muscle tissue. The premature differentiation of lateral muscle tissue suggested that SHH might induce presomitic mesoderm to differentiate early.
  • flh embryos for muscle differentiation and found that it occurs in an altered location.
  • Cells in the embryonic midline not those in the adaxial position are the first to differentiate in flh embryos. This differentiation is immediately beneath the presumptive floorplate that expresses SHH sporadically. Despite the unusual location of these muscle cells, they express slow but not fast MyHC, spread dorsally around the neural tube and ventrally in the midline and appear able to undergo lateral migration to take up a normal position beneath the ectoderm by 24 hours of development.
  • MyHC slow but not fast MyHC
  • SHH receptor Stone, et al. 1996), and is up-regulated adjacent to residual shh expression in flh embryos both at somitic and presomitic antero-posterior positions (Concordet et al., 1996). Therefore, mesodermal cells are first exposed to SHH long before muscle differentiation commences. Moreover, event the most recently differentiated muscle cells in flh embryos frequently do not express high levels of ptcl mRNA, despite adjacent mesoderm expressing ptcl abundantly. Thus, although SHH induces ptcl locally along the entire length of the flh embryo, there is a delay after SHH exposure before the appearance of differentiated slow muscle cells.
  • Sonic hedgehog induces adaxial slow myoblasts
  • Muscle is formed in two steps: mesodermal commitment to the proliferative myoblast, followed by terminal differentiation into the post-mitotic muscle fiber.
  • SHH is responsible for induction of slow muscle precursor cells, rather than the terminal differentiation of slow muscle er se.
  • the muscle-specific transcription factor myoD is initially detectable in adaxial precursors located adjacent to s z/z-expressing cells within the embryonic shield several hours before their terminal differentiation at around the time of somitogenesis (Weinberg et al. 1996). This expression oimyaD prior to terminal differentiation is also detected in adaxial cells at later stages when posterior somites arise from the tail bud.
  • zebrafish mutants like boz and ntl that lack slow muscle also lack the early adaxial myoD expression, and this correlates with a lack of axial SHH (Concordet et al., 1996; Odenthal et al., 1996; Weinberg, et al. 1996; Schier et al., 1997).
  • SHH signaling can induce premature myoD in lateral presomitic cells (Concordet et al., 1996; Hammerschmidt et al, 1996; Weinberg et al., 1996).
  • MyoD is the first MRF to be expressed in birds
  • Myf-5 is the earliest MRF to appear at high levels in mammalian somites (Ott et al., 1991)
  • Pax-3 can induce myogenesis (Tajbakhsh et al., 1997; Maroto et al., 1997).
  • SHH induces adaxial myoblasts that adopt a slow muscle fate.
  • SHH is not necessary for fast muscle formation.
  • Boz fish that lack SHH produce abundant fast muscle throughout the somite.
  • the normal izzyo -expressing myoblasts stripes across the posterior border of the somite form at the normal time just prior to somitogenesis in embryos that lack shh expression (Odenthal et al., 1996).
  • MyoD may mark commitment to a myoblast fate irrespective of the type of myoblast formed.
  • the delay is due to an intrinsically timed maturation of the somitic cells.
  • cell division is not extensive in zebrafish somites (Kimmel and Warga, 1987)
  • SHH might induce myoblasts committed to division followed by differentiation as it can be a somitic mitogen (Fan et al., 1995).
  • Mammalian myoblasts show such behavior in vitro (Quirm et al., 1985), which is reminiscent of the induction of division followed by terminal differentiation in Drosophila lamina ganglia neurons in response to retinal neuron-derived hedgehog (Huang and Kunes, 1996).
  • an extracellular signal(s) may control terminal differentiation.
  • zebrafish embryos may also express and embryonic myosin in both slow and fast fibers as the immunoreaction with the our all-myosin antibody was stronger than with the specific slow and fast antibodies.
  • both adaxial and non-adaxial cells react from their inception with an antibody that detects embryonic myosin (Devoto et al., 1996).
  • Amniote secondary fibers form overlying the neuromuscular junctions of primary fibers and it has been suggested that signals from the forming neuromuscular junction region may be required to initiate secondary fiber formation (Duxson et al., 1989). This is not the case in the zebrafish as absence of differentiated slow primary fibers does not prevent differentiation of fast muscle despite the striking correlation between the lateral migration of slow fibers and the differentiation of fast fibers. The converse relationship, that fast fiber differentiation might cause slow fiber migration, remains a possibility. Nevertheless, the close similarities between fish and amniote fiber generation suggest that the common ancestor had two steps of muscle patterning: early fibers being slow and later fast. There are further analogies between amniote and fish myogenesis.
  • Amniote primary fibers are of several distinct fiber types that prefigure later muscle characteristics (Crow and Stockdale, 1986), even though all express some form of slow MyHC (Kelly and Rubinstein, 1980; Vivarelli et al., 1988; Page et al., 1992; Hughes et al., 1993).
  • Slow adaxial cells in the zebrafish are also composed of two sub-populations, the muscle pioneer cells which express engrailed, and the non-pioneer adaxials. Engrailed proteins also mark a sub-population of muscle cells in the jaw muscle of the zebrafish (Hatta et al., 1990).
  • shh' ⁇ ⁇ mice have deficits in sclerotomal derivatives (Chiang et al., 1996).
  • over-expression of shh induces ectopic myoD expression, suggesting a myogenic action (Johnson et al., 1994; Concordet et al., 1996; Weinbert et al., 1996).
  • shh' ⁇ ' show defects in medial muscle formation (Chiang et al., 1996) and notochord can induce avian myogenesis (Pownall et al., 1996). So SHH May regulate formation of both ventral and more dorsal somitic tissues.
  • MRFs are the earliest known definitive myogenic markers. Expression of at least one MRF is obligatory for myogenesis in mice (Rudnicki et al., 1993). MRFs are expressed at low levels in presomitic mesoderm which has the capacity to form muscle in dissociated cell culture (George-Weinstein et al., 1994, Lin- Jones and Hauschka, 1996).
  • zebrafish SHH may function in vivo to suppress overt myogenic phenotypes in the lateral compartment that generates limb and body wall muscle and may have no homologous process in most zebrafish somites. So generation of further diversity within the dorsomedial myogenic compartment could be a role of SHH in amniote myogenesis. Distinct populations of slow and fast fibers may be present in amniote myogenesis. Distinct populations of slow and fast fibers may be present in amniote myotome (Dhoot. 1994). In this paper, we have shown that in zebrafish SHH regulates formation of myotomal slow muscle.
  • Much slow muscle in amniote limbs is located near developing bone that expresses Indian hedgehog (Vortkamp et al., 1996; Bitgood and McMahone. 1995).
  • Indian hedgehog Vortkamp et al., 1996; Bitgood and McMahone. 1995.
  • motoneurons which strongly influence muscle development, can express shh (Bitgood and McMahone 1995; Stone et al., 1996), raising the possibility that diverse hedgehog proteins may regulate muscle fiber diversification.
  • myf-5 is induced by the neural tube and MyoD by the dorsal ectoderm in mouse paraxial mesoderm. Development. 122: 429-437. Cossu, G., S. Tajbakhsh and M. Buckingham. 1996b. How is myogenesis initiated in the embryo? Trends Genet. 12: 218-222. Crow, M.T. and F.E. Stockdale. 1986. Myosin expression and specialization among the earliest muscle fibres of the developing avian limb. Dev.Biol. 113: 238-254. Currie. P.D. and P.W. Ingham. 1996.
  • Felsenfeld A.L.. M. Curry and C.B. Kimmcl. 19 1.
  • the fub-1 mutation blocks initial myofibril formation in zebrafish muscle pioneer cells.
  • Dev.Biol. 148 23-30.
  • Halves of epithelial somites and segmental plate show distinct muscle differentiation behavior in vitro compared to entire somites and segmental plate.
  • Dev. Biol 172 625-639. Gardahaut, M.F.. J. Fo ⁇ taine-Perus. T. Rouaud, E. Bandman and R. Ferrand. 1992.
  • Protein kinase A is a common negative regulator of hedgehog signalling in the vertebrate embryo. Genes Dev. 10: 647- 658.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Marine Sciences & Fisheries (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
EP98943462A 1997-08-29 1998-08-28 Regulierung von muskelgewebe mittels hedgehog-ähnlichen polypeptiden und dazu geeignete formulierungen und anwendungen Withdrawn EP1009424A2 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US5739497P 1997-08-29 1997-08-29
US57394P 1997-08-29
PCT/US1998/017922 WO1999010004A2 (en) 1997-08-29 1998-08-28 Regulation of muscle tissues by hedgehog-like polypeptides, and formulations and uses related thereto

Publications (1)

Publication Number Publication Date
EP1009424A2 true EP1009424A2 (de) 2000-06-21

Family

ID=22010324

Family Applications (1)

Application Number Title Priority Date Filing Date
EP98943462A Withdrawn EP1009424A2 (de) 1997-08-29 1998-08-28 Regulierung von muskelgewebe mittels hedgehog-ähnlichen polypeptiden und dazu geeignete formulierungen und anwendungen

Country Status (4)

Country Link
EP (1) EP1009424A2 (de)
AU (1) AU9125298A (de)
CA (1) CA2302780A1 (de)
WO (1) WO1999010004A2 (de)

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU8173098A (en) 1997-06-27 1999-01-19 Ontogeny, Inc. Neuroprotective methods and reagents
US7144997B2 (en) 1997-07-24 2006-12-05 Curis, Inc. Vertebrate embryonic patterning-inducing proteins, compositions and uses related therto
US6891082B2 (en) * 1997-08-01 2005-05-10 The Johns Hopkins University School Of Medicine Transgenic non-human animals expressing a truncated activintype II receptor
US6639051B2 (en) 1997-10-20 2003-10-28 Curis, Inc. Regulation of epithelial tissue by hedgehog-like polypeptides, and formulations and uses related thereto
US20030083242A1 (en) * 1998-11-06 2003-05-01 Alphonse Galdes Methods and compositions for treating or preventing peripheral neuropathies
US6884770B1 (en) 1998-11-06 2005-04-26 Curis, Inc. Methods and compositions for treating or preventing peripheral neuropathies
AU2724500A (en) * 1999-01-14 2000-08-01 Eli Lilly And Company Use of desert hedgehog protein
WO2000073337A1 (en) * 1999-06-01 2000-12-07 Biogen, Inc. Polymer conjugates of hedgehog proteins and uses
DE60017157T2 (de) * 1999-09-16 2005-12-22 Curis, Inc., Cambridge Vermittler von igel signalwegen, deren zusammenstellungen und verwendungen
US20070021493A1 (en) 1999-09-16 2007-01-25 Curis, Inc. Mediators of hedgehog signaling pathways, compositions and uses related thereto
US8852937B2 (en) 2000-03-30 2014-10-07 Curis, Inc. Small organic molecule regulators of cell proliferation
US7115653B2 (en) 2000-03-30 2006-10-03 Curis, Inc. Small organic molecule regulators of cell proliferation
US6683192B2 (en) * 2000-03-30 2004-01-27 Curis, Inc. Small organic molecule regulators of cell proliferation
US6683108B1 (en) * 2000-03-30 2004-01-27 Curis, Inc. Agonists of hedgehog signaling pathways and uses related thereto
US7498304B2 (en) 2000-06-16 2009-03-03 Curis, Inc. Angiogenesis-modulating compositions and uses
US7708998B2 (en) 2000-10-13 2010-05-04 Curis, Inc. Methods of inhibiting unwanted cell proliferation using hedgehog antagonists
AU2001296844B2 (en) 2000-10-13 2006-07-20 Curis, Inc. Hedgehog antagonists, methods and uses related thereto
WO2005097207A2 (en) 2004-03-26 2005-10-20 Curis, Inc. Rna interference modulators of hedgehog signaling and uses thereof
EP2247288A1 (de) * 2008-01-29 2010-11-10 Novartis AG Verwendung von hedgehog-agonisten zur behandlung von muskelskelett-erkrankungen

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE3942114C2 (de) * 1988-12-26 1997-09-04 Hiroyoshi Hidaka Verbindungen mit einer Wirkung für eine gleichmäßige Gefäßmuskelentspannung und pharmazeutische Zusammensetzung, die diese enthält
EP0741783B1 (de) * 1993-12-30 2009-03-18 President And Fellows Of Harvard College "hedgehog-like" proteine von wirbeltieren, die embryonale strukturen induzieren
US6281332B1 (en) * 1994-12-02 2001-08-28 The Johns Hopkins University School Of Medicine Hedgehog-derived polypeptides
WO1997004761A1 (en) * 1995-07-28 1997-02-13 Trustees Of Boston University Methods and compositions for treating cell proliferative disorders
WO1998048830A1 (en) * 1997-04-28 1998-11-05 Trustees Of The University Of Pennsylvania Method for promoting myogenesis using osteogenic proteins

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO9910004A3 *

Also Published As

Publication number Publication date
CA2302780A1 (en) 1999-03-04
WO1999010004A2 (en) 1999-03-04
WO1999010004A3 (en) 1999-05-27
AU9125298A (en) 1999-03-16

Similar Documents

Publication Publication Date Title
US6639051B2 (en) Regulation of epithelial tissue by hedgehog-like polypeptides, and formulations and uses related thereto
AU776265B2 (en) Methods and compositions for treating or preventing peripheral neuropathies
US20110124580A1 (en) Regulation of lung tissue by hedgehog-like polypeptides, and formulations and uses related thereto
US20040171533A1 (en) Methods and compositions for regulating adiopocytes
US20070048286A1 (en) Method of treating dopaminergic and GABA-nergic disorders
EP1009424A2 (de) Regulierung von muskelgewebe mittels hedgehog-ähnlichen polypeptiden und dazu geeignete formulierungen und anwendungen
EP1646395B1 (de) Verfahren und zusammensetzungen zur regulierung der lymphozytenaktivität
US6884770B1 (en) Methods and compositions for treating or preventing peripheral neuropathies
US6951839B1 (en) Methods and compositions for regulating lymphocyte activity

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20000329

AK Designated contracting states

Kind code of ref document: A2

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

RIN1 Information on inventor provided before grant (corrected)

Inventor name: HUGHES, SIMON, M.

Inventor name: INGHAM, PHILIP, W.

Inventor name: CURRIE, PETER, D.

Inventor name: BLADGEN, CHRIS, S.

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: CURIS, INC.

17Q First examination report despatched

Effective date: 20020606

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20021217

REG Reference to a national code

Ref country code: HK

Ref legal event code: WD

Ref document number: 1029743

Country of ref document: HK